AU2017203999B2 - Tfpi inhibitors and methods of use - Google Patents

Tfpi inhibitors and methods of use Download PDF

Info

Publication number
AU2017203999B2
AU2017203999B2 AU2017203999A AU2017203999A AU2017203999B2 AU 2017203999 B2 AU2017203999 B2 AU 2017203999B2 AU 2017203999 A AU2017203999 A AU 2017203999A AU 2017203999 A AU2017203999 A AU 2017203999A AU 2017203999 B2 AU2017203999 B2 AU 2017203999B2
Authority
AU
Australia
Prior art keywords
peptide
amino acid
group
tfpi
acid selected
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
AU2017203999A
Other versions
AU2017203999A1 (en
Inventor
Michael Dockal
Hartmut Ehrlich
Thomas Polakowski
Ulf Reimer
Ulrich Reineke
Freidrich SCHEIFLINGER
Eberhard Schneider
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Takeda Pharmaceutical Co Ltd
Original Assignee
Takeda Pharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2009327338A external-priority patent/AU2009327338B2/en
Priority claimed from AU2015234306A external-priority patent/AU2015234306B2/en
Application filed by Takeda Pharmaceutical Co Ltd filed Critical Takeda Pharmaceutical Co Ltd
Priority to AU2017203999A priority Critical patent/AU2017203999B2/en
Publication of AU2017203999A1 publication Critical patent/AU2017203999A1/en
Application granted granted Critical
Publication of AU2017203999B2 publication Critical patent/AU2017203999B2/en
Assigned to TAKEDA PHARMACEUTICAL COMPANY LIMITED reassignment TAKEDA PHARMACEUTICAL COMPANY LIMITED Request for Assignment Assignors: Baxalta GmbH, BAXALTA INCORPORATED
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Landscapes

  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The invention provides peptides that bind Tissue Factor Pathway Inhibitor (TFPI), including TFPI-inhibitory peptides, and compositions thereof. The peptides may be used to inhibit a TFPI, enhance thrombin formation in a clotting factor-deficient subject, increase blood clot formation in a subject, and/or treat a blood coagulation disorder in a subject.

Description

[0001] The invention generally relates to peptides that bind Tissue Factor Pathway Inhibitor (TFPI) and uses thereof.
CROSS REFERENCE TO RELATED APPLICATIONS AND INCORPORATION BY REFERENCE [0002] This application claims priority to U.S. Provisional Patent Application No. 61/139,272, filed December 19, 2008, the disclosure of which is incorporated by reference in its entirety.
BACKGROUND OF THE INVENTION [0003] Hemostasis relies on the complex coagulation cascade, wherein a series of events mediated by blood clotting factors lead to conversion of prothrombin to thrombin. Factor X (FX) activation is the central event of both the intrinsic and extrinsic pathways of the coagulation cascade. The extrinsic pathway has been proposed as the primary activator of the coagulation cascade (Mackmanet al., Arterioscler. Thromb. Case. Biol., 27, 1687-1693 (2007)). Circulating Tissue Factor (TF) and activated Factor VII (FVlla) interact to form the “extrinsic complex,” which mediates activation of FX, The coagulation cascade is amplified by the intrinsic pathway, during which successive activation of factors ΧΠ, XI, IX, and VIII results in formation of the “intrinsic” FIXa-FVIUa complex that also mediates FX activation. Activated FX promotes thrombin formation, which is required for the body to create fibrin and effectively curb bleeding.
[0004] Severe bleeding disorders, such as hemophilia, result from disruption of the blood coagulation cascade. Hemophilia A, the most common type of hemophilia, stems from a deficiency in factor VIII, while hemophilia B is associated with deficiencies in Factor IX (FIX), Hemophilia C is caused by a deficiency in Factor XI (FXI) (Cawthern et ah, Blood, 91(\2), 4581-4592 (1998)). There is currently no cure for hemophilia and other clotting diseases. Factor replacement therapy is the most common treatment for blood coagulation disorders. However, blood clotting factors typically are cleared from the bloodstream shortly after administration. To be effective, a patient must receive frequent intravenous infusions of
2017203999 14 Jun 2017 plasma-derived or recombinant factor concentrates, which is uncomfortable, requires clinical settings, is expensive, and is time consuming. In addition, therapeutic efficacy of factor replacement therapy can diminish drastically upon formation of inhibitory antibodies. Approximately 30% of patients with severe hemophilia A develop inhibitory antibodies that neutralize Factor Vlll (FVII1) (Peerlinck and Hermans, Haemophilia, 12, 579-590 (2006)). Few therapeutic options exist for patients with anti-Factor antibodies.
[0005] Thus, there exists a need in the art for compositions and methods for treating blood coagulation disorders. The invention provides such compositions and methods.
SUMMARY OF THE INVENTION [0006] The invention provides peptides that bind to Tissue Factor Pathway Inhibitor (TFPI), including TFPI antagonistic peptides having the ability to modulate the blood coagulation cascade. For example, the invention provides a peptide comprising the amino acid sequence X7XQi9X|0XiiXi2X13X14X1 9X20X21 (SEQ ID NO: 3109), wherein
X7 is selected from the group consisting of L, P, K, S, W, V, N, and Q;
Xs is selected from the group consisting of L, R, N, F, and I;
X<) is selected from the group consisting of Y, V, P, and C;
Xjo is selected from the group consisting of F, L, and G;
Xj 1 is selected from the group consisting of L, W, V, A, Μ, T, and S;
Xj2 is selected from the group consisting of T, F, V, R, A, D, L, E, S, and Y;
Xj3 is selected from the group consisting of I, M, G, Q, D, and R;
X14 is selected from the group consisting of G, W, Y, L, M, and H:
X15 is selected from the group consisting of N, P, F, Η, K, and Y;
Xi6 is selected from the group consisting of M, D, E, V, G, and K;
X17 is selected from the group consisting of G, 1, R, S, T, and L;
Xie is selected from the group consisting of M, K, L, and I;
X19 is selected from the group consisting of Y, G, R, and S;
X20 is selected from the group consisting of A, E, S, C, and Y; and X21 is selected from the group consisting of A, V, K, and E.
[0007] in one aspect, the peptide comprises one or more N-terminal amino acid(s) directly linked to X7, wherein the N-terminal amino acid(s) comprise the amino acid sequence selected from the group consisting of
2017203999 14 Jun 2017
Χό,
XsXft,
X4X5X6,
X3X4X5X0 (SEQ ID NO: 3110),
X2X3X4X5X6(SEQ ID NO: 3111), and X1X2X3X4X5X0 (SEQ ID NO: 3112), wherein
Xi is selected from the group consisting of T and G; X2 is selected from the group consisting of F and V; X3 is selected from the group consisting of V, W, Y, and F; X4 is selected from the group consisting of D, Q, and S; X5 is selected from the group consisting of E, T, N, and S; and X(l is selected from the group consisting of R, Η, K, and A.
[0008] Alternatively or in addition, the peptide comprises one or more C-terminal amino acids directly linked to X2J, wherein the C-terminal amino acid(s) comprise the amino acid sequence selected from the group consisting of
X22,
Xxz 22-A-23,
X
22^23^24?
X22X23X24X25 (SEQ ID NO: 3113),
X22X23X24X25X26 (SEQ ID NO: 3114), and X22X23X24X25X26X27 (SEQ ID NO: 3115), wherein
X22 is selected from the group consisting of Q, 1, E, W, R, L, and N: X23 is selected from the group consisting of L, V, M, and R; X24 is selected from the group consisting of K, L, A, and Y; X25 is F; X2fi is G; and X27 is T.
[0009] In one aspect, the invention provides a peptide comprising the amino acid sequence set foith in SEQ ID NOs: 1 -7, such as a peptide comprising the amino acid sequence set forth in any one of JBT0132, JBT0303, JBT0193. JBT0178, JBT0120, and JBT0224, which inhibits TFPI activity within the blood coagulation cascade. The invention also provides a peptide that binds TFPI comprising an amino acid sequence of at least 60% identity to the sequence Phe-Gln-Ser-Lys-Gly-Asn-Val-Phe-Val-Asp-Gly-Tyr-Phe-Glu-Arg-Leu-Arg-AlaLys-Leu (FQSKGNVFVDGYFERLRAKL) (SEQ ID NO: 32).
[0010] In addition, the invention provides a peptide that binds TFPI, wherein the peptide comprises the structure of formula (I): X1001-X1002-X1003-X100AX1005-X1006-XI007ΧΙ008-Χ1009-Χ1010-Χ10Π-ΧΙ0Ι2-ΧΙ013-Χ1014-Χ1015-Χ1Ό16-Χ1017-Χ1018-Χ1019X1020 (SEQ ID NO: 3116). In formula (I),
2017203999 14 Jun 2017
X1001 is an amino acid selected from the group consisting of Bhf, C, D, F, G, Η, I, K, L, Μ, N, Nmf, Q, R, T, V, W and Y;
X1002 is an amino acid selected from the group consisting of G and Q;
X1003 is an amino acid selected from the group consisting of A, Aib, Bhs, C, D, E, F, G, Η, I,
K, L, Μ, N, P, Q, R, S, T, V, W and Y;
X1004 is an amino acid selected from the group consisting of A, Aib, Bhk, C, D, E, F, G, Η, I, K, k, L, Μ, N, Nmk, P, Q, R, S, T, V, W and Y;
X1005 is an amino acid selected from the group consisting of a, A, Aib, Bal, C, D, d, E, F, G, Η, K, k, L, Μ, N, Nmg, p, Q, R, S, T, V, W and Y;
X1006 is an amino acid selected from the group consisting of A, Aib, Btq, C, D, E, F, G, Η, I,
K, L, Μ, N, Q, R, S, T, V, W and Y;
X1007 is an amino acid selected from the group consisting of A, F, I, K, L, Nmv, P, G, S, V, W and Y;
X1008 is an amino acid selected from the group consisting of F, Η, K, W and Y;
X1009 is an amino acid selected from the group consisting of A, Aib, f, I, K, S, T and V;
X1010 is an amino acid selected from the group consisting of A, Aib, C, D, E, F, G, Η, I, K, L,
Μ, N, Nmf, P, G, R, S, T, V, W and Y;
X1011 is an amino acid selected from the group consisting of Aib, C, K, G and Nmg;
X1012is Y;
X1013 is an amino acid selected from the group consisting of A, Aib, C, E, F, G, Η, K, L, M, Q,
R, W and Y;
X1014 is an amino acid selected from the group consisting of A, Aib, Bhe, C, D, E, F, G, Η, I,
K, L, Μ, N, P, Q, R, S, T, V, W and Y;
X1015 is an amino acid selected from the group consisting of (omega-methyl)-R, D, E, K and
R;
X1016 is L;
X1017 is an amino acid selected from the group consisting of (omega-methyl)-R, A, Aib, Bhr,
C, Cha, Cit, D, Dab, Dap, E, Eag, Eew, F, G, H, Har, Hci, Hie, I, K, L, Μ, N, Nle, Nva, Opa, Orn, Q, R,
S, T, V, W and Y;
2017203999 14 Jun 2017
X1018 is an amino acid selected from the group consisting of A, Bal, C, D, E, F, G, Η, I, K, L, Μ, N, Q, R, S, T, V, W and Y;
XJ019 is an amino acid selected from the group consisting of Bhk. K, R and V; and
XI020 is either present or absent, whereby, in case X1020 is present, it is an amino acid selected from the group consisting of Aib, Bhl, C, F, G, H, 1, K, L, Nml, Q. R, S, T, V, W and Y.
[0011] In one aspect, the peptide that binds TFPI comprises the structure of formula (III): XI001-Q-XI003-X1004-X1005-X1006-I/V-X1008-V-X1010-G-Y-C/F-X1014-R-L-X1017X1018-K-K/L (III) (SEQ ID NO: 3117). In formula (III), X1001, X1003, XI004, X1005, X1006, X1008, X1010, X1014, XI017 and XI018 are each independently selected from any amino acid.
[0012] The invention further provides a TFPl-binding peptide comprising the structure of formula (V): X2001-X2002-X2003-X2004-X2005-X2006-[X2007-X2008-X2009-X2010X2O11-X2OI2-X2O13-X2O14-X2OI5-X2016-X2O17-X2O18]-X2O19-X2O2O-X2O21-X2O22X2023 (V) (SEQ ID NO: 3118). In formula (V), X2001, X2002, and X2023 independently are either present or absent. When present. X2001 is an amino acid selected from the group consisting of A, D, E, F, G, H, I, K, L, P, R, S, T, V and W; andX2002 is an amino acid selected from the group consisting of A, D, E, F, G, Η, I, K, F, Μ, P, R, S, T, V and W. Additionally,
X2003 is an amino acid selected from the group consisting of A, F, I, K, L, R, S, T, V, W and Y;
X2004 is an amino acid selected from the group consisting of A, D, E, F, G, I, K, L,
R, S, T, V and W;
X2005 is W;
X2006 is an amino acid selected from the group consisting of F, Η, I, K. L, R V and W;
X2007 is an amino acid selected from the group consisting of C, Hey, Dap and K, preferably selected from the group consisting of C and Hey;
X2OO8 is an amino acid selected from the group consisting of A, G, R, S and T;
X2009 is an amino acid selected from the group consisting of a. A, 1, K, L, M, m, Nle, p, R, and V;
2017203999 14 Jun 2017
X2010 is an amino acid selected from the group consisting of A, G, I, K, L, P, R, S, T and V;
X2011 is an amino acid selected from the group consisting of D, E, G, S and T;
X2012 is an amino acid selected from the group consisting of A, a, D, d, E, e, F, f, G, I, K, k, L, 1, M, m, Nle, nle, P, p. R, r, S, s, T, t, V, v. W and w;
X2013 is an amino acid selected from the group consisting of A, D, d, E, e, F, G, I, K,
L, R, S, s, Τ, V and W:
X2014 is an amino acid selected from the group consisting of A, D, E, F, G, I, K, L,
M, R, S, Τ, V and W;
X2015 is an amino acid selected from the group consisting of A, D, E, F, G, I. K, L, M, Nle, R, S, Τ, V and W;
X2016 is an amino acid selected from the group consisting of A, D, E, F, I, K, L, M, Nle, R, S, Τ, V, W and Y;
X2017 is an amino acid selected from the group consisting of A, D, E, F, G, 1. K, L,
R, S, Τ, V, W and Y;
X2018 is an amino acid selected from the group consisting of C and D (preferably X2O18isC);
X2019 is an amino acid selected from the group consisting of A, F, I, L, S, Τ, V and W;
X2020 is an amino acid selected from the group consisting of F and W;
X2021 is an amino acid selected from the group consisting of 1, L and V; and
X2022 is an amino acid selected from the group consisting of A, D, E, F, G, 1, K, L,
P, R, S, Τ, V, and W.
When X2023 is present in the peptide, X2023 is an amino acid selected from the group consisting of A, D, E, F, G, I, K, L, R, S, Τ, V, W and Y. The peptide comprises a cyclic structure generated by a linkage between X2007 and X2018, indicated in Formula (V) by brackets.
[0013] The invention also provides a peptide that binds TFP1, wherein the peptide comprises the structure of formula (VI): X200J-X2002-F/Y-K-W-F/H-[C-X2008-M/VX2010-D-X2012-X2013-G-1/T-X20I6-S/T-CJ-A/V-W-V-X2022-X2023 (VI) (SEQ ID NO:
3119). In formula (VI), X2001, X2002 and X2023 are each independently present or absent.
