WO2020029179A1 - Anti-factor xi antibodies - Google Patents

Anti-factor xi antibodies Download PDF

Info

Publication number
WO2020029179A1
WO2020029179A1 PCT/CN2018/099638 CN2018099638W WO2020029179A1 WO 2020029179 A1 WO2020029179 A1 WO 2020029179A1 CN 2018099638 W CN2018099638 W CN 2018099638W WO 2020029179 A1 WO2020029179 A1 WO 2020029179A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
fxi
antibodies
thrombosis
Prior art date
Application number
PCT/CN2018/099638
Other languages
French (fr)
Inventor
Wenyi Wang
Quan Yu
Xiaowu Liu
John Liuzhong Xu
Zhiqiang Du
Original Assignee
Shanghai Benemae Pharmaceutical Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Benemae Pharmaceutical Corporation filed Critical Shanghai Benemae Pharmaceutical Corporation
Priority to BR112021002472-7A priority Critical patent/BR112021002472A2/en
Priority to AU2018436195A priority patent/AU2018436195A1/en
Priority to CN202310301532.8A priority patent/CN116554334A/en
Priority to MX2021001613A priority patent/MX2021001613A/en
Priority to CA3108708A priority patent/CA3108708A1/en
Priority to CN202111466510.4A priority patent/CN114478781B/en
Priority to CN201880098606.XA priority patent/CN113227150B/en
Priority to EP18929685.8A priority patent/EP3833692A1/en
Priority to KR1020217006922A priority patent/KR20210042352A/en
Priority to JP2021506623A priority patent/JP2021534098A/en
Priority to PCT/CN2018/099638 priority patent/WO2020029179A1/en
Priority to CN202111467365.1A priority patent/CN114478782B/en
Priority to US16/537,427 priority patent/US11958911B2/en
Publication of WO2020029179A1 publication Critical patent/WO2020029179A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/36Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against blood coagulation factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This disclosure relates to antibodies capable of binding to the coagulation factor XI (FXI) and/or its activated form factor XIa (FXIa) , and to fragments of FXI and/or FXIa, and uses thereof, including uses as anticoagulation agents for treating thrombosis that do not compromise hemostasis.
  • FXI coagulation factor XI
  • FXIa activated form factor XIa
  • Thrombosis is a condition that involves blood clotting in a blood vessel, thereby blocking or obstructing blood flow in the affected area. This condition can lead to serious complications if the blood clots travel along the circulatory system to a crucial body part such as heart, brain, and lungs, causing heart attack, stroke, pulmonary embolism, etc.
  • Thrombosis is the major cause of most strokes and myocardial infarctions, deep vein thrombosis (DVT) , pulmonary embolism, and other cardiovascular events.
  • DVT deep vein thrombosis
  • 1, 2 Thrombosis can be treated or prevented by anticoagulants such as low-molecular-weight heparin, warfarin, and Factor Xa direct inhibitors.
  • anticoagulants such as low-molecular-weight heparin, warfarin, and Factor Xa direct inhibitors.
  • the most common adverse effect of these currently available therapies is impairing haemostasis.
  • antibodies that bind to coagulation factor XI (FXI) and/or its activated form factor XIa (FXIa) , and to fragments of FXI and/or FXIa.
  • the antibodies are monoclonal antibodies.
  • the antibodies are recombinant antibodies.
  • the antibodies are humanized antibodies.
  • the antibodies are immunologically active portions of immunoglobulin molecules, e.g., Fabs, Fvs, or scFvs.
  • the antibodies bind to the A3 domain of FXI and/or FXIa.
  • the antibodies include one or more CDRs consisting of or comprising the amino acid sequences of SEQ ID NOs: 11-16, 27-32, 43-48, 59-64, 75-80, 91-96, 107-112, 123-128, 139-144, 155-160, 171-176, and 187-192.
  • the pharmaceutical composition comprises one or more anti-FXI and/or anti-FXIa antibodies as disclosed herein.
  • the pharmaceutical composition further comprises one or more pharmaceutically acceptable adjuvants, carriers, excipients, preservatives, or a combination thereof.
  • nucleic acid encoding an anti-FXI and/or anti-FXIa antibody as disclosed herein, or a functional fragment of either antibody, as well as a vector comprising the nucleic acid, and a host cell comprising the vector.
  • the vector is an expression vector that is capable of producing the antibody or a functional fragment thereof encoded by the nucleic acid in a host cell.
  • kits comprising one or more anti-FXI and/or anti-FXIa antibodies as disclosed herein for use in treating and/or preventing thrombosis and/or complications or conditions associated with thrombosis.
  • the kit comprises a pharmaceutical composition comprising one or more anti-FXI and/or anti-FXIa antibodies as disclosed herein for use in treating and/or preventing thrombosis and/or complications or conditions associated with thrombosis.
  • the kit further comprises instructions for use.
  • the method includes administering to a subject in need thereof a therapeutically effective amount of one or more anti-FXI and/or anti-FXIa antibodies as disclosed herein.
  • the method includes administering to a subject in need thereof a therapeutically effective amount of a pharmaceutical composition containing an anti-FXI antibody, an anti-FXIa antibody, or a functional fragment of either antibody.
  • an anti-FXI and/or anti-FXIa antibody as disclosed herein formulating a medicament for treating and/or preventing thrombosis and/or complications or conditions associated with thrombosis.
  • an anti-FXI and/or anti-FXIa antibody as disclosed herein.
  • the method entails the steps of transforming a host cell with a vector comprising a nucleic acid encoding the antibody, and expressing the antibody in the host cell.
  • the method can further include purifying the expressed antibody from the host cell.
  • the purified antibody can be subjected to modifications such that the modified recombinant antibody retains the activity of the corresponding human antibody.
  • an antibody disclosed herein can be produced from culturing a hybridoma.
  • Figures 1A-1E illustrate the effects of five anti-FXI antibodies via APTT assay in human plasma.
  • Human plasma supplemented with five different antibodies at a concentration ranging from 0 to 400 nM were tested in an APTT assay as described in Example 3.
  • the five antibodies tested included 19F6 (A) , 34F8 (B) , 42A5 (C) , 1A6 (D) , and 14E11 (E) .
  • Antibodies 1A6 and 14E11 were used as positive controls in this experiment.
  • Figures 2A-2C illustrate the effects of antibodies 19F6 (A) , 34F8 (B) , and 42A5 (C) on the APTT assay in monkey plasma.
  • Figures 3A-3F illustrates SPR sensorgrams for FXI binding to immobilized h-19F6 (A) , h-34F8 (B) , and h-42A5 (C) , as well as SPR sensorgrams for FXIa binding to immobilized h-19F6 (D) , h-34F8 (E) , and h-42A5 (F) .
  • Data were fit with 1: 1 binding model, and curve fits at test concentrations of FXI (0.005 -1 ng/mL) are shown overlaid on the sensorgrams. Each curve indicates a different test concentration of FXI or FXIa.
  • Figures 4A-4C illustrate the concentration-response curves of antibodies h-19F6 (A) , h-34F8 (B) , and h-42A5 (C) inhibiting human FXIa from hydrolyzing S-2366.
  • Figures 5A-5B illustrate the inhibitory effects of antibodies h-19F6 (A) and h-42A5 (B) on FXIa-mediated activation of FIX to FIXa.
  • Human FIX 200 nM was incubated with FXIa (5 nM) in PBS with 5 mM CaCl 2 at room temperature with 1 ⁇ M h-19F6 or h-42A5. At the indicated intervals, samples were collected and the FIX as well as FIXa was determined by Western blots using goat anti-human FIX IgG (Affinity Biologicals) .
  • Figures 5C-5D illustrate the inhibitory effects of antibodies h-19F6 (C) and h-42A5 (D) on FXIIa-mediated activation of FXI to FXIa.
  • Human FXI 500 nM was incubated with FXIIa (50 nM) in the presence of 1 ⁇ M of h-19F6 or h-42A5.
  • FXI, as well as FXIa light chain, which represents FXIa production, at indicated time points was determined by Western blots.
  • a human IgG4 (1 ⁇ M) was used as the control.
  • Figures 6A-6C illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on APTT in cynomolgus monkeys.
  • the monkeys were intravenously administered with indicated doses of h-34F8 (A) , h-19F6 (B) , and h-42A5 (C) .
  • Ex vivo clotting time APTT was determined at pre-dose (time 0) , and 0.5, 1, 3, 6, 12, and 24 hours post-dose.
  • Figures 7A-7C illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on PT in cynomolgus monkeys.
  • Monkeys were intravenously administered with the indicated doses of h-34F8 (A) , h-19F6 (B) , and h-42A5 (C) .
  • Ex vivo clotting time PT was determined at pre-dose (time 0) , and 0.5, 1, 3, 6, 12, and 24 hours post-dose.
  • Figures 8A-8C illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on AV shunt thrombosis in cynomolgus monkeys.
  • Figures 9A-9C illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on bleeding time in cynomolgus monkeys.
  • Figures 10A-10B illustrate the antithrombotic effects of antibodies h-34F8, h-19F6, and h-42A5.
  • the time to 80%thrombotic occlusion (A) and to 100%thrombotic occlusion (B) were determined by monitoring the blood flow velocity.
  • Figures 11A-11D illustrate that the treatment with antibodies h-34F8, h-19F6, or h-42A5 did not prolong the bleeding time in monkeys.
  • the individual bleeding time in h-34F8, h-19F6, and h-42A5 treated group is shown in (A) , (B) and (C) , respectively.
  • the bleed time change upon vehicle, h-34F8, h-19F6, or h-42A5 treatment is shown in (D) .
  • Figures 12A-12B illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on clotting times of monkey plasma.
  • the APTT changes and PT changes are shown in (A) and (B) , respectively. **P ⁇ 0.01 and ***P ⁇ 0.001 vs. vehicle.
  • Figure 13 illustrates the amino acid sequence of human FXI (SEQ ID NO: 203) .
  • Figures 14A-14B illustrate the effects of modified antibodies h-19F6 (A) , and h-42A5 (B) on APTT in cynomolgus monkeys.
  • the monkeys were intravenously administered with indicated doses of modified h-19F6 and h-42A5.
  • Ex vivo clotting time APTT was determined at pre-dose (time 0) , and 0.5, 2, 6, 12, 24, 48, 96, 168, 240, and 336 hours post-dose.
  • Figures 15A-15B illustrate the effects of modified antibodies h-19F6 (A) , and h-42A5 (B) on PT in cynomolgus monkeys.
  • Monkeys were intravenously administered with the indicated doses of modified h-19F6 and h-42A5.
  • Ex vivo clotting time PT was determined at pre-dose (time 0) , and 0.5, 2, 6, 12, 24, 48, 96, 168, 240, and 336 hours post-dose.
  • Figures 16A-16B illustrate the effects of h-19F6 and h-42A5 on APTT and PT in human plasma.
  • Figure 16A shows the effects of h-19F6 and h-42A5 on APTT in human plasma.
  • Figure 16B shows the effects of h-19F6 and h-42A5 on PT in human plasma.
  • Figure 17 shows the binding specificity of test antibodies to human FXI.
  • 10 ⁇ L of human standard plasma or FXI-deficient plasma were served as FXI-positive and FXI-negative controls.
  • Figure 18 shows the effects of h-19F6 and h-42A5 in AV shunt thrombosis models on bleeding times recorded at pre-dose and 1-hour post-dose.
  • Figures 19A-19D show the binding properties of h-19F6 and h-42A5 to human FXI.
  • Figure 19A shows sensorgrams for h-19F6 captured on a sensor chip subjected to flows of indicated concentrations of FXI.
  • Figure 19B shows sensorgrams for h-42A5 captured on a sensor chip subjected to flows of indicated concentrations of FXI.
  • Figure 19C shows antibodies captured when test antibodies (5 ⁇ g/mL) flew through a sensor chip immobilized with equal amounts of 4 mutant FXIs in which the A1, A2, A3, or A4 domain was replaced with the corresponding domain from prekallikrein. A reported anti-FXI antibody, O1A6, was also tested a positive control.
  • Figure 19D shows that FXI was immobilized on a sensor chip.
  • H-19F6 and h-42A5 (5 ⁇ g/ml) were successively injected into flow cells on the sensor surface at a flow rate of 30 ⁇ l/minute, and the response change was monitored. The experiment was performed twice, and a representative result is depicted.
  • Figures 20A-20B show the binding properties of h-19F6 and h-42A5 to human FXIa.
  • Figure 20A shows sensorgrams for h-19F6 captured on a sensor chip subjected to flows of indicated concentrations of FXIa.
  • Figure 20B shows sensorgrams for h-42A5 captured on a sensor chip subjected to flows of indicated concentrations of FXIa.
  • the intrinsic pathway also called the contact activation pathway, is initiated by contact with a surface interface and results in activation of FXII.
  • the intrinsic pathway also involves FXI, FIX and FVIII.
  • the extrinsic pathway also called the tissue factor (TF) pathway, is initiated by vascular injury and results in the formation of an activated complex of TF-FVIIa. These two pathways meet and activate the common pathway, leading to conversion of prothrombin to thrombin and eventually the formation of cross-linked fibrin clot.
  • An ideal anticoagulant should be efficacious in preventing thrombosis without compromising haemostasis.
  • FXI can be activated by factor XIIa via the intrinsic pathway to FXIa, which in turn activates factor IX.
  • Epidemiological studies have suggested that FXI deficiency in humans is associated with decreased risk of venous thromboembolism and stroke, whereas increased FXI levels are associated with increased risk. 9-11
  • FXI-deficient humans show a very low bleeding tendency. 12, 13
  • mice deficient in FXI are protected against many types of thrombosis without increased bleeding.
  • small-molecule inhibitors, antibodies and antisense oligonucleotides that inhibit FXI have demonstrated antithrombotic properties with no bleeding risk in many animal models of thrombosis.
  • the antibodies disclosed herein binds to FXI and/or FXIa and target the intrinsic pathway of blood coagulation.
  • the structure of FXI and FXI’s involvement in blood coagulation have been reported in various publications. 33
  • FXI-deficient mice have been studied by many research teams and have displayed remarkable antithrombotic phenotypes in several models, including FeCl 3 -induced arterial and deep vein thrombosis models, a pulmonary embolism model, and a cerebral artery occlusion model. 14, 17, 22, 23
  • VTE venous thromboembolism
  • FXI thromboembolism
  • FXI-deficient mice do not show excessive bleeding, as their tail-bleeding times are comparable to those of wild-type animals. 23, 24 In addition, severely FXI-deficient patients do not exhibit spontaneous bleeding, although they may display a variable bleeding tendency during surgical operations. 12, 13 Combination of two or more anti-thrombotics are widely used clinically. A previous study showed that aspirin caused a similar bleeding tendency in wild-type and FXI-deficient mice, suggesting that targeting FXI might still be safe even in the presence of other anti-thrombotic therapies. 14
  • FXI/FXIa is a safe drug target for treating thrombosis-related diseases without compromising haemostasis.
  • many approaches have been applied to target FXI/FXIa for developing therapeutics for treating thrombosis, such as antibodies, oligonucleotides, and small-molecule inhibitors.
  • antibody-type blockers of FXI/FXIa were generated.
  • the advantages of antibodies include fast-acting properties and a low frequency of dosing, and a major weakness of antibodies is their potential immunogenicity. 25 At least two test antibodies were humanized before conducting in vivo studies.
  • FXI/FXIa inhibitors have prolonged APTT and exhibited antithrombotic effects in different models.
  • Anti-FXI antibody 14E11 increased APTT by approximately 1.3-fold and reduced thrombosis in exteriorized femoral arteriovenous shunts in baboons. 17
  • An antisense oligonucleotide inhibiting FXI expression reduced plasma FXI levels by approximately 50%and decreased thrombus formation in baboons. 26, 27
  • an orally bioavailable small-molecule FXIa inhibitor, ONO-5450598 significantly inhibited thrombosis formation in monkey models of thrombosis.
  • Bleeding risk is the most concerning issue in developing antithrombotic agents. As previously mentioned, FXI-deficient patients may show a bleeding tendency under surgical settings. It is unclear to what extent plasma FXI activity inhibition is still safe in terms of bleeding risk. As demonstrated in the working examples, the bleeding risk of intensive inhibition of FXI/FXIa by h-19F6 and h-42A5 was tested in the same monkeys used in thrombosis experiments. In AV shunt thrombosis animals, no bleeding tendency was observed as the treating dose of h-19F6 or h-42A5 escalated, suggesting that bleeding risk may be independent of the extent of FXI inhibition.
  • antibodies that bind to FXI, FXIa, and/or a fragment of FXI or FXIa and inhibit the formation of blood clot are capable of binding to FXI, FXIa, and/or a fragment of FXI or FXIa (e.g., a fragment comprising the A3 domain) and exhibiting an inhibitory effect at a concentration that is much lower than the maximum safety dose.
  • a dose of the antibody between 0.1 mg/kg i.v. and 3 mg/kg i.v. exhibits an inhibitory effect on conversion of FXI to FXIa in cynomolgus monkeys.
  • the antibodies disclosed herein can be used as anticoagulation agents with superior safety due to their minimal risk of causing bleeding versus conventional anticoagulation agents such as heparin.
  • anti-human FXI antibodies were generated by immunizing rats with human FXI to identify antibodies with anticoagulation properties. A dozen such antibodies were identified, and some of which were humanized for further development.
  • the humanized rat anti-human FXI antibodies such as h-19F6 and h-42A5 antibodies, were characterized in vitro and in vivo. In the in vitro studies, the humanized antibodies inhibited activated FXI (FXIa) -mediated hydrolysis of factor IX but not factor XIIa-induced FXI activation.
  • the binding properties of the antibodies to FXI were determined, and the dissociation constants (KD) for h-19F6 and h-42A5 were 22 pM and 35 pM, respectively. These two antibodies bind different sites in the A3 domain of FXI.
  • two distinct primate thrombosis models were used to evaluate the anti-thrombotic effects and bleeding risks of the humanized antibodies.
  • arteriovenous (AV) shunt thrombosis models both antibodies dose-dependently decreased thrombus formation without causing bleeding.
  • AV arteriovenous
  • FeCl3-induced thrombosis models both antibodies extended the time to thrombosis-mediated vessel occlusion, and neither antibody increased bleeding. The two antibodies showed anti-thrombotic efficacy without compromising haemostasis in primates, further confirming that targeting FXI can be used for treating thrombosis.
  • composition or method includes at least the recited elements.
  • consisting essentially of means that the composition or method includes the recited elements, and may further include one or more additional elements that do not materially affect the novel and basic characteristics of the composition or method.
  • a composition consisting essentially of recited elements may include those recited elements plus one or more trace contaminants from the isolation and purification method, pharmaceutically acceptable carriers such as phosphate buffered saline, preservatives, and the like.
  • Consisting of means the composition or method includes only the recited elements. Embodiments defined by each of the transitional terms are within the scope of this invention.
  • an antibody refers to an immunoglobulin molecule or an immunologically active portion thereof that specifically binds to, or is immunologically reactive with a particular antigen, for example, FXI, FXIa, or a particular domain or fragment of FXI or FXIa, e.g., the A3 domain.
  • an antibody for use in the present methods, compositions, and kits is a full-length immunoglobulin molecule, which comprises two heavy chains and two light chains, with each heavy and light chain containing three complementary determining regions (CDRs) .
  • antibody in addition to natural antibodies, also includes genetically engineered or otherwise modified forms of immunoglobulins, such as synthetic antibodies, intrabodies, chimeric antibodies, fully human antibodies, humanized antibodies, peptibodies and heteroconjugate antibodies (e.g., bispecific antibodies, multispecific antibodies, dual-specific antibodies, anti-idiotypic antibodies, diabodies, triabodies, and tetrabodies) .
  • the antibodies disclosed herein can be monoclonal antibodies or polyclonal antibodies.
  • an antibody is an immunologically active portion of an immunoglobulin molecule
  • the antibody may be, for example, a Fab, Fab’, Fv, Fab’F (ab’) 2 , disulfide-linked Fv, single chain Fv antibody (scFv) , single domain antibody (dAb) , or diabody.
  • the antibodies disclosed herein, including those that are immunologically active portion of an immunoglobulin molecule, retain the ability to bind a specific antigen, for example FXI or FXIa, or to bind a specific fragment of FXI or FXIa such as the A3 domain.
  • the anti-FXI and/or anti-FXIa antibodies disclosed herein have undergone post-translational modifications such as phosphorylation, methylation, acetylation, ubiquitination, nitrosylation, glycosylation, or lipidation associated with expression in a mammalian cell line, including a human or a non-human host cell.
  • post-translational modifications such as phosphorylation, methylation, acetylation, ubiquitination, nitrosylation, glycosylation, or lipidation associated with expression in a mammalian cell line, including a human or a non-human host cell.
  • polynucleotides or nucleic acids encoding the anti-FXI and/or anti-FXIa antibodies disclosed herein include DNA, mRNA, cDNA, plasmid DNA.
  • the nucleic acid encoding the antibody or a functional fragment thereof disclosed herein can be cloned into a vector, such as a pTT5 mammalian expression vector, which may further include a promoter and/or other transcriptional or translational control elements such that the nucleic acid can be expressed to produce the antibody or the functional fragment thereof.
  • nucleic acid (DNA) and/or amino acid (PRT) sequences including the sequences of the VH and VL and CDRs, of some examples of the antibodies disclosed herein are listed in Table 1 below.
  • humanized anti-FXI and/or anti-FXIa antibodies are provided in certain embodiments herein.
  • Various techniques are known in the art for humanizing antibodies from non-human species such that the antibodies are modified to increase their similarity to antibodies naturally occurring in humans.
  • Six CDRs are present in each antigen binding domain of a natural antibody. These CDRs are short, non-contiguous sequences of amino acids that are specifically positioned to form the antigen binding domain as the antibody assumes its three dimensional configuration.
  • the remainder of the amino acids in the antigen binding domains referred to as “framework” regions, show less inter-molecular variability and form a scaffold to allow correct positioning of the CDRs.
  • the antibodies or fragments disclosed herein have conserved sequences for CDR3 regions.
  • humanization of the antibodies disclosed herein can be accomplished by CDR grafting of monoclonal antibodies produced by immunizing mice or rats.
  • the CDRs of a mouse monoclonal antibody can be grafted into a human framework, which is subsequently joined to a human constant region to obtain a humanized antibody.
  • the human germline antibody sequence database, the protein data bank (PDB) , the INN (International Nonproprietary Names) database, and other suitable databases can be searched and the most similar frameworks to the antibodies can be identified by the search.
  • some back mutations to the donor residues are made in the human acceptor frameworks.
  • the variable regions are linked to a human IgG constant region.
  • human IgG1, IgG2, IgG3 and IgG4 Fc domains can be used. It is within the purview of one of ordinary skill in the art to humanize a monoclonal antibody produced by a non-human species based on the existing technology.
  • the antibodies provided herein include variants of the sequences disclosed herein that contain one or more mutations in their amino acid sequences while retaining binding affinity for FXI, FXIa, and/or a fragment thereof (e.g., a fragment comprising the A3 domain) .
  • the antibodies include a variable region having an amino acid sequence that is at least 85%, at least 90%, at least 95%, at least 98%, or at least 99%identical to a sequence selected from the group consisting of SEQ ID NOs: 9, 10, 25, 26, 41, 42, 57, 58, 73, 74, 89, 90, 105, 106, 121, 122, 137, 138, 153, 154, 169, 170, 185, 186, and 197-209, or a fragment thereof that retains binding affinity for FXI, FXIa, and/or a fragment thereof.
  • nucleic acids encoding antibodies that bind to FXI, FXIa, and/or a fragment thereof (e.g., a fragment comprising the A3 domain) .
  • the nucleic acids encoding the antibodies include a variable region having a nucleic acid sequence that is at least 85%, at least 90%, at least 95%, at least 98%, or at least 99%identical to a sequence selected from the group consisting of SEQ ID NOs: 1, 2, 17, 18, 33, 34, 49, 50, 65, 66, 81, 82, 97, 98, 113, 114, 129, 130, 145, 146, 161, 162, 177, and 178, or a fragment thereof that encodes a polypeptide with binding affinity for FXI, FXIa, and/or a fragment thereof.
  • the antibodies are further subjected to a strategic Chemistry, Manufacturing, and Control (CMC) development such that the novel antibodies such as monoclonal antibodies or humanized monoclonal antibodies disclosed herein are advanced from discovery to human clinical trials, and then to the pharmaceutical market.
  • CMC strategic Chemistry, Manufacturing, and Control
  • the modifications further improve the properties of the antibodies without compromising the immunological functions of the antibodies.
  • a CMC modified antibody is more stable under various temperatures (e.g., 4°C, 25°C, and 37°C) for an extended period of time (e.g., 3 days, 7 days, 14 days and 28 days) and under repeated freeze/thaw cycles (e.g., -40°C/25°C for up to 5 cycles) comparing to the unmodified antibody.
  • the CMC modified antibodies have an acceptable solubility.
  • certain amino acids can be potential oxidation and glycosylation sites. These amino acid residues at the potential oxidation, deamidation, or glycosylation sites may be mutated and additional residues in the proximity can also be mutated and/or optimized to maintain the 3D structure and function of a particular antibody.
  • one or more amino acid residues in a CDR region having the potential of oxidation, deamidation, or glycosylation are mutated to improve the stability of the antibody or a fragment thereof without compromising the immunological functions.
  • one or more Met residues in a CDR region having the potential of oxidation are mutated.
  • one or more Asn residues in a CDR region having the potential of deamidation are mutated.
  • the antibodies disclosed herein can be formulated into pharmaceutical compositions.
  • the pharmaceutical compositions may further comprise one or more pharmaceutically acceptable carriers, excipients, preservatives, or a combination thereof.
  • the pharmaceutical compositions can have various formulations, e.g., injectable formulations, lyophilized formulations, liquid formulations, etc. Depending on the formulation and administration route, one would select suitable additives, such as adjuvants, carriers, excipients, preservatives. 34
  • the pharmaceutical composition can be included in a kit with an instruction for using the composition.
  • a method of treating and/or preventing thrombosis in a subject suffering from thrombosis and/or at an elevated risk of developing thrombosis is also provided.
  • a method of inhibiting the formation of blood clots in a subject entail administering a therapeutically effective amount of an anti-FXI and/or FXIa antibody provided herein to intervene in the intrinsic pathway.
  • these methods comprise administering a pharmaceutical composition comprising an anti-FXI and/or anti-FXIa antibody as provided herein to the subject.
  • thrombosis causes or is associated with a number of complications or conditions, such as embolic stroke, venous thrombosis such as venous thromboembolism (VTE) , deep vein thrombosis (DVT) , and pulmonary embolism (PE) , arterial thrombosis such as acute coronary syndrome (ACS) , coronary artery disease (CAD) , and peripheral artery disease (PAD) .
  • embolic stroke venous thrombosis such as venous thromboembolism (VTE) , deep vein thrombosis (DVT) , and pulmonary embolism (PE)
  • VTE venous thromboembolism
  • VVT deep vein thrombosis
  • PE pulmonary embolism
  • PE pulmonary embolism
  • ACS acute coronary syndrome
  • CAD coronary artery disease
  • PAD peripheral artery disease
  • thrombosis Other conditions associated with thrombosis include, for example, high risk of VTE in surgical patients, immobilized patients, patients with cancer, patients with heart failure, pregnant patients, or patients having other medical conditions that may cause thrombosis.
  • the methods disclosed herein relate to a preventive anticoagulant therapy, that is, thromboprophylaxis. These methods entail administering to a subject suffering from a thrombosis-related complication disclosed above a therapeutically effective amount of an anti-FXI and/or FXIa antibody as disclosed herein or a therapeutically effective amount of a pharmaceutical composition comprising the anti-FXI and/or FXIa antibody.
  • the antibody or pharmaceutical composition can be administered either alone or in combination with any other therapy for treating or preventing the thrombosis-related complications or conditions.
  • the antibodies disclosed herein can be used as a secondary therapy in combination with other therapeutic agents for treating sepsis, such as antibiotics.
  • the term “subject” refers to mammalian subject, preferably a human.
  • a "subject in need thereof” refers to a subject who has been diagnosed with thrombosis or complications or conditions associated with thrombosis, or is at an elevated risk of developing thrombosis or complications or conditions associated with thrombosis.
  • the phrases “subject” and “patient” are used interchangeably herein.
  • treat, ” “treating, ” and “treatment” as used herein with regard to a condition refers to alleviating the condition partially or entirely, preventing the condition, decreasing the likelihood of occurrence or recurrence of the condition, slowing the progression or development of the condition, or eliminating, reducing, or slowing the development of one or more symptoms associated with the condition.
  • treating may refer to preventing or slowing the existing blood clot from growing larger, and/or preventing or slowing the formation of blood clot.
  • the term “treat, ” “treating, ” or “treatment” means that the subject has a reduced number or size of blood clots comparing to a subject without being administered with the antibodies or functional fragments thereof. In some embodiments, the term “treat, ” “treating, ” or “treatment” means that one or more symptoms of thrombosis and/or thrombosis-related conditions or complications are alleviated in a subject receiving an antibody or pharmaceutical composition as disclosed herein comparing to a subject who does not receive such treatment.
  • a “therapeutically effective amount” of an antibody or pharmaceutical composition as used herein is an amount of the antibody or pharmaceutical composition that produces a desired therapeutic effect in a subject, such as treating and/or preventing thrombosis.
  • the therapeutically effective amount is an amount of the antibody or pharmaceutical composition that yields maximum therapeutic effect.
  • the therapeutically effective amount yields a therapeutic effect that is less than the maximum therapeutic effect.
  • a therapeutically effective amount may be an amount that produces a therapeutic effect while avoiding one or more side effects associated with a dosage that yields maximum therapeutic effect.
  • a therapeutically effective amount for a particular composition will vary based on a variety of factors, including but not limited to the characteristics of the therapeutic composition (e.g., activity, pharmacokinetics, pharmacodynamics, and bioavailability) , the physiological condition of the subject (e.g., age, body weight, sex, disease type and stage, medical history, general physical condition, responsiveness to a given dosage, and other present medications) , the nature of any pharmaceutically acceptable carriers, excipients, and preservatives in the composition, and the route of administration.
  • the characteristics of the therapeutic composition e.g., activity, pharmacokinetics, pharmacodynamics, and bioavailability
  • the physiological condition of the subject e.g., age, body weight, sex, disease type and stage, medical history, general physical condition, responsiveness to a given dosage, and other present medications
  • the nature of any pharmaceutically acceptable carriers, excipients, and preservatives in the composition e.g., the nature of any pharmaceutically acceptable
  • a therapeutically effective amount of an antibody disclosed herein is in the range from about 0.01 mg/kg to about 30 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, from about 1 mg/kg to about 5 mg/kg.
  • the antibody or pharmaceutical composition can be administered continuously or intermittently, for an immediate release, controlled release or sustained release. Additionally, the antibody or pharmaceutical composition can be administered three times a day, twice a day, or once a day for a period of 3 days, 5 days, 7 days, 10 days, 2 weeks, 3 weeks, or 4 weeks. The antibody or pharmaceutical composition may be administered over a pre-determined time period. Alternatively, the antibody or pharmaceutical composition may be administered until a particular therapeutic benchmark is reached. In certain embodiments, the methods provided herein include a step of evaluating one or more therapeutic benchmarks to determine whether to continue administration of the antibody or pharmaceutical composition.
  • these methods entail the steps of cloning a nucleic acid encoding an anti-FXI and/or anti-FXIa antibody into a vector, transforming a host cell with the vector, and culturing the host cell to express the antibody.
  • the expressed antibody can be purified from the host cell using any known technique.
  • Various expression vectors such as pTT5 vector, and pcDNA3 vector, as well as various host cell lines such as CHO cells (e.g. CHO-K1 and ExpiCHO) , and HEK193T cells, can be used.
  • antibodies produced by the method disclosed above may have been subjected to one or more post-translational modifications.
  • Antibody preparation Animal immunization and hybridoma screening were performed at Genscript Inc. (Nanjing, China) , and the procedures that were applied to animals in this protocol were approved by the Genscript Institutional Animal Care and Use Committee. The experiment was performed in accordance with the approved guidelines. Wistar rats were immunized with human FXI, and splenocytes from animals with a good immune response were collected for the preparation of hybridomas, which were subjected to subcloning by limiting dilution. Finally, several monoclonal hybridoma clones expressing the desired anti-FXI antibodies, including 19F6, h-34F8 and 42A5, were obtained by using ELISA and functional screening.
  • 19F6, h-34F8 and 42A5 were subjected to humanization, resulting in three humanized antibodies, h-19F6 , h-34F8 and h-42A5, in an IgG4 form. These three humanized antibodies were produced in a transient mammalian expression system and purified by Protein G chromatography.
  • APTT Activated partial thromboplastin time
  • PT prothrombin time
  • monkey plasma diluted with an equal volume of phosphate-buffered saline (PBS) was used instead of the above mentioned human plasma-antibody mixture.
  • FXI activation by FXIIa Human FXI (500 nM) was pre-incubated at room temperature with 1 ⁇ M control IgG4 or h-19F6 or h-34F8 or h-42A5 in PBS for 5 minutes. At time zero, FXIIa, HK, and kaolin were added so that the final concentrations were FXI (250 nM) , FXIIa (50 nM) , HK (100 nM) , and kaolin (0.5 mg/mL) . At 0, 30, 60, 120 min intervals, 50- ⁇ L samples were collected into dodecyl sulfate sample buffer.
  • FXIa-mediated FIX activation Human FIX (200 nM) was incubated with FXIa (5 nM) in PBS containing 5 mM CaCl 2 at room temperature in the presence of 1 ⁇ M control lgG4, h-19F6, h-34F8, or h-42A5. At intervals of 0, 15, 30, 45, and 60 min, 50- ⁇ L samples were collected into dodecyl sulfate sample buffer. Samples were size-fractionated on 10%non-reducing gels and transferred to polyvinylidene fluoride membranes. Western blotting was performed to determine the FIX as well as FIXa levels using goat anti-human FIX IgG (Affinity Biologicals) . The image results were acquired using a ChemiDocMP Imaging System with Image Lab Software (Bio-Rad) .
  • SPR Surface plasmon resonance
  • test antibodies on FXI were first generated by replacing each apple domain (A1, A2, A3, and A4) with the corresponding domains from human prekallikrein. Equal amounts of each mutant were immobilized on a CM5 sensor chip, and test antibodies (33.3nM) were allowed to flow through the chip for 180 s for association and then for 1200 s for dissociation. The amounts of each antibody captured were recorded in response units (RU) using the Biacore Evaluation Software.
  • Epitope binding results of the test antibodies were also analyzed using the Biacore T200 system. Briefly, wild-type FXI with 6 ⁇ His tag was pre-immobilized on a CM5 sensor chip (GE Healthcare) , and h-19F6, h-34F8, or h-42A5 (5 ⁇ g/ml) was successively injected into flow cells on the sensor surface at a flow rate of 30 ⁇ l/minute to monitor the change in response.
  • Plasma samples were diluted with an equal volume of phosphate buffered saline (pH 7.4) and then subjected to APTT and PT analysis on an automatic analyser (CA660, Sysmex Inc. ) .
  • AV shunt thrombosis and bleeding time test A 30-min post-test antibody treatment was administered via intravenous bolus in cynomolgus monkeys. A tail vein bleeding time test was then performed, followed by thrombosis induction. Thrombosis was induced by connecting a shunt device between the femoral arterial and femoral venous cannulas containing a pre-weighed 10-cm-long thread. Blood was allowed to flow through the shunt for 10 min. The thrombus formed on the thread was weighed. Immediately after the removal of the shunt, blood samples were collected, and the next higher level of test antibody was administered. This bleeding/thrombosis process was carried out four times to dose the vehicle and three escalating levels (0.1, 0.3, 1 mg/kg) of test antibody in the same animal.
  • a 2-mL syringe was inserted into the tail vein of the animals.
  • the elapsed time was recorded manually as the bleeding time.
  • Ferric chloride (FeCl 3 ) -induced thrombosis and bleeding time test This animal experiment was performed at PharmaLegacy Laboratories Inc. (Shanghai, China) , and the procedures that were applied to animals in this protocol were approved by the PharmaLegacy Institutional Animal Care and Use Committee. The experiment was performed in accordance with the approved guidelines. Cynomolgus monkeys were pre-anaesthetized with 1.5 mg/kg of Zoletil, intubated, and ventilated with a respirator. Anaesthesia was maintained with isoflurane. Blood pressure, heart rate, and body temperature were monitored throughout the entire procedure.
  • the vehicle or 0.3 mg/kg of h-19F6, h-34F8, or h-42A5 was administered through a limb vein 2 hours before FeCl 3 application.
  • the left femoral artery was exposed and isolated via blunt dissection.
  • a Doppler flow probe was set up on the artery, and blood flow was continuously recorded.
  • blood flow was measured for at least 5 minutes.
  • two pieces of filter paper pre-soaked with 40%FeCl 3 were applied to the adventitial surface of the vessel upstream of the probe for 10 minutes. After the filter paper was removed, the site of application was washed with saline. Blood flow was continuously measured until it decreased to 0.
  • test antibodies (h-19F6, h-42A5, and 14E11) were first biotinylated using EZ-Link TM Sulfo-NHS-LC-Biotinylation Kit (Cat No. 21435, Thermo Fisher Inc. ) . These antibodies (25 ⁇ g each) were incubated with 200 ⁇ L of human standard plasma (Siemens Inc. ) or FXI-deficient plasma (Hyphen Biomed Inc. ) for 1h. Then 50 ⁇ L of Streptavidin-coated beads (Dynabeads TM M-280 Streptavidin, Thermo Fisher Inc.
  • mice and Wistar rats were immunized with human FXI, and splenocytes from the animals with good immune response were collected for the preparation of hybridomas, which were subjected to subcloning by limiting dilution. Twelve monoclonal hybridoma clones expressing desired anti-FXI antibodies 3G12, 5B2, 7C9, 7F1, 13F4, 19F6, 21F12, 34F8, 38E4, 42A5, 42F4, and 45H1 were obtained by using capture ELISA and functional screening.
  • V L and V H variable region of the light (V L ) and heavy chain (V H ) of these antibodies.
  • cDNAs encoding V L and V H were cloned from the corresponding hybridoma cells by standard RT-PCR procedures.
  • the V L and V H sequences of exemplary antibodies, including the sequences of CDRs, are shown in Table 1.
  • Example 3 Determination of anti-coagulation activity in human plasma using activated partial thromboplastin time (APTT) assay and prothrombin time (PT) assay
  • APTT assay measures the activity of the intrinsic and common pathways of coagulation; whereas PT assay measures the activity of the extrinsic and common pathways of coagulation.
  • the antibodies tested in these experiments were 19F6, 34F8, 42A5, 1A6 and 14E11. Antibodies 1A6 and 14E11 were used as positive controls in this experiment.
  • the sequences of the variable regions of the control antibodies were obtained from U.S. Patent No. 8,388,959 and U.S. Patent Application Publication No. 2013/0171144 and reformatted to IgG4. These antibodies were then expressed using ExpiCHO cell system.
  • the APTT assay and PT assay were performed as described above.
  • Example 4 Determination of the anti-coagulation activity in the plasma of non-human species using activated partial thromboplastin time (APTT) assay
  • the affinity of anti-FXI/FXIa antibodies to FXI/FXIa were determined using surface plasmon resonance (SPR) technology performed on the BIAcore T200 instrument.
  • the humanized antibodies were constructed by linking the variable regions of the antibodies disclosed herein to human IgG4 Fc domain and the recombinants were expressed in CHO cells. These antibodies were captured onto a Biacore CM5 sensor chip that was pre-immobilized with an anti-human IgG antibody.
  • the dissociation constants (K D ) of h-19F6, h-34F8, and h-42A5 to FXI and FXIa were calculated and detailed in Table 4.
  • the affinities of each antibody to FXI and FXIa are considered to be the same since the difference between them is less than 10 times.
  • the binding sites of 19F6 and 42A5 on FXI were determined using the SPR technology. Briefly, human IgG capture antibody was pre-immobilized on a Biacore CM5 sensor chip, and recombinant h-19F6 or h-42A5 was captured by flowing through the chip. An equal amount (15 relative units) of h-19F6 and h-42A5 was captured through adjustment of the antibody flowing time. Then wild type FXI or chimeric FXI in which individual apple domain was replaced with the corresponding domain from the human prekallikrein (FXI/PK chimeras) was allowed to flow through the chip for 180 seconds for association with h-19F6 or h-42A5, followed by a time period of 1800 seconds for dissociation.
  • FXI/PK chimeras human prekallikrein
  • the binding data was analyzed in a high performance kinetic mode as only one concentration of FXI, wild-type or chimeric, was tested in the SPR assay. Results showed that both h-19F6 and h-42A5 bound FXI as well as FXI/PK chimeras except when the A3 domain of FXI was replaced with the corresponding PK domain, indicating that part or the complete epitope of h-19F6 and h-42A5 on FXI is located in the A3 domain.
  • Human FXIa activity was determined by measuring the cleavage of a specific, chromogenic substrate, S-2366 (Diapharma Inc. ) .
  • S-2366 Diapharma Inc.
  • antibodies h-19F6, h-34F8 and h-42A5 were pre-incubated for 5 minutes at room temperature with a final concentration of 5 nM of FXIa in PBS (phosphate buffer saline) .
  • PBS phosphate buffer saline
  • S-2366 phosphate buffer saline
  • S-2366 phosphate buffer saline
  • Data were analyzed using the GraphPad Prism software and are shown in Figure 4.
  • the calculated apparent Ki for h-19F6, h-34F8, and h-42A5 are 0.67, 2.08, and 1.43 nM, respectively. Therefore, all three antibodies tested exhibited satisfying inhibitory effects on FXIa at a relatively low concentration.
  • the FXIa-mediated FIX activation was performed as described above.
  • Anti-FXI antibodies may modulate the intrinsic pathway by inhibiting FXI activation and/or by inhibiting FXIa activity.
  • both h-19F6 and h-42A5 reduced FIX activation in a concentration-dependent manner.
  • the inhibitory effect of these two antibodies on FXIa activity was further confirmed by using a chromogenic substrate of FXIa, S-2366. Both antibodies concentration-dependently inhibited the hydrolysis of S-2366 ( Figure 4) .
  • Example 10 Evaluation of the effects of anti-FXI antibodies on clotting time in cynomolgus monkeys
  • APTT test 50 ⁇ L of diluted plasma sample and 25 ⁇ L of APTT reagent (SMN 10445709, Symens Inc. ) were mixed and incubated at 37 °C for 4 min. Then 25 ⁇ L of CaCl 2 Solution (25 mM, SMN 10446232, Symens Inc. ) was added and time to clot formation was determined.
  • APTT reagent 25 mM, SMN 10446232, Symens Inc.
  • PT test 50 ⁇ L of diluted plasma sample was mixed with equal volume of PT reagent (SMN 10446442, Symens Inc. ) and incubated at 37°C and time to clot formation was determined. All three antibodies tested demonstrated dose-dependently increased APTT as shown in Figure 6 and none of them affected PT as shown in Figure 7.
  • Example 11 Evaluation of the effects of anti-FXI antibodies in arteriovenous (AV) shunt thrombosis and tail vein bleeding models in cynomolgus monkeys
  • Both thrombosis and bleeding time were assessed in the same animal for multiple doses of each antibody tested.
  • the antibodies included in this experiment were h-34F8, h-19F6, and h42A5. Briefly, bleeding time and thrombosis were sequentially evaluated at pre-dose and 30 minutes following each administration of the antibody. The bleeding/thrombosis assessments were conducted four times: pre-dose and post-dose at three escalating dose levels (0.1, 0.3 and 1 mg/kg) .
  • a shunt device containing a pre-weighed 10-cm long silk thread was applied to connecting the femoral arterial and femoral venous cannulae, and blood was allowed to flow through the shunt for 10 minutes. Then the thread was removed from the shunt and weighed again. Clot weight on the thread was calculated as the difference of the thread weight before and after blood flow.
  • a 2-mL syringe was inserted into the tail vein of the animals.
  • the elapsed time was recorded manually as the bleeding time.
  • Example 12 Evaluation of the effects of anti-FXI antibodies on ferric chloride–induced artery thrombosis and template bleeding time in cynomolgus monkeys
  • Cynomolgus monkeys were pre-anesthetized with 1.5 mg/kg of Zoletil, intubated, and ventilated with a respirator. Anesthesia was maintained with isoflurane. The blood pressure, heart rate, and body temperature were monitored throughout the entire procedure. The antibodies tested, including h-34F8, h-19F6, and h-42A5, or the vehicle control were administered through limb vein by injection 2 hours before FeCl 3 application. The left femoral artery was exposed and isolated via blunt dissection. A Doppler flow probe was set up on the artery and the blood flow was continuously recorded. Before applying FeCl 3 , the blood flow was measured for at least 5 minutes.
  • Example 13 Evaluation of the effects of modified anti-FXI antibodies on clotting time in cynomolgus monkeys for an extended period of time
  • modified antibodies disclosed herein did not show any adverse effects of prolonged bleeding while effectively inhibiting the intrinsic pathway of coagulation for an extended period of time, up to 14 days.
  • Example 14 Effects on clotting times of standard human plasma
  • Example 15 Binding properties of h-19F6 and h-42A5 to FXI
  • the affinities of h-19F6 and h-42A5 to FXI were determined using surface plasmon resonance (SPR) technology.
  • the test antibodies were captured on a sensor chip, and then indicated concentrations of FXI were allowed to flow through the chip.
  • Sensorgrams for h-19F6 ( Figure 19A) and h-42A5 ( Figure 19B) were obtained.
  • the dissociation constants for h-19F6 and h-42A5 were 22 and 36 pM, respectively ( Figures 19A and 19B) .
  • FXI is a homodimer consisting of 4 tandem apple domains (A1-4) and a catalytic domain.
  • A1-4 tandem apple domains
  • Four mutants of FXI were generated by replacing each apple domain with corresponding domains from human prekallikrein and tested the binding properties of h-19F6 or h-42A5 to the 4 mutants of FXI using SPR.
  • Equal amounts of the 4 mutant FXIs in which the A1, A2, A3, or A4 domain was replaced with the corresponding domain from prekallikrein were immobilized on a sensor chip, and test antibodies (5 ⁇ g/mL) were allowed to flow through the chip for association. The amounts of each antibody captured were recorded.
  • Example 16 Binding properties of h-19F6 and h-42A5 to FXIa
  • the antibodies bound to FXIa with the good affinities with which they bound to FXI ( Figures 20A and 20B) .
  • the affinities of h-19F6 and h-42A5 to FXI were determined using surface plasmon resonance (SPR) technology.
  • the dissociation constants for h-19F6 and h-42A5 were 26 and 81 pM, respectively ( Figures 20A and 20B) .
  • the test antibodies were captured on a sensor chip, and then indicated concentrations of FXIa were allowed to flow through the chip. Sensorgrams for h-19F6 ( Figure 20A) and h-42A5 ( Figure 20B) were obtained.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Hematology (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Diabetes (AREA)
  • Biophysics (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

Disclosed are antibodies thereof that bind to coagulation factor XI (FXI) and/or its activated form factor XIa (FXIa), or to fragments of FXI and/or FXIa, and compositions containing the antibodies. Also disclosed are methods of preparing the antibodies and use of the antibodies for treating and/or preventing coagulation associated conditions such as thrombosis and complications or conditions associated with thrombosis.

Description

ANTI-FACTOR XI ANTIBODIES TECHNICAL FIELD
This disclosure relates to antibodies capable of binding to the coagulation factor XI (FXI) and/or its activated form factor XIa (FXIa) , and to fragments of FXI and/or FXIa, and uses thereof, including uses as anticoagulation agents for treating thrombosis that do not compromise hemostasis.
BACKGROUND
Thrombosis is a condition that involves blood clotting in a blood vessel, thereby blocking or obstructing blood flow in the affected area. This condition can lead to serious complications if the blood clots travel along the circulatory system to a crucial body part such as heart, brain, and lungs, causing heart attack, stroke, pulmonary embolism, etc. Thrombosis is the major cause of most strokes and myocardial infarctions, deep vein thrombosis (DVT) , pulmonary embolism, and other cardiovascular events.  1, 2 Thrombosis can be treated or prevented by anticoagulants such as low-molecular-weight heparin, warfarin, and Factor Xa direct inhibitors. The most common adverse effect of these currently available therapies is impairing haemostasis.  3-5 Therefore, these therapies are limited by the dose and patient compliance because patients are required to be closely monitored after the treatment.
There is a need for an effective thrombosis therapy or prophylaxis with minimal side effects. This disclosure satisfies the need in the art.
SUMMARY
Provided herein in certain embodiments are antibodies that bind to coagulation factor XI (FXI) and/or its activated form factor XIa (FXIa) , and to fragments of FXI and/or FXIa. In some embodiments, the antibodies are monoclonal antibodies. In some embodiments, the antibodies are recombinant antibodies. In some embodiments, the antibodies are humanized antibodies. In some embodiments, the antibodies are immunologically active portions of immunoglobulin molecules, e.g., Fabs, Fvs, or scFvs. In some embodiments, the antibodies bind to the A3 domain of FXI and/or FXIa. In some embodiments, the antibodies include one or more  CDRs consisting of or comprising the amino acid sequences of SEQ ID NOs: 11-16, 27-32, 43-48, 59-64, 75-80, 91-96, 107-112, 123-128, 139-144, 155-160, 171-176, and 187-192.
Provided herein is a pharmaceutical composition for treating and/or preventing thrombosis and/or complications or conditions associated with thrombosis. The pharmaceutical composition comprises one or more anti-FXI and/or anti-FXIa antibodies as disclosed herein. In some embodiments, the pharmaceutical composition further comprises one or more pharmaceutically acceptable adjuvants, carriers, excipients, preservatives, or a combination thereof.
Provided herein is a nucleic acid encoding an anti-FXI and/or anti-FXIa antibody as disclosed herein, or a functional fragment of either antibody, as well as a vector comprising the nucleic acid, and a host cell comprising the vector. In some embodiments, the vector is an expression vector that is capable of producing the antibody or a functional fragment thereof encoded by the nucleic acid in a host cell.
Provided herein is a kit comprising one or more anti-FXI and/or anti-FXIa antibodies as disclosed herein for use in treating and/or preventing thrombosis and/or complications or conditions associated with thrombosis. Alternatively, the kit comprises a pharmaceutical composition comprising one or more anti-FXI and/or anti-FXIa antibodies as disclosed herein for use in treating and/or preventing thrombosis and/or complications or conditions associated with thrombosis. In certain embodiments, the kit further comprises instructions for use.
Provided herein is a method of treating and/or preventing thrombosis and/or complications or conditions associated with thrombosis. The method includes administering to a subject in need thereof a therapeutically effective amount of one or more anti-FXI and/or anti-FXIa antibodies as disclosed herein. Alternatively, the method includes administering to a subject in need thereof a therapeutically effective amount of a pharmaceutical composition containing an anti-FXI antibody, an anti-FXIa antibody, or a functional fragment of either antibody.
Provided herein is a use of an anti-FXI and/or anti-FXIa antibody as disclosed herein formulating a medicament for treating and/or preventing thrombosis and/or complications or conditions associated with thrombosis.
Provided herein is a method of producing an anti-FXI and/or anti-FXIa antibody as disclosed herein. The method entails the steps of transforming a host cell with a vector comprising a nucleic acid encoding the antibody, and expressing the antibody in the host cell. The method can further include purifying the expressed antibody from the host cell. Additionally, the purified antibody can be subjected to modifications such that the modified recombinant antibody retains the activity of the corresponding human antibody. Alternatively, an antibody disclosed herein can be produced from culturing a hybridoma.
BRIEF DESCRIPTION OF THE DRAWINGS
Figures 1A-1E illustrate the effects of five anti-FXI antibodies via APTT assay in human plasma. Human plasma supplemented with five different antibodies at a concentration ranging from 0 to 400 nM were tested in an APTT assay as described in Example 3. The five antibodies tested included 19F6 (A) , 34F8 (B) , 42A5 (C) , 1A6 (D) , and 14E11 (E) . Antibodies 1A6 and 14E11 were used as positive controls in this experiment.
Figures 2A-2C illustrate the effects of antibodies 19F6 (A) , 34F8 (B) , and 42A5 (C) on the APTT assay in monkey plasma. The monkey plasma supplemented with three different antibodies at a concentration ranging from 0 to 400 nM were tested in an APTT assay as described in Example 4.
Figures 3A-3F illustrates SPR sensorgrams for FXI binding to immobilized h-19F6 (A) , h-34F8 (B) , and h-42A5 (C) , as well as SPR sensorgrams for FXIa binding to immobilized h-19F6 (D) , h-34F8 (E) , and h-42A5 (F) . Data were fit with 1: 1 binding model, and curve fits at test concentrations of FXI (0.005 -1 ng/mL) are shown overlaid on the sensorgrams. Each curve indicates a different test concentration of FXI or FXIa.
Figures 4A-4C illustrate the concentration-response curves of antibodies h-19F6 (A) , h-34F8 (B) , and h-42A5 (C) inhibiting human FXIa from hydrolyzing S-2366.
Figures 5A-5B illustrate the inhibitory effects of antibodies h-19F6 (A) and h-42A5 (B) on FXIa-mediated activation of FIX to FIXa. Human FIX (200 nM) was incubated with FXIa (5 nM) in PBS with 5 mM CaCl 2 at room temperature with 1 μM h-19F6 or h-42A5. At the indicated intervals, samples were collected and the FIX as well as FIXa was determined by Western blots using goat anti-human FIX IgG (Affinity Biologicals) . Figures 5C-5D illustrate the inhibitory effects of antibodies h-19F6 (C) and h-42A5 (D) on FXIIa-mediated activation of FXI to FXIa. Human FXI (500 nM) was incubated with FXIIa (50 nM) in the presence of 1 μM of h-19F6 or h-42A5. FXI, as well as FXIa light chain, which represents FXIa production, at indicated time points was determined by Western blots. A human IgG4 (1 μM) was used as the control.
Figures 6A-6C illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on APTT in cynomolgus monkeys. The monkeys were intravenously administered with indicated doses of h-34F8 (A) , h-19F6 (B) , and h-42A5 (C) . Ex vivo clotting time APTT was determined at pre-dose (time 0) , and 0.5, 1, 3, 6, 12, and 24 hours post-dose.
Figures 7A-7C illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on PT in cynomolgus monkeys. Monkeys were intravenously administered with the indicated doses of h-34F8 (A) , h-19F6 (B) , and h-42A5 (C) . Ex vivo clotting time PT was determined at pre-dose (time 0) , and 0.5, 1, 3, 6, 12, and 24 hours post-dose.
Figures 8A-8C illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on AV shunt thrombosis in cynomolgus monkeys. Escalating levels of h-34F8 (A) , h-19F6 (B) , or h-42A5 (C) were intravenously administered to monkeys (n=3 for h-34F8 and h-19F6; n=4 for h-42A5) , changes of clot weight from pre-dose were determined in monkey model of AV shunt thrombosis. *P < 0.05, **P < 0.01 and ***P < 0.001 vs. Vehicle.
Figures 9A-9C illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on bleeding time in cynomolgus monkeys. Escalating levels of h-34F8 (A) , h-19F6 (B) , or h-42A5 (C) were intravenously administered to monkeys (n=3 for 34F8 and h-19F6; n=4 for h-42A5) , bleeding time was assessed at pre-dose and at 30 min post each dose.
Figures 10A-10B illustrate the antithrombotic effects of antibodies h-34F8, h-19F6, and h-42A5. Four groups of monkeys (n = 5) were intravenously administered with the vehicle, 0.3 mg/kg of h-34F8, h-19F6, or h-42A5, for 2 hours, and FeCl 3 was applied on the left femoral artery of each animal to induce thrombosis. The time to 80%thrombotic occlusion (A) and to 100%thrombotic occlusion (B) were determined by monitoring the blood flow velocity. *P <0.05 and **P < 0.01 vs. vehicle.
Figures 11A-11D illustrate that the treatment with antibodies h-34F8, h-19F6, or h-42A5 did not prolong the bleeding time in monkeys. Four groups of monkeys (n = 5) were intravenously administered with the vehicle, 0.3 mg/kg of h-34F8, h-19F6, or h-42A5, and template bleeding time was measured pre-dose and 1 hour post-dose. The individual bleeding time in h-34F8, h-19F6, and h-42A5 treated group is shown in (A) , (B) and (C) , respectively. The bleed time change upon vehicle, h-34F8, h-19F6, or h-42A5 treatment is shown in (D) .
Figures 12A-12B illustrate the effects of antibodies h-34F8, h-19F6, and h-42A5 on clotting times of monkey plasma. Four groups of monkeys (n = 5) were intravenously administered with the vehicle, 0.3 mg/kg of h-34F8, h-19F6, and h-42A5, respectively, and blood was collected pre-dose and about 3 hours post-dose for plasma preparation and clotting time APTT and PT determination. The APTT changes and PT changes are shown in (A) and (B) , respectively. **P < 0.01 and ***P < 0.001 vs. vehicle.
Figure 13 illustrates the amino acid sequence of human FXI (SEQ ID NO: 203) .
Figures 14A-14B illustrate the effects of modified antibodies h-19F6 (A) , and h-42A5 (B) on APTT in cynomolgus monkeys. The monkeys were intravenously administered with indicated doses of modified h-19F6 and h-42A5. Ex vivo clotting time APTT was determined at pre-dose (time 0) , and 0.5, 2, 6, 12, 24, 48, 96, 168, 240, and 336 hours post-dose.
Figures 15A-15B illustrate the effects of modified antibodies h-19F6 (A) , and h-42A5 (B) on PT in cynomolgus monkeys. Monkeys were intravenously administered with the indicated doses of modified h-19F6 and h-42A5. Ex vivo clotting time PT was determined at pre-dose (time 0) , and 0.5, 2, 6, 12, 24, 48, 96, 168, 240, and 336 hours post-dose.
Figures 16A-16B illustrate the effects of h-19F6 and h-42A5 on APTT and PT in human plasma. Figure 16A shows the effects of h-19F6 and h-42A5 on APTT in human plasma. Figure 16B shows the effects of h-19F6 and h-42A5 on PT in human plasma.
Figure 17 shows the binding specificity of test antibodies to human FXI. In Western blotting, 10 μL of human standard plasma or FXI-deficient plasma were served as FXI-positive and FXI-negative controls.
Figure 18 shows the effects of h-19F6 and h-42A5 in AV shunt thrombosis models on bleeding times recorded at pre-dose and 1-hour post-dose.
Figures 19A-19D show the binding properties of h-19F6 and h-42A5 to human FXI. Figure 19A shows sensorgrams for h-19F6 captured on a sensor chip subjected to flows of indicated concentrations of FXI. Figure 19B shows sensorgrams for h-42A5 captured on a sensor chip subjected to flows of indicated concentrations of FXI. Figure 19C shows antibodies captured when test antibodies (5 μg/mL) flew through a sensor chip immobilized with equal amounts of 4 mutant FXIs in which the A1, A2, A3, or A4 domain was replaced with the corresponding domain from prekallikrein. A reported anti-FXI antibody, O1A6, was also tested a positive control. Figure 19D shows that FXI was immobilized on a sensor chip. H-19F6 and h-42A5 (5 μg/ml) were successively injected into flow cells on the sensor surface at a flow rate of 30 μl/minute, and the response change was monitored. The experiment was performed twice, and a representative result is depicted.
Figures 20A-20B show the binding properties of h-19F6 and h-42A5 to human FXIa. Figure 20A shows sensorgrams for h-19F6 captured on a sensor chip subjected to flows of indicated concentrations of FXIa. Figure 20B shows sensorgrams for h-42A5 captured on a sensor chip subjected to flows of indicated concentrations of FXIa.
DETAILED DESCRIPTION
The following description of the invention is merely intended to illustrate various embodiments of the invention. As such, the specific modifications discussed are not to be construed as limitations on the scope of the invention. It will be apparent to one skilled in the art that various equivalents, changes, and modifications may be made without departing from the  scope of the invention, and it is understood that such equivalent embodiments are to be included herein.
Both intrinsic pathway and extrinsic pathway are involved in in vivo blood coagulation cascades. The intrinsic pathway, also called the contact activation pathway, is initiated by contact with a surface interface and results in activation of FXII. The intrinsic pathway also involves FXI, FIX and FVIII. The extrinsic pathway, also called the tissue factor (TF) pathway, is initiated by vascular injury and results in the formation of an activated complex of TF-FVIIa. These two pathways meet and activate the common pathway, leading to conversion of prothrombin to thrombin and eventually the formation of cross-linked fibrin clot. An ideal anticoagulant should be efficacious in preventing thrombosis without compromising haemostasis. Several lines of evidence suggest that the intrinsic coagulation pathway is important for the amplification phase of coagulation, whereas extrinsic and common pathways are more heavily involved in the initiation and propagation phases.  5-8 These findings indicate that the intrinsic pathway plays a minor role during normal haemostasis and that the inhibition of intrinsic pathway may provide antithrombotic benefits with low bleeding risk. FXI, a component of the intrinsic pathway, has recently become an attractive target, as it may have the potential to elicit anti-thrombosis effects without affecting bleeding.  3, 5, 6
FXI can be activated by factor XIIa via the intrinsic pathway to FXIa, which in turn activates factor IX. Epidemiological studies have suggested that FXI deficiency in humans is associated with decreased risk of venous thromboembolism and stroke, whereas increased FXI levels are associated with increased risk.  9-11 In addition, FXI-deficient humans show a very low bleeding tendency.  12, 13 Furthermore, mice deficient in FXI are protected against many types of thrombosis without increased bleeding.  14 Moreover, small-molecule inhibitors, antibodies and antisense oligonucleotides that inhibit FXI have demonstrated antithrombotic properties with no bleeding risk in many animal models of thrombosis.
The antibodies disclosed herein binds to FXI and/or FXIa and target the intrinsic pathway of blood coagulation. The structure of FXI and FXI’s involvement in blood coagulation have been reported in various publications.  33
Animal and clinical studies have suggested a robust association between FXI and thrombosis. FXI-deficient mice have been studied by many research teams and have displayed remarkable antithrombotic phenotypes in several models, including FeCl 3-induced arterial and deep vein thrombosis models, a pulmonary embolism model, and a cerebral artery occlusion model.  14, 17, 22, 23 In human epidemiological studies, patients with congenital FXI deficiency are insusceptible to venous thromboembolism (VTE) orischaemic stroke, and subjects with higher levels of FXI are at greater risk for VTE and ischaemic stroke than those with lower levels.  9-11 For physiological haemostasis, the role of FXI appears dispensable. FXI-deficient mice do not show excessive bleeding, as their tail-bleeding times are comparable to those of wild-type animals.  23, 24 In addition, severely FXI-deficient patients do not exhibit spontaneous bleeding, although they may display a variable bleeding tendency during surgical operations.  12, 13 Combination of two or more anti-thrombotics are widely used clinically. A previous study showed that aspirin caused a similar bleeding tendency in wild-type and FXI-deficient mice, suggesting that targeting FXI might still be safe even in the presence of other anti-thrombotic therapies.  14
All of the above-mentioned findings indicate that FXI/FXIa is a safe drug target for treating thrombosis-related diseases without compromising haemostasis. Thus, many approaches have been applied to target FXI/FXIa for developing therapeutics for treating thrombosis, such as antibodies, oligonucleotides, and small-molecule inhibitors.  5 As described herein, antibody-type blockers of FXI/FXIa were generated. The advantages of antibodies include fast-acting properties and a low frequency of dosing, and a major weakness of antibodies is their potential immunogenicity.  25 At least two test antibodies were humanized before conducting in vivo studies. Two humanized antibodies, h-19F6 and h-42A5, demonstrated very high affinity to human FXI/FXIa. Interestingly, they bind different regions but the same domain (A3) of FXI. Without bound by any theory, the antibodies might both inhibit FXIa activity but have no effect on FXI activation mediated by either FXIIa or thrombin.
Various types of FXI/FXIa inhibitors have prolonged APTT and exhibited antithrombotic effects in different models. Anti-FXI antibody 14E11 increased APTT by approximately 1.3-fold and reduced thrombosis in exteriorized femoral arteriovenous shunts  in baboons.  17 An antisense oligonucleotide inhibiting FXI expression reduced plasma FXI levels by approximately 50%and decreased thrombus formation in baboons.  26, 27 In addition, an orally bioavailable small-molecule FXIa inhibitor, ONO-5450598, significantly inhibited thrombosis formation in monkey models of thrombosis.  28 Furthermore, the antithrombotic effects of therapeutics targeting FXI/FXIa have also been confirmed in many non-primate animal models, such as mouse and rabbit thrombosis models.  19, 29-31 A recent clinical trial showed that an antisense oligonucleotide targeting FXI prevented venous thrombosis in patients undergoing knee arthroplasty.  32 As demonstrated in the working examples, two distinctive primate thrombosis models were used to evaluate the antithrombotic effects of h-19F6 and h-42A5. In the AV shunt thrombosis models, both antibodies decreased thrombosis formation in a dose-dependent manner. In FeCl 3-induced thrombosis models, both antibodies extended the time for thrombosis-led vessel occlusion. These results provide further evidence of the anti-thrombotic roles of FXI/FXIa inhibitors. The dose-dependent reduction of thrombosis formation for h-19F6 and h-42A5 in AV shunt thrombosis models suggest that thrombosis formation may negatively correlate with the degree of FXI inhibition, which can be indicated by APTT prolongation. Because h-42A5 is more potent than h-19F6 in prolonging APTT, the comparison of antithrombotic effects between h-42A5 and h-19F6 in the FeCl 3-induced thrombosis models could also lead to such an indication. Thus, more intense inhibition of FXI/FXIa, as indicated by a longer APTT, by FXI/FXIa inhibitors may result in better anti-thrombotic outcomes.
Bleeding risk is the most concerning issue in developing antithrombotic agents. As previously mentioned, FXI-deficient patients may show a bleeding tendency under surgical settings. It is unclear to what extent plasma FXI activity inhibition is still safe in terms of bleeding risk. As demonstrated in the working examples, the bleeding risk of intensive inhibition of FXI/FXIa by h-19F6 and h-42A5 was tested in the same monkeys used in thrombosis experiments. In AV shunt thrombosis animals, no bleeding tendency was observed as the treating dose of h-19F6 or h-42A5 escalated, suggesting that bleeding risk may be independent of the extent of FXI inhibition. In FeCl 3-induced thrombosis animals, neither h-19F6 nor h-42A5 treatment caused excessive bleeding. h-42A5 treatment resulted in an approximately 2-fold elevation of plasma APTT, which indicated more than 99%FXI  inhibition. Previous studies have never evaluated bleeding risk under such intensive APTT-prolongation and high-FXI-inhibition conditions. The antisense oligonucleotide ISIS416856 only caused 30%elevation of APTT when its bleeding risk was evaluated.  26 In other bleeding risk-evaluation studies in primates, a high potent anti-FXI antibody, aXIMab, caused an approximately1-fold increase in APTT (from 30.5 s to 65.6 s) .  26 Thus, the results described herein demonstrate that intensive inhibition of FXI/FXIa does not increase bleeding risk in primates. Thus, FXI can be used as a drug target for thrombosis treatment.
Anti-FXI or Anti-FXIa Antibodies
Provided herein are antibodies that bind to FXI, FXIa, and/or a fragment of FXI or FXIa and inhibit the formation of blood clot. These antibodies are capable of binding to FXI, FXIa, and/or a fragment of FXI or FXIa (e.g., a fragment comprising the A3 domain) and exhibiting an inhibitory effect at a concentration that is much lower than the maximum safety dose. For example, in some embodiments a dose of the antibody between 0.1 mg/kg i.v. and 3 mg/kg i.v. exhibits an inhibitory effect on conversion of FXI to FXIa in cynomolgus monkeys. Moreover, the antibodies disclosed herein can be used as anticoagulation agents with superior safety due to their minimal risk of causing bleeding versus conventional anticoagulation agents such as heparin.
As demonstrated in the working examples, many anti-human FXI antibodies were generated by immunizing rats with human FXI to identify antibodies with anticoagulation properties. A dozen such antibodies were identified, and some of which were humanized for further development. The humanized rat anti-human FXI antibodies, such as h-19F6 and h-42A5 antibodies, were characterized in vitro and in vivo. In the in vitro studies, the humanized antibodies inhibited activated FXI (FXIa) -mediated hydrolysis of factor IX but not factor XIIa-induced FXI activation. The binding properties of the antibodies to FXI were determined, and the dissociation constants (KD) for h-19F6 and h-42A5 were 22 pM and 35 pM, respectively. These two antibodies bind different sites in the A3 domain of FXI. In the in vivo studies, two distinct primate thrombosis models were used to evaluate the anti-thrombotic effects and bleeding risks of the humanized antibodies. In arteriovenous (AV) shunt thrombosis models, both antibodies dose-dependently decreased thrombus formation without causing bleeding. In FeCl3-induced thrombosis models, both antibodies extended the time to thrombosis-mediated  vessel occlusion, and neither antibody increased bleeding. The two antibodies showed anti-thrombotic efficacy without compromising haemostasis in primates, further confirming that targeting FXI can be used for treating thrombosis.
As used herein, the term “comprising” with regard to a composition or method means that the composition or method includes at least the recited elements. The term "consisting essentially of" means that the composition or method includes the recited elements, and may further include one or more additional elements that do not materially affect the novel and basic characteristics of the composition or method. For example, a composition consisting essentially of recited elements may include those recited elements plus one or more trace contaminants from the isolation and purification method, pharmaceutically acceptable carriers such as phosphate buffered saline, preservatives, and the like. The term “consisting of” means the composition or method includes only the recited elements. Embodiments defined by each of the transitional terms are within the scope of this invention.
The term “antibody” as used herein refers to an immunoglobulin molecule or an immunologically active portion thereof that specifically binds to, or is immunologically reactive with a particular antigen, for example, FXI, FXIa, or a particular domain or fragment of FXI or FXIa, e.g., the A3 domain. In certain embodiments an antibody for use in the present methods, compositions, and kits is a full-length immunoglobulin molecule, which comprises two heavy chains and two light chains, with each heavy and light chain containing three complementary determining regions (CDRs) . The term “antibody, ” in addition to natural antibodies, also includes genetically engineered or otherwise modified forms of immunoglobulins, such as synthetic antibodies, intrabodies, chimeric antibodies, fully human antibodies, humanized antibodies, peptibodies and heteroconjugate antibodies (e.g., bispecific antibodies, multispecific antibodies, dual-specific antibodies, anti-idiotypic antibodies, diabodies, triabodies, and tetrabodies) . The antibodies disclosed herein can be monoclonal antibodies or polyclonal antibodies. In those embodiments where an antibody is an immunologically active portion of an immunoglobulin molecule, the antibody may be, for example, a Fab, Fab’, Fv, Fab’F (ab’)  2, disulfide-linked Fv, single chain Fv antibody (scFv) , single domain antibody (dAb) , or diabody. The antibodies disclosed herein, including those that are immunologically active portion of an  immunoglobulin molecule, retain the ability to bind a specific antigen, for example FXI or FXIa, or to bind a specific fragment of FXI or FXIa such as the A3 domain.
In some embodiments, the anti-FXI and/or anti-FXIa antibodies disclosed herein have undergone post-translational modifications such as phosphorylation, methylation, acetylation, ubiquitination, nitrosylation, glycosylation, or lipidation associated with expression in a mammalian cell line, including a human or a non-human host cell. Techniques for producing recombinant antibodies and for in vitro and in vivo modifications of recombinant antibodies are known in the art. See, e.g., Liu et al., mAbs 6 (5) : 1145-1154 (2014) , the content of which is incorporated by reference.
Also disclosed are polynucleotides or nucleic acids encoding the anti-FXI and/or anti-FXIa antibodies disclosed herein. In some embodiments, the polynucleotide or nucleic acid includes DNA, mRNA, cDNA, plasmid DNA. The nucleic acid encoding the antibody or a functional fragment thereof disclosed herein can be cloned into a vector, such as a pTT5 mammalian expression vector, which may further include a promoter and/or other transcriptional or translational control elements such that the nucleic acid can be expressed to produce the antibody or the functional fragment thereof.
The nucleic acid (DNA) and/or amino acid (PRT) sequences, including the sequences of the VH and VL and CDRs, of some examples of the antibodies disclosed herein are listed in Table 1 below.
Table 1: Antibody Sequences
Figure PCTCN2018099638-appb-000001
Figure PCTCN2018099638-appb-000002
Figure PCTCN2018099638-appb-000003
Figure PCTCN2018099638-appb-000004
Figure PCTCN2018099638-appb-000005
Figure PCTCN2018099638-appb-000006
Figure PCTCN2018099638-appb-000007
Figure PCTCN2018099638-appb-000008
Figure PCTCN2018099638-appb-000009
Figure PCTCN2018099638-appb-000010
Figure PCTCN2018099638-appb-000011
Figure PCTCN2018099638-appb-000012
Figure PCTCN2018099638-appb-000013
Figure PCTCN2018099638-appb-000014
Figure PCTCN2018099638-appb-000015
Figure PCTCN2018099638-appb-000016
Provided in certain embodiments herein are humanized anti-FXI and/or anti-FXIa antibodies. Various techniques are known in the art for humanizing antibodies from non-human species such that the antibodies are modified to increase their similarity to antibodies naturally occurring in humans. Six CDRs are present in each antigen binding domain of a natural antibody. These CDRs are short, non-contiguous sequences of amino acids that are specifically positioned to form the antigen binding domain as the antibody assumes its three dimensional configuration. The remainder of the amino acids in the antigen binding domains, referred to as “framework” regions, show less inter-molecular variability and form a scaffold to allow correct positioning of the CDRs. In some embodiments, the antibodies or fragments disclosed herein have conserved sequences for CDR3 regions.
For example, humanization of the antibodies disclosed herein can be accomplished by CDR grafting of monoclonal antibodies produced by immunizing mice or rats. The CDRs of a mouse monoclonal antibody can be grafted into a human framework, which is subsequently joined to a human constant region to obtain a humanized antibody. Briefly, the human germline  antibody sequence database, the protein data bank (PDB) , the INN (International Nonproprietary Names) database, and other suitable databases can be searched and the most similar frameworks to the antibodies can be identified by the search. In addition, some back mutations to the donor residues are made in the human acceptor frameworks. In some embodiments, the variable regions are linked to a human IgG constant region. For example, human IgG1, IgG2, IgG3 and IgG4 Fc domains can be used. It is within the purview of one of ordinary skill in the art to humanize a monoclonal antibody produced by a non-human species based on the existing technology.
The sequences of the variable regions of several example humanized antibodies are shown in Table 2 below.
Table 2: Sequences of humanized antibodies
Figure PCTCN2018099638-appb-000017
The antibodies provided herein include variants of the sequences disclosed herein that contain one or more mutations in their amino acid sequences while retaining binding affinity for FXI, FXIa, and/or a fragment thereof (e.g., a fragment comprising the A3 domain) . In some embodiments, the antibodies include a variable region having an amino acid sequence that is at least 85%, at least 90%, at least 95%, at least 98%, or at least 99%identical to a sequence selected from the group consisting of SEQ ID NOs: 9, 10, 25, 26, 41, 42, 57, 58, 73, 74, 89, 90, 105, 106, 121, 122, 137, 138, 153, 154, 169, 170, 185, 186, and 197-209, or a fragment thereof that retains binding affinity for FXI, FXIa, and/or a fragment thereof.
Also included in this disclosure are variants of nucleic acids encoding antibodies that bind to FXI, FXIa, and/or a fragment thereof (e.g., a fragment comprising the A3 domain) . In some embodiments, the nucleic acids encoding the antibodies include a variable region having a nucleic acid sequence that is at least 85%, at least 90%, at least 95%, at least 98%, or at least 99%identical to a sequence selected from the group consisting of SEQ ID NOs: 1, 2, 17, 18, 33, 34, 49, 50, 65, 66, 81, 82, 97, 98, 113, 114, 129, 130, 145, 146, 161, 162, 177, and 178, or a fragment thereof that encodes a polypeptide with binding affinity for FXI, FXIa, and/or a fragment thereof.
In some embodiments, the antibodies are further subjected to a strategic Chemistry, Manufacturing, and Control (CMC) development such that the novel antibodies such as monoclonal antibodies or humanized monoclonal antibodies disclosed herein are advanced from discovery to human clinical trials, and then to the pharmaceutical market. The modifications further improve the properties of the antibodies without compromising the immunological functions of the antibodies. In certain embodiments, a CMC modified antibody is more stable under various temperatures (e.g., 4℃, 25℃, and 37℃) for an extended period of time (e.g., 3 days, 7 days, 14 days and 28 days) and under repeated freeze/thaw cycles (e.g., -40℃/25℃ for up to 5 cycles) comparing to the unmodified antibody. Additionally, the CMC modified antibodies have an acceptable solubility. For example, for a given sequence of a light chain or a heavy chain, certain amino acids can be potential oxidation and glycosylation sites. These amino acid residues at the potential oxidation, deamidation, or glycosylation sites may be mutated and additional residues in the proximity can also be mutated and/or optimized to maintain the 3D structure and function of a particular antibody. In some embodiments, one or more amino acid residues in a CDR region having the potential of oxidation, deamidation, or glycosylation are  mutated to improve the stability of the antibody or a fragment thereof without compromising the immunological functions. In some embodiments, one or more Met residues in a CDR region having the potential of oxidation are mutated. In some embodiments, one or more Asn residues in a CDR region having the potential of deamidation are mutated.
The sequences of the variable regions of several example CMC optimized, humanized antibodies are shown in Table 3 below.
Table 3: Sequences of CMC optimized, humanized antibodies
Figure PCTCN2018099638-appb-000018
Pharmaceutical Compositions
The antibodies disclosed herein can be formulated into pharmaceutical compositions. The pharmaceutical compositions may further comprise one or more pharmaceutically acceptable carriers, excipients, preservatives, or a combination thereof. The pharmaceutical compositions can have various formulations, e.g., injectable formulations, lyophilized formulations, liquid  formulations, etc. Depending on the formulation and administration route, one would select suitable additives, such as adjuvants, carriers, excipients, preservatives.  34
The pharmaceutical composition can be included in a kit with an instruction for using the composition.
Methods of Treatment
Provided herein is a method of treating and/or preventing thrombosis in a subject suffering from thrombosis and/or at an elevated risk of developing thrombosis. Also provided is a method of inhibiting the formation of blood clots in a subject. These methods entail administering a therapeutically effective amount of an anti-FXI and/or FXIa antibody provided herein to intervene in the intrinsic pathway. In some embodiments, these methods comprise administering a pharmaceutical composition comprising an anti-FXI and/or anti-FXIa antibody as provided herein to the subject.
The methods disclosed herein can be used to prevent and/or treat complications or conditions associated with thrombosis in a subject in need thereof. Thrombosis causes or is associated with a number of complications or conditions, such as embolic stroke, venous thrombosis such as venous thromboembolism (VTE) , deep vein thrombosis (DVT) , and pulmonary embolism (PE) , arterial thrombosis such as acute coronary syndrome (ACS) , coronary artery disease (CAD) , and peripheral artery disease (PAD) . Other conditions associated with thrombosis include, for example, high risk of VTE in surgical patients, immobilized patients, patients with cancer, patients with heart failure, pregnant patients, or patients having other medical conditions that may cause thrombosis. The methods disclosed herein relate to a preventive anticoagulant therapy, that is, thromboprophylaxis. These methods entail administering to a subject suffering from a thrombosis-related complication disclosed above a therapeutically effective amount of an anti-FXI and/or FXIa antibody as disclosed herein or a therapeutically effective amount of a pharmaceutical composition comprising the anti-FXI and/or FXIa antibody. The antibody or pharmaceutical composition can be administered either alone or in combination with any other therapy for treating or preventing the thrombosis-related complications or conditions.
Also provided is a method of treating and/or preventing sepsis in a subject in need thereof. It was attempted to administer anticoagulants to sepsis patients to improve mortality or morbidity. However, the attempt was unsuccessful due to the undesired bleeding caused by anticoagulants. The antibodies disclosed herein can be used as a secondary therapy in combination with other therapeutic agents for treating sepsis, such as antibiotics.
As used herein, the term “subject” refers to mammalian subject, preferably a human. A "subject in need thereof" refers to a subject who has been diagnosed with thrombosis or complications or conditions associated with thrombosis, or is at an elevated risk of developing thrombosis or complications or conditions associated with thrombosis. The phrases “subject” and “patient” are used interchangeably herein.
The terms “treat, ” “treating, ” and “treatment” as used herein with regard to a condition refers to alleviating the condition partially or entirely, preventing the condition, decreasing the likelihood of occurrence or recurrence of the condition, slowing the progression or development of the condition, or eliminating, reducing, or slowing the development of one or more symptoms associated with the condition. With regard to thrombosis and/or complications or conditions associated with thrombosis, "treating" may refer to preventing or slowing the existing blood clot from growing larger, and/or preventing or slowing the formation of blood clot. In some embodiments, the term “treat, ” “treating, ” or “treatment” means that the subject has a reduced number or size of blood clots comparing to a subject without being administered with the antibodies or functional fragments thereof. In some embodiments, the term “treat, ” “treating, ” or “treatment” means that one or more symptoms of thrombosis and/or thrombosis-related conditions or complications are alleviated in a subject receiving an antibody or pharmaceutical composition as disclosed herein comparing to a subject who does not receive such treatment.
A “therapeutically effective amount” of an antibody or pharmaceutical composition as used herein is an amount of the antibody or pharmaceutical composition that produces a desired therapeutic effect in a subject, such as treating and/or preventing thrombosis. In certain embodiments, the therapeutically effective amount is an amount of the antibody or pharmaceutical composition that yields maximum therapeutic effect. In other embodiments, the therapeutically effective amount yields a therapeutic effect that is less than the maximum therapeutic effect. For example, a therapeutically effective amount may be an amount that  produces a therapeutic effect while avoiding one or more side effects associated with a dosage that yields maximum therapeutic effect. A therapeutically effective amount for a particular composition will vary based on a variety of factors, including but not limited to the characteristics of the therapeutic composition (e.g., activity, pharmacokinetics, pharmacodynamics, and bioavailability) , the physiological condition of the subject (e.g., age, body weight, sex, disease type and stage, medical history, general physical condition, responsiveness to a given dosage, and other present medications) , the nature of any pharmaceutically acceptable carriers, excipients, and preservatives in the composition, and the route of administration. One skilled in the clinical and pharmacological arts will be able to determine a therapeutically effective amount through routine experimentation, namely by monitoring a subject’s response to administration of the antibody or the pharmaceutical composition and adjusting the dosage accordingly. For additional guidance, see, e.g., Remington: The Science and Practice of Pharmacy, 22 nd Edition, Pharmaceutical Press, London, 2012, and Goodman &Gilman’s The Pharmacological Basis of Therapeutics, 12 th Edition, McGraw-Hill, New York, NY, 2011, the entire disclosures of which are incorporated by reference herein.
In some embodiments, a therapeutically effective amount of an antibody disclosed herein is in the range from about 0.01 mg/kg to about 30 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, from about 1 mg/kg to about 5 mg/kg.
It is within the purview of one of ordinary skill in the art to select a suitable administration route, such as subcutaneous administration, intravenous administration, intramuscular administration, intradermal administration, intrathecal administration, or intraperitoneal administration. For treating a subject in need thereof, the antibody or pharmaceutical composition can be administered continuously or intermittently, for an immediate release, controlled release or sustained release. Additionally, the antibody or pharmaceutical composition can be administered three times a day, twice a day, or once a day for a period of 3 days, 5 days, 7 days, 10 days, 2 weeks, 3 weeks, or 4 weeks. The antibody or pharmaceutical composition may be administered over a pre-determined time period. Alternatively, the antibody or pharmaceutical composition may be administered until a particular therapeutic benchmark is reached. In certain embodiments, the methods provided herein include a step of evaluating one  or more therapeutic benchmarks to determine whether to continue administration of the antibody or pharmaceutical composition.
Method of Producing Antibodies
Also provided herein are methods of producing the anti-FXI and/or anti-FXIa antibodies disclosed herein. In certain embodiments, these methods entail the steps of cloning a nucleic acid encoding an anti-FXI and/or anti-FXIa antibody into a vector, transforming a host cell with the vector, and culturing the host cell to express the antibody. The expressed antibody can be purified from the host cell using any known technique. Various expression vectors such as pTT5 vector, and pcDNA3 vector, as well as various host cell lines such as CHO cells (e.g. CHO-K1 and ExpiCHO) , and HEK193T cells, can be used.
Also encompassed by this disclosure are antibodies produced by the method disclosed above. The antibodies may have been subjected to one or more post-translational modifications.
The following examples are provided to better illustrate the embodiments and are not to be interpreted as limiting the scope of any claimed embodiment. To the extent that specific materials are mentioned, it is merely for purposes of illustration and is not intended to limit the invention. One skilled in the art may develop equivalent means or reactants without the exercise of inventive capacity and without departing from the scope of the invention. It will be understood that many variations can be made in the procedures herein described while still remaining within the bounds of the present invention. It is the intention of the inventors that such variations are included within the scope of the invention.
Examples
Example 1: Materials and methods
Materials. Human FXI (Cat No. HFXI 1111) , FXIa (Cat No. HFXIa 1111a) , FXIIa (HFXIIa 1212a) , and FIX (Cat No. HFIX 1009) were purchased from Enzyme Research Laboratory (IN, USA) .
Antibody preparation. Animal immunization and hybridoma screening were performed at Genscript Inc. (Nanjing, China) , and the procedures that were applied to  animals in this protocol were approved by the Genscript Institutional Animal Care and Use Committee. The experiment was performed in accordance with the approved guidelines. Wistar rats were immunized with human FXI, and splenocytes from animals with a good immune response were collected for the preparation of hybridomas, which were subjected to subcloning by limiting dilution. Finally, several monoclonal hybridoma clones expressing the desired anti-FXI antibodies, including 19F6, h-34F8 and 42A5, were obtained by using ELISA and functional screening. After determining the amino acid sequences of their variable regions, 19F6, h-34F8 and 42A5 were subjected to humanization, resulting in three humanized antibodies, h-19F6 , h-34F8 and h-42A5, in an IgG4 form. These three humanized antibodies were produced in a transient mammalian expression system and purified by Protein G chromatography.
Activated partial thromboplastin time (APTT) and prothrombin time (PT) . Standard human plasma purchased from Symens Inc. was mixed with equal volume of various antibodies at various concentrations from 0 to 400 nM for 5 minutes before being tested on a CA600 analyzer. In the APTT assay, 50 μL of the plasma-antibody mixture and 25 μL of APTT reagent (SMN 10445709, Symens Inc. ) were mixed at 37 ℃ for 4 min. Then 25 μL of CaCl 2 Solution (25 mM, SMN 10446232, Symens Inc. ) was added and time to clot formation was determined. In the PT assay, 50 μL of the plasma-antibody mixture was mixed with an equal volume of PT reagent (SMN 10446442, Symens Inc. ) at 37 ℃ and time to clot formation was determined.
The effects of the antibodies on APTT and PT in monkey plasma were also evaluated using the same methods applied to human plasma. In these assays, monkey plasma diluted with an equal volume of phosphate-buffered saline (PBS) was used instead of the above mentioned human plasma-antibody mixture.
FXI activation by FXIIa. Human FXI (500 nM) was pre-incubated at room temperature with 1μM control IgG4 or h-19F6 or h-34F8 or h-42A5 in PBS for 5 minutes. At time zero, FXIIa, HK, and kaolin were added so that the final concentrations were FXI (250 nM) , FXIIa (50 nM) , HK (100 nM) , and kaolin (0.5 mg/mL) . At 0, 30, 60, 120 min intervals, 50-μL samples were collected into dodecyl sulfate sample buffer. Samples were size-fractionated on 10%non-reducing gels and transferred to polyvinylidene fluoride  membranes. Western blotting was performed to determine the FXI as well as FXIa-light chain levels using a mouse anti-human FXI IgG (1C5, an in-house made antibody binding the C-terminal of FXI) . The image results were acquired using a ChemiDocMP Imaging System with Image Lab Software (Bio-Rad) .
FXIa-mediated FIX activation. Human FIX (200 nM) was incubated with FXIa (5 nM) in PBS containing 5 mM CaCl 2 at room temperature in the presence of 1 μM control lgG4, h-19F6, h-34F8, or h-42A5. At intervals of 0, 15, 30, 45, and 60 min, 50-μL samples were collected into dodecyl sulfate sample buffer. Samples were size-fractionated on 10%non-reducing gels and transferred to polyvinylidene fluoride membranes. Western blotting was performed to determine the FIX as well as FIXa levels using goat anti-human FIX IgG (Affinity Biologicals) . The image results were acquired using a ChemiDocMP Imaging System with Image Lab Software (Bio-Rad) .
Surface plasmon resonance (SPR) . The interaction of the antibodies with FXI was determined by the SPR assay on a Biacore T200 system (Biacore, GE Healthcare) . Briefly, human IgG capture antibody (Biacore, GE Healthcare) was pre-immobilized on a CM5 sensor chip (GE Healthcare) , and test antibodies were captured by flowing through the chip. The final amount of the test antibodies captured was adjusted to an equal amount of 15 response units (RU) by adjusting the capture time. Then, antigen FXI was allowed to flow through the chip for 180 s for association and then for 1200s for dissociation. FXI was tested at concentrations of 0.063, 0.313, 0.625, 1.25, 3.125 and 6.25 nM. The data were collected and the affinities between the test antibodies and FXI were analyzed using the Biacore Evaluation Software.
To determine the binding sites of test antibodies on FXI, four mutants of FXI tagged with 6×His at the C terminal were first generated by replacing each apple domain (A1, A2, A3, and A4) with the corresponding domains from human prekallikrein. Equal amounts of each mutant were immobilized on a CM5 sensor chip, and test antibodies (33.3nM) were allowed to flow through the chip for 180 s for association and then for 1200 s for dissociation. The amounts of each antibody captured were recorded in response units (RU) using the Biacore Evaluation Software.
Epitope binding results of the test antibodies were also analyzed using the Biacore T200 system. Briefly, wild-type FXI with 6×His tag was pre-immobilized on a CM5 sensor chip (GE Healthcare) , and h-19F6, h-34F8, or h-42A5 (5 μg/ml) was successively injected into flow cells on the sensor surface at a flow rate of 30 μl/minute to monitor the change in response.
Pharmacodynamics in cynomolgus monkeys. This animal experiment and the following AV thrombosis experiment were performed at Wincon Inc. (Nanning, China) , and the procedures applied to animals in this protocol were approved by the Wincon Institutional Animal Care and Use Committee. The experiments were performed in accordance with the approved guidelines. Animals received an intravenous bolus injection of different doses of h-19F6, h-34F8, or h-42A5. Two mL of blood from the superficial veins of upper limb was collected into a collection tube containing 3.2%sodium citrate at pre-dose (time 0) and at 0.5 h, 1 h, 3 h, 6 h, 12 h, and 24 h post-dose. Then, tubes were mixed by gentle inversion ten times at room temperature. Plasma was collected in labelled tubes and stored at -20 ℃until clotting time analysis. Plasma samples were diluted with an equal volume of phosphate buffered saline (pH 7.4) and then subjected to APTT and PT analysis on an automatic analyser (CA660, Sysmex Inc. ) .
AV shunt thrombosis and bleeding time test. A 30-min post-test antibody treatment was administered via intravenous bolus in cynomolgus monkeys. A tail vein bleeding time test was then performed, followed by thrombosis induction. Thrombosis was induced by connecting a shunt device between the femoral arterial and femoral venous cannulas containing a pre-weighed 10-cm-long thread. Blood was allowed to flow through the shunt for 10 min. The thrombus formed on the thread was weighed. Immediately after the removal of the shunt, blood samples were collected, and the next higher level of test antibody was administered. This bleeding/thrombosis process was carried out four times to dose the vehicle and three escalating levels (0.1, 0.3, 1 mg/kg) of test antibody in the same animal.
For bleeding time evaluation, a 2-mL syringe was inserted into the tail vein of the animals. When the volume of blood in the syringe stopped increasing, the elapsed time was recorded manually as the bleeding time.
Ferric chloride (FeCl 3) -induced thrombosis and bleeding time test. This animal experiment was performed at PharmaLegacy Laboratories Inc. (Shanghai, China) , and the procedures that were applied to animals in this protocol were approved by the PharmaLegacy Institutional Animal Care and Use Committee. The experiment was performed in accordance with the approved guidelines. Cynomolgus monkeys were pre-anaesthetized with 1.5 mg/kg of Zoletil, intubated, and ventilated with a respirator. Anaesthesia was maintained with isoflurane. Blood pressure, heart rate, and body temperature were monitored throughout the entire procedure. The vehicle or 0.3 mg/kg of h-19F6, h-34F8, or h-42A5 was administered through a limb vein 2 hours before FeCl 3 application. The left femoral artery was exposed and isolated via blunt dissection. A Doppler flow probe was set up on the artery, and blood flow was continuously recorded. Before applying FeCl 3, blood flow was measured for at least 5 minutes. Then, two pieces of filter paper pre-soaked with 40%FeCl 3 were applied to the adventitial surface of the vessel upstream of the probe for 10 minutes. After the filter paper was removed, the site of application was washed with saline. Blood flow was continuously measured until it decreased to 0. The time to 80%occlusion (blood flow reduced to 20%of the baseline blood flow) and the time to 100%occlusion (blood flow reduced to 0) were recorded. In the same animals, haemostasis was evaluated using the Surgicutt device and bleeding time was manually recorded at pre-dose and 1-hour post-dose. At the end of the study (approximately 3 hours post-dose) , blood samples were collected.
The binding specificity of test antibodies to human FXI in plasma. Test antibodies (h-19F6, h-42A5, and 14E11) were first biotinylated using EZ-Link TM Sulfo-NHS-LC-Biotinylation Kit (Cat No. 21435, Thermo Fisher Inc. ) . These antibodies (25 μg each) were incubated with 200 μL of human standard plasma (Siemens Inc. ) or FXI-deficient plasma (Hyphen Biomed Inc. ) for 1h. Then 50 μL of Streptavidin-coated beads (Dynabeads TM M-280 Streptavidin, Thermo Fisher Inc. ) were added to the mixture to extract the biotin-containing antigen-antibody complex. After washing with PBS for 3 times, the antigen-antibody complex was then eluted and subjected to Western blotting using a mouse anti-human FXI IgG (1C5, an in-house made antibody binding the C-terminal of FXI) as the primary antibody. The image results were acquired using a ChemiDocMP Imaging System  with Image Lab Software (Bio-Rad) . In Western blotting, 10 μL of human standard plasma and FXI-deficient plasma served as FXI-positive and FXI-negative controls, respectively.
Statistical analysis. Numerical data from multiple experiments are presented as means ± standard error of mean (SEM) . One-way ANOVA followed by Dunnett’s multiple comparisons test was used for the statistical analysis of thrombus weight in the AV shunt experiment and both bleeding time tests. The Kruskal-Wallis rank test was performed for the statistical analysis of occlusion times in the FeCl 3-induced thrombosis experiment. A value of P﹤0.05 was considered statistically significant.
Example 2: Generation and sequencing of anti-FXI antibodies
BALB/c mice and Wistar rats were immunized with human FXI, and splenocytes from the animals with good immune response were collected for the preparation of hybridomas, which were subjected to subcloning by limiting dilution. Twelve monoclonal hybridoma clones expressing desired anti-FXI antibodies 3G12, 5B2, 7C9, 7F1, 13F4, 19F6, 21F12, 34F8, 38E4, 42A5, 42F4, and 45H1 were obtained by using capture ELISA and functional screening.
To determine the amino acid and nucleotide sequences of the variable region of the light (V L) and heavy chain (V H) of these antibodies, cDNAs encoding V L and V H were cloned from the corresponding hybridoma cells by standard RT-PCR procedures. The V L and V H sequences of exemplary antibodies, including the sequences of CDRs, are shown in Table 1.
Example 3: Determination of anti-coagulation activity in human plasma using activated partial thromboplastin time (APTT) assay and prothrombin time (PT) assay
APTT assay measures the activity of the intrinsic and common pathways of coagulation; whereas PT assay measures the activity of the extrinsic and common pathways of coagulation. The antibodies tested in these experiments were 19F6, 34F8, 42A5, 1A6 and 14E11. Antibodies 1A6 and 14E11 were used as positive controls in this experiment. The sequences of the variable regions of the control antibodies were obtained from U.S. Patent No. 8,388,959 and U.S. Patent Application Publication No. 2013/0171144 and reformatted to IgG4. These antibodies were then expressed using ExpiCHO cell system. The APTT assay and PT assay were performed as described above.
As shown in Figure 1, all antibodies tested increased APTT in a concentration-dependent manner at a relatively low concentration, for example, up to 100 nM (or up to 200 nM for 14E11) ; whereas none of these antibodies had a significant effect on PT (data not shown) . These results indicate that all of the antibodies tested inhibited the intrinsic pathway of coagulation but not the extrinsic pathway.
Example 4: Determination of the anti-coagulation activity in the plasma of non-human species using activated partial thromboplastin time (APTT) assay
Effects of various antibodies, including 19F6, 34F8 and 42A5, on coagulation were assessed in the mouse, rat, and monkey plasma using the same method as described in Example 3. None of the antibodies tested had any effect on APTT in the mouse and rat plasma, but all of them, at a relatively low concentration, concentration-dependently, increased APTT in the monkey plasma as shown in Figure 2, indicating that the antibodies tested had cross-activity with monkey FXI/FXIa, but not with mouse or rat FXI/FXIa.
Example 5: Humanization of anti-FXI antibodies
The use of murine monoclonal antibodies directly as therapeutics has been hindered by the short half-life and the elicitation of the human anti-murine antibody responses. One solution to this problem is to humanize the murine antibodies. Some antibodies were subjected to humanization by CDR grafting. Suitable human acceptor frameworks for both V L and V H of each murine antibody were identified and varying numbers of back mutations were introduced to the selected human frameworks to maintain the structure and/or function of the resulting antibody. If the affinity and function of these humanized antibodies were not substantially inferior to the corresponding unmodified antibodies, the modified antibodies were considered successfully humanized. Three humanized V H and V L sequences of 19F6, 34F8 and 42A5, described as h-19F6, h-34F8, and h-42A5, respectively, are shown in Table 2.
Example 6: Determination of the affinity of anti-FXI antibodies to human FXI
The affinity of anti-FXI/FXIa antibodies to FXI/FXIa were determined using surface plasmon resonance (SPR) technology performed on the BIAcore T200 instrument. The humanized antibodies were constructed by linking the variable regions of the antibodies disclosed  herein to human IgG4 Fc domain and the recombinants were expressed in CHO cells. These antibodies were captured onto a Biacore CM5 sensor chip that was pre-immobilized with an anti-human IgG antibody.
Then different concentrations of the purified antigen FXI or FXIa (0.005-1 μg/mL) were allowed to flow through the CM5 chip for 180 s for association with the anti-FXI/FXIa antibody, followed by a time of 1800 s for dissociation. The binding data was collected and the affinity between FXI/FXIa and the test antibodies was analyzed using the Biacore Evaluation Software provided by GE Healthcare. The SPR sensorgrams of FXI/FXIa binding to immobilized h-19F6, h-34F8, and h-42A5 are shown in Figure 3. As shown in Figure 3, the response (RU) for each antibody became higher with escalating concentrations of FXI or FXIa. The dissociation constants (K D) of h-19F6, h-34F8, and h-42A5 to FXI and FXIa were calculated and detailed in Table 4. The affinities of each antibody to FXI and FXIa are considered to be the same since the difference between them is less than 10 times.
Table 4: K D values of the antibodies to FXI and FXIa
Figure PCTCN2018099638-appb-000019
Example 7: Determination of the binding site of anti-FXI antibodies on FXI
The binding sites of 19F6 and 42A5 on FXI were determined using the SPR technology. Briefly, human IgG capture antibody was pre-immobilized on a Biacore CM5 sensor chip, and recombinant h-19F6 or h-42A5 was captured by flowing through the chip. An equal amount (15 relative units) of h-19F6 and h-42A5 was captured through adjustment of the antibody flowing time. Then wild type FXI or chimeric FXI in which individual apple domain was replaced with the corresponding domain from the human prekallikrein (FXI/PK chimeras) was allowed to flow through the chip for 180 seconds for association with h-19F6 or h-42A5, followed by a time period of 1800 seconds for dissociation. The binding data was analyzed in a high performance kinetic mode as only one concentration of FXI, wild-type or chimeric, was  tested in the SPR assay. Results showed that both h-19F6 and h-42A5 bound FXI as well as FXI/PK chimeras except when the A3 domain of FXI was replaced with the corresponding PK domain, indicating that part or the complete epitope of h-19F6 and h-42A5 on FXI is located in the A3 domain.
Example 8: Functional neutralization of FXIa by the antibodies
Human FXIa activity was determined by measuring the cleavage of a specific, chromogenic substrate, S-2366 (Diapharma Inc. ) . For testing the inhibitory activity of the antibodies, antibodies h-19F6, h-34F8 and h-42A5 were pre-incubated for 5 minutes at room temperature with a final concentration of 5 nM of FXIa in PBS (phosphate buffer saline) . Then an equal volume of 1 mM of S-2366 was added to initiate the FXIa cleavage reaction and changes in absorbance at 405 nm was monitored continuously using a M5 e plate reader (Molecular Devices Inc. ) . Data were analyzed using the GraphPad Prism software and are shown in Figure 4. The calculated apparent Ki for h-19F6, h-34F8, and h-42A5 are 0.67, 2.08, and 1.43 nM, respectively. Therefore, all three antibodies tested exhibited satisfying inhibitory effects on FXIa at a relatively low concentration.
Example 9: Inhibition of FXIa-mediated FIX activation by the antibodies
The FXIa-mediated FIX activation was performed as described above. Anti-FXI antibodies may modulate the intrinsic pathway by inhibiting FXI activation and/or by inhibiting FXIa activity. First the effects of the two antibodies h-19F6 and h-42A5 on FXIIa-mediated activation of FXI were tested and it was found that neither h-19F6 nor h-42A5 prevented the conversion of FXI to FXIa mediated by FXIIa (Figures 5C and 5D) . Then, the effect of these two antibodies on FXIa activity was tested using FIX as the substrate. As shown in Figures 5A and 5B, both h-19F6 and h-42A5 reduced FIX activation in a concentration-dependent manner. The inhibitory effect of these two antibodies on FXIa activity was further confirmed by using a chromogenic substrate of FXIa, S-2366. Both antibodies concentration-dependently inhibited the hydrolysis of S-2366 (Figure 4) .
Example 10: Evaluation of the effects of anti-FXI antibodies on clotting time in cynomolgus monkeys
To find proper animal species for in vivo experiments, the cross-reactivity of the antibodies for mouse, rat, and monkey FXI was tested by the APTT assay. The antibodies prolonged APTT in monkey plasma but not in mouse or rat plasma (data not shown) . Thus, monkey models were chosen for evaluating the pharmacodynamic effects of the three antibodies on clotting times prior to efficacy studies on thrombosis in vivo. Cynomolgus monkeys were intravenously administered with indicated doses of various antibodies. Blood from the superficial veins of the upper limb was collected at pre-dose and at 0.5, 1, 3, 6, 12, and 24 hours post-dose, and citrated plasma was prepared for APTT and PT determination. In the APTT test, 50 μL of diluted plasma sample and 25 μL of APTT reagent (SMN 10445709, Symens Inc. ) were mixed and incubated at 37 ℃ for 4 min. Then 25 μL of CaCl 2 Solution (25 mM, SMN 10446232, Symens Inc. ) was added and time to clot formation was determined. In the PT test, 50 μL of diluted plasma sample was mixed with equal volume of PT reagent (SMN 10446442, Symens Inc. ) and incubated at 37℃ and time to clot formation was determined. All three antibodies tested demonstrated dose-dependently increased APTT as shown in Figure 6 and none of them affected PT as shown in Figure 7.
Both h-19F6 and h-42A5 dose-dependently prolonged APTT (Figures 6B and 6C) . Notably, h-42A5 increased APTT more strongly than h-19F6 did at the same dose levels (0.3 and 1 mg/kg) , consistent with the antibodies’in vitro effects on human APTT (Figure 16A) . In addition, neither antibody affected the PT in vivo (Figures 7B and 7C) .
Example 11: Evaluation of the effects of anti-FXI antibodies in arteriovenous (AV) shunt thrombosis and tail vein bleeding models in cynomolgus monkeys
Both thrombosis and bleeding time were assessed in the same animal for multiple doses of each antibody tested. The antibodies included in this experiment were h-34F8, h-19F6, and h42A5. Briefly, bleeding time and thrombosis were sequentially evaluated at pre-dose and 30 minutes following each administration of the antibody. The bleeding/thrombosis assessments were conducted four times: pre-dose and post-dose at three escalating dose levels (0.1, 0.3 and 1 mg/kg) .
For AV shunt thrombosis, a shunt device containing a pre-weighed 10-cm long silk thread was applied to connecting the femoral arterial and femoral venous cannulae, and blood was allowed to flow through the shunt for 10 minutes. Then the thread was removed from the shunt and weighed again. Clot weight on the thread was calculated as the difference of the thread weight before and after blood flow.
For bleeding time evaluation, a 2-mL syringe was inserted into the tail vein of the animals. When the volume of blood in the syringe stopped increasing, the elapsed time was recorded manually as the bleeding time.
All antibodies dose-dependently reduced the thrombus weight as shown in Figure 8 and none of them prolonged the tail vein bleeding time as shown in Figure 9. Effects of h-19F6 and h-42A5 on thrombosis and haemostasis were evaluated in monkey models of AV shunt thrombosis and tail vein bleeding. Intravenous injection of h-19F6 resulted in a dose-dependent reduction of clot weight, and a significant reduction was observed at 1 mg/kg dose (Figure 8B) . Regarding h-42A5-treated animals, clot weight was significantly reduced at all test dose levels in a dose-dependent manner (Figure 8C) . No significant change in bleeding time was noted following treatment with h-19F6 or h-42A5 (Figures 9B and 9C) .
Example 12: Evaluation of the effects of anti-FXI antibodies on ferric chloride–induced artery thrombosis and template bleeding time in cynomolgus monkeys
Cynomolgus monkeys were pre-anesthetized with 1.5 mg/kg of Zoletil, intubated, and ventilated with a respirator. Anesthesia was maintained with isoflurane. The blood pressure, heart rate, and body temperature were monitored throughout the entire procedure. The antibodies tested, including h-34F8, h-19F6, and h-42A5, or the vehicle control were administered through limb vein by injection 2 hours before FeCl 3 application. The left femoral artery was exposed and isolated via blunt dissection. A Doppler flow probe was set up on the artery and the blood flow was continuously recorded. Before applying FeCl 3, the blood flow was measured for at least 5 minutes. Then two pieces of filter paper pre-soaked with FeCl 3 were applied to the adventitial surface of the vessel upstream from the probe for 10 minutes. After the filter paper was removed, the site of application was washed with saline. Blood flow was continuously measured until it decreased to 0. The time to 80%occlusion (blood flow reduced to 20%of the baseline blood  flow) and the time to 100%occlusion (blood flow reduced to 0) were recorded. In the same animal, template bleeding time was assessed at pre-dose and 1 hour post-dose.
The effects of all three antibodies on FeCl 3-induced arterial thrombosis were investigated. Four groups of monkeys were treated with the vehicle control, h-34F8, h-19F6, or h-42A5 for 2 hours, respectively, and FeCl 3 was applied on the left femoral artery of each animal to induce thrombosis. Downstream blood flow velocity was monitored. The time to 80%and to 100%thrombotic occlusion in the vehicle control group was 14.66 ± 1.30 min and 18.50 ± 1.76 min, respectively. Pretreatment with 0.3 mg/kg of h-34F8 or h-42A5 significantly delayed the time to 80%occlusion to 59.53 ± 16.95 min and 40.80 ±7.94 min, and the time to 100%occlusion to 70.40 ± 20.76 min and 50.61 ±9.48 min, respectively, as shown in Figure 10. Prolongation of the time to 80%occlusion (26.43 ± 5.72 min) and to 100%occlusion (32.78 ±5.09 min) was also observed in monkeys treated with h-19F6, although there was no statistically significant difference when compared to the vehicle control group as shown in Figure 10.
The effects of the antibodies on haemostasis was assessed in terms of template bleeding time. No significant difference was noted between pre-dose and 1 hour post-dose for each test article (Figure 11A, 11B, and 11C) . The bleeding time change following h-34F8, h-19F6, and h-42A5 treatment was not different from that following the vehicle control treatment (Figure 11D) .
The effects of h-19F6 and h-42A5 antibodies on haemostasis were assessed by a skin laceration-caused bleeding test in the same animals with FeCl 3-induced arterial thrombosis (n=5 per group) . The bleeding times were recorded at pre-dose and 1-hour post-dose. No significant difference in bleeding time was observed between the pre-dose and 1-hour post-dose for either antibody or among the three groups 1-hour post-dose (Figure 18) .
The effects of the antibodies on ex vivo clotting times of monkey plasma were also evaluated. As expected, treatments with 0.3 mg/kg of h-34F8, h-19F6, and h-42A5 significantly prolonged APTT by 3.29 ± 0.20, 1.67 ± 0.09, and 2.87 ± 0.10 fold, respectively, while no increase in APTT was observed upon vehicle control treatment, as shown in Figure 12A. In addition, treatments with h-34F8, h-19F6, or h-42A5 had no effect on PT as shown in Figure 12B.
Thus, it was unexpectedly discovered that the antibodies disclosed herein did not have any adverse effects of prolonged bleeding while effectively inhibiting the intrinsic pathway of coagulation.
Example 13: Evaluation of the effects of modified anti-FXI antibodies on clotting time in cynomolgus monkeys for an extended period of time
Two additional CMC optimized, humanized anti-FXI antibodies, shown in Figures 14 and 15 as “modified h-19F6” and “modified h-42A5, ” were evaluated for their effects on clotting time in cynomolgus monkeys for an extended period of time, e.g., for up to 14 days, by APTT and PT assays as described in Example 10. The heavy chain and light chain sequences of these two antibodies are shown in Table 3. Cynomolgus monkeys were intravenously administered with 0.6 mg/kg or 2.0 mg/kg of the tested antibodies. Blood from the superficial veins of the upper limb was collected at pre-dose and at 0.5 hour, 2 hours, 6 hours, 12 hours, 24 hours, 48 hours, 96 hours, 168 hours, 240 hours, 336 hours post-dose. Both modified antibodies tested demonstrated dose-dependently increased APTT as shown in Figure 14 and none of them affected PT as shown in Figure 15. Both modified antibodies demonstrated efficacy for an extended period, up to 7 days, up to 10 days, or up to 14 days.
Thus, it was unexpectedly discovered that the modified antibodies disclosed herein did not show any adverse effects of prolonged bleeding while effectively inhibiting the intrinsic pathway of coagulation for an extended period of time, up to 14 days.
Example 14: Effects on clotting times of standard human plasma
Antibodies h-19F6 and h-42A5 were added to normal human plasma, after which the APTT (Figure 16A) and PT (Figure 16B) were determined (N = 3) . Both h-19F6 and h-42A5 antibodies prolonged the activated partial thromboplastin time (APTT) of standard human plasma in a concentration-dependent manner (Figure 16A) . The maximum levels of inhibition of FXI activity in the plasma for h-19F6 and h-42A5 were approximately 97%and 99.5%, respectively, based on the correlation curve between plasma FXI level and APTT established (data not shown) . Neither antibody affected the PT of human plasma (Figure 16B) .
Example 15: Binding properties of h-19F6 and h-42A5 to FXI
The binding specificity of h-19F6 and h-42A5 to FXI were first verified as they reacted with FXI in standard human plasma and no reaction was detected in human FXI-deficient plasma (Figure 17) . Biotinylated test antibodies were incubated with human normal plasma or FXI-deficient plasma. The FXI-antibody complex in the plasma was eluted and subject to Western blotting using a mouse anti-human FXI IgG as the primary antibody. In Western blotting, 10 μL of human standard plasma or FXI-deficient plasma were served as FXI-positive and FXI-negative controls. A previously reported anti-FXI antibody, 14E11 17, showed the same binding profile as the two antibodies did (Figure 17) .
The affinities of h-19F6 and h-42A5 to FXI were determined using surface plasmon resonance (SPR) technology. The test antibodies were captured on a sensor chip, and then indicated concentrations of FXI were allowed to flow through the chip. Sensorgrams for h-19F6 (Figure 19A) and h-42A5 (Figure 19B) were obtained. The dissociation constants for h-19F6 and h-42A5 were 22 and 36 pM, respectively (Figures 19A and 19B) .
The binding sites of these two antibodies on FXI were then determined. FXI is a homodimer consisting of 4 tandem apple domains (A1-4) and a catalytic domain. Four mutants of FXI were generated by replacing each apple domain with corresponding domains from human prekallikrein and tested the binding properties of h-19F6 or h-42A5 to the 4 mutants of FXI using SPR. Equal amounts of the 4 mutant FXIs in which the A1, A2, A3, or A4 domain was replaced with the corresponding domain from prekallikrein were immobilized on a sensor chip, and test antibodies (5 μg/mL) were allowed to flow through the chip for association. The amounts of each antibody captured were recorded. The experiment was performed twice, and a representative result is depicted. Unexpectedly, both antibodies predominantly bound to the A3 domain of FXI, as replacement of the A3 domain resulted in much less binding of either antibody compared with the replacement of the other 3 apple domains (Figure 19C) . Another antibody, O1A6, a reported anti-FXI antibody used as a positive control, also specifically bound to the A3 domain of FXI, consistent with a previous study.  21 However, it was hypothesized that h-19F6 and h-42A5 bind different sites of FXI because they have comparable affinities to FXI but quite different inhibitory potencies with respect to FXI activity, as indicated in Figure 16. This hypothesis was tested  by epitope binding using a Biacore T200 system. Indeed, the binding of h-19F6 to FXI did not prevent the following binding of h-42A5 to FXI, indicating that the two antibodies bind different sites in the A3 domain of FXI (Figure 19D) . Then the flowing order of these two antibodies were changed and it was found that binding of h-42A5 to FXI did not prevent the following binding of h-19F6 to FXI (data not shown) .
Example 16: Binding properties of h-19F6 and h-42A5 to FXIa
The antibodies bound to FXIa with the good affinities with which they bound to FXI (Figures 20A and 20B) . The affinities of h-19F6 and h-42A5 to FXI were determined using surface plasmon resonance (SPR) technology. The dissociation constants for h-19F6 and h-42A5 were 26 and 81 pM, respectively (Figures 20A and 20B) . The test antibodies were captured on a sensor chip, and then indicated concentrations of FXIa were allowed to flow through the chip. Sensorgrams for h-19F6 (Figure 20A) and h-42A5 (Figure 20B) were obtained.
References
The references, patents and published patent applications listed below, and all references cited in the specification above are hereby incorporated by reference in their entirety, as if fully set forth herein.
1 Raskob, G.E. et al. Thrombosis: a major contributor to global disease burden. Arterioscler Thromb VascBiol 34, 2363-2371 (2014) .
2 Gomez-Outes, A., Garcia-Fuentes, M. &Suarez-Gea, M.L. Discovery methods of coagulation-inhibiting drugs. Expert Opin Drug Discov 12, 1195-1205 (2017) .
3 Weitz, J. I. &Fredenburgh, J.C. Factors XI and XII as Targets for New Anticoagulants. Front Med (Lausanne) 4, 19 (2017) .
4 Muller, F., Gailani, D. &Renne, T. Factor XI and XII as antithrombotic targets. Curr Opin Hematol 18, 349-355 (2011) .
5 Al-Horani, R.A. &Desai, U.R. Factor XIa inhibitors: A review of the patent literature. Expert Opin Ther Pat 26, 323-345 (2016) .
6 Chen, Z., Seiffert, D. &Hawes, B. Inhibition of Factor XI activity as a promising antithrombotic strategy. Drug Discov Today 19, 1435-1439 (2014) .
7 Gailani, D. &Gruber, A. Factor XI as a Therapeutic Target. Arterioscler Thromb Vasc Biol 36, 1316-1322 (2016) .
8 Puy, C., Rigg, R.A. &McCarty, O.J. The hemostatic role of factor XI. Thromb Res 141 Suppl 2, S8-S11 (2016) .
9 Seligsohn, U. Factor XI deficiency in humans. J Thromb Haemost 7 Suppl 1, 84-87 (2009) .
10 Preis, M. et al. Factor XI deficiency is associated with lower risk for cardiovascular and venous thromboembolism events. Blood 129, 1210-1215 (2017) .
11 Meijers J.C., Tekelenburg W.L., Bouma B.N., Bertina R.M., Rosendaal F.R. High levels of coagulation factor XI as a risk factor for venous thrombosis. N Engl J Med 342, 696–701 (2000) .
12 Peyvandi, F. et al. Coagulation factor activity and clinical bleeding severity in rare bleeding disorders: results from the European Network of Rare Bleeding Disorders. J Thromb Haemost 10, 615-621 (2012) .
13 Salomon, O. &Seligsohn, U. New observations on factor XI deficiency. Haemophilia 10 Suppl 4, 184-187 (2004) .
14 Wang, X. et al. Effects of factor IX or factor XI deficiency on ferric chloride-induced carotid artery occlusion in mice. J Thromb Haemost 3, 695-702 (2005) .
15 Wang, X. et al. Inhibition of Factor XIa Reduces the Frequency of Cerebral Microembolic Signals Derived from Carotid Arterial Thrombosis in Rabbits. J Pharmacol Exp Ther 360, 476-483 (2017) .
16 Wong, P.C., Crain, E.J., Watson, C.A. &Schumacher, W.A.A small-molecule factor XIa inhibitor produces antithrombotic efficacy with minimal bleeding time prolongation in rabbits. J Thromb Thrombolysis32, 129-137 (2011) .
17 Cheng, Q. et al. A role for factor XIIa-mediated factor XI activation in thrombus formation in vivo. Blood 116, 3981-3989 (2010) .
18 Takahashi, M. et al. Inhibition of factor XI reduces thrombus formation in rabbit jugular vein under endothelial denudation and/or blood stasis. Thromb Res 125, 464-470 (2010) .
19 Yau, J.W. et al. Selective depletion of factor XI or factor XII with antisense oligonucleotides attenuates catheter thrombosis in rabbits. Blood 123, 2102-2107 (2014) .
20 Crosby, J.R. et al. Antithrombotic effect of antisense factor XI oligonucleotide treatment in primates. Arterioscler Thromb Vasc Biol 33, 1670-1678 (2013) .
21 Kravtsov D.V. et al. Factor XI contributes to thrombin generation in the absence of factor XII. Blood 114, 452-458 (2009) .
22 Wang, X. et al. Effects of factor XI deficiency on ferric chloride-induced vena cava thrombosis in mice. J Thromb Haemost 4, 1982-1988 (2006) .
23 Kleinschnitz, C. et al. Targeting coagulation factor XII provides protection from pathological thrombosis in cerebral ischemia without interfering with hemostasis. J Exp Med 203, 513-518 (2006) .
24 Gailani, D., Lasky, N.M. &Broze, G.J., Jr. A murine model of factor XI deficiency. Blood Coagul Fibrinolysis 8, 134-144 (1997) .
25 Beck, A., Wurch, T., Bailly, C. &Corvaia, N. Strategies and challenges for the next generation of therapeutic antibodies. Nat Rev Immunol 10, 345-352 (2010) .
26 Tucker, E.I. et al. Prevention of vascular graft occlusion and thrombus-associated thrombin generation by inhibition of factor XI. Blood 113, 936-944 (2009) .
27 Younis, H.S. et al. Antisense inhibition of coagulation factor XI prolongs APTT without increased bleeding risk in cynomolgus monkeys. Blood 119, 2401-2408 (2012) .
28 Kouyama S, O.T., Hagio T, et al. Discovery of ONO-5450598, a highly orally bioavailable small molecule factor XIa inhibitor: the pharmacokinetic and pharmacological profiles. Res Pract Thromb Haemost 1 Suppl 1: PB 2139 (2017) .
29 Wong, P.C. et al. In vitro, antithrombotic and bleeding time studies of BMS-654457, a small-molecule, reversible and direct inhibitor of factor XIa. J Thromb Thrombolysis 40, 416-423 (2015) .
30 David, T. et al. Factor XIa-specific IgG and a reversal agent to probe factor XI function in thrombosis and hemostasis. Sci Transl Med 8, 353ra112 (2016) .
31 van Montfoort, M.L. et al. Two novel inhibitory anti-human factor XI antibodies prevent cessation of blood flow in a murine venous thrombosis model. Thromb Haemost 110, 1065-1073 (2013) .
32 Buller, H.R. et al. Factor XI antisense oligonucleotide for prevention of venous thrombosis. N Engl J Med 372, 232-240 (2015) .
33. Emsley et al., Blood 115 (13) : 2569-2577 (2010) .
34. Wang et al., J. Pharm. Sciences 96 (1) : 1-26 (2007) .

Claims (17)

  1. An isolated anti-FXI or anti-FXIa antibody that specifically binds to human FXI or FXIa, wherein the antibody comprises an immunoglobulin light chain variable domain comprising three CDRs selected from the group consisting of SEQ ID NOs: 11-13, 27-29, 43-45, 59-61, 75-77, 91-93, 107-109, 123-125, 139-141, 155-157, 171-173, 187-189, 197, 199, 201, 204, 206, and 208, and sequences sharing at least 90%identity, or an immunoglobulin heavy chain variable domain comprising three CDRs selected from the group consisting of SEQ ID NOs: 14-16, 30-32, 46-48, 62-64, 78-80, 94-96, 110-112, 126-128, 142-144, 158-160, 174-176, 190-192, 198, 200, 202, 205, 207, and 209, and sequences sharing at least 90%identity, or an immunologically active portion thereof.
  2. The antibody of claim 1, wherein the antibody specifically binds to the A3 domain of the human FXI or FXIa.
  3. The antibody of claim 1, wherein the antibody comprises an immunoglobulin light chain variable domain selected from the group consisting of SEQ ID NO: 9, SEQ ID NO: 25, SEQ ID NO: 41, SEQ ID NO: 57, SEQ ID NO: 73, SEQ ID NO: 89, SEQ ID NO: 105, SEQ ID NO: 121, SEQ ID NO: 137, SEQ ID NO: 153, SEQ ID NO: 169, SEQ ID NO: 185, SEQ ID NO; 197, SEQ ID NO: 199, SEQ ID NO: 201, SEQ ID NO: 204, SEQ ID NO: 206, and SEQ ID NO: 208, and sequences sharing at least 90%identity.
  4. The antibody of claim 1, wherein the antibody comprises an immunoglobulin heavy chain variable domain selected from the group consisting of SEQ ID NO: 10, SEQ ID NO: 26, SEQ ID NO: 42, SEQ ID NO: 58, SEQ ID NO: 74, SEQ ID NO: 90, SEQ ID NO: 106, SEQ ID NO: 122, SEQ ID NO: 138, SEQ ID NO: 154, SEQ ID NO: 170, SEQ ID NO: 186, SEQ ID NO: 198, SEQ ID NO: 200, SEQ ID NO; 202, SEQ ID NO: 205, SEQ ID NO: 207, and SEQ ID NO: 209, and sequences sharing at least 90%identity.
  5. A pharmaceutical composition comprising the antibody of any one of claims 1-4.
  6. A method of inhibiting the formation of blood clots in a subject comprising administering to the subject a therapeutically effective amount of the antibody of any one of claims 1-4.
  7. A method of inhibiting the formation of blood clots in a subject comprising administering to the subject a therapeutically effective amount of the pharmaceutical composition of claim 5.
  8. A method of treating or preventing thrombosis or a complication or condition associated with thrombosis comprising administering to a subject a therapeutically effective amount of the antibody of any one of claims 1-4, wherein the administration does not compromise hemostasis of the subject.
  9. A method of treating or preventing thrombosis or a complication or condition associated with thrombosis comprising administering to a subject a therapeutically effective amount of the pharmaceutical composition of claim 5, wherein the administration does not compromise hemostasis of the subject.
  10. A method of treating or preventing sepsis comprising administering to a subject a therapeutically effective amount of the antibody of any one of claims 1-4, wherein the administration does not compromise hemostasis of the subject.
  11. A method of treating or preventing sepsis comprising administering to a subject a therapeutically effective amount of the pharmaceutical composition of claim 5, wherein the administration does not compromise hemostasis of the subject.
  12. A method of producing the antibody of any one of claims 1-4, comprising expressing a nucleic acid encoding the antibody cloned in an expression vector in a host cell.
  13. The method of claim 12, further comprising purifying the expressed antibody from the host cell.
  14. The method of claim 12, wherein the expression vector is a pTT5 vector or pcDNA3 vector.
  15. The method of claim 12, wherein the host cell is a CHO cell or an HEK193T cell.
  16. An antibody or a functional fragment thereof produced by the method of any one of claims 12-15, or an immunologically active portion thereof.
  17. The antibody of claim 16, wherein the antibody has been post-translationally modified.
PCT/CN2018/099638 2017-02-10 2018-08-09 Anti-factor xi antibodies WO2020029179A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
BR112021002472-7A BR112021002472A2 (en) 2018-08-09 2018-08-09 anti-factor xi antibodies
AU2018436195A AU2018436195A1 (en) 2018-08-09 2018-08-09 Anti-factor XI antibodies
CN202310301532.8A CN116554334A (en) 2018-08-09 2018-08-09 Anticoagulant factor XI antibodies
MX2021001613A MX2021001613A (en) 2018-08-09 2018-08-09 Anti-factor xi antibodies.
CA3108708A CA3108708A1 (en) 2018-08-09 2018-08-09 Anti-factor xi antibodies
CN202111466510.4A CN114478781B (en) 2018-08-09 2018-08-09 Anticoagulant factor XI antibodies
CN201880098606.XA CN113227150B (en) 2018-08-09 2018-08-09 Anticoagulant factor XI antibodies
EP18929685.8A EP3833692A1 (en) 2018-08-09 2018-08-09 Anti-factor xi antibodies
KR1020217006922A KR20210042352A (en) 2018-08-09 2018-08-09 Anti-factor XI antibody
JP2021506623A JP2021534098A (en) 2018-08-09 2018-08-09 Anti-factor XI antibody
PCT/CN2018/099638 WO2020029179A1 (en) 2018-08-09 2018-08-09 Anti-factor xi antibodies
CN202111467365.1A CN114478782B (en) 2018-08-09 2018-08-09 Anticoagulant factor XI antibodies
US16/537,427 US11958911B2 (en) 2017-02-10 2019-08-09 Anti-coagulation factor XI antibody

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2018/099638 WO2020029179A1 (en) 2018-08-09 2018-08-09 Anti-factor xi antibodies

Related Child Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/119856 Continuation WO2018145533A1 (en) 2017-02-10 2017-12-29 Anti-coagulation factor xi antibody

Publications (1)

Publication Number Publication Date
WO2020029179A1 true WO2020029179A1 (en) 2020-02-13

Family

ID=69413757

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/099638 WO2020029179A1 (en) 2017-02-10 2018-08-09 Anti-factor xi antibodies

Country Status (9)

Country Link
EP (1) EP3833692A1 (en)
JP (1) JP2021534098A (en)
KR (1) KR20210042352A (en)
CN (4) CN114478782B (en)
AU (1) AU2018436195A1 (en)
BR (1) BR112021002472A2 (en)
CA (1) CA3108708A1 (en)
MX (1) MX2021001613A (en)
WO (1) WO2020029179A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3914624A4 (en) * 2019-01-21 2022-10-19 Aronora, Inc. Novel humanized antibodies against factor xi having anti-thrombotic and anti-inflammatory effects and uses thereof

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11958911B2 (en) 2017-02-10 2024-04-16 Shanghai Benemae Pharmaceutical Anti-coagulation factor XI antibody
JP2021534098A (en) * 2018-08-09 2021-12-09 上海仁会生物制▲やく▼股▲ふん▼有限公司Shanghai Benemae Pharmaceutical Corporation Anti-factor XI antibody
US20230343982A1 (en) 2021-03-31 2023-10-26 Lg Energy Solution, Ltd. Mono-Cell Manufacturing Apparatus with Gloss Meter and Manufacturing Method Using the Same
CN116410258A (en) * 2023-04-13 2023-07-11 上海太阳生物技术有限公司 Factor XI deficiency plasma protective agent and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009067660A2 (en) * 2007-11-21 2009-05-28 Oregon Health & Science University Anti-factor xi monoclonal antibodies and methods of use thereof
WO2017162791A1 (en) * 2016-03-23 2017-09-28 Prothix Bv Monoclonal antibodies against the active site of factor xi and uses thereof
WO2018145533A1 (en) * 2017-02-10 2018-08-16 上海仁会生物制药股份有限公司 Anti-coagulation factor xi antibody

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2014202009A1 (en) * 2007-11-21 2014-05-01 Oregon Health & Science University Anti-factor xi monoclonal antibodies and methods of use thereof
ES2702365T3 (en) * 2008-06-19 2019-02-28 Prothix Bv Use of anti-factor XI antibodies for the prevention or treatment of thrombus formation
LT2373691T (en) * 2008-12-18 2019-05-27 Oregon Health & Science University Anti-fxi antibodies and methods of use
HUE040580T2 (en) * 2012-05-10 2019-03-28 Bayer Pharma AG Antibodies capable of binding to the coagulation factor xi and/or its activated form factor xia and uses thereof
JOP20200312A1 (en) * 2015-06-26 2017-06-16 Novartis Ag Factor xi antibodies and methods of use
KR20180104036A (en) * 2016-01-22 2018-09-19 머크 샤프 앤드 돔 코포레이션 Anti-coagulation factor XI antibody
JP2021534098A (en) * 2018-08-09 2021-12-09 上海仁会生物制▲やく▼股▲ふん▼有限公司Shanghai Benemae Pharmaceutical Corporation Anti-factor XI antibody

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009067660A2 (en) * 2007-11-21 2009-05-28 Oregon Health & Science University Anti-factor xi monoclonal antibodies and methods of use thereof
WO2017162791A1 (en) * 2016-03-23 2017-09-28 Prothix Bv Monoclonal antibodies against the active site of factor xi and uses thereof
WO2018145533A1 (en) * 2017-02-10 2018-08-16 上海仁会生物制药股份有限公司 Anti-coagulation factor xi antibody

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3914624A4 (en) * 2019-01-21 2022-10-19 Aronora, Inc. Novel humanized antibodies against factor xi having anti-thrombotic and anti-inflammatory effects and uses thereof

Also Published As

Publication number Publication date
CN114478781A (en) 2022-05-13
CN114478781B (en) 2024-04-02
BR112021002472A2 (en) 2021-07-27
AU2018436195A1 (en) 2021-02-18
CN116554334A (en) 2023-08-08
EP3833692A1 (en) 2021-06-16
KR20210042352A (en) 2021-04-19
CN113227150A (en) 2021-08-06
MX2021001613A (en) 2021-04-28
JP2021534098A (en) 2021-12-09
CN114478782A (en) 2022-05-13
CA3108708A1 (en) 2020-02-13
CN113227150B (en) 2023-07-28
CN114478782B (en) 2024-04-02

Similar Documents

Publication Publication Date Title
WO2020029179A1 (en) Anti-factor xi antibodies
EP3581587A1 (en) Anti-coagulation factor xi antibody
JP7366988B2 (en) Anticoagulant factor XI antibody
JP2024054266A (en) Tissue factor pathway inhibitor antibodies and uses thereof
JP7022081B2 (en) Anticoagulation factor XI antibody
JP2019534282A (en) Anti-IL-33 antibody and use thereof
US11958911B2 (en) Anti-coagulation factor XI antibody
WO2022002249A1 (en) Anti-fxi/fxia antibody, antigen-binding fragment thereof, and pharmaceutical use thereof
RU2800719C2 (en) Antibodies to factor xi
WO2023044280A1 (en) Anti-il-1-beta antibodies
NZ786444A (en) Anti-coagulation factor xi antibodies

Legal Events

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

Ref document number: 18929685

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3108708

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2021506623

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018436195

Country of ref document: AU

Date of ref document: 20180809

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021002472

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217006922

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2018929685

Country of ref document: EP

Effective date: 20210309

ENP Entry into the national phase

Ref document number: 112021002472

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210209