NL2036011B1 - Molecules for reversing anti-coagulant activity of direct oral anticoagulants - Google Patents
Molecules for reversing anti-coagulant activity of direct oral anticoagulants Download PDFInfo
- Publication number
- NL2036011B1 NL2036011B1 NL2036011A NL2036011A NL2036011B1 NL 2036011 B1 NL2036011 B1 NL 2036011B1 NL 2036011 A NL2036011 A NL 2036011A NL 2036011 A NL2036011 A NL 2036011A NL 2036011 B1 NL2036011 B1 NL 2036011B1
- Authority
- NL
- Netherlands
- Prior art keywords
- seq
- doac
- polypeptide
- cdr
- amino acid
- Prior art date
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
- A61P7/04—Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/44—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
- A61K2039/507—Comprising a combination of two or more separate antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/76—Albumins
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/22—Immunoglobulins specific features characterized by taxonomic origin from camelids, e.g. camel, llama or dromedary
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/31—Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/35—Valency
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/56—Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
- C07K2317/569—Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/76—Antagonist effect on antigen, e.g. neutralization or inhibition of binding
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Pharmacology & Pharmacy (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biophysics (AREA)
- Biochemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Genetics & Genomics (AREA)
- Animal Behavior & Ethology (AREA)
- Epidemiology (AREA)
- Mycology (AREA)
- Microbiology (AREA)
- Diabetes (AREA)
- Hematology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Peptides Or Proteins (AREA)
Abstract
The present invention relates to binding molecules capable of reversing the anti-coagulant activity of direct oral anticoagulants in-vivo and in-vitro. The present invention, more particularly concerns binding molecules for reversing the factor Xa inhibition activity of a factor Xa inhibitor such as in a patient to whom a factor Xa inhibitor is administered. The present invention is further related to binding molecules capable of reversing the anti-coagulant activity of direct oral anticoagulants, for use in medicine and diagnostics. More particularly, the present invention concerns binding molecules for use in the treatment or prevention of bleeding, and for use in the detection and quantification of such molecules, e.g. in human plasma. The present invention further provides a method for detecting a factor Xa inhibitor in a 10 sample.
Description
MOLECULES FOR REVERSING ANTI-COAGULANT ACTIVITY OF DIRECT ORAL
ANTICOAGULANTS
The present invention relates to binding molecules capable of reversing the anti-coagulant activity of direct oral anticoagulants in vivo and in vitro. The present invention, more particularly concerns binding molecules for reversing inhibition of the factor Xa by a factor Xa inhibitor, possibly in a patient to whom a factor Xa inhibitor was administered. The present invention further relates to binding molecules capable of reversing the anti-coagulant activity of direct oral anticoagulants, for use in medicine and diagnostics. More particularly, the present invention concerns binding molecules for use in the treatment or prevention of bleeding, and for use in the detection and quantification of such molecules, e.g. in human plasma. The present invention further provides a method for detecting a factor
Xa inhibitor in a sample.
Anticoagulants are developed for the prevention and reduction of recurrent venous thromboembolism, stroke prevention in patients with non-valvular atrial fibrillation, and for reducing the incidence of recurrent ischemic events and death in patients with acute coronary syndrome.
Anticoagulation therapy is also required in patients having clotting disorders, restricted mobility or undergoing medical surgery.
An example of oral anticoagulants are vitamin K antagonists (VKAs) such as warfarin which is indicated in the long-term antithrombotic therapy by acting on the vitamin K-dependent synthesis of coagulation factors including factor VII, factor X, factor IX, factor ll, protein C and protein S. The risk of bleeding in patients administered with warfarin is the most common side effect. The prevention of bleeding requires constant tailored dosing based on frequent international normalized ratio (INR) monitoring. Still, major and fatal bleedings that can occur are the main concerns of the warfarin use.
Direct oral anticoagulants ("DOACSs”) are provided as alternatives to warfarin and there are, to date, four US - approved direct oral anticoagulants called non-vitamin-K-antagonist oral anticoagulants (NOACs) or DOACs. DOACs encompass four direct activated factor X ("fXa”) inhibitors: apixaban, betrixaban, edoxaban and rivaroxaban. Betrixaban is approved for recurrent venous thromboembolism (VTE) whereas other fXas are approved for reducing the risk of stroke and systemic embolism in patients with nonvalvular AF (NVAF), for treating deep vein thrombosis (DVT) and pulmonary embolism (PE), and for DVT prophylaxis. Rivaroxaban is the first approved DOAC in Europe by EMA for the prevention of venous thromboembolic events (VTE) in adult patients who have undergone elective total hip replacement surgery or total knee replacement surgery. Then, the direct fXa inhibitor apixaban is approved in 2011. The indication for the prevention of stroke and systemic embolism in adult patients with nonvalvular atrial fibrillation was granted for all these molecules with additional indications being approved later. Edoxaban is approved for prevention in NVAF, for treatment and prevention of deep vein thrombosis and pulmonary embolism, as well as prevention of recurrences in adults. Less major and fatal bleeding events occur upon the administration of DOACs.
DOACs are usually preferred in comparison to vitamin K antagonists such as warfarin due to occurrence of less side effects and rapid anticoagulant effect. The use of DOACs does not require blood monitoring and they can be administered in fixed doses. They display several advantages in view of the
VKAs including comparatively stable pharmacokinetic and pharmacodynamic profiles, eliminating the need for repeated coagulation assay monitoring and dose adjustments, and leading to minimal drug and food interactions. Therefore, DOACs over time became the first-choice drugs particularly for some indications. For example, for patients with nonvalvular atrial fibrillation apixaban is the most prescribed
DOAC.
Yet, despite these advantages, DOACs also cause side effects and display some disadvantages. It is reported in clinical trials that patients having atrial fibrillation to whom fXa inhibitors are administered, displayed major bleeding at an annualized rate of 2.1 to 3.5%. Clinical experience has confirmed these results. Anticoagulation treatment causing major bleeding remains associated with an increased risk of death and thrombotic events unrelated to the type of anticoagulant used.
Furthermore, patients to whom a DOAC is administered may have an increased risk of bleeding, particularly in case of an emergency surgery (Siegal et al. Siegal DM, Curnutte JT, Connolly SJ, Lu G,
Conley PB, Wiens BL, Mathur VS, Castillo J, Bronson MD, Leeds JM, Mar FA, Gold A, Crowther MA.
Andexanet Alfa for the Reversal of Factor Xa Inhibitor Activity. N Engl J Med. 2015 Dec 17;373(25):2413-24. doi: 10.1056/NEJMoa1510991. Epub 2015 Nov 11. PMID: 26559317.).
The effect of the anticoagulants needs to be reversed or eliminated or needs to be in homeostatic status in case of an emergency, an uncontrolled bleeding or a surgical need. The discontinuation of the administration of such anticoagulants does not immediately provide this homeostatic status in blood and thus there is a demand for agents and/or molecules which can reverse or neutralize or inhibit the effects of anticoagulants, in particular of DOACs.
Major bleeding is reported in phase III trials for atrial fibrillation and for venous thromboembolism patients administered with rivaroxaban and apixaban, in particular. The data from the clinics show that rivaroxaban and warfarin are comparable regarding the rate of adverse effects observed including stroke, systemic embolism and major bleeding (Sarfori et al.; Sartori & Cosmi,
Andexanet alfa to reverse the anticoagulant activity of factor Xa inhibitors: a review of design, development and potential place in therapy, Journal of Thrombosis and Thrombolysis (2018) 45:345- 352, doi.org/10.1007/s11239-018-1617-2).
These adverse effects of DOACs and their increased use in clinics pose a strong need for reversal agents (‘antidotes’) which can inhibit and/or neutralize the anticoagulant activity of DOACSs.
Discontinuing the DOAC will result in the elimination of the drug from the blood circulation in relatively short times due to the short half-lives of the DOACs ranging from 5 to 17 hours. However, in cases of emergencies including an undesired bleeding such as a life-threatening major bleeding, or non-elective major surgery anticoagulation, a faster method for reversing or neutralizing the effects of DOACs is desired.
Andexanet alfa is developed with the objective of inhibiting the anticoagulant activity of DOACs (rivaroxaban, apixaban, edoxaban, betrixaban) and other fXa indirect inhibitors including low molecular weight heparin (LMWH) and fondaparinux, to restore the coagulation. Andexanet alfa is a decoy protein which lacks the factor X Gla domain, membrane-binding-carboxyglutamic acid domain crucial for its binding to phospholipids. Accordingly, Andexanet alfa competes with the native factor Xa for binding to a DOAC administered to the patient, and Andexanet alfa neutralizes a DOAC by impairing the binding of the DOAC to the native factor Xa, which fXa can in turn generate thrombin (Sartori et al.).
Andexanet alfa sequesters the anticoagulant effects of DOACs including rivaroxaban, apixaban, and betrixaban and reverses their anticoagulant effect in a dose-dependent manner.
Andexanet alfa can interact with activated antithrombin Ill and therefore can also completely reverse the effects of enoxaparin or fondaparinux in a dose-dependent manner. Additionally, however,
Andexanet alfa can bind and inhibit the activity of tissue factor pathway inhibitor (TFPI) and increases the tissue factor-initiated thrombin generation (Nihar R. Desai & David Cornutt (2019) Reversal agents for direct oral anticoagulants: considerations for hospital physicians and intensivists, Hospital Practice, 47:3, 113-122 (“Nihar et al.”)).
Clinical studies report that Andexanet alfa can cause thrombotic events. The patients administered with Andexanet alfa are at risk to develop thrombosis due to the pro-coagulant effect of
Andexanet alfa mediated by inhibition of TFPI. In bleeding patients, the duration of this effect is unknown. In healthy volunteers administered with Andexanet alfa, increase in the coagulation markers such as F1+2, TAT, and D-dimer and dose-dependent decreases in TFPI is reported. Yet, the clinical studies are conducted only among healthy volunteers (ANNEXA-A and ANNEXA-R clinical trials) and patients with acute major bleeding (ANNEXA-4 clinical trial). Data on patients undergoing emergency surgery or invasive procedures are not available. The FDA boxed warning states that Andexanet alfa treatment has been associated with serious and life-threatening adverse events (AEs), including arterial and venous thromboembolic events; ischemic events, including myocardial infarction and ischemic stroke; cardiac arrest; and sudden death. Physicians are advised to monitor for thromboembolic events and initiate anticoagulation when appropriate. Monitoring for symptoms and signs that precede cardiac arrest and providing treatment, as needed, is also advised.
The ability of Andexanet alfa for inhibiting more than one DOAC and its interaction with other actors of the coagulation cascade can cause undesired or unpredictable side effects and also requires the administration of Andexanet alfa in high doses. High concentrations of Andexanet alfa in the circulation are required, relating to a high proportion of Andexanet alfa cross-reacting with antithrombin
III (ATI) which is highly abundant in blood, and with TFPI, which results in a loss of factor Xa scavenging function for Andexanet alfa.
Reversal treatments give rise to some potential complications, for example, incomplete reversal can cause thrombotic events, and death. Major limitations of the treatment with Andexanet alfa is the hampered and limited availability of Andexanet alfa and cumbersome and technically difficult manufacturing protocol (also resulting in relatively high costs) of Andexanet alfa, individually and both in combination severely hampering supply and accessibility of the therapeutic drug for patients in need thereof. Therefore, Andexanet alfa is not reliably and sustainably affordable and applicable as antidote for the majority of emergency settings or in surgery rooms.
In view of the above, there is a strong need for a treatment modality for specifically inhibiting, or reversing, or neutralizing the effects of direct oral anticoagulation agents, causing less further side effects particularly in patients with a bleeding disorder, a bleeding patient, and in patients in need of (an emergency) surgery. The treatment modality is required to provide rapid and complete reversal of the anticoagulant activity of a DOAC. In case of emergency, it is crucial to know whether the patient is treated with a DOAC. This generates a need for method for detecting the presence of a DOAC in the circulation of a patient, particularly if the detailed history of the patient is unknown.
In view of the above, there is a need for a therapy for inhibiting the anticoagulant activity of
DOACs.
The use of Andexanet alfa in diagnostic tests has also been investigated. Andexanet alfa could perhaps be a potential tool to reverse DOAC function in patient plasma to 0%, similar to the patient's physiological coagulation profile. Andexanet alfa is however not reliable for this diagnostic application, since spiking resulted in partial FXa activity reversal profiles of apixaban (22%), betrixaban (56%), edoxaban (28%), and rivaroxaban (49%) (Siddiqui et al., Reversal of Factor Xa Inhibitors by Andexanet
Alfa May Increase Thrombogenesis Compared to Pretreatment Values, Clinical and Applied
Thrombosis/Hemostasis, Volume 25: 1-7 (2019) doi: 10.1177/1076029619863493}. In addition,
Siddiqui et al. established that Andexanet alfa cannot be used for a complete reversal of DOAC profiles in thrombin generation (TG), because Andexanet alfa spiking results in an increased TG in comparison to saline, due to complete inhibition of tissue factor pathway inhibitor (TFPI).
In view of the above, there is a need for a diagnostic tool for testing for the occurrence of inhibition of coagulation by a DOAC in for example human plasma.
It is an objective of the present invention to provide such therapy. It is further an objective of the present invention to provide diagnosis methods for detecting presence of a DOAC in a human blood sample.
The present invention resides in the finding that inhibition of activated factor X by a direct oral anti-coagulant (DOAC) capable of inhibiting factor Xa such as rivaroxaban, edoxaban and apixaban can be efficiently and completely reversed by a VHH acting as an antidote for the DOAC. Such VHH is raised in a camelid such as for example llama upon immunization of the llama with a small-molecule
DOAC. VHH capable of specific binding and specific inhibition of a selected DOAC therewith restoring factor Xa activity in e.g. plasma, is established with regard to DOACs rivaroxaban, edoxaban and apixaban. Surprisingly, the inventors succeeded in providing a series of different VHHs, each series capable of specifically binding and inhibiting a certain selected DOAC capable of inhibiting fXa, selected from rivaroxaban, edoxaban and apixaban. Therewith, the invention provides the possibility to reverse 5 the fXa inhibiting activity of a certain DOAC by administering such DOAC specific VHH to a patient to whom the DOAC is administered, For example, a patient to whom apixaban is previously administered can be treated with a VHH specific for inhibiting the fXa activity inhibiting activity of apixaban, therewith neutralizing the anti-coagulant activity of apixaban and restoring the Xa activity. Similarly, for the VHHs specific for inhibiting the fXa activity inhibiting activity of one of rivaroxaban, edoxaban and betrixaban.
The present invention therefore aims to provide molecules (“antidotes”) which specifically bind to a DOAC that inhibits fXa without any cross reactivity to other DOACs not being administered to the patient, for the rapid and complete reversal of the fXa inhibitory activity of the DOAC administered. The desired antidote should preferably not act on other proteins which are implicated in the coagulation cascade, for preventing undesired effects or prolonged anticoagulant effects.
The present invention aims to provide molecules (“antidotes”) which specifically bind to a selected DOAC that inhibits fXa without any cross reactivity to other DOACs, such as other DOACs not administered to a human subject (patient) to whom the selected DOAC is administered, for amongst others the purpose of the detection and quantification of the fXa inhibitory activity of the selected DOAC administered to the subject or present in a sample of ((human) patient) blood, plasma and any other body fluid. The antidote does preferably not act on other proteins than activated factor X, which are implicated in the coagulation cascade.
The present invention relates to a binding molecule, preferably a polypeptide, which specifically binds to a DOAC for inhibiting activated factor X (fXa), such as a DOAC present in the blood circulation of a subject or, in-vitro, in a body fluid taken from a (human) subject, for diagnostic purposes. The binding molecule according to the present invention (the polypeptide of the invention) is for example administered in amounts sufficient to inhibit the effects of the DOAC and to reverse the anticoagulation therapy provided by the said DOAC, therewith restoring the normal homeostasis in said subject by reversing the activated factor X inhibition by the DOAC. The administration of the polypeptide of the invention to a subject in need thereof is particularly required when the subject such as a human patient needs to undergo surgery with urge or in cases when the effect of a DOAC administered to the subject should be neutralized or when the DOAC should be eliminated from the circulation of the subject, for example during an emergency occasion including an undesired (excessive) bleeding or other adverse effects due to the DOAC.
The present invention relates to a polypeptide comprising or consisting of at least one antibody which antibody specifically binds to a first direct oral anticoagulant (“DOAC"} which DOAC is an inhibitor of activated factor X (“Xa”). The polypeptide preferably comprises or consists of a single domain antibody (sdab), more preferably a VHH.
The present invention also relates to a polypeptide comprising an antibody such as a single variable domain that specifically binds to a DOAC that inhibits fXa. More particularly, the invention relates to a polypeptide comprising a single variable domain that specifically binds to a DOAC that inhibits Xa, wherein the single variable domain comprises a combination of three complementarity determining region (CDR) sequences (CDR 1, CDR 2, CDR 3) selected from the group consisting of the CDR sequences with SEQ ID NO: 1-50 (listed in Table A and Tables B-D, displayed below). In a preferred embodiment, the polypeptide selectively binds (at least or only, preferably only) any one of edoxaban, apixaban and rivaroxaban.
In an embodiment of the invention, the polypeptide comprises sdab('s) specifically binding to at least one DOAC (capable of inhibiting activated factor X) and wherein the polypeptide has one or more of the characteristics selected from the group consisting of: a) in human plasma, the molar ratio between the polypeptide and a DOAC, required for inhibiting the fXa inhibiting activity of the DOAC with at least 50%, is a ratio selected from 1:1 — 20:1, such as 1:1 for inhibiting the fXa inhibiting activity of edoxaban, 4:1 — 16:1 for inhibiting the fXa inhibiting activity of apixaban, and 3:1 — 16:1 for inhibiting the fXa inhibiting activity of rivaroxaban; b) the polypeptide at room temperature has an association rate constant (ka) of 1.00e+08 1/Ms or higher for binding to a DOAC, as determined with surface plasmon resonance with immobilized DOAC, such as 1.00e+06 1/Ms — 1.00e+08 1/Ms for binding to apixaban, edoxaban, rivaroxaban or betrixaban, preferably apixaban, and c) the polypeptide at room temperature has a dissociation constant (ks) of 1.00e-01 1/s or lower, when bound to a DOAC, as determined with surface plasmon resonance with immobilized DOAC, such as 1.00e-01 1/s — 1.00e-03 1/s for dissociating from apixaban, edoxaban, rivaroxaban or betrixaban, preferably apixaban; and/or d) the polypeptide at room temperature has an affinity (Ko) of 1.00e-8 nM or lower, for binding to the first, second and/or third DOAC, wherein the first DOAC is edoxaban, the second
DOAC is apixaban and the third DOAC is rivaroxaban, as determined with surface plasmon resonance with immobilized DOAC, such as 1.00e-8 nM — 1.00e-10 nM for binding to apixaban, edoxaban, rivaroxaban or betrixaban, preferably apixaban; and/or e) the polypeptide at room temperature has an affinity (Ko) of 1.00e-8 nM or lower, for binding to the first, second and/or third DOAC, wherein the first DOAC is edoxaban, the second
DOAC is apixaban and the third DOAC is rivaroxaban, as determined with ELISA with immobilized albumin-DOAC, such as 1.00e-8 nM — 1.00e-10 nM for binding to edoxaban, 1.00e-8 nM — 1.00e-10 nM for binding to apixaban, and
1.00e-8 nM — 1.00e-10 nM for binding to rivaroxaban.
An aspect of the invention relates to a mixture of at least two polypeptides according to the invention, wherein a first polypeptide specifically binds to a first DOAC and a second polypeptide specifically binds to a second DOAC, different from the first DOAC, and, if present, a third polypeptide specifically binds to a third DOAC different from the first DOAC and different from the second DOAC.
A further aspect of the invention relates to a pharmaceutical composition comprising a polypeptide according to the invention and a pharmaceutically acceptable carrier and/or a pharmaceutically acceptable diluent.
A further aspect of the invention relates to a pharmaceutical composition comprising the mixture of at least two polypeptides according to the invention and a pharmaceutically acceptable carrier and/or a pharmaceutically acceptable diluent.
An embodiment is the polypeptide of the invention, the mixture of polypeptides of the invention or a pharmaceutical composition of the invention, wherein the (first) DOAC, (second) DOAC and (third)
DOAC are selected from edoxaban, apixaban and rivaroxaban.
An aspect of the invention relates to the polypeptide according to the invention, the mixture of polypeptides of the invention or a pharmaceutical composition according to the invention, for use as a medicament.
A further aspect of the invention relates to the polypeptide according to the invention, the mixture of polypeptides of the invention or the pharmaceutical composition according to the invention, for use in the treatment of inhibited coagulation in a patient or in the treatment of inhibited fXa activity in a patient and/or for use in the prevention or treatment of bleeding or major bleeding in a patient or uncontrolled bleeding in a patient, and/or for use in the reversal of anti-coagulant activity in a patient to whom an anti-coagulant such as an inhibitor of activated factor X, for example a DOAC, is administered.
In an embodiment of the invention, the polypeptide according to the invention, the mixture of polypeptides of the invention or the pharmaceutical composition according to the invention, is for use according to the invention, wherein a DOAC that is an inhibitor of fXa is previously administered to said patient, before the polypeptide, the mixture of polypeptides or the pharmaceutical composition is administered to said patient, and wherein the polypeptide or at least one of the polypeptides comprised by the mixture of polypeptides or comprised by the pharmaceutical composition specifically binds to said DOAC.
In an embodiment of the invention, for said use, the DOAC is selected from edoxaban, apixaban and rivaroxaban.
An aspect of the invention relates to a nucleic acid molecule comprising a nucleotide sequence encoding the polypeptide according to the invention, wherein the nucleotide sequence encoding the polypeptide further preferably encodes a signal peptide operably linked to the polypeptide, and wherein the nucleic acid molecule further preferably comprises regulatory elements conducive to the expression of the polypeptide, which regulatory elements are operably linked to the nucleotide sequence.
A further aspect relates to a host cell comprising a nucleic acid molecule according to the invention.
An aspect of the invention relates to a method for producing a polypeptide according to the invention, the method comprising the steps of: a) culturing a host cell of the invention under conditions conducive to the expression of the polypeptide; and, b) recovery of the polypeptide.
An aspect of the invention relates to a method for producing a pharmaceutical composition of the invention, wherein the method comprises the steps of: a) producing at least one polypeptide of the invention in a method according to the invention; and, b) formulating the polypeptide(s} with a pharmaceutically acceptable carrier and/or pharmaceutically acceptable diluent to obtain a pharmaceutical composition.
An aspect of the invention relates to the use of the polypeptide(s) according to the invention or a pharmaceutical composition according to the invention for in vitro detection or quantification of a DOAC in a sample.
In an embodiment of the invention, in said use e the DOAC is any one of edoxaban, apixaban and rivaroxaban; and/or e the sample is a plasma sample obtained from a patient; and/or e the sample is obtained from a patient to whom previously a DOAC is administered; and/or e the sample is obtained from a patient who suffers from bleeding, major bleeding or uncontrolled bleeding, or is at risk for bleeding, major bleeding or uncontrolled bleeding; and/or e the sample is obtained from a patient who needs to be subjected to surgery or is or has been subjected to surgery.
A further aspect of the invention relates to a kit comprising the polypeptide(s) of the invention or the pharmaceutical composition of the invention, and optionally further comprising instructions for administration of the polypeptide(s) or the pharmaceutical composition, such as administration to a patient in need of inhibition of fXa inhibition by a previously administered DOAC and/or to a patient at risk of bleeding, major bleeding or uncontrolled bleeding, or suffering from bleeding, major bleeding or uncontrolled bleeding, or to a patient according to the invention.
The antidote (polypeptide) according to the present invention preferably reverses selectively function of a selected DOAC to 0% in factor Xa measurements in patient plasma. Comparison of factor
Xa tests in patient plasma with and without spiked antidotes is an diagnostic tool to determine the patient's individual DOAC function response.
An additional diagnostic application of the current invention is to distinguish which specific
DOAC is administered to the (human) patient.
An aspect of the invention relates to a detection method for determining the presence of a DOAC inhibitor of fXa in a sample obtained from a subject, the method comprising the step of contacting the sample with the polypeptide of the invention, wherein the polypeptide is used in detecting the DOAC inhibitor of fXa in the sample.
In an embodiment of the invention, the detection method comprises the steps of: - providing the sample obtained from the subject, - assessing a reference value associated with fXa activity in the sample or in a first aliquot of the sample; - contacting the sample or a second aliquot of the sample with the polypeptide of the invention and assessing a first test value after the contacting; wherein finding a difference between the first test value and the reference value indicates the presence of the DOAC inhibitor of fXa in the sample obtained from the subject.
An aspect of the invention relates to a kit and/or a kit of parts for performing the detection method of the invention, the kit or kit of parts comprising at least the polypeptide of the invention, and further optionally comprising one or more reagents for assessing the reference value and/or the first test value.
It will be apparent to those skilled in the art, based on the present disclosure, that the main aspects of the invention, as defined here above and below, are based on the same innovative concepts.
The innovative concepts presented herein will be described with respect to particular embodiments.
Unless stated otherwise and/or unless something else is apparent from the context, the particular embodiments described herein below apply, indiscriminately, to each and every one of the aspects of the invention as defined herein above. Particular embodiments described herein should be regarded as descriptive and not limiting beyond of what is described in the claims. The embodiments as described herein can operate in combination and cooperation, unless specified otherwise.
The term “lag time” has its regular scientific meaning and here refers to the time needed for the first amounts of thrombin to be generated in a thrombin generation test. Peak thrombin generation levels is the maximum amount of active thrombin that is formed in a calibrated thrombin generation test. This
Peak thrombin concentration represents the balance between pro-coagulant and anti-coagulant factors in blood plasma. An example of a suitable test for assessing the lag time of thrombin generation is: the calibrated automated thrombinogram CAT assay method invented by Synapse Research Institute and disclosed in WO2018151602 A1 and described in Hemker et al. (2009) (Hemker HC, Giesen P, Al Dieri
R, Regnault V, de Smedt E, Wagenvoord R, Lecompte T, Béguin S. Calibrated automated thrombin generation measurement in clotting plasma. Pathophysiol Haemost Thromb. 2003;33(1):4-15. doi: 10.1159/00007 1636. PMID: 12853707 and Coen Hemker H., Hemker PW, Al Dieri R. The technique of measuring thrombin generation with fluorescent substrates: 4. The H-transform, a mathematical procedure to obtain thrombin concentrations without external calibration. Thromb Haemost. 2009; 101(01): 171-177 doi: 10.1160/TH08-09-0562).
As used herein, the term “anticoagulant” has its regular scientific meaning and here refers to a chemical substance which prevents or reduces coagulation of blood and extends the clotting time in- vivo and in-vitro. Anticoagulants are commonly known as blood thinners.
The term “direct oral anticoagulant” or “DOAC” has its regular scientific meaning and here refers to a (small molecule) therapeutic which is taken orally to prevent blood clot formation by directly inhibiting certain coagulation factors, and here specifically factor Xa, unless specified otherwise.
In an embodiment, the direct oral anticoagulant is or comprises a small molecule therapeutic (such as a therapeutic capable of inhibiting fXa). Preferably, the small molecule therapeutic has a molecular weight of 750 Dalton (Da) or less, preferably less than 650 Da, preferably less than 600 Da, more preferably less than 550 Da.
In an embodiment, the direct oral anticoagulant is in a pharmaceutically acceptable form including any salts, prodrugs thereof.
Typical direct oral anticoagulants (DOACs) include apixaban, edoxaban, rivaroxaban and betrixaban.
Apixaban is known from US 6,967,208 B2, which discloses lactam-containing compounds and derivatives thereof which are inhibitors of trypsin-like serine protease enzymes, especially factor Xa, under the name 1-(4-methoxyphenyi)-7-oxo-6-(4-(2-oxopiperidin-l-yl)phenyl)-4,5,8,7-tetrahydro-1H- pyrazolo[3,4-c]pyridine-3-carboxamide. Reference is made to Pinto et al. (2007) “Discovery of I-(4- methoxyphenyl)-7-oxo-6-(4-(2-oxopiperidin-I-yhphenyl)-4,5,6, 7-tetrahydro-!H-pyrazolo{f3, 4-c]pyridine- 3-carboxamide (apixaban, BMS-562247), a highly potent, selective, efficacious, and orally bioavailable inhibitor of blood coagulation factor Xa" Med. Chem. 55(22):5339-5356. Its molecular formula is
CasH25Ns04, which corresponds to a molecular weight of 459.50 g/mol {CAS 503612-47-3). Apixaban has the following structural formula (I):
O
TS
N / a ir hl N
N ” \ _
OC
(!)
Apixaban is a selective Factor Xa inhibitor and it inhibits free and clot-bound fXa, and inhibits thrombinase activity without requiring antithrombin II! (ATIII} for antithrombotic activity. Apixaban having no direct effect on platelet aggregation, indirectly inhibits platelet aggregation induced by thrombin. By inhibiting fXa, apixaban decreases thrombin generation and thrombus development. As a result of fXa inhibition, apixaban prolongs clotting tests such as prothrombin time (PT), INR, and activated partial thromboplastin time (aPTT).
Edoxaban is another selective fXa inhibitor which is provided as edoxaban tosylate monohydrate in a pharmaceutical formulation, and is disclosed in US 7,365,205 B2. The chemical name of edoxaban is N'-{5-chioropyridin-2-y-N-[{15,2R 48 }-4-{dimethyicarbamoyi}-2-{8-methyl- 4H 85H,6H,7H-[1,3lthiazolof 5 4-clpyridine-2amido}cyclohexylijethanediamide. Its molecular formula is
CaaHaoliN7O2S which corresponds to a molecular weight of 548.06 gimol {CAS d430449-70-5),
Edaxaban has the following structural formula (11):
NL
{ N /
NN / “TN
NH
~
IT
HN
/ \ ú 3 /
AN | / / ~~ a S G ‚NN 7 \ & (mn
Edoxaban tosylate monohydrate has the empirical formula C24Ha30CIN704S:C7Hs03S:H20 representing a molecular weight of 738.27 (CAS 480449-71-6).
Edoxaban is indicated for the treatment of deep vein thrombosis (DVT) and pulmonary embolism (PE) following 5 to 10 days of initial therapy with a parenteral anticoagulant. Edoxaban, like apixaban, acts selectively on fXa and inhibits free fXa, and prothrombinase activity and thrombin-
induced platelet aggregation, which results in the reduction of thrombin generation and thrombus formation.
Rivaroxaban is known from US 7,157,456 B2 disclosing novel oxazolidinone derivatives used in the field of blood coagulation. The chemical name of rivaroxaban is 5-Chloro-N-({(5S)-2-ox0-3-[4-(3- oxo-4-morpholinyl)phenyl]-1,3-oxazolidin-5-yl}methyl)-2-thicphenecarboxamide. Its molecular formula is CishHisG/N:055S and the molecular weight is 435.88 g/mol (CAS 366789-02-8). Rivaroxaban has the following structural formula (III):
Q
Q
8S
N \ Cl * J 0 N N re
Q
(nn
Rivaroxaban inhibits free fXa and prothrombinase activity and results in decreasing thrombin generation. It is indicated to reduce the risk of stroke and systemic embolism in adult patients with nonvalvular atrial fibrillation.
Betrixaban is disclosed in US 6,376,515 B2 and US 6,835,739 B2 which relates to novel compounds which are potent and highly selective inhibitors of isolated factor Xa or fXa when assembled in the prothrombinase complex. The chemical name of betrixaban is N-{5-chloropyridin-2-yl}-2-[4-(N,N- dimethylcarbamimidoyl)benzamido]-5-methoxybenzamide. Its molecular formula is C23H220INsC: and the molecular weight is 451.971 g/mol {CAS 330842-05-7). Betrixaban has the following structural formuia (IV):
Ci 9
NH y © ‚NH
Sy —_ EN a ~~ =
O Na
I
(IV)
Betrixaban selectively binds to the active site of fXa and does not require a co-factor such as
Antithrombin Ill for its inhibitory activity. It inhibits free fXa and prothrombinase activity by directly acting on fXa and it decreases thrombin generation. Betrixaban is indicated for the prophylaxis of venous thromboembolism (VTE) in adult patients hospitalized for an acute medical illness who are at risk for thromboembolic complications due to moderate or severe restricted mobility and other risk factors for
VTE.
The term “major bleeding” has its regular scientific meaning and here refers to the definition according to the Society on Thrombosis and Haemostasis, as published in 2019 in the appendix “Appendix 10 Bleeding Classification System Definitions” of the paper Dual Antiplatelet Therapy
Following Percutaneous Coronary Intervention: Clinical and Economic Impact of Standard Versus
Extended Duration by the Canadian Agency for Drugs and Technologies in Health (Bookshelf ID:
NBK542934, www.ncbi.nim.nih.gov/books/NBK542934/#:~:text=Major%3A%20fatal %20bleeding%2C%20and%2F, or%20greater)%200r%20more%2C%200r): fatal bleeding, and/or symptomatic bleeding in a critical area or organ, such as intracranial, intraspinal, intraocular, retroperitoneal, intra-articular or pericardial, or intramuscular with compartment syndrome, and/or bleeding causing a fall in hemoglobin levels of 1.24 mmol/L (20 g/L or greater} or more, or leading to a transfusion of 2 U or more of whole blood or red cells.
The term “reversal agent” or “reversing agent” or “antidote” or “neutralizing agent” or “normalize factor Xa inhibitor” or “noXi” refers to a molecule which inhibits or partially inhibits, for example at least 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or at least 99%, inhibits the activity of a DOAC by specifically binding to said DOAC in-vivo and in-vitro.
The term “reversing” or “neutralizing” or “normalizing” the effects of DOACs refers to eliminating specifically the effects of a DOAC or refer to restoring fully or partially the Factor Xa activity by inhibiting or partially inhibiting, for example at least 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or at least 99%, specifically the activity of the DOAC by binding the DOAC which in turn is unable to inhibit Factor
Xa. For instance, the Factor Xa activity can be restored to at least 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or at least 99%, upon binding of a reversal agent according to the invention to a DOAC.
As used herein, the term “antibody or a binding fragment thereof or a binding domain thereof” refers to a polypeptide that includes at least one immunoglobulin variable domain or at least one antigenic determinant, e.g., paratope that specifically binds to an antigen. In some embodiments, an antibody is a full-length antibody. In some embodiments, an antibody is a chimeric antibody. In some embodiments, an antibody is a humanized antibody. However, in some embodiments, an antibody is a
Fab fragment, a F(ab’) fragment, a F(ab')2 fragment, a Fv fragment or a scFv fragment. In some embodiments, an antibody is a nanobody (single domain antibody; sdab) derived from a camelid antibody, such as from a llama antibody, for example and preferably from llama heavy-chain only antibody (also referred to as single-chain antibody and heavy-chain antibody), or a nanobody derived from a shark antibody. In some embodiments, an antibody is a diabody. In some embodiments, an antibody comprises a framework having a human germline sequence. In another embodiment, an antibody comprises a heavy chain constant domain selected from the group consisting of IgG, 1gG1,
IgG2, 1IgG2A, IgG2B, IgG2C, IgG3, 1gG4, IgA1, IgA2, IgD, IgM, and IgE constant domains. In some embodiments, an antibody comprises a heavy (H) chain variable region (abbreviated herein as VH), and/or (e.g., and) a light (L) chain variable region {abbreviated herein as VL). In some embodiments, an antibody comprises a constant domain, e.g., an Fc region. An immunoglobulin constant domain refers to a heavy or light chain constant domain. Human IgG heavy chain and light chain constant domain amino acid sequences and their functional variations are known. With respect to the heavy chain, in some embodiments, the heavy chain of an antibody described herein can be an alpha (a), delta (D), epsilon (e), gamma (g) or mu (m) heavy chain. In some embodiments, the heavy chain of an antibody described herein can comprise a human alpha (a), delta (D), epsilon (e), gamma (g) or mu (m) heavy chain. In a particular embodiment, an antibody described herein comprises a human gamma 1
CHI, CH2, and/or (e.g., and) CH3 domain. In some embodiments, the amino acid sequence of the VH domain comprises the amino acid sequence of a human gamma (g) heavy chain constant region, such as any known in the art. Non-limiting examples of human constant region sequences have been described in the art, e.g., see U.S. Pat. No. 5,693,780 and Kabat EA et al, (1991) ("Sequence of proteins of immunological interest”, US Public Health Services, NIH Bethesda, Md. , Publication No. 91) supra.
In some embodiments, the VH domain comprises an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or at least 99% identical to any of the variable chain constant regions provided herein. In some embodiments, an antibody is modified, e.g., modified via glycosylation, phosphorylation, sumoylation, and/or (e.g., and) methylation. In some embodiments, an antibody is a glycosylated antibody, which is conjugated to one or more sugar or carbohydrate molecules. In some embodiments, the one or more sugar or carbohydrate molecule are conjugated to the antibody via N- glycosylation, O-glycosylation, C-glycosylation, glypiation (GPI anchor attachment), and/or (e.g., and)
phosphoglycosylation. In some embodiments, the one or more sugar or carbohydrate molecule are monosaccharides, disaccharides, oligosaccharides, or glycans. In some embodiments, the one or more sugar or carbohydrate molecule is a branched oligosaccharide or a branched glycan. In some embodiments, the one or more sugar or carbohydrate molecule includes a mannose unit, a glucose unit, an N-acetylglucosamine unit, an N-acetylgalactosamine unit, a galactose unit, a fucose unit, or a phospholipid unit. In some embodiments, an antibody is a construct that comprises a polypeptide comprising one or more antigen binding fragments of the disclosure linked to a linker polypeptide or an immunoglobulin constant domain. Linker polypeptides comprise two or more amino acid residues joined by peptide bonds and are used to link one or more antigen binding portions. Examples of linker polypeptides have been reported {see e.g., Holliger, P, et al. (1993) Proc. Natl. Acad. Sci. USA 90:6444- 6448; Poljak, R. J., et al. (1994) Structure 2:1121-1123). Still further, an antibody may be part of a larger immunoadhesion molecule, formed by covalent or noncovalent association of the antibody or antibody portion with one or more other proteins or peptides. Examples of such immunoadhesion molecules include use of the streptavidin core region to make a tetrameric scFv molecule (Kipriyanov, S. M., et al. (1995) Human Antibodies and Hybridomas 6:93-101) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molecules (Kipriyanov, S. M., et al. (1994) Mol. Immunol. 31:1047-1058).
The term “single domain antibody”, or “sdAb”, in short, or ‘nanobody’, has its regular scientific meaning and here refers to an antibody fragment consisting of a single monomeric variable antibody domain, unless referred to as more than one monomeric variable antibody domain such as for example in the context of a bivalent sdAb, which comprises two of such monomeric variable antibody domains in tandem. A bivalent nanobody is a molecule comprising two single domain antibodies targeting a
DOAC, such as fXa inhibiting DOACs edoxaban, apixaban, rivaroxaban, betrixaban. Preferably the
DOAC is edoxaban, apixaban or rivaroxaban A bivalent nanobody is also named a bivalent single domain antibody. Preferably the two different single domain antibodies are directly covalently bound or covalently bound through an intermediate molecule that is covalently bound to the two different single domain antibodies, such as an amino-acid linker sequence, for example the linker with the sequence according to SEQ ID NO: 104 (AAGGGGSGGGGSAAA). Preferably the intermediate molecule of the bivalent nanobody has a molecular weight of less than 10,000 Dalton, more preferably less than 5000
Dalton, even more preferably less than 2000 Dalton, most preferably less than 1500 Dalton. Preferably, the intermediate molecule is an amino-acid linker sequence.
A "single domain antibody" is an antibody or antibody fragment consisting only of heavy chains and devoid of light chains as are known e.g. from Camelids. A single domain antibody is thus an antibody comprising a “single variable domain" wherein the antigen binding site is present on, and formed by, the single variable domain (also referred to as an "immunoglobulin single variable domain" or "ISVD"). This sets single variable domains apart from "conventional" immunoglobulins or their fragments, wherein two immunoglobulin domains, typically a heavy chain variable domain (VH) and a light chain variable domain (VL), interact to form an antigen binding site. The term "single domain antibody” as used herein includes antibodies or antibody fragments comprising the single variable domains of camelid heavy chain antibodies (VHHs), also referred to as nanobodies, domain antibodies (dAbs), and single domain antibodies derived from shark (IgNAR domains). The term "single domain antibody” as used herein can refer to polypeptides either comprising or consisting of a single variable domain. In preferred embodiments the single domain antibody comprises or consists of VHH domain(s).
Generally, single variable domains will be amino acid sequences that essentially consist of 4 framework regions (FR1 to FR4 respectively) and 3 complementarity determining regions (CDR1 to
CDR3 respectively). Such single variable domains and fragments are most preferably such that they comprise an immunoglobulin fold or are capable for forming, under suitable conditions, an immunoglobulin fold. Thus, the amino acid sequence and structure of an immunoglobulin variable domain sequence, in particular a single variable domain can be considered - without however being limited thereto - to be comprised of four framework regions or "FR’s", which are referred to in the art and herein as "Framework region 1" or "FR1"; as "Framework region 2" or "FR2"; as "Framework region 3" or "FR3"; and as "Framework region 4" or "FR4", respectively; which framework regions are interrupted by three complementary determining regions or "CDR's", which are referred to in the art as "Complementarity Determining Region 1" or "CDR1"; as "Complementarity Determining Region 2" or "CDR2"; and as "Complementarity Determining Region 3" or "CDR3", respectively. The CDRs may also be referred to as "hypervariable regions" (HVRs). The total number of amino acid residues in a single variable domain can be in the region of 110-120, is preferably 112-115, and is most preferably 113. It should however be noted that parts, fragments, analogues or derivatives of a single variable domain are not particularly limited as to their length and/or size, as long as such parts, fragments, analogues or derivatives meet the further requirements outlined herein and are also preferably suitable for the purposes described herein.
Thus, in the meaning of the present invention, the term "single domain antibody" comprises polypeptides which are derived from a non-human source, preferably a camelid, preferably a camel heavy chain antibody or a llama heavy chain antibody. They may be humanized, as previously described, e.g. in WO 08/101985 and WO 08/142164. Moreover, the term comprises polypeptides derived from non-camelid sources, e.g. mouse or human, which have been "camelized", as previously described, e.g. in WO 08/101985 and WO 08/142164. The term "single domain antibody” encompasses immunoglobulin sequences of different origin, comprising mouse, rat, rabbit, donkey, human and camelid immunoglobulin sequences. It also includes fully human, humanized or chimeric immunoglobulin sequences. For example, it comprises camelid immunoglobulin sequences and humanized camelid immunoglobulin sequences, or camelized single variable domains, e.g. camelized dAb as described by Ward et al (see for example WO 94/04678 and Davies and Riechmann 1994, Febs
Lett. 339: 285 and 1996, Protein Engineering 9: 531).
The amino acid residues of a single variable domain (or conventional variable domain) are numbered according to the general numbering for VH domains given by Kabat et al. ("Sequence of proteins of immunological interest”, US Public Health Services, NIH Bethesda, Md. , Publication No. 91), as applied to VHH domains from Camelids by Riechmann and Muyldermans (1999, J. Immunol.
Methods; 231: 25-38; see for example Fig. 2 of said reference). According to this numbering, FR1 of a
VHH comprises the amino acid residues at positions 1-30, CDR1 of a VHH comprises the amino acid residues at positions 31-36, FR2 of a VHH comprises the amino acids at positions 36-49, CDR2 of a
VHH comprises the amino acid residues at positions 50-65, FR3 of a VHH comprises the amino acid residues at positions 66-94, CDR3 of a VHH comprises the amino acid residues at positions 95-102, and FR4 of a VHH comprises the amino acid residues at positions 103-113. In this respect, it should be noted that as is well known in the art for VH domains and for VHH domains the total number of amino acid residues in each of the CDRs may vary and may not correspond to the total number of amino acid residues indicated by the Kabat numbering (that is, one or more positions according to the Kabat numbering may not be occupied in the actual sequence, or the actual sequence may contain more amino acid residues than the number allowed for by the Kabat numbering). This means that, generally, the numbering according to Kabat may or may not correspond to the actual numbering of the amino acid residues in the actual sequence. Generally, however, it can be said that, according to the numbering of Kabat and irrespective of the number of amino acid residues in the CDRs, position 1 according to the Kabat numbering corresponds to the start of FR1 and vice versa, position 36 according to the Kabat numbering corresponds to the start of FR2 and vice versa, position 66 according to the
Kabat numbering corresponds to the start of FR3 and vice versa, and position 103 according to the
Kabat numbering corresponds to the start of FR4.
Alternative methods for numbering the amino acid residues of VH domains, which methods can also be applied in an analogous manner to VHH domains from Camelids, are the method described by
Chothia et al.(1989, Nature 342, 877-883), the so-called "AbM definition” and the so-called “contact definition”. However, in the present description, claims and figures, the numbering according to Kabat as applied to VHH domains by Riechmann and Muyldermans will be followed, unless indicated otherwise.
For a general description of single domain antibodies and the single variable domains (VHH domains) thereof, reference is inter alia made to the following references, which are mentioned as general background art: WO 94/04678, WO 95/04079, WO 96/34103, WO 94/25591, WO 99/37681,
WO 00/40968, WO 00/43507, WO 00/65057, WO 01/40310, WO 01/44301, EP 1134231, WO 02/48193, WO 97/49805, WO 01/21817, WO 03/035694, WO 03/054016, WO 03/055527 WO 03/050531, WO 01/90190, WO 03/025020; WO 04/041867, WO 04/041862, WO 04/041865, WO 04/041863 and WO 04/062551 and Hassanzadeh-Ghassabeh et al. (2013, Nanomedicine, 8(6):1013— 1026).
Generally, it should be noted that the term “single variable domain” or “single domain antibody” as used herein in its broadest sense is not limited to a specific biological source or to a specific method of preparation. For example, single variable domain as used in the invention can be obtained (1) by isolating the single variable domain of a naturally occurring single domain antibody; (2) by expression of a nucleotide sequence encoding a naturally occurring single variable domain; (3) by “humanization" (as described below) of a naturally occurring single variable domain or by expression of a nucleic acid encoding a such humanized single variable domain; (4) by "camelization” of a naturally occurring Vu domain from any animal species, in particular a species of mammal, such as from a human being, or by expression of a nucleic acid encoding such a camelized Vu domain; (5) using synthetic or semi- synthetic techniques for preparing proteins, polypeptides or other amino acid sequences; (6) by preparing a nucleic acid encoding a single variable domain using techniques for nucleic acid synthesis, followed by expression of the nucleic acid thus obtained; and/or (7) by any combination of the foregoing.
Suitable methods and techniques for performing the foregoing are state of the art and therefore known to the skilled person.
One particularly preferred class of single domain antibodies for use in the invention comprises single variable domains with an amino acid sequence that corresponds to the amino acid sequence of a naturally occurring single variable domain, but that has been "humanized", i.e. by replacing one or more amino acid residues in the amino acid sequence of said naturally occurring single variable domain sequence by one or more of the amino acid residues that occur at the corresponding position(s) in a Vn domain from a conventional 4-chain antibody from a human being. This can be performed in a manner known per se, which will be clear to the skilled person, for example on the basis of the prior art on humanization including e.g. Jones et al. (Nature 321:522-525, 1986); Riechmann et al., (Nature 332:323-329, 1988); Presta (Curr. Op. Struct. Biol. 2:593-596, 1992), Vaswani and Hamilton (Ann.
Allergy, Asthma and Immunol., 1:105-115 1998); Harris (Biochem. Soc. Transactions, 23:1035-1038, 1995); Hurle and Gross (Curr. Op. Biotech., 5:428-433, 1994), and specific prior art relating to humanization of VHHs such as e.g. Vincke et al. (2009, J. Biol. Chem. 284:3273-3284). Again, it should be noted that such humanized single variable domain of the invention can be obtained in any suitable manner known per se and thus are not strictly limited to polypeptides that have been obtained using a polypeptide that comprises a naturally occurring single variable domain as a starting material.
The terms “homology”, “sequence identity” and the like are used interchangeably herein.
Sequence identity is herein defined as a relationship between two or more amino acid (polypeptide or protein) sequences or two or more nucleic acid (polynucleotide) sequences, as determined by comparing the sequences. In the art, "identity" and “similarity” also means the degree of sequence relatedness between amino acid or nucleic acid sequences, as the case may be, as determined by the match between strings of such sequences. "Identity" and "similarity" can be readily calculated by known methods. “Sequence identity” and “sequence similarity” can be determined by alignment of two peptide or two nucleotide sequences using global or local alignment algorithms, depending on the length of the two sequences. Sequences of similar lengths are preferably aligned using global alignment algorithms (e.g. Needleman Wunsch) which align the sequences optimally over the entire length, while sequences of substantially different lengths are preferably aligned using local alignment algorithms (e.g. Smith
Waterman). Sequences may then be referred to as "substantially identical” or “essentially similar” when they (when optimally aligned by for example the programs GAP or BESTFIT using default parameters) share at least a certain minimal percentage of sequence identity (as defined below). GAP uses the
Needleman and Wunsch global alignment algorithm to align two sequences over their entire length (full length), maximizing the number of matches and minimizing the number of gaps. A global alignment is suitably used to determine sequence identity when the two sequences have similar lengths. Generally,
the GAP default parameters are used, with a gap creation penalty = 50 (nucleotides) / 8 (proteins) and gap extension penalty = 3 (nucleotides) / 2 (proteins). For nucleotides the default scoring matrix used is nwsgapdna and for proteins the default scoring matrix is Blosum62 (Henikoff & Henikoff, 1992, PNAS 89, 915-919). Sequence alignments and scores for percentage sequence identity may be determined using computer programs, such as the GCG Wisconsin Package, Version 10.3, available from Accelrys
Inc., 9685 Scranton Road, San Diego, CA 92121-3752 USA, or using open source software, such as the program “needle” (using the global Needleman Wunsch algorithm) or “water” (using the local Smith
Waterman algorithm) in EmbossWIN version 2.10.0, using the same parameters as for GAP above, or using the default settings (both for ‘needle’ and for ‘water’ and both for protein and for DNA alignments, the default Gap opening penalty is 10.0 and the default gap extension penalty is 0.5; default scoring matrices are Blossum62 for proteins and DNAFull for DNA). When sequences have a substantially different overall length, local alignments, such as those using the Smith Waterman algorithm, are preferred.
Alternatively, percentage similarity or identity may be determined by searching against public databases, using algorithms such as FASTA, BLAST, etc. Thus, the nucleic acid and protein sequences of the present invention can further be used as a “query sequence” to perform a search against public databases to, for example, identify other family members or related sequences. Such searches can be performed using the BLASTn and BLASTx programs (version 2.0) of Altschul, et al. (1990) J. Mol. Biol. 215:403—10. BLAST nucleotide searches can be performed with the NBLAST program, score = 100, wordlength = 12 to obtain nucleotide sequences homologous to oxidoreductase nucleic acid molecules of the invention. BLAST protein searches can be performed with the BLASTx program, score = 50, wordlength = 3 to obtain amino acid sequences homologous to protein molecules of the invention. To obtain gapped alignments for comparison purposes, Gapped BLAST can be utilized as described in
Altschul et al., (1997) Nucleic Acids Res. 25(17): 3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., BLASTx and BLASTN) can be used.
See the homepage of the National Center for Biotechnology Information at http://www.ncbi.nlm.nih.gov/.
A “variant” of a sequence, in particular a CDR sequence, typically comprises one or more amino acid substitutions. These variant CDRs are functional variants having an amino acid sequence different from the reference sequence but maintaining or exhibiting the same functional activity as the reference sequence of the single variable domain. The same functional activity refers to the binding specificity and/or affinity of the single variable domain consisting of one or more variant CDRs to its target molecule, i.e. a DOAC.
Optionally, in determining the degree of amino acid similarity, the skilled person may also take into account so-called "conservative" amino acid substitutions, as will be clear to the skilled person.
Conservative amino acid substitutions refer to the interchangeability of residues having similar side chains. Examples of classes of amino acid residues for conservative substitutions are given in the
Tables T1 below.
Table T1
Hydrophilic Uncharged Residues Ser (8), Thr (T), Asn (N), and ee
Aliphatic Uncharged Residues Gly (G), Ala (A), Val (V), Leu (L), ee
Alternative conservative amino acid residue substitution classes are displayed in the Table T2, here below.
Table T2
Ee ° or & 00000 os Ny le 000000 4 & 0 « 00 sr rrr 0M 000 ¢ = ry | ww
Alternative Physical and Functional Classifications of Amino Acid Residues are displayed in the
Table T3.
Table T3
Hydrophobic residues ACF GHILLMRT VW and em em
A “nucleic acid construct” or “nucleic acid vector” is herein understood to mean a man-made nucleic acid molecule resulting from the use of recombinant DNA technology. The term “nucleic acid construct” therefore does not include naturally occurring nucleic acid molecules although a nucleic acid construct may comprise (parts of) naturally occurring nucleic acid molecules. The terms “expression vector” or “expression construct" refer to nucleic acid molecules that are capable of effecting expression of a nucleotide sequence or gene in host cells or host organisms compatible with such expression vectors or constructs. These expression vectors typically include regulatory sequence elements that are operably linked to the nucleotide sequence to be expressed to effect its expression. Such regulatory elements usually at least include suitable transcription regulatory sequences and optionally, 3' transcription termination signals. Additional elements necessary or helpful in effecting expression may also be present, such as expression enhancer elements. The expression vector will be introduced into a suitable host cell and be able to effect expression of the coding sequence in an in vitro cell culture of the host cell. The expression vector will be suitable for replication in the host cell or organism of the invention whereas an expression construct will usually integrate in the host cell's genome for it to be maintained. Techniques for the introduction of nucleic acid into cells are well established in the art and any suitable technique may be employed, in accordance with the particular circumstances. For eukaryotic cells, suitable techniques may include calcium phosphate transfection, DEAE-Dextran, electroporation, liposome-mediated transfection and transduction using retrovirus or other virus, e.g. adenovirus, AAV, lentivirus or vaccinia. For microbial, e.g. bacterial, cells, suitable techniques may include calcium chloride transformation, electroporation and transfection using bacteriophage. The introduced nucleic acid may be on an extra-chromosomal vector within the cell or the nucleic acid may be integrated into the genome of the host cell. Integration may be promoted by inclusion of sequences within the nucleic acid or vector which promote recombination with the genome, in accordance with standard techniques. The introduction may be followed by expression of the nucleic acid to produce the encoded fusion protein. In some embodiments, host cells (which may include cells actually transformed although more likely the cells will be descendants of the transformed cells) may be cultured in vitro under conditions for expression of the nucleic acid, so that the encoded fusion protein polypeptide is produced, when an inducible promoter is used, expression may require the activation of the inducible promoter.
As used herein, the term “operably linked” refers to a linkage of polynucleotide elements in a functional relationship. A nucleic acid is “operably linked” when it is placed into a functional relationship with another nucleic acid sequence. For instance, a transcription regulatory sequence is operably linked to a coding sequence if it affects the transcription of the coding sequence. Operably linked means that the DNA sequences being linked are typically contiguous and, where necessary to join two protein encoding regions, contiguous and in reading frame.
The terms “protein” or “polypeptide” are used interchangeably and refer to molecules consisting of a chain of amino acids, without reference to a specific mode of action, size, 3-dimensional structure or origin.
The term "specificity" or “specific” or “specifically” refers to the number of different types of antigens or antigenic determinants to which a particular immunoglobulin sequence (antibody part of a polypeptide of the invention, such as an sdab, preferably a VHH domain), antigen-binding molecule or antigen-binding protein (such as a single domain antibody of the invention) can bind. The specificity of an antigen-binding molecule can be determined based on affinity and/or avidity. The affinity, represented by the equilibrium constant for the dissociation of an antigen with an antigen-binding protein (Ko), is a measure for the binding strength between an antigenic determinant and an antigen-binding site on the antigen-binding protein. Alternatively, the affinity can also be expressed as the affinity constant (Ka), which is 1/Kb. Affinity can be determined in a manner known per se, depending on the specific combination of antigen-binding protein and antigen of interest, here the fXa inhibiting DOACSs. Avidity is herein understood to refer to the strength of binding of a target molecule with multiple binding sites by a larger complex of binding agents, i.e. the strength of binding of multivalent binding.
Typically, a single domain antibody of the invention, such as a VHH, will bind the target molecule (i.e. a DOAC) with a dissociation constant (Kp) of about 107 to 10712 M or less, and preferably 10°8 to 1072 M or less, or even more preferably 10-'° or less and/or with a binding affinity of at least 107 M, preferably at least 108 M, more preferably at least 10° M, such as at least 10719, 1071, 10712 M or more.
Any Kb value greater than 10+ M (i.e. more than 100 uM) is generally considered to indicate non-specific binding. Preferably, a single domain antibody of the invention will bind to the target molecule with an affinity less than 10 nM or less than 5 nM, more preferably less than 1 nM, such as less than 500, 200, 100, 50, 10 or 5 pM. A variety of methods of measuring binding affinity are known in the art, any of which can be used for purposes of the present invention. Specific illustrative embodiments are described in the following.
A" Kp "or" Kp value" can be measured by using an ELISA as described in the Examples herein or by using surface plasmon resonance assays for example using a BlAcore™-T200 (BlAcore, Inc.,
Piscataway, NJ) at 25°C with immobilized antigen CMS chips at ~10 - 50 response units (RU). Briefly, carboxymethylated dextran biosensor chips (CMS, BIAcore Inc.) are activated with N-ethyl-N'-(3- dimethylaminopropyl)-carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions. Antigen (DOAC) is diluted with 10 mM sodium acetate, pH 4.8, into 5 pg/ml (~0.2 uM) before injection at a flow rate of 5 ul/minute to achieve approximately 10 response units (RU) of coupled protein. Following the injection of antigen, 1 M ethanolamine is injected to block unreacted groups. For kinetics measurements, two-fold serial dilutions of the polypeptide of the invention (0.78 nM to 500 nM) is injected in PBS with 0.05% Tween 20 (PBST) at 25°C at a flow rate of approximately 25 pl/min, Association rates (Kon) and dissociation rates (ker) are calculated using a simple one-to-one
Langmuir binding model (BlAcore Evaluation Software version 3.2) by simultaneous fitting the association and dissociation sensorgram. The equilibrium dissociation constant (Kp) is calculated as the ratio kof/kon. See, e.g., Chen, Y., et al., (1999) J. Mol Biol 293:865-881.
An "on-rate" or "rate of association" or "association rate" or "kon" according to this invention can also be determined with the same surface plasmon resonance technique described above using for example a BlIAcore™-T200 (BlAcore, Inc., Piscataway, NJ) as described above.
As used herein, with "an effective amount” is meant the amount of an agent required to ameliorate the symptoms of a disease relative to an untreated patient, here for example bleeding, major bleeding or uncontrolled bleeding. The effective amount of active agent(s) used to practice the present invention (here, the polypeptide of the invention) for therapeutic treatment of, for example bleeding, varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the attending physician or veterinarian will decide the appropriate amount and dosage regimen. Such amount is referred to as an "effective" amount, which may be determined as milligram active ingredient per kilogram body weight (mg/kg). Thus, in connection with the administration of a drug which, in the context of the current disclosure, is "effective against" a disease or condition indicates that administration in a clinically appropriate manner results in a beneficial effect for at least a statistically significant fraction of patients, such as an improvement of symptoms, a cure, a reduction in at least one disease sign or symptom, extension of life, improvement in quality of life, or other effect generally recognized as positive by medical doctors familiar with treating the particular type of disease or condition.
The use of a substance as a medicament as described in this document can also be interpreted as the use of said substance in the manufacture of a medicament. Similarly, whenever a substance is used for treatment or as a medicament, it can also be used for the manufacture of a medicament for treatment. Products for use as a medicament described herein can be used in methods of treatments, wherein such methods of treatment comprise the administration of the product for use.
The phrases "pharmaceutical or pharmacologically acceptable" refers to molecular entities and compositions that do not produce or produce acceptable adverse, allergic or other untoward reaction when administered to an animal, such as, for example, a human, as appropriate. Whether certain adverse effects are acceptable is determined based on the severity of the disease. The preparation of a pharmaceutical composition that contains at least one polypeptide comprising e.g. a single domain antibody of the invention or additional active ingredient will be known to those of skill in the art in light of the present disclosure, as exemplified by Remington: The Science and Practice of Pharmacy” (Ed.
Adeboye Adejare, 23rd edition, 2020, Elsevier Inc.), incorporated herein by reference. Moreover, for animal (e.g., human) administration, it will be understood that preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biological Standards.
As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drugs, drug stabilizers, gels, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, such like materials and combinations thereof, as would be known to one of ordinary skill in the art (see, for example, Remington: The Science and Practice of Pharmacy” (Ed. Adeboye Adejare, 23rd edition, 2020, Elsevier Inc.), incorporated herein by reference). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated.
As used herein, the terms “administering” or “administration” means to provide a polypeptide of the invention or a pharmaceutical composition comprising such polypeptide to a subject in a manner that is physiologically and/or (e.g., and) pharmacologically useful {e.g., to treat a condition in the subject).
The term “comprising”, used in the claims, should not be interpreted as being restricted to for example the elements or the method steps or the constituents of a compositions listed thereafter; it does not exclude other elements or method steps or constituents in a certain composition. It needs to be interpreted as specifying the presence of the stated features, integers, (method) steps or components as referred to, but does not preclude the presence or addition of one or more other features, integers, steps or components, or groups thereof. Thus, the scope of the expression “a method comprising steps A and B” should not be limited to a method consisting only of steps A and B, rather with respect to the present invention, the only enumerated steps of the method are A and B, and further the claim should be interpreted as including equivalents of those method steps. Thus, the scope of the expression “a composition comprising components A and B” should not be limited to a composition consisting only of components A and B, rather with respect to the present invention, the only enumerated components of the composition are A and B, and further the claim should be interpreted as including equivalents of those components.
The terms “activated factor X”, “factor Xa”, “FXa”, “Xa” and “fXa” have their regular scientific meaning and all refer to the same serine endopeptidase: factor X that is activated upon hydrolysis by serine proteases in for example one of the two principal pathways of the initiation of blood coagulation, being the ‘extrinsic pathway’ (by factor VII/VIla in complex with Tissue Factor) and the ‘intrinsic pathway’ (by factor |Xa in complex with its cofactor factor Villa), or via for example a snake venom, Activated factor X can cleave prothrombin at two cleavage sites in the prothrombin amino-acid sequence, providing active thrombin.
In addition, reference to an element or a component by the indefinite article "a" or "an" does not exclude the possibility that more than one of the element or component are present, unless the context clearly requires that there is one and only one of the elements or components. The indefinite article "a" or "an" thus usually means "at least one".
The use of terms in brackets in the text, with the exception of chemical and/or mathematical formulae, usually means that the term within brackets specifies a possible option or a possible meaning and should thus not be considered limiting.
The embodiments as described herein can operate in combination and cooperation, unless specified otherwise. Furthermore, the various embodiments, although referred to as “preferred” or “e.g.” or “for example” or “in particular” and the like are to be construed as exemplary manners in which the disclosed herein concepts may be implemented rather than as limiting.
Any reference to nucleotide sequences or amino acid sequences accessible in public sequence databases herein refers to the version of the sequence entry as available on the filing date of this document.
The present invention has been described above and below with reference to a number of exemplary embodiments, including the embodiments as shown in the drawings. Modifications and alternative implementations of some molecules or elements are possible, and are included in the scope of protection as defined in the appended claims.
Figure 1A, 1B, 1C, 1D: Inhibition of Edoxaban (25 nM (A), 50 nM (B), 400 nM (C), 1000 nM (D))} by VHH (SEQ ID NO: 52; edo_1) binding specifically to edoxaban in a chromogenic STA® Liquid Anti-Xa (LAX) test;
Figure 2: Normalization of thrombin generation at 300 nM edoxaban with various concentrations of VHH (0, 300, 600, 1200, 2400 nM edo_1) binding specifically to edoxaban;
Figure 3: Inhibition of 300 nM edoxaban with various concentrations of VHH (edo_1) binding specifically to edoxaban observed via the decrease in coagulation lag time;
Figure 4: Inhibition of anti-coagulant activity of 300 nM edoxaban with various concentrations of VHH (edo_1) binding specifically to edoxaban observed via the increase in Peak Thrombin values;
Figure 5: Normalization of thrombin generation at 300 nM edoxaban with various concentrations of VHH (SEQ ID NO: 98; edo_22, bispecific) binding specifically to both edoxaban and human serum albumin;
Figure 6: Inhibition of anti-coagulant activity of 300 nM edoxaban with various concentrations of VHH (edo 22, bispecific) binding specifically to both edoxaban and albumin, observed via the decrease in lag time;
Figure 7: Inhibition of anti-coagulant activity of 300 nM edoxaban with various concentrations of VHH (edo_22, bispecific) binding specifically to both edoxaban and albumin observed via the increase in
Peak Thrombin values;
Figure 8A, 8B: Thrombin generation lag time compensation in plasma of patients who are on treatment with edoxaban, by various concentrations of VHHs (edo_1 and edo_22, respectively) binding specifically to edoxaban (edo_1, A) and both edoxaban and albumin (edo_22, B), respectively;
Figure 9A, 9B: Thrombin generation Peak (TG-Peak) thrombin values compensation in plasma of patients who are on treatment with edoxaban by various concentrations of VHHs (edo_1 and edo_22, respectively) binding specifically to edoxaban (A) and both edoxaban and albumin (B), respectively;
Figure 10: Inhibition of apixaban (1088 nM) by VHH (SEQ ID NO: 64; api_1) binding specifically to apixaban and by VHH (SEQ ID NO: 100; api_31) binding specifically to both apixaban and albumin in a chromogenic STA® Liquid Anti-Xa (LAX) test;
Figure 11: Normalization of thrombin generation in NPP plasma at a concentration of 400 nM apixaban with various concentrations of VHH (0, 400, 800, 1600, 3200 nM api_1) binding specifically to apixaban;
Figure 12: Inhibition of 400 nM apixaban in NPP plasma with various concentrations of VHH (api_1) binding specifically to apixaban, assessed as the decrease in lag time for thrombin generation;
Figure 13: Inhibition of 400 nM apixaban in NPP plasma with various concentrations of VHH (api_1) binding specifically to apixaban, assessed as the increase in Peak Thrombin values;
Figure 14: Normalization of thrombin generation at 400 nM apixaban in NPP plasma, with various concentrations of VHH (SEQ ID NO: 100; api_31, bispecific) binding specifically to both apixaban and human serum albumin;
Figure 15: Inhibition of 400 nM apixaban in NPP plasma with various concentrations of VHH (api_31, bispecific) binding specifically to both apixaban and albumin, as assessed by measuring the decrease in lag time for thrombin generation;
Figure 16: Inhibition of 400 nM apixaban in NPP plasma with various concentrations of VHH (api_31, bispecific) binding specifically to both apixaban and albumin, as assessed by determining the increase in Peak Thrombin values;
Figure 17 A, B: Inhibition of apixaban with a single dose (‘spike’) of 1600 nM of VHH (api_1) binding specifically to apixaban in plasma of patients who are on treatment with apixaban. Fig. 17A and Fig. 17B are clinical measurements of factor Xa activity (calculated to apixaban concentrations) in plasma of patients who are treated with apixaban. In those patient plasma samples api_1 was added to test the apixaban-neutralizing activity of api_1 in the patients who were on apixaban treatment;
Fig, 18: Binding of apixaban inhibiting VHH domain with SEQ ID NO: 64 (api_1) to apixaban as assessed in a surface plasmon resonance (SPR) measurement (replicate 1 of 3 is shown). The SPR measurements are done to test the binding characteristics of api_1 to an albumin-apixaban coated surface;
Figure 19: Normalization of thrombin generation in NPP plasma containing 600 nM rivaroxaban, with various concentrations of VHH (SEQ ID NO: 79; riva_1) binding specifically to rivaroxaban;
Figure 20: Inhibition of anti-coagulant activity of 600 nM rivaroxaban in NPP plasma with various concentrations of VHH (riva_1) binding specifically to rivaroxaban, as assessed by measuring the decrease in lag time for thrombin generation;
Figure 21: Inhibition of anti-coagulant activity of 600 nM rivaroxaban in NPP plasma with various concentrations of VHH (riva_1) binding specifically to rivaroxaban, determined by measuring the increase in Peak Thrombin values.
Figure 22: Results of the Chromogenic STA® liquid anti-Xa (LAX) test (diagnostica stago) (A, B). The reference measurement is human normal pooled plasma (NPP) without added apixaban and without added api_32 (two linked apixaban binding VHH domains, with SEQ ID NO: 101) (referred to as “plasma™) (A, B). All the other four measurements contain 218 nM apixaban (A) and 1088 nM apixaban (B) with O- (no VHH added), 4-, 8-, and 16-fold molar excess concentrations of added api_32.
In a first aspect of the invention, the invention provides a polypeptide comprising or consisting of at least one antibody, such as a single domain antibody (‘'sdab’), that specifically binds a DOAC which is capable of inhibiting activated factor X (fXa). Herewith, such polypeptide acts as an antidote for such fXa inhibiting DOAC. Typically, binding to the DOAC means that the antibody binds to a specific DOAC only, without displaying any cross-reactivity to other DOAC:s. Preferably, the polypeptide comprising the antibody binds to any pharmaceutically acceptable form of a DOAC. Such specific binding of the polypeptide comprising the antibody to its specific DOAC allows the polypeptide to inhibit or neutralize the DOAC as is further described hereinbelow. This way, the polypeptide comprising the antibody such as a sdab acts as a specific antidote for the selected specific DOAC.
A polypeptide comprising or consisting of an antibody of the invention is for example a polypeptide comprising or consisting of at least one sdab, wherein such sdab may be obtained from a (llama) heavy chain-only antibody, wherein the complete antigen binding site is present on, and formed by, the sdab.
In one embodiment, the polypeptide comprises or consists of an antibody wherein the antibody comprises or consists of at least one sdab that specifically binds the DOAC and that is a variable VHH domain (VHH' or ‘VHH domain’) obtained or obtainable from a camelid, preferably llama, heavy chain antibody or a variable IgNAR domain obtained or obtainable from a shark single domain antibody.
Preferably the at least one sdab comprised by or being the polypeptide is a VHH or at least one VHH obtained or obtainable from a camelid heavy chain antibody, more preferably from a llama heavy chain antibody.
An aspect of the invention relates to a polypeptide comprising or consisting of at least one antibody which antibody specifically binds to a first direct oral anticoagulant (‘DOAC") which DOAC is an inhibitor of activated factor X (“fXa").
The first DOAC for inhibiting fXa is a DOAC selected from the known DOACs capable of silencing or inhibiting the activity of fXa: apixaban, rivaroxaban, edoxaban and betrixaban. Preferably, the polypeptide specifically binds selectively to a DOAC selected from apixaban, rivaroxaban and edoxaban. That is to say, the invention provides a polypeptide comprising or consisting of an antibody that specifically binds to either apixaban, or rivaroxaban, or edoxaban, or betrixaban, preferably, to either apixaban, or rivaroxaban, or edoxaban. Such specificity for binding of the polypeptide to a selected specific DOAC provides several benefits. For example, a patient in need of treatment with an antidote for a specific DOAC which is previously administered to the patient, can be selectively treated with only the antidote capable of neutralizing or inhibiting the effect of the DOAC with which the patient is treated. The antidote is the polypeptide of the invention capable of specifically inhibiting the fXa inhibiting activity of the DOAC with which the patient is treated. After the patient has been treated with the polypeptide specific for the DOAC, if deemed necessary, the patient can start treatment with a second DOAC capable of inhibiting fXa, at any time after the polypeptide specific for inhibiting the first
DOAC has been administered. For example, to a patient to whom a first polypeptide of the invention specific for inhibiting a first DOAC capable of inhibiting fXa is administered before said patient is subjected to a surgical procedure, a second DOAC capable of inhibiting fXa different from the first
DOAC can be administered, resulting in (restored) anti-coagulant activity in the patient by presence of the second DOAC. The first polypeptide specific for the first DOAC (antidote for the first DOAC) does not act as an antidote for the second DOAC and thus does not inhibit the fXa inhibiting activity of the second DOAC. Therewith, the patient in general need of anti-coagulant therapy can undergo the surgical procedure after the first polypeptide is administered, temporarily blocking the anti-coagulant therapeutic effect of a DOAC, and restoring anti-coagulant therapy by administering yet a second DOAC different from the first DOAC and not capable of binding to the polypeptide that was administered to the patient before surgery.
In an embodiment of the invention the at least one antibody comprised by the polypeptide comprises or consists of: an IgG, IgG1, IgG2, IgG2A, IgG2B, IgG2C, IgG3, IgG4, IgA1, IgA2, IgD, IgM or IgE or a binding fragment thereof or a binding domain thereof, or a Fab fragment, a F(ab’) fragment, a F(ab')2 fragment, an Fv fragment or an scFv fragment, or a diabody, or a single domain antibody (“sdab”), for example an antigen binding fragment of heavy chain only antibody (variable domain of heavy chain only antibody,
VHH) and/or a sdab derived from a camelid antibody or a shark antibody. Preferred is a polypeptide comprising at least one antibody, wherein the antibody comprises or consists of an sdab, preferably a
VHH. Preferred is a VHH derived from a llama, e.g. obtained from a llama antibody or heavy-chain only antibody, preferably a heavy-chain only antibody, after immunization of the llama with a selected DOAC to which the VHH can specifically bind. Of course, such VHH can also be obtained after immunization of a different camelid species. Although the antibody can be any one of the here above listed antibodies or a binding domain thereof or a binding fragment thereof, the antibody being or comprising at least one
VHH is preferred. VHH domains have several advantages. Also due to their size, (scaled up) production and purification resulting in clinical-grade amounts suitable for use as a medicament is feasible. In addition, a VHH domain is relatively stable and resistant to denaturation under physiological conditions such as the conditions in regular pharmaceutical compositions and in the circulation. Moreover, due to their size VHH domains can be rapidly cleared from the circulation. This latter aspect can be beneficial when a patient who is on DOAC treatment should be subjected to for example emergency surgery procedures. Administering a polypeptide of the invention that can bind and inhibit the fXa inhibiting activity of the first DOAC, to such a patient in need of an antidote to reverse anti-coagulant activity of the DOAC, will not only result in (rapid) neutralization of the anti-coagulant activity of the DOAC in the circulation of the patient, but will also result in rapid clearance of the VHH-comprising polypeptide with the DOAC bound thereto, therewith effectively clearing the circulation in relatively short time from the
DOAC. A further advantage of rapid clearance of (intravenously) administered polypeptide of the invention comprising or consisting of a VHH to a patient is that after such surgical procedure has been conducted on the patient, anti-coagulant treatment with the same first DOAC can start rapidly, without the necessity to wait for a prolonged time before the administered VHH is cleared from the circulation.
To the surprise of the inventors, they naticed that a polypeptide of the invention is capable of neutralizing and reversing DOAC activity in patient plasma of a patient to whom the DOAC is previously administered, within 10 minutes after the polypeptide is contacted with the plasma. If the polypeptide is subsequently rapidly cleared by the kidney, the patient is free of free and polypeptide-scavenged DOAC within a short period after the start of the polypeptide administration. For example, when the polypeptide consists of a single VHH or of two VHH domains in tandem, the molecular size is about 15 kDa or 30 kDa, being a size which is small enough for fast uptake and clearance from the body by the renal system.
In an embodiment of the invention the at least one antibody comprises or consists of at least one sdab that specifically binds to the first DOAC. Preferred is a polypeptide that comprises or consists of at least one sdab, such as 1-9 sdab’s. Preferably, the sdab(s) is/are VHH. The at least one sdab is an antidote that specifically binds a single DOAC that is capable of inhibiting fXa. In embodiments, the polypeptide comprises or consists of more than one sdab such as VHH, capable of binding to more than one DOAC capable of inhibiting fXa, such as two, three or four different DOACS, for example selected from rivaroxaban, apixaban, edoxaban, betrixaban, preferably from rivaroxaban, apixaban and edoxaban.
In an embodiment of the invention the at least one antibody is a VHH, preferably a llama VHH. Also suitable are VHH derived from a different camelid species such as camel or alpaca. The inventors surprisingly found that upon immunization of llama’s with the separate small-molecule DOACs rivaroxaban, apixaban and edoxaban, when conjugated with albumin, in these llama’s immune responses specific for the selected free unconjugated small-molecule DOACs were elicited. That is to say, llama’s immunized with rivaroxaban comprised heavy-chain only antibodies specific for free rivaroxaban (no cross-reaction and binding to albumin) while no binding to any of the other DOACs capable of inhibiting fXa was observed. The same result was achieved with the llama’s immunized with apixaban, or with edoxaban. Moreover, from llama’s immunized with rivaroxaban at least four different clusters comprising related VHH sequences could be established (see Table D). The related VHH amino-acid sequences within a cluster comprise one or a few amino-acid substitutions in the constant regions and/or in CDR(s). For Ilama's immunized with apixaban, two different clusters of related VHH amino-acid sequences were obtained (Table C), for llama’s immunized with edoxaban, at least three different clusters of highly homologous VHH were obtained (Table B). Within clusters, affinity of the VHH for binding to the specific DOAC is very similar and the same is determined for VHH in different clusters specific for the same DOAC. As said, the VHHs bind specifically to a selected single DOAC, without any binding specificity for a further DOAC. The inventors thus established a series of VHHs all capable of specific binding and inhibition of a single selected DOAC. VHHs binding to edoxaban inhibit (reverse) the fXa inhibiting activity of edoxaban, while leaving the fXa inhibiting activity of apixaban, rivaroxaban and betrixaban unaltered. Similar results are established for the VHHs binding to apixaban or the VHHs binding to rivaroxaban. Herewith, the shortcoming of Andexanet alfa, being that multiple different
DOACs capable of inhibiting fXa all bind to Andexanet alfa, is overcome. When a patient is treated with a certain DOAC, and when said patient is need of reversal of anti-coagulant therapy based on the
DOAC, administering the polypeptide of the invention comprising a VHH that specifically binds and inhibits said DOAC to the patient effectively reverses the anti-coagulant therapy.
Preferably, the polypeptide of the invention comprises or consists of at least one antibody, wherein the antibody preferably is a sdab. Such at least one sdab preferably is a VHH, such as one or more than one VHH. According to the invention, a single VHH specifically binds to a single type of
DOAC capable of inhibiting fXa, such as any one of rivaroxaban, edoxaban, apixaban and betrixaban, preferably any one of rivaroxaban, edoxaban and apixaban. Combining more than one VHH that all separately specifically can bind to a specific selected DOAC molecule such as any one of rivaroxaban, edoxaban and apixaban, results in a multivalent polypeptide of the invention which is monospecific for a selected DOAC. Such multivalent polypeptide may comprise or consist of a string of linearly linked
VHH wherein the more than one VHH are all the same VHH. For example, the polypeptide can comprise any number between and including 2 to 10 VHH of the same clone (having the same amino-acid sequence). Typically, the consecutively linked VHH in the polypeptide are linked via an amino-acid linker sequence. Alternatively, the more than one VHH comprised by the polypeptide all specifically bind to the same DOAC, wherein at least two of the more than one VHH have a different amino-acid sequence.
For example, referring to Table A, displaying various clusters of VHH with amino-acid sequences with high sequence homology in an indicated cluster, the polypeptide specifically binds to edoxaban and comprises at least one copy each of one or more varying VHH with an amino acid sequence selected from cluster 1 and/or at least one copy each of one or more varying VHH with an amino acid sequence selected from cluster 2 and/or at least one copy each one or more varying VHH with an amino acid sequence selected from cluster 3. For example, again referring to Table A, the polypeptide specifically binds to apixaban and comprises at least one copy each of one or more varying VHH with an amino acid sequence selected from cluster 1 and/or one or more copies of the VHH with an amino acid sequence in cluster 2. For example, again referring to Table A, the polypeptide specifically binds to rivaroxaban and comprises at least one copy each of one or more varying VHH with an amino acid sequence selected from cluster 1 and/or at least one copy each of one or more varying VHH with an amino acid sequence selected from cluster 2 and/or at least one copy each of one or more varying VHH with an amino acid sequence selected from cluster 3, and/or at least one copy each of one or more varying VHH with an amino acid sequence selected from cluster 4. For example, such multivalent mono- specific polypeptide comprises two to ten VHH with an identical amino-acid sequence or with at least two different amino-acid sequences, such as two to five different amino-acid sequences. The advantage of such multivalent (mono-specific) polypeptide is its capability to bind and capture and therewith neutralize or inhibit multiple DOAC molecules, with the maximum number of DOAC molecules of which the activity can be reversed is equal to the number of VHH domains in the polypeptide. Such polypeptide therewith displays a higher DOAC inhibitory activity on a molar basis compared to a polypeptide comprising less VHH domains specifically binding to the same DOAC such as a polypeptide comprising a single VHH. In addition, such a multivalent polypeptide has a longer plasma half-life when administered to a patient, due to its larger size and/or due to its larger molecular weight. Specially, when the number of VHH copies or number of VHH domains comprised by the polypeptide results in a molecular mass and size of the polypeptide that prevents rapid clearance of the polypeptide from the circulation in the kidney, can improve the DOAC inhibiting efficacy of the polypeptide due to its extended half life compared to a polypeptide of the invention comprising less VHH domains. The polypeptide of the invention allows for fine-tuning between maximally desired DOAC binding capacity in terms of for example the number of DOAC molecules that can be bound by a single polypeptide molecule, and desired plasma half-life once the polypeptide is administered to a patient.
In an embodiment of the invention the polypeptide comprises or consists of at least two sdab’s, preferably conjugated to each other via an amino-acid linker sequence such as linker with amino-acid sequence AAGGGGSGGGGSAAA (SEQ ID NO: 104). Preferably, the at least two sdab's are VHH, such as the VHHs displayed in Table A and capable of specifically binding to a DOAC selected from edoxaban, apixaban and rivaroxaban. For example, the at least two sdab's are capable of binding at least one, or two, or three, or four DOACs selected from edoxaban, apixaban, betrixaban and rivaroxaban, preferably, the at least two sdab’s are capable of binding at least one, or two, or three
DOACs selected from edoxaban, apixaban and rivaroxaban. In certain embodiments, the polypeptide comprises at least two VHHs wherein each VHH is capable of binding to the same one DOAC, preferably any one of edoxaban, apixaban, betrixaban and rivaroxaban, more preferably anyu one of edoxaban, apixaban and rivaroxaban.
In an embodiment of the invention the at least two sdab's specifically bind to the same first DOAC, or wherein a first sdab of the at least two sdab’s specifically binds to the first DOAC and a second sdab of the at least two sdab’s specifically binds to a second DOAC which is different from the first DOAC and wherein the second DOAC is an inhibitor of fXa.
Occasionally, a patient can be presented who is in urgent need of a surgical procedure, and/or who is suffering from bleeding or major bleeding for example, for which patient it is not known whether the patient has been previously administered with an anti-coagulant therapeutic substance such as a DOAC capable of inhibiting fXa. If for example in such situation it is known that the patient has been treated with a DOAC but the specific DOAC capable of inhibiting fXa is not known, rapid reversal of the anti- coagulant activity of such DOAC can be required (life saving). In order to be able to do so, a polypeptide comprising at least one VHH specifically binding to a first DOAC (for example edoxaban) and comprising at least one VHH specifically binding to a second DOAC, for example selected from apixaban, rivaroxaban and betrixaban, preferably apixaban and rivaroxaban, can be administered to the patient in such emergency situation. Such a polypeptide can comprise any combination of at least two VHH, wherein the at least one first VHH specifically binds to a first DOAC and the second of the at least one VHH specifically binds to a second DOAC, the first and second DOAC preferably selected from edoxaban, apixaban, rivaroxaban and betrixaban, more preferably, edoxaban, apixaban, rivaroxaban. If a patient is for example treated with either edoxaban or apixaban, being it unknown which of the two DOACSs is actually administered to the patient, a polypeptide comprising at least one of a first sdab specifically binding to edoxaban and comprising at least one of a second sdab specifically binding to apixaban, can be administered to the patient in order to be sure that the DOAC present in the circulation of the patient is efficiently inhibited by the polypeptide. For example, may anti-coagulant therapy again be required for example after a surgical procedure or when (excessive) bleeding is halted,
in such circumstance it is possible to administer yet a third DOAC different from the first DOAC and second DOAC, on short notice (no waiting time required for clearance of the administered multi-specific polypeptide from the circulation of the patient). Of course, it will be appreciated by the skilled person that also the same DOAC (first or second DOAC) can again be administered if required. The dose of the DOAC can for example be temporarily higher than the dose previously administered in order to saturate any circulating polypeptide, and/or the first or second DOAC can again be administered to the patient, starting after a waiting time sufficiently long for clearance of the polypeptide from the circulation.
This way, such multi-specific polypeptide, which polypeptide can also be multivalent for one or two of the different DOACS, as here above described, provides for flexibility in reversing the known or unknown
DOAC which was (likely) previously administered to a patient, and provides for flexibility in time to restart anti-coagulant therapy and/or flexibility in which anti-coagulant treatment is (re-)started. Such flexibility is for example absent for Andexanet alfa, which non-specifically binds to any DOAC capable of inhibiting fXa, such as edoxaban, apixaban and rivaroxaban. Once Andexanet alfa is administered to a patient, any fXa binding and inhibiting DOAC will be captured and inhibited upon binding to Andexanet alfa.
In an embodiment of the invention the at least two sdab’s are three, four, five, six, seven, eight or nine sdab’s, wherein - the three, four, five, six, seven, eight or nine sdab’s specifically bind to the same first DOAC, or - at least one sdab of the three to nine sdab’s specifically binds to the first DOAC and at least one of the three to nine sdab's specifically binds to a second DOAC, or - at least one sdab of the three to nine sdab’s specifically binds to the first DOAC and at least one of the three to nine sdab’s specifically binds to a second DOAC and at least one of the three to nine sdab’s specifically binds to a third DOAC, wherein the second DOAC is different from the first DOAC and the third DOAC is different from the first and second DOAC, and wherein the second DOAC and the third DOAC are inhibitors of fXa. Preferably, the sdab’s are VHHs. More preferably, the sdab’s are VHHs selected from the VHH sequences with SEQ ID NOs as listed in Table A.
As detailed here above before, for the polypeptide comprising a first sdab capable of binding and inhibiting a first DOAC and comprising a second sdab capable of binding and inhibiting a second DOAC different from the first DOAC, the polypeptide can also comprise yet a third sdab capable of binding to a third DOAC different from both the first and second DOAC. Advantages are rather the same: during circumstances in which it is not known which DOAC has been administered to a patient who is in need of immediate reversal of such anti-coagulant therapy, a multispecific polypeptide specifically binding to more than one different DOAC, such as two or three different DOACs, can be administered to the patient in order to maximize the chance that anti-coagulation therapy relating to previously administered DOAC capable of inhibiting fXa, is successfully and fast reversed and/or inhibited. For example, the polypeptide comprises at least one VHH specifically binding to edoxaban, at least one VHH specifically binding to apixaban and at least one VHH specifically binding to rivaroxaban. The multispecific polypeptide is for example multivalent for one, two or three of the three different DOACs: the polypeptide can comprise more than one copy of a VHH specifically binding to a DOAC, and/or for a specific DOAC the polypeptide comprises more than one different sdab's, preferably VHHs, e.g. selected from Table
A.
In an embodiment of the invention - the polypeptide comprises an albumin-binding antibody, preferably capable of specifically binding to human serum albumin, or - the polypeptide comprises albumin, preferably human serum albumin, bound to the at least one sdab or to the at least two sdab('s), preferably covalently bound to the at least one sdab or to the at least two sdab('s), optionally bound to the at least one sdab or to the at least two sdab('s) via a linker such as an amino-acid linker sequence.
Preferably, the sdab(s) is/are VHH domains. The albumin binding antibody is for example an sdab, such as a VHH specifically binding to human serum albumin. Of course, multiple copies of such VHH can be comprised by the polypeptide, and/or different VHHs all capable of binding to human serum albumin are comprised by the polypeptide. It is preferred that the polypeptide comprises a single copy of a VHH that specially binds to human serum albumin. The advantage of such a polypeptide is its increased half-life in the circulation of a patient once the polypeptide is administered to said patient. It is preferred that the albumin or the albumin-binding antibody, such as a VHH, are linked to the antibody comprised by the polypeptide via an amino-acid linker sequence. The antibody preferably is a single
VHH or are multiple VHHs, such as two or more VHHs in a multivalent polypeptide of the invention.
In an embodiment of the invention the albumin-binding antibody comprises or consists of an albumin-binding sdab, preferably an albumin binding VHH, comprising a combination of complementarity determining region (“CDR”) 1, CDR 2 and CDR 3 amino-acid sequences GYISDAYY (SEQ ID NO: 105}, ITHGTNTY (SEQ ID NO: 106) and AVLETRSYSFRY (SEQ ID NO: 107), or CDR 1 amino-acid sequences, CDR 2 amino-acid sequences and CDR 3 amino-acid sequences that have at least 95% sequence identity with CDR 1-, CDR 2- and CDR 3 amino-acid sequences with SEQ ID NO: 105, SEQ ID NO 106 and SEQ ID NO: 107, wherein preferably the albumin binding sdab is conjugated with the at least one antibody comprised by the polypeptide via an amino-acid linker sequence such as linker with amino-acid sequence AAGGGGSGGGGSAAA (SEQ ID NO: 104).
In an embodiment of the invention the first DOAC, second DOAC and third DOAC is a small molecule therapeutic, preferably with a molecular weight of 1200 Dalton (Da) or less and/or preferably with a molecular weight of 250 Da or more, more preferably with a molecular weight of 400 Da— 600
Da. Such DOAC is capable of inhibiting fXa.
In an embodiment of the invention any one or more of the first DOAC, second DOAC and third
DOAC(s) is/are selected from the group comprising or consisting of apixaban, edoxaban, rivaroxaban and betrixaban, preferably apixaban, edoxaban and rivaroxaban. Examples of polypeptides capable of binding and inhibiting such a first, second and third DOAC are tabulated in Table A, for edoxaban, apixaban and rivaroxaban. Surprisingly, the inventors succeeded in providing multiple different VHH series (clusters comprising homologous VHHSs) for each of these three DOACs. Such different VHHs are capable of specifically binding and inhibiting a selected DOAC therewith reversing the anti- coagulant activity of such DOAC. Thus, the inventors provide for the first time a series of VHH that in combination can reverse the anti-coagulant activity of the DOACs apixaban, edoxaban, rivaroxaban.
Either, a VHH can be selected to inhibit specifically a selected DOAC, or multiple VHHs can be selected to inhibit specifically more than one selected DOAC, either concomitantly or sequentially.
In an embodiment of the invention the polypeptide comprises or consists of a sdab, wherein said at least one sdab comprises a combination of CDR 1-, CDR 2- and CDR 3 amino-acid sequences, or wherein the at least two sdab’s, preferably any one of two to nine sdab’s comprise(s) the combination of CDR 1-, CDR 2- and CDR 3 amino-acid sequences, wherein said combination of CDR 1, CDR 2 and
CDR 3 specifically binds to edoxaban and wherein the CDR 1 is selected from amino-acid sequences GFAFDDYA (SEQ ID NO: 1) and GFTFDDYA (SEQ ID NO: 2), the CDR 2 is amino-acid sequence ISSSDGSTY (SEQ ID NO: 3), and the CDR 3 is selected from amino-acid sequences AAVPRTMYSRWGCGVRPYYYGMDY (SEQ ID NO: 4), AAVPRTMNSRWGCGVRPYYYGMDY (SEQ ID NO: 5) and
AAVPRTMYSRWGCGVRPYYHGMDY (SEQ ID NO: 6); and/or the CDR 1 is amino-acid sequence GRTFSINV (SEQ ID NO: 7), the CDR 2 is selected from amino-acid sequences IWWSGGASQ (SEQ ID NO: 8) and
TWWSGGASQ (SEQ ID NO: 9}, and the CDR 3 is selected from amino-acid sequences AAGPMFSMDYRRVNY (SEQ ID NO: 10),
AAGPVFSMDYRRVNY (SEQ ID NO: 11), AAGPMFSMDYTRVNY (SEQ ID NO: 12) and
AAGPMFSMDYRRVNH (SEQ ID NO: 13); and/or the CDR 1 is amino-acid sequence GRTFSSYH (SEQ ID NO: 14), the CDR 2 is selected from amino-acid sequences ITRGGGVTY (SEQ ID NO: 15) and
IARGGGVTY (SEQ ID NO: 16), and the CDR 3 is amino-acid sequence AADAIWNQVRWMETKYTY (SEQ ID NO: 17); and/or specifically binds to apixaban and wherein the CDR 1 is selected from amino-acid sequences GRIVSIYS (SEQ ID NO: 18), GRIVGIYS (SEQ ID NO: 19) and GRIVSTYS (SEQ ID NO: 20), the CDR 2 is selected from amino-acid sequences ISWNGAETD (SEQ ID NO: 21),
ISWNGGETD (SEQ ID NO: 22), and ISWNGAETQ (SEQ ID NO: 23), the CDR 3 is selected from amino-acid sequences AKPQSHFYDGSWRRASAYDD (SEQ ID
NO: 24), AKPQSHFSDGSWRRASAYDD (SEQ ID NO: 25), AKPQSHFYDGSWRRALAYDD (SEQ ID NO: 26) and AKPQSHFYDGSWRRASAYGD (SEQ ID NO: 27);
and/or the CDR 1 is amino-acid sequence RRTFRSYA (SEQ ID NO: 28), the CDR 2 is amino-acid sequence TTWIISSTY (SEQ ID NO: 29), and the CDR 3 is amino-acid sequence AARVRSGSGQYTLPGHYDY (SEQ ID NO: 30); and/or specifically binds to rivaroxaban and wherein the CDR 1 is amino-acid sequence TRISSLTV (SEQ ID NO: 31), the CDR 2 is amino-acid sequence LTRFGLAG (SEQ ID NO: 32), and the CDR 3 is selected from amino-acid sequences NVKTLGGADY (SEQ ID NO: 33) and
NAKTLGGADY (SEQ ID NO: 34); and/or the CDR 1 is amino-acid sequence GRTFTSYT (SEQ ID NO: 35), the CDR 2 is amino-acid sequence ISWSYWNGDSTW (SEQ ID NO: 36), and the CDR 3 is selected from amino-acid sequences AARPSARITSRRSDYDY (SEQ ID NO: 37) and VARPSARITSRRSDYDY (SEQ ID NO: 38); and/or the CDR 1 is selected from amino-acid sequences GRTFSTLA (SEQ ID NO: 39) and
GRTFGTLA (SEQ ID NO: 40) the CDR 2 is amino-acid sequence IIRNSLSTY (SEQ ID NO: 41), and the CDR 3 is amino-acid sequence AAGRWEAVRTNTPDY (SEQ ID NO: 42); and/or the CDR 1 is selected from amino-acid sequences GRTFSTLA (SEQ ID NO: 43), GRTSSTLA (SEQ ID NO: 44) and GRTFSLLA (SEQ ID NO: 45), the CDR 2 is selected from amino-acid sequences IIRNSISTY (SEQ ID NO: 46) and
ITRNSISTY (SEQ ID NO: 47) the CDR 3 is selected from amino-acid sequences AAGRWEAVRTNTPDY (SEQ ID NO: 48),
AAGRWEAVRTDTPDY (SEQ ID NO: 49) and AAGRWEAVRTITPDY (SEQ ID NO: 50); and/or specifically binds to betrixaban.
After immunization of separate llama’s with either edoxaban, or apixaban, or rivaroxaban, and development of VHH producing clones therefrom, a large series of VHHs was obtained comprising multiple different VHHs specifically binding to edoxaban and inhibiting the anti-coagulant activity of edoxaban, and multiple different VHHs specifically binding to apixaban and inhibiting the anti-coagulant activity of apixaban, and multiple different VHHs specifically binding to rivaroxaban and inhibiting the anti-coagulant activity of rivaroxaban. As outlined in Table A, below. Several clusters of homologous
VHHs within a cluster were obtained for each of the DOACs. This provides for the opportunity to select and combine for each specific DOAC VHHs with the same amino-acid sequence, with homologous amino-acid sequences and/or with varying amino-acid sequences (when selected from different clusters of VHHs specific for the same DOAC). Half-life of multivalent polypeptides of the invention comprising
VHHs specific for the same DOAC though having varying amino-acid sequences may contribute to extended half-life of the polypeptide in the circulation of a patient, compared to a multivalent polypeptide comprising or consisting of multiple copies of the same VHH.
In an embodiment of the invention the polypeptide comprises or consists of a sdab, wherein the sdab('s) comprise(s) any one or more of the combination(s) of CDR 1-, CDR 2-, and CDR 3 amino-acid sequences, that specifically bind(s) to edoxaban and is/are selected from the group of combinations of CDR 1- , CDR 2-, CDR 3 amino-acid sequences consisting of:
GFAFDDYA (SEQ ID NO: 1), ISSSDGSTY (SEQ ID NO: 3) and AAVPRTMYSRWGCGVRPYYYGMDY (SEQ ID NO: 4);
GFAFDDYA (SEQ ID NO: 1), ISSSDGSTY (SEQ ID NO: 3) and AAVPRTMNSRWGCGVRPYYYGMDY (SEQ ID NO: 5);
GFAFDDYA (SEQ ID NO: 1), ISSSDGSTY (SEQ ID NO: 3) and AAVPRTMYSRWGCGVRPYYHGMDY (SEQ ID NO: 6);
GFTFDDYA (SEQ ID NO: 2), ISSSDGSTY (SEQ ID NO: 3) and AAVPRTMYSRWGCGVRPYYYGMDY (SEQ ID NO: 4);
GRTFSINV (SEQ ID NO: 7), IWWSGGASQ (SEQ ID NO: 8) and AAGPMFSMDYRRVNY (SEQ ID NO: 10);
GRTFSINV (SEQ ID NO: 7), TWWSGGASQ (SEQ ID NO: 9) and AAGPMFSMDYRRVNY (SEQ ID NO: 10);
GRTFSINV (SEQ ID NO: 7}, IWWSGGASQ (SEQ ID NO: 8) and AAGPVFSMDYRRVNY (SEQ ID NO: 11);
GRTFSINV (SEQ ID NO: 7), IWWSGGASQ (SEQ ID NO: 8) and AAGPMFSMDYTRVNY (SEQ ID NO: 12);
GRTFSINV (SEQ ID NO: 7), IWWSGGASQ (SEQ ID NO: 8) and AAGPMFSMDYRRVNH (SEQ ID NO: 13);
GRTFSSYH (SEQ ID NO: 14), ITRGGGVTY (SEQ ID NO: 15) and AADAIWNQVRWMETKYTY (SEQ
ID NO: 17); and/or
GRTFSSYH (SEQ ID NO: 14), IARGGGVTY (SEQ ID NO: 16) and AADAIWNQVRWMETKYTY (SEQ
ID NO: 17); and/or that specifically bind(s) to apixaban and is/are selected from the group of combinations of CDR 1-, CDR 2-, CDR 3 amino-acid sequences consisting of:
GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRASAYDD (SEQ
ID NO: 24);
GRIVGIYS (SEQ ID NO: 19), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRASAYDD (SEQ
ID NO: 24);
GRIVSTYS (SEQ ID NO: 20), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRASAYDD (SEQ ID NO: 24);
GRIVSIYS (SEQ ID NO: 18), ISWNGGETD (SEQ ID NO: 22) and AKPQSHFYDGSWRRASAYDD (SEQ
ID NO: 24);
GRIVSIYS (SEQ ID NO: 18), ISWNGAETQ (SEQ ID NO: 23) and AKPQSHFYDGSWRRASAYDD (SEQ
ID NO: 24};
GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFSDGSWRRASAYDD (SEQ
ID NO: 25);
GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRALAYDD (SEQ
ID NO: 26);
GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRASAYGD (SEQ
ID NQ: 27); and/or
RRTFRSYA (SEQ ID NO: 28), TTWIISSTY (SEQ ID NO: 29) and AARVRSGSGQYTLPGHYDY (SEQ
ID NO: 30); and/or that specifically bind(s) to rivaroxaban and is/are selected from the group of combinations of CDR 1-,
CDR 2-, CDR 3 amino-acid sequences consisting of:
TRISSLTV (SEQ ID NO: 31), LTRFGLAG (SEQ ID NO: 32) and NVKTLGGADY (SEQ ID NO: 33);
TRISSLTV (SEQ ID NO: 31), LTRFGLAG (SEQ ID NO: 32) and NAKTLGGADY (SEQ ID NO: 34};
GRTFTSYT (SEQ ID NO: 35), ISWSYWNGDSTW (SEQ ID NO: 38) and AARPSARITSRRSDYDY (SEQ ID NO: 37);
GRTFTSYT (SEQ ID NO: 35), ISWSYWNGDSTW (SEQ ID NO: 36) and VARPSARITSRRSDYDY (SEQ ID NO: 38);
GRTFSTLA (SEQ ID NO: 39), IRNSLSTY (SEQ ID NO: 41) and AAGRWEAVRTNTPDY (SEQ ID NO: 42),
GRTFGTLA (SEQ ID NO: 40), IRNSLSTY (SEQ ID NO: 41) and AAGRWEAVRTNTPDY (SEQ ID NO: 42);
GRTFSTLA (SEQ ID NO: 43), IRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTNTPDY (SEQ ID NO: 48);
GRTSSTLA (SEQ ID NO: 44}, IRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTNTPDY (SEQ ID NO: 48);
GRTFSLLA (SEQ ID NO: 45), IIRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTNTPDY (SEQ ID NO: 48);
GRTFSTLA (SEQ ID NO: 43), ITRNSISTY (SEQ ID NO: 47) and AAGRWEAVRTNTPDY (SEQ ID NO: 48);
GRTFSTLA (SEQ ID NO: 43), IRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTDTPDY (SEQ ID NO: 49) and/or
GRTFSTLA (SEQ ID NO: 43), IIRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTITPDY (SEQ ID NO: 50).
In an embodiment of the invention the polypeptide comprises or consists of a sdab, wherein the sdab(‘s) comprise(s) - the combination of CDR 1-, CDR 2-, CDR 3 amino-acid sequences GFAFDDYA (SEQ ID NO: 1), ISSSDGSTY (SEQ ID NO: 3) and AAVPRTMYSRWGCGVRPYYYGMDY (SEQ ID NO: 4), that specifically binds to edoxaban; and/or - the combination of CDR 1-, CDR 2-, CDR 3 amino-acid sequences GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRASAYDD (SEQ ID NO: 24), that specifically binds to apixaban; and/or - the combination of CDR 1-, CDR 2-, CDR 3 amino-acid sequences TRISSLTV (SEQ ID NO: 31), LTRFGLAG (SEQ ID NO: 32) and NVKTLGGADY (SEQ ID NO: 33), that specifically binds to rivaroxaban.
In an embodiment of the invention the polypeptide comprises or consists of a sdab, wherein the sdab(‘s) is/are (a) VHH(s), preferably llama VHH(s), wherein the VHH(s) either specifically bind(s) to edoxaban and comprise(s) or consist(s} of any one or more of the amino-acid sequences selected from SEQ ID NO: 51 — SEQ ID NO: 63, SEQ ID NO: 98 and SEQ
ID NO: 99 (as detailed in Table A), preferably SEQ ID NO: 51, SEQ ID NO: 99 or SEQ ID NO: 98, more preferably SEQ ID NO: 51; and/or specifically bind(s) to apixaban and comprise(s) or consist(s) of any one or more of the amino-acid sequences selected from SEQ ID NO: 64 — SEQ ID NO: 78, SEQ ID NO: 100 and SEQ
ID NO: 101 (as detailed in Table A), preferably SEQ ID NO: 64, SEQ ID NO: 101 or SEQ ID NO: 100, more preferably SEQ ID NO: 64; and/or specifically bind(s) to rivaroxaban and comprise(s) or consist(s) of any one or more of the amino-acid sequences selected from SEQ ID NO: 79 — SEQ ID NO: 97, SEQ ID NO: 102 and SEQ
ID NO: 103 (as detailed in Table A), preferably SEQ ID NO: 79, SEQ ID NO: 103 or SEQ ID NO: 102, more preferably SEQ ID NO: 79.
In an embodiment of the invention the polypeptide comprises or consists of a sdab, wherein the sdab(’s) is/are (a) VHH(s), preferably llama VHH(s), wherein the VHH(s) either specifically bind(s) to edoxaban and comprise(s) or consist(s) of an amino-acid sequence selected from SEQ ID NO: 51 and SEQ ID NO: 53, preferably SEQ ID NO: 51; and/or specifically bind(s} to apixaban and comprise(s) or consist(s} of any one or more of the amino-acid sequences selected from SEQ ID NO: 64, SEQ ID NO: 70, SEQ ID NO: 76 and SEQ
ID NO: 77, preferably SEQ ID NO: 64; and/or specifically bind(s) to rivaroxaban and comprise(s) or consist(s) of any one or more of the amino-acid sequences selected from SEQ ID NO: 79, SEQ ID NO: 80, SEQ ID NO: 81 and SEQ
ID NO: 82, preferably SEQ ID NO: 79.
In an embodiment of the invention the polypeptide comprises or consists of a sdab, wherein the sdab('s) comprise(s) or consist(s) of (an) amino acid sequence(s) that has/have at least 95% sequence identity with any one or more of the amino-acid sequences with SEQ ID NO: 51 — SEQ ID NO 103, and/or wherein the sdab(‘s) comprise(s} an N-terminal His-tag and/or a C-terminal His-tag, preferably a C-terminal His-tag, more preferably said His-tag consisting of 6 histidine residues.
In an embodiment of the invention the polypeptide comprises or consists of a sdab, wherein the sdab('s} is/are (a) VHH(s), wherein the VHH domain(s) specifically bind(s) to edoxaban and comprise(s) or consist(s) of an amino-acid sequence with SEQ
ID NO: 51 or SEQ ID NO: 53, preferably SEQ ID NO: 51, or an amino acid sequence that has at least 95% sequence identity with SEQ ID NO: 51 or SEQ ID NO: 53, preferably SEQ ID NO: 51; and/or specifically bind(s) to apixaban and comprise(s) or consist(s) of an amino-acid sequence with any one of SEQ ID NO: 64, SEQ ID NO: 70, SEQ ID NO: 76 and SEQ ID NO: 77, preferably
SEQ ID NO: 64, or an amino acid sequence that has at least 95% sequence identity with SEQ ID
NO: 64, SEQ ID NO: 70, SEQ ID NO: 76 and SEQ ID NO: 77, preferably SEQ ID NO: 64; and/or specifically bind(s) to rivaroxaban and comprise(s) or consist({s} of an amino-acid sequence with any one of SEQ ID NO: 79, SEQ ID NO: 80, SEQ ID NO: 81 and SEQ ID NO: 82, preferably SEQ ID NO: 79, or an amino acid sequence that has at least 95% sequence identity with
SEQ ID NO: 79, SEQ ID NO: 80, SEQ ID NO: 81 and SEQ ID NO: 82, preferably SEQ ID NO: 79, or the polypeptide comprises or consists of a sdab, wherein the polypeptide consists of an amino-acid sequence with any one of SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 98 and SEQ
ID NO: 99, preferably SEQ ID NO: 51, or an amino acid sequence that has at least 95% sequence identity with any one of SEQ ID NO: 51, SEQ ID NO: 53, SEQ ID NO: 98 and SEQ ID NO: 99, preferably SEQ ID NO: 51; or consists of an amino-acid sequence with any one of SEQ ID NO: 64, SEQ ID NO: 70, SEQ ID NO: 76, SEQ ID NO: 77, SEQ ID NO: 100 and SEQ ID NO: 101, preferably SEQ ID NO: 64, or an amino acid sequence that has at least 95% sequence identity with any one of SEQ ID NO: 64, SEQ ID
NO: 70, SEQ ID NO: 76, SEQ ID NO: 77, SEQ ID NO: 100 and SEQ ID NO: 101, preferably SEQ
ID NO: 64; or consists of an amino-acid sequence with any one of SEQ ID NO: 79, SEQ ID NO: 80, SEQ ID NO: 81, SEQ ID NO: 82, SEQ ID NO: 102 and SEQ ID NO: 103, preferably SEQ ID NO: 79, or an amino acid sequence that has at least 95% sequence identity with any one of SEQ ID NO: 79, SEQ ID
NO: 80, SEQ ID NO: 81, SEQ ID NO: 82, SEQ ID NO: 102 and SEQ ID NO: 103, preferably SEQ
ID NO: 79.
In an embodiment of the invention the polypeptide comprises sdab('s) specifically binding to at least one DOAC (capable of inhibiting fXa) and the polypeptide has one or more of the characteristics selected from the group consisting of: a) in human plasma, the molar ratio between the polypeptide and a DOAC, required for inhibiting the fXa inhibiting activity of the DOAC with at least 50%, is a ratio selected from 1:1 — 20:1, such as 1:1 for inhibiting the fXa inhibiting activity of edoxaban, 4:1 — 16:1 for inhibiting the fXa inhibiting activity of apixaban, and 3:1 — 16:1 for inhibiting the fXa inhibiting activity of rivaroxaban; b) the polypeptide at room temperature has an association rate constant (ka) of 1.00e+08 1/Ms or higher for binding to a DOAC, as determined with surface plasmon resonance with immobilized DOAC, such as 1.00e+06 1/Ms — 1.00e+08 1/Ms for binding to apixaban, edoxaban, rivaroxaban or betrixaban, preferably apixaban, and c) the polypeptide at room temperature has a dissociation constant (ks) of 1.00e-01 1/s or lower, when bound to a DOAC, as determined with surface plasmon resonance with immobilized DOAC, such as 1.00e-01 1/s — 1.00e-03 1/s for dissociating from apixaban, edoxaban, rivaroxaban or betrixaban, preferably apixaban; and/or d) the polypeptide at room temperature has an affinity (Ko) of 1.00e-8 nM or lower, for binding to the first, second and/or third DOAC, wherein the first DOAC is edoxaban, the second
DOAC is apixaban and the third DOAC is rivaroxaban, as determined with surface plasmon resonance with immobilized DOAC, such as 1.00e-8 nM — 1.00e-10 nM for binding to apixaban, edoxaban, rivaroxaban or betrixaban, preferably apixaban; and/or e) the polypeptide at room temperature has an affinity (Ko) of 1.00e-8 nM or lower, for binding to the first, second and/or third DOAC, wherein the first DOAC is edoxaban, the second
DOAC is apixaban and the third DOAC is rivaroxaban, as determined with ELISA with immobilized albumin-DOAC, such as 1.00e-8 nM — 1.00e-10 nM for binding to edoxaban, 1.00e-8 nM — 1.00e-10 nM for binding to apixaban, and
1.00e-8 nM — 1.00e-10 nM for binding to rivaroxaban.
In an embodiment of the invention the polypeptide comprises sdab('s) specifically binding to at least one DOAC (capable of inhibiting fXa) and the polypeptide has one or more of the characteristics selected from the group consisting of; in human plasma, the molar ratio between the polypeptide and a DOAC, required for inhibiting the fXa inhibiting activity of the DOAC with at least 50%, is a ratio selected from 1:1 — 20:1, such as 1:1 for inhibiting the fXa inhibiting activity of edoxaban, 4:1 — 16:1 for inhibiting the fXa inhibiting activity of apixaban, and 3:1 — 16:1 for inhibiting the fXa inhibiting activity of rivaroxaban, wherein the ratio is for example determined by measuring the concentration of the polypeptide required to inhibit DOAC activity and to reverse the inhibition of fXa by the DOAC, in a plasma sample which comprises the DOAC. Such ratio is for example determined using a liquid anti-Xa (LAX) test as outlined in the Examples section here below. Such ratio is for example determined using a calibrated automated thrombinogram CAT assay method for measuring thrombin generation, as outlined in the Examples section here below. Such ratio is for example determined by measuring the coagulation lag time, as outlined in the Examples section here below. Such ratio is for example determined by measuring the peak thrombin values (normalization of thrombin generation under influence of the polypeptide) or (the compensation by the polypeptide of) the thrombin generation lag time, as outlined in the Examples section here below.
Furthermore or alternatively, in an embodiment of the invention the polypeptide comprises sdab(‘s) specifically binding to at least one DOAC (capable of inhibiting fXa) and the polypeptide has one or more of the characteristics selected from the group consisting of: a) the polypeptide does not bind to TFPI; b) the polypeptide does not inhibit the activity of TFPI; c) the polypeptide does not induce a pro-coagulant effect when contacted with blood or plasma of a subject; and d) the polypeptide does not bind to antithrombin [Il (ATI).
Absence of binding of the polypeptide to TFPI or to ATIII can be assessed by applying protein- protein binding assays known to the skilled person, such as, but not limited to, ELISA and surface plasmon resonance. In an embodiment, the polypeptide is or comprises at least one VHH specifically binding to any one of edoxaban, apixaban, betrixaban, rivaroxaban, preferably any one of edoxaban, apixaban, rivaroxaban. Such at least one VHH does not have a binding site for binding to either TFPI or ATI. Therewith, the polypeptide and the VHH(s) comprised by the polypeptide do not interfere with
TFPI activity and/or ATIII activity in a (plasma) sample or in the circulation of a subject. Due to the specificity of the VHH, the advantage of using the polypeptide of the invention for reversing anti- coagulant activity of a DOAC, is the absence of the (risk for) side effects that could be due to binding of the VHH to TFPI or ATIII.
An aspect of the invention relates to a mixture of at least two polypeptides of the invention, wherein a first polypeptide specifically binds to a first DOAC (capable of inhibiting fXa) and a second polypeptide specifically binds to a second DOAC (capable of inhibiting fXa), different from the first DOAC, and, if present, a third polypeptide specifically binds to a third DOAC (capable of inhibiting fXa) different from the first DOAC and different from the second DOAC.
Similarly to the advantages described here above for a polypeptide comprising at least one antibody such as a first VHH specifically binding to a first DOAC, at least one antibody such as a second VHH specifically binding to a second DOAC, a mixture of polypeptides the mixture comprising a first polypeptide specifically binding to a first DOAC and a second polypeptide specifically binding to a second DOAC, has the advantage of the capability to bind and reverse the anti-coagulant activity of both the first and second DOAC simultaneously. For a patient or blood sample or plasma sample for which it is not know which DOAC of the first or second DOAC is administered to the patient or is present in the sample, the mixture of at least two polypeptides provides for the possibility to inhibit and reverse the anticoagulant activity of the unknown first or second DOAC, without the necessity to know the identity of the DOAC present in the circulation of the patient or in the sample. Such mixture of polypeptides is thus suitable for immediate neutralization of an anti-coagulant activity of a DOAC of unknown identity, the DOAC inhibiting fXa and known to be selected from e.g. edoxaban, apixaban and rivaroxaban, wherein the mixture of polypeptides comprises at least one polypeptide each that specifically binds and inhibits the fXa inhibiting activity of each DOAC that is (putatively) present in a sample or in the circulation of a patient in need of treatment with an antidote capable of reversing anti- coagulant activity of a DOAC that inhibits fXa.
An aspect of the invention relates to a pharmaceutical composition comprising a polypeptide of the invention and a pharmaceutically acceptable carrier and/or a pharmaceutically acceptable diluent.
An aspect of the invention relates to a pharmaceutical composition comprising the mixture of at least two polypeptides and a pharmaceutically acceptable carrier and/or a pharmaceutically acceptable diluent.
As outlined here above, the polypeptide is capable of specifically binding and inhibiting a DOAC that inhibits fXa, therewith reversing the anti-coagulant activity of the DOAC. The DOAC for example and preferably being any one or more of edoxaban, apixaban and rivaroxaban {(multi-specific polypeptide and/or mixture of polypeptides with polypeptides in the mixture that have specificity for binding to a first DOAC or to a second DOAC), or any one of edoxaban, apixaban and rivaroxaban {mone-specific polypeptide). Therefore, the polypeptide or the mixture of polypeptides is suitable for reversing anti-coagulant activity of a DOAC in a patient to whom the DOAC is previously administered.
The polypeptide can be administered to a patient who is treated with a DOAC capable of inhibiting fXa, for the purpose of inhibiting the DOAC and therewith reversing the anti-coagulant activity of the DOAC.
Advantageously, the polypeptide(s) do not bind to TFPI or ATIII, and therewith do not interfere with coagulation pathways otherwise than reversing the DOAC activity and reinstating active fXa. To the surprise of the inventors, the polypeptide(s) is/are not only capable of binding specifically to a selected
DOAC that is either immobilized or in solution in the absence of coagulation factors such as factor X(a).
In addition, the inventors established that the polypeptide(s) is/are also binding to the DOAC that is bound to fXa and therewith that is inhibiting fXa. Binding of the polypeptide(s) to fXa-bound DOAC results in the DOAC being released from fXa and in reversing the fXa inhibiting activity of the DOAC.
For example, in plasma comprising DOAC, fXa is inhibited, as established in a coagulation lag time test and/or a thrombin generation test and/or a liquid anti-Xa (LAX) test, and as compared with fXa activity as determined for control plasma in the absence of the DOAC. Contacting the polypeptide(s) of the invention with the plasma of patients to whom a DOAC was administered now results in reversing the fXa-inhibiting activity of the DOAC and restores fXa back to normal levels seen for plasma lacking such
DOAC. Based on these findings, the polypeptide(s) of the invention provide for a treatment modality suitable for reversing anti-coagulant therapy in a patient in need thereof. Such a patient is for example a subject to whom a DOAC has been administered. The DOAC is for example any one or more of edoxaban, apixaban, betrixaban and rivaroxaban, preferably edoxaban, apixaban and rivaroxaban (multi-specific polypeptide and/or mixture of polypeptides with polypeptides in the mixture that have specificity for binding to a first DOAC or to a second DOAC), or any one of edoxaban, apixaban, betrixaban and rivaroxaban, preferably edoxaban, apixaban and rivaroxaban (mono-specific polypeptide).
An aspect of the invention relates to the polypeptide of the invention, the mixture of polypeptides of the invention or the pharmaceutical composition of the invention, wherein the first DOAC, second DOAC and third DOAC are selected from edoxaban, apixaban and rivaroxaban.
An aspect of the invention relates to the polypeptide of the invention, for use as a medicament.
An aspect of the invention relates to the mixture of polypeptides of the invention, for use as a medicament.
An aspect of the invention relates to the pharmaceutical composition of the invention, for use as a medicament.
Generally stated, the invention concerns a therapeutic method of treatment of a subject suffering from - inhibited fXa activity and/or anti-coagulant therapy due to administered DOAC to the patient; said therapeutic method of treatment comprising: the administration, to said subject, of a polypeptide of the invention which is capable of reversing, neutralizing and/or inhibiting a DOAC (capable of inhibiting fXa) that is administered to the subject; wherein the administration of the polypeptide results in inhibition of the fXa inhibiting activity of the
DOAC and in (increased and/or restored) fXa activity.
A particular aspect of the invention concerns a therapeutic method of treatment of a subject suffering from - inhibited coagulation - bleeding or major bleeding - uncontrolled bleeding and/or
- anti-coagulant activity in the subject to whom an anti-coagulant such as an inhibitor of activated factor X, for example a DOAC (capable of inhibiting fXa), is administered; said therapeutic method of treatment comprising: the administration, to said subject, of a polypeptide of the invention which is capable of reversing, neutralizing and/or inhibiting a DOAC (capable of inhibiting fXa) that is administered to the subject; wherein the administration of the polypeptide results in inhibition of the fXa inhibiting activity of the
DOAC and in (increased and/or restored) fXa activity.
The DOAC is preferably any one (or more than one, for combined mono-specific polypeptides and/or for a multi-specific polypeptide) of edoxaban, apixaban, betrixaban and rivaroxaban, preferably, edoxaban, apixaban and rivaroxaban. The subject is for example a human patient in need of reversing or inhibiting anti-coagulant therapy, such as a patient to whom a DOAC is previously administered and who for example is (fatally, excessively or major) bleeding or for example will or needs to undergo surgical procedures (such as for example hip replacement, or for example due to injury and/or an accident).
An aspect of the invention relates to the polypeptide of the invention, for use in the treatment of inhibited coagulation in a patient or in the treatment of inhibited fXa activity in a patient and/or for use in the prevention or treatment of bleeding or major bleeding in a patient or uncontrolled bleeding in a patient, and/or for use in the reversal of anti-coagulant activity in a patient to whom an anti-coagulant such as an inhibitor of activated factor X, for example a DOAC (capable of inhibiting fXa), is administered. The patient is for example and preferably a human subject. The DOAC is selected from edoxaban, apixaban, betrixaban and rivaroxaban, preferably from edoxaban, apixaban and rivaroxaban.
An aspect of the invention relates to the mixture of polypeptides of the invention, for use in the treatment of inhibited coagulation in a patient or in the treatment of inhibited fXa activity in a patient and/or for use in the prevention or treatment of bleeding or major bleeding in a patient or uncontrolled bleeding in a patient, and/or for use in the reversal of anti-coagulant activity in a patient to whom an anti-coagulant such as an inhibitor of activated factor X, for example a DOAC (capable of inhibiting fXa), is administered.
One of the advantages of such a mixture of mono-specific or multi-specific polypeptides for use in the treatment of a bleeding patient and/or of a patient who will or has to be subjected to a surgical procedure, is that more than a single DOAC is inhibited in the circulation of said patient when administered to said patient after a DOAC has been administered to said patient. When it is not known (in time) which DOAC has been administered previously to a patient in need of reversal of anti-coagulant therapy, administering a composition comprising polypeptides capable of inhibiting more than one
DOAC such as the mixture of polypeptides of the invention, has the advantage that at least the one unknown DOAC that has been previously administered is inhibited. Preferably, such a mixture of polypeptides of the invention (for use according to the invention) comprises at least the antibodies, preferably being sdabs such as VHH, that specifically bind to the DOACs that are authorized for administration to human subjects in need of anticoagulant therapy. For example, the mixture of polypeptides comprise polypeptides specifically binding to edoxaban and apixaban, or edoxaban and rivaroxaban, or apixaban and rivaroxaban, or edoxaban, apixaban and rivaroxaban. In an embodiment, the mixture comprises polypeptides capable of specifically inhibiting at least two different DOACs that are authorized for human use, such as two, three, four or five different DOACs. In a preferred embodiment, the mixture of polypeptides comprises a first polypeptide binding to a first DOAC and a second polypeptide binding to a second DOAC, for use according to the invention. For example, when the patient needs temporarily reversal of anti-coagulant therapy, for example for a few hours (during a surgical procedure for example), anti-coagulant therapy can start again after those for example few hours by administering a third DOAC for which the mixture of polypeptides does not comprise a polypeptide that binds to said third DOAC.
An aspect of the invention relates to the pharmaceutical composition of the invention, for use in the treatment of inhibited coagulation in a patient or in the treatment of inhibited fXa activity in a patient and/or for use in the prevention or treatment of bleeding or major bleeding in a patient or uncontrolled bleeding in a patient, and/or for use in the reversal of anti-coagulant activity in a patient to whom an anti-coagulant such as an inhibitor of activated factor X, for example a DOAC (capable of inhibiting fXa), is administered.
An embodiment is the polypeptide according to the invention, the mixture of polypeptides of the invention or the pharmaceutical composition according to the invention, for use according to the invention as here above described, wherein a DOAC that is an inhibitor of fXa is previously administered to said patient, before the polypeptide, the mixture of polypeptides or the pharmaceutical composition is administered to said patient, and wherein the polypeptide or at least one of the polypeptides comprised by the mixture of polypeptides or comprised by the pharmaceutical composition specifically binds to said DOAC.
An embodiment is the polypeptide according to the invention, the mixture of polypeptides of the invention or the pharmaceutical composition according to the invention, for use according to the invention as here above described, wherein the DOAC is selected from edoxaban, apixaban and rivaroxaban.
A further aspect of the invention concerns the use of the polypeptide of the invention in the manufacture of a medicament for use in a therapeutic method of treatment of a subject suffering from a disease or condition related to: - inhibited fXa activity - inhibited coagulation - bleeding or major bleeding - uncontrolled bleeding and/or - anti-coagulant activity in the subject to whom an anti-coagulant such as an inhibitor of activated factor X, for example a DOAC (capable of inhibiting fXa), is administered; said therapeutic method of treatment comprising:
the administration, to said subject, of a polypeptide of the invention which is capable of reversing, neutralizing and/or inhibiting a DOAC (capable of inhibiting fXa) that is administered to the subject; wherein the administration of the polypeptide results in inhibition of the fXa inhibiting activity of the
DOAC and in (increased and/or restored) Xa activity.
The DOAC is preferably any one (or more than one, for combined mono-specific polypeptides and/or for a multi-specific polypeptide) of edoxaban, apixaban, betrixaban and rivaroxaban, preferably, edoxaban, apixaban and rivaroxaban. The subject is for example a human patient in need of reversing or inhibiting anti-coagulant therapy, such as a patient to whom a DOAC is previously administered and who for example is (fatally, excessively or major) bleeding or for example will or needs to undergo surgical procedures (such as for example hip replacement, or for example due to injury and/or an accident).
A pharmaceutical composition for (treatment of) - inhibited fXa activity - inhibited coagulation - bleeding or major bleeding - uncontrolled bleeding and/or - anti-coagulant activity in the subject to whom an anti-coagulant such as an inhibitor of activated factor X, for example a DOAC (capable of inhibiting fXa), is administered, comprising the polypeptide of the invention or the mixture of polypeptides of the invention as an active ingredient.
In an embodiment, the polypeptide of the invention is an antidote of a DOAC (capable of inhibiting fXa) or fXa inhibitor (DOAC (capable of inhibiting fXa)) inhibiting agent as an active ingredient or a (pharmaceutical) composition comprising the antidote comprising the polypeptide of the invention as an active ingredient or the mixture of polypeptides of the invention as an active ingredient.
Anticoagulants such as the DOACs according to the invention (that inhibit fXa) are developed for the prevention and reduction of recurrent venous thromboembolism, stroke prevention in patients with non-valvular atrial fibrillation, and for reducing the incidence of recurrent ischemic events and death in patients with acute coronary syndrome. Anticoagulation therapy is also required in patients having clotting disorders, restricted mobility or undergoing medical surgery.
Therefore, in embodiments of the invention, the polypeptide is for use in the reversal of anticoagulant therapy in a patient to whom a DOAC has been administered, and who is for example suffering from bleeding and/or in need of (further) surgical procedures, wherein said patient is suffering from, and/or treated for prevention against (treated for prophylaxis of), or prevention of recurrence of any one or more of; recurrent venous thromboembolism, stroke, for example when the patient has non-valvular atrial fibrillation, recurrent ischemic events, systemic embolism, for example when the patient has nonvalvular AF (NVAF),
death, e.g. when the patient suffers from acute coronary syndrome, clotting disorders, and restricted mobility, deep vein thrombosis (DVT), venous thromboembolic events (VTE), for example when a(n) (adult) patient has undergone elective total hip replacement surgery or total knee replacement surgery, pulmonary embolism (PE), or who is undergoing medical surgery.
Discontinuing anticoagulant therapy based on a DOAC, that is to say no further administration of the DOAC to the patient, will result in the elimination of the drug (therapeutic molecule) from the blood circulation in relatively short times due to the short half-lives of the DOACs (edoxaban, apixaban, rivaroxaban, for example) ranging from 5 to 17 hours. However, in cases of emergencies including an undesired (major) bleeding such as a life-threatening major bleeding, or non-elective major surgery anticoagulation, a faster method for reversing or neutralizing the effects of DOACs is desired.
An advantage of the polypeptide of the invention is the relatively fast anticoagulant reversal time when the polypeptide is added to a blood sample such as plasma of a patient, or when added to the circulation of the patient in need of reversal of DOAC therapy, the reversal time being in the order of minutes to half an hour, such as 5 — 20 minutes, for example 10 minutes or less, such as 7-10 minutes. The patient is for example a human patient in need of inhibition (reversal) of DOAC fXa inhibitory activity. The patient may suffer from any one or more of: recurrent venous thromboembolism, stroke, for example when the patient has non-valvular atrial fibrillation, recurrent ischemic events, systemic embolism, for example when the patient has nonvalvular AF (NVAF), acute coronary syndrome, clotting disorders, and restricted mobility, deep vein thrombosis (DVT), venous thromboembolic events (VTE), for example when a(n) (adult) patient has undergone elective total hip replacement surgery or total knee replacement surgery, pulmonary embolism (PE), and the patient is for example in need of a surgical procedure, and/or is bleeding, necessitating an intervention to stop the bleeding.
Therefore, one of the many advantages of the polypeptide of the invention is the relatively short time for reversing anti-coagulant activity of a DOAC administered to a patient, the short time being in the order of minutes such as 5-15 minutes, for example 7-12 minutes, or less than 10 minutes. In addition to this advantage, polypeptides of the invention comprising or consisting of a single sdab such as a VHH, or comprising or consisting of two or three sdabs, such as VHHs, have the advantage that the clearance time in the circulation is also relatively short due to the size of the polypeptide (being less than 50 kDa, and even about 15 kDa for a single sdab / VHH). Therefore, due to the relatively fast
DOAC reversing activity of the polypeptide and/or due to the fast clearance from the circulation, either alone or in combination, these two advantages provide for an advantageous polypeptide when fast onset of anticoagulant therapy is needed and/or desired, for example during a bleeding episode or when the patient is in need of (emergency) surgical treatment, and when thereafter, {re-)start of anti-coagulant therapy is required or desired with a DOAC such as the very same DOAC of which the activity was reversed upon treatment with the polypeptide.
As outlined before, since the polypeptide of the invention does not bind to TFPI and does not inhibit the activity of TFPI, an advantage of the polypeptide is that administering the polypeptide to a patient in need thereof who is treated with a DOAC to which the polypeptide binds, does not result in an increase of tissue factor-initiated thrombin generation. Therewith, an advantage is that the polypeptide does not have the undesired pro-coagulant side effect of inducing thrombin generation.
Such side effect may result in (an increased risk for) any one or more of: recurrent venous thromboembolism, stroke, for example when the patient has or previously had non-valvular atrial fibrillation, recurrent ischemic events, systemic embolism, for example when the patient has or previously had nonvalvular AF (NVAF), acute coronary syndrome, clotting disorders, and restricted mobility, deep vein thrombosis (DVT), venous thromboembolic events (VTE), for example when a(n) (adult) patient has to undergo or has undergone elective total hip replacement surgery or total knee replacement surgery, pulmonary embolism (PE),
An advantage of the monospecific polypeptide of the invention is that it only specifically binds to a certain selected DOAC without binding to a different DOAC, wherein the binding to the first DOAC results in reversal of the anti-coagulant activity of the first DOAC. Administering a second DOAC will result in anti-coagulant activity irrespective whether the polypeptide specific for binding and reversing the first DOAC is (to some extent) in the circulation of the patient in need of anti-coagulant therapy (for example after major bleeding is halted and/or a surgical procedure has been completed). Reversing specifically a selected first DOAC with a polypeptide and starting anti-coagulant therapy with a second
DOAC to which the polypeptide does not bind is a major advantage of the polypeptide. It provides flexibility to the practitioner and patient with regard to (temporarily) reversing a current first DOAC therapy and with regard to the possibility to (re-)start DOAC therapy with a second DOAC different from the first DOAC at whish at any moment after the polypeptide is administered to the patient and independent of whether the polypeptide without capability to bind to the second DOAC is still present in the circulation.
The ability of Andexanet alfa for inhibiting more than one DOAC and its interaction with other actors of the coagulation cascade can cause undesired or unpredictable side effects and also requires the administration of Andexanet alfa in high doses. High concentrations of Andexanet alfa in the circulation are required, relating to a high proportion of Andexanet alfa cross-reacting with antithrombin
IN (ATI) which is highly abundant in blood, and with TFPI, which results in a loss of factor Xa scavenging function for Andexanet alfa. Therefore, the polypeptide of the invention has several further advantages. Since the polypeptide comprising or consisting of VHH(s) does not bind to TFPI and does not bind to ATIII, the polypeptide is effective at lower dose. Any polypeptide administered to a patient is available for binding to the DOAC for which the polypeptide has binding specificity.
The present invention also provides a method of treating bleeding such as major bleeding, the method comprising the step of administering a medicament comprising a polypeptide according to the invention to a patient in need thereof, preferably administering an effective dose of said medicament to a patient in need thereof, preferably a human patient at risk of bleeding, such as a patient who is previously treated with a DOAC of which the activity can be reversed or inhibited by the polypeptide.
Also provided is the use of a polypeptide of the invention or a composition comprising at least one polypeptide according to the invention for the manufacture of a medicament. An embodiment is the use of a polypeptide of the invention or a (pharmaceutical) composition comprising at least one polypeptide according to the invention for the manufacture of a medicament for reversing the anti- coagulant activity of a DOAC in a patient to whom the DOAC was previously administered. The patient may suffer from bleeding or may be at risk for suffering from bleeding, for example when the patient is about to be subjected to surgical procedure.
The inventors established that the DOAC neutralizing effect of the polypeptide of the invention was already apparent within 10 minutes in human plasma, after mixing the plasma with the polypeptide, such as a polypeptide binding to any one of edoxaban, apixaban and rivaroxaban. Moreover, the rapid anti-coagulant reversing activity of the polypeptide is illustrative for a rapid and complete DOAC neutralizing effect in the patient's circulation when the polypeptide is administered to the patient intravenously at a dose sufficient to establish a concentration of the polypeptide in the plasma of the patient. For example, when the patient is treated with edoxaban, resulting in a certain plasma concentration in the circulation of edoxaban, a dose of the polypeptide of the invention for binding to edoxaban, resulting in a polypeptide concentration of one to fourfold the edoxaban concentration in the circulation, is sufficient and enough for effectively reversing edoxaban anti-coagulant activity. For example, when the patient is treated with edoxaban, resulting in a plasma concentration in the circulation of about 400 nM edoxaban, a dose of the polypeptide of the invention for binding to edoxaban, resulting in a polypeptide concentration of 400 — 1600 nM in the circulation, is sufficient and enough for effectively reversing edoxaban anti-coagulant activity. For example, when the patient is treated with apixaban, resulting in a certain plasma concentration in the circulation of apixaban, a dose of the polypeptide of the invention for binding to apixaban, resulting in a polypeptide concentration of about fourfold the apixaban concentration in the circulation, is sufficient and enough for effectively reversing apixaban anti-coagulant activity. For example, when the patient is treated with apixaban, resulting in a plasma concentration in the circulation of about 400 nM apixaban, a dose of the polypeptide of the invention for binding to apixaban, resulting in a polypeptide concentration of 400 — 1600 nM in the circulation, is sufficient and enough for effectively reversing apixaban anti-coagulant activity.
An aspect of the invention relates to a nucleic acid molecule comprising a nucleotide sequence encoding the polypeptide according to the invention, wherein the nucleotide sequence encoding the polypeptide further preferably encodes a signal peptide operably linked to the polypeptide, and wherein the nucleic acid molecule further preferably comprises regulatory elements conducive to the expression of the polypeptide, which regulatory elements are operably linked to the nucleotide sequence.
An aspect of the invention relates to a host cell comprising such an aforementioned nucleic acid molecule.
An aspect of the invention relates to a method for producing a polypeptide of the invention, the method comprising the steps of: a) culturing the host cell as here above mentioned under conditions conducive to the expression of the polypeptide; and, b) recovery of the polypeptide.
An aspect of the invention relates to a method for producing a pharmaceutical composition of the invention, wherein the method comprises the steps of: a) producing at least one polypeptide of the invention in the method for producing a polypeptide of the invention; and, b) formulating the polypeptide(s) with a pharmaceutically acceptable carrier and/or pharmaceutically acceptable diluent to obtain a pharmaceutical composition.
An aspect of the invention relates to the use of the polypeptide(s) of the invention or the pharmaceutical composition of the invention for in vitro detection or quantification of a DOAC (capable of inhibiting fXa) in a sample.
An embodiment of the invention is the use according to the invention, wherein: e the DOAC is any one of edoxaban, apixaban, betrixaban and rivaroxaban, preferably any one of edoxaban, apixaban and rivaroxaban; and/or e the sample is a plasma sample obtained from a (human) patient; and/or e the sample is obtained from a (human) patient to whom previously a DOAC is administered; and/or e the sample is obtained from a (human) patient who suffers from bleeding, major bleeding or uncontrolled bleeding, or is at risk for bleeding, major bleeding or uncontrolled bleeding; and/or e the sample is obtained from a (human) patient who needs to be subjected to surgery or is or has been subjected to surgery.
In an embodiment of the invention, the use relates to a sample from a (human) patient in need of inhibition (reversal) of DOAC fXa inhibitory activity. The patient may suffer from any one or more of: recurrent venous thromboembolism, stroke, for example when the patient has non-valvular atrial fibrillation, recurrent ischemic events, systemic embolism, for example when the patient has nonvalvular AF (NVAF), acute coronary syndrome, clotting disorders, and restricted mobility, deep vein thrombosis (DVT), venous thromboembolic events (VTE), for example when a(n) (adult) patient has undergone elective total hip replacement surgery or total knee replacement surgery, pulmonary embolism (PE), and the patient is for example in need of a surgical procedure, and/or is bleeding, necessitating an intervention to stop the bleeding.
An aspect of the invention relates to a kit comprising the polypeptide according to the invention or the mixture of polypeptides according to the invention or the (pharmaceutical) composition according to the invention, and optionally further comprising instructions for administration of the polypeptide(s) or the pharmaceutical composition, such as administration to a patient in need of inhibition of fXa inhibition by a previously administered DOAC (capable of inhibiting fXa) and/or to a patient at risk of bleeding, major bleeding or uncontrolled bleeding, or suffering from bleeding, major bleeding or uncontrolled bleeding, or to a patient to whom previously a DOAC is administered; and/or a patient who needs to be subjected to surgery or is or has been subjected to surgery.In an embodiment, the kit of the invention further comprises a second DOAC to which the polypeptide comprised by the kit does not bind. For example, the kit is herewith suitable for administering the polypeptide to a patient in need of reversal of anti-coagulant therapy, wherein the patient is previously administered with the DOAC to which the polypeptide binds, whereas the same kit is also suitable for administering the second DOAC to the same patient, after the polypeptide has been administered, in order to re-start anti-coagulation therapy at a suitable moment. For example, the kit is applied for arresting bleeding in a patient suffering from bleeding and being treated with a first DOAC (for example in an emergency situation or when the patient needs to undergo surgery such as a hip replacement surgery): the polypeptide binding to the first DOAC is administered and the patient can be treated for the bleeding episode or can be subjected to a surgical procedure. If the kit also comprises the second DOAC, after treatment of the bleeding or after the surgery, the patient can be administered with the second DOAC in order to restore the anti- coagulant therapy, yet with a different DOAC than before. Since the polypeptide acts fast (reverses the anti-coagulant activity of the first DOAC in a short time) and is cleared from the circulation relatively fast, the kit provides for the option to arrest anti-coagulation therapy for a relatively short period of time, and when the second DOAC is comprised by the kit, re-start anti-coagulation therapy at any time after the polypeptide has been administered.
In a further aspect, provided herein is a detection method for determining the presence of a
DOAC inhibitor of fXa (DOAC capable of inhibiting fXa) in a sample obtained from a subject, the method comprising the step of contacting the sample with the polypeptide according any one of the disclosed herein embodiments, wherein the polypeptide is used in detecting the DOAC inhibitor of fXa in the sample and/or is used as a detection agent.
Uses of target-recognising polypeptides in immunodiagnostic and other detection methods for analysing biological samples is known in the art and a multitude of detection set-ups have been developed to date. An overview of an exemplary number of them can e.g. be found in Gubala at al., "Immunodiagnostics and immunosensor design (IUPAC Technical Report)" Pure and Applied
Chemistry, vol. 86, no. 10, 2014, pp. 1539-1571. DOI 10.1515/pac-2013-1027; which can be adapted for the described herein diagnostic purposes and which are herewith incorporated by reference.
For example, in direct modes of target detection, the principle of immune-sandwiching can be used in different configurations incorporating the disclosed herein polypeptides. Usually, the polypeptides will be contacted with a sample obtained from a subject, advantageously being a liquid biopsy sample, likely comprising the subject's blood or at least plasma suspected to contain the DOAC inhibitor which can be bound by the selected one or more of the polypeptides of the present disclosure.
Depending on the chosen detection principle and assay format, the polypeptides can be labelled with signal-generating moieties, such as fluorophores, or can be recognised by secondary binding molecules labelled with such signal-generating moieties. The recognition by the secondary binding molecules can be direct or aided by known in the art high-affinity based interactions like in the pen-strep system or modifications thereof.
As it will be readily known to those skilled in the art, it is possible to design immunodetection strategies recognising several different DOAC inhibitors of fXa in one assay, wherein distinct and/or at least discernible signals will be associated with different binding events governed by the presented herein polypeptides exhibiting different DOAC-specificities. Depending on the selected assay design, such multiple DOAC recognition reactions can be performed in different aliquots of the sample obtained from the subject, possibly in parallel, or in a multiplex setting in one reaction volume, for example utilising one label per one specific DOAC-binding event. Possibly, the disclosed herein polypeptides can be provided as part of a microchip, possibly being a micro- and/or nanofluidic chip, advantageously configured to generate electrochemical or other signals in response to target DOAC binding.
Alternatively, detection principle can utilise the ability of the disclosed herein polypeptides to displace or to sterically prevent the association of the DOAC inhibitor with fXa, thus resulting in restoration or at least in increasing of fXa activity, which can be measured in the sample obtained from the subject. There currently exist several assays capable of assessing the extent of fXa activity in a sample, which can be incorporated into possible embodiments of the disclosed herein detection methods.
In one such general embodiment, a detection method is disclosed herein, comprising the steps of: - providing the sample obtained from the subject, - assessing a reference value associated with fXa activity in the sample or in a first aliquot of the sample; - contacting the sample or a second aliquot of the sample with the polypeptide of the invention and assessing a first test value after the contacting; wherein finding a difference between the first test value and the reference value indicates the presence of the DOAC inhibitor of fXa in the sample obtained from the subject.
As used herein, the term fXa activity in the sample is to be construed as relating to any specific or all forms of activated factor X (fXa), i.e. unbound, bound to another factor, or present in a larger complex of factors, which forms may exist naturally or be man-made for use as e.g. controls in detection methods and/or in therapeutic applications. For example, it is known that DOAC inhibitors like edoxaban, apixaban, and rivaroxaban inhibit substrate conversion by fXa and fXa bound to a2- macroglobulin (fXa-a2M). Hence, depending on the selection of reagents and signal-detection strategies, possible embodiments of the presented herein detection method can measure one or both of fXa or e.g. fXa-a2M activity.
The assessing of the reference value and/or the first test value and/or any subsequent reference values (in case control reagents or further polypeptides according to the present disclosure are included in the detection method) can, for example, be performed by a visual assessment, i.e., by the eye of the user performing the detection method. In such a case, it is possible that the finding of the difference which indicates successful detection of the DOAC inhibitor of fXa in the sample is self-evident as the user will immediately see it as, depending on the assay set-up, e.g. a perceivable generation of colour in the test sample only or by occurrence of a difference in colour or in amount of a generated precipitate between the reference and test sample.
In an embodiment, a detection method is provided, wherein the assessing of the reference value and/or of the first test value comprises a chromogenic assay, a light-generating assay, preferably being a fluorescence-generating assay, or an electric signal-generating assay, possibly on a chip.
Alternatively, the assessing can be aided by a device comprising a detector that is suitable for capturing the signals from the sample and/or signal-generating reagents introduced to the sample before the assessing is done. For example, these reagents can comprise signal-generating substrates of fXa.
The detector-aided mode of assessing is advantageous as it can not only confer superior sensitivity of detection as compared to the human eye but also can capture signals not visible to the latter. Hence, in an embodiment, a detection method is provided, wherein the assessing of the reference value and/or of the first test value comprises a chromogenic assay, a light-generating assay, preferably being a fluorescence-generating assay, or an electric signal-generating assay, possibly on a chip.
As explained above, there currently exists many assay types that can be used for assessing the reference and the first and possibly further test values. In possible embodiment, a detection method is provided, wherein the assessing of the reference value and/or of the first test value comprises low- molecular-weight heparin (LMWH) assay and/or heparin calibrated anti-fXa assay and/or an assay involving a substrate that generates or quenches a signal following cleavage by fXa.
As the skilled person will know, depending on the choice of reagents and the selected assay format, which will determine how the values associated with fXa activity are assessed, the finding of the difference can be an instantaneous act happening in the user's consciousness or can be an additional step comprising a conscious act of comparing the first test value and the reference value, possibly aided by a computing device comprising a processing unit. In an embodiment, the step of the comparing can be performed at the place of the assessing of the reference value and/or the first and/or any subsequent (if present) test values. Alternatively, the assessing of the values can be done in one place, after which the more or less processed values can be sent to another location like an analysis centre or a portable device of a medical professional, after which sending the step of comparing will take place, ultimately resulting in finding the difference indicative of the presence of the DOAC inhibitor of fXa or not. Hence, in an advantageous embodiment, the diagnostic method is provided further comprising the step of comparing the first test value and the reference value
As already explained above, the step of comparing may be seen as non-essential as the finding of the difference can in certain embodiments be seen as happening in the subconsciousness of the user, especially when the difference is immediately self-evident, or as it can be separated in space and time from the act of assessing the reference and test values. However, as any form of more or less subconscious comparing will usually be involved, in some embodiments the comparing can be performed during a visual assessment by a person performing the method, which e.g. involves a reaction that generates a colour or a change in colour that is perceivable by the human eye.
Alternatively, as already explained, the reference value and of the first test value can be measured by a device comprising an appropriate detector. A good example of a detector is one capable of capturing fluorescent or other light signals.
The detector-aided mode of assessing the values associated with fXa activity will usually entail attribution of numerical values to the reference and test values as measured by the detector; i.e. it will usually entail quantification. This is because for machine processability, the detector-equipped device converts the captured signals to numerical values. Such values can be communicated to and compared,
e.g. subtracted, by the user, or, advantageously, can be compared by a computing device comprising a processing unit. In line with the above, in an advantageous embodiment, a detection method is provided, wherein the step of comparing is performed by a computing device comprising a processing unit. In such instance, the computing device can further and advantageously comprise instructions for aiding the decision whether the difference is or should be considered found. Hence, in a possible embodiment of the computer-aided detection method, the computing device comprises instructions for deciding whether the difference between the first test value and the reference value is found.
Thanks to the quantification of the reference and/or the test values during the detector-aided assessing, the disclosed herein detection methods can further be amendable to (usually computer- implemented) mathematical operations for attaining greater reliability or for obtaining more information about the sample, for example by plotting dose-response curves and/or by investigation of kinetics of restoring fXa activity after the contacting with the polypeptide.
Hence, in an advantageous embodiment, a detection method is provided, wherein the assessing of the first test value further comprises evaluation of the change of the first test value over time, preferably wherein the detection method further comprises assessment of fXa activity (or reactivation) Kinetics after the contacting with the polypeptide, most preferably comprising assessment of chromogen or fluorescent signal detection kinetics.
Other examples of computer-implemented mathematical operations may e.g. include determination of detection thresholds, measurement transformations or corrections versus potential background noise to better determine the assessed values, and/or setting of cut-offs for aiding the decision whether the difference obtained from comparing the test and reference values is sufficiently substantial or statistically significant to be considered as a reliable confirmation of an increased Xa activity after the contacting of the sample with the polypeptide. For example, in an embodiment, a detection method can be provided, wherein the difference is only considered indicative of the presence of the anti-factor Xa DOAC above a pre-determined, pre-set, or computed threshold value.
In a specific embodiment and as already explained above, the detection method can be designed such that the reference value and the first test value are assessed and captured or quantified at one place, for example in an ambulance, and sent, e.g. over network to a device at another location e.g. in a hospital, where the step of comparing the values to find the difference between the first test value and the reference value is performed and the decision is made whether the difference, if found, is indicative of a successful detection of the DOAC inhibitor of fXa in the sample. Consequently, in a related embodiment, a detection method is provided, wherein the presence of the DOAC inhibitor of fXa in the sample obtained from the subject is indicated by the difference that is considered confirmatory to an increased fXa activity after the contacting with the polypeptide.
As the presented herein detection methods find particular use in relation to syndromes related pathology of blood coagulation as well as anticoagulation therapy, in particularly preferred embodiments, a detection method is provided, wherein the sample is a liquid biopsy sample and/or comprises blood plasma. Examples of suitable samples include plasma samples or a whole blood samples.
The presented herein detection methods can be combined with and/or aid other analytical procedures performed in relation to the subject's health, in particular, concerning blood characteristics and/or medical procedures related thereto. For example, in an embodiment, a detection method of the disclosure can be provided, further comprising the step of estimating the plasma concentration of the
DOAC inhibitor of fXa and/or the step of evaluation of anticoagulant activity in the subject's blood.
In a further advantageous embodiment, the detection method is provided, in combination with or further comprising the step of aiding a member of a medical personnel in planning the timing of surgery or other therapeutic intervention within the subject. In a further embodiment, a diagnostic method can be provided further combined with or comprising the step of aiding a member of a medical personnel in determining the suitability of the subject for thrombolytic therapy and/or administration of an inhibitor of a coagulation reversal agent, such inhibitor being a polypeptide according to any one of the disclosed herein embodiments and/or further coagulation reversal agents, for example other ones than those detected and/or administered to the subject once anticoagulation therapy would need to be restarted.
The presented herein detection methods have the advantage of being easily scalable to include multiple measurement options such as additional measurement conditions or repetitions of one measurement condition, like duplicates or triplicates of one reaction condition for achieving better reliability and avoiding false positives or false negatives. These can for example be provided in a multi- well format, including different detection polypeptides in different wells and/or at different concentrations of one or more of the polypeptides, and/or or at different concentrations of the reaction agents like the signal-generating substrates if fXa.
In line with the above, in a particularly advantageous embodiment, a detection method is provided, further comprising - contacting the sample or the second aliquot or a third aliquot of the sample with a second polypeptide capable of specifically binding to at least a further one DOAC inhibitor of fXa and assessing a second test value, wherein finding a difference between the second test value and the reference value indicates the presence of the further one DOAC inhibitor of fXa in the sample obtained from the subject.
The above described format with multiple different polypeptides of the disclosure having different DOAC-specificities and recognition properties, can easily be adapted to provide a detection method comprising the DOAC identification.
In a preferred embodiment, a detection method is provided, further comprising identification of the DOAC inhibitor of fXa present in the sample, wherein the identification comprises the contacting of the sample or the second aliquot of the sample with the polypeptide that specifically binds to exactly one DOAC inhibitor of fXa, preferably wherein the identified DOAC inhibitor of fXa is selected from apixaban, edoxaban, and rivaroxaban.
Due to the specificity of the binding between a selected polypeptide of the present disclosure and the recognised by it DOAC inhibitor of fXa, it will be apparent that finding of the difference between the reference value assessed from the untreated sample and the first test value assessed from the sample contacted with the polypeptide that specifically binds to exactly one DOAC inhibitor of fXa, can only be contributed to restoring the fXa activity through successful binding of this specific inhibitor by the polypeptide. Consequently, finding said value difference after the contacting of the sample with the polypeptide that specifically binds to exactly one DOAC inhibitor of fXa, identified this DOAC inhibitor of fXa as being present in the sample obtained from the subject.
For example, a possible embodiment of present detection method can include three different polypeptides according to the present disclosure, one specific to apixaban, the second one to edoxaban, and the third one to rivaroxaban. Each of the polypeptides is to be contacted in for example a multi-well format with an aliquot of a (plasma) sample obtained from a subject and with e.g. several solutions containing different concentrations of a signal-generating substrate of fXa, wherein each solution is provided in e.g. duplicates or triplicates (2- or 3-well repetitions per concentration condition), per each one of the three polypeptides and per reference condition (control and calibration benchmark) in which no polypeptide capable of binding a DOAC inhibitor of fXa is provided. Such exemplary design of a possible embodiment of the presented herein detection method would not only allow to identify which one or more of the DOAC inhibitors of fXa is present in the subject's blood but also would provide a dose-response curve for each one of the substrate concentrations.
In a specific embodiment, a diagnostic method is provided, wherein the polypeptide that specifically binds to exactly one DOAC inhibitor of fXa is selected from any one or more of; i. a polypeptide that specifically binds to edoxaban and comprise any one or more of the following combinations of CDRs:
GFAFDDYA (SEQ ID NO: 1), ISSSDGSTY (SEQ ID NO: 3) and AAVPRTMYSRWGCGVRPYYYGMDY (SEQID NO: 4);
GFAFDDYA (SEQ ID NO: 1), ISSSDGSTY (SEQ ID NO: 3) and AAVPRTMNSRWGCGVRPYYYGMDY (SEQ ID NO: 5);
GFAFDDYA (SEQ ID NO: 1), ISSSDGSTY (SEQ ID NO: 3) and AAVPRTMYSRWGCGVRPYYHGMDY (SEQ ID NO: 6);
GFTFDDYA (SEQ ID NO: 2), ISSSDGSTY (SEQ ID NO: 3) and AAVPRTMYSRWGCGVRPYYYGMDY (SEQ ID NO: 4);
GRTFSINV (SEQ ID NO: 7), IWWSGGASQ (SEQ ID NO: 8) and AAGPMFSMDYRRVNY (SEQ ID NO: 10);
GRTFSINV (SEQ ID NO: 7), TWWSGGASQ (SEQ ID NO: 9) and AAGPMFSMDYRRVNY (SEQ ID NO: 10);
GRTFSINV (SEQ ID NO: 7), IWWSGGASQ (SEQ ID NO: 8) and AAGPVFSMDYRRVNY (SEQ ID NO: 11);
GRTFSINV (SEQ ID NO: 7), IWWSGGASQ (SEQ ID NO: 8) and AAGPMFSMDYTRVNY (SEQ ID NO: 12);
GRTFSINV (SEQ ID NO: 7), IWWSGGASQ (SEQ ID NO: 8) and AAGPMFSMDYRRVNH (SEQ ID NO: 13);
GRTFSSYH (SEQ ID NO: 14), ITRGGGVTY (SEQ ID NO: 15) and AADAIWNQVRWMETKYTY (SEQ
ID NO: 17); or
GRTFSSYH (SEQ ID NO: 14), IARGGGVTY (SEQ ID NO: 16) and AADAIWNQVRWMETKYTY (SEQ
ID NO: 17), preferably GFAFDDYA (SEQ ID NO: 1), ISSSDGSTY (SEQ ID NO: 3) and
AAVPRTMYSRWGCGVRPYYYGMDY (SEQ ID NO: 4) ii. a polypeptide that specifically binds to apixaban and comprise any one or more of the following combinations of CDRs:
GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRASAYDD (SEQ
ID NO: 24);
GRIVGIYS (SEQ ID NO: 19), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRASAYDD (SEQ
ID NO: 24);
GRIVSTYS (SEQ ID NO: 20), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRASAYDD (SEQID NO: 24),
GRIVSIYS (SEQ ID NO: 18), ISWNGGETD (SEQ ID NO: 22) and AKPQSHFYDGSWRRASAYDD (SEQ
ID NO: 24);
GRIVSIYS (SEQ ID NO: 18), ISWNGAETQ (SEQ ID NO: 23) and AKPQSHFYDGSWRRASAYDD (SEQ
ID NO: 24};
GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFSDGSWRRASAYDD (SEQ
ID NO: 25);
GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRALAYDD (SEQ
ID NO: 26);
GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and AKPQSHFYDGSWRRASAYGD (SEQ
ID NO: 27); or
RRTFRSYA (SEQ ID NO: 28), TTWIISSTY (SEQ ID NO: 29) and AARVRSGSGQYTLPGHYDY (SEQ
ID NO: 30),
preferably GRIVSIYS (SEQ ID NO: 18), ISWNGAETD (SEQ ID NO: 21) and
AKPQSHFYDGSWRRASAYDD (SEQ ID NO: 24) iii. a polypeptide that specifically binds to rivaroxaban and comprise any one or more of the following combinations of CDRs:
TRISSLTV (SEQ ID NO: 31), LTRFGLAG (SEQ ID NO: 32) and NVKTLGGADY (SEQ ID NO: 33);
TRISSLTV (SEQ ID NO: 31), LTRFGLAG (SEQ ID NO: 32) and NAKTLGGADY (SEQ ID NO: 34);
GRTFTSYT (SEQ ID NO: 35), ISWSYWNGDSTW (SEQ ID NO: 36) and AARPSARITSRRSDYDY (SEQ ID NO: 37),
GRTFTSYT (SEQ ID NO: 35), ISWSYWNGDSTW (SEQ ID NO: 36) and VARPSARITSRRSDYDY (SEQID NO: 38);
GRTFSTLA (SEQ ID NO: 39), IRNSLSTY (SEQ ID NO: 41) and AAGRWEAVRTNTPDY (SEQ ID NO: 42);
GRTFGTLA (SEQ ID NO: 40), IRNSLSTY (SEQ ID NO: 41) and AAGRWEAVRTNTPDY (SEQ ID NO: 42);
GRTFSTLA (SEQ ID NO: 43), IRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTNTPDY (SEQ ID NO: 48);
GRTSSTLA (SEQ ID NO: 44), IRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTNTPDY (SEQ ID NO: 48);
GRTFSLLA (SEQ ID NO: 45), IRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTNTPDY (SEQ ID NO: 48);
GRTFSTLA (SEQ ID NO: 43), ITRNSISTY (SEQ ID NO: 47) and AAGRWEAVRTNTPDY (SEQ ID NO: 48);
GRTFSTLA (SEQ ID NO: 43), IRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTDTPDY (SEQ ID NO: 49); or
GRTFSTLA (SEQ ID NO: 43), IIRNSISTY (SEQ ID NO: 46) and AAGRWEAVRTITPDY (SEQ ID NO: 50), preferably TRISSLTV (SEQ ID NO: 31), LTRFGLAG (SEQ ID NO: 32) and NVKTLGGADY (SEQ ID
NO: 33).
In a related specific embodiment, a diagnostic method is provided wherein the polypeptide that specifically binds to exactly one DOAC inhibitor of fXa is selected from any one or more of: (i) a polypeptide that specifically binds to edoxaban and comprise any one or more of the of the following binding domains represented by SEQ ID NO: 51 — SEQ ID NO: 63, SEQ ID NO: 98 and SEQ ID NO: 99, preferably SEQ ID NO: 51 or SEQ ID NO: 98, more preferably SEQ ID
NO: 51 ii) a polypeptide that specifically binds to apixaban and comprise any one or more of the of the following binding domains represented by SEQ ID NO: 64 — SEQ ID NO: 78, SEQ ID NO: 100 and SEQ ID NO: 101, preferably SEQ ID NO: 64 or SEQ ID NO: 100, more preferably SEQ ID
NO: 64 (iii) a polypeptide that specifically binds to rivaroxaban and comprise any one or more of the of the following binding domains represented by SEQ ID NO: 79 — SEQ ID NO: 97, SEQ ID NO: 102 and SEQ ID NO: 103, preferably SEQ ID NO: 79 or SEQ ID NO: 102, more preferably SEQ ID
NO: 79.
Last but not least, in a further aspect related to the above described detection methods, also provided herein is a kit and/or a kit of parts for performing the detection method of the disclosure, the kit and/or the kit of parts comprising at least one or more of the polypeptide(s) as described herein, and further optionally comprising one or more reagents for assessing the reference value and/or the first test value. In a possible further embodiment, the kit and/or the kit of parts can further be provided as a multi-well plate or strip preloaded with at least some of the reagents and/or the at least one or more of the polypeptide(s), e.g. possibly in a dried and/or lyophilised form; wherein, optionally the kit comprises a chip or a micro- and/or nano-fluidic device, advantageously pre-loaded with at least some of the reagents and/or the at least one or more of the polypeptide(s).
While the invention has been described in terms of several embodiments, it is contemplated that alternatives, modifications, permutations and equivalents thereof will become apparent to one having ordinary skill in the art upon reading the specification and upon study of the drawings. The invention is not limited in any way to the illustrated embodiments. Changes can be made without departing from the scope which is defined by the appended claims.
The present invention has been described above with reference to a number of exemplary embodiments. Modifications are possible, and are included in the scope of protection as defined in the appended claims. The invention is further illustrated by the following examples, which should not be interpreted as limiting the present invention in any way.
Materials and methods
Abbreviations
FXa or fXa: activated Factor X, factor Xa
NPP: Normal plasma pool
PPP: Platelet poor plasma
TG: Thrombin generation
MATERIALS
TEST COMPOUNDS
Description Cat. Code / Product
No.
Daiichi-Sankyo EMEAMIC/I002629
Apixaban Bristol- Myers | EMEA/H/C/002148
Squibb
Chromogenic STA® liquid | Stago diagnostica | 00311 anti-Xa (LAX) test
METHODS
Immunization
Three sets of two Llamas are immunized in three rounds with a single DOAC: edoxaban, apixaban or rivaroxaban. After the last immunization (day 43), RNA is isolated from peripheral blood lymphocytes.
The integrity of the RNA was confirmed by the clear visibility of intact 283 and 18S rRNA on an 1% agarose gel.
Phage library reconstruction
A total of 40 ug RNA was transcribed into cDNA using a reverse transcriptase reaction with random hexamer primers (Thermo Fisher Scientific), followed by purification using a PCR clean-up kit (Macherey Nagel). cDNAs encoding for heavy chain immunoglobulin domains (both conventional and heavy chain-only) were amplified by PCR using proprietary primer sets. This resulted in two types of
DNA fragments (sizes of ~900 bp and ~700 bp), which represent the conventional and heavy chain- only antibody cDNAs respectively. The ~700 bp fragments (CH1-VHH of the heavy chain-only antibodies) were excised from the gel, purified and used as a template for a nested PCR that introduced flanking Sfil restriction site. Amplified nested-PCR fragments were then purified as before and subsequently digested with Sfil and BstEll. Digested VHH genes were ligated in frame with genelll into the pQ81 phagemid vector and transformed into TG1 by electroporation. To assess the transformation efficiency, the transformed TG1 were titrated using 10-fold dilutions and then spotted on LB-agar plates supplemented with 100 pg/ml ampicillin and 2% glucose. The number of transformants was estimated using the formula: # of transformants = ({# of colonies) * (dilution) * 3 (ml}) / (0.005 (ml; spotted volume))
The insert frequency was determined by randomly picking 24 clones from each library and performing a colony PCR using primers that anneal within the pQ81 vector. In this PCR, successfully inserted VHH fragments would yield PCR-fragments of ~700bp, compared to a ~300bp for empty plasmids. The insert frequency for both libraries was 100%. Libraries were grown overnight, spun down and resuspended in 2xYT medium supplemented with 20% glycerol, 2% glucose and 100 ug/ml ampicillin. OD600 was determined and libraries were stored at -80°C.
Selections of VHH domains binding to a DOAC and screening for VHH domains
Phages were produced according to established method known to the skilled person. For the 1st round of selections, 20 yl of the precipitated phages of each library were pre-blocked and pre-incubated with 10x molar excess of bovine serum albumin (BSA) and applied to wells coated with a DOAC conjugated to BSA. This corresponds to -10e9 phages per well and >10-fold the diversity of the libraries. For both libraries phages were eluted from the DOAC conjugated to BSA-coated wells. Counter selection with
BSA shows a decrease in the amount of eluted phages. Rescued outputs from the selection on 5 pg/ml of a DOAC conjugated to BSA {with counter selection with BSA) were used for phage production for the next round of selection.
For the 2nd round of selection, 1pl of the precipitated phages (pre-incubated with 10x molar excess
BSA) was applied to wells coated with a DOAC conjugated to BSA-coated. This corresponds to ~108 phages per well.
Rescued outputs of the 2nd round of selection on directly coated DOAC conjugated to BSA were plated out and single clones were picked to create master plate. From this master plate, periplasmic extracts containing monoclonal VHH were produced in deep-well plates and binding of these VHH was determined by ELISA. A large number of binders (clones) that bind to a DOAC conjugated to BSA and that do not cross-react to BSA, were obtained, showing the specific binding to the DOAC only (See
Table A; VHH sequences with SEQ ID NO: 51-97).
After careful analysis of the peri ELISA data, all clones were sequenced to determine the diversity of the VHH clones that specifically bind to a DOAC. The sequence alignment showed a diversity of at least three different VHH sequence clusters for each of the three DOACs.
VHH domain expression and purification
Representative sequences were selected and cloned in E.coli to produce VHH’s from each binding cluster.
Characterization of anti-DOAC VHH domains
Chromogenic STA® liquid anti-Xa (LAX) test (diagnostica stago) 30 pL of spiked-plasma diluted 4-fold in Owren-Koller® were mixed with 150 pL of chromogenic substrate (CBS 02.44 consisting of MAPA-Gly-Arg-pNA .HCI) and incubated during 240 sec. Then, 150
UL of bovine fXa pre-warmed at 37°C were added, starting the measurement. DOAC levels expressed as drug concentration-equivalent (ng/mL) were measured with STA-Liquid anti-Xa (Diagnostica Stago) for apixaban, edoxaban, and rivaroxaban. In brief, edoxaban, apixaban and rivaroxaban concentrations are determined by Factor Xa formation, which is measured with a factor Xa specific chromogenic substrate. Cleavage of this substrate results in factor Xa concentration dependent increased light absorption. DOAC concentration is determined from a calibration curve of a series of DOAC concentrations.
Thrombin Generation
Thrombin generation is measured with the calibrated automated thrombinogram CAT assay method invented by Synapse Research Institute and disclosed in WO2018151602 A1 and described in Hemker et al. (2009) (Hemker HC, Giesen P Al Dieri R, Regnault V, de Smedt E, Wagenvoord R, Lecompte T,
Béguin S. Calibrated automated thrombin generation measurement in clotting plasma. Pathophysiol
Haemost Thromb. 2003;33(1):4-15. doi: 10.1159/000071636. PMID: 12853707 and Coen Hemker H.,
Hemker PW, Al Dieri R. The technique of measuring thrombin generation with fluorescent substrates: 4. The H-transform, a mathematical procedure to obtain thrombin concentrations without external calibration. Thromb Haemost. 2009; 101(01): 171-177 doi: 10.1160/TH08-09-0562). Measurements are performed in triplicate while, in parallel a calibrator was tested for each plasma sample to correct for the inner filter effect, donor-to-donor variability in plasma transparency, substrate depletion, and instrumental differences. The effects of all DOACS and the DOAC binding VHH domains (also referred to as anti-dotes) were investigated in a standardized reaction mix of 5 pM TF/4 uM Phospholipids, 5% bovine serum albumin, 16.7 mM Calcium and ZGGR-AMC substrate.
All tests were done in a 96 well plate and reaction kinetics were done in a Thrombinoscope plate reader (Thrombinoscope BV, Maastricht, The Netherlands) and the kinetic data were analyzed with thrombinoscope software (Hemker H. et al., 2009). Measurements were conducted for a duration of 60 minutes, with data recorded at 20-second intervals. Standard parameters were derived and calculated from the thrombin generation curves: lag time, time to peak, peak height, .
Dose-dependent immune assay
The binding capacity of VHH’s to BSA-conjugated edoxaban, apixaban and rivaroxaban was measured by ELISA. BSA-conjugated edoxaban, apixaban or rivaroxaban were coated on an ELISA plate and a buffer with a titration of VHH concentrations of (0.01, 0.1, 1, 10, 100 and 1000 nM) was added. The lowest detectable concentration VHH determined the affinity of the VHH's.
Substrate conversion by a2M for estimating the inhibition of a DOAC by an anti-DOAC VHH
Edoxaban, apixaban, and rivaroxaban inhibit substrate conversion by FXa and by FXa bound to a2- macroglobulin (FXa-a2M). The current assay measures the reduction of FXa-a2M activity (on fluorogenic substrate ZGGR-AMC) by the presence of DOAC in the tested sample. The amount of activity inhibition is directly correlated to the amount of edoxaban, apixaban or rivaroxaban in the sample.
The test consists of two FXa-a2M solutions (30 and 90 nM) which are used respectively to measure low (50-500 nM) and high (500-1000 nM) plasma edoxaban, apixaban or rivaroxaban concentrations.
In each experiment, a dose-response curve is measured for both FXa-a2M solutions. Each well (calibration curve or testing well) contains 20 uL FXa-a2M (either 30 or 90 nM), 80 uL of 1:8 prediluted plasma, and 20 ul of ZGGR-AMC in BSA60 (2.5 mM) is added just prior to the measurement.
In each experiment, two dose-response curves are measured; one at 30 nM FXa-a2M (50-500 nM edoxaban, apixaban or rivaroxaban) and the other at 90 nM FXa-a2M (500-1000 nM Rivaroxaban).The dose-response series are prepared in pooled normal plasma to correct for the possible influence of the presence of plasma in the measurement wells. In addition, up to 18 test samples can be measured. All sample measurements are performed in duplicate and at two FXa-a2M concentrations (4 wells per sample).
Binding experiment showing that the bivalent polypeptide comprising a DOAC inhibiting VHH and an anti-albumin VHH binds to human serum albumin and inhibits the targeted DOAC.
To modulate (increase) the half-life of a VHH domain, such VHH can be linked (e.g. at the cDNA level, which is preferred) with an albumin binding protein or (antibody) domain such as (preferably) an albumin binding VHH. DOAC binding VHH domains were linked with a human serum albumin binding VHH a the cDNA level, and polypeptides comprising such DOAC binding VHH domain and the human serum albumin binding VHH domain were obtained at the protein level. These bi-specific polypeptides were tested for their binding to DOAC, and this binding was compared to the binding of the DOAC binding
VHH. In addition, inhibitory activity of the bi-specific polypeptides was tested when inhibition of factor
Xa inhibition by a DOAC is concerned. For this purpose, thrombin generation tests were performed as here-above outlined. The amino-acid sequences of such bivalent polypeptides (SEQ ID NO: 98, SEQ
ID NO: 100, SEQ ID NO: 102), as well as the sequences of the human serum albumin binding sdab (VHH) (SEQ ID NO: 108) and the CDR1-3 amino-acid sequences of said sdab (SEQ ID NO: 105-107 for CDR1, CDR2, CDR3, respectively) are displayed in the Table A, provided here below.
Multivalent polypeptides comprising multiple copies of a DOAC binding VHH
One way of improving efficacy of reversal of fXa inhibiting activity of a DOAC is by providing a polypeptide comprising multiple copies of VHH domains that specifically bind to said DOAC and that inhibit the fXa inhibiting activity of said DOAC. To that end, bivalent polypeptides comprising two copies of VHH domains specifically binding to a DOAC, were produced, based on VHH domains selected for their specific binding to a DOAC and their effective inhibition of the DOAC. Such mono-specific bivalent polypeptides prepared and tested are edoxaban inhibiting VHH-VHH with clone id edo_23 (SEQ ID NO: 99), apixaban inhibiting VHH-VHH tandem with clone id api_32 (SEQ ID NO: 101) and rivaroxaban inhibiting VHH-VHH tandem with clone id riva_40 (SEQ ID NO: 103). Also refer to the Table A, here below, for the VHH sequences and the CDR1, CDR2, CDR3 sequences and the linker sequence linking the two VHH domains together (SEQ ID NO: 104). As will be understood by those skilled in the art, equally suitable are polypeptides comprising more than two copies of the same VHH domain, or for example a linear string of multiple DOAC inhibiting VHH domains, wherein the VHH domains either bind to the same DOAC and have different amino-acid sequences, or bind to two or more different DOACs (multi-specific polypeptides suitable for inhibiting more than one DOAC, or suitable for detecting presence of more than one DOAC in e.g. patient plasma samples).
Determining the specificity of the DOAC binding of VHH domains capable of binding to a DOAC and capable of inhibiting the fXa inhibiting activity of a DOAC, and determining the affinity of
DOAC binding VHH’s for DOAC binding, as established with surface plasmon resonance (SPR) measurements
DOACs edoxaban, apixaban, rivaroxaban and betrixaban were immobilized on SPR chips and binding of VHH's selected for their binding to a DOAC as established in an ELISA set-up, was established.
Specificity of the binding of a VHH selected for binding to a first DOAC, was established by testing the binding to said first DOAC and the binding to the further three DOACSs. In addition, association constant, dissociation constant as well as the Ko were determined.
RESULTS
After immunization of Llama’s with fXa inhibiting DOACs edoxaban, apixaban or rivaroxaban, clones expressing specific DOAC binding and inhibiting VHH domains were obtained for each of the three different DOACs as here above described.
In the Table A here below, SEQ ID NO's are given for all clones providing VHH’s that specifically bind to the indicated DOAC. Here, ‘edo’ refers to VHH’s specifically binding to edoxaban, ‘api’ refers to VHH's specifically binding to apixaban and ‘riva’ refers to VHH's specifically binding to rivaroxaban.
Table A: SEQ ID NO:'s and clone ID's for VHH's that specifically bind to edoxaban (‘edo_nn’), apixaban (‘api_nn’) or rivaroxaban (‘riva_nn’), and CDR1-3 amino-acid sequences present in those VHH’s.
Clone =F ee ope. pee FP
Memeweenwenwew 10 peewee PF een
Ow nnn
Ee
Ee PF
Ee i A
I i A
I
I i pee mee pee emer" ope. nnn"
I
I
I
Ien
Mee
AKPQSHFYDGSWRRASAYDD 24
Ke FF
Ke F ee 7 oo..." oe esse PF oe
Oe ees eee oe. meee PF ewer eee PF oo. nnn" oe." mer emewswe 1"
Oe
Ee
On ome ome fees F
AAGRWEAVRTITPDY 50 edo_1 EVOLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSC
ISSSDGSTYYADSVKGRFTISSDNAKNTVYLQMNSLRPEDTAVYYCAAVPRT | 51
MYSRWGCGVRPYYYGMDYWGKGTLVTVSS edo 2 EVQLVESGGGLVQAGGSLRLSCAASGFTFDDYAIGWFRQAPWKEREGVSC
ISSSDGSTYYADSVKGRFTISSDNAKNTVYLQMNSLRPEDTAVYYCAAVPRT | 52
MYSRWGCGVRPYYYGMDYWGKGTLVTVSS edo 3 EVOLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPGKEREGVSCI
SSSDGSTYYADSVKGRFTISSDNAKNTVYLQMNSLRPEDTAVYYCAAVPRT | 53
MYSRWGCGVRPYYYGMDYWGKGTLVTVSS edo 4 EVQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSC
ISSSDGSTYYADSVKGRFTISSDNAKNTVYLQMNSLRPEDTAVYYCAAVPRT | 54
MNSRWGCGVRPYYYGMDYWGKGTLVTVSS
EVQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSC
ISSSDGSTYYADSVKGRFTISSDNAKNTVYLQMNSLRPEDTAVYYCAAVPRT | 55 edo 5 MYSRWGCGVRPYYHGMDYWGKGTLVTVSS
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAI
WWSGGASQYADSVKGRFSISKDNAKNTMFLQMNSLKPEDTAVYYCAAGP | 56 edo_6 MFSMDYRRVNYWGQGTQVTVSS
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAT
WWSGGASQYADSVKGRFSISKDNAKNTMFLGMNSLKPEDTAVYYCAAGP | 57 edo_7 MFSMDYRRVNYWGQGTQVTVSS
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAI
WWSGGASQYADSVKGRFSISKDNAKNMMFLQMNSLKPEDTAVYYCAAGP | 58 edo_8 MFSMDYRRVYNYWGQGTQVTVSS
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAI
WWSGGASQYADSVKGRFSISKDNAKNTMFLQMNSLKPEDTAVYYCAAGPV | 59 edo_9 FSMDYRRVNYWGQGTQVTVSS
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAI
WWSGGASQYADSVKGRFSISKDNAKNTMFLOMNSLKPEDTAVYYCAAGP edo_10 | MFSMDYTRVNYWGQGTQVTVSS
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAI
WWSGGASQYADSVKGRFSISKDNAKNTMFLQMNSLKPEDTAVYYCAAGP | 61 edo_11 | MFSMDYRRVNHWGQGTQVTVSS
EVQLVESGGGLVQAGGSLRLSCTASGRTFSSYHMAWFRQAPGKERESVAA
ITRGGGVTYYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAADAIW | 62 edo 12 | NQVRWMETKYTYSGQGTQOVTVSS
EVQLVESGGGLVQAGGSLRLSCTASGRTFSSYHMAWFRQAPGKERESVAA
IARGGGVTYYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAADAI 63 edo_13 | WNQVRWMETKYTYSGQGTQVTVSS api_1 EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 64
SHFYDGSWRRASAYDDWGQGTQVTVSS api_2 EVQLVESGGGLVRAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 65
SHFYDGSWRRASAYDDWGQGTQVTVSS api_3 EVQLVESGGGLVEAGGSLRLSCAASGRIVSIYSMAWFRGAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ
SHFYDGSWRRASAYDDWGQGTQVTVSS api 4 EVQLVESGGGLVOAGGSLRLSCAAPGRIVSIYSMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 67
SHFYDGSWRRASAYDDWGQGTQVTVSS api_5 EVQLVESGGGLVQAGGSLRLSCAASGRIVGIYSMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ
SHFYDGSWRRASAYDDWGQGTQVTVSS
EVQOLVESGGGLVQAGGSLRLSCAASGRIVSTYSMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ
SHFYDGSWRRASAYDDWGQGTQVTVSS api_7 EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAYFRQAPGKVREFVAAIS
WNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQS | 70
HFYDGSWRRASAYDDWGQGTQVTVSS api_8 EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAI
SWNGGETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 71
SHFYDGSWRRASAYDDWGQGTQVTVSS
EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAI
SWNGAETQYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 72
SHFYDGSWRRASAYDDWGQGTQVTVSS api_10 EVQLVESGGGLVQAGGSLRLSCAASGRIVSIY SMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 73
SHSYDGSWRRASAYDDWGQGTQVTVSS api_11 EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 74
SHFYDGSWRRALAYDDWGQGTQVTVSS api_12 EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 75
SHFYDGSWRRASAYGDWGQGTQVTVSS api_13 EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 76
SHFYDGSWRRASAYDDCGQGTQVTVSS api_14 EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ | 77
SHFYDGSWRRASAYDDWCQGTQVTVSS
EVQLVESGGGLVOAGGSLRLSCAASRRTFRSYATAWFRQAPGKERVFVAG api_15 TTWIISSTYYADSVNGRFTISRDNAENTVYLQMNSLKPEDTAVYYCAARVRS | 78
GSGQYTLPGHYDYWGQGTQVTVSS riva_1 EVOLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRGAPGEQREVVATL
TRFGLAGYADSVKGRFTISADHAKLTLYLHMNNLKPADTAVYYCNVKTLGGA | 79
DYWGQGTQVTVSS riva_2 EVQLVESGGDSVOPGGSLRISCTASTRISSLTVMGWYRQAPGVOREVVATL
TRFGLAGYADSVKGRFTISADHAKLTLYLHMNNLKPADTAVYYCNVKTLGGA
DYWGQGTQVTVSS riva_3 EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATL
TRFGLAGYADPVKGRFTISADHAKLTLYLHMNNLKPADTAVYYCNVKTLGGA | 81
DYWGOGTQVTVSS riva_4 EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATL
TRFGLAGYADSVKDRFTISADHAKLTLYLHMNNLKPADTAVYYCNVKTLGGA | 82
DYWGQGTQVTVSS riva_5 EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATL
TRFGLAGYADSVKGRFTISADHAKLTLYLHMNNLKPADTAVYYCNAKTLGGA | 83
DYWGQGTQVTVSS riva_6 EVQLVESGGGLVQAGGSLRLSCAASGRTFTSYTLGWFRQAPEKEREFVGG
ISWSYWNGDSTWYADSVKGRFTVSTDNAKKTAYLQMNSLKPEDTAVYCAA | 84
RPSARITSRRSDYDYWGQGTQVTVSS riva_7 EVQLVESGGGLVQAGGSLRLSCAASGRTFTSYTLGWFRQAPEKEREFVGG
ISWSYWNGDSTWYADSVKGRFTVSTDNAKKTAYLQMNSLKPEDTAVYCVA | 85
RPSARITSRRSDYDYWGQGTQVTVSS riva_8 EVQLVESGGGLVQAGGSLKLSCAASGRTFSTLAMGWFRQAPGKEREFVAG
IRNSLSTYYSDSVKGRFTISGDNAKNTVYLQMNSLNHEDTAVYYCAAGRWE
AVRTNTPDYWGQGTQVTVSS riva_9 EVOLVESGGGLVQAGGSLKLSCAASGRTFGTLAMGWFRQAPGKEREFVA
GIIRNSLSTYYSDSVKGRFTISGDNAKNTVYLQMNSLNHEDTAVYYCAAGR 87
WEAVRTNTPDYWGQGTQVTVSS riva_10 | EVQLVESGGGLVQAGGSLKLSCAASGRTFSTLAMGWFRQAPGKEREFVAG
IRNSLSTYYSDAVKGRFTISGDNAKNTVYLQMNSLNHEDTAVYYCAAGRWE | 88
AVRTNTPDYWGQGTQVTVSS riva_11 EVQLVESGGGLVOAGGSLKLSCAASGRTFSTLAMGWFRQAPGKEREFVAG
IRNSLSTYYSDSVKGRFTISGGNAKNTVYLQMNSLNHEDTAVYYCAAGRW
EAVRTNTPDYWGQGTQVTVSS riva_12 | EVQLVESGGGLVQOAGGSLRLSCAASGRTFSTLAWGWFRGAPGKEREFVA
GIIRNSISTYYSDSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAAGRW
EAVRTNTPDYWGQGTQVTVSS riva_13 | EVOLVESGGGLVQAGGSLRLSCAASGRTSSTLAWGWFRQAPGKEREFVA
GIIRNSISTYYSDSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAAGRW | 91
EAVRTNTPDYWGQGTQVTVSS riva_14 | EVQLVESGGGLVQAGGSLRLSCAASGRTFSLLAWGWFRQAPGKEREFVAG
IRNSISTYYSDSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAAGRWE | 92
AVRTNTPDYWGQGTQVTVSS riva_15 | EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRGAPGKEREFVA
GITRNSISTYYSDSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAAGRW | 93
EAVRTNTPDYWGQGTQVTVSS riva_16 | EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRQAPGKEREFVA
GIIRNSISTYYSDSVKGRFTISRDNAKNMVYLQMNSLKPEDTAVYYCAAGRW | 94
EAVRTNTPDYWGQGTQVTVSS riva_17 | EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRQAPGKEREFVA
GIIRNSISTYYSDSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYHCAAGRW | 95
EAVRTNTPDYWGQGTQVTVSS fiva_18 | EVGLVESGGGLVGAGGSLRLSCAASGRIFSTLAWGWERGAPGKEREFVA
GIIRNSISTYYSDSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAAGRW
EAVRTDTPDYWGQGTQVTVSS fiva_19 | EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRQAPGKEREFVA
GIIRNSISTYYSDSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAAGRW | 97
EAVRTITPDYWGQGTQVTVSS
EVQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSC
ISSSDGSTYYADSVKGRFTISSDNAKNTVYLQMNSLRPEDTAVYYCAAVPRT
MYSRWGCGVRPYYYGMDYWGKGTLVTVSSSAAGGGGSGGGGSAAAQV
94922 | QLQESGGGLVOAGGSLRLSCAASGYISDAYYMGWYRQAPGKEREFVATIT
HGTNTYYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAVLETRSY
SFRYWGQGTQVTVSSLEHHHHHH
EVQLVESGGGIVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSC
ISSSDGSTYYADSVKGRFTISSDNAKNTVYLQMNSLRPEDTAVYYCAAVPRT
MYSRWGCGVRPYYYGMDYWGKGTLVTVSSSAAGGGGSGGGGSAAA
94023 | EyYQLVESGGGLVOAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSC
ISSSDGSTYYADSVKGRFTISSDNAKNTVYLQMNSLRPEDTAVYYCAAVPRT
MYSRWGCGVRPYYYGMDYWGKGTLVTVSS
EVQLVESGGGLVQAGGSLRLSCAASGRIVSIY SMAWFRQAPGKVREFVAA
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ
SHFYDGSWRRASAYDDWGQGTQVTVSSAAGGGGSGGGGSAAAQVQLGE oP 31 | SGGGLVQAGGSLRLSCAASGYISDAYYMGWYR@APGKEREFvATITHGTN | 19
TYYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAVLETRSYSFRY
WGQGTOVTVSSLEHHHHHH
EVALVESGGGLVQAGGSLRLSCAASGRIVSIY SMAWFRQAPGKVREF VAAL
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ
Opi 32 SHFYDGSWRRASAYDDWGQGTQVTVSSAAGGGGSGGGGSAAA 01 7 EVOLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFROAPGKVREFVAAI
SWNGAETDYVDYVQGRFTASRDNAKNTMYLQMNNLKPEDTATYYCAKPQ
SHFYDGSWRRASAYDDWGQGTQVTVSS
EVAQLVESGGDSVOPGGSLRISCTASTRISSLTVMGWYRQAPGEGREVVATL rva_39 | TREGLAGYADSVKGRFTISADHAKLTLYLHMNNLKPADTAVYYCNVKTLGGA | 102
DYWGQGTQVTVSS
AAGGGGSGGGGSAAAQVQLQESGGGLVQAGGSLRLSCAASGYISDAYYM
GWYRQAPGKEREFVATITHGTNTYYADSVKGRFTISRDNAKNTVYLQMNSL
KPEDTAVYYCAVLETRSYSFRYWGQGTQVTVSSLEHHHHHH
EVOLVESGGDSVOPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATL
TRFGLAGYADSVKGRFTISADHAKLTLYLHMNNLKPADTAVYYCNVKTLGGA va 40 DYWGQGTOVTVSSAAGGGGSGGGGSAAA 103 riva 7 EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATL
TRFGLAGYADSVKGRFTISADHAKLTLYLHMNNLKPADTAVYYCNVKTLGGA
DYWGQGTQVTVSS
Be AAGGGGSGGGGSAAA
QVQLQESGGGLVQAGGSLRLSCAASGYISDAYYMGWYRQAPGKEREFVAT
ITHGTNTYYADSVKGRFTISRDNAKNTVYLQMNSLKPEDTAVYYCAVLETRS | 108
YSFRYWGQGTQVTVSSLEHHHHHH
SEQ ID NO: 104 relates to the linker sequence applied for linking a DOAC binding VHH with a human serum albumin binding VHH (SEQ ID NO: 108, with CDR1, CDR2 and CDR3 with sequences according to SEQ ID NO: 105-107, respectively, and with a C-terminal 6x His-tag), providing bispecific VHH-VHH tandems (SEQ ID NO: 98 (edo_22), SEQ ID NO: 100 (api_31) and SEQ ID NO: 102 (riva_39). The same linker sequence with SEQ ID NO: 104 is also introduced in the bivalent tandem of two identical
VHH's binding to the same DOAC, linking the two VHH's together: for edoxaban binding tandem, SEQ
ID NO: 99 (edo_23), for apixaban binding, SEQ ID NO: 101 (api_32), and for rivaroxaban binding, SEQ
ID NO: 103 (riva_40).
Afterimmunization with edoxaban and selection of clones expressing edoxaban binding VHH's, amino- acid sequence alignment revealed three clusters of similar VHH sequences, with highly homologous
CDRs and highly homologues VHH amino-acid sequences. VHH domains with SEQ ID NO: 51-55 form a first cluster; SEQ ID NO: 56-61 are a second cluster; SEQ ID NO: 62 and 63 are a third cluster. In
Table B, the SEQ ID NO's for the CDR1, CDR2 and CDR3 sequences (see Table A) are given for each edoxaban binding VHH, per cluster.
Table B: clusters of VHH specifically binding to edoxaban [ene Jee [ee [wade [Sam [ew [ewe jem ww
Swe en] eee
CE Em ees ree
TE mE ees ower eee ee
EEE |E fees
Ce wees ee
EE [ee asf [OL eee] eee
After immunization with apixaban and selection of clones expressing apixaban binding VHH's, amino- acid sequence alignment revealed two clusters of similar VHH sequences, with highly homelogous
CDRs and highly homologues VHH amino-acid sequences. VHH domains with SEQ ID NO: 64-77 form a first cluster; SEQ ID NO: 78 is a second cluster. In Table C, the SEQ ID NO’s for the CDR1, CDR2 and CDR3 sequences (see Table A} are given for each apixaban binding VHH, per cluster.
Table C: clusters of VHH specifically binding to apixaban
Joe Jeane [ease Jom om om jam wn oewp
KH NO NN eea] eea
EEE
B EL; mms om ww Jes
Eee
OE [aas
Ee
BE ea
B mm Jme
EEE ae] eee
After immunization with rivaroxaban and selection of clones expressing rivaroxaban binding VHH's, amino-acid sequence alignment revealed four clusters of similar VHH sequences, with highly homologous CDRs and highly homologues VHH amino-acid sequences. VHH domains with SEQ ID
NO: 79-83 form a first cluster; SEQ ID NO: 84 and 85 is a second cluster; SEQ ID NO: 86-89 form a third cluster; SEQ ID NO: 90-97 form a fourth cluster. In Table D, the SEQ ID NO’s for the CDR1, CDR2 and CDR3 sequences (see Table A for the sequences) are given for each rivaroxaban binding VHH, per cluster.
Table D: clusters of VHH specifically binding to rivaroxaban [ene Tew Jensen] [eNom Jew [ww [ced ewe Oe
Ee
EE ee ee
TETE awe
CE wee eee
CRT aas] [me Je Je | [as ee eee oe [ee [EO err owed | |] sm mw en
Cw ms er wen
Tee
Emm me
Emm
EE mE me
Cm www wee ee
EDOXABAN
Dose-dependent immune assay for estimating the affinity of VHH binding fo a DOAC
Table 1 Selection of representative VHH's in the binding clusters 1-3 for binding to edoxaban
Relative max binding (OD) | Affinity (nM)
Ke a | 8
In Table 1, for five representative VHH domains capable of specifically binding to edoxaban, the affinity in nM is presented, as determined with an ELISA.
Diagnostic anti factor Xa (DOAC) test of Stago diagnostica to investigate the anti-DOAC function of edoxaban-binding Vin
The figures 1A, 1B, 1C and 1D show the results obtained with the Chromogenic STA® liquid anti-Xa (LAX) test (diagnostica stago) and VHH obtained from clone edo_1, specific for edoxaban binding. The reference measurement ‘NPP’ in each of figures 1A-D is NPP without edoxaban and without edo_1 (plasma (no edoxaban)). Measurements in figures 1A-D concern plasma which contains 25 nM, 50 nM, 400 nM or 1000 nM edoxaban, respectively, with the indicated concentrations series of edo_1 (0, 25, 50, 100 nM edo_1 for figure 1A; 0, 50, 100, 200 nM edo-1 for figure 1B; 0, 400, 800, 1600 nM edo_1 for figure 1C; 0 (‘'edoxaban’), 1000, 2000, 4000 nM edo _1 for figure 1D). The results illustrate that 25 nM, 50 nM, 400 nM or 1000 nM edoxaban inhibits the Chromogenic STA® liquid anti-Xa (LAX) test.
Furthermore, addition of edo_1 completely restores the fXa inhibitory effects of edoxaban; adding edo_1 to NPP spiked with edoxaban leads to a return of the thrombin generation as determined with the
Chromogenic STA® liquid anti-Xa (LAX) test to the reference value measured with NPP without edo_1 and without edoxaban spike in the Chromogenic STA® liquid anti-Xa (LAX) test. Typically, about a four times higher concentration of edo_1 is sufficient for complete inhibition of the fXa inhibitory effect of a dose of edoxaban added to NPP, at all edoxaban concentrations tested.
Thrombin Generation
The figure 2 shows the thrombin generation curves. The reference thrombin generation curve is plasma without edoxaban and without edo 1 (plasma (no edoxaban)). All the further samples analyzed contain 300 nM edoxaban with indicated variable concentrations of edo_1 (0, 300, 600, 1200, 2400 nM). The results illustrate that 300 nM edoxaban inhibits the thrombin generation (peak shifts to the right and the peak is lower). Furthermore, addition of edo 1 completely restores the effects of edoxaban; all concentrations of edo_1 lead to a return of the thrombin generation curves into the same pattern as the reference thrombin generation curves, indicative for complete inhibition of the fXa inhibitory effect of edoxaban.
The figure 3 shows the lag times for thrombin generation, derived from thrombin generation curves obtained with plasma samples from three independent human donors. The reference lag time is derived from human plasma without edoxaban spike and without edo_1 (plasma). All the other bars relate to plasma samples that contain 300 nM edoxaban with indicated variable concentrations of edo_1 (0, 300, 600, 1200, 2400 nM). The results illustrate that 300 nM edoxaban increases the lag time almost threefold. Furthermore, addition of edo_1 already at a 1 : 1 molar ratio between edo_1 and edoxaban completely restores the thrombin generation and neutralizes the fXa inhibitory effects of edoxaban.
The figure 4 shows the Peak Thrombin values, derived from thrombin generation curves from three independent healthy human donors. The reference Peak value (‘plasma’) is derived from human plasma without edoxaban and without edo_1 (plasma). All the other bars relate to the same human plasma sample now spiked withn 300 nM edoxaban and with indicated variable concentrations of edo 1 (0, 300, 600, 1200, 2400 nM). The results illustrate that 300 nM edoxaban decreases the Peak Thrombin
Generation. Furthermore, addition of edo_1 already at a 1 : 1 molar ratio between edo_1 and edoxaban completely restores the Peak Thrombin value and effectively neutralizes or reverses the fXa inhibitory effects of edoxaban.
The figure 5 shows the thrombin generation curves obtained with testing the edoxaban neutralizing activity of the bi-specific polypeptide ‘edo_22’ (SEQ ID NO: 98) comprising the first VHH edo_1 (SEQ
ID NO: 51) and the second VHH which specifically binds human serum albumin (SEQ ID NO: 108) linked together via amino-acid linker sequence with SEQ ID NO: 104. The reference thrombin generation curve is obtained with plasma without edoxaban and without edo 22 (plasma (no edoxaban)). All the other curves are measured with plasma containing 300 nM edoxaban with indicated variable concentrations of edo 22 (0, 300, 600, 1200, 2400 nM). The results illustrate that 300 nM edoxaban inhibits the thrombin generation (peak shifts to the right and the peak is lower). Furthermore, addition of 4 fold excess edo _ 22 (1200 nM) completely restores the thrombin generation and reverses the inhibitory effects of edoxaban on fXa; 4 fold excess concentrations of edo_22 lead to a return of the thrombin generation curves into the same thrombin generation pattern as the reference thrombin generation curves.
The figure 6 shows the lag times for thrombin generation, derived from thrombin generation curves obtained with a plasma sample obtained from three independent healthy human donors. The reference lag time is derived with human plasma without edoxaban and without edo_22 (referred to as ‘plasma’).
For details on the edo_22 polypeptide, refer to the description relating to figure 5, here above. All the other lag time measurements are performed with the human plasma containing 300 nM edoxaban with indicated variable concentrations of edo 22 (0, 300, 600, 1200, 2400 nM). The results illustrate that 300 nM edoxaban increases the lag time about threefold. Furthermore, addition of edo_22 restores the lag time to a value similar to the lag time obtained with the human plasma control. The fXa inhibiting effects of edoxaban are inhibited and reversed in a dose dependent manner, and at a 4 fold excess concentration (1200 nM), edo_22 completely compensated (restored) the lag time to the same value as the reference lag time. This shows that edo_22 effectively inhibits the fXa inhibiting effect of the
DOAC.
The figure 7 shows the Peak Thrombin values, derived from thrombin generation curves from three independent donors. The reference Peak Thrombin values is derived from plasma without edoxaban and without edo_ 22 (plasma). All the other bars are obtained with human plasma samples that contain 300 nM edoxaban with indicated variable concentrations of edo_22 (0, 300, 600, 1200, 2400 nM). The results illustrate that 300 nM edoxaban decreases the Peak Thrombin Generation more than fourfold.
Furthermore, addition of edo 22 restores the thrombin generation to a level also seen for the plasma control, and neutralizes the fXa inhibiting effects of edoxaban. At a 8 fold excess concentration (2400 nM), edo_22 completely compensated the Peak thrombin values to the same value as the reference
Peak Thrombin values. This shows that edo_22 effectively inhibits the fXa inhibiting effect of the DOAC.
All the above results show that a VHH binding specifically to edoxaban and a bispecific VHH binding specifically both to edoxaban and human serum albumin are equally potent when inhibiting the DOAC, namely edoxaban, is considered. At a molar ratio of 1:1 — 1:8 for antidote versus DOAC, effective reversal of the fXa inhibiting activity of the DOAC is established.
Edoxaban Treated Patients
Edo 1 in plasma samples of human patients, treated with edoxaban
The figure 8A illustrates the thrombin generation lag time in plasma of human patients who are on treatment with edoxaban, without spiked edo 1 (‘plasma’), with 400 nM edo 1, spiked to plasma and with 1600 nM edo _1, spiked to plasma.
The figure 8B illustrates the thrombin generation lag time in plasma of patients who are on treatment with edoxaban, without spiked edo_1 (‘PPP’), with 400 nM edo_22 (bispecific), spiked to plasma and with 1600 nM edo_1, spiked to plasma.
The figure 9A illustrates the Peak Thrombin values with said plasma of human patients who are on treatment with edoxaban, without spiked edo _1, with 400 nM edo _1, spiked to plasma and with 1600 nM edo_1, spiked to plasma.
The figure 9B illustrates the Peak Thrombin values with said plasma of patients who are on treatment with edoxaban, without spiked edo_1, with 400 nM edo_22 (bispecific), spiked to plasma and with 1600 nM edo_1, spiked to plasma.
It is observed that both edo_1 and edo_22 are able to compensate lag time in patients treated with edoxaban independently of the levels of edoxaban in their plasmas.
Since the DOAC neutralizing effect was already established within 10 minutes after mixing the plasma with the polypeptide edo_1 or edo_22, these examples are illustrative for a rapid and complete DOAC neutralizing effect in the patient's circulation when the polypeptide is administered to the patient intravenously at a dose sufficient to establish a concentration of 400 — 1600 nM of the polypeptide in the plasma.
Synthesis of a bivalent molecule comprising two edoxaban binding VHH domains
A bivalent molecule riva_23 was synthesized on the cDNA level and on the protein level (SEQ ID NO: 99), comprising two linked VHH domains edo_1 (SEQ ID NO: 51). Such a bivalent molecule is suitable for increasing the half-life of the molecule in the human blood circulation. The bivalent VHH is about twice as large as a single VHH domain and therewith increases the IC50 for clearance from the circulation. In addition, the bivalent molecule can bind two copies of edoxaban, therewith having an increased binding capacity at the same molar dose. For, for example, purification purposes, the bivalent molecule is provided with a His-tag such as a C-terminal His-tag, such as a 6x His-tag.
A bi-valent VHH may be bi-specific and mono-specific. A bi-specific VHH may be bi-specific for different targets or for different epitopes on the same target. Typically, the bi-valent VHH tandem is specific for the same DOAC, and comprises two copies of the same VHH specific for the same DOAC, or comprises a first VHH specific for a DOAC and a second VHH different from the first DOAC and also specific for the same DOAC to which the first VHH is capable of specifically binding.
A bi-valent VHH directed to the same epitope (the same DOAC) may improve the binding to the target because binding of the target (the DOAC) (especially to small VHHs) is transient; therefore there is always an equilibrium between bound (e.g. to the selected DOAC) and unbound VHHs and targets (free molecules of selected DOAC). Bi-valent VHHs can improve the binding-equilibrium between VHH and target (selected DOAC to which the VHH specifically binds) for example (and without wishing to be bound by any theory) because the target (selected DOAC) will be exchanged between the two binding sites provided by the two VHH domains.
A bi-valent/bi-specific VHH is prepared to bind the VHH to a carrier-protein (albumin). This improves the pharmacokinetics of the VHH.
APIXABAN
Dose-dependent immune assay for estimating the affinity of VHH binding to a DOAC
Table 2 Selection of representative VHH’s in the binding clusters 1-3 for binding to apixaban
Relative max binding (OD) Affinity (nM)
In Table 2, for a representative VHH domain capable of specifically binding to apixaban, the affinity in nM is presented, as determined with an ELISA.
Diagnostic anti factor Xa (DOAC) test of Stago diagnostica to investigate the anti-DOAC function (inhibitory activity towards fXa inhibition by the DOAC) of apixaban-binding Vun
The figure 10 shows the results obtained with the Chromogenic STA® liquid anti-Xa (LAX) test (diagnostica stago), with human plasma control (‘plasma’), plasma spiked with 1088 nM apixaban, and plasma spiked with 1088 nM apixaban and a concentration series of either polypeptide api_1 (SEQ ID
NO: 64) which specifically binds to apixaban, or polypeptide api_31 (SEQ ID NO: 100) which specifically binds to apixaban and to human serum albumin (bi-specific polypeptide comprising 2 VHH domains, the first VHH domain being api_1 with SEQ ID NO:64 and the second VHH domain being the albumin- binding VHH with SEQ ID NO: 108). The reference measurement is performed with NPP human plasma without apixaban and without api_1 or api_31 (‘plasma’). All the other plasma samples contain 1088 nM apixaban with indicated variable concentrations of api_1 or api_31. The results illustrate that both api_1 and api_31 inhibit the Chromogenic STA® liquid anti-Xa (LAX) test measurement, as indicated with the apixaban concentration (nM). Furthermore, addition of a fourfold, eightfold or sixteen-fold molar excess api_1 or api_31 almost completely restores the fXa inhibitory effects of apixaban; all tested concentrations of api_1 and api_31 resulted in at least partial reversal of the Chromogenic STA® liquid anti-Xa (LAX) test result into the direction of the reference Chromogenic STA® liquid anti-Xa (LAX) test value.
Thrombin Generation
The figure 11 shows the thrombin generation curves obtained with control plasma, plasma spiked with 400 nM apixaban, and plasma spiked with 400 nM apixaban, to which a concentration series of polypeptide api_1 is added. The reference thrombin generation curve is obtained for human plasma without apixaban and without api_1 (plasma (no apixaban)). All the other curves are obtained with human plasma samples that contain 400 nM apixaban with the indicated variable concentrations of api_1. The results illustrate that 400 nM apixaban inhibits the thrombin generation (peak shifts to the right and the peak is lower). Furthermore, addition of 8 fold excess api_1 (3200 nM) completely restores the thrombin generation (reverses fXa inhibition) and reverses the fXa inhibiting effects of apixaban; 8 fold molar excess concentrations of api_1 leads to a return of the thrombin generation curves to a level that is the same as the thrombin generation obtained with the reference plasma sample.
The figure 12 shows the lag times, derived from thrombin generation curves from a plasma sample obtained from three independent healthy human donors. The reference lag time is derived from plasma without apixaban and without api_1 (plasma). All the other lag time measurements are obtained with the plasma sample that now contains 400 nM apixaban with the indicated variable concentrations of api_1. The results illustrate that 400 nM apixaban increases the lag time more than two-fold.
Furthermore, addition of api_1 restores the fXa activity as displayed by the lag time for thrombin generation reaching values comparable to the value obtained for the control human plasma sample.
That is to say, the api_1 effectively inhibits the fXa inhibiting effects of apixaban, and at a 8 fold molar excess concentration (3200 nM), api_1 completely compensated the prolonged lag time under influence of the DOAC back to the short lag time which is the same as the reference value as obtained with the reference plasma sample without DOAC.
The figure 13 shows the peak thrombin values, derived from thrombin generation curves with the same healthy human plasma sample obtained from three independent donors. The reference peak thrombin value is derived from plasma without apixaban and without api 1 (plasma). All the other thrombin peak measurements are determined with the plasma sample that contains 400 nM apixaban with indicated variable concentrations of api_1. The results illustrate that 400 nM apixaban decreases the peak thrombin values. Furthermore, addition of api_1 restores the Peak Thrombin value back to the Peak
Thrombin value measured for the positive control, i.e. the plasma sample that is not spiked with the
DOAC. That is to say, the fXa inhibiting effects of apixaban is reversed under influence of the polypeptide api_1, and at a 8 fold molar excess concentration (3200 nM), ap_1 completely compensated (restored) the peak thrombin values to the same extent as the peak thrombin value obtained with the reference plasma sample.
The figure 14 shows the thrombin generation curves obtained with control healthy human donor plasma (plasma (no apixaban)’), and the plasma spiked with 400 nM apixaban and a concentration series of bivalent polypeptide api_31 comprising an apixaban binding VHH and a human serum albumin binding
VHH (SEQ ID NO: 100), as outlined here above. The reference thrombin generation curve is obtained with human plasma sample without apixaban and without api_31 (plasma (no apixaban). All the other curves are obtained by measuring thrombin generation with the plasma sample containing 400 nM apixaban with the indicated variable concentrations of api 31. The results illustrate that 400 nM apixaban inhibits the thrombin generation (peak shifts to the right and the peak is lower). Furthermore, addition of 2-4 fold molar excess of api_31 (800-1600 nM) completely restores the thrombin generation (fXa activity) and reverses the fXa inhibiting effects of apixaban; 2-4 fold molar excess concentrations of api_31 lead to a return of the thrombin generation curves into the same pattern as the reference thrombin generation curves obtained with positive control, i.e. the control plasma without apixaban and without the apixaban inhibiting polypeptide api_31.
The figure 15 shows the lag times for thrombin generation, derived from thrombin generation curves obtained with positive control human plasma sample obtained from three independent donors. The reference lag time is derived from plasma without apixaban and without api_31 (plasma). All the other lag time measurements are obtained from the plasma sample that contains 400 nM apixaban with the indicated variable concentrations of api_31. The results illustrate that 400 nM apixaban increases the lag time at least twofold. Furthermore, addition of api _31 restores the thrombin generation back to control plasma sample values and fXa inhibiting effects of apixaban are effectively reversed, and at a 4 fold molar excess concentration (1600 nM), api_31 completely compensated the prolonged lag time obtained under influence of apixaban in the plasma sample back to the same value as obtained for the lag time with the reference plasma sample.
The figure 16 shows the peak thrombin values, derived from thrombin generation curves obtained with the same healthy human plasma sample obtained from three independent donors. The reference peak thrombin value is derived from plasma without apixaban and without api_31 (plasma). All the other thrombin peak measurements are determined with the plasma sample that contains 400 nM apixaban with indicated variable concentrations of api_31. The results illustrate that 400 nM apixaban decreases the peak thrombin values. Furthermore, addition of api 31 restores the Peak Thrombin value back to the Peak Thrombin value measured for the positive control, i.e. the plasma sample that is not spiked with the DOAC. That is to say, the fXa inhibiting effects of apixaban is reversed under influence of the polypeptide api_31, and at a 2 fold molar excess concentration (800 nM), api_31 already completely compensated (restored) the peak thrombin values to the same extent as the peak thrombin value obtained with the reference plasma sample.
Apixaban Treated Patients
Api 1 in plasma samples of human patients, treated with apixaban
The figure 17A and 17B illustrate the Sta® factor Xa test in plasma samples of human patients who are on treatment with apixaban, without spiked api 1 and with api_1, spiked to plasma. The same measurements have been performed for the bivalent polypeptide api_31. The concentration of the apixaban-inhibiting polypeptides api_1 and api_31 tested in the assay is 1600 nM (4-fold excess of the peak values).
It Is observed that both api_1 (Fig. 17A, B) and api_31 (not shown) are able to compensate the prolonged lag time for thrombin generation under influence of apixaban in patients treated with apixaban independently of the levels of apixaban in their plasmas.
Since the DOAC neutralizing effect was already established within 10 minutes after mixing the plasma with the polypeptide api_1 or api_31, these examples are illustrative for a rapid and complete DOAC neutralizing effect in the patient's circulation when the polypeptide is administered to the patient intravenously at a dose sufficient to establish a concentration of about 1600 nM of the polypeptide in the plasma.
Apixaban concentration can be determined in patient plasma with a STAR-Max DOAC test: Factor Xa formation is measured with a factor Xa specific chromogenic substrate. Cleavage of this substrate results in factor Xa concentration-dependent increased light absorption. DOAC concentration is determined from a calibration curve of a series of DOAC concentrations.
Figure 18 displays the results of the surface plasmon resonance (SPR) measurements obtained with immobilized apixaban and apixaban-inhibiting VHH domain api_1 (SEQ ID NO: 64). Based on the measurements, the ka, ka and KD values were determined (see Table E). Three replicates were measured. Importantly, in the similar SPR measurements, api_1 did not bind to any of the DOACs edoxaban, rivaroxaban and betrixaban (data not shown), showing the specificity of the api_1 polypeptide for apixaban binding.
TABLE E. Kinetics and affinity constants for binding of polypeptide api_1 to apixaban (surface plasmon resonance data)
Replicate Ka (1/Ms) ka (1/s) Ko (M) 1.00e+07 8.45e-03 8.42e-10 1.39e+07 1.69e-02 1.22e-09 1.33e+07 1.48e-02 1.11e-09 1.24e+07 1.34e-02 1.06e-09
Synthesis of a bivalent molecule comprising two apixaban binding VHH domains
A bivalent molecule api_32 was synthesized on the cDNA level and on the protein level (SEQ ID NO: 101), comprising two linked VHH domains api_1 (SEQ ID NO: 64). Such a bivalent molecule is suitable for increasing the half-life of the molecule in the human blood circulation. The bivalent VHH is about twice as large as a single VHH domain and therewith increases the IC50 for clearance from the circulation. In addition, the bivalent molecule can bind two copies of apixaban, therewith having an increased binding capacity at the same molar dose. For, for example, purification purposes, the bivalent molecule is provided with a His-tag such as a C-terminal His-tag, such as a 6x His-tag.
Testing the DOAC inhibiting activity of bivalent VHH specific for apixaban, in human plasma spiked with apixaban
Apixaban concentrations were measured in normal pooled plasma (NPP) without added apixaban and without api_32 (two linked apixaban binding VHH domains, with SEQ ID NO: 101) (referred to as sample “plasma” in Figure 22). For the other measurements, the plasma was spiked with apixaban to a final concentration of 218 nM apixaban (Figure 22A) and 1088 nM apixaban (Figure 22B}; with 0-, 4-, 8-, and
16-fold excess concentrations of added api_32. Thirty uL spiked-plasma was diluted 4-fold in Owren-
Koller® and mixed with 150 pL of chromogenic substrate (CBS 02.44 consisting of MAPA-Gly-Arg-pNA
HCI) and incubated during 240 seconds. Then, 150 jL of bovine fXa pre-warmed at 37°C were added, starting the measurement. Results are expressed in OD/min and measurements were performed on
STAR.
Results
The figure 22A and 22B shows results of the here-above outlined Chromogenic STA® liquid anti-Xa (LAX) test (diagnostica stago) (A, B). The reference measurement is human normal pooled plasma (NPP) without added apixaban and without added api_32 (two linked apixaban binding VHH domains, with SEQ ID NO: 101) (referred to as “plasma”) (A, B). All the other four measurements contain 218 nM apixaban (A) and 1088 nM apixaban (B) with 0-, 4-, 8-, and 16-fold excess concentrations of added api_32. The results illustrate that apixaban spiking increases the apixaban concentration measurement by Chromogenic STA® liquid anti-Xa (LAX) test. Addition of 16-fold excess api_32 completely inhibits the effects of apixaban; as shown by the return of the Chromogenic STA® liquid anti-Xa (LAX) test value into the level as measured for the NPP reference in the Chromogenic STA® liquid anti-Xa (LAX) test.
RIVAROXABAN
Table 3 Selection of representative VHH's in the binding clusters 1, 2 and 4 for binding to rivaroxaban
Relative max binding (OD) | Affinity (nM)
ND: not determined
In Table 3, for several representative VHH domains capable of specifically binding to rivaroxaban, the binding of the VHH domains to rivaroxaban is presented, as determined with an ELISA.
Diagnostic anti factor Xa (DOAC) test of Stago diagnostica to investigate the anti-DOAC function (inhibitory activity towards fXa inhibition by the DOAC) of rivaroxaban-binding Va
The figure 19 shows the thrombin generation curves obtained with control human plasma, said plasma spiked with 600 nM rivaroxaban, and said spiked plasma to which a concentration series of rivaroxaban inhibiting polypeptide riva_1 (SEQ ID NO: 79) was added. The reference thrombin generation curve is obtained with human plasma without rivaroxaban and without riva_1 (plasma (no rivaroxaban)). All the other curves are obtained with said human plasma sample that how contained 600 nM rivaroxaban with the indicated variable concentrations of riva_1. The results illustrate that 600 nM rivaroxaban inhibits the thrombin generation (peak shifts to the right and the peak is lower). Furthermore, addition of 16 fold molar excess riva_1 (9600 nM) completely restores the thrombin generation and reverses the fXa inhibiting effects of rivaroxaban; 16 fold molar excess concentrations of riva_1 lead to a return of the thrombin generation curves into the same pattern as the reference thrombin generation curves (peak shift back to the short thrombin generation time obtained for positive control plasma sample, and peak maximum for thrombin generation being about the same as that obtained with the control plasma sample).
The figure 20 shows the lag times, derived from thrombin generation curves obtained with positive control plasma sample obtained from three independent human donors, said plasma sample now spiked with 600 nM rivaroxaban, and said spiked plasma sample to which is further added a concentration series of the rivaroxaban inhibiting polypeptide riva_1. The reference lag time is derived from human plasma sample without rivaroxaban and without riva_1 (plasma). All the other lag time measurements are obtained with said plasma sample that now contains 600 nM rivaroxaban with the indicated variable concentrations of riva_1. The results illustrate that 600 nM rivaroxaban increases the lag time at least threefold. Furthermore, addition of riva_1 restores the lag time for thrombin generation back to the value obtained with the positive control plasma sample. The fXa inhibiting effects of rivaroxaban are reversed in a dose dependent manner, and at a 16 fold molar excess concentration (9600 nM), riva_1 completely compensated the prolonged lag time obtained under influence of the
DOAC back to the same value for the lag time obtained with the positive control.
The figure 21 shows the peak thrombin values, derived from thrombin generation curves obtained with the same healthy human plasma sample obtained from three independent donors. The reference peak thrombin value is derived from plasma without rivaroxaban and without riva_1 (plasma). All the other peak thrombin value measurements contain 600 nM rivaroxaban with variable concentrations of riva_1.
The results illustrate that 600 nM rivaroxaban reduces the peak thrombin value. Furthermore, addition of riva_1 restores the peak thrombin value and reverses the fXa inhibiting effects of rivaroxaban, and at a 16 fold molar excess concentration (9600 nM), riva_1 completely compensated the peak thrombin value obtained under influence of the DOAC back to the same value as the reference peak thrombin value obtained with the positive control plasma sample.
Synthesis of a bispecific molecule comprising rivaroxaban binding capacity and albumin binding capacity
A bispecific molecule was synthesized on the cDNA level and on the protein level (SEQ ID NO: 102; referred to as riva_39), comprising the VHH riva_1 (SEQ ID NO: 79) and the subsequent VHH domain with SEQ ID NO: 108. Such a bispecific molecule is suitable for increasing the half-life of the molecule in the human blood circulation. The albumin-binding VHH binds to albumin present in the circulation and therewith increases the IC50 for clearance from the circulation. For, for example, purification purposes, the bispecific molecule is provided with a His-tag such as a C-terminal His-tag, such as a 6x
His-tag.
Synthesis of a bivalent molecule comprising two rivaroxaban binding VHH domains
A bivalent molecule riva_40 was synthesized on the cDNA level and on the protein level (SEQ ID NO: 103), comprising two linked VHH domains riva_1 (SEQ ID NO: 79). Such a bivalent molecule is suitable for increasing the half-life of the molecule in the human blood circulation. The bivalent VHH is about twice as large as a single VHH domain and therewith increases the IC50 for clearance from the circulation. In addition, the bivalent molecule can bind two copies of rivaroxaban, therewith having an increased binding capacity at the same molar dose. For, for example, purification purposes, the bivalent molecule is provided with a His-tag such as a C-terminal His-tag, such as a 6x His-tag.
i “odsml varsion=sl QN evooing="UNNS8N TL ë <!DOCTYPE ST26SequenceListing PUBLIC "-//WIPO//DTD Sequence Listing 1.3//EN" "ST26SequenceListing V1 3.dtd"> 2 <“ST26fequencelieting colgiseiiveeTerilanguegalode="an" dudVarzlion="Vi_ 3% IilaName=
NPEZOOZSOINL sequange listing Sinal amd? softwaredanm:="WIPS Sequana scinwarevVersian= v.30 SYaniiet LonData= 2023-08-11 % > 4 <Applicationidentification>
NE <IPOfficefoderNL</IPOfficetoder
Z <ApplicationMunbertents></ApplicationNunba riers 7 <FilingDater</FilingDaie> © </hppiicationidentification> 3 <&ppliceantFilekeference>P62003907NL</ApnlicantFilsReferencex
Lj <ApplicaniName languagaelode=tan>Synapse Research Institute</ApplicantNams>
Gk <InvenitionTiitle languagslode="an'>MOLECULES FOR REVERSING ANTI-COAGULANT ACTIVITY
OF DIRECT ORAL ANTICOAGULANTS</InveniionTitle>
La <SequenceTlobalduantity>108</Zequencalaotaluantity>
LS <Sequencebata seguanceiiNucbev=niND> id <INSDEeg> ih <INSDSeq length>»8</INSDSeg length» id <INSDSeq moltype>AAc/INSDSeq molLypex
Lj <INSDSeq division»PAT</INSDSeg divisions
LE <INSDSeg feabture-tablel in <INSDFesture> 20 <INSDFearure key>source</INIDFeature key»
ZL “INSDFeature Location>l..8</INSDFeelure location»
ZE <“INSDPFeacure ouals:»> 22 <INSDQualifier»> 24 <INSDQualifier name>mol type</INsSDQualifier nama» 25 <INSDuualifier valuerprotein</INSDOualifisr valus> 28 </INSDQualifisan» 2 <INSDQuaiifler id="gloS852> aj <INSDOQualifier namerorganism“/INSDQualifier name eis CINZDQualifiler value>synthetic construct </INSDQualifier value> 20 </INSDQualifier> zi </INSDFeature guals> 22 x“/INSDFeacturex IJ 33 </INEDEeg feature-tabled> 34 <INSDSeq segquence>GFAFDDYA</INSDSeq sequenced 3% </INSDSear 34 <j Gaemquencebata> 37 <Gequencelala segquenceliNuubsc=tg> 28 <INSDSeq> 23 <INSDSeq iength>8</INSDSeng lengih> 40 <INSDSeg moliype>AAd/INSDSeqg moltype»>
Gd <INSDSeq division>PAT</INSDSea division»
Ee <INSDieq festure-table> 43 <INSDFeature> ah “INSDFearture keyrsourced/INIDFeaturs Key» a4 <INSDFeecture locabtion>l..B</INSDFealtire location» dd <INSDFeature guals> u 37 <INSDOualifier>
G8 <INSDQualifier name>mol type</INSDQualifiar DATE >
G9 <INSDQualifier valuesprotein/INSDqvalifier valued 59 </INSDQuali fier!»
CINSDQualifiler id="glijr>
DE “INSDOualifier namevorganism /INSDQualifier named 53 <INSDQualifier value>synthetic construct </INSDQualifier values
Bd </INSDQualifier> -
Sl </INSDFaature quals> hE </INSDFeature> - 5 </INSDSeg feature-tabhlad 3 <INSDSeq sequence>GFTFDDYA</IN3SDSeg sequencer 2 </INSUSeq> u
Li </EegquencebData> el “SequenceData seguencellsNumben="3"> 52 <INSDSeq> 2 <INSDSegq lengitb>b9S4d/INSDEeg lengih> 4 <INSDSeg moltype>AA</INSDSeqg moliype:» 5 <INSDSeq division>PAT</INSDSeqg division» ce <INSDSeq festure-table>
£7 <INSD¥eature»
Gi <INSDFeature key>source</INSDFeature key»
OG <INSDFeature locations»l..9S</INSDFeature locations»
Fh <INSDFesture duels»
Gl SINEZDOualifiso>
Ta <INSDQualifier namedmol type</INSDOualifisr name> 3 <INSDQualifier valuerprotein“/INSDgualifier value»
Th </INSDOualifier:>
TE CINSDQualifler io=vglily>
TE <INSDQUalifiler naemerorganism/INSDOualifier name
TE <INSDQualifier value>synthetic construct </INSDQualifier valued 38 </INSDQualiËier>
Ta </INSDFaature cualss» </INSDPeature>
SL </INSDSeg feature-teblex> od “INSDSeqg sequencerISSSDGSTY</INSDSeq sequence
GS </INSDSeg> ee! </SeguenceData> <Sequencalata sopancalbiumber="4v>
Zó VINSDSeq» wy <INSDZeq length>24</INSDSeq lengths 33 <INSDSeq moltype>AA</INSDSeqg moltypes
So <INSDSeq division>PAT4/INSDSeg division oi “INSDSeq fealure-table>
Di <INSDFeature»
Ad <INSDFeature kevy>source</INSDPFeature key: 332 <INSDFesture locationrl..24</INSDFeature locations 34 CINSDFeature quals> u
GH <INSDQualifier>
QE <INSDQualifier namermol type</INSDQualifier name> ui CINMEDOualifier valuesprotein</INSDLQualifier valued
Ld <SINSDOualifier> af LINSDGualifiler in=Ngilar>
LO <INSDQualifier name>organism</INSDQualifier names
AEE <INSDQualifier value>synthetic construct <SINSDOualifier value»
LOR </INSDQuali fier:
HOE </INSDFeature quals>
LOG </INSDEFeature>
LoL </INSDSeg faature-table>
LUE “INSDSeq sequsrce>AAVPRTMYSRWGCGVRPYYYGMDY“ /INSDSeq sequence 107 </INEDSea> 108 </SeguencaData ins <SequenceData samencoelósuben=NSN>
LAD <INSDSeq>
LAE <INSDSeq lengih>24</INSDSeg length»
LL <INSDSeq moliype>AA</INSDSeq moltype»
LLS “INSDSeq division»PAT</ INDE division
Lie <“INSDSeq feature-itable> 115 <INSDFearture:» iia <INSDFeature keyrsource</INSDFeature key> 1a <INSDFeature locations>l..24</INSDFeature Locations
LAE <INSDFeature guals> ie <INSDQualifier>
Lal CINEIDOualifier namermol type“ /INSDUualifier name>
Loi <INZDQualiflsr values>protein</IN3LQualifier valued
LEE </INSDOualifier> 123 <INSDQualifier id="glijn> ize <INSDQualifier name>organism</INSDQualifisr names 185 <INBDQualifier valuersynthetic construct </INSDOualifier value ize </INSDQuali fier!»
LET </INSDFearture qualss
LEG </INSDFeacturer
LADE </INSDSeg feature-itablex 13h <INSDSeg sequsnce>AAVPRTMNSRWGCGVRPYYYGMDY-/INSISeqg sequence 1a: </INSDSeq> u 152 </SecuenceDatas 133 <Sequencelata saguencalliunbaer="gt» 134 <iNSDSed>
LEG <INSDSeq lengih>24</INSDSeqg lengths
LE CINZDSeq moltype»AAS/INSDSey moltype> 157 <“INSDSeg division>PAT/INSDSecg division» 1H <INSDSeq feature-itableX 133 CINEDEeatures ijd <INSDFeeture keyrsource“/INSDFeature key> ijl <INSDFeature locationsl1..24</INSDFeature Location»
LAL {INEDFeature gualis>
TAS <INSDQualifiers
Lid <INSDQualifier name>mol type“ /INSDQualifier name»
Lan <INSDQualifier valuse>protein</INaTQualifier value» ide </INSDOQualijiier> ijj CINSDOualifier id="gilgr> ijs <INSDuualifier namevorganism“/INSDQuali fier name> ide <INSDQualifier valuersynthetic construct </INSDOualijier value»
LEG </INSDOualifier:>
LEL </INSDFealture cauels»>
HN </INSDFealure»> </INSDSeg feature-tabler
RINE <INSDSeq zsequencs>AAVPRTMYSRWGCGVRPYYHGMDY-/INSDieq sequence 155 </INSDSeqy
L3G </SeguenceDatas
LE <Sequencebata zegvencaiDNumber=njn)»
LEE <INSDSeq>
Lh CINEDSeq length>8</INSDSeq lengths»
LED <INSDSeq moltype>dAA/INSDSeg moltype> ial <INSDSeq diviglon»>PAT</INSDSeg divisions ief <INSDSeq Ieatureriabier 153 “INSDFeaturex
Lhd <INSDFeature key>source“/INSDFealure key> 18% <INSDFearure locations»l..8</INSDFeature locations
LO <INSDFearure guals:»
LET <CINSDQualifiers
LEE <INSDQuUalifiler namebmol type /INSDOualifier name
LES <INSDQualifier value»protein</INSDOQualijier value» ijn </INSDQualifier>
KE JINSDoualifier id="gil5ns
LHR <INSDGualifier namerorganism</INSDQualifisr name> 1 <INSDQualifier valuersynthetic construct </INSDQuali fier values 174 </INSUQualifiers>
LEE </INSDFeature duels»
HE <JINSDFeature»
ATE </INSD8eq featura-tabler ie <IN3DSeq zequence>GRTFSINV</INSDSed sequence irs </INSDSeg:> i848 </Segquaencebata>
LE <Segquencebalta seguantailiicghar=ngns
TER CINSDSag»
Las <INSDSeq length>9</INSDSeq length? isd <INSDSeq moltype>AA“/INSDSeq moltype>
Eh <INSDSeq division»PAT</INSDSeqg division: ind <INSDSeq fearvurertabier»
LET <INSDFeabture> 18s <INSDFsature key>source</INZDFsature key>
TRG CINEDFsature location>l..9</INSDFeature location
Lau CINEDFeature gualse
RE CINBDOualifierns ied <INSDQualifier nams>mol type“ /INSDOualifier name ijl <INSDQualifier valuerprotein</INSDQualifier value» 134 <ATNaSDQuali fis» 185 <INSDQualifier ìd="glist2>
RES <INSDQualifier namerorganism“/INSDQualifier name>
Lar <INSDOQualifier valuersynthetic construct <{/INSDQuali jier valuex> 1e </INEDOualifiers> 145 </INSDPeature vuals> 200 </INSDFeaturer - 231 </INSDEeg Zearture-tabier 202 <INSDSeq sequence>IWWSGGASQ</INSDSeq sequence> 203 </INSDSec:> 204 <SBequencelata>
Sal <Hegquencelbata seguansellMonhar=nat >
Li LINSDSeq»
ZOT <INSDSeq iength>9«/INSDSeg length> zE <INSDSeq moltype>AA</INSDSeq moliype> 203 <INSDSeq division»PAT</INSDSeg divisions
ZLD <INSDSeg feabture-tablel 2 <INSDFesture> aE <INSDFearure key>source</INIDFeature key»
SL “INSDFeature location>l..9</INSDFealture location»
FRR <“INSDPFeacure gqualis> ih <INSDQualifier»> zie <INSDgualifier name>mol type:/INSDOQuali fier name>
SAT <INSDuualifier valuerprotein</INSDOualifisr valus>
Zie </INSDQvalifier: ale <INSDQuaiifler id="glij"2>
EA CINMEDOualifier namerorganism</INSDQualifier name oak <INZDQualiflsr value>synthetie construct </INSDQualifier value> zee </INSDOuali fier» zal </INSDFeature guals> 22d x“/INSDFeacturex
Zal </INSDSeg feature-tabled> 226 <INSDSeq sequence>TWWSGGASQ</INSDSeq sequence» nav </INSDSear
Las <j Gaemquencebata>
Ln <“SequsrnceData segquenceliNuubsc=NiGr>
L300 <INSDSeq> zji <INSDSeq length2l5</INSDSeq length> 2322 <INSDSeq moliype>AA/INIDIeq molLype»> zij <INSDSeq division>PAT</INSDSeg division» 2A <INSDieq festure-table>
EARS <INSDFeature>
EEE “INSDFearture keyrsourced/INIDFeaturs Key» 2 <INSDFeecture lcocaition>l.. 15</IN3DFeature location»
Zu <INSDFesture gualis>
San <INSDOualifier> zän <INSDuualifier name>mol type“/INSDQuali fier name> 23 <INSDQualifier valuesprotein/INSDqvalifier valued 242 </INSDQuali fier!»
ZA CINSDQualifiler id="gligv>
Sad CINZDQuallfier namerorganism</IN3DQualifier name>
Lis <INSDQualifier value>synthetic construct </INSDQualifier values
ERED </INSDQualifier> 247 </INSDFeature gquals> 24% </INSDFeature> - 24% </INSDSeg feature-tabhlad
Zn <“INSDSeqg sequence >AAGPMFSMDYRRVNY </ INSDSeqg sequence
SL </INSUE aa»
LE </SeqenceData>
Ze “SequenceData seguencellsumben=N2L®> 254 <INSDSeqg> 255 <INSDSeq lengih>15</INSDSeq length> 256 <INSDSeq moliype>AA</INSDSeq moltype> 25% <INEDZeq division>PAT</INSDEsqg division»
ZnS <INSDSeq festure-table> dille <INSDFeature»
ED <INSDFeature key>source</INSDFeature key» ded <INSDPeature locations. .15:/INSDFeature location» zó2 <INSDFesture duels»
Zan CINSDQuUalifiso> 264 <INSDQualifier namedmol type</INSDOualifisr name> 28% <INSDQualifier valuerprotein</INSDgualifier values
REE </INSDOualifier:> ni’ CINSDQualifler io=vglisdys
ZES <INSDQUalifiler naemerorganism/INSDOualifier name
ZE <INSDQualifier value>synthetic construct </INSDQualifier valued 278 </INSDQualiËier>
SE </INSDFaature cualss»
ZEE </INSDFeature>
EAE </INSDSeg feature-tabled> an “INSDSeq sequence >AAGPVFSMDYRRVNY</INSDSeq sequencer
E75 </INSDSeg> did </SeguenceData>
Ea) <Sequencalata sopeancalliumber="313%> zin CINEDSeqr 208 <INSDSey lenghh>15</INSDSeq length 280 <INSDSeq moltype>AA</INSDSeqg moltypes
ZR <INSDSeq division>PAT4/INSDSeg division
SE “INSDSeq feature-table>
En LINSDFeature> did <INSDFeature kevy>source</INSDPFeature key:
SED <INSDFesture Location>l..1B</INSDFeature location» 288 CINSDFeature quals> u 287 <INSDOualifier> 288 <INSDQualifier namermol type</INSDQualifier name>
SE CINMEDOualifier valuesprotein</INSDLQualifier valued
REE <SINSDOualifier> a LINSDGualifler in=Ngigd®> zieë <INSDQualifier name>organism</INSDQualifier names 232 <INSDQualifier value>synthetic construct <SINSDOualifier value» and </INSDQvalifier: - 29% </INSDFeature quals> nue </INSDFeature> an </INSDSeg faature-table>
LE “INSDSeq sequence>AAGPMFSMDYTRVNY</INSDSeq sequence»
ZA </INSDSeg> </SeguenceDatar
GDL <SequenceData samencelósubern=NL3nx>
TOE <INSDSeq> 33 <INSDSeq lengih>15</INSDSeg length»
IO <INSDSeq moliype>AA</INSDSeq moltyper
SOL CINEDSeq division»PAT</ INDE division
SLE <INSDSeq feature-itable>
SO <INSDFeature:> 308 <INSDFeature keyrsource“/INSDFeeture key> 23 <INSDFeature location>l..15</INSDFeature location»
TLG <INSDFeature guals>
SAE <INSDQualiifisr>
SLE <INSDOQualifier namermol type“ /INSDUualifier name>
LS v“INSDQualifiern valuesprotein“/INSDgualifier value» sld </INSDOualilfier> 418 <INSDQualifier id="glg22n>
ERE <INSDQualifier name>organism</INSDQualifisr names
Si <INBDQualifier valuersynthetic construct </INSDOualifier value
SLS </INSDQuali fier!»
IL </INSDFearture qualss
G0 </IN3DFaaturey
SAL </INSDSeg feature-itablex
SZ <INSDSeg sequsnce»>AAGPMFSMDYRRVNH</INSDSeg sequence» 282 </INSDSeq> u
S54 <fSeguenceDalar
TE: <SequenceData zeqvencslnNuber="ian»
ILE <iNSDSed>
Sj <“INSDSeq length>8</INSDSeg length»
Sas “INSDSeqg moliypevAA</INSDSeq moltype>
Sz <“INSDSeg division>PAT/INSDSecg division»
SED <INSDSeq feature-itableX
Sak LINSDFeature> 2322 <INSDFeature keyssource“/INSDFealure hay» 2d <INSDFeature location»>l..8</TINSDFeature location» zijd <INSDFeature quals> u
EIR <INSDQualifiers
BRE CINEDQUalifiler name>mol type“ /INSDgualifier named
HRT <INSDQualifier valuse>protein</INaTQualifier value» 448 </INSDOQualijier>
S38 CINSDOualifier id="glz2r>
San <INSDuualifier namedorganism</INSDOualifisr name>
BE <INSDQualifier valuersynthetic construct </INSDOualijier value»
BAD <fINSDUualifler»>
RE </INSDFeaturs gquals>
Hdd </INBDFeaturne»
Lan </INSDSeg festure-tabler
REE <INSDSeq saquence>GRTFSSYH/INEDIaq sequence 247 </INSDSeas IJ ian </SecuenceDala»>
TAS <Sequencebata zegvencaiDNumber=nijtD>
IHG <INSDSeq>
SDL “INSDSeg length>9</INSDSeg lengths»
SLE “INSDSeq moltype>dAA/INSDSeg moltype> 353 <INSDSeq diviglon»>PAT</INSDSeg divisions 254 <INSDSeq Ieatureriabier 255 CINEDFeaturae»
Ihe <INSDFeature key>source“/INSDFealure key>
RN <INSDPFeature iocation»l..9S</INSDPeeture location»
IRB <INSDFearure guals:» dn <CINSDQualifiers
HED <INSDQuUalifiler namebmol type /INSDOualifier name
SE <INSDQualifier value»protein</INSDOQualijier value» 282 </INSDQualifier> 267 CINSZDOualifiler 1d="qRRizve
TE <INSDQualifier namerorganism“/INSDQuali fier name> zel <INSDQualifier value»synthetic construct </INSDQuali fier values
IEE </INSLQualifiers se </INSDFeature duels»
SER </INSDFeature:> 353 </INSDSeg feature-tabler
SFG <INSDSeq zeguenc=e>ITRGGGVTY</INSDSeq sequencer 371 </INSDSeq>
IEA </Segquaencebata>
IES LSequencebalta seqguancailiioghar=i8% >
RE CINSDSag»
STE <INSDSeq length»9S</INSDSeg length»
STe <INSDSeq moltype>AA“/INSDSeq moltype>
ST <INSDSeq division>PAT/INSDSeg divisions zie <INSDSeq fearvurertabier» ijd <INSDFeabure> 380 <INEDFeature key>source:/INSDFeature kKey>
IRE CINSDFeature iccation»>l..9</INSDFeature location
SEE CINEDFeature guals> ses <INSDQOualifier» dad <INSDQualifier nams>mol type“ /INSDOualifier name
SED <INSDQualifier value>protein</INSDQualifisr value» 388 </INSDQuali jier
DE <INSDQualifier 1d=V"gldvs
TRY <INSDQualifier namerorganism“/INSDQualifier name>
Sin <INSDOQualifier valuersynthetiec construct <{/INSDQuali jier valuex>
Ze </INSDOualifier>
Eh </INSDFPeature guals> 232 </INSDFeaturer u 232 </INSDEeg Zearture-tabier
IHG <INSDSeq sequences>IARGGGVTI</INSDSeq sequence 355 </INSDSeg>
ERE <SBequencelata> hE <HGegquencelbata seguanselilMonhar=niTYs
SEG LINSDSeq»
SD <INSDSeq length>18</INSDSeq length> 40340 <INSDSeq moltype>AA</INSDSeq moliype>
SDL <INSDSeq division»PAT</INSDSeg divisions 402 <INBDZeq feabture-tablel 305 <INSDFesture> 404 <INSDFearure key>sources/INSDFeature key> 4050 “INSDFearure location». 18</INSD¥Featiure location 405 <“INSDPFeacure gqualis> dT <INSDQualifier»> 409 <INSDgualifier name>mol type</INSDOualifier name> 408 <INSDuualifier valuerprotein</INSDOualifisr valus> 410 </INSDQualifisan»
AL <INSDQuaiifler id="gl2512>
ALE <INSDOQualifier namerorganism“/INSDQualifier name
413 CINEDOUalifisr value>synthetie construct </INSDQualifier value>
Ald </INSDOQualijiier> 419 </INSDFeature guals> ais x“/INSDFeacturex IJ
ALT </INEDEeg feature-tabled> 418 <INSDSed sequence>AADAIWNQVRWMETKYTY</INSDSeg sequenced
AL </INSDSear dE <j Gaemquencebata>
TEL <“SequsrnceData segquenceliNuubsc=NiSN> dee <IiNSsDseqd> dz <INSDSeq length>8</INSDSeg length» did <INSDSeq moliype>AA/INIDIeq molLype»> 42% <INSDSeq division>PAT</INSDSeg division» 326 <INSDieq festure-table> 427 <INSDFeature> 4nd “INSDFearture keyrsourced/INIDFeaturs Key» ds <INSDFeecture loceciorsl..8</INSDFeature location» 450 <INSDFesture gualis>
Zi: <INSDOualifier> an <INSDuualifier name>mol type</INSDOualifisr name> 4273 <INSDQualifier valuesprotein/INSDqvalifier valued 434 </INSDQuali fier!»
AI CINSDQualifiler id="gldi&v>
AEE CINZDQuallfier namerorganism</IN3DQualifier name> 457 <INSDQualifier value>synthetic construct </INSDQualifier values 458 </INSDQuali fier» u 458 </INSDFeature gquals> 444 </INSDFeature>
AAL </INSDSeg feature-tabhlad
AAA <INSDSeq sequence>GRIVSIYS</INSDSag seqience> 443 </INSDE ag» dad </EegquencebData> dd “SequenceData seguencellsumben="i8®> ada <INSDSeq> 337 <INSDSeq Lengihb>8</INSDSeqg lengih> 338 <INSDSeq moliype>AA</INSDSeq moltype»r> 448 <INSDSeq division>PAT</INSDSeqg division»
AG <INSDSeq festure-table> 450 <INSDFeaturer
TE <“INSDFearure key>source</INSDFeature key» dal <INSDFeature lccationsl..8</INSDFealture location» 454 <INSDFesture duels» 43% CINSZDOualifier> 458 <INSDQualifier namedmol type</INSDOualifisr name>
An <INSDQualifier valuerprotein</INSDgualifier values 453 </INSDOualifier:> ann CINSDQualifler io=ngladys
AE <INSDQUalifiler naemerorganism/INSDOualifier name dE <INSDQualifier value>synthetic construct </INSDQualifier valued 442 <ATNaSDQuali fis» 457 </INSDFaature cualss» 484 </INSDFeature>
Aas </INSDSeg feature-tabled> 444 “INSDSed sequsnce>GRIVGIYS</INSDSeq zequence> 167 <SINBDE eg
AR </SeguenceData> 4573 <Sequencalata sopeancalliumber="208> 4770 CINEDSeqr
ATE <INSDSey lenghh>8</INEDIag length»
AE <INSDSeq moltype>AA< /INSDSeg moltype:»
AUS <INSDSeq division>PAT4/INSDSeg division 44 CINEDSeq feature-table> 475 <INSDFeaturer 478 <INSDFeature kevyrsource</INSDFesture kev: 477 <INSDFeature location>l..8</IN3DFeature locations» 4778 <INSD¥eature guals> 478 CINSDQualifier>
ARQ <INSDQualifier namermol type</INSDQualifier name>
ARE CINMEDOualifier valuesprotein</INSDLQualifier valued
AEL <SINSDOualifier> 45% LINSDGualifler ia=ngl28y>
Af <INSDQualifier name>organism</INSDQualifier names 450 <INSDQualifier value>synthetic construct <SINSDOualifier value» 488 </INSDQualifisan»
ART </INSDFeature quals>
ABS </INSDFeature>
GE </INSDSeg faature-table> dol “INSDSeq sequerice>GRIVSTYS“/INSDSeq sequence> do: </INSDSea> 432 </SeguenceDatar 433 <Seduencepata sopuencallilumber="2340> 484 <INSDSeq> 48% <INSDSeq lengith>9S</INSDSeg length»
Ant <INSDSeq moliype>AA</INSDSeq moltype» qa CINEDSeq division»PAT</ INDE division dee <INSDSeq feature-itable>
BENE] <INSDFeature»> 304 <INSDFesture keyrsource“/INSDFealure key»
SR <INSDFeature locaftion>l..9S</INSDF Feature locations
SOL “INSDFeature guals>
RIE <INSDQualifier>
SG CINEIDOualifier namermol type“ /INSDUualifier name>
SUL <INZDQualiflsr values>protein</IN3LQualifier valued zE </INSDOualilfier>
LOT <INSDQualifier id="gl2S8'> 508 <INSDQualifier name>organism</INSDQualifisr names 508 <INBDQualifier valuersynthetic construct </INSDOualifier value
SLD </INSDQuali fier!» u
LL </INSDFearture qualss
Liz </INSDFaaturel
LLS </INSDSeg feature-itablex
Lid <INSDSeq sequence>ISWNGAETD/INSDSeq sequenced 315 </INSDSeg>
Sia <fSeguenceDalar
SA <SequenceData saguencalMiunbaer="23% 2%
SAR <INSDSeq>
LL CINEDSeq length>9</INSDSeg length»
Lal CINZDSeq moltype»AAS/INSDSey moltype>
SEL <“INSDSeg division>PAT/INSDSecg division»
SZ <INSDSeq feature-itableX
SEZ LIMIDFeaturas
Did <INSDFeature keyssource“/INSDFealure hay» sal <INSDFeature location»l..9</INSDFeature location»
Lae {INEDFeature gualis>
Sa <INSDQualifiers a CINEDQUalifiler name>mol type“ /INSDgualifier named
La <INSDQualifier valuse>protein</INaTQualifier value»
SEO </INSDOQualijier>
S53 CINSDOualifier id="gi3gr>
S22 <INSDuualifier namedorganism</INSDOualifisr name>
BI <INSDQualifier valuersynthetic construct </INSDOualijier value»
RI </INSDOualifier:>
LL </INSDFeaturs gquals>
IEE </INBDFeaturne» u
DST </INSDSeg feature-tabler 538 <INSDSeq sequencs>ISWNGGETD/INSDSeq sequence»
B29 <STHEDS eq
Ja </SecuenceDala»>
SAL <SequenceData zegvencaiDNumber=N23"D
SAE <INSDSeq>
LAS CINEDSeq length>9</THNSDSeq lengths»
Saba <INSDSeq moltype>dAA/INSDSeg moltype>
Sa <INSDSeq diviglon»>PAT</INSDSeg divisions
S49 <INSDSeq Ieatureriabier baj CINEDFeaturae»
Han <INSDFeature key>source“/INSDFealure key>
SAS <INSDPFeature iocation»l..9S</INSDPeeture location»
LEO <INSDFearure guals:»
DL <CINSDQualifiers
LEE <INSDQuUalifiler namebmol type /INSDOualifier name 555 <INSDQualifier value»protein</INSDOQualijier value» 554 </INSDQualifier> 35% CINSZDOualifiler 1d="qll3ive
She <INSDQualifier namerorganism“/INSDQuali fier name>
RR <INSDQualifier valuersynthetic construct </INSDQuali fier values
LG </INSLQualifiers
LIT </INSDFeature duels»
SED </INSDFeature> 353 </INSD8eq featura-tabler
DAZ <INSDSeq szequenca>ISWNGAETQ</INIDSeq sequencer
B47 “/INSDSeoa>
Sed </SegusncsDara>
LOL LSequencebalta seqguantailiioghar="24% >
LEE CINSDSag» 267 “INSDSeq length>20</INSkSeq Length>
DEE <INSDSeq moltype>AA“/INSDSeq moltype> 383 <INSDSeq division»PAT</INSDSeqg division:
DEG <INSDSeq feature-takbled
BL <INSDFeabure>
Sil <INEDFeature key>source:/INSDFeature kKey>
LE <INSDFearure location>l..20</INSDFeature location
LEA CINEDFeature gualse
LYE CINBDOualifierns
UE <INSDQualifier nams>mol type</INZDQualiifier name
DUT <INSDQualifier valuerprotein</INSDQualifier value» sR <ATNaSDQuali fis»
BO <INSDQualifier ìd="gl38N2>
SEO <INSDQualifier namerorganism“/INSDQualifier name>
SR <INSDOQualifier valuersynthetic construct <{/INSDQuali jier valuex>
Dad </INSDOualilfier>
BED </INSDFPeature guals>
BEd </INSDFeaturer u 385 </INSDEeg Zearture-tabier sn <INSDSeq sequence>AKPQSHFYDGSWRRASAYDD/INSDSea sequence 587 <“/INSDSeg>
SRE <SBequencelata>
Lh <HGegquencelbata seguanselilMonhar=na8Y >
Sen <INSDEeg>
Shi <INSDSeq length>20</INSDSeq length> 337 <INSDSeq moltype>AA</INSDSeq moliype> 532 <INSDSeq division>PAT/THSDSeq divisions
BR <INBDZeq feabture-tablel
La <INSDFesture>
Lo <INSDFearure key>source</INIDFeature key»
La “INSDFearure location>l..20</INSD¥Featiure location
LE <“INSDPearure duals»
Es <INSDQualifier»>
SA <INSDgualifier name>mol type:/INSDOQuali fier name>
GD: <INSDuualifier valuesprotein</INSDQvali fier valus>
ADE </INSDQvalifier: 553 <INSDQuaiifler id="gl3312>
Sá <INSDOQualifier namerorganism“/INSDQualifier name
Sl CINZDQualifiler value>synthetic construct </INSDQvalifier valus>
SE </INSDOualiiier>
LOT </INSDFeature guals>
S08 x“/INSDFeacturex
Hag </INEDEeg feature-tabled>
S10 <“INSDSed sequence>AKPQSHSYDGSWRRASAYDD«/INSISeq sequence»
SLI </INSDSeg>
GLE <j Gaemquencebata> £13 <Gequencelala sequanceliNuubsc=tgSN> aid <IiNSsDseqd> £15 <INSDSeq iength>20</INSDSeq length> ald <INSDSeq moliype>AA/INIDIeq molLype»> ai <INSDSeq division>PAT</INSDSeg division»
S18 <INSDSeq feature-table>
SLY <INSDFeature>
GE “INSDFearture keyrsourced/INIDFeaturs Key» dE <“INSDFeature Llocabtion»l..20/INSDFeature location»
SRE <INSDFeature gualis>
SEL <INSDOualifier>
BEd <INSDuualifier name>mol type</INSDOualifisr name> dak <INSDQualifier valuesprotein/INSDqvalifier valued
ARE </INSDQuali fier!» aa CINSDQualifiler iad=vqlldys
SEG v“INSDQualifier namerorganism</IN3DQualifier name> [Sas <INSDQualifier value>synthetic construct </INSDQualifier value
S35 </INSDQualifier> an </INSDFeature gquals>
A22 </INSDFealurex> - 533 </INSDSey feahure-table>
G34 <INEDSeq sequence>AKPQSHFYDGSWRRALAYDD/INSDSeq sequence»
SSD </INSDE ag»
L348 </EegquencebData>
ST “SequenceData seguencellsNumben="g"r>
HEE <INSDSeq> 433 <INSDSegq lengtb>b20</INSDSeq length>
HAG <INSDSeq moliype>AA</INSDSeq moltype>
SAL <INEDSeq divisior>PAT«</INSDSeg division»
SAR <INSDSeq festure-table>
Sas <INSD¥eature»
IS <INSDFeature key>source</INSDFeature key»
S45 <INSDFeature location>l..20</IN3DFeature location»
SEEN <INSDFesture duels» aa SINEZDOualifiso> din <INSDQualifier name»mol type“ /INSDQuali fier name> sis <INSDQualifier valuerprotein</INSDgualifier values an </INSDOualifier:>
SEL CINSDQualifler io=vglls®ys
Si <INSDQUalifiler naemerorganism/INSDOualifier name
FA <INSDQualifier value>synthetic construct </INSDQualifier valued
And </INSDQualiËier> aH </INSDFaature cualss» 556 </INSDPeature>
SRY </INSDSeg feature-teblex>
GLE CINZDSeq sequence >AKPQSHFYDGSWRRASAYGD</ INIDIeq sequence
SLD </INSDSeg>
S80 </SeguenceData>
G61 <SedquenceData sequencernNumbLer=Ng8N> 862 CINIDSeq>
A43 <INSDSey lenghh>8</INEDIag length» 594 <INSDSeq moltype>AA</INSDSeqg moltypes
SoL <INSDSeq division>PAT4/INSDSeg division [ES CINEDSeq feature-table>
E87 <INSDFeature»
LEE <INSDFeature keyirsource</INSDFeature key: 5553 <INSDFesture location>»l..8</INSDFeature location»
Ag <INSD¥eature guals>
SL <INSDQualifier>
SPE <INSDQualifier namermol type“ /INSDQgualifier name>
Sis <INSDOQualifier valuesprotein</INSDLQualifier valued
Ga <SINSDOualifier>
SUD LINSDGualifiler in="gi3Sr>
Ee <INSDQualifier name>organism</INSDQualifier names
HF <INSDQualifier value>synthetic construct <SINSDOualifier value» aia </INSDQualifisan» -
Sis </INSDFeature quals>
SSD </INSDEFeature> u
SRE </INSDSeg faature-table>
Lo “INSDSeq sequence>RRTFRSYA/INSkSeq sequence
LEE </INSDSea>
LE </SeguenceDatar
Sh <Seduencepata sopuencallilumber="280%
HES <INSDSeq>
SE <INSDSeq lengith>9S</INSDSeg length» sss <INSDSeq moliype>AA</INSDSeq moltype»
GR CINEDSeq division»PAT</ INDE division
Sah <INSDSeq feature-itable> del <INSDFearture:» £32 <INSDFesture keyrsource“/INSDFealure key»
BE <INSDFeature location>l..9S</INSDFeature locations
Hh <INSDFeature guals>
Aa <INSDQualifier> aud <INMEDQualifier namermol type</INsDoualifier name>
HF CINZDOQuallifier valuesprotein“/INSDgualifier value»
Eng </INSDOualilfier>
Ee <INSDQualifier id="glS7n>
TOG <INSDQualifier name>organism</INSDQualifisr names
EN <INBDQualifier valuersynthetic construct </INSDOualifier value
Tan </INSDQuali fier!»
RES </INSDFearture qualss
Gd </INSUFeature
Tuk </INSDSeg feature-itablex
TOE <INSDSeq sequsnces>TTWIISSTY/INSDSeq sequence 307 </INSDSeq> u an <fSeguenceDalar
Tag <SequenceData saguencalMiunbaer="30"%
TAO <INSDSeq>
LL <INSDSeq lengih>19</INSDSeq length
VLE CINZDSeq moltype»AAS/INSDSey moltype>
TLS <“INSDSeg division>PAT/INSDSecg division»
Tid <INSDSeq feature-itableX
Tin CINEDEeatures jlá <INSDFeature keyssource“/INSDFealure hay»
TAT <INSDFeature locationsl1..19</INSDFeature Location»
FLS <INSDFeature guals>
Tw <INSDQualifiers
Fai CINEDQUalifiler name>mol type“ /INSDgualifier named
EL <INSDQualifier valuse>protein</INaTQualifier value»
Tey </INSDOQualijier>
BEL CINSDOualifier id="giig0> id <INSDuualifier namevorganism“/INSDQuali fier name>
Tan <INSDQualifier valuersynthetic construct </INSDOualifier value»
TRE </INSDOualifier:>
Va </INSDFealture guals>
Grn </INSDFealure»>
TER </INSDSeg feature-tabler
IE <INSDSeq sequence>AARVRSGSGQYTLPGHYDY</INSDSeg sequence)»
Toi </INSDSeqy
TER </SeguenceDatas
TEE <Sequencebata zegvencaiDNumber=n3iND»
RG <INSDSeq>
TRL CINEDSeq length>8</INSDSeq lengths»
TEE <INSDSeq moltype>dAA/INSDSeg moltype>
TST <INSDSeq division»>PAT</INSDSeg divisions ian <INSDSeq feature-itablel
Tas “INSDFeaturex
Tan <INSDFeature key>source“/INSDFealure key>
JAL <INSDFearure locations»l..8</INSDFeature locations
TAL <INSDFearure guals:»
TAR <CINSDQualifiers
Gad <INSDQuUalifiler namebmol type /INSDOualifier name
Tah <INSDQualifier value>protein</INsDQualifier valued
Taw </INSDQualifier>
AT JINSDoualifier id="g1jn12 jn <INSDQualifier namerorganism“/INSDQuali fier name>
JAG <INSDQualifier value»synthetic construct </INSDOQuali fier valuer>
TE </INSUQualifiers>
JEL </INSDFeature duels»
Fn </INSDEeaturer
THE </INSD8eq featura-tabler
Th <INSDSeq zequence>TRISSLTV</INSDSedg sequence
FRA </INSDSeg:>
Tae </Segquaencebata>
TRY LSequencebalta seqguantailiioghar="32%2>
TLE CINSDSag»
TL <INSDSeq length>8</INSDSeq length?
TE <INSDSeq moltype>AA“/INSDSeq moltype>
TE <INSDSeq divislon»>PATC/INSDSeg divisions
Taz <INSDSeq feature-takbled
Tal <INSDFeabture>
Tod <INEDFeature key>source:/INSDFeature kKey>
TO CINEDFsature location>l.. 8</INSDFeature location
TOG CINEDFeature gualse
ET <INSDQOualifier»
TEE <INSDQualifier nams>mol type“ /INSDOualifier name
TES <INSDQualifier value>protein</INSDQualifisr value»
TIN </INSDQuali fier»
TL <INSDQualifier Ld=Vgl adv
EL <INSDQualifier namerorganism“/INSDQualifier name>
TE <INSDOQualifier valuersynthetic construct <{/INSDQuali jier valuex> 7d </INEDOualifiers>
TIES </INSDPeature guals>
TEA </INSDFeature> u
TTT </INSDEeg Zearture-tabier iis <INSDSeq sequence>LTRFGLAG</INSDSedq sequenced
TG </INSDSeg>
TRO <SBequencelata>
TRA <Segquencebata seguantaLiNomhaer="33%>
Tak LINSDSeq»
TEE <INSDSeq length>l0</INSISeq length>
ThE <INSDSeq moltype>AA</INSDSeq moliype>
TES <INSDSeq division>PAT/THSDSeq divisions
IEG <INBDZeq feabture-tablel a <INSDFeature>
TRE <INSDFearure key>source</INIDFeature key»
GL JIiNSDFeature lccation>l..10/INSDFeature Location
Fh <“INSDPFeacure gqualis> a <INSDQualifier»>
TY <INSDgualifier name>mol type</INSDOualifier name>
ERR <INSDuualifier valuerprotein</INSDOualifisr valus>
Tog </INSDQualifisan»
Tan <INSDQualifiler id="gl4ln>
TE <INSDOQualifier namerorganism“/INSDQualifier name
Tad CINZDQualifiler value>synthetic construct </INSDQvalifier valus>
TE </INSDOQualijier>
TRE </INSDFeature guals>
S00 </INSDFealure» u
SOL </INSDSeg feature-tabled>
SOE <INSDSed sequence>NVKTLGGADY</INSDSeg zequence>
SOS </INSDSe G>
Fld <j Gaemquencebata> ain <“SequsrnceData segquenceliNuubsc=NS4dN>
LE <IiNSsDseqd>
HOT <INSDSeq length>lO0</INSDSeq length>
S08 <INSDSeq moliype>AA/INIDIeq molLype»>
SOD <INSDSeq division>PAT</INSDSeg division»
SEG <INSDieq festure-table>
SL <INSDFeature> sl “INSDFsature keyrsourece</INIDFeaturs key»
SLS <INSDFeature locabtion>l..l0</IN3DFeature location»
Hid <INSDFeature gualis>
SLD <INSDOualifier>
Sia <INSDuualifier name>mol type</INSDOualifisr name>
SA <INSDQualifier valuesprotein/INSDqvalifier valued
SLE </INSDQuali fier!»
RG CINSDQualifiler ia=vqlddy>
GE v“INSDQualifier namerorganism</IN3DQualifier name>
BEL <INSDQualifier value>synthetic construct </INSDQualifier value
SEE </INSDQualifier> 222 <SINSDFaature guals>
Sad </INSDFeaiture»> 22% </INSDSey feahure-table>
BE <INSDSeq sequence>NAKTLGGADY</INSDSeqg zeguence>
Ra </INBUE aa»
GLE </SeqenceData>
GD “SequenceData seguencellsumben="3S®>
Ban <INSDSeq>
Zij <INSDSeq Lengihb>8</INSDSeqg lengih>
Zia <INSDSeq moliype>AA</INSDSeq moltype»r> 233 <INEDSeq divisior>PAT«</INSDSeg division» 334 <INSDSeq festure-table>
SL <INSD¥eature»
GG <INSDFeature key>source</INSDFeature key»
NET <INSDFeature location>l..8«</INSDIFesature locations»
HIER <INSDFesture duels» 228 SINSDOualifie>
S40 <INSDQualifier namedmol type</INSDOualifisr name>
SAL <INSDQualifier valuerprotein</INSDgualifier values
BAL <fINSDUualifler»>
Gis CINSDQualifler io=ngldlys
Dd <INSDQUalifiler naemerorganism/INSDOualifier name
Sh <INSDQualifier value>synthetic construct </INSDQualifier valued 248 <ATNaSDQuali fis»
Sa </INSDFaature cualss» 248 </INSDPeature> sas </INSDSeg feature-teblex>
BLU “INSDSeq sequance»GRTFTSYT</ INSDIeq sequence
FL <SINBDE eg 45E </SeguenceData> 252 <Sequencalata sequencelnNumbLer=NB&r> nid CINEDSeqr 25H <INSDSey lenghh>12</IN3DSeq length
BEG <INSDSeq moltype>AA</INSDSeqg moltypes
BDT <INSDSeq division>PAT4/INSDSeg division
BLE CINEDSeq feature-table>
SL <INSDFeature»
SEO <INSDFeature keyirsource</INSDFeature key:
Si: <INSDFesture locaticon»l1..12</INSDFeature Location»
Daf <INSD¥eature guals> ga <INSDQualifier>
Bad <INSDQualifier namermol type</INSDQualifier name>
BES CINMEDOualifier valuesprotein</INSDLQualifier valued
BEE <SINSDOualifier>
LET LINSDGualifiler in=Ngiddr>
SEE <INSDQualifier name>organism</INSDQualifier names 253 <INSDQualifier value>synthetic construct <SINSDOualifier value» 27 </INSDQvalifier:
ST </INSDFeature quals>
REL </INSDEFeature>
Ss </INSDSeg faature-table> aid “INSDSeq sequence>ISWSYWNGDSTW</INSDSed sequence» 5TH </INSDSeq>
Did </SeguenceDatar
SE <Seduencepata samencoalóNuber=s23n>
SIE <INSDSeq>
Sie <INSDSeq length>l17</INSDSeqg lengths
SEG <INSDSeq moliype>AA</INSDSeq moltype»
SEL “INSDSeq division»PAT</ INDE division
Go <INSDSeq feature-itable>
NEE <INSDFearure:x»
SESE <INSDFesture keyrsource“/INSDFealure key» £85 <INSDFeature locationsl..17</INSDFeature Locations 256 “INSDFeature guals> 287 <INSDQualifier>
BEE CINEIDOualifier namermol type“ /INSDUualifier name>
Gon <INZDQualiflsr values>protein</IN3LQualifier valued
HE </INSDOualilfier>
Sel <INSDQualifier id="gl45n>
SR <INSDQualifier name>organism</INSDQualifisr names 232 <INBDQualifier valuersynthetic construct </INSDOualifier value
SOA </INSDQuali fier!» u
Ban </INSDFearture qualss
Bag </INSDFaaturel
Sa </INSDSeg feature-itablex
Hh <INSDSeq sequencs>AARPSARITSRRSDYDY/INSDSeq sequenced
HEE </INEDSeg>
S00 <fSeguenceDalar
BOL <SequenceData ssqvueanoslDNumsern=N38N)>
SOE <iNSDSed>
GOS <INSDSeq lengih>17</INSDSeq lengths
Gig CINZDSeq moltype»AAS/INSDSey moltype>
ELEN <INSDSeq division»PATL/INIDSeg division»
DE <INSDSeq feature-itableX
BOT LIMIDFeaturas 308 <INSDFeature keyssource“/INSDFealure hay» 808 <INSDFeature location»l1..17</INSDFeature location
SEG <INSDFeature guals>
SL <INSDQualifiers
Sz CINZDQuUalifier namermol type /INSDgualifier name> “13 <INSDQualifier valuse>protein</INaTQualifier value»
Aid </INSDOQualijier>
BLD CINSDOualifier id="gldër>
S16 <INSDuualifier namedorganism</INSDOualifisr name>
SQA <INSDQualifier valuersynthetic construct </INSDOualijier value»
WLS <fINSDUualifler»> lis x“/INSDFeature gquals>
Ge </INSDFeature»
GEL </INSDSeg feature-tabler
ER <INSDSeq zsequence>VARPSARITSRRSDYDY</INSDSeq sequence» 522 </INSDSeas IJ
Gad </SecuenceDala»>
Ga <Sequencebata zegvencoaiDNumber=n3S"D>
DIE <iNSDSeg>
GET “INSDSeg length>8</INSDSeg lengths»
GLE “INSDSeq moltype>dAA/INSDSeg moltype>
Ged <INSDSeq division»>PAT</INSDSeg divisions 334 <INSDSeq Ieatureriabier
Zij CINEDFeaturae»
ER <INSDFeature key>source“/INSDFealure key>
G13 <INSDPFeature iocation»l..8</INSDPeeture location»
BIg <INSDFeature quals> u
GE <CINSDQualifiers
GEE <INSDQuUalifier namebmol type /INSDQualifier name> wi <INSDQualifier value>protein</INsDQualifier valued 338 </INSDQualifier> 328 JINSDoualifier id="giá7ns
G40 <INSDQualifier namerorganism“/INSDQuali fier name>
SA <INSDQualifier value»synthetic construct </INSDOQuali fier valuer>
GE <SINSDOualifier>
Ga </INSDFeature duels» id </INSDFeature:> 340 </INSDSeg feature-tabler 334 <INSDSeq zequence>GRTFSTLA</INSDSed sequence 847 “/INSDSeoa>
S48 </Segquaencebata>
LA LSequencebalta seqguancailiicghar=m40% >
SLO CINSDSag»
SLL <INSDSeq length>8</INSDSeq length?
Dh <INSDSeq moltype>AA“/INSDSeq moltype> 352 <INSDSeq divislon»>PATC/INSDSeg divisions 354 <INSDSeq feature-takbled 25% <INSDFeabure>
ES <INEDFeature key>source:/INSDFeature kKey> wij “INSDFeature iccation»>l..8</INSDFeature location»
ERE CINEDFeature gualse
Lh CINBDOualifierns
El <INSDQualifier nams>mol type</INZDQualiifier name
HE <INSDQualifier value>protein</INSDQualifisr value»
BAZ <ATNaSDQuali fis»
Ga <INSDQualifier Ld=Vglagye
God <INSDQualifier namerorganism“/INSDQualifier name>
SEN <INSDOQualifier valuersynthetic construct
<{/INSDQuali jier valuex>
LEE </INSDOualilfier>
GET </INSDPearure guals>
ZEE </INSDFearure»> 343 </INSDEeg Zearture-tabier
ZIE <INSDSeq sequences>GRTFGTLAC/INSDSedq sequence»
ST </INSDSeg>
SEL </SemtenceDala>
SES <HGegquencelbata seguanselilMonhar=nqiv>
Ga LINSDSeq>
Gij <INSDSeq iength>9«/INSDSeg length> 3a <INSDSeq moltype>AA</INSDSeq moliype>
DIT CINBDSeq divisionsPAT</INSDSeg division»
ZI <INSDSeg feabture-tablel
SG <INSDFeature
WR <INSDFearure key source; INSDFeature key»
GR “INSDFeature location>l..9</INSDFealture location
Gi <“INSDPFeacure gqualis>
WEE <INSDQualifier»> zie <INSDguelifier nams>mol type“/INSDQualifier name>
SEG <INSDQualifier valuerprotein</INSDQualifisr value» 326 </INSDQvalifier:
GE <INSDQuaiifler id="gl4S52>
URE CIMNEDOuAalifieyr namerorganism</INSDQualifier name>
GE “INSDoualifier value>synthetic construct </INSDQvalifier valus>
ERI </INSDQualifier>
BEE </INSDFeature guals> 332 </INSDFeature> 837 </INEDSeg featurs-table>
Gud <INSDSeq sequence>IIRNSLSTY</INSDSeq sequencer
Ges </INSDSeg>
Sag <j Gaemquencebata>
Ga “Sequencebata seguanteliMuahao="44%>
Gee <INSDSed> 333 <INSDSeq length2l5</INSDSeq length>
LDS <INSDSeq maltype>AA</INSDSeq molLype»>
LOD <INSDSeq division>PAT</INSDSeg division»
LOE <INSDieq festure-table>
LOS “INSDFeatucer
Lod CINEDFeature keyvsource</INSDFeature key»
GOS “INSDFeabture locabtion>l..15</INSDFeature location»
Lone <INSDIesture guals>
Lon SINSDOualifier>
Lijns <INSDQualifier name>mol type“ /INSDQuali fier names
La <INSDQualifler valuerprotein</INSDQualifisr value>
LOL </INSDQualifiern>
LOLI CINSDQualifler id="gl5ör>
LOLs SINEDOualifiler namerorganism</INSDQualifier name>
TELE <INSDQualifier value>synthetic construct </INSDQualifier value 1314 </INSDQualifier> 1915 </INSDFeature guals> nie </INSDPeatune>
LOT </INSDSeq feature-tabled
Loi <INSDSeqg sequence >AAGRWEAVRTNTPDY </ INSDSeg saguencel
POLE </INSDE ag»
LEG </EegquencebData>
LEL “SequenceData seguencellsNumben="g3e>
LOER <INSDSeq> 1533 <IN3SDSeq Lengihb>8</INSDSeqg lengih>
Lid <INSDSeq moliype>AA</INSDSeq moltype»r>
Loa <IMEDZeq division>PAT«/INSDSeg division
Lone <INSDSeq festure-table>
Laan <INSDFeaturer zs <INSDFeature key>source</INSDFeature key»
Logs <INSDFeature Llocation>l..8</INSDFeature location:
IAG <INSDFesture duels»
LORE SINEZDOualifiso>
LED CINBDOualifier namearmol type</INSDQualifisn name>
LOS <INSDQualifier valuerprotein</INSDQualifier values» 103d </INSDOualifier:>
LOS CINSDQualifler io=vgdSivs
Liss <INSDQUalifiler naemerorganism/INSDOualifier name
IRE <INSDQualifier value>synthetic construct </INSDQualifier valued ijze </INSDQualiËier>
Lijs </INSDFaature cualss» 104 </INSDFeature>
LOA </INSDSeg feature-tabled>
Lads “INSDSeq sequsnce»GRTFSTLA“/INSD3eq zequence>
Las </INSDSeg>
Luda </SeguenceData>
Ian <Sequencalata sopeancallbiumber="449%> ide “INSDSeg» 13a <INSDSey lenghh>8</INEDIag length» 134s <INSDSeq moltype>AA</INSDSeqg moltypes
TOA <INSDSeq division>PAT4/INSDSeg division
LoL CINEDSeq feature-table>
EECA <INSDFeaturer
LEE <INSDFeature kevyrsource</INSDFesture kev: 1as2 <INSDFeature location>l..8</IN3DFeature locations» 154 <INSD¥eature guals>
LOD <INSDOuealijilen»
LNE <INSDQualifier namermol type“ /INSDQgualifier name>
LEET <INSDOQualifier valuesprotein</INSDLQualifier valued
LORE <SINSDOualifier>
REN LINSDGualifiler in="gi52r>
LOE <INSDQualifier name>organism</INSDQualifier names 105 <INSDQualifier value>synthetic construct </INSDQualifier value» 1082 </INSDQualifisan» 1083 </INSDFearure guals>
Lood </INSDFeature>
LOD </INSDSeg faature-table>
Lees “INSDSeq sequerice>GRTSSTLA“/INSDSeq sequence
LOE </INSDSea> eae </SeguenceDatar 16S <SequenceData samencelósubern=Nd5n>
LOG <INSDSeq>
LOL <INSDSeq lengith>8</INSDSeg length»
LOE <INSDSeq moliype>AA</INSDSeq moltyper
LOUS “INSDSeq division»PAT</ INDE division
Luid <INSDSeq feature-itable>
LTD <INSDFeature»> ave <INSDFeature keyssource“/INSDFealure keys» 1a <INSDFeature location»l..B</INIDFeature locations 13s <INSDFeature guals>
LOT <INSDQualifier>
LORE <INSDOQualifier namermol type“ /INSDUualifier name>
LOST v“INSDQualifiern valuesprotein“/INSDgualifier value»
LEE </INSDOualilfier>
OER <INSDQualifier id="gl53n>
Lus <INSDQualifier name>organism</INSDQualifisr names 198% <INBDQualifier valuersynthetic construct </INSDOualifier value» ang </INSDQuali fier!» u
LOST </INSDFearture qualss
LOS </INSDFaaturel u
LSD </INSDSeg feature-itablex
Lea <INSDSeq sequsnce>GRTFSLLA/INSDSeq sequence 1631 </INSDSeg> u 1432 </SeguenceData> 1383 <SequenceData saguencalliunbaer="48" >
Lied <iNSDSed>
Lous CINEDSeq length>9</INSDSeg length»
Long CINZDSeq moltype»AAS/INSDSey moltype>
LEE <INSDSeq division»PATL/INIDSeg division»
Loan <INSDSeq feature-itableX 143% LIMIDFeaturas
Lins <INSDFeature hey>source</IN3DFeature key>
HEE <INSDFeature location»l..9</INSDFeature locations
LIE <INSDFeature quals> u
Ls <INSDguelifiler»
BREE CINEDQUalifiler name>mol type“ /INSDgualifier named
LOG <INSDQualifier valuse>protein</INaTQualifier value»
Las </INSDOuaiifiers
Tiny CINSDOualifier 1d=svgll4any
Lijs <INSDuualifier namevorganism“/INSDQuali fier name> ils <INSDQualifier valuersynthetic construct </INSDOualifier valued
LLG </INSDOualifier:>
TELT </INSDFealture cauels»>
LILLE </INSDFaature» u iia </INSDSeg feature-tabler iid <INSDSeq sequencs>IIRNSISTY/INSDSeq sequence» iis </INSDSeq tiie </SeguenceDatas
LAAT <Sequencebata zaogquencaildiunbhay=m47T00
Lil <iNSDSeg>
LLL “INSDSeq length>9S</INSDSeg length»
Lie “INSDSeq moltype>dAA/INSDSeg moltype>
LL: <INSDSeq division»>PAT</INSDSeg divisions 1142 <INSDSeq Ieatureriabier
Lie: CINEDFeaturae»
Lida <INSDFeature key>source“/INSDFealure key>
Liet <INSDPFeature iocation»l..9S</INSDPeeture locetion>
Le <INSDFearure guals:»
LET <CINSDQualifiers
LEE <INSDouelifier namermol type /INSDOualifier name
LE <INSDQualifier value>protein</INsDQualifier valued 1130 </INSDQualifier>
EC CINSZDOualifiler 1d="gqRB8ve
Lian <INSDQualifier namerorganism“/INSDQuali fier name>
LLS <INSDQualifier value»synthetic construct </INSDQuali fier values
LE </INSDOualifiers
LEE </INSDFeature duels»
LEG </INESDFeatures 1137 </INSD8eq featura-tabler iiae <INSDSeq szequenca>ITRNSISTY</INIDSeq sequencer 113s </INSDSeq> 114C </Segquaencebata>
LAAT LSequencebalta seqguantailiioghar="48% >
Lan CINSDSag»
LLAZ <INSDSeq length>15</INSkSeq Length>
Lida <INSDSeq moltype>AA“/INSDSeq moltype> iiëdh <INSDSeq division>PAT/INSDSeg divisions fide <INSDSeq feature-takbled
Lian <INSDFeabure>
LAAT <INSDFsature key>source</INZDFsature key>
LRA <INSDFearure location>l..15</INSDFeature location
ALG <INSZDFeature gqualsh
TLE: CINBDOualifierns
LL58 <INSDQualifier nams>mol type“ /INSDOualifier name 1152 <INSDQualifier value>protein</INSDQualifisr value» 1154 <ATNaSDQuali fis»
Lins <INSDQualifier ìd="gl5S"2>
LE <INSDQualifier namerorganism“/INSDQualifier name>
Lan <INSDOQualifier valuersynthetic construct <{/INSDQuali jier valuex>
Line </INSDQualifiers
Lins </INSDFreaturs guals> 1180 </INSDFeaturer iad </INSDEeg Zearture-tabier
Lina <INSDSeq sequences>AAGRWEAVRINTPDY</INSDSed sequence
Lies </INSDSeg>
LOA <SBequencelata>
LLS “Sequsrcebala seconde liMNgacev="g8s>
Lies LINSDSeq>
Lian <INSDSeq length>l5</INSDSeq length> 11a <INSDSeq moltype>AA</INSDSeq moliype> 116% <INSDSeq division>PAT/THSDSeq divisions
LITC <INBDZeq feabture-tablel
LATE <INSDFesture>
LATE <INSDFearure key>source</INIDFeature key»
LATE “INSDFearure location». .15</INSDFeature location
LLjd <“INSDPFeacure ouals:»>
LETS <INSDQualifier> iT “INSDgvelifier named>mol type</INSDQualifier name>
Li <INSDuualifier valuerprotein</INSDOualifisr valus>
Tas </INSDQualifisan»
LLS <INSDQuaiifler id="gl5Ys2>
LiRG <INSDOQualifier namerorganism“/INSDQualifier name
PRE CINEDOUalifisr value>synthetie construct </INSDQualifier value>
Lie </INSDQualifier> i182 </INSDFeature guals>
Pid x“/INSDFeacturex
LAE </INEDEeg feature-tabled>
LAE <INSDSeq sequence>AAGRWEAVRTDTPDY</INSDSeg sequencer
Lan </INSDSear
LLS <j Gaemquencebata>
Tied <Gequencelala seguantellManhao=2847>
Lise <IiNSsDseqd>
Lind <INSDSeq length2l5</INSDSeq length>
Iisa <INSDSeq moliyperAA/INIDSeq moltype> rien <INSDSeq division>PAT</INSDSea divisions
Lied <INSDieq festure-table>
LL <INSDFeature>
EES “INSDFearture keyrsourced/INIDFeaturs Key»
RRC <INSDFeecture lcocaition>l.. 15</IN3DFeature location» ilse <INSDFeature guals> u ize CINEDOualifier>
Lene <INSDQualifler named>mol type“ /INSDQuali fier names 12a <INSDQualifier valuesprotein/INSDqvalifier valued oan </INSDQuali fier!»
LEDS CINSDQualifiler id="gl5gv>
Ladd “INSDOualifier namevorganism /INSDQualifier named zal <INSDQualifier value>synthetic construct </INSDQualifier values
LEDá </INSDQualifier> - lua </INSDFaature quals> 1208 </INSDFeature> - 1209 </INSDSeg feature-tabhlad
LLG <INSDSeqg sequence >AAGRWEAVRTITPDY</ INSDSeg saguencel
Lal </INSUE aa»
TELE </EegquencebData>
Lai “SequenceData seguencellsumben="SL®>
IES <INSDSeqg> bell <INSDSeg lengith>l3L</INSDSeq length»
RENE <INSDSeq moelivpe>AA/INSDSeq moltyper ral <INEDZeq division>PAT</INSDEsqg division» ital <INSDSeq festure-table>
LES <INSDFeature»
EEG <INSDFeature key>source</INSDFeature key»
Lat <INSDPeature locations. .131</INSDF Feature location>
LEER <INSDFesture duels»
Lia) JINSDOualifier>
Rt) <INSDOQualifier name>mol type“ /INSDQguali fier name»
Leal <INSDQualifier valuerprotein</INSDgualifier values
Laze <fINSDUualifler»>
ZET CINSDQualifler io=vgdiSsys
EEn <INSDQUalifiler naemerorganism/INSDOualifier name
LE <INSDQualifier value>synthetic construct </INSDQualifier valued
LER </INSDQuali jier
LE: </INSDFeature gualss» 223: </INSDFeature>
L233 </INSDSeg feature-tabled>
Ed <INSDSeq sequsncer
EVQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSCISSSDGSTYYADSVKGRFTISSDN
AKNTVYLQMNSLRPEDTAVYYCAAVPRTMYSRWGCGVRPYYYGMDYWGKGTLVTVSS
</INSDSeg sequences
IES </INSDSeg>
IEEE </SeguenceData 1337 <Sequencebata zegvencainNunmber=nB2tD 123 <INSDSeq>
Lass “INSDSeqg length>131</INSDSeq lengths
LEA <INSDSeq moltype>dAA/INSDSeg moltype>
Ld: <INSDSeq division»PAT</INSDSeg diviglon» 1E42 <INSDSeq Ieatureriabier 1dd4n CINEDFeaturae»
Lis <INSDFeature key>source“/INSDFealure key>
LEAL <IMaDFeature location>l..131</INSDFeature location» 124d <INSDFearure guals:»
LET <CINSDQualifiers
TEAR <INSDQualifier name>mol_ type /INSDQualiiier named izin <INSDgualifier valie>protein</INSDOQualifier values» 1550 </INSDQualifier> u
TES CINSZDOualifiler 1d="gql Sgn»
LEBD <INSDGualifier namerorganism</INSDQualifisr name>
Lass <INSDQualifier value»>synthetic construct </INSDQualifier valuer tana <SINSDOualifier>
ELE </INSDFeature duels»
Lamia </INSDFeature>
TEST </INSD8eq featura-tabler 1dhe <INSLSeq zequenca>
EVQLVESGGGLVQAGGSLRLSCAASGFTFDDYAIGWFRQAPWKEREGVSCISSSDGSTYYADSVKGRFTISSDN
AKNTVYLQMNSLRPEDTAVYYCAAVPRTMYSRWGCGVRPYYYGMDYWGKGTLVTVSS
</INSDSeq sequencer
Tan </INSUE aa»
LEE </EegquencebData>
Ll “SequenceData seguencellsumben="S3®> 1852 <INSDSeq> idan <INSDSeq lengihrl31</INSDSeq lengih>
Ldn <INSDSeg moltype>AA</INSDSeqg moliype:» 128% <INSDSeqg divisiom>PAT:/INSDSeg divisions
L288 <INSDSeq festure-table>
Laan <INSDFeature» zen <INSDFeature key>source</INSDFeature key»
Lid <INSDFeature focation>l..131</IN3DF¥eature location>
LETS <INSDFesture duels»
LEL <INSDOualifier>
LED <INSDgualifier namexmol type</INSDQualifier name>
LETS <INSDQualifier valuerprotein“/INSDgualifier value» 12rd </INSDOualifier:>
LENS CINSDQualifler iQ=Ngl8LN>
Lie <INSDQualifier name>organism</INiDQualifier named
Lai <INSDQualifier value>synthetic construct </INSDQualifier valued igre </INSDQuali jier rave </INSDFeature guals> 1280 </INSDFeature>
Lani </INSDSeg feature-tabled>
ERA “INSDSed sequencer
EVQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPGKEREGVSCISSSDGSTYYADSVKGRFTISSDN
AKNTVYLQMNSLRPEDTAVYYCAAVPRTMYSRWGCGVRPYYYGMDYWGKGTLVTVSS
</INSDSeg sequences
AEE c/rNeDSeqy
LEE </SeguenceDatas
LER <Sequencebata zaogquencaildiunbhar="84"0 lane <INSDSeq>
Lawn CINZDSeq length>131</INSDSeq lengths zed <INSDSeq moltype>dAA/INSDSeg moltype>
LEED <INSDSeq divislon»PAT</INIDSeg division
LxB <INSDSeq Ieatureriabier
LE BL CINEDFeaturae» rasa <INSDFeature key>source“/INSDFealure key>
Lass <IMaDFeature location»l..131</INSDPeaiure location»
LE <INSDFearure guals:» aul <CINSDQualifiers pach <INSDQualifier name>mol_ type /INSDQualiiier named
Lal <INSDQualifier valie>protein</INSDOQualifier values» 1EE0 </INSDQualifier> - - 1E38 CINSZDOualifiler 1d="ql8ave ize <INSDGualifier nansrorganism“/INSDQuali fien name>
La <INSDQualifier value»>synthetic construct </INSDQualifier values
TELE <SINSDOualifier>
L303 </INSDFeature duels» 1504 </INSDFeature> 1305 </INSD8eq featura-tabler ijje <INSLSeq zequenca>
EVQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSCISSSDGSTYYADSVKGRFTISSDN
AKNTVYLQMNSLRPEDTAVYYCAAVPRTMNSRWGCGVRPYYYGMDYWGKGTLVTVSS
</INSDSeq sequencer
LRT </INSUE aa» xu </EegquencebData>
LS08 “SequenceData seguencellsumben="SSn> isis <INSDSeq> 131A <INSDSeq lengihrl31</INSDSeq lengih>
TILE <INSDSeg moltype>AA</INSDSeqg moliype:»
TALE <INSDSeq divisior>PAT</INSDSeq division»
LLG <INSDSeq festure-table>
LEDS <INSDFeature»
TELE <INSDFeature key>source</INSDFeature key» sid <INSDFeature focation>l..131</IN3DF¥eature location isis <INSDFealure guals> zie CINSDQUalifler> 220 <INSDgualifier namexmol type“/INSDgualifier name> 1320 <INSDQualifier valuerprotein</INSDQualifier value»
LE <fINSDUualifler»>
LEES CINSDQualifler in=Ngl83>
Lake <INSDQualifier name>organism</INiDQualifier named
LEER <INSDQualifier value>synthetic construct </INSDQualifier valued ijze </INSDQuali jier rae </INSDFeature guals> 13238 </INSDFeature>
Lan </INSDSeg feature-tabled>
LEG “INSDSed sequencer
EVQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSCISSSDGSTYYADSVKGRFTISSDN
AKNTVYLQMNSLRPEDTAVYYCAAVPRTMYSRWGCGVRPYYHGMDYWGKGTLVTVSS
</INSDSeg sequences izz: </INSDSegs
Lid </SequenceDatas
LIES {Sequencebata zaguencaidilunbay="538"0
L3G <INSDSeq>
LERE CINZDSeq length>122</INSDSeq lengths
RCICE <INSDSeq moltype>dAA/INSDSeg moltype> 155 <INSDSeq divislon»PAT</INIDSeg division 13a <INSDSeq Ieatureriabier 122s CINEDFeaturae»
Lise <INSDFeature key>source“/INSDFealure key>
LEA <IMaDFeature location>l..122</INSDFeature location»
LAE <INSDFearure guals:»
Laas <CINSDQualifiers
Txdd <INSDQualifier name>mol_ type /INSDQualiiier named
L545 <INSDQualifier value>protein</INsDQualifier valued lida </INSDQualifier> 154 CINSZDOualifiler 1d="gql eens 1248 <INSDGualifier namerorganism</INSDQualifisr name>
Ide <INSDQualifier valuersynthetic construct </INSDQuali fier values
LBG <SINSDOualifier>
TELE </INSDFeature duels»
L552 </INSDFeature> 1382 </INSD8eq featura-tabler 1354 <INSLSeq zequenca>
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAIWWSGGASQYADSVKGRFSISKDN
AKNTMFLQMNSLKPEDTAVYYCAAGPMFSMDYRRVNYWGQGTQVTVSS</INSDSeg sequence
Lans </INSDSeg> u
RL <j Gaemquencebata>
ERC <Gequencelala segquenceliNuubsc=NSyN> 15a <IiNSsDseqd> 135% <INSDSeq length>122</INSDSeq length> 1360 <INSDSeg moliype>AAd/INSDSeqg moltype»> aad <INSDSeg division>PAT</INSDSeq division»
Lied <INSDSeq feature-table>
Lis <INSDFeature>
Lied CINEDFeature keyvsource</INSDFeature key»
LEED “INSDFeabture locabtion>l..122</IN3DFeature location» isde <INSDIesture guals> 1357 <INSDOualifier> 1588 <INSDQualifler named>mol type</INSDRualifizp names
Laas <INSDQualifier valuevprotein</INSDQualifisn wvalus>
IAT </INSDQuali fier!»
LOL CINSDQualifiler id="gls5r>
LRE v“INSDQualifier namerorganism</IN3DQualifier name>
EYE <INSDQualifier value>synthetic construct </INSDQualifier value» ist </INSDQualiËier> u
LS <SINSDFaature guals> are </INSDFeature> - ian </INSDSeg feature-tabhlad
Lig <INSDSeq sequence’
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAATWWSGGASQYADSVKGRFSISKDN
AKNTMFLQMNSLKPEDTAVYYCAAGPMFSMDYRRVNYWGQGTQOVTVSS</INSDSeg sequence» 157s </INSDSeg> 13EG </SeguenceDatar 1381 <Seduencepata sopuencallilumber="38"%
LEL <INSDSeq»>
LIER <INSDSeq lengih>122</INSDSeq iength»
Lind <INSDSeq moliype>AA</INSDSeq moltype»
TEED CINZDSeq divizion»PAT</INSDS=q division»
LEE <INSDSeq feature-itable>
Laud CINZEDFezture>
Lins <INSDFesture keyrsource“/INSDFealure key» 1388 <INSDFeature location>l..122</INSDFeaturs location
Lis <INSDFeature guals> ian <INSDQualifier> aan <INMEDQualifier namermol type</INsDoualifier name>
Taus CINZDOQuallifier valuesprotein“/INSDgualifier value»
Lsdd </INSDOualilfier>
Leh <INSDQualifier id="glëë>
Lize <INSDQualifier name>organism</INSDQualifisr names 1a <INBDQualifier valuersynthetic construct </INSDOualifier value» 138s </INSDQuali fier!» ian </INSDFearture qualss
LAL </INSDFaaturel u
Laud </INSDSeg feature-itablex
LAGE <INSDSeq sequencer
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAIWWSGGASQYADSVKGRFSISKDN
ARNMMFLOMNSLKPEDTAVYYCAAGPMF SMDYRRVNYWGQGTQVTVSS</ I NSLS aq seguence> 1403 </INSDSeg> 1404 </Segquaencebata>
LAOS <SSequencebala seqguantailiioghar="88%
Lane <INSDSeq> any <INSDSeq length>122</INSD3eq Length>
Lan <INSDSeq moltype>AA“/INSDSeq moltype> 1408 <INSDSeq divislon»>PATC/INSDSeg divisions
ALG <INSDSeg feature-table> IJ
LEE <INSDPeature> ral <IMaDFesature key>source“/INSDFeature key>
ALS <INSDFearure location>l..122</INSDFeatiure location»
Laid <INSDFeature quals> IJ
TALS <INSboualifier?
Lidl <INSDQualifier name>mol type“ /INSDQualifier name» 14 <INSDQualifier value>protein</INSDQualifisr value» i418 </INSDQuali jier ijle <INSDQualifier ìd="gl87N2> 1420 <INSDQualifier namevorganism“/INSDQualifier name>
LAT <INSDOQualifier valuersynthetiec construct <{/INSDQuali jier valuex>
Laze </INSDOualilfier>
HE </INSDFreaturs guals> 1474 </INSDFeaturer u
Lint </INSDEeg Zearture-tabier
Lize <INSISeq sequence>
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAIWWSGGASQYADSVKGRFSISKDN
AKNTMFLQMNSLKPEDTAVYYCAAGPVFSMDYRRVNYWGQGTQOVTVSS</ INSDSeg sequencer
Tan </INSUE aa»
Taxa </EegquencebData>
Lak “SequenceData seguencellNumben="89r> 1430 <INSDSeq>
LAZ <INSDSeq lengibh>122</INSDSeq lengih>
Lid <INSDSeq moliype>AA</INSDSeq moltype»r> 14S <INEDZeq division>PAT</INSDEsqg division»
LAG <INSDSeq festure-table>
LARD <INSDFeature»
TAN <INSDFeature key>source</INSDFeature key»
Last <INSDFeature focation>l..122</INSDF¥eature location 14a <INSDFesture duels» ijs <INSDQualifier> 1440 <INSDOQualifier name>mol type“ /INSDQguali fier name» 1440 <INSDQualifier valuerprotein</INSDQualifier value»
LAAT </INSDOualifier:>
Taal CINSDQualifler io=vglaly>
Tadd <INSDQUalifiler naemerorganism/INSDOualifier name adn <INSDQualifier value>synthetic construct </INSDQualifier valued 1448 <ATNaSDQuali fis» aay </INSDFeature guals» 1448 </INSDFeature>
Lad </INSDSeg feature-tabled>
TAU “INSDSed sequencer
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAIWWSGGASQYADSVKGRFSISKDN
AKNTMFLQMNSLKPEDTAVYYCAAGPMFSMDYTRVNYWGQGTQOVTVSS<./INSDSeg zequence> 1451 </INSDSeg> 1452 </SeguenceData> 1453 <SequenceData saguencaliunbaer="a3" >
Ladd <iNSDSed>
LAS <INSDSeq length>122</INSDSeq length»
TALE CINZDSeq moltype»AAS/INSDSey moltype>
TAT <INSDSeq diviglon»PAT</INSDSeqg division
Lane <INSDSeq feature-itableX 1459 <INSDFeature> 1880 <IN3DFeature key>source“/INSDFeature key>
Lia <INSDFeature location»>1..122</INSDFeacture location»
Lie: <INSDFeature guals>
LAGS <INSDQualifiers
Tad <INSDQualifier name>mol type“ /INSDQualifier name»
TAS <INSDQualifier valuse>protein</INaTQualifier value» 1a </INSDOualifier> u aay CINSDOualifier id="glós0>
Lijns <INSDQualifler namedorganism</INSDQualifisr name> dae <INSDQualifier valuersynthetic construct </INSDOualijier value»
LATS </INSDOualifier:>
ATI “{INSDFeature quals>
LATE </INSDFeature» u
LAD </INSDSeg feature-tabler
Lid <INSDSeq sequence>
EVQLVESGGGLVQAGDSLRLSCTPSGRTFSINVVGWFRQAPGKEREFVAAIWWSGGASQYADSVKGRFSISKDN
AKNTMFLQMNSLKPEDTAVYYCAAGPMFSMDYRRVNHWGQGTQOVTVSS/INSDSeq sequence»
Lan </INSDSeg> iat <SBequencelata>
LAT “Sequsricebala seguanselilMonhar=n88Y >
TATE LINSDSeq>
LATE <INSDSeq iength>125-/INSDSeq Lengih>
Lind <INSDSeq moltype>AA</INSDSeq moliype> aR CINBDSeq division»PAT</IN3D3eq division»
LEL <INSDSeg feabture-tablel 1483 <INSDFeature>
Lag <INSDFeature keyrsource</INIiDiFezaturs key>
LAR CINZDFeature location>l..125</IN3SD¥Feature location»
TAT <“INSDPFeacure gqualis>
TART <INSDQualifier»>
Lijns “INSDgvelifier named>mol type</INSDQualifier name> ase <INSDQualifier valuerprotein</INSDQualifisr value» 1480 </INSDQuali fier: an <INSDQuaiifler i1d=vgliovs fas CINMEDOualifier namerorganism</INSDQualifier name dvs CINEDOUalifisr value>synthetie construct </INSDQualifier value>
Land </INSDQualifier> 1435 </INSDFeature guals> 143s x“/INSDFeacturex 148 </INSDSeg feature-tabled> 148g <INSDSeq sequence
EVQLVESGGGLVQAGGSLRLSCTASGRTFSSYHMAWFRQAPGKERE SVAAITRGGGVTYYADSVKGRFTISRDN
AKNTVYLQMNSLKPEDTAVY YCAADAIWNQVRWMETKYTY SGQGTQVTVSS</ IN33 eg sequence 1443 <SINBDE eg
Lie </SeguenceData> 1&0: <Sequencalata sopeancallilumber="88%> 15892 CINIDSeq>
Lin: <INSDSeq lengith>125</INSDSeq length»
R04 <INSDSeq moliype>AA/INSDSeq moltypbe»>
TROL CINEDSeq division>PAT</INSDEaq division» na “INSDSeq fealure-table>
LL <INSDFeaturer
Lua <INSDFeature key>source</INSDFeature key» 150% <INSDFesture location>l..125</INSDieaturs location» 130 <INSD¥eature guals>
LSL <INSDoualifier>
ALE <INSDQualifier namermol type“ /INSDgualifier name>
LEL Ss <INSDOQualifier valuesprotein</INSDLQualifier valued
ELA <SINSDOualifier>
LEE LINSDGualifiler pa=sngdiiv>
ERE <INSDQualifier name>organism</INSDQualifier names
Ln <INSDQualifier value>synthetic construct <SINSDOualifier value» ijle </INSDQvalifier: u
HRY </INSDFeature quals>
LEG </INSDFeature> u
EL </INSDSeg faature-table>
Lede <“INSDSeq sequenced
EVQLVESGGGLVQAGGSLRLSCTASGRTFSSYHMAWFRQAPGKERE SVAAIARGGGVTYYADSVKGRFTISRDN
AKNTVYLQMNSLKPEDTAVYYCAADAIWNQVRWMETKYTYSGQOGTQOVTVSS</ INSDSeq sequence 15232 <STHEDS eq 1nad </SequenceDatas han {Sequencebata zaguencaidiiunbay="84%0 tho <INSDSeq> nen CINZDSeq length>127</INSDSeq lengths
LEE <INSDSeq moltype>dAA/INSDSeg moltype>
LLdS <INSDSeq division»PAT</INSDSeg diviglon»
Laan <INSDSeq Ieatureriabier 152% CINEDFeaturae» 1532 <INSDFeature key>source“/INSDFealure key>
LEE <IMaDFeature location>l..127</INSDFeature location»
LAS <INSDFearure guals:»
Len <CINSDQualifiers
ERE <INSDQualifier name>mol_ type /INSDQualiiier named
Lan <INSDQualifier value>protein</INsDQualifier valued inss </INSDQuali fier» 1558 CINSDOuaLLTLier LAELIA 15340 <INSDGualifier namerorganism</INSDQualifisr name>
LRA <INSDQualifier valuersynthetic construct </INSDQualifier value»
TEAR <SINSDOualifier>
TRA </INSDFeacure duals»
Lia </TNIDFezturer 1545 </INSD8eq featura-tabler isde <INSLSeq zequenca>
EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS< /INSDSeg sequence» 1547 </INSDSeg> u
Lid <j Gaemquencebata>
LAR <Gequencelala seguantellianha o="E8Y >
Lise <IiNSsDseqd> 155: <INSDSeq length»r127</INIDSeq length> 1352 <INSDSeg moliype>AAd/INSDSeqg moltype»> 1353 <INSDSeg division>PAT</INSDSeq division»
Lisa <INSDSeq feature-table>
Ls <INSDFeature>
Lanne CINEDFeature keyvsource</INSDFeature key»
Tan “INSDFeabture locabtion>l..127</IN3DFeature location»
LuER <INSDIesture guals> 1558 <INSDOualifier> 1550 <INSDQualifler named>mol type</INSDRualifizp names hal <INSDQualifier valuevprotein</INSDQualifisn wvalus> 1har </INSDQuali fier!»
LEGS CINSDQualifiler iad=vqliivs
Lead CINZDQuallfier namerorganism</IN3DQualifier name>
Lael <INSDQualifier value>synthetic construct </INSDQualifier value 18a </INSDQualifier> - ina <SINSDFaature guals> han </INSDFeatune> - hey </INSDSey feahure-table>
LRG <INSDSeq sequence’
EVQLVESGGGLVRAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLOMNNLKPEDTATYYCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS </INEDS eg sequenced
Ln </INSDSeg> 1572 </SeguenceDatar ijf <Seduencepata samencoalóNuber="gón>
Lsa <INSDSeq>
LL <INSDSeq lengith>127</INSDSeq iength»
LEE <INSDSeq moliype>AA</INSDSeq moltyper
RET CINZDSeq divizion»PAT</INSDS=q division»
TLR <INSDSeq feature-itable>
LETS <INSDFearture:» 15E <INSDFesture keyrsource“/INSDFealure key»
LEL <INSDF Feature locationsl..127</INSDFealture location»
BER <INSDFeature guals>
SES <INSDQualifier>
EERE <INMEDQualifier namermol type</INsDoualifier name>
Lael CINZDOQuallifier valuesprotein“/INSDgualifier value»
TLE </INSDOualilfier>
LET <INSDQualifier id="gljar> 1558 <INSDQualifier name>organism</INSDQualifisr names 1388 <INBDQualifier valuersynthetic construct </INSDoualifier valuer ieu </INSDQuali fier!»
LAST </INSDFearture qualss
Lau </INSUFeature -
Lud </INSDSeg feature-itablex
EE <INSDSeq sequencer
EVQLVESGGGLVEAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLOMNNLKPEDTATY YCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS</ INSDS eq sequence
Lan </INSDSeg> = aed </Segquaencebata>
Laan LSequencebalta seqguantailiioghar="8¥Y >»
Thal CINSDSag»
Lud <INSDSeq length>127</INSD3eq Length>
LaLa <INSDSeq moltype>AA“/INSDSeq moltype>
Len: <INSDSeq division>PAT/INSDSeg divisions 1452 <INSDSeg feature-table> IJ 1H <TNSDFeaburae> 1504 <IMaDFesature key>source“/INSDFeature key>
LOOT <INSDFeature location>l..127</INSDFeature locations
Leas <INSDFeature quals> IJ
TEL <INSboualifier? ene <INSDQualifier name>mol type“ /INSDQualifier name»
LEDS <INSDQualifier value>protein</INSDQualifisr value» iel </INSDQuali jier
Lal <INSDQualifier ìd="gl75N2> iad <INSDQualifier namevorganism“/INSDQualifier name>
Zels <INSDOQualifier valuersynthetiec construct <{/INSDQuali jier valuex>
End </INSDOualilfier> ies </INSDFreaturs guals> igre </INSDFeaturer - 1617 </INSDEeg Zearture-tabier 1a LINSDSeg sequenced
EVQLVESGGGLVQOAGGSLRLSCAAPGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS“/INSUS2eq sequence»
Tal </INSUE aa»
TEE </SegquenceData>
Lak “SequenceData seguencellNumben="&g®> 1882 <INSDSeqg> 1633 <INSDSeq lengihrl27</INSDSeq lengih>
REAR <INSDSeq moliype>AA</INSDSeq moltype> 162% <INEDZeq division>PAT</INSDEsqg division»
Lend <INSDSeq festure-table> tan’ <INSDFeaturer exe <INSDFeature key>source</INSDFeature key»
Leds <INSDFeature focation>l..127</INSD¥eature location>
Leaf <INSDFesture duels»
Leal JINSDOualifier> ió3ë <INSDOQualifier name>mol type“ /INSDQguali fier name» 18733 <INSDQualifier valuerprotein</INSDQualifier value»
Lead <fINSDUualifler»>
Taal CINSDQualifler io=ngliays
TERE <INSDQUalifiler naemerorganism/INSDOualifier name
Len <INSDQualifier value>synthetic construct </INSDQualifier valued 1628 <ATNaSDQuali fis» -
Lai </INSDFeature gualss» 1840 </INSDPeature>
Leal </INSDSeg feature-tabled>
Lodz “INSDSed sequencer
EVQLVESGGGLVQOAGGSLRLSCAASGRIVGIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS</INSDSeq sequenced iad </INSDSeg>
Ladd </SeguenceData> 18a <SequenceData saguencalMiiunbaer="88" > 184d <iNSDSed>
Lean <INSDSeq length>127</INSDSeq length»
Leds “INSDSeqg moliypevAA</INSDSeq moltype>
TEAR <INSDSeq diviglon»PAT</INSDSeqg division
Lana <INSDSeq feature-itableX rah: <INSDFeaturer> 1452 <IN3DFeature key>source“/INSDFeature key>
Les <INSDFeature location»l..127</INSDFeacture location
Lad {INEDFeature gualis>
LEE <INSDQualifiers
Lane <INSDQualifier name>mol type“ /INSDQualifier name»
TEL <INSDQualifier valuse>protein</INaTQualifier value»
Lene </INZDOualifier> u rasa CINSDOualifier id="gij7n> read <INSDQualifler namedorganism</INSDQualifisr name> ian <INSDQualifier valuersynthetic construct </INSDOualijier value»
Laan </INSDOualifier:>
Lens “{INSDFeature quals>
Leed </INSDFeature»> u
LEES </INSDSeg feature-tabler 1888 <INSDSeq sequence>
EVQLVESGGGLVQAGGSLRLSCAASGRIVSTYSMAWFRQAPGKVREFVAATISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPED TATY YCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS< / INSDS ey sequence» 188 </INSDSeq>
LEE <SBequencelata>
Lens “Sequsrcebala seguansellMonhar=nTaY >
LETH LINSDSeq>
LEE <INSDSeq length>127/INSDSeq Lengih> e772 <INSDSeq moltype>AA</INSDSeq moliype> 14773 CINBDSeq division»PAT</IN3D3eq division»
Lad <INSISeq feature-table>
LHL <INSDFeature
Leid <INSDFeature keyvsource</INSDFeature key>
LET CINZDFeature location>l..127«</IN3SDFeature location»
LEE <“INSDPearure duals»
Les <INSDQualifier»>
Land “INSDgvelifier named>mol type</INSDQualifier name> rani <INSDQualifier valuerprotein</INSDQualifisr value» 1680 </INSDQuali fier: 188s <INSDQuaiifler id="g17S8"2>
Leid <INSDOQualifier namerorganism“/INSDQualifier name
LEED CINEDOUalifisr value>synthetie construct </INSDQualifier value>
Lesa </INSDQualifier> ian’ </INSDFeature guals> aaa x“/INSDFeacturex ==
LOES </INSDSeg feature-tabled>
Lang <INSDSeq sequence
EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAYFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPED TATY YCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS</ INSU ag sequence
TEL <SINEDE E>
Len? </SeguenceData>
LE: <SedquenceData sequencernNumbLer=NJ Les i634 CINIDSeq>
REA <INSDEeq lengith>127</IN3DSeq length»
Lang <INSDSeq moltype>AA< /INSDSeg moltype:»
Loa CINEDSeq division>PAT</INSDEaq division»
Lens “INSDSeq fealure-table>
LSD LINSDFeature:»
Ln <INSDFeature key>source</INSDFeature key» 170: <INSDFesture location»l..127</INSDFeature location» 12 <INSD¥eature guals>
LDS <INSDoualifier>
Ld <INSDQualifier namermol type“ /INSDgualifier name>
LOE CINMEDOualifier valuesprotein</INSDLQualifier valued
LEDE <SINSDOualifier>
TEL LINSDGualifler ia=sngdisy> vias <INSDQualifier name>organism</INSDQualifier names 170% <INSDQualifier value>synthetic construct <SINSDOualifier value»
ITA </INSDQualifisan» -
LIL </INSDFeature quals> iE </INSDFeature>
TELE </INSDSeg faature-table> ind <“INSDSeq sequenced
EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGGETDYVDYVQGRFTASRDN
AKNTMY LOMNNLKPEDTATY YCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS</ INIDSeyg sequence
ITA </INSDSea: rie </SeguenceDatas
LA <Sequencebata zegvencaiDNunmber=nNj2ND
Lil <iNSDSeg>
LLS “INSDSeqg length>127</INSDSeq lengths
Lei “INSDSeq moltype>dAA/INSDSeg moltype>
Lj: <INSDSeq division»PAT</INSDSeg diviglon» 17E2 <INSDSeq Ieatureriabier 17a CINEDFeaturae»
Lia <INSDFeature key>source“/INSDFealure key>
LEL <IMaDFeature location»l..127</INSDPeaiure location» ine <INSDFeacure guals»
LET <CINSDQualifiers
LEE <INSDQualifier name>mol_ type /INSDQualiiier named
Lies <INSDQualifier value>protein</INsDQualifier valued 1730 </INSDQualifier>
LUZ JINSDoualifier id="gi8012
LEED <INSDGualifier namerorganism</INSDQualifisr name>
LE <INSDQualifier valuersynthetic construct </INSDQuali fier values
LE </INSLQualifiers
ERE </INSDFeature duels»
Liss </INSDFeature>
ITE </INSD8eq featura-tabler ivan <INSLSeq zequenca>
EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETQYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS< /INSDSeg sequence»
LIE </INSDSeg> u
Lia <j Gaemquencebata>
LEA <Gequencelala segquenceliNuubsc=NIi3N>
Lia <IiNSsDseqd> 17a <INSDSeq length»r127</INIDSeq length> 1744 <INSDSeg moliype>AAd/INSDSeqg moltype»> iran <INSDSeg division>PAT</INSDSeq division» hae <INSDSeq feature-table>
Lav <INSDFeature>
ES CINEDFeature keyvsource</INSDFeature key»
TEAR “INSDFeabture locabtion>l..127</IN3DFeature location»
LPOG <INSDIesture guals>
LB! SINSDOualifier>
LB <INSDQualifler named>mol type“ /INSDQuali fier names
LDS <INSDQualifier valuerprotein</INSDQuali fien value»
LRA </INSDQuali fier!»
TER CINSDQualifiler iad="gql8iv>
Line CINZDQuallfier namerorganism</IN3DQualifier name>
VERE <INSDQualifier value>synthetic construct </INSDQualifier value»
TES </INSDOualifise> 1ise <SINSDFaature guals>
TAG </INSDFeatune> - ia </INSDSey feahure-table>
LISE <INSDSeq sequence’
EVQLVESGGGLVQOAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLOMNNLKPEDTATYYCAKPQSHSYDGSWRRASAYDDWGQGTQVTVSS </INEDS eg sequenced
Lian << INEDSeg>
Lind </SeguenceDatar ian <Seduencepata samencoalóNuber="Pa4n>
Lind <INSDSeq>
Lav <INSDSeq lengith>127</INSDSeq iength»
Lid <INSDSeq moliype>AA</INSDSeq moltype»
LUSS CINZDSeq divizion»PAT</INSDS=q division»
LEG <INSDSeq feature-itable>
LFF <INSDFearture:»
Ld <INSDFesture keyrsource“/INSDFealure key»
Luis <INSDF Feature locationsl..127</INSDFealture location»
A <INSDFeature guals>
LUL <INSDQualifier>
LIFE <INSDQualifier namermol type“ /INSD{ualifier name>
EE CINZDOQuallifier valuesprotein“/INSDgualifier value»
ETE </INSDOualilfier>
LITE <INSDQualifier ial=vgllav>
LTES <INSDQualifier name>organism</INSDQualifisr names
LER <INBDQualifier valuersynthetic construct </INSDOualifier value» ial </INSDQuali fier!»
LIBS </INSDFearture qualss
Lea </INSUFeature -
TEE </INSDSeg feature-itablex
Lies <INSDSeq sequencer
EVQLVESGGGLVQOAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPQSHFYDGSWRRALAYDDWGQOGTQOVTVSS</INSDSeg sequence
LI <{INSDSeg> u
LUSS </SegusncsDara>
Lin LSequencebalta seqguancaliliioghar=nTEY >
LSD <INSDSeq>
TE <INSDSeq length>127</INSD3eq Length>
Lise <INSDSeq moltype>AA“/INSDSeq moltype>
Ten <INSDSeq divislon»>PATC/INSDSeg divisions 1754 <INSDSeg feature-table> IJ
LD <INSDFeature>
Lin <IMaDFesature key>source“/INSDFeature key>
LIST <INSDFeature locationsl1..127</INSDFeature location:
Lies “IiNSDFeanure gqualsh
TERR <INSboualifier?
Lain <INSDQualifier name>mol type“ /INSDQualifier name»
LED! <INSDQualifier valuerprotein</INSDQualifier value» uaz </INSDQuali jier 18a <INSDQualifier 1d=Vgladvs
LSA <INSDQualifier namevorganism“/INSDQualifier name>
ADs <INSDOQualifier valuersynthetiec construct <{/INSDQuali jier valuex>
THLE </INSDOualilfier>
Lean </INSDFreaturs guals>
Lene </INSDFeaturer - inns </INSDEeg Zearture-tabier
LELO <INSISeq sequence>
EVQLVESGGGLVQOAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPQSHFYDGSWRRASAYGDWGQGTQVTVSS“/INSUS2eq sequence»
Lani </ INSEE o>
Tei </EegquencebData>
Laid “SequenceData seguanoelidhunbhe o=YT809>
Eid <INSDSeq>
IHG <INSDSeg lengith>127</INSDSeq length» inie <INSDSeg moltype>AA</INSDSeqg moliype:»
LSL <INEDSeq divisior>PAT«</INSDSeg division»
Leie <INSDSeq festure-table>
Tan <INSD¥eature»
Lee <“INSDFearure key>source</INSDFeature key»
Lei <INSDFeature location>l..127</INSDFeature location
Lied <INSDFesture duels» ee! <INSDQualifier> 188d <INSDOQualifier name>mol type“ /INSDQguali fier name» 182s <INSDQualifier valuerprotein</INSDgualifier values
Leze <fINSDUualifler»>
LBT CINSDQualifler io=vglfdys»
LEE <INSDQUalifiler naemerorganism/INSDOualifier name
Laws <INSDQualifier value>synthetic construct </INSDQualifier valued
R20 <ATNaSDQuali fis»
HEA </INSDFeature guals» ien: </INSDFeature>
TRE </INSDSeg feature-teblex>
LEE <INSDSeq sequsncer
EVQLVESGGGLVQOAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPQSHFYDGSWRRASAYDDCGQGTQVTVSS</INSDSeg sequence)» 1535 </INSDSegy u inde <fSeguenceDalar env <SequenceData saguencalliunber="FFns 1938 <iNSDSed>
LOSE <INSDSeq length>127</INSDSeg length»
LBA CINEDSeqg moliypevAA/INSDSeg moltype>
Tad <INSDSeq division»PATL/INIDSeg division»
Ladd <INSDSeq feature-table> 1542 CINEDEeatures indd <INSDFeeture keyrsource“/INSDFeature key> ean <INSDFeacture location>l..127</INSDFeaturs Localion> 194d <INZDFeature qualss u
LAT <“INSDQualifier:»
Laas SINEDOualifiler name>mol type</IN3DQualifier name»
Lain <INSDQualifier valuse>protein</INaTQualifier value»
LEO </INZDOualifier> u
ERI CINSDOualifier id="gign0>
TEE <INSDuualifier namedorganism</INSDOualifisr name> 1953 <INSDQualifier valuersynthetic construct </INSDOualijier value»
Land <fINSDUualifler»>
Tani </INSDFealture guals>
Tana </INSDFealure»>
Lan </INSDSeg feature-tabler 1369 <INSDSeq sequence>
EVQLVESGGGLVQOAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPQSHFYDGSWRRASAYDDWCQOGTQVTVSS /INSDSe a gecuence> 188g </INSDSeq> 1REG <SBequencelata>
Rol <HGegquencelbata seguanselilMonhao=nTRY > eed LINSDSeq»
Leal <INSDSeq length»126/INSDSeq lengih> load <INSDSeq molityperAA/INIDSeqg molLypex
IESE <INSDSeq division>PAT/THSDSeq divisions
REA) <INBDZeq feabture-tablel 19a <INSDFeature>
Lise <INSDFearure key>sources/INSDFeature key> nan JIiNSDFeature location>l..l26</IN3DFsature location» eG <“INSDPFeacure gqualis>
Lat <INSDQualifier»>
Tai <INSDQualifier name>mol type</INsSDQualifier nama»
LEER <INSDuualifier valuerprotein</INSDOualifisr valus> 1874 </INSDQualifisan»
RE <INSDQuaiifler id=vglasgys
Lee CINEIDOualifier namerorganism“/INSDUualifier name>
RA CINZDQualifiler value>synthetic construct </INSDQualifier value>
LET </INSDQualifier> 187e </INSDFeature guals>
TES x“/INSDFeacturex ==
LEE </INSDSeg feature-tabled>
LEE <INSDSeq sequence»
EVQLVESGGGLVQAGGSLRLSCAASRRTFRSYATAWFRQAPGKERVFVAGTTWIISSTYYADSVNGRFTISRDN
AENTVYLQMNSLKPEDTAVYYCAARVRSGSGQYTLPGHYDYWGQGTQVTVSS</LN3LE ag sequence
TEE <SINEDE E>
HRCERS </SeguenceData> loud <SeduenceData sapuiencelliumber=2Tans
E88 CINIDSeq> rae <INSDSeq lengith>ll6</INSDSeq length»
Ess <INSDSeq moltype>AA< /INSDSeg moltype:»
TERE CINEDSeq division>PAT</INSDEaq division»
LES “INSDSeq fealure-table>
Tul <INSDFeaturer
Lene <INSDFeature key>source</INSDFeature key» 193? <INSDFesture location»l..116</INSDFeature location»
LEE <INSD¥eature guals>
LES <INSDoualifier> iead <INSDQualifier namermol type</INSDQualifier name>
Laan CINMEDOualifier valuesprotein</INSDLQualifier valued
LES <SINSDOualifier>
Tae <INSDQualifier aad=ngil8Ty>
Leid <INSDQualifier name>organism</INSDQualifier names
Loi <INSDQualifier value>synthetic construct <SINSDOualifier value» 1902 </INSDQualifisan» -
RCE </INSDFearure guals> 1804 </INSDFeature>
Lens </INSDSeg faature-table>
Les <“INSDSeq sequenced
EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATLTRFGLAGYADSVKGRFTISADHA
KLTLYLHMNNLKPADTAVYYCNVKTLGGADYWGQGTQVTVSS</INiliZeq seguance> ina </INSDSea:
E08 </SequenceDatas
LEG <Sequencebata zegvencaiDNunmber=ng9">
LSG <INSDSeq> eld CINZDSeq length>116</INSDSeq lengths
Leie “INSDSeq moltype>dAA/INSDSeg moltype> iel <INSDSeq division»PAT</INSDSeg diviglon» isië <INSDSeq Ieatureriabier 131% CINSDFeaturae>
IEEE <INSDFeature key>source“/INSDFealure key>
LEAT <IMaDFeature location>l..116</INSDFeature location»
Leie <INSDFearure guals:»
Tenn <CINSDQualifiers
REN <INSDQualifier name>mol_ type /INSDQualiiier named
LGE: <INSDQualifier value»protein</INSDOQualijier value»
TEER </INSDQualifier> ijn JINSDoualifier id="gi8812 is2á <INSDGualifier nansrorganism“/INSDQuali fien name>
LEZ <INSDQualifier valuersynthetic construct </INSDQuali fier values
Lene <SINSDOualifier>
Tend </INSDFeature duels»
Lae </INSDFeature>
Los </INSD8eq featura-tabler 1020 <INSLSeq zequenca>
EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGVQREVVATLTRFGLAGYADSVKGRFTISADHA
KLTLYLHMNNLKPADTAVYYCNVKTLGGADYWGQGTQVTVSS<,/INSDS eq sequencer
LEI </INSDS eg u
LOSE </ZaquenceData>
Less <“SequsrnceData sequanceliNuubsc=NSIN>
Laid <IiNSsDseqd> 1335 <INSDSeq lengthr1l6</INIDSeq length>
Ljie <INSDSeg moltype>AAc/INSDSeq moltype»> 1s <INSDSeg division>PAT</INSDSeq division»
LS3s <INSDSeq feature-table>
LS <INSDFeature>
Leal CINEDFeature keyvsource</INSDFeature key»
Lodi “INSDFeabture Location». .116</INSDFeature location»
Leda <INSDIesture guals>
Lsa <INSDOualifier> indd <INSDQualifler named>mol type“ /INSDQuali fier names 18a <INSDQualifier valuevprotein</INSDQualifisn wvalus> 14d </INSDQuali fier!»
Lear CINSDQualifler id="gl8S>
Ladd v“INSDQualifier namerorganism</IN3DQualifier name>
Leid <INSDQualifier value>synthetic construct </INSDQualifier value
EEG </INSDQualifier> -
ERE <SINSDFaature guals> 1853 </INSDPeatune> -
Las </INSDSeg feature-tabhlad
Leng <INSDSeq sequence’
EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATLTRFGLAGYADPVKGRFTISADHA
KLTLYLHMNNLKPADTAVYYCNVKTLGGADYWGQGTQOVTVSS /INSDSeg sequence
Lunn << INEDSeg> 1358 </SeguenceDatar inbd <Seduencepata samencoalóNuber="gZ2n> 1558 <INSDSeq>
LER <INSDSeq lengih>116</INSDSeq iength»
LSG <INSDSeq moliype>AA</INSDSeq moltype»
LeSi CINZDSeq divizion»PAT</INSDS=q division»
Lead <INSDSeq feature-itable>
Lee <INSDFearture:» ijs <INSDFesture keyrsource“/INSDFealure key» 13a <INSDFeature location>l..116</INSDFeaturs location
Lisse <INSDFeature guals> 19a <INSDQualifier>
SSS <INSDQualifier namermol type“ /INSD{ualifier name>
Laan CINZDOQuallifier valuesprotein“/INSDgualifier value»
Len </INSDOualilfier>
LET <INSDgualifier ii="g19807>
LET <INSDQualifier name>organism</INSDQualifisr names
LEER <INBDQualifier valuersynthetic construct </INSDOualifier value»
HS </INSDQuali fier!» 18 </INSDFearture qualss eve </INSUFaature -
Leij </INSDSeg feature-itablex
Lie <INSDSeq sequencer
EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATLTRFGLAGYADSVKDRFTISADHA
KLTLYLHMNNLKPADTAVYYCNVKTLGGADYWGQGTQVTVSS</ I NSDS a seguence»
LRT </INSDSeg> IJ 198 </Segquaencebata>
LOST <SSequencebala seqguantailDiioghar="83%2>
Les CINSDSag»
Less “INSDSeq length»+116</INSDSeg Length>
Lend <INSDSeq moltype>AA“/INSDSeq moltype> 1nER <INSDSeq divislon»>PATC/INSDSeg divisions isu <INSDSeg feature-table> IJ
Lae <TNSDFeaburae>
LEE <IMaDFesature key>source“/INSDFeature key>
Lon CINSDFesature locations»l..116</IN3DFeature locations»
Len <INSDFeature quals> IJ
Tend CINBDOualifierns
Leh <INSDQualifier name>mol type“ /INSDQualifier name» 1532 <INSDQualifier valuerprotein</INSDQualifier value» imag <ATNaSDQuali fis»
Ines <INSDQualifier ìd="gl8ins
Lane <INSDQualifier namevorganism“/INSDQualifier name>
Lewy <INSDOQualifier valuersynthetic construct <{/INSDQuali jier valuex>
Leds </INSDOualilfier>
HRCI </INSDFPeature guals>
SEVERE </INSDFeature> u
SOR </INSDEeg Zearture-tabier
ZD LINSDSeg sequenced
EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATLTRFGLAGYADSVKGRFTISADHA
KLTLYLHMNNLKPADTAVYYCNAKTLGGADYWGQGTOVTVSS</INSD3eg sequence»
2003 </INSUE aa»
Zuid </SeqenceData>
FREER “SequenceData seguencellNumben="S84*> zon <INSDSeq>
A <INSDSeq lengihrl26</INIDSeq lengih> 20a <INSDSeq moelivpe>AA/INSDSeq moltyper 200% <INSDSeqg divisiom>PAT:/INSDSeg divisions
ZELE <INSDSeq festure-table> 2001 <INSDFeature»
ZULE <“INSDFearure key>source</INSDFeature key»
ULE <INSDFeature focation>l..126</IN3DF¥eature location
Auld <INSDFesture duels»
BOL <INSDQualifier> 2018 <INSDOQualifier name>mol type“ /INSDQguali fier name»
EAN <INSDQualifier valuerprotein</INSDQualifier value» 2000 </INSDOualifier:>
ZOLL CINSDQualifler iQ=NglSZN>
ZOE <INSDQUalifiler naemerorganism/INSDOualifier name
EL <INSDQualifier value>synthetic construct </INSDQualifier value»
EOE2 <ATNaSDQuali fis» -
ZES </INSDFeature gualss» 2024 </INSDFeature> 202% </INSDSeg feature-tabled>
ZOE “INSDSed sequencer
EVQLVESGGGLVQAGGSLRLSCAASGRTFTSYTLGWFRQAPEKEREFVGGISWSYWNGDSTWYADSVKGRFTVS
TDNAKKTAYLQMNSLKPEDTAVYCAARPSARITSRRSDYDYWGQGTQVTVSS</ INSDSeq seguence»
SET </INSDSeg>
SER </SeguenceData> 250 <SequenceData saguencalMiunbaer="R8"
SURG <iNSDSed> 20731 <INSDSeq length>126</INSDSeq length» 20% CINZDSeq moltype»AAS/INSDSey moltype> 2GRE <INSDSeq diviglon»PAT</INSDSeqg division
ZUS <INSDSeq feature-itableX
San <INSDFeature> die <INSDF Feature key>source“/INSDFeature key> 2a <INSDFeature location»>1..126</INSDFeacture location» 20s <INSDFeature guals>
ZOL <INSDguelifiler»
ZO <INSDQualifier name>mol type“ /INSDQualifier name»
ZOL <INSDQualifier valuse>protein</INaTQualifier value»
LOAD </INSDOualifier> u
San CINSDOualifier id="gi983n> did d <INSDQualifler namedorganism</INSDQualifisr name> 204% <INSDQualifier valuersynthetic construct </INSDOualijier value» 2046 </INSDOualifier:> 2047 “{INSDFeature quals>
ZuiE </INSDFeature» u
TAR </INSDSeg feature-tabler
SGD <INSDSeq sequence>
EVQLVESGGGLVQAGGSLRLSCAASGRTFTSYTLGWFRQAPEKEREFVGGISWSYWNGDSTWYADSVKGRFTVS
TDNAKKTAYLQMNSLKPEDTAVYCVARPSARITSRRSDYDYWGQGTQVTVSS< /INSDSeq sequencer
ZOL </INSDSeg>
Ze </SemtenceDala> 2053 <HGegquencelbata seguansellMNonhao="88Y >
GLE LINSDSeq»
ZUDd <INSDSeq iength>122/INSDSeq Lenghth> 20h <INSDSeq moltype>AA</INSDSeq moliype>
HORT CINBDSeq division»PAT</IN3D3eq division»
ZIS <INSDSeg feabture-tablel
RARER <INSDFesture>
AREY <INSDFeature keyrsource</INIiDiFezaturs key> 2081 CINZDFeature location>l..122«</IN3D¥Feature location»
Ged <“INSDPearure duals»
ZES <INSDQualifier»> zoas “INSDgvelifier named>mol type</INSDQualifier name>
HORE <INSDQualifier valuerprotein</INSDQualifisr value» 2058 </INSDQvalifier: 2067 <INSDQuaiifler id="glS4r> 208d CINMEDOualifier namerorganism</INSDQualifier name
20a CINZDQualifiler value>synthetic construct </INSDQualifier value>
ZT </INSDQualifier>
AUT </INSDFeature guals>
ZUER2 x“/INSDFeacturex
CARI </INSDSeg feature-tabled>
ZOT <INSDSeq sequence»
EVQLVESGGGLVQAGGSLKLSCAASGRTFSTLAMGWFRQAPGKEREFVAGIIRNSLSTYYSDSVKGRFTISGDN
AKNTVYLQMNSLNHEDTAVYYCAAGRWEAVRTNTPDYWGQGTQOVTVSS</INSDSeqg sequencer
ZGT </INSDSeg>
ZOT </SeguenceData>
AUT <Sequencalata sopeancalliumber="{7T8>
AEE CINIDSeq>
ZOT <INSDSeq lengith>122</IN3DSeq length»
ORG <INSDSeq moltype>AA< /INSDSeg moltype:»
ZOE <INSDSeq division>PAT</INSDS=q division> 200% “INSDSeq fealure-table> 20nd <INSDFeaturer
FEE <INSDFeature key>source</INSDFeature key»
SUES <INSDFesture location>l..122</INSDFeaturs location»
FORE <INSD¥eature guals>
ZOET <INSDoualifier> 2083 <INSDQualifier namermol type</INSDQualifier name>
SOR CINMEDOualifier valuesprotein</INSDLQualifier valued
ZO <SINSDOualifier>
ZO LINSDGualifler in=Ngi35r>
Zine <INSDQualifier name>organism</INSDQualifier names
FER <INSDQualifier value>synthetic construct <SINSDOualifier value» 2004 </INSDQualifisan» - 208% </INSDFearure guals> 20% </INSDFeature>
ZST </INSDSeg faature-table>
Gua <“INSDSeq sequenced
EVQLVESGGGLVQAGGSLKLSCAASGRTFGTLAMGWFRQAPGKEREFVAGIIRNSLSTYYSDSVKGRFTISGDN
AKNTVYLQMNSLNHEDTAVYYCAAGRWEAVRTNTPDYWGQGTQOVTVSS< /INSDSeqg zeguence> <STHEDS eq 2100 </SeguenceDatas
SRO <Sequencebata zaogquencailiunhay="88%0
Sid <iNSDSeg>
ZL “INSDSeqg length>122</INSDSeq lengths
SLE <INSDSeq moltype>dAA/INSDSeg moltype>
ZAGER <INSDSeq divislon»PAT</INIDSeg division
Side <INSDSeq Ieatureriabier
ERE <JINEDFeature> ian <INSDFeature key>source“/INSDFealure key> 210% <IMaDFeature location>l..122</INSDFeature location»
ZAL <INSDFeacure guals»
ZOLL <CINSDQualifiers
ZLLE <INSDQualifier name>mol_ type /INSDQualiiier named 2113 <INSDQualifier value>protein</INsDQualifier valued 11d </INSDQualifier>
EIL JINSDoualifier id="giS8nz 2118 <INSDGualifier namerorganism</INSDQualifisr name> 2147 <INSDQualifier valuersynthetic construct </INSDQuali fier values
BIL <SINSDOualifier>
SALLE </INSDFeature duels»
TLE </INSDFeature:>
Re </INSD8eq featura-tabler
SiR <INSLSeq zequenca>
EVQLVESGGGLVQAGGSLKLSCAASGRTFSTLAMGWFRQAPGKEREFVAGIIRNSLSTYYSDAVKGRFTISGDN
ARNTVYLOMNSLNHEDTAVYYCAAGRWEAVRTNTPDYWGQGTQVTVSS</INIDSey sequence 210% </INSDS eg u
Zina <j Gaemquencebata>
ZES <Sequencebata geguanteildNuahao="88"> zld <IiNSsDseqd>
ALE <INSDSeq length>122</INSDSeq length>
Sian <INSDSeg moliype>AAd/INSDSeqg moltype»>
Zia <INSDSeg division>PAT</INSDSeg division»
ALG <INSDSeq feature-table> 2131 <INSDFeature>
ZEGE CINEDFeature keyvsource</INSDFeature key»
ZANE “INSDFeabture locabtion>l..122</IN3DFeature location» isd <INSDIesture guals>
ERIE CINEDOualifier>
Sie <INSDQualifler named>mol type</INSDRualifizp names
ZL <INSDQualifier valuerprotein</INSDQuali fien value»
REE </INSDQuali fier!» 213% CINSDQualifler iad=vgqlevys
ZL v“INSDQualifier namerorganism</IN3DQualifier name>
ZOL <INSDQualifier value>synthetic construct </INSDQualifier value 214 </INSDQualifier> -
Dian <SINSDFaature guals> 2144 </INSDPeatune> 214% <GINZDSeyg feature-tabled
Zit <INSDSeq sequence’
EVQLVESGGGLVQAGGSLKLSCAASGRTFSTLAMGWFRQAPGKEREFVAGIIRNSLSTYYSDSVKGRFTISGGN
AKNTVYLQMNSLNHEDTAVYYCAAGRWEAVRTNTPDYWGQGTQVTVSS</INSDSe G sequencer zld << INIDSeg> 2149 </SeguenceDatar
Side <SequenceData samencelósubern=NS9n> 2150 <INSDSeq> 215A <INSDSeq lengih>122</INSDSeq iength»
Ze <INSDSeq moliype>AA</INSDSeq moltype»
ALLE CINZDSeq divizion»PAT</INSDS=q division»
SLE <INSDSeq feature-itable> zis <INSDFeature:>
S158 <INSDFesture keyrsource“/INSDFealure key»
Sin <INSDFeature location>l..122</INSDFeaturs location 2i5w <INSDFeature guals>
Dinu <INSDQualifier>
SLAG <INMEDQualifier namermol type</INsDoualifier name>
Zhe CINZDOQuallifier valuesprotein“/INSDgualifier value»
ZLES </INSDOualilfier>
Zin <INSDQualifier id="g138%> zie <INSDQualifier name>organism</INSDQualifisr names
ZAG <INBDQualifier valuersynthetic construct </INSDOualifier value» 218¢ </INSDQualifiern> 2187 </INSDFearture qualss
PARES </INSUFaature u
LET </INSDSeg feature-itablex
ATG <INSDSeq sequencer
EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRQAPGKEREFVAGITRNSISTYYSDSVKGRFTISRDN
ARNTVYLOMNSLKPEDTAVYYCAAGRWEAVRTNTPDYWGQGTQVTVSS</ I NSLS aq seqguanca>
ZIT </INSDSeg> u 21% </Segquaencebata>
ZS <SSequencebala seqgsenceinNvumbsc=nSis> 20a CINSDSag»
ZTE <INSDSeq length>122</INSD3eq Length>
ZTE <INSDSeq moltype>AA“/INSDSeq moltype> 210 <INSDSeq divislon»>PATC/INSDSeg divisions zis <INSDSeg feature-table> IJ 21GG CINSDFeabure>
ZIRT <IMaDFesature key>source“/INSDFeature key>
ZERIT <INSDFeature location>l..122</INSDFeature location:
Zin <INSZDFeature gqualsh
ZIE: <INSboualifier? ik <INSDQualifier name>mol type“ /INSDQualifier name»
ZiSh <INSDQualifier valuerprotein</INSDQualifier value»
S188 </INSDQuali jier
ZIS <INSDQualifier ìd="gl8Sn2>
ERR <INSDQualifier namevorganism“/INSDQualifier name>
DERG <INSDOQualifier valuersynthetic construct <{/INSDQuali jier valuex>
Zen </INSDOualifier> ial </INSDFPeature guals>
AER </INSDFeature> u
Sie </INSDEeg Zearture-tabier 218d <INSDZeq sequences
EVQLVESGGGLVQAGGSLRLSCAASGRTSSTLAWGWFRQAPGKEREFVAGIIRNSISTYYSDSVKGRFTISRDN
AKNTVYLQMNSLKPEDTAVYYCAAGRWEAVRTNTPDYWGQGTQVTVSS</INSDSe G sequence’
ZL </INSUE aa»
SLES </EegquencebData> lin <Sequencelata seguanoelidhunhe n=Y8320> zine <INSDSeq> zins <INSDSeq lengibh>122</INSDSeq lengih> 2200 <INSDSeq moliype>AA</INSDSeq moltype> 224 <INEDZeq division>PAT</INSDEsqg division» 220% <INSDSeq festure-table> 2203 <INSDFeature»
Add <“INSDFearure key>source</INSDFeature key»
ZZD <INSDFeature Location»1..122</INSDFeature location>
Zend <INSDFesture duels»
ZED JINSDOualifier>
ZED8 <INSDOQualifier name>mol type“ /INSDQguali fier name» 220% <INSDQualifier valuerprotein</INSDQualifier value»
BELG <fINSDUualifler»>
ZE CINSDQualifler io=ngadays
Zedd <INSDQUalifiler naemerorganism/INSDOualifier name zele <INSDQualifier value>synthetic construct </INSDQualifier value»
Eid </INSDQuali fier» u
ZE </INSDFeature gualss» 2216 </INSDFeature>
ZEIL </INSDSeg feature-tabled> 221s “INSDSed sequencer
EVQLVESGGGLVQAGGSLRLSCAASGRTFSLLAWGWFRQAPGKEREFVAGIIRNSISTYYSDSVKGRFTISRDN
AKNTVYLQMNSLKPEDTAVYYCAAGRWEAVRTNTPDYWGOQGTQVTVSS /INSDSeg sequence» zEis </INSDSeg> u
RENE </SeguenceData>
ZEEL <SequenceData saguencalMiunbaer="233"2 228% <iNSDSed> 2223 <INSDSeq length>122</INSDSeq length»
Zana CINZDSeq moltype»AAS/INSDSey moltype>
ZELs <INSDSeq diviglon»PAT</INSDSeqg division:
EEE <INSDSeq feature-itableX
AEE <INSDFeature>
HEE <IN3DFeature key>source“/INSDFeature key>
ZENG <INSDFeature location»>1..122</INSDFeacture location» 220 CINSDFeature gualis>
ZES <INSDguelifiler»
ZES <INSDQualifier name>mol type“ /INSDQualifier name»
Zed <INSDQualifier valuse>protein</INaTQualifier value»
Gms </INSDOualiiier> u u
SEER CINSDOualifier 1d=svgRQiT»
EES <INSDQualifler namedorganism</INSDQualifisr name>
ZAT <INSDQualifier valuersynthetic construct </INSDOualijier value»
ZES <fINSDUualifler»> 22s “{INSDFeature quals>
Zin </INSDFeature» u
Zed: </INSDSeg feature-tabler
Sud <INSDSeq sequence>
EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRQAPGKEREFVAGITRNSISTYYSDSVKGRFTISRDN
AKNTVYLQMNSLKPEDTAVYYCAAGRWEAVRTINTPDYWGQGTQOVTVSS/INSDSeq sequence» 2243 </INSDSeqg> 2244 <SBequencelata> 2248 <Sequencebata segoantailomhao="R8Y
LAA LINSDSeq» zj <INSDSeq iength>122/INSDSeq Lenghth> zoas <INSDSeq moltype>AA</INSDSeq moliype>
Zed <INSDSeq division»PAT</INSDSeg divisions
ZES <INSDSeg feabture-tablel
ZET <INSDFesture> 22% <INSDFeature keyrsource</INIiDiFezaturs key> 2253 CINZDFeature location>l..122«</IN3D¥Feature location»
Land <“INSDPearure duals»
ZeDh <INSDQualifier»>
SERENE “INSDgvelifier named>mol type</INSDQualifier name>
Pan <INSDQualifier valuerprotein</INSDQualifisr value» 2258 </INSDQvalifier:
DEE <INSDQuaiifler id="g208"2>
SEE CINMEDOualifier namerorganism</INSDQualifier name
Za CINZDQualifiler value>synthetic construct </INSDQualifier value> zee </INSDOualiiier> u
SEER </INSDFeature guals> dnd x“/INSDFeacturex ==
ZE </INSDSeg feature-tabled> 2266 <INSDSeq sequence»
EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRQAPGKEREFVAGIIRNSISTYYSDSVKGRFTISRDN
AKNMVYLQMNSLKPEDTAVYYCAAGRWEAVRTNTPDYWGQGTQOVTVSS</INSDSeqg sequencer
ZZE </INSDSeg>
ZEER </SeguenceData>
SE50 <Sequencalata sopeancallilumber="880
AEG CINIDSeq>
ZET <INSDSeq lengith>122</IN3DSeq length»
ZAT <INSDSeq moltype>AA< /INSDSeg moltype:»
ZES CINEDSeq division>PAT“/LNSUSen division» 22a “INSDSeq fealure-table>
ZES LINSDFeature:»
ZES <INSDFeature key>source</INSDFeature key»
Ea <INSDFesture location>l..122</INSDFeaturs location»
ZEER <INSD¥eature guals> 22 <INSDoualifier> 2280 <INSDQualifier namermol type“ /INSDgualifier name>
ZEN <INSDOQualifier valuesprotein</INSDLQualifier valued
BEEK <SINSDOualifier>
PR LINSDGualifler ia=ngdQlv> sok <INSDQualifier name>organism</INSDQualifier names
SEER <INSDQualifier value>synthetic construct <SINSDOualifier value»
ZEEE </INSDQualifisan» - 2287 </INSDFeature quals> 2255 </INSDFeature>
ZAR </INSDSeg faature-table>
LEE <“INSDSeq sequenced
EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRQAPGKEREFVAGIIRNSISTYYSDSVKGRFTISRDN
AKNTVYLQMNSLKPEDTAVYHCAAGRWEAVRTNTPDYWGQGTQOVTVSS< /INSDSeqg zeguence>
ZED </INSDSeag:
Zas: </SequenceDatas
AES <Sequencebata zegvencaiDNunber="S8N>
Laud <iNSDSeg> 2258 CINZDSeq length>122</INSDSeq lengths
LEE <INSDSeq moltype>dAA/INSDSeg moltype>
ZEGT <INSDSeq division»PAT</INSDSeg diviglon»
Sens <INSDSeq Ieatureriabier
FESS <JINEDFeature> 2200 <INSDFeature key>source“/INSDFealure key> 220 <IMaDFeature location>l..122</INSDFeature location»
ZOE <INSDFeacure guals»
ZEUS <CINSDQualifiers
Zaid <INSDQualifier name>mol_ type /INSDQualiiier named
ER <INSDQualifier value>protein</INsDQualifier valued
EEA </INSDQualifier>
HEAT CINSZDOuallifler 1d=7gRQ4v» 22488 <INSDGualifier namerorganism</INSDQualifisr name> 230% <INSDQualifier valuersynthetic construct </INSDQuali fier values
ZELE <SINSDOualifier>
ALE </INSDFeature duels» 2512 </INSDFeature>
A312 </INSD8eq featura-tabler
S314 <INSLSeq zequenca>
EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRQAPGKEREFVAGIIRNSISTYYSDSVKGRFTISRDN
ARNTVYLOMNSLKPEDTAVYYCAAGRWEAVRTDTPDYWGQGTQVTVSS</INIDSey sequence 2s </INSDS eg u
Z3LE <j Gaemquencebata> 2317 <Sequencebata geguanteildNuaha =F >
SAE <IiNSsDseqd>
A310 <INSDSeq length>122</INSDSeq length>
SIE <INSDSeg moliype>AAd/INSDSeqg moltype»>
ZE <INSDSeg division>PAT</INSDSeg division»
ZIEL <INSDSeq feature-table> 2323 <INSDFeature> daad CINEDFeature keyvsource</INSDFeature key»
ZAL “INSDFeabture Location». .1224/INSDFeature location»
ZIS <INSDIesture guals>
EERE SINSDOualifier>
HOES <INSDQualifler named>mol type</INSDRualifizp names 22 <INSDQualifier valuevprotein</INSDQualifisn wvalus> 2330 </INSDQualifiern>
ZS CINSDQualifler id="g2o5> 23% CINZDQuallfier namerorganism</IN3DQualifier name>
ZEEE <INSDQualifier value>synthetic construct </INSDQualifier values
EEG </INSDQualifier> -
ECA <SINSDFaature guals> 2226 </INSDFearure> IJ 2337 </INSDSeg feature-tabhlad zis <INSDSeq sequence’
EVQLVESGGGLVQAGGSLRLSCAASGRTFSTLAWGWFRQAPGKEREFVAGIIRNSISTYYSDSVKGRFTISRDN
AKNTVYLQMNSLKPEDTAVYYCAAGRWEAVRTITPDYWGQGTQOVTVSS</INSDSeg sequence»
TEES << INIDSeg>
ASE0 </SeguenceDatar
PCS <SequenceData samencelósubern=NS8n> 2282 <INSDSeg> 22453 <INSDSeq lengih>273</INSDSeq iength» 2A <INSDSeq moliype>AA</INSDSeq moltyper 234d CINZDSeq divizion»PAT</INSDS=q division»
LEA <INSDSeq feature-itable>
FIAT <INSDFeature»> 25498 <INSDFesture keyrsource“/INSDFealure key»
S248 <INSDFeature location>l..273</INSDFeaturs location 2250 <INSDFeature guals> 2351 <INSDQualiifisr>
Zan <INMEDQualifier namermol type</INsDoualifier name>
ZEDS v“INSDQualifiern valuesprotein“/INSDgualifier value» zand </INSDOualilfier>
EN <INSDQualifier id="g3Q80>
A356 <INSDQualifier name>organism</INSDQualifisr names
PEN <INBDQualifier valuersynthetic construct </INSDOualifier value» 2203 </INSDQuali fier!» 235% </INSDFearture qualss 2304 </INSDFaaturel u
AEL </INSDSeg feature-itablex
ERT <INSDSeq sequencer
EVQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSCISSSDGSTYYADSVKGRFTISSDN
AKNTVYLQMNSLRPEDTAVYYCAAVPRTMYSRWGCGVRPYYYGMDYWGKGTLVTVSSSAAGGGGSGGGGSAAA9
VQLQESGGGLVQAGGSLRLSCAASGYISDAYYMGWYRQAPGKEREFVATITHGTNTYYADSVKGRFTISRDNAK
NTVYLOMNSLKPEDTAVYYCAVLE TRSY SFRYWGQGTQVTVSSLEHHHHHH</ INSDS eq sequence 276% </INSDEeg> u
Zie <j Gaemquencebata> 2368 <Sequencebata geguanteildNuahao="880%>
FEES <IiNSsDseqd> zSó7 <INSDSeq length>278</INSDSeq length>
PCAARY <INSDSeq moliyperAA/INIDSeq moltype> 2200 <INSDSeg division>PAT</INSDSeq division» 237 <INSDSeq feature-table>
ZTL <INSDFeature>
ZEE CINEDFeature keyvsource</INSDFeature key» 2303 “INSDFeabture locabtion>l..278</IN3DFeature location» 2574 <INSDIesture guals>
ETH LINEDOualifiar>
ATE <INSDQualifler named>mol type</INSDRualifizp names
AIH <INSDQualifier valuevprotein</INSDQualifisn wvalus> 22S </INSDQuali fier!» 23 CINSDQualifler id="g207v>
ZAG v“INSDQualifier namerorganism</IN3DQualifier name>
ZEEE <INSDQualifier value>synthetic construct </INSDQualifier values
SEER </INSDQualifier> -
EAE <SINSDFaature guals> 2284 </INSDFeature> - 228 </INEDSeg feature-tabled
ZIRE <INSDSeq sequence’
EVQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSCISSSDGSTYYADSVKGRFTISSDN
AKNTVYLQMNSLRPEDTAVYYCAAVPRTMYSRWGCGVRPYYYGMDYWGKGTLVTVSSSAAGGGGSGGGGSAAAE
VQLVESGGGLVQAGGSLRLSCAASGFAFDDYAIGWFRQAPWKEREGVSCISSSDGSTYYADSVKGRFTISSDNA
KNTVYLOMNSLRPEDTAVYYCAAVPRTMY SRWGCGVRPYYYGMDYWGKGTLVTVSS
</INSDEeg sequenced 229 </INSDSeg> 2383 </Segquaencebata> 2 <Sequencebalta seqguantaiDiicghae=l00% > 23uG CINSDSag»
Laud “INSDSeq length>268</INSD3eq Length>
EEE <INSDSeq moltype>AA“/INSDSeq moltype> 2332 <INSDSeq division»PAT</INSDSeqg division:
RCAC Re! <INSLSeq feature-itabled
ERA <INSDFeabure> 2388 <IMaDFesature key>source“/INSDFeature key> zie <INSDFearure location>l..268</IN3DFeatiuire location» djs <INSDFeature quals> IJ
Zaan <INSDQOualifier»
ALG <INSDQualifier name>mol type“ /INSDQualifier name» zin: <INSDQualifier valuerprotein</INSDQualifier value»
S402 </INSDQuali jier 2400 <INSDQualifier ìd="g208N2> 2404 <INSDQualifier namevorganism“/INSDQualifier name>
SAGE <INSDOQualifier valuersynthetic construct <{/INSDQuali jier valuex>
EE </INSDOualifier>
TAT </INSDPeature guals> zine </INSDFeaturer
EERE </IN3DSeq featura-table>
ZAL <INSISeq sequence>
EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPOQSHFYDGSWRRASAYDDWGOGTOVTVSSAAGGGGSGGGGSAAAQVQOLOE
SGGGLVQAGGSLRLSCAASGYISDAYYMGWYRQAPGKEREFVATITHGTNTYYADSVKGRFTISRDNAKNTVYL
QMNSLKPEDTAVYYCAVLETRSYSFRYWGQGTQVTVSSLEHHHHHH</INSDSeq sequence»
Zaid << INIDSeg> 2412 </SeguenceDatar
EI <SequenceData samenoelósubern=NLQins 291d <INSDSeq»> 2ALL <INSDSeq lengih>269</INSDSeq iength»
Zale <INSDSeq moliype>AA</INSDSeq moltype» 2410 CINZDSeq divizion»PAT</INSDS=q division» 2418 <INSDSeq feature-itable>
FALE <INSDFeature»>
S4E0 <INSDFesture keyrsource“/INSDFealure key»
ZAL <INSDF Feature location>l..269</INIDFeaturs location» 2422 <INSDFeature guals> 242% <INSDQualiifisr>
Zama <INMEDQualifier namermol type</INsDoualifier name>
ZALES CINZDOQuallifier valuesprotein“/INSDgualifier value»
FALE </INSDOualilfier>
LEN <INSDQualifier id="g2085> 2478 <INSDQualifier name>organism</INSDQualifisr names sane <INBDQualifier valuersynthetic construct </INSDOualifier value» 2436 </INSDQuali fier!» 24731 </INSDFearture qualss
Za </INSLFeature»
LAE </INSDSeq feature-fablex asd <INSDSeq sequencer
EVQLVESGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDN
AKNTMYLQMNNLKPEDTATYYCAKPOQSHFYDGSWRRASAYDDWGOGTOVTVSSAAGGGGSGGGGSAAAEVQOLVE
SGGGLVQAGGSLRLSCAASGRIVSIYSMAWFRQAPGKVREFVAAISWNGAETDYVDYVQGRFTASRDNAKNTMY
LQMNNLKPEDTATYYCAKPQSHFYDGSWRRASAYDDWGQGTQVTVSS-/INSDSeq seguence> 24735 </INSDSeq> 2A <j Gaemquencebata>
LAT <Gequencelala seguanceiliManhas or="183% >
EERE <IiNSsDseqd> 2430 <INSDSeq length>257</INSDSeq length>
SAG <INSDSeq moliype>AA/INIDIeq molLype»> zis <INSDSeq division>PAT</INSDSeg division» 244 <INSDSeq feature-table> 24473 <INSDFeature> aad CINEDFeature keyvsource</INSDFeature key»
ZAAG “INSDFeabture Location». .257</INSDFeature location»
AES <INSDIesture guals> zij SINSDOualifier>
EE <INSDQualifler named>mol type“ /INSDQuali fier names 2449 <INSDQualifier valuevprotein</INSDQualifisn wvalus> 2450 </INSDQualifiern> 2451 CINSDQualifler id="g2ijr>
ZANE CINZDQuallfier namerorganism</IN3DQualifier name>
ZANT <INSDQualifier value>synthetic construct </INSDQualifier value» z45d </INSDOualifise>
SALE <SINSDFaature guals> 2458 </INSDFeatune> oo 2457 </INSDSeg feature-tabhlad 24% <INSDSeq sequence’
EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATLTRFGLAGYADSVKGRFTISADHA
KLTLYLHMNNLKPADTAVYYCNVKTLGGADYWGQGTQVTVS SAAGGGGSGGGGSAAAQVQLOE SGGGLVQAGGS
LRLSCAASGYISDAYYMGWYRQAPGKEREFVATITHGTNTYYADSVKGRFTISRDNAKNTVYLOMNSLKPEDTA
VYYCAVLETRSYSFRYWGQGTQVTVSSLEHHHHHH</ INSDSed sequencer z2äjne </INSDSeag: 2480 </SeguenceDatas 248% <Sequencebata zegvenaoaiDNumber=NiO3"> 248 <iNSDSeg>
ZAG x“INSDSeq length>247</INSDSeg length»
Land <INSDSeg moltype>AA/INSDZeq woltype>
EEE <INSDSeq divislon»PAT</INIDSeg division
Adan <INSDSeq Ieatureriabier
FART LINSDFeatura> 2488 <INSDFeature key>source“/INSDFealure key> 240% <IMaDFeature location>l..247</INSDFeature location»
ZA <INSDFeacure guals»
ZAL <CINSDQualifiers
ZAT: <INSDQualifier name>mol_ type /INSDQualiiier named
SATE <INSDQualifier value>protein</INsDQualifier valued 247d </INSDQualifier>
Gn JINSDoualifier id="gZllns 2474 <INSDGualifier nansrorganism“/INSDQuali fien name> 247% <INSDQualifier valuersynthetic construct </INSDQuali fier values 247 <SINSDOualifier>
LATE </INSDFeature duels»
AED </INSDEeaturer
SAE </INSD8eq featura-tabler
ERE <INSLSeq zequenca>
EVQLVESGGDSVQPGGSLRISCTASTRISSLTVMGWYRQAPGEQREVVATLTRFGLAGYADSVKGRFTISADHA
KLTLYLHMNNLKPADTAVYYCNVKTLGGADYWGQGTQVTVS SAAGGGGSGGGGSAAAEVQLVE SGGDSVQPGGS
LRISCTASTRISSLTVMGWYRQAPGEQREVVATLTRFGLAGYADSVKGRFTISADHAKLTLYLHMNNLKPADTA
VYYCNVKTLGGADYWGQGTQVTVSS</IN3DS eg sequencer
TARE <SINEDE E> zikd </SeguenceData>
AES <Sequencalata sepeancallilumber="304n»
EEE VINSDSeq» a8 <INSDZeq Length>15</INSDSeq lengths 248g <INSDSeq moltype>AA< /INSDSeg moltype:» 24 CINEDSeq division>PAT</INSDEaq division»
ZA “INSDSeq fealure-table>
FEN <INSDFeature» zite <INSDFeature key>source</INSDFeature key»
Zia <INSDFeature location»... I5</INSDFeature location»
Panag <INSD¥eature guals> 248% <INSDoualifier> 2484 <INSDQualifier namermol type</INSDQualifier name>
ZAT <INSDOQualifier valuesprotein</INSDLQualifier valued 24% <SINSDOualifier>
ZANE LINSDGualifler in=Ngijdr>
SLOG <INSDQualifier name>organism</INSDQualifier names
SI <INSDQualifier value>synthetic construct <SINSDOualifier value»
ZBO </INSDQvalifier: - 234% </INSDFearure guals> 2504 </INSDEFeature>
Zn </INSDSeg faature-table>
ZLD “INSDSeq sequsrce>AAGGGGSGGGGSAAA“ /INSDSeq sequence»
SLi </INSDSea>
ESSERE </SeguencaDatar
S508 <SequenceData samencoelósubern=NLQnnL 2510 <INSDSeq>
ZL <INSDSeq lengith>8</INSDSeg length»
ZELE <INSDSeq moliype>AA</INSDSeq moltype»
ZL CINEDSeq division»PAT</ INDE division
Zil <INSDSeq feature-itable>
SLAB <INSDFeature:>
Soie <INSDFeature keyrsource“/INSDFeeture key>
HES <INSDFeature Location>l..8</INEDFeature locations
ZLD “INSDFeature guals> 231% <INSDQualiifisr>
SRG CINEIDOualifier namermol type“ /INSDUualifier name>
Pa CINZDOQuallifier valuesprotein“/INSDgualifier value»
PAR </INSDOualifier>
Zig <INSDQualifier id="g312N>
Dig <INSDQualifier name>organism</INSDQualifisr names
FLEE <INBDQualifier valuersynthetic construct </INSDOuallfier value»
IRE </INSDQualifiern> u
ZAT </INSDFearture qualss
ZES </INSDPaalure: 2h </INSDSeg feature-itablex
EERE <INSDSeq sequsnce>GYISDAYY-/INSDSeq sequenced
LOR </INSDSeqy u
FRIES </SecuenceDatas 253% <SequenceData zoguancalDiumben="108" >
SRA <iNSDSed> 2G CINEDSeq length>8</INSDSeg length» 2h CINZDSeq moltype»AAS/INSDSey moltype>
EE <“INSDSeg division>PAT/INSDSecg division»
ERE <INSDSeq feature-itableX
SDE LINSDFeature> 2540 <INSDFeature hey>source</IN3DFeature key> 2540 <INSDFeature iocation>l..8</IN3DFeature locations
SSA <INSDFeature guals> 254% <INSDQualifiers
Zand CINEDQUalifiler name>mol type“ /INSDgualifier named
Zan <INSDQualifier valuse>protein</INaTQualifier value»
FLAS </INSDOQualijiier> nA CINSDOualifier 1d=swgRi3Ty» has <INSDuualifier namedorganism</INSDOualifisr name> 2549 <INSDQualifier valuersynthetic construct </INSDOualijier value»
ZENG </INSDOualifier:>
ZEE </INSDFeature guals>
LEE </INSDFeature»> zine </INSDSeg feature-itabler 2854 <INSDSeq sequenca>ITHGTNTY/INIDSaqg seguenced 2355 </INSDSeas IJ 255 </SeguenceDatas
A7 <SequenceDats zegvenaoaiDNumber=ni03ns> 25% <iNSDSeg> a: “INSDSeq lengih>12</INSDSeg Length>
EATERS <INSDSeq moltype>dAA/INSDSeg moltype>
ESET <INSDSeq diviglon»>PAT</INSDSeg divisions
S552 <INSDSeq Ieatureriabier
SEAR “INSDFeature> 2504 <INSDFeature key>source“/INSDFealure key> 238% <INSDFeature iocation>l..12</INSDFeature location»
EIN <INSDFeacure guals»
ZT <CINSDQualifiers
ZEEE <INSDouelifier namermol type /INSDOualifier name
Shen <INSDQualifier value>protein</INsDQualifier valued
ZDF </INSDQualifier>
GDL JINSDoualifier id="gZlarn 57E <INSDQualifier namerorganism“/INSDQuali fier name>
SRT <INSDQualifier valuersynthetic construct </INSDQuali fier value»
Zed <SINSDOualifier>
ZIS </INSDFeacure duals»
LETS <C/INSDFeatures
SET </INSD8eq featura-tabler 25s <INSLSeq seguence>AVLETRSYSFRI</INSDSeq sequences 2578 </INSDSeg:> 2580 </SegusncsDara> 255 <Sequencebalta seqguantaiDiicghar="108% >
AER CINSDSag»
Lhe <INSDSeq length»+126</INSDSeg Length>
Ehud <INSDSeq moltype>AA“/INSDSeq moltype> 23ED <INSDSeq division»PAT</INSDSeqg division: 2358 <INSDSeq feature-takbled
SNR <INSDFealure>
EERERES <IMaDFesature key>source“/INSDFeature key>
AREIET <INSDFearure location>l..126</IN3DFeatiure location»
ZES <INSDFeature quals> IJ
ZIG <INSDQOualifier» zie <INSDQualifier name>mol type“ /INSDQualifier name»
Eine <INSDQualifier valuerprotein</INSDQualifier value» 2534 <ATNaSDQuali fis» 250% <INSDQualifier ìd="g2i5Ns> 2584 <INSDQualifier namevorganism“/INSDQualifier name>
ZST <INSDOQualifier valuersynthetiec construct <{/INSDQuali jier valuex> zZLeE </INSDOualilfier> zien </INSDPearure guals>
ZEDD </INSDFeaturer u
ZED. </INSDEeg Zearture-tabier 2502 <INSDZeq sequences
QOVOLGQESGGGLVQAGGSLRLSCAASGYISDAYYMGWYRQAPGKEREFVATITHGTNTYYADSVKGRFTISRDNA
KNTVYLQMNSLKPEDTAVYYCAVLETRSYSFRYWGQGTQVTVSSLEHHHHHH</INSDSe dq sequences 2803 </INSUE aa» aad </EegquencebData>
ZED </ST25SequenceListing>
Claims (50)
Priority Applications (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
NL2036011A NL2036011B1 (en) | 2023-10-12 | 2023-10-12 | Molecules for reversing anti-coagulant activity of direct oral anticoagulants |
PCT/EP2024/078690 WO2025078605A1 (en) | 2023-10-12 | 2024-10-11 | Molecules for reversing anti-coagulant activity of direct oral anticoagulants |
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
NL2036011A NL2036011B1 (en) | 2023-10-12 | 2023-10-12 | Molecules for reversing anti-coagulant activity of direct oral anticoagulants |
Publications (1)
Publication Number | Publication Date |
---|---|
NL2036011B1 true NL2036011B1 (en) | 2025-04-30 |
Family
ID=93119815
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
NL2036011A NL2036011B1 (en) | 2023-10-12 | 2023-10-12 | Molecules for reversing anti-coagulant activity of direct oral anticoagulants |
Country Status (2)
Country | Link |
---|---|
NL (1) | NL2036011B1 (en) |
WO (1) | WO2025078605A1 (en) |
Family Cites Families (35)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
MX9204374A (en) | 1991-07-25 | 1993-03-01 | Idec Pharma Corp | RECOMBINANT ANTIBODY AND METHOD FOR ITS PRODUCTION. |
EP1589107B1 (en) | 1992-08-21 | 2009-12-23 | Vrije Universiteit Brussel | Immunoglobulins devoid of light chains |
DK0698097T3 (en) | 1993-04-29 | 2001-10-08 | Unilever Nv | Production of antibodies or (functionalized) fragments thereof derived from Camelidae heavy chain immunoglobulins |
FR2708622B1 (en) | 1993-08-02 | 1997-04-18 | Raymond Hamers | Recombinant vector containing a sequence of a structural lipoprotein gene for the expression of nucleotide sequences. |
EP0739981A1 (en) | 1995-04-25 | 1996-10-30 | Vrije Universiteit Brussel | Variable fragments of immunoglobulins - use for therapeutic or veterinary purposes |
JP2000515002A (en) | 1996-06-27 | 2000-11-14 | フラームス・インテルウニフェルジテール・インスティテュート・フォール・ビオテヒノロジー・ヴェーゼットウェー | Recognition molecules that specifically interact with the active site or cleft of the target molecule |
AU3596599A (en) | 1998-01-26 | 1999-08-09 | Unilever Plc | Method for producing antibody fragments |
AU3041100A (en) | 1999-01-05 | 2000-07-24 | Unilever Plc | Binding of antibody fragments to solid supports |
ATE276359T1 (en) | 1999-01-19 | 2004-10-15 | Unilever Nv | METHOD FOR PRODUCING ANTIBODY FRAGMENTS |
WO2000065057A1 (en) | 1999-04-22 | 2000-11-02 | Unilever Plc | Inhibition of viral infection using monovalent antigen-binding proteins |
US6479280B1 (en) | 1999-09-24 | 2002-11-12 | Vlaams Interuniversitair Institutuut Voor Biotechnologie Vzw | Recombinant phages capable of entering host cells via specific interaction with an artificial receptor |
ES2331051T3 (en) | 1999-11-29 | 2009-12-21 | Bac Ip B.V. | IMMOBILIZATION OF MOLECULES OF UNION OF ANTIGENS OF A DOMAIN. |
US7358096B1 (en) | 1999-11-29 | 2008-04-15 | Conopco, Inc. | Immobilisation of proteins |
DE19962924A1 (en) | 1999-12-24 | 2001-07-05 | Bayer Ag | Substituted oxazolidinones and their use |
DE60115394T2 (en) | 2000-02-29 | 2006-10-19 | Millennium Pharmaceuticals, Inc., Cambridge | BENZAMID AND SIMILAR INHIBITORS FROM FACTOR XA |
ES2324280T3 (en) | 2000-03-14 | 2009-08-04 | Unilever N.V. | VARIABLE DOMAINS OF THE HEAVY ANTIBODY CHAIN AGAINST HUMAN DIETETIC LIPASSES AND THEIR USES. |
WO2001090190A2 (en) | 2000-05-26 | 2001-11-29 | National Research Council Of Canada | Single-domain antigen-binding antibody fragments derived from llama antibodies |
ES2368623T3 (en) | 2000-12-13 | 2011-11-18 | Bac Ip B.V. | PROTEIN MATRICES OF VARIABLE DOMAINS OF CAMILIDAE HEAVY CHAIN IMMUNOGLOBULIN. |
US7365205B2 (en) | 2001-06-20 | 2008-04-29 | Daiichi Sankyo Company, Limited | Diamine derivatives |
EP1433793A4 (en) | 2001-09-13 | 2006-01-25 | Inst Antibodies Co Ltd | Method of constructing camel antibody library |
TWI331526B (en) | 2001-09-21 | 2010-10-11 | Bristol Myers Squibb Pharma Co | Lactam-containing compounds and derivatives thereof as factor xa inhibitors |
JP2005289809A (en) | 2001-10-24 | 2005-10-20 | Vlaams Interuniversitair Inst Voor Biotechnologie Vzw (Vib Vzw) | Mutant heavy chain antibody |
EP1456410A2 (en) | 2001-12-11 | 2004-09-15 | AlgoNomics N.V. | Method for displaying loops from immunoglobulin domains in different contexts |
AU2002360068B2 (en) | 2001-12-21 | 2009-09-03 | Vlaams Interuniversitair Instituut Voor Biotechnologie Vzw | Method for cloning of variable domain sequences |
JP2005517674A (en) | 2002-01-03 | 2005-06-16 | フラームス・インテルウニフェルシタイル・インステイチュート・フォール・ビオテヒノロヒー・ヴェーゼットウェー | Novel immunoconjugates useful for tumor treatment |
EP2301967A3 (en) | 2002-11-08 | 2011-07-13 | Ablynx N.V. | Single domain antibodies for nasal administration |
EP2267032A3 (en) | 2002-11-08 | 2011-11-09 | Ablynx N.V. | Method of administering therapeutic polypeptides, and polypeptides therefor |
EP2390270A1 (en) | 2003-01-10 | 2011-11-30 | Ablynx N.V. | Therapeutic polypeptides, homologues thereof, fragments thereof and for use in modulating platelet-mediated aggregation |
AU2008219216A1 (en) | 2007-02-21 | 2008-08-28 | Ablynx N.V. | Amino acid sequences directed against vascular endothelial growth factor and polypeptides comprising the same for the treatment of conditions and diseases characterized by excessive and/or pathological angiogenesis or neovascularization |
NZ581097A (en) | 2007-05-24 | 2012-03-30 | Ablynx Nv | Amino acid sequences directed against rank-l and polypeptides comprising the same for the treatment of bone diseases and disorders |
WO2010063113A1 (en) | 2008-12-05 | 2010-06-10 | University Of Guelph | Anti-cobra toxin antibody fragments and method of producing a vhh library |
AR079944A1 (en) | 2010-01-20 | 2012-02-29 | Boehringer Ingelheim Int | NEUTRALIZING ANTIBODY OF THE ACTIVITY OF AN ANTICOAGULANT |
CA2824885A1 (en) * | 2011-01-19 | 2012-07-26 | Bayer Intellectual Property Gmbh | Binding proteins to inhibitors of coagulation factors |
EP3363912B1 (en) | 2017-02-17 | 2020-10-28 | Synapse B.V. | Self-calibrated assay for determining proteolytic enzyme generation, dedicated means and uses thereof |
TW202045545A (en) | 2019-02-22 | 2020-12-16 | 美商安維塔生物科學股份有限公司 | Albumin binding antibodies and use thereof |
-
2023
- 2023-10-12 NL NL2036011A patent/NL2036011B1/en active
-
2024
- 2024-10-11 WO PCT/EP2024/078690 patent/WO2025078605A1/en unknown
Also Published As
Publication number | Publication date |
---|---|
WO2025078605A1 (en) | 2025-04-17 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP7146871B2 (en) | plasma kallikrein-binding protein | |
JP7587550B2 (en) | Factor XI Antibodies and Methods of Use | |
US10544216B2 (en) | P2X7 receptor antagonists and agonists | |
CN105037543B (en) | Therapeutic DLL4 binding proteins | |
EP1558646A2 (en) | Single domain antibodies directed against interferon- gamma and uses thereof | |
TW201136606A (en) | Anticoagulant antidotes | |
JP2019521978A (en) | Anticoagulation factor XI antibody | |
WO2022037527A1 (en) | Bcma-binding single variable structural domain and antigen-binding molecule | |
NL2036011B1 (en) | Molecules for reversing anti-coagulant activity of direct oral anticoagulants | |
AU2019204295B2 (en) | Plasma kallikrein binding proteins | |
CA3199648A1 (en) | Il-7 binding proteins and their use in medical therapy | |
JP7419544B2 (en) | Anti-IL-1β antibody | |
WO2019096294A1 (en) | Thrombin antibody, antigen-binding fragment and pharmaceutical use thereof | |
TW202413441A (en) | Use of tnf-alpha binding proteins and il-7 binding proteins in medical treatment | |
TW201245226A (en) | Binding proteins to inhibitors of coagulation factors |