2017203999 14 Jun 2017
X2OO8, X2010, X2012, X2013, X2016 and X2022, as well as X2001, X2002, and X2O23 when present, are each independently selected from any amino acid. The peptide comprises a cyclic structure generated by a linkage between X2007 and X2018, indicated in Formula (VI) by brackets, [0014] in one aspect, the invention provides a peptide that binds TFP1, wherein the peptide comprises the structure of formula (VIII): X300]-X3002-X3003-X3004-X3005-X3006X3007-X3008-X3009-X3010-X30J1-X3012-X3013-X3014-X3015-X3016-X3017-X3018X3019-X3020-X302I (VIII) (SEQ ID NO: 3120). In formula (VIII), X3001 and X3002 are. each independently present or absent. When present, X3001 is an amino acid selected from the group consisting of A, C, D, F, G, I, K, L. Μ, N, P, Q, R, S, T, W, E, H and Y; and X3OO2 is an amino acid selected from the group consisting of A, C, D, F, Η, K, Μ, N, P, R, S, T. W, Y, G, 1 and L. With respect to the remainder of formula (VIII),
X3OO3 is an amino acid selected from the group consisting of A, C. D, E, F, G. Η, I,
R, L, Μ, N, P, Q, R, S, T, W and Y;
X3004 is an amino acid selected from the group consisting of A, C, D, E, F, G, Η, I,
K. L, Μ, N, Q, R, S, T, V. W, Y and P;
X3005 is an amino acid selected from the group consisting of C, D, F, G, Η, I, K, L,
Μ, N, P, R, S, T, V, W and Y;
X3006 is an amino acid selected from the group consisting of A, W, C, K, P, R and H;
X3007 is an amino acid selected from the group consisting of Q, A, C, F, G, H, 1, K.
L, N, R, S, T, W and Y;
X3OO8 is an amino acid selected from the group consisting of A, C, F, G, Η, K, L, M, N, P, Q, R, S, T, V, W, Y and 1;
X3009 is an amino acid selected from the group consisting of A, C, F, G, Η, J, L, M,
R, S, T, V, W, Y and K;
X3010 is an amino acid selected from the group consisting of A, C, F, G. Η, I, K. L,
Μ, N, P, Q, R, S, T, V, W and Y;
X3011 is an amino acid selected from the group consisting of A, G, I, K, L, Μ, N, Q,
R, S, T, V, W, Y, C, F and H;
X3O32 is an amino acid selected from, the group consisting of A, C, Η, I, K, L and R;
ο
CM
2017203999 26 Apr
X3013 is an amino acid selected from the group consisting of A, C, F, G, Η, K, L, M, R, S, V, W, Y and I;
X3014 is an amino acid selected from the group consisting of A, C, F, G, Η, I, L, M, N, Q, R, S, T, V, W, Y and K;
X3015 is an amino acid selected from the group consisting of A, K and R;
X3016 is an amino acid selected from the group consisting of A, F, K and R;
X3O17 is an amino acid selected from the group consisting of A, C, F, G. 1, K, L, N,
Q, R, S,T, V, W, Y, H, AandM;
X3018 is an amino acid selected from the group consisting of A, C, F, I, K. L, M, Q,
R, V. W and Y;
X3019 is an amino acid selected from the group consisting of A, C, D, E, F, G, Η, K. L, N, P, Q, R, V, W, Y and I;
X3020 is an amino acid selected from the group consisting of A, C, F, G, Η, K, L, M, N, Q, R, V, W, Y, 1 and P; and
X3021. is an amino acid selected from the group consisting of A, C, H, I, K, L, Μ. N. P, Q, R, T, V, W, Y, F and G.
[0015] Additionally, the invention provides a TFPl-binding peptide comprising the structure of formula (IX): X3001-X3002-X3003-X3004-X3005-X3006-X3007-X3008X3009-X3010-X3011-H-X3013-X3014-K/R-R-X3017-X3018-X3019-X3020-X3023 (IX) (SEQ ID NO: 3121), wherein X3001, X3002, X3OO3, X3004, X3OO5, X3OO6, X3007, X3OO8, X3OO9, X3010, X301.3, X30I3, X3014, X30I7, X3018, X30I9, X3O2O and X3021 are each independently selected from any amino acid. In addition, the invention includes a peptide that binds TFPI, wherein the peptide comprises an amino acid sequence having at least 60% identity to the sequence of formula (X): Ac-G YASFPWFVQLHVHK.RSWEMA-NH2 (SEQ ID NO: 2023). In the context of the disclosure, any peptide encompassed by any of formulas (1) to (X) and any TFPl-binding peptide described herein is also referred to as “the peptide of the invention” and as “a peptide as described herein.” [0016] In some embodiments, the peptide of the invention binds TFPI-1 (e.g,, TFPI-la) and, optionally, improves TFPl-regulated thrombin generation in the absence of FVIJI, FIX, and/or FX1. A composition (e.g., a pharmaceutical composition) comprising the peptide also is provided.
2017203999 14 Jun 2017 [0017] In addition, the invention provides methods of using the peptide of the invention. For example, the invention provides a method of inhibiting a TFPI comprising contacting the TFPI with a peptide as described herein. The invention also provides a method of enhancing thrombin formation in a clotting factor-deficient subject, a method for increasing blood clot formation in a subject, and a method of heating a blood coagulation disorder in a subject.
The methods are, in their entirety, also referred to herein as, e.g., “the method of the invention.” The methods comprise administering to the subject a peptide as provided herein in an amount effective to enhance thrombin formation, an amount effective to enhance blood clot formation, or an amount effective to treat the blood coagulation disorder in the subject. Unless explicitly indicated to the contrary, the description provided herein with respect to one peptide of the invention or method of the invention applies to each and every peptide of the invention and method of the invention, respectively. Further aspects of the invention include use of the peptide of the invention for the manufacture of a medicament, a method for targeting a cell displaying TFPI, a method for treating or diagnosing a subject suffering from a disease or at risk of suffering from a disease, and a method of purifying TFPI. The preceding methods are, in their entirety, also referred to herein as, e.g., “the method of the invention.”
DESCRIPTION OF THE FIGURES [0018] Figure 1 is an illustration of the blood coagulation cascade, [0019] Figure 2 is an illustration of the secondary structure of Tissue Factor Pathway
Inhibitor-1.
[0020] Figure 3 is an illustration of the formation of a quaternary complex comprising Tissue Factor, Factor Xa (FXa), Factor Vila (FVIIa), and TFPI.
[0021] Figure 4 is a listing of amino acid sequences of various TFPI-inhibitory peptides denoting amino acid substitutions (bolded and underlined) in reference to peptide JBTO293. [0022] Figure 5 is an illustration of mRNA display selection of TFPI-inhibitor peptides.
[0023] Figure 6A is an illustration of the EC50 binding ELISA and Figure 6B is an illustration of the IC50 ELISA described in Example I.
[0024] Figure 7 is a binding ELISA curve comparing % OD (y-axis) and concentration [11M] (x-axis) for biotinylated peptide JBT0132.
2017203999 14 Jun 2017 [0025] Figures 8A-8D are competition ELISA curves comparing % OD (y-axis) and concentration [nM] (x-axis) for exemplary peptides of the invention.
[0026] Figures 9A and 9B are sensorgrams plotting RU (y-axis) against time in seconds (x axis) for peptides JBT0120 and JBT0132.
[0027] Figures 1ΌΑ and JOB are sensorgrams plotting RU (y-axis) against time in seconds (x-axis) for peptide JBT0120 interaction with Tissue Factor Pathway Inhibitor-1 and Tissue Factor Pathway Inhibitor-2.
[0028] Figures 11A and 1.1B are graphs comparing amount of thrombin generated (nM) (yaxis) and time in minutes (x-axis) for peptide JBT0120 and peptide JBTO132 in a plasmabased assay.
[0029] Figures 12-18 are tables listing the amino acid sequences of various TFPIinhibitory peptides; ECso and percent inhibition of TFPI observed in the FXa inhibition assay; ECso and percent inhibition of TFPI observed in the extrinsic tenase inhibition assay; and FEIBA, Factor VIII (FVIII) Immunate, or Factor IX (FIX) equivalent activities (mU/mL) in plasma-based assays. denotes negative controls.
[0030] Figures 19-21 are tables listing the results from BIAcore analysis of several TFPIbinding peptides. denotes negative controls.
[0031] Figures 22-30 are tables listing the amino acid sequences of various TFPI-binding peptides; EC30 and percent inhibition of TFPI observed in the FXa inhibition assay; ECso and percent inhibition of TFPI observed in the extrinsic tenase inhibition assay; and FEIBA,
FVIII Immunate. or FIX equivalent activities (mU/mL) in plasma-based assays. denotes negative controls.
[0032] Figure 31 is a graph comparing a pharmacokinetic characteristic (concentration of peptide (y-axis) versus time after administration (x-axis)) of a PEGylated TFPI-binding peptide to the pharmacokinetic characteristic of same peptide lacking PEG. The peptides were administered intravenously to C57BI6 mice at a dose of 10 mg/kg. Three biological samples were analyzed for the presence of peptide at each time point.
[0033] Figures 32-39 are tables listing the amino acid sequences and ICso or EC50 values of various peptides of the invention. denotes negative controls.
2017203999 14 Jun 2017
DETAILED DESCRIPTION OF THE INVENTION [0034] The invention provides peptides that block the inhibitory activity of Tissue Factor Pathway Inhibitor-1 (herein referred to as TFPI) within the blood coagulation cascade. Upon vascular injury, Tissue Factor (TF) complexes with Factor Vila to form the “extrinsic complex” or “extrinsic tenase complex,” which activates Factors IX and X (Figure 1). TFPI. is the main natural regulator of TF/FVIIa extrinsic complex activity and by extension, plays a role in controlling thrombin generation (Panteleev et al., Eur. J. Biochem., 249, 2016-2031 (2002)). TFPI is a 43 kDa serine protease inhibitor comprising three Kunitz-type inhibitory domains (Figure 2). Kanitz domain 1 of TFPI binds FVIJa and Kunitz domain 2 binds FXa, enabling the inhibitor to form a quaternary FXa-TFPI-FVIIa-TF complex that blocks activity of the TF/FVIIa extrinsic complex (Figure 3). TFPI binding of FXa also downregulates the common pathway of the coagulation cascade, during which FXa converts prothrombin to thrombin (Audu et al., Anesth. Analg., 103(A), 841-845 (2006)). The invention provides, e.g., TFPl-inhibitory peptides that block TFPI’s inhibitory action on the blood coagulation cascade, thereby enhancing thrombin fomaation.
[0035] The amino acid sequences of several TFPI-binding peptides are provided herein. Conventional amino acids are identified according to their standard, one-letter or three-letter codes, as set forth in Table 1.
TABLE 1
3-3etter 1-letter Amino acids 3-letter 1 -letter Amino acids
codes code codes code
Ala A Alanine Met M Methionine
Cys C Cysteine Asn N Asparagine
Asp D Aspartic acid Pro P Proline
Glu E Glutamic acid Gln Q Glutamine
Phe F Phenylalanine Arg R Arginine
Gly G Glycine Ser S Serine
2017203999 14 Jun2017
His H Histidine Thr T Threonine
lie I Isoleucine Val V Valine
Lys K Lysine Tip w Tryptophan
Leu L Leucine Tyr Y Tyrosine
wwBWBewjwKwwwRTOwwwjTO^ [0036] Non-conventional amino acids and additional peptide building blocks are identified according to a three-let ter code (with the exception of Ttds. which is a common four-letter abbreviation) found in Table 2,
TABLE 2
Name Abbreviation Structure
2-aminobutyric acid Abu H2N^ C J
Ξ ^OH
A
2-Amino-isobutyric acid Aib h2n o X
A A.
Bal - nh2
β-Alanine λ HO
3-Homoglutamatic acid Bhe HO =0
h2n*—-< [Si OH
C
2017203999 14 Jun 2017
Name Abbreviation Structure
β -Homopheny] alanine Bhf 0
HOX h;X JL
β-Homolysine Bhk H2N J-OH
nh2 o
β-Homolencine Bhl / 7H
C- p° nh2
β-Homoasparagine Bhn % h2n. J .0
HO r
β-Homoglutamine Bhq nh2 Y --x. r
nh2 OH
β-Homoarginine Bhr nh2
H Ύ*νη NHj
β-Homoserine Bhs HO
1 nh2 c )H
β-Homotyrosine Bhy o
HO^ (
kz k ‘nh2
2017203999 14 Jun2017
Name Abbreviation Structure
β-Homoaspartic acid Bhd °%^°H ΎΥ O NHj
β-HomovaIine Bhv, Btl NH, 0 γ/χ.
β-Homoasparagin Bhn, Btq 0 nh2 0
(S)-Cyclohexylalanine Cha o ‘'f OH Ό
(S)-Citrullin Cit HO < A-/ >-NH /A v_ O NHg \==O HjN
(S)-2,4-Diaminobutyric acid Dab /...............' OH H2N
(S)-Diaminopropionic acid Dap h2n ,o V/ H2N-OH
(S )-2-Proparg y 1 gl ycine Eag OH
]4
2017203999 14 Jun 2017
Name Abbreviation Structure
(S)-N(omega)-nitro-arginine Eew o II 0 HN 0 a H
L-homophenylalanine Hfe h2n «.-ΪΑΆ Ό
(S)-Homo-arginine Har NHa NH2 HN N OH
< S )-Homo-citru 11 me Hci Ά ., . / \ '- // an '—\ r\ '-' OH
(S)-Homo-cysteine Hey O nh2
(S)-2~Amino-5-methyl-hexanoic acid Hie OH nh2
(S)-Homo-lysine Hly H2NZAzZX/A/ ΗΟ^^^Ο
2017203999 14 Jun 2017
Name Abbreviation Structure
(S)-Norleucine Nie „0-^-7
(S)-/V-Methyla]anine Nma ja OH
(S)-/V-Methy3-Aspartic acid Nmd o °Y':H H
(S)-N-Methyl-glutamic acid Nme .° ί/ OH H
(S)-N-Methyl-phenyla]anine Nmf
A-Methyl-giycine Nmg 1 ° hn< Jt V\H
(S)-A-Methyl-Iysine Nmk O °H H
(S)-A-Methyl-leucine Nml ο<γΑΟΗ 1 ks? H
2017203999 14 Jun 2017
Name Abbreviation Structure
(S)- N-Methyl - arginine Nmr H
(S )-N-Methyl-seri ne Nms HO ^==^0 NH /
(S)-N-Methyl-valine Nmv
(S)-N-Methyl-tyrosine Nmy HO-ft 2-\ & \=J HN OH \
(S)-2-Amino-pentanoic acid Nva
(S)-2~PyridyI~aIanine Opa o haj
(S)-Omi thine Om OH NHe
L-phenylglycin Phg OH
2017203999 14 Jun 2017
Name Abbreviation Structure
4-Phenyl-butyric acid PhPrCO 0 OH
Polyethylene glycol PEG
Selenomethionine Sent
c 3
ΌΗ
nh2
1,2,3,4-L- Tic
tetrahydroisoquinolinecarboxybc o
acid
ΟΗ
\ XNH
(13-Amino-4,7,10-trioxa- Ttds
tridecayl)~succinamic acid
'’χ-*'
L
ο
Carboxyfluorescein FAM
(Ε) η -AJH
Xv+X Τ r Τ
(27
-Γ\
o
ΗΟ
[0037] The amino acid sequences of the peptides provided herein are depicted in typical peptide sequence format, as would be understood by the ordinary skilled artisan. For example, the three-letter code or one-letter code of a conventional amino acid, or the threeletter code or abbreviation for a non-conventional amino acid, indicates the presence of the amino acid in a specified position within the peptide sequence. The code for each non18
2017203999 14 Jun 2017 conventional amino acid is connected to the code for the next and/or previous amino acid in the sequence by a hyphen. Adjacent amino acids are connected by a chemical bond (typically an amide bond). The formation of the chemical bond removes a hydroxyl group from the 1carboxyl group of the amino acid when it is located to the left of the adjacent amino acid (e.g,, Hle-adjacent amino acid), and removes a hydrogen from the amino group of the amino acid when it is located on the right of the adjacent amino acid (e.g., adjacent amino acid-Hle). it is understood that both modifications can apply to the same amino acid and apply to adjacent conventional amino acids present in amino acid sequences without hyphens explicitly illustrated. Where an amino acid contains more than one amino and/or carboxy group in the amino acid side chain, the 2- or 3-amino group and/or the 1-carboxy group generally are used for the formation of peptide bonds. For non-conventional amino acids, a 3-letter code was used where the first letter indicates the stereochemistry of the C-a-atom.
For example, a capital first letter indicates that the L-form of the amino acid is present in the peptide sequence, while a lower case first letter indicating that the D-form of the correspondent amino acid is present in the peptide sequence. When one-letter code is used, a lower case letter represents a D-amino acid, while an upper case letter represents an L-amino acid. Unless indicated to the contrary, the amino acid sequences are presented herein in N- to C-terminus direction.
[0038] The C-termini of several TFPI-binding peptide sequences described herein are explicitly illustrated by inclusion of an OH, NH?, or an abbreviation for a specific terminating amine linked to the C-terminal amino acid code via a hyphen. The N-termini of several peptides described herein are explicitly illustrated by inclusion of a hydrogen (for a free Nterminus), or an abbreviation for a specific terminating carboxylic acid or other chemical group linked to the N-terminal amino acid code via a hyphen.
[0039] The invention provides a peptide comprising the amino acid sequence
X7XRX9X10X1 ]XιτΧ i’X11X’ίΧιι',Χ 1 tX;sX(SEQ ID NO: 3109), wherein (using single letter codes for amino acids)
X7 is selected from the group consisting of L, P, K, S, W, V, N, and Q;
X8 is selected from the group consisting of L, R, N, F, and 1;
X9 is selected from the group consisting of Y, V, P, and C;
X10 is selected from the group consisting of F, L, and G;
Xi i is selected from the group consisting of L, W, V. A. Μ, T, and S;
XS2 is selected from the group consisting of T, F, V, R, A, D, L, E, S, and Y;
X,3 is selected from the group consisting of I, M, G, Q, D, and R;
2017203999 14 Jun 2017
Xu is selected from the group consisting of G, W, Y, L, M, and H;
Xi5 is selected from the group consisting of N, P, F, Η, K, and Y;
X)6 is selected from the group consisting of M, D, Ε, V, G, and K;
Xi7 is selected from the group consisting of G, 1, R, S, T, and L;
Xis is selected from the group consisting of Μ, K, L, and 1;
Xi9 is selected from the group consisting of Y, G, R, and S;
X20 is selected from the group consisting of A, E, S, C, and Y; and X21 is selected from the group consisting of A, V, K, and E.
[0040] In addition to the core structure set forth above, X7-X2i, other structures that are specifically contemplated are those in which one or more additional amino acids are attached to the core structure (e.g., linked to the N-terminus or the C-terminus of the amino acid sequence X7-X2i). Thus, the invention includes peptides comprising the core structure and further comprising one or more N-terminal amino acid(s) comprising an amino acid sequence selected from the group consisting of:
X(1,
ΧΖ1Χ5ΧΛ,
XjXjXsXbiSEQ ID NO: 3110),
X2X3X4X5X6 (SEQ ID NO: 3111), and X,X2X3X4X5X6 (SEQ ID NO: 3112);
wherein X(, is directly linked to X? of the core structure amino acid sequence, and
Xί is selected from the group consisting of T and G;
X2 is selected from the group consisting of F and V;
X3 is selected from the group consisting of V, W, Y, and F;
X4 is selected from the group consisting of D, Q, and S;
X5 is selected from the group consisting of Ε, T, N, and S; and Χή is selected from the group consisting of R. Η, K, and A.
The peptide of the invention in one aspect comprises or consists of the amino acid sequence QSKKNVFVFGYFERLRAK (SEQ ID NO: I).
[0041] In another embodiment, the peptide of the invention comprising the core structure comprises one or more C-terminal amino acid(s) comprising an amino acid sequence selected from the group consisting of:
X22,
X22X23^
2017203999 14 Jun 2017
X
22Λ23Λ24,
X22X23X24X25 (SEQ ID NO: 3113),
X22X23X24X25X26 (SEQ ID NO: 3114), and X22X2.1X24X25X26X27 (SEQ ID NO: 3115), wherein X22 is directly linked to X?i of the core structure amino acid sequence, and
X22 is selected from the group consisting of Q, I, E, W, R, L, and N:
X23 is selected from the group consisting of L, V, M, and R;
X24 is selected from the group consisting of K, L, A, and Y;
X25 is F;
X26 is G; and X27 is T.
[0042] In one aspect, the peptide of the invention comprises or consists of the amino acid sequence VIVFTFRHNKLIGYERRY (SEQ ID NO: 4), It is also contemplated that the peptide of the invention comprises additional amino acids at both the N-terminus and the Cterminus of the core structure. In this aspect, the peptide comprises or consists of the amino acid sequence TFVDERLLYFLTJGNMGMYAAQLKF (SEQ ID NO: 3), GVWQTHPRYFWTMWPDIKGEV1VLFGT (SEQ ID NO: 5),
KWFCGMRDMKGTMSCVWVKF (SEQ ID NO: 6), or ASFPLAVQLHVSKRSKEMA (SEQ ID NO: 7).
[0043] The invention further includes peptides comprising the amino acid sequence XQGX.s-F-Xy-NVF-XnXn-GY-XisXiirRLRAK-Xzz (SEQ ID NO: 2), wherein X3 is Y or F; X4 is Q or S; X3 is N or S; X7 is K, N, or Q; Xj j is V, A, S, or T; XJ2 is F, A, D, L, Q, S, or Y X15 is F, K, or Y; X]<, is E or D; and Xi? is L or N.
[0044] In addition, the invention provides a peptide that binds TFPI, wherein the peptide comprises the structure of formula (I): X1001-X1002-X1003-X1004-X1005-X1006-X1007X1008-X1009-X1010-X1011-X1012-X1013-X1014-X1015-XI016-X1017-X10J8-X1019X1020 (SEQ ID NO: 3116). In formula (J),
XI001 is an amino acid selected from the group consisting of Bhf, C, D, F, G, Η, I, K, L, Μ, N, Nmf, Q, R, T, V, W and Y;
X1002 is an amino acid selected from the group consisting of G, K and Q:
XI003 is an amino acid selected from the group consisting of A, Aib, Bhs, C, D, E, F, G, Η, I, K, L, Μ, N, P, Q, R, S, T, V,W and Y;
2017203999 14 Jun 2017
X1004 is an amino acid selected from the group consisting of, A, Aib, Bhk. C, D, E.
F, G, Η, 1, K, k, L, Μ, N, Nmk, P, Q, R. S, T, V, W and Y;
XI005 is an amino acid selected from the group consisting of a, A, Aib, Bal, C, D, d, E, F, G, Η, K, k, L, Μ. N, Nmg, p, Q, R, S. T, V, W and Y;
X1006 is an amino acid selected from the group consisting of A, Aib, Btq, C, D, E, F,
G, Η, I, K, L, Μ. N, Q, R, S T, V, W and Y;
X1007 ss an amino acid selected from the group consisting of A, F, G, I, K, L, Nmv, P, Q, S, V, W and Y;
X1008 is an amino acid selected from the group consisting of F, Η, K, W and Y;
XI009 is an amino acid selected from the group consisting of A, Aib, f, I, K, S, T and V;
X1010 is an amino acid selected from the group consisting of A, Aib, C, D, E, F, G, Η, I, K, L, Μ, N, Nmf, P, Q, R, S, T, V, W and Y;
X1011 is an amino acid selected from the group consisting of Aib, C, K, G and Nmg;
X1012is Y;
X1013 is an amino acid selected from the group consisting of A, Aib, C, E, F, G, H,
K, L, M, Q, R, W and Y;
X1014 is an amino acid selected from the group consisting of A, Aib, Bhe, C, D, E, F,
G, Η, I, K, L, Μ, N, P, Q, R, S, T, V, W and Y;
X1015 is an amino acid selected from the group consisting of (omega-methyl)-R, D,
E, K and R;
X1016is L;
X1017 is an amino acid selected from the group consisting of (omega-methyl)-R. A, Aib, Bhr, C, Cha, Cit, D, Dab, Dap, E, Eag, Eew, F, G, H, Har, Hci, Hie, I, K, L, Μ, N, Nle, Nva, Opa, Orn, Q, R, S, T, V, W and Y;
X1018 is an amino acid selected from the group consisting of A, Bal, C, D, E, F, G,
H, 1, K, L, Μ, N, Q, R, S, T, V, W and Y; and
X1019 is an amino acid selected from the group consisting of Bhk, K, R and V.
X1020 is either present or absent in formula (1) (i.e., in some instances, the peptide of the invention comprises the structure X1001-X10O2-X1OO3-X1004-X1005-X1006-X100722
2017203999 14 Jun 2017
X1008-X1010-X1011-X1012-X1013-X1014-X10I5-X1016-X1017-X1018-X1019 (SEQ ID
NO: 3116)). When XI020 is present, it is an amino acid selected from die group consisting of Aib, Bhl, C, F, G, H, 1, K, L, Nml, Q, R, S, Τ, V, W and Y.
[0045] For example, the peptide of the invention comprises the structure of formula (I) wherein X1001 is an amino acid selected from the group consisting of C, F, 1, K, L, Nmf, V, M. W and Y; X1002 is Q; X1003 is an amino acid selected from the group consisting of A,
C, D, Ε, Η. K, Μ, I, N, Q, R, S, T and V; XI004 is an amino acid selected from the group consisting of A, Aib, C, D, E, G, H, F, 1, K, k, L, Μ, N, Nmk, P, Q, R, S, V, W and Y; X1005 is an amino acid selected from the group consisting of a. A, Aib, Bal, C, d, E, D, F, G, Η, K, k, L, Μ, N, Nmg, p, Q, R, S, T and Y; X1006 is an amino acid selected from the group consisting of A, Btq, C, D, G, I, K, H, L, Μ, N, Q, R, S, V and Y; X1007 is an amino acid selected from the group consisting of I, K, L, Q, V and Y; XI008 is an amino acid selected from the group consisting of F, H and Y; XI009 is an amino acid selected from the group consisting of f, I and V; X1030 is an amino acid selected from the group consisting of A, D,
E, F, G, Η, K, L, Μ, Ν, P, Q, R, S, Τ, V, W and Y; X1011 is an amino acid selected from the group consisting of G and Nmg; X1012 is Y; X1013 is an amino acid selected from the group consisting of Aib, C, F, H, L, W and Y; X1014 is an amino acid selected from the group consisting of A, Aib, Bhe, C, D, Ε, H, 1, K, L, Μ, N, Q, R, S, Τ, V, W and Y; X1015 is an amino acid selected from the group consisting of E and R; Χ1Ό16 is L; X1017 is an amino acid selected from the group consisting of (omega-methyl )-R, A, Aib, Bhr, C, Cha, Cit, Dab. Dap, Bag, Eew, F, H, Har, Hci, Hie, I, K, L, Μ, N, Nle, Nva, Opa, Orn, R, S, Τ, V and Y; X1018 is an amino acid selected from the group consisting of A, C, D, E, F, I, K, L, Μ, N, Q, R, V and W; X1019 is an amino acid selected from the group consisting of K and R; and X3020 is an amino acid selected from the group consisting of Aib, Bhl, F, K, L, R and W (when XI020 is present in the peptide).
[0046] In one aspect, the peptide of the invention comprises the structure of formula (1) wherein X1001 is an amino acid selected from the group consisting of F, L, Y and Μ; X1002 is Q: X1003 is an amino acid selected from the group consisting of M, Q, R, S, T and C; X1004 is an amino acid selected from the group consisting of Aib, K, L, P, R, E, G, I, Y, M and W; X1005 is an amino acid selected from the group consisting of a, Aib, D, d, G, Η, K, k, N, Nmg, p, Q, R, A. E, C and Μ; X1006 is an amino acid selected from the group consisting of A, C, D, G, Η, K, N, Q, R, S and Μ; XI007 is an amino acid selected from the group consisting of I and V; X1008 is an amino acid selected from the group consisting of F, H and Y; XI009 is V; XI010 is an amino acid selected from the group consisting of A, D, Ε, K, M,
2017203999 14 Jun 2017
N, Q, R, F, Η, P, S, V, W and Y; X1011 is G; X1012 is Y; X10I3 is C or F; X1014 is an amino acid selected from the group consisting of A. C, D, E. K, L, Μ, N, Q. R. Τ, V and Aib; X1015 is R; X1016 is L; X1017 is an amino acid selected from the group consisting of A, Aib, C, Cha, Dab, Dap, Eag, Eew, H, Har, Hci, Hie, K, Nle, Nva, Opa, Orn, R, 1, L, S and M; X1018 is an amino acid selected from the group consisting of A, L, N, M and R; X1019 is K; and XI020 is K or L.
[0047] When amino acid X1020 is absent from formula (I), the peptide of the invention in one aspect further comprises amino acid XI OCX) at the N-terminus of formula (I), such that the peptide comprises or consists of the structure of formula (II); X J000-X1001-X1002Χ1003-Χ1004-Χ1005-Χ1006-Χ1007-Χ1008-Χ1009-Χ1010-Χ10Π-Χ1012-ΧΙ013-Χ1014Χ1Ό15-Χ1Ο16-Χ1Ο17-Χ1ΟΙ8-Χ1Ο19 (II) (SEQ ID NO: 3122). When X1000 is present in the peptide, X1000 is an amino acid selected from the group consisting of A, E, and P, while the amino acids of X1001-X1019 are as defined above.
[0048] In an additional aspect, the TFPI-binding peptide of the invention comprises the structure of formula (ΠΙ): X1001-Q-X1003-X1004-XI005-X1006-I/V-X1008-V-X10IO-GY-C/F-XJ014-R-L-X1017-X1018-K-K/L (III) (SEQ ID NO: 3117). X1001, X1003, ΧΊ004, X1005, X3OO6, ΧΊ008, X1030, X1014, X10I7 and X10.18 in formula (III) are each independently selected from any amino acid. For example, in formula (111),
XI001 is optionally an amino acid selected from the group consisting of Bhf, C, D, F, G, H, 1, K. L, Μ, N, Nmf, Q, R, T, V.W and Y, such as an amino acid selected from the group consisting of C, F, I, K, L, Nmf, V, M, W and Y (e.g,, an amino acid selected from the group consisting of F, L, Y and M);
X I003 is optionally an amino acid selected from the group consisting of A, Aib, Bhs, C, D, E, F, G, H. I, K, L, Μ, N, P, Q, R, S, T, V,W and Y, such as an amino acid selected from the group consisting of A, C, D, E, Η, K, Μ, I, N, Q, R, S, T and V (e.g., the amino acid isM, Q, R, S, TorC);
X1004 is optionally an amino acid selected from the group consisting of. A, Aib, Bhk, C, D, E, F, G, Η, I, K, k, L, Μ, N, Nmk, P, Q, R, S, Τ, V, W and Y, such as an amino acid selected from the group consisting of A, Aib, C, D, E, G, H, F, I, K, k, L, Μ, N, Nmk, P, Q,
R, S, V, W and Y (e.g.. an amino acid selected from the group consisting of Aib. K, L, P, R,
E, G, I, Y, M and W);
XJ005 is optionally an amino acid selected from the group consisting of a, A, Aib,
Bal, C, D, d, E, F, G, Η, K, k, L, Μ, N, Nmg, p, Q, R, S, Τ, V, W and Y, such as an amino
2017203999 14 Jun 2017 acid selected from the group consisting of a, A. Aib, Bal, C, d, E, D, F, G, Η, K, k, L, Μ, N. Nmg, p, Q, R, S, T and Y (e.g., the amino acid is a, Aib, D, d, G, Η, K, k, N, Nmg, p, Q, R,
A, E, CorM);
X3OO6 is optionally an amino acid selected from the group consisting of A, Aib, Btq, C, D, E, F, G, H, 1, K, L, Μ, N, Q, R, S, Τ, V, W and Y, such as an amino acid selected from the group consisting of A, Btq, C, D, G, I, K, H, L, Μ, N, Q, R, S, V and Y (e.g., an amino acid selected from the group consisting of A, C, D, G, Η, K, N, Q, R, S and M);
X1008 is optionally an amino acid selected from the group consisting of F, Η, K, W and Y, such as an amino acid selected from the group consisting of F, H and Y;
X1010 is optionally an amino acid selected from the group consisting of A, Aib, C, D, E, F, G, Η, I, K, L, Μ, N, Nmf, P, Q, R, S, Τ, V, W and Y, such as an amino acid selected from the group consisting of A, D, E, F, G, Η, K, L, Μ, N, P, Q, R, S, Τ, V, W and Y (e.g., an amino acid selected from the group consisting of A, D, E, K, Μ, N, Q, R, F, Η, P, S, V, W and Y);
X1014 is optionally an amino acid selected from the group consisting of A, Aib, Bhe, C, D, E, F, G, Η, I, K, L, Μ, N, P, Q, R, S, Τ, V, W and Y, such as an amino acid selected from the group consisting of A, Aib, Bhe, C, D, E, H, 1, K, L, Μ, N, Q. R, S, Τ, V, W and Y (e.g.. A, C, D, E, K, L, Μ, N, Q, R, Τ, V or Aib);
X1017 is optionally an amino acid selected from the group consisting of (omegamethyl)-R, A, Aib, Bhr, C, Cha, Cit, D, Dab, Dap, E, Eag, Eew, F, G, H, Har, HcL Hie, 1. K, L, Μ, N, Nle, Nva, Opa, Orn, Q, R, S, Τ, V, W and Y, such as an amino acid selected from the group consisting of (omega-methyJ)-R, A, Aib, Bhr, C, Cha, Cit, Dab, Dap, Eag, Eew, F, H, Har, Hci, Hie, I, K, L, Μ, N, Nle, Nva, Opa, Ora, R, S, Τ, V and Y (e.g., an amino acid selected from the group consisting of A, Aib, C, Cha, Dab, Dap, Eag, Eew, H, Har, Hci, Hie,
K. Nle, Nva, Opa, Orn, R, 1, L, S and M); and/or
XI018 is optionally an amino acid selected from the group consisting of A, Bal, C, D, E, F, G, Η, I, K, L, Μ, N, Q, R, S, Τ, V, W and Y, such as an amino acid selected from the group consisting of A, C, D, E, F, I, K, L, Μ, N, Q, R, V and W (e.g., an amino acid selected from the group consisting of A, L, N, M and R).
[0049] In some embodiments, the peptide of the invention comprises one or more additional amino acid residues attached to the N- or C-terminus of the amino acid sequence. For example, the peptide comprising the structure of any one of formulas (1)-(111), in some embodiments, further comprises one or more N-terminal amino acid(s) directly linked to
2017203999 14 Jun 2017
X1001, wherein the N-terminal amino acid(s) comprise the amino acid sequence selected from the group consisting of
XI000,
X999-X1000,
X998-X999-X1000,
X997-X998-X999-X1000 (SEQ ID NO: 3123),
X996-X997-X998-X999-X1000 (SEQ ID NO: 3124),
X995-X996-X997-X998-X999-X1000 (SEQ ID NO: 3125),
X994-X995-X996-X997-X998-X999-X1000 (SEQ ID NO: 3126),
X993-X994-X995-X996-X997-X998-X999-X1000 (SEQ ID NO: 3127),
X992-X993-X994-X995-X996-X997-X998-X999-X1000 (SEQ ID NO: 3128),
X991-X992-X993-X994-X995-X996-X997-X998-X999-X1000 (SEQ ID NO: 3129), and
X990-X991-X992-X993-X994-X995-X996-X997-X998-X999-XJ000 {SEQ ID NO:
3130).
When the peptide comprises one or more N-terminal amino acids, XI000 is A or K; X999 is V or K; X998 is Q or K: X997 is L or K; X996 is R or K; X995 is G or K; X994 is V or K; X993 is G or K; X992 is S or K; X993 is K; and X990 is K.
[0050] In addition to the core structures set forth in formulas (1)-(111), other structures that are specifically contemplated are those in which one or more additional amino acids are attached to the C-terminus of the core stmcture directly linked to XI020. For example, the C-terminal addition optionally comprises an amino acid sequence selected from the group consisting of X1021, X102J-X1022, XI021-XI 022-X1023, and XI021-XI 022-X 1023X1024 (SEQ ID NO: 3131), wherein X102I is Tor K; X1022 is S or K; and X1023 and XI024 are K.
[0051] The invention further includes a TEPI-binding peptide comprising or consisting of an amino acid sequence having at least 60% identity (e.g., at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95 % or 100% identity) to the amino acid sequence Ac-FQSK-Nmg-NVFVDGYFERL-Aib-AKL-NH2 (formula IV) (SEQ ID NO: 164). In some instances, the peptide comprises or consists of the amino acid sequence of any one of formulas (1)-(111), as described herein. The invention also includes a peptide
2017203999 14 Jun 2017 comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 8-978 (e.g., a peptide comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NOs: 8-741 and 962-972 (such as SEQ ID NOs: 8-741, 962-968, 971, or 972) and/or selected from the group consisting of 742-961 (such as SEQ ID NOs: 744-961) and/or selected from the group consisting of SEQ ID NOs: 973-978).
[0052] The peptides of the invention, in some instances, comprise intramolecular disulfide bonds. In this regard, the peptide comprising the structure of formulas (1)-(111) contains at least two cysteine residues (e.g., the peptide contains two cysteine residues) that are spaced apart by at least three amino acid residues such that the cysteines form an intramolecular disulfide bond. In some instances, the cysteines are spaced apart by more than three amino acid residues. For example, in the peptide comprising the structure of formulas (I), (II), or (III), any two of X1000. X1001, X1003, XI004, X1005, X1006, X1010, X1011, X1013, X10I4, X1017, X1018, XT020 andX1021 are optionally cysteines capable of forming a disulfide bridge. Accordingly, in some aspects, the peptide contains two cysteine residues; one of X1000, X1005, X1010 and X1014 is cysteine, and one of X1006, X1010, X1017 and X I021 is a cysteine. The invention contemplates all of the possible combinations of cysteine pairs, e.g., X1000 and X1006 are C; XJOOO and X1010 are C; X1000 and X1017 are C; X1005 and X1017 are C; X1010 and X1017 are C; X1010 and X1021 are C; or X1014 and XI021 areC.
[0053] The invention further provides a peptide that binds TFPI, the peptide comprising the structure of formula (V): X2001 -X2002-X2003-X2004-X2005-X2006-[X2007-X2008X2009-X2010-X2011-X2012-X2013-X2014-X2015-X2016-X2017-X2018]-X2019-X2020X2021-X2022-X2023 (V) (SEQ ID NO: 3118), wherein the peptide forms a cyclic structure generated by a linkage, e.g., a disulfide bond, between X2007 and X2018 (denoted as brackets within formula (V)). In formula (V), X2001, X2002, and X2023 are independently either present or absent. When present, X2001 is an amino acid selected from the group consisting of A, D, E, F, G, Η, I, K, L, P, R, S, T, V and W: X2002 an amino acid selected from the group consisting of A, D, E, F, G, Η, I, K, L, Μ, P, R, S, T, V and W; and X2023 is an amino acid selected from the group consisting of A, D, E, F, G, J, K, L, R, S, T, V. W and Y, In addition,
X2003 is an amino acid selected from the group consisting of A. F, 1, K, L, R, S, T, V, W and Y;
2017203999 14 Jun2017
X2004 is an amino acid selected from the group consisting of A, D, E, F, G, I, K, L, R, S.T. VandW;
X2OO5 is W;
X2006 is an amino acid selected from the group consisting of F. Η, I, K, L. R, V and W;
X2007 is an amino acid selected from the group consisting of C, Hey, Dap and K (e.g., C or Hey);
X2OO8 is an amino acid selected from the group consisting of A, G, R, S and T;
X2OO9 is an amino acid selected from the group consisting of a. A, 1, K, L, M, m, Nle, p, R, Sem, and V;
X2010 is an amino acid selected from the group consisting of A, G, I, K, L, P, R, S, T and V;
X2011 is an amino acid selected from the group consisting of D, E, G, S, and T;
X2012 is an amino acid selected from the group consisting of A, a, D, d, E, e, F, f, G, I, K, lc, L, 1, M, m, Nle, nle, P, p, R, r, S, s, Sem, T, t, V, v, W and w;
X2O13 is an amino acid selected from the group consisting of A, D, d, E, e, F, G, I, K,
L, R, S, s, Τ, V and W;
X2014 is an amino acid selected from the group consisting of A, D, E, F, G, I, K, L,
M, R, S, Τ, V and W;
X20I5 is an amino acid selected from the group consisting of A, D. E, F, G, 1, K, L, M, Nle, R, S, Τ, V and W;
X2016 is an amino acid selected from the group consisting of A, D, E, F, I, K, L, M, Nle, R, S, Sem, Τ, V, W and Y;
X2O37 is an amino acid selected from the group consisting of A, D, E, F, G, I, K, L,
R, S, Τ, V, W and Y;
X2018 is an amino acid selected from the group consisting of C and D (e.g., X2018 is C);
X2019 is an amino acid selected from the group consisting of A, F, I, L, S, Τ, V and W;
X2020 is an amino acid selected from the group consisting of F and W;
2017203999 14 Jun 2017
X2021 is an amino acid selected from the group consisting of I, L and V: and
X2022 is an amino acid selected from the group consisting of A, D, E, F, G, I, K, L, P, R, S, Τ, V, and W.
[0054] In some instances, in the peptide of the invention comprising the structure of formula (V),
X2001 is optionally an amino acid selected from the group consisting of A, D, F, G, Η, K, L, P and S, such as an amino acid selected from the group consisting of A, D, F, G, H,
K, L and S (when X2001 is present).;
X2002 is optionally an amino acid selected from the group consisting of A, D, F, G, Η, K, L, P, R and S, such as an amino acid selected from the group consisting of A, F, Η, K,
L, M, R and S (e.g., H, F, M or R) (when X2002 is present);
X2003 is optionally an amino acid selected from the group consisting of A, F, K, L, S and Y, such as an amino acid selected from the group consisting of F, S and Y (e.g., F or Y);
X2004 is optionally an amino acid selected from the group consisting of A, D, F, G, K, L and S (e.g., K);
X2OO5 is optionally W;
X2006 is optionally an amino acid selected from the group consisting of F, Η, K and L (e.g., F orH);
X2007 is optionally an amino acid selected from the group consisting of C and HcY (e.g,, X2007 is C);
X200S is optionally an amino acid selected from the group consisting of A, G and S;
X2009 is optionally an amino acid selected from the group consisting of a, A, K, L,
V, M, m, Nle, Sem, and p, such as an amino acid selected from the group consisting of M, Nle, p and V (e.g., M, Sem, or V);
X2010 is optionally an amino acid selected from the group consisting of A, G, K, L,
P, R and S, such as an amino acid selected from the group consisting of A. K, L, P, R and S (e.g., K, P, or R);
X2011 is optionally an amino acid selected from the group consisting of D, G and S (e.g., D or S);
X2012 is optionally an amino acid selected from the group consisting of A, a, D, d, F, f, G, K, k, L, 1, M, m, Nle, P, S and s, such as an amino acid selected from the group
2017203999 14 Jun 2017 consisting of D, d, F, f. G, K, k, L. 1, M, Nle, P, S, and Sem (e.g., an amino acid selected from the group consisting of F, L, I, Sem, and M);
X2013 is optionally an amino acid selected from the group consisting of A, D, d, F,
G, K, L, S and s, such as an amino acid selected from the group consisting of A, D, F, G. K.
L and S (e.g., D, G, K, or S);
X2014 is optionally an amino acid selected from the group consisting of D, F, G, K, L and S (e.g., D or G);
X2015 is optionally an amino acid selected from the group consisting of A, D, F, G, 1, K, L, M, Nle, S and T (e.g., I or T);
X2016 is optionally an amino acid selected from the group consisting of D, F, K, L,
M, Nle, S, and Y, such as an amino acid selected from the group consisting of D, F, K, L, M, Nle, S, Sem, and Y (e.g., D, F, M, Sem, or Y);
X2017 is optionally an amino acid selected from the group consisting of A, D, F, G,
K, L, S, T and Y (e.g., S or T);
X2018 is optionally C;
X2019 is optionally an amino acid selected from the group consisting of A, F, L, S and V (e.g., A or V);
X2020 is optionally an amino acid selected from the group consisting of F and W (e.g., W);
X2021 is optionally an amino acid selected from the group consisting of L and V (e.g., V);
X2022 is optionally an amino acid selected from the group consisting of A, D, F, G,
K, L, P, R, S and W. such as an amino acid selected from the group consisting of A, F, G, K,
L, P, R, S and W (e.g., an amino acid selected from the group consisting of F, L, K, R, P and W); and
X2023 is optionally an amino acid selected from the group consisting of A, D, F, G,
K, L, M, S and Y, such as an amino acid selected from the group consisting of A, D, F, G, L
M, S and Y (e.g.. an amino acid sequence selected from the group consisting of A, D, F, M, S and Y) (when X2023 is present).
[0055] The invention further includes a peptide that binds TFPI, wherein the peptide comprises the structure of formula (VI): X200J -X2002-F/Y-K-W-F/H-[C-X2008-M/V30
2017203999 14 Jun2017
X2010-D-X2012-X2013-G-I/T-X2016-S/T-C]-A/V-W-V-X2022-X2023 (VI) (SEQ ID NO: 3119). In the peptide comprising the structure of formula (VI), X2001, X2002 and X2023 are each independently present or absent. If X2001, X2002, and/or X2023 are present, any of X2001, X2002 and X2023 is independently selected from any amino acid. In addition, X2008, X2010, X2012, X.2013, X2016, and X2022 are each independently selected from any amino acid.
[0056] In some aspects, in the peptide of formula (VI),
X2001 is optionally an amino acid selected from the group consisting of A, D, E, F,
G, Η, I, K, L, P, R, S, Τ, V and W, such as an amino acid selected from the group consisting of A, D, F, G, H, K. L, P and S (e.g., an amino acid selected from the group consisting of A, D, F, G, Η, K, L and S) (when X2001 is present);
X2002 is optionally an amino acid selected from the group consisting of A, D, E, F,
G, Η, I, K, L, Μ, P, R, S, Τ, V and W, such as an amino acid selected from the group consisting of A, D. F, G, Η, K, L, Μ, P, R and S (e.g,, an amino acid selected from the group consisting of A, F, Η, K, L, M, R and S, such as H, F, M, or R) (when X2002 is present);
X2OO8 is optionally an amino acid selected from the group consisting of A, G, R, S and T, such as an amino acid selected from the group consisting of A, G and S;
X2010 is optionally an amino acid selected from the group consisting of A, G, I, K, L, P, R, S, T and V, such as an amino acid selected from the group consisting of A, G, K, L, P,
R and S (e.g., an amino acid selected from the group consisting of A, K, L. P, R and S, such as K, P or R);
X2012 is optionally an amino acid selected from the group consisting of A, a, D, d, E, e, F, f, G, I, I, K, k, L, 1, M, m, Nle, nle, P, p, R, r, S, s, Sem, T, t, V, v , W and w, such as an amino acid selected from the group consisting of A, a, D, d, F, f, G, K, k, L, 1, M, m, Nle, P,
S, s, and Sem (e.g., an amino acid selected from the group consisting of D, d, F, f, G, K, k, L,
1, M, Nle, P, S, and Sem, such as F, L, 1, Sem, or M);
X2013 is optionally an amino acid selected from the group consisting of A, D, d, E, e, F, G, I, K, L, R, S, s, Τ, V and W, such as an amino acid selected from the group consisting of A, D, d, F, G, K, L, S and s (e.g,, an amino acid selected from the group consisting of A,
D, F, G, K, L and S, such as D, G, K or S);
X2016 is optionally an amino acid selected from the group consisting of A, D, E, F. I, K, L, M, Nle, R, S, Sem, Τ, V, W and Y, such as an amino acid selected from the group
2017203999 14 Jun 2017 consisting of D, F, K, L. M, Nle, S Sem, and Y (e.g., an amino acid selected from the group consisting of D, F, K, L, M, Nle, S, Sem, such as F, Sem, or M);
X2022 is optionally an amino acid selected from the group consisting of A, D, E, F, G, 1, K, L, P, R, S, Τ, V, and W, such as an amino acid selected from the group consisting of A, D, F, G, K, L, P, R, S and W (e.g.. an amino acid selected from the group consisting of A,
F, G, K, L, P, R, S and W, such as F, L, K, R, P or W); and/or
X2023 is optionally an amino acid selected from the group consisting of A, D, E, F,
G, 1, K, L, R, M, S, Τ, V, W and Y, such as an amino acid selected from the group consisting of A, D, F, G, K, L, M, S and Y (e.g., an amino acid selected from the group consisting of A, D, F, G, L M, S and Y, such as A, D, F, M, S or Y) (when X2023 is present).
[0057] The TFPFbinding peptide of the invention, in one aspect, comprises an amino acid sequence having at least 60% identity (e.g., at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95% or 100% identity) to the sequence of formula VII: Ac-FYYKWH[CGMRDMKGTMSC]AWVKF-NH2 (VII) (SEQ ID NO: 1040). Optionally, the peptide comprises or consists of the amino acid sequence of formula (V)-(VII) as defined herein. The invention also includes a peptide comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NOs: 1001-1293 (e.g., a peptide comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NOs: 1001-1212 and 1290-1291 (such as SEQ ID NOs: 1001-120, 1290, or 1291) and/or selected from the group consisting of SEQ ID NOs; 1213-1289 and/or selected from the group consisting of 1292 and 1293).
[0058] The invention further provides a TFPI-binding peptide comprising the structure of formula (VIII): X300I-X3002-X3003-X3004-X3005-X3006-X3007-X3008-X3009-X3010X3011-X3012-X3013-X3014-X3015-X3016-X3017-X3018-X3019-X3020-X3021 (VIII) (SEQ ID NO: 3120). In formula (VIII), X3001 and X3002 are independently either present or absent in the peptide. If present, X3001 is an amino acid selected from the group consisting of A, C, D, F, G, 1, K, L, Μ, N, P, Q, R, S, T, W, Ε, H and Y; and X3002 is an amino acid selected from the group consisting of A, C, D, F, Η, K, Μ, N, P, R, S, T, W, Y, G, I and L. In addition,
X3003 is an amino acid selected from the group consisting of A, C, D, E, F, G, H, 1,
K, L, Μ, N, P, Q, R, S, T, W and Y;
X3004 is an amino acid selected from the group consisting of A, C, D, E, F, G, H, I,
K, L, Μ, N, Q, R, S, Τ, V, W, Y and P;
2017203999 14 Jun 2017
X3OO5 is an amino acid selected from the group consisting of C, D, F, G, Η, I, K. L, Μ, N, P, R, S, T. V, W and Y;
X3006 is an amino acid selected from the group consisting of A, W, C, K, P, R and H;
X3OO7 is an amino acid selected from the group consisting of Q, A, C, E G, Η, I, K,
L, N, R, S, T, W and Y;
X3OO8 is an amino acid selected from the group consisting of A, C. F, G, Η. K, L, M, N, P, Q, R, S, T, V, W, Y and I;
X3009 is an amino acid selected from the group consisting of A, C, E G, H. 1, L, M,
R, S,T, V,W, Y and K;
X3010 is an amino acid selected from the group consisting of A, C, F, G, HL I, K, L, Μ, N, P, Q, R, S, T, V, W and Y;
X3011 is an amino acid selected from the group consisting of A, G, 1, K, L, Μ, N, Q, R. S,T, V, W, Y,C,FandH;
X3O12 is an amino acid selected from the group consisting of A, C, H, I, K. L and R;
X3013 is an amino acid selected from the group consisting of A, C, F, G. Η, K, L, M, R. S, V, W, Y and T;
X3014 is an amino acid selected from the group consisting of A, CL F, G, H, 1, L, M,
N. Q, R, S, T, V, W, Y and K;
X3015 is an amino acid selected from the group consisting of A, K and R;
X3016 is an amino acid selected from the group consisting of A, E K and R;
X3017 is an amino acid selected from the group consisting of A, C, F, G. I, K, L, N,
Q. R, S, T, V, W, Y, H, A and M;
X3018 is an amino acid selected from the group consisting of A, C, F, F K. L, M, Q,
R, V, W and Y;
X3019 is an amino acid selected from the group consisting of A, C, D, E. F, G, Η, K,
L, N, P, Q. R, V, W, Y and I;
X3020 is an amino acid selected from the group consisting of A, C, F, G, Η, K, L, M. N, Q, R, V, W, Y, 1 and P; and
X3O23 is an amino acid selected from the group consisting of A, C. Η, I, K, L, Μ, N,
P, Q, R, T, V, W, Y, F and G.
2017203999 14 Jun 2017 [0059] In some aspects of the invention, the peptide comprises the sequence of formula (VIII), wherein
X3001 is optionally an amino acid selected from the group consisting of A, C, D, G, I, K, L, Μ, N, P, Q, R, S, T, W. E, H and Y, such as an amino acid selected from the group consisting of A, C, D, G, K, L, Μ, N, P, R, S, T, E, H and Y (when X3001 is present);
X3OO2 is optionally an amino selected from the group consisting of C, F, Η, K, R, S, W, Y, G, 1 and L, such as an amino acid selected from the group consisting of C, K, R, W, Y, G, I and L (when X3002 is present);
X3OO3 is optionally an amino acid selected from the group consisting of A, C, D, F,
G, Η, I, K, L, Μ, N, P, Q, R, S, T and W, such as an amino acid selected from the group consisting of A, C, G, H, I, K, L, M, R, S, T and W;
X3004 is optionally an amino acid selected from the group consisting of A, C, D, G,
FL 1, K, L, Μ, N, R, S, T, V and P, such as an amino acid selected from the group consisting of A, C, G, Η, I, K, L, Μ, N, R, S, T and P;
X3005 is optionally an amino acid selected from the group consisting of C, F, Η, I, K, M, R, T, W and Y, such as an amino acid selected from the group consisting of C, F. Η, K, R and W;
X3006 is optionally an amino acid selected from the group consisting of P, H and A;
X3007 is optionally an amino acid selected from the group consisting of C, G, R, W,
A and L, such as an amino acid selected from the group consisting of L, C, R and W;
X3OO8 is optionally an amino acid selected from the group consisting of A, C, F, G,
Η, K, L, Μ, N, Q, R, T, V, W, Y and I, such as an amino acid selected from the group consisting of A, C, F, Η, K, R, V, W, Y and I;
X3009 is optionally an amino acid selected from the group consisting of C, I, R, V and K, such as an amino acid selected from the group consisting of C, R, V and K;
X3010 is optionally an amino acid selected from the group consisting of A, C. G, Η, I,
K, L, M, Q, R, S and T, such as an amino acid selected from the group consisting of A, C, K,
L, Q, R and S;
X3011 is optionally an amino acid selected from the group consisting of A, I, K, L,
M, R, S, V, W, C, F and H, such as an amino acid selected from the group consisting of I, K,
L, M, R, V, W, C, F and H;
2017203999 14 Jun 2017
X3012 is optionally an amino acid selected from the group consisting of H and R (e.g., H);
X3O13 is optionally an amino acid selected from the group consisting of C, F, K. L, M, R, V and I, such as an amino acid selected from the group consisting of C, K, R, V and I;
X3014 is optionally an amino acid selected from the group consisting of A, M, C, F, Η, I, L, N, R, S, V, W and K, such as an amino acid selected from the group consisting of A, S, C, F, Η, I, R and K;
X3O15 is optionally K or R;
X3016 is optionally K or R;
X3O17 is optionally an amino acid selected from the group consisting of A, C, F, G, I, K, L, N, Q, R, S, T, V, W, H, A and M, such as an amino acid selected from the group consisting of C, G, I, K, L, N, Q, R, S, T, V, H, A and M;
X3O18 is optionally an amino acid selected from the group consisting of A, K, C, I, L, R and W (e.g., K, C, I, R, or W);
X3OI9 is optionally an amino acid selected from the group consisting of A, C, E, H,
K, N, Q, R and I, such as an amino acid selected from the group consisting of C, E, Η, K, R and I;
X3020 is optionally an amino acid selected from the group consisting of C, H, L, M,
R, V, I and P (e.g., C, Μ. I, or P); and
X302 J is optionally an amino acid selected from the group consisting of A, C, Η, I, K,
L, Μ, N, Q, R, V, W, Y, F and G, such as an amino acid selected from the group consisting of A, C, Η, I, K, L, Μ, N, Q, R, V, W, F and G.
[0060] The invention further provides a peptide that binds TFPi and comprises the structure of formula (IX): X3001-X3002-X3003-X3004-X3005-X3006-X3007-X3008X3009-X30J0-X3011-H-X3013-X3014-K/R-R-X3017-X3018-X3019-X3020-X3021 (IX) (SEQ ID NO: 3121), In formula (IX), X3001 and X3OO2 are independently either present or absent in the peptide. If present, X3001 and/or X3002 are independently selected from any amino acid. Likewise, X3003, X3004, X3OO5, X3006, X3OO7, X3008, X3009, X30I0,
X3011, X3013, X3014, X3017, X3018, X3019, X3020 and X3021 are each independently selected from any amino acid. When present, X300I is optionally an amino acid selected from the group consisting of A, C, D, F, G, I, K, L, Μ, N, P, Q, R, S, T, W, E, H and Y, such as an amino acid selected from the group consisting of A, C, D, G, I, K, L, Μ, N, P, Q, R, S,
2017203999 14 Jun 2017
T, W, E, H and Y (e.g., an amino acid selected from the group consisting of A, C, D, G, K, L, Μ, N, P, R, S, T, E, H and Y). Likewise, when present, X3002 is optionally an amino acid selected from the group consisting of A, C, D, F, Η, K, Μ, N, P, R, S, T, W, Y, G, J and L, such as an amino acid selected from the group consisting of C, F, Η, K, R, S, W, Y, G, I and L (e.g., an amino acid selected from the group consisting of C, K, R, W, Y, G, I and L), Also with respect to formula (IX),
X3OO3 is optionally an amino acid selected from the group consisting of A, C, D, E,
F, G. Η, I, K, L, Μ, N, P, Q, R, S, T, W, and Y, such as an amino acid selected from the group consisting of A, C, D, F, G, Η. I, K, L, Μ, N, P, Q, R, S, T and W (e.g., an amino acid selected from the group consisting of A, C, G, Η, I, K, L, M, R, S, T and W);
X3004 is optionally an amino acid selected from the group consisting of A, C, D, E,
F, G, Η, I, K, L, Μ, N, Q, R, S, T, V, W, Y and P, such as an amino acid selected from the group consisting of A, C. D, G, H, 1, K, L, Μ, N, R, S, T, V and P (e.g., an amino acid selected from the group consisting of A, C, G, Η, I, K. L, Μ, N, R, S, T and P);
X3OO5 is optionally an amino acid selected from the group consisting of C, D, F, G,
Η, I, K, L, Μ, N, P, R, S, T, V, W and Y, such as an amino acid selected from the group consisting of C, F, Η, I, K, M, R, T, W and Y (e.g., an amino acid selected from the group consisting of C, F, Η, K, R and W);
X3006 is optionally an amino acid selected from the group consisting of A, W, C, K,
P, R and H, such as an amino acid selected from the group consisting of P, H and A;
X3007 is optionally an amino acid selected from the group consisting of Q, A, C, F,
G, Η, I, K, L, N, R, S, T, W and Y, such as an amino acid selected from the group consisting of C, G. R, W, A and L (e.g., L, C, R or W);
X3008 is optionally an amino acid selected from the group consisting of A, C, F, G.
Η, K, L, Μ, N, P, Q, R, S, T, V, W, Y and I, such as an amino acid selected from the group consisting of A, C, F, G, Η, K. L, Μ, N, Q, R, T, V, W, Y and 1 (e.g., an amino acid selected from the group consisting of A, C, F, Η, K, R, V, W, Y and I);
X3009 is optionally an amino acid selected from the group consisting of A, C, F, G,
Η, I, L, M, R, S, T, V, W, Y and K, such as an amino acid selected from the group consisting of C, I, R, V and K (e.g., C, R, V or K);
X3010 is optionally an amino acid selected from the group consisting of A, C, F, G,
Η, I, K, L, Μ, N. P, Q, R, S, T, V, W and Y, such as an amino acid selected from the group
2017203999 14 Jun 2017 consisting of A, C, G, Η, I, K, L, M, Q, R, S and T (e.g., an amino acid selected from the group consisting of A, C, K. L, Q, R and S);
X3011 is optionally an amino acid selected from the group consisting of A, G, I, K, L, Μ, N, Q, R, S, Τ, V, W, Y, C. F and H, such as an amino acid selected from the group consisting of A, I, K, L. ML R, S, V, W, C, F and H (e.g., an amino acid selected from the group consisting of I, K, L, M, R, V, W, C, F and H);
X3O13 is optionally an amino acid selected from the group consisting of A, C, F, G,
Η, K, L, M, R, S, V, W, Y and I, such as an amino acid selected from the group consisting of C, F, K, L, M, R, V, and I {e.g., C, K, R, V, or I);
X3014 is optionally an amino acid selected from the group consisting of A, C, F, G,
Η, Ϊ, L, Μ, N, Q, R, S, Τ, V, W, Y and K, such as an amino acid selected from the group consisting of A, M, C, F, Η, I, L, N, R, S, V, W and K (e.g., an amino acid selected from the group consisting of A, S, C, F, H, 1, R and K);
X3017 is optionally an amino acid selected from the group consisting of A, C, F, G, I,
K, L, N, Q, R, S, Τ, V, W, Y, H, A and M, such as an amino acid selected from the group consisting of A, C, F, G, I, K, L, N, Q, R, S, Τ, V, W, H, A and M {e.g., an amino acid selected from the group consisting of C, G, I, K, L, N, Q, R, S, Τ, V, H, A and M);
X3018 is optionally an amino acid selected from the group consisting of A, C, F, 1, K,
L, M, Q, R, V, W and Y, such as an amino acid selected from the group consisting of A, K,
C, 1, L, R and W (e.g., K, C, I, R, or W);
X3019 is optionally an amino acid selected from the group consisting of A, C, D, E,
F, G, Η, K, L, N, P, Q, R, V, W, Y and I, such as an amino acid selected from the group consisting of A, C, E, Η, K, N, Q, R and I (e.g., C, E, Η, K, R, or I);
X3020 is optionally an amino acid selected from the group consisting of A, C, F, G,
Η, K, L, Μ, N, Q, R, V, W, Y, 1 and P, such as an amino acid selected from the group consisting of C, H, L, M, R. V, I and P (e.g., C, Μ, I, or P); and/or
X3021 is optionally an amino acid selected from the group consisting of A, C, H, 1, K, L, ML N, P, Q, R, Τ, V, W, Y, F and G, such as an amino acid selected from the group consisting of A, C, Η. I, K, L, Μ, N, Q, R, V, W, Y, F and G (e.g,, an amino acid selected from the group consisting of A, C, Η, I, K, L, Ml, N, Q, R, V, W, F and G).
[0061] The TFPI-binding peptide of the invention comprises, in some aspects, an amino acid sequence having at least 60% identity (e.g., at least 65%, at least 70%, at least 75%, at
2017203999 26 Apr 2019 least 80%, at least 85%, at least 90%, at least 95% or 100% identity) to the sequence of formula (X); Ac-GYASFPWFVQLHVHKRSWEMA-NH2 (X) (SEQ ID NO; 2023). Optionally, the peptide comprises or consists of the amino acid sequence of formula (VIII)(IX) as defined herein. As used herein, “at least 60% identity” and similar terms encompass any integer from, e.g., 60%, to 100%. such as 60%, 61%, 62%, and the like. Also, the term “at least [percentage] identity” encompasses any percentage that is greater than or equal to the number of identical amino acids divided by the total number of amino acids of the peptide of the invention ([at least percentage identity] > [number of identical amino acids] / [total number of amino acids of the peptide of the invention]).
[0062] The invention also includes a peptide comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NOs: 2001-2498 (e.g., a peptide comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NOs; 2001-2296 and 2498 (such as SEQ ID NOs: 2001-2126, 2128-2296, or 2498) and/or selected from the group consisting of SEQ ID NOs: 2297-2497 (such as SEQ ID NOs; 2298-2497)). The invention further provides a peptide comprising or consisting of the amino acid sequence selected from the group consisting of SEQ ID NOs; 3001-3108 (e.g., a peptide comprising or consisting of the amino acid sequence selected fiom the group consisting of SEQ ID NOs: 3001-3064 (such as SEQ ID NOs: 3001-3048, 3051-3053, 3055, or 3057-3064) and/or selected fiom the group consisting of SEQ ID NOs: 3065-3084 (such as SEQ ID NOs: 3066-3084) and/or selected from the group consisting of SEQ ID NOs; 3085-3108).
[0063] The peptide of SEQ ID NOs: 1 -7 also, in some aspects, comprises one or more amino acids attached at the N- or C-terminus of SEQ ID NOs: 1-7. For example, the invention includes a peptide comprising or consisting of the amino acid sequence of
JBT0047, JBT0051, JBTOO55, JBT013J, JBT0I32, JBT0133, JBT0J55, JBT0158, JBT0162,
JBT0163, JBT0164, JBT0I66, JBT0169, JBT0170, JBT0171, JBT0J74, JBT0175, or
JBT0293, all of which comprise the amino acid sequence of SEQ ID NO: 1. Exemplary peptides comprising the amino acid sequence of SEQ ID NO: 2 include peptides comprising or consisting of the amino acid sequence of JBT0294, JBT0295, JBT0296, JBT0297,
JBT0298, JBT0299, JBT0300, JBT0301, JBT0302, JBTO3O3, JBT0304, JBTO3O5, JBTO306,
JBT0307, JBTO3O8, JBT0309, JBT03I0, or JBT0311. Exemplary peptides comprising the amino acid sequence of SEQ ID NO: 3 comprise or consist of the amino acid sequence of
JBT0049, JBTOO53, JBT0057, JBT0190, JBT0I93, or JBT0197. The invention further includes a peptide comprising or consisting of the amino acid sequence of JBT0050,
JBT0054. JBTOO58, JBT0129, JBT0130, JBT0205. JBT0208. JBT02JJ. JBT0212, JBT0217.
2017203999 14 Jun 2017
JBT0218, or JBT0219, all of which include the amino acid sequence of SEQ ID NO: 4, Exemplary peptides comprising SEQ ID NO: 5 include those comprising or consisting of the amino acid sequence of JBT0101, JBT0052, JB70103, JBT0178, or JBT0182. The invention additionally includes a peptide comprising or consisting of the amino acid sequence of JBT0120, JBT0124, JBT0247. JBT0248, JBT0251, or JBT0252, each of which include the amino acid sequence of SEQ ID NO: 6. A peptide including the amino acid sequence of SEQ ID NO: 7, e.g., a peptide comprising or consisting of the amino acid sequence of JBT0122, JBT0126. JBT0221. JBT0224. JBT0225, JBT0226, JBT0228, JBT0232, or JBTO233, also provided by the invention. The peptides described herein are set forth in Table 5 of Example 1 and in Figures 12-18.
[0064] In certain embodiments, the peptide of the invention comprises or consists of the amino acid sequence of JBT0047, JBT0049, JBTO1OJ, JBT0I20, or JBT0122 or any of the inventive peptides described herein (e.g., a peptide comprising or consisting of the amino acid sequence of any one of SEQ ID NOs: 1-3108, such as a peptide comprising or consisting of the amino acid sequence of any one of SEQ ID NOs: 8-741, 744-968, 971-978, 1001-1210, 1213-1289, 1290-1293, 2001-2126, 2128-2296, 2298-2498, 3001-3048, 3051-3053, 3055, 3057-3064, and 3067-3108), or a variant of any of the foregoing. By “variant” is meant a peptide comprising one or more amino acid substitutions, amino acid deletions, or amino acid additions to a parent amino acid sequence. Variants include, but are not limited to, peptides having an amino acid sequence that is at least 60%, 65%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to any of the amino acid sequences provided herein while retaining the ability to bind ΤΕΡΪ and/or inhibit TFPI activity. In one embodiment, the peptide comprises or consists of the amino acid sequence of JBT0132, JBT0303, JBT0193, JBT0178, JBT0120, or JBT0224.
[0065] In one aspect, the peptide of the invention consists of 40 amino acids or less, such as 35 amino acids or less. Optionally, the peptide of the invention consists of 25 amino acids or less, or 10 amino acids or less. In various embodiments, the peptide comprises 15-35 amino acid residues (e.g., 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31,32, 33, 34. or 35 amino acid residues). However, it is also contemplated that a peptide described herein comprising one or more deletions is suitable in the context of the invention so long as the peptide blocks TFPI inhibition of the coagulation cascade and, optionally, binds TFPI. In one aspect, the peptide comprising one or more deletions is suitable in the context of the invention so long as the peptide binds TFPI and, optionally, blocks TFPI inhibition of the
2017203999 14 Jun 2017 coagulation cascade. In some aspects, amino acids are removed from within the amino acid sequence, at the N-terminus, and/or at the C-terminus. Such peptide fragments can comprise 3-14 amino acid residues (e.g., 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 amino acid residues).
[0066] Optionally, the peptide of the invention comprises one or more amino acid substitutions (with reference to any of the amino acid sequences provided herein) that do not destroy the ability of the peptide to bind and/or inhibit TFPI. For instance, peptides comprising or consisting of the amino acid sequence selected from the group consisting of JBT0294, JBT0295, JBT0296, JBT0297, JBT0298, JBT0299, JBT0300, JBT0301, JBT0302, JBT0303, JBT0304, JBT0305, JBT0306, JBTO3O7, JBT0308, JBT0309, JBT0310, or JBT0311 are substitutional mutants of the amino acid sequence of JBT0293 (the amino acid sequence of SEQ ID NO: 1 directly linked to a phenylalanine residue at the N-terminus and a lysine reside at the C-terminus) (see Figure 4). Amino acid substitutions include, but are not limited to, those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinities, and/or (4) confer or modify other physiochemical or functional properties on a peptide. In one aspect, the substitution is a conservative substitution, wherein an amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined within the art, and include amino acids with basic side chains (e.g., lysine, arginine, and histidine), acidic side chains (e.g., aspartic acid and glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, and cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, and tryptophan), beta-branched side chains (e.g., threonine, valine, and isoleucine) and aromatic side chains (e.g.. tyrosine, phenylalanine, tryptophan, and histidine). It will be appreciated, however, that a practitioner is not limited to creating conservative substitutions so long as the resulting peptide retains the ability to downregulate, in whole or in part, TFPI activity. The invention also embraces TFPI-inhibitory peptides comprising atypical, non-naturally occurring amino acids, which are well known in the art. Exeniplaty non-naturally occurring amino acids include ornithine, citrulline, hydroxyproline, homoserine, phenylglycine, taurine, iodotyrosine, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, 2-amino butyric acid, y-amino butyric acid, 2-amino isobutyric acid, 3-amino propionic acid, norleucine, norvaline, sarcosine, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, β-alanine, a fluoro-amino acid, a 3-methyl amino acid, α-C-methyl amino acid, a Ν-methyl amino acid, 2amino-isobutyric acid, β-homoglutamatic acid, β-homophenylalanine. β-homolystne, β40
2017203999 14 Jun2017 homoleucine, β-homoasparagine, β-homoglutamine, β-homoarginine, β-homoserine, βhomotyrosine, β-homoaspartic acid, β-homovaline, β-homoas paragin, (S)-cyclohexylalanine, (S)-citrullin, (S)-2,4-diaminobutyric acid, (S)-2,4-diaminobutyric acid, (S)-diaminopropionic acid, (S)-2-propargylglycine, (S)-N(omega)-nitro-arginine, L-homophenylalanine, S)-homoarginine, (S)-homo-citrulIine, (S)-homo-cysteine, (S)-2-amino-5-methyLhexanoic acid, (S)homo-lysine, (S)-norleucine, (S)-jV-methylalanine, (S)-/V-methyl-aspartic acid, (S) -N- methyl glutamic acid, (S)-(V-methyl-phenylalanine, N-methyl-glycine, (S)-N-methyl-lysine, (S)-jVmethyl-leucine, (S)-A-methyl-arginine, (S)-jV-methyl-serine, (S)-V-methyl-valine, (S)-A7methyl-tyrosine, (S)-2-amino-pentanoic acid, (S)-2-pyridyl-aIanine, (S)-ornithine, Lphenylglycin. 4-phenyl-butyric acid and selenomethionine. The individual amino acids may have either L or D stereochemistry when appropriate, although the L stereochemistry is typically employed for all of the amino acids in the peptide.
[0067] The invention further includes TFPI-inhibitory peptide variants comprising one or more amino acids inserted within an amino acid sequence provided herein and/or attached to the N-terminus or C-terminus. In one aspect, the peptide further comprises one or more amino acids that facilitate synthesis, handling, or use of the peptide, including, but not limited to, one or two lysines at the N-terminus and/or C-terminus to increase solubility of the peptide. Suitable fusion proteins include, but are not limited to, proteins comprising a TFPIinhibitory peptide linked to one or more polypeptides, polypeptide fragments, or amino acids not generally recognized to be part of the protein sequence. In one aspect, a fusion peptide comprises the entire amino acid sequences of two or more peptides or, alternatively, comprises portions (fragments) of two or more peptides. In addition to all or part of the TFPI-inhibitory peptides described herein, a fusion protein optionally includes all or part of any suitable peptide comprising a desired biological activity/function. Indeed, in some aspects, a TFPI-inhibitory peptide is operably linked to, for instance, one or more of the following: a peptide with long circulating half life, a marker protein, a peptide that facilitates purification of the TFPI-inhibitory peptide, a peptide sequence that promotes formation of multimeric proteins, or a fragment of any of the foregoing. In one embodiment, two or more TFPI-inhibitory peptides are fused together, linked by a multimerization domain, or attached via chemical linkage to generate a TFPI-inhibitory peptide complex. The TFPI-inhibitor peptides may be the same or different.
[0068] “Derivatives” are included in the invention and include TFPI-inhibitory peptides that have been chemically modified in some manner distinct from addition, deletion, or substitution of amino acids. In this regard, a peptide of the invention provided herein is
2017203999 14 Jun 2017 chemically bonded with polymem, lipids, other organic moieties, and/or inorganic moieties. Examples of peptide and protein modifications are given in Hermanson, Bioconjugate Techniques, Academic Press, (1996). Derivatives are prepared in some situations to increase solubility, stability, absorption, or circulating half-life. Various chemical modifications eliminate or attenuate any undesirable side effect of the agent, in this regard, the invention includes TFPI-inhibitory peptides covalently modified to include one or more water soluble polymer attachments. Useful polymers known in the art include, but are not limited to, polyethylene glycol (PEG) (e.g., PEG approximately 40 kD or 1 kD in size), polyoxyethylene glycol, polypropylene glycol, monomethoxy-polyethylene glycol, dextran, cellulose, poly-(Nvinyl pyiTolidone)-polyethylene glycol, propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer, polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohol, as well as mixtures of any of the foregoing. In one aspect, the peptide of the invention is a PEGylated peptide. For further discussion of water soluble polymer attachments, see U.S. Patent Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192; and 4,179,337. In another aspect, a peptide derivative includes a targeting moiety specific for a particular cell type, tissue, and/or organ. Alternatively, the peptide is linked to one or more chemical moieties that facilitate purification, detection, multimerization, and characterization of peptide activity. An exemplary chemical moiety is biotin, in addition, in one aspect, the peptides of the invention are acylated at the N-terminal amino acid of the peptide. In another aspect, the peptides of the invention are amidated at the C-terminal amino acid of the peptide. In a still further aspect, the peptides of the invention are acylated at the N-terminai amino acid of the peptide and are amidated at the C-terminal amino acid of the peptide.
[0069] Derivatives also include peptides comprising modified or non-proteinogenic amino acids or a modified linker group (see, e.g., Grant, Synthetic Peptides: A User's Guide, Oxford University Press (1992)), Modified amino acids include, for example, amino acids wherein the amino and/or carboxyl group is replaced by another group. Non-limiting examples include modified amino acids incorporating thioamides, ureas, thioureas, acylhydrazides, esters, olefines, sulfonamides, phosphoric acid amides, ketones, alcohols, boronic acid amides, benzodiazepines and other aromatic or non-aromatic heterocycles (see Estiarte et al.. Burgers Medicinal Chemistry, 6th edition, Volume 1, Part 4, John Wiley & Sons, New York (2002)). Modified amino acids are often connected to the peptide with at least one of the above mentioned functional groups instead of an amide bond. Non-proteinogenic amino acids include, but are not limited, to β-alanine (Bal), norvaline (Nva), norleucine (Nle), 4aminobutyric acid (γ-Abu), 2-aminoisobutyric acid (Aib), 6-aminohexanoic acid (ε-Ahx),
2017203999 14 Jun 2017 ornithine (Orn). hydro xyproline (Hyp), taurine, sarcosine, citrulline (Cit), cysteic acid (Coh), cyclohexylalanine (Cha), methioninesulfoxide (Meo), methionine sulfone (Moo), homoserinemethylester (Hsm), propargylglycine (Eag), 5-fluorotryptophan (5Fw), 6fluorotryptophan (6Fw), 3 Ydimethoxyphenyl-alanine (Ear), 3 jft-difluorophenyhlanlne (Dff), 4'-fluoiOphenyl-alanine (Pff), 1-naphthyl-alanine (INi), 1-methyltryptophan (lMw), penicillamine (Pen), homoserine (Hse). t-butylglycine, t-butylalanine, phenylglycine (Phg), benzothienylalanine (Bta), L-homo-cysteine (Hey), N-methyl-phenylalanine (Nmf), 2thienylalanine (Thi), 3,3-diphenylalanine (Ebw), homophenylalanine (Hfe) and S-benzyl-Lcysteine (Ece). These and other non-proteinogenic amino acids may exist as D- or Lisomers. Examples of modified linkers include, but are not limited to, the flexible linker 4,7,10-trioxa-l,13-tridecanediamine (Ttds), glycine, 6-aminohexanoic acid, beta-alanine (Bal), and combinations of Ttds, glycine, 6-aminohexanoic acid and Bal.
[0Θ70] Homologues of the amino acids constituting the peptides of the invention may be as set forth in Table 3.
TABLE 3
Amino Acid Exemplary homologues
A Aib, Bal, Eag, Nma, Abu, G, M, Nva, Nle
C S, A, Hey, M, L, I, V, Nmc, β-Cysteine
D E, Homoglutamic acid, γ-Hydroxy-glutamic acid. γ-Carboxy- glutamic acid, Nmd, β-Aspartic acid, N, Q, Cysteic acid
E D, Glu, Homoglutamic acid, γ-Hydroxy-glutamic acid, γ-Carboxy- glutamic acid, α-Aminoadipic acid, Nme, β-glutamic acid, Q, N, Cysteic acid
F Hfe, Nmf, β-Phenylalanine, Phg, Bhf, Thienyl alanine, Benzothienylalanine, Bromophenyl alanine. Iodophenylalanione, Chlorophenylalanine, Methylphenyialanine, Nitrophenylalanine, Y, W, Naphtylalanine, Tic
G A, Nmg
H Nmh, l-Methylhistidine, 3-Methylhistidine, Thienylalanine
1 L, V, Hie, Nva, Nle, β-Isoleucine, Nml, M, Nmi
2017203999 14 Jun 2017
κ Nmk, R, Nmr, β-Lysine, Dab, Dap, p-(l-Piperazinyl)-alanine, 2,6Diamino-4-hexynoic acid, delta-Hydroxy-lysine, Har, omega- Hydroxy-norarginine, omega-Amino-arginine, omega-Methyl- arginine, p-(2-Pyridyj)-alanine, p-(3-PyridyI)-alanine, 3-Amino- tyrosine, 4-Amino-phenylalanine, Hci, Cit
L 1, V, Hie, Nle, Nva, β-Isoleucine, Nml, M
Μ I, V, Hie, Nva, R, Har, Nmm, Methioninesulfone
N Nmn, β-Asparagine, Q, Nmq, β-Glutamine, Cys(3-propionic acid amide)-OH, Cys(O2-3-propionic acid amide)-OH
P Azetidine-2-carboxylic acid, Hyp, ot-Methyl-methionine, 4- Hydroxy-piperidine-2-carboxylic acid, Pip, OC-Methyl-Pro
Q N, Nmn, Nmq, β-Glutamine, Cys(3-propionic acid amide)-QH, Cys(02-3-propionic acid aniide)-OH
R Nmk, K, Nmr, β-Lysine, Dab, Dap, Ora, PANPiperazinyiy-alanine, 2,6-Diamino-4-hexynoic acid, delta-Hydroxy-lysine, Har, omega- Hydroxy-norarginine, omega-Amino-arginine, omega-Methyl- arginine, β-(2~Pyridyl)-alanine, 3-(3-Pyridy1)-alanine, 3-Amino- tyrosine, 4-Amino-phenylalanine, Hci, Cit, Hie, L, Nle, M
S T, Hse, β-Serine, C, β-Cyano-alanine, alio-Threonine
T S, Homothreonine, β-Threonine, al Io-Threonine
V L, I, Hie, Nva, Nle, β-Valine, Nmv, M, Nmi, Nml
w Nmw, β-Tryptophan, F, Hfe, Nmf, β-Phenylalanine, Phg, Bhf, Thienylalanine, Benzothienylalanine, Bromophenylalanine, lodophenylalanine. Chlorophenyl alanine, Methylphenylalanine, Nitrophenylalanine. Y, Naphtylalanine, Tic
Y Nmy, β-Tyrosine,, F, Hfe, Nmf. β-Phenylalanine, Phg, Bhf, Thienylalanine, Benzothienylalanine, Bromophenylalanine, lodophenylalanine, Chlorophenylalanine, Methylphenylalanine, Mitrophenylalanine, W, Naphtylalanine, Tic
2017203999 14 Jun 2017 [0071] In some embodiments, the peptide (CO-NH) linkages joining amino acids within the peptide of the invention are reversed to create a “retro-modified” peptide, i.e., a peptide comprising amino acid residues assembled in the opposite direction (NH-CO bonds) compared to the reference peptide. The retro-modified peptide comprises the same amino acid chirality as the reference peptide. An “inverso-modified” peptide is a peptide of the invention comprising amino acid residues assembled in the same direction as a reference peptide, but the chirality of the amino acids is inverted. Thus, where the reference peptide comprises L-amino acids, the “inverso-modified” peptide comprises D-amino acids, and vice versa. Inverso-modified peptides comprise CO-NH peptide bonds. A “retro-inverso modified” peptide refers to a peptide comprising amino acid residues assembled in the opposite direction and which have inverted chirality. A retro-inverso analogue has reversed termini and reversed direction of peptide bonds (i.e., NH-CO), while approximately maintaining the side chain topology found in the reference peptide. Retro-inverso peptidomimetics are made using standard methods, including the methods described in Meziere et al, J. Immunol,, 159, 3230-3237 (1997), incorporated herein by reference. Partial retro-inverso peptides are peptides in which only part of the amino acid sequence is reversed and replaced with enantiomeric amino acid residues.
[0072] TFPI-binding peptides of the invention (e.g., TFPI inhibitor peptides) are made in a variety of ways. In one aspect, the peptides are synthesized by solid phase synthesis techniques including those described in Merrifield, J. Am, Chem. Soc., 85, 2149 (1963);
Davis et al., Biochem. Inti,, 10, 394-414 (1985); Larsen et al,,,/. Am, Chem. See., 115, 6247 (1993); Smith et al., J. Peptide Protein Res., 44, 183 (1994); O'Donnell et al.,,/. Am, Chem. Soc., 118, 6070 (1996); Stewart and Young, Solid Phase Peptide Synthesis, Freeman (1969); Finn et al.. The Proteins, 3rd ed., vol. 2, pp. 105-253 (1976); and Erickson et al., The Proteins, 3rd ed., vol. 2, pp. 257-527 (1976). Alternatively, the TFPI-binding peptide (e.g., the TFPJ-inhibitory peptide) is expressed recombinantly by introducing a nucleic acid encoding a TFPI-binding peptide (e.g,, a TFPI-mhibitory peptide) into host cells, which are cultured to express the peptide. Such peptides are purified from the cell culture using standard protein purification techniques.
[0073] The invention also encompasses a nucleic acid comprising a nucleic acid sequence encoding a TFPI-inhibitory peptide of the invention. Methods of preparing DNA and/or RNA molecules are well known in the art. In one aspect, a DNA/RNA molecule encoding a peptide provided herein is generated using chemical synthesis techniques and/or using polymerase chain reaction (PCR). If desired, a TFPI-inhibitory peptide coding sequence is
2017203999 14 Jun 2017 incorporated into an expression vector. One of ordinary skill in the art will appreciate that any of a number of expression vectors known in the art are suitable in the context of the invention, such as, but not limited to, plasmids, plasmid-liposome complexes, and viral vectors. Any of these expression vectors are prepared using standard recombinant DNA techniques described in, e.g., Sambrook et al.. Molecular Cloning, a Laboratory Manual, 2d edition, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. <1989), and Ausubel et al„ Current Protocols in Molecular Biology, Greene Publishing Associates and John Wiley & Sons, New York, N.Y. (1994). Optionally, the nucleic acid is operably linked to one or more regulatory sequences, such as a promoter, activator, enhancer, cap signal, polyadenylation signal, or other signal involved with the control of transcription or translation.
[0074] Any of the TFPl-inhibitory peptides of the invention or nucleic acids encoding the peptides also is provided in a composition (e.g., a pharmaceutical composition). Tn this regard, the peptide is formulated with a physiologically-acceptable (i.e,, pharmacologicallyacceptable) carrier, buffer, excipient, or diluent, as described further herein. Optionally, the peptide is in the form of a physiologically acceptable salt, which is encompassed by the invention. “Physiologically acceptable salts” means any salts that are pharmaceutically acceptable. Some examples of appropriate salts include acetate, hydrochloride, hydrobromide, sulfate, citrate, tartrate, glycolate, and oxalate. If desired, the composition comprises one or more additional pharmaceutically-effective agents.
[0075] The peptide provided herein optionally inhibits at least one Tissue Factor Pathway Inhibitor-1 (e.g., TFPI-la) activity such as, but not limited to, an activity that downregulates the blood coagulation cascade. Without being bound by any specific mechanism of action, a proposed mechanism of inhibition may involve preventing formation of the quaternary TFFVIIA-FXA-TFPI complex. The peptide may inhibit binding of TFPI to FXa (e.g., inhibit binding of TFPI Kunitz domain 2 to Factor Xa), the TF/FVIIa complex (e.g., inhibit binding of TFPI Kunitz domain I to the TF/FVIIa complex), TF alone, and/or FVlIa alone. With TFPI activity diminished, TF and FVlIa are free to activate FX which, in turn, enhances conversion of prothrombin to thrombin, [0076] In one aspect, the peptide of the invention exhibits TFPI antagonistic activity in mode! and/or plasmatic systems. An exemplary model system for determining TFPlinhibitory activity is the extrinsic tenase assay, which tests the ability of candidate peptides to restore extrinsic complex-mediated FX activation in the presence of TFPI (which is a natural inhibitor of the FX activation reaction) (see, e.g., Lindhout et al., Thromb. Haemost., 74, 910915 (1995)). Another model system for characterizing TFPl-inhibitory activity is the FXa
2017203999 14 Jun 2017 inhibition assay, wherein FXa activity is measured in the presence of TFPI (see Sprecher et al., PNAS, 91, 3353-3357 (1994)). The extrinsic tenase assay and the FXa inhibition assay are further described in Example 3. Optionally, the peptide of the invention enhances FX activation in the presence of TFPI with a half maximal effective concentration (EC;o) of less than or equal to 1 x 104M, less than or equal to 1 x ΙΟ’5 M, less than or equal to 1 x 106M, or less than or equal to 1 x 10’7 M.
[0077] In one aspect, TFPI-antagonist activity is characterized in a plasma-based assay. Thrombin formation is triggered in plasma substantially lacking FVIII or FIX activity (e.g., the residual coagulation factor activity is lower than 1.%) in the presence of a candidate peptide. Thrombin formation can be detected using a fluorogenic or chromogenic substrate, as described in Example 4. A system for measuring thrombin activity is provided by Thrombinoscope BV (Maastricht, The Netherlands). Prothrombin conversion is measured using, e.g., a Thrombograph™ (Thermo Scientific, Waltham, MA), and the resulting data is compiled into a Calibrated Automatic Thrombogram generated by Thrombinoscope™ software available from Thrombinoscope B V. In certain embodiments, the TFPI-inhibitory peptide increases the amount of peak thrombin generated during the assay and/or decreases the time required to achieve peak thrombin formation. For example, the peptide improves TFPI-regulated thrombin generation in the absence of FVIII (e.g., in FVJlI-depleted plasma) to at least 1% of the level of TFPI-dependent thrombin generation in normal plasma. Generally, normal (unafflicted) plasma contains about 0.5 U/mL to about 2 U/mL Factor VIII, Accordingly, in some instances, a TFPI-inhibitor peptide will enhance thrombin formation in the absence of FVIII to at least about 1 % of that observed in the presence of 0.5 U/mL to 2 U/mL FVIII. In further embodiments, the peptide enhances thrombin formation in the absence of Factor VIII to at least about 2%, at least about 3%, at least about 5%, at least about 7%, or at least about 10% of the level of thrombin formation in normal plasma, i.e., in the presence of physiological levels of Factor VIII. In various aspects, the peptide is administered to an animal model of thrombin deficiency or hemophilia to characterize TFPI inhibitory activity in vivo. Such in vivo models are known in the art and include for example, mice administered anti-FVID antibodies to induce hemophilia A (Tranholm et al., Blood, 102, 3615-3620 (2003)); coagulation factor knock-out models such as, but not limited to, FVIII knock-out mice (Bi et al., Nat. Genet., 10{ i), 119-121 (1995)) and FIX knock-out mice (Wang et al,, PNAS, 94(23), 11563-66 (1997)); induced hemophilia-A in rabbits (Shen etal,, Blood, 42(4), 509-521 (1973)); and Chapel Hill HA dogs (Lozier et al., PNAS, 99, 1299112996 (2002)).
2017203999 14 Jun2017 [0078] While not being bound to any particular theory or mechanism, the peptide of the invention provided herein may inhibit TFPI activity by blocking (competitively or allosterically) binding of TFPI and FXa, Alternatively or in addition, the peptide may inhibit binding of TFPI with a Tissue Factor (TF)/Factor Vila complex. Thus, in certain aspects, the peptide specifically binds TFPI. Various peptides bind TFPI from any source including, but not limited to, mouse, rat, rabbit, dog, cat, cow, horse, pig, guinea pig, and primate, In one embodiment, the peptide binds human TFPI, Optionally, the TFPI-inhibitory peptide binds TFPI from more than one species (i.e., the peptide is cross-reactive among multiple species). In certain aspects, the peptide binds TFPI with a dissociation constant (Κ») of less than or equal to 1 χ 10’1 M. less than or equal to 1 χ 10'5 M, less than or equal to 1 χ IO'6 M, or less than or equal to 1 x 10’7M. Affinity may be determined using, for example and without limitation, any one, two, or more of a variety of techniques, such as affinity ELISA assay, a competitive ELISA assay, and/or surface plasmon resonance (BIAcore™) assay. When characterized using a competitive (IC50) ELISA assay, the peptide of the invention optionally demonstrates an IC50 of less than or equal to about 50,000 nM, For example, the peptide demonstrates an IC50 of less than or equal to about 10,000 nM, such as an IC50 of less than or equal to about 5,000 nM, less than or equal to about 1,000 nM. or less than or equal to about 500 nM. In one aspect, the peptide demonstrates an IC50 of less than or equal to about 250 nM, less than or equal to about 100 nM, or less than or equal to about 50 nM. Exemplary peptides and their IC50 values are provided in Figures 32-39; in some instances, the peptides are classified into Groups A, B, C, D, E, F, and G (see Table 4 in Example 1) based on their IC50 values. In various aspects, the invention provides peptides falling within Groups A, B,
C, D, E, F, and/or G as defined in Table 4. Affinity may also be determined by a kinetic method or an equilibrium/solution method. Such methods are described in further detail herein or known in the ail.
[0079] As with all binding agents and binding assays, one of skill in the art recognizes that the various moieties to which a binding agent should not detectably bind in order to be biologically (e.g., therapeutically) effective would be exhaustive and impractical to list. Therefore, the term “specifically binds” refers to the ability of a peptide to bind TFPI with greater affinity than it binds to an unrelated control protein that is not TFPI, For example, the peptide may bind to TFPI with an affinity that is at least, 5, 10, 15, 25, 50, 100, 250, 500,
1000, or 10,000 times greater than the affinity for a control protein. In some embodiments, the peptide binds TFPI with greater affinity than it binds to an “anti-target,” a protein or other naturally occurring substance in humans to which binding of the peptide might lead to
2017203999 14 Jun 2017 adverse effects. Several classes of peptides or proteins are potential anti-targets. Because TFPI-inhibitory peptides exert their activity in the blood stream and/or at the endothelium, plasma proteins represent potential anti-targets. Proteins containing Kunitz domains (KDs) are potential anti-targets because KDs of different proteins share a significant similarity. Tissue Factor Pathway lnhibitor-2 (TFPI-2) is highly similar to TFPI-la and, like TFPI-la, contains KDs (Sprecher et al., PNAS, 91, 3353-3357 (1994)). Thus, in one aspect, the peptide of the invention binds to TFPI with an affinity that is at least 5, 10, 15, 25, or 50 times greater than the affinity for an anti-target, such as TFPI-2.
[0080] The invention further includes a method of inhibiting Tissue Factor Pathway Inhibitor-! (TFPI). The method comprises contacting TFPI with a TFPl-binding peptide as described herein. Any degree of TFPl-actlvity inhibition is contemplated. For example, a TFPI-inhibitory peptide reduces TFPI-inhibition of the extrinsic pathway at least about 5% (e.g.. at least about 10%, at least about 25%, or at least about 30%). In some embodiments, the TFPI-inhibitory peptide reduces TFPI activity within the extrinsic pathway at least about 50%, at least about 75%, or at least about 90% compared to TFPI activity in the absence of the peptide.
[0081] The invention further includes a method for targeting biological structures (including, but not limited to, cell surfaces and endothelial lining) where TFPI is located.
The method comprises contacting the biological structure (e.g.. including, without limitation, a cell displaying TFPI on the cell surface) with a TFPl-binding peptide described herein, optionally conjugated to a moiety that adds additional functionality to the peptide. The moiety can be a dye (such as a fluorescence dye), a radionuclide or a radionuclide-containing complex, a protein (e.g., an enzyme, a toxin, or an antibody) or a cytotoxic agent. For example, the peptide is linked or conjugated to an effector moiety that facilitates peptide detection and/or purification and/or comprises therapeutic properties. In one aspect, the TFPl-binding peptide or peptide conjugate is administered to a mammal to target a TFPIdisplaying cell within the mammal. Optionally, the method further comprises detecting binding of the TFPl-binding peptide to TFPI. The method is useful for therapy and diagnostic of disease where TFPI is a suitable diagnostic marker or TFPI-expressing cells are a target for a therapeutic approach.
[0082] In some aspects, peptide-TFPl binding is detected indirectly. In this regal’d, the peptide is contacted with an interaction partner that binds the peptide of invention without significantly interfering with peptide-TFPI binding, and the interaction partner is detected. Exemplary interaction partners include, but are not limited to, antibodies, antigen-binding
2017203999 14 Jun 2017 antibody fragments, anticalins and antibody mimetics, aptamers, and spiegelmers. Optionally, the interaction partner comprises a detection moiety to facilitate detection of an interaction partner-peptide complex. Methods of detecting, e.g., antibodies and fragments thereof, are well understood in the art. Similarly, detection moieties are widely used in the art to identify biological substances and include, for example, dye (e.g., fluorescent dye), radionuclides and radionuclide-containing complexes, and enzymes.
[0083] Thus, the invention provides a method for diagnosing a subject suffering from a disease or disorder, or at risk of suffering from a disease or disorder, wherein the disease or disorder is associated with or caused by aberrant TFPI activity. The method comprises administering to the subject the TFPI-binding peptide of the invention and detecting the TFPI-peptide complex. In some instances, the peptide of the invention is conjugated to a detectable moiety, and the method comprises detecting the detectable moiety. In other instances, the method comprises administering to the subject a TFPI-binding peptide interaction partner that binds the TFPI-binding peptide, and detecting the interaction partner. If desired, the interaction partner comprises or is conjugated to a detectable moiety, and the detectable moiety is detected. The presence of the detectable moiety indicates the presence of TFPI, thereby allowing diagnosis of a disease or disorder associated with TFPI (e.g., a disease or disorder which (i) can be treated by inhibiting TFPI or (ii) comprises symptoms which can be ameliorated or prevented by inhibiting TFPI). If administration of the peptide to the subject is not desired, a biological sample is obtained from the subject, contacted with the TFPI-binding peptide as described herein, and TFPI-peptide complexes are detected.
[0084] The peptides of the invention bind TFPI and, therefore, are useful for purifying TFPI or recombinant TFPI from a biological sample (e.g., a biological fluid, such as serum), fermentation extract, tissue preparations, culture medium, and the like. The invention includes methods of using the TFPI-binding in the commercial production of TFPI or in a method of characterizing TFPI molecules. For example, the invention includes a method of purifying TFPI. The method comprises contacting a sample containing TFPI with a peptide as defined herein under conditions appropriate to form a complex between TFPI and the peptide; removing the complex from the sample: and, optionally, dissociating the complex to release TFPI. Exemplary conditions appropriate to form a complex between TFPI and the peptide are disclosed in the Examples, and such conditions can be easily modified to dissociate the TFPI-peptide complex. In some embodiments, the peptide is immobilized to a support, e.g., a solid support, to facilitate recovery of TFPI. For example, in one embodiment, the peptide is immobilized to chromatography stationary phase (e.g., silica,
2017203999 14 Jun 2017 affinity chromatography beads, or chromatography resins), a sample comprising TFPI is applied to the stationary phase such that TFPl-peptide complexes are formed, the remainder of the sample is removed from the stationary phase, and TFPI is elnted from the stationaiy phase. In this regard, the peptides of the invention are, in one aspect, suitable for use in affinity chromatography techniques.
[0085] A method of enhancing thrombin formation in a clotting factor-deficient subject also is provided. The method comprises administering to the subject a peptide provided herein under conditions effective to inhibit TFPI. In this regard, the TFPI-inhibitory peptide is administered in an amount and under conditions effective to enhance thrombin formation in the subject. By “clotting factor-deficient” is meant that the subject suffers from a deficiency in one or more blood factors required for thrombin formation, such as FVIII, FIX, or FXI. Indeed, in one embodiment, the subject is deficient in FVIII. Alternatively or in addition, the subject is deficient in Factor IX, Clotting factor deficiencies are identified by examining the amount of factor in a clinical sample. Practitioners classify hemophilia according to the magnitude of clotting factor deficiency. Subjects suffering from mild hemophilia have approximately 5% to 30% of the normal amount (] U/ml) of Factor VIII or Factor IX. Moderate hemophilia is characterized by approximately 1 % to 5% of normal Factor VIII, Factor IX, or Factor Xl levels, while subjects suffering from severe hemophilia have less than 1% of the normal amount of Factor VIII, Factor IX, or Factor XI. Deficiencies can be identified indirectly by activated partial thromboplastin time (APTT) testing. APTT testing measures the length of time required for a blood clot to form, which is longer for patients with Factor VIII Deficiency (hemophilia A), Factor IX Deficiency (hemophilia B), and Factor XI Deficiency (hemophilia C) compared to patients with normal clotting factor levels. Almost 100% of patients with severe and moderate Factor VIII deficiency can be diagnosed with an APTT. The invention further includes enhancing thrombin formation in a subject that does not suffer from a clotting factor deficiency. The method comprises administering to a subject (e.g., a subject comprising normal, physiological levels of clotting factor) a peptide provided herein under conditions effective to enhance thrombin formation, [0086] In one aspect, the TFPI-inhibitory peptide is used for increasing blood dot formation in a subject. The method of increasing blood clot formation comprises administering to the subject a peptide described herein in an amount and under conditions effective to increase blood clot formation. It will be appreciated that the method need not completely restore the coagulation cascade to achieve a beneficial (e.g., therapeutic) effect. Any enhancement or increase in thrombin or blood clot formation that reduces the onset or
2017203999 14 Jun 2017 severity of symptoms associated with clotting factor deficiencies is contemplated. Methods of determining the efficacy of the method in promoting thrombin formation and blood clotting are known in the ait and described herein.
[0087] The invention further includes a method of treating a blood coagulation disorder in a subject, the method comprising administering to the subject one or more TFPl-inhibitory peptides, such as any one or more of the peptides described herein, in an amount and under conditions effective to treat the blood coagulation disorder in the subject. In one aspect, the peptide is not a naturally-occurring peptide that inhibits TFPI activity. “Coagulation disorders” include bleeding disorders caused by deficient blood coagulation factor activity and deficient platelet activity. Blood coagulation factors include, but are not limited to, Factor V (FV), FVII, FV1II, FIX, FX, FXI, FXIIJ, FII (responsible for hypoprothrombinemia), and von Willebrand’s factor. Factor deficiencies are caused by, for instance, a shortened in v/w-half-life of the factor, altered binding properties of the factor, genetic defects of the factor, and a reduced plasma concentration of the factor. Coagulation disorders can be congenital or acquired. Potential genetic defects include deletions, additions and/or substitution within a nucleotide sequence encoding a clotting factor whose absence, presence, and/or substitution, respectively, has a negative impact on the clotting factor’s activity. Coagulation disorders also stem from development of inhibitors or autoimmunity (e.g., antibodies) against clotting factors. In one example, the coagulation disorder is hemophilia A. Alternatively, the coagulation disorder is hemophilia B or hemophilia C.
[0088] Platelet disorders are caused by deficient platelet function or abnormally low platelet number in circulation. Low plateiet count may be due to. for instance, underproduction, platelet sequestration, or uncontrolled patent destruction.
Thrombocytopenia (platelet deficiencies) may be present for various reasons, including chemotherapy and other drug therapy, radiation therapy, surgery, accidental blood loss, and other disease conditions. Exemplary disease conditions that involve thrombocytopenia are: aplastic anemia; idiopathic or immune thrombocytopenia (ITP), including idiopathic thrombocytopenic purpura associated with breast cancer; HIV-associated ITP and HIVreiated thrombotic thrombocytopenic purpura; metastatic tumors which result in thrombocytopenia; systemic lupus erythematosus, including neonatal lupus syndrome splenomegaly; Fanconi’s syndrome; vitamin BI2 deficiency; folic acid deficiency; MayHegglin anomaly; Wiskott-Aldrich syndrome; chronic liver disease; myelodysplastic syndrome associated with thrombocytopenia; paroxysmal nocturnal hemoglobinuria; acute profound thrombocytopenia following C7E3 Fab (Abciximab) therapy; alioimmune
2017203999 14 Jun 2017 thrombocytopenia, including maternal alloimmune thrombocytopenia; thrombocytopenia associated with antiphospholipid antibodies and thrombosis; autoimmune thrombocytopenia; drug-induced immune thrombocytopenia, including carboplatin-induced thrombocytopenia and heparin-induced thrombocytopenia; feta] thrombocytopenia; gestational thrombocytopenia; Hughes' syndrome; lupoid thrombocytopenia; accidental and/or massive blood loss; myeloproliferative disorders; thrombocytopenia in patients with malignancies; thrombotic thrombocytopenia purpura, including thrombotic microangiopathy manifesting as thrombotic thrombocytopenic purpura/hemolytic uremic syndrome in cancer patients; posttransfusion purpura (PTP); autoimmune hemolytic anemia; occult jejunal diverticulum perforation; pure red cell aplasia; autoimmune thrombocytopenia; nephropathia epidemica; rifampicin-associated acute renal failure; Paris-Trousseau thrombocytopenia; neonatal alloimmune thrombocytopenia; paroxysmal nocturnal hemoglobinuria; hematologic changes in stomach cancer; hemolytic uremic syndromes (e.g,, uremic conditions in childhood); and hematologic manifestations related to viral infection including hepatitis A virus and CMVassociated thrombocytopenia. Platelet disorders also include, but are not limited to, Von Willebrand Disease, para neoplastic platelet dysfunction, Glanzman’s thrombasthenia, and Bernard-Soulier disease. Additional bleeding disorders amenable to treatment with a TFPIinhibitory peptide include, but are not limited to, hemorrhagic conditions induced by trauma; a deficiency in one or more contact factors, such as FXL FX1I, prekailikrein, and high molecular weight kininogen (HMWK); vitamin K deficiency; a fibrinogen disorder, including afibrinogenemia, hypofibrinogenemia, and dysfibrinogenemia; and alpha2-antiplasmin deficiency, In one embodiment, the TFPJ-inhibitory peptide is used to treat excessive bleeding, such as excessive bleeding caused by surgery, trauma, intracerebral hemorrhage, liver disease, renal disease, thrombocytopenia, platelet dysfunction, hematomas, internal hemorrhage, hemarthroses, hypothermia, menstruation, pregnancy, and Dengue hemorrhagic fever. All of the above are considered “blood coagulation disorders” in the context of the disclosure.
[0089] In one aspect, the TFPl-inhibitory peptide of the invention is used to reverse the effects (in whole or in pari) of one or more anticoagulants in a subject. Numerous anticoagulants are known in the art and include, for instance, heparin; coumarin derivatives, such as warfarin or dicumarol; TFPI; AT III; lupus anticoagulant: nematode anticoagulant peptide (NAPc2); FVIIa inhibitors; active-site blocked FVIIa (FVllai); active-site blocked FIXa (FIXai); FIXa inhibitors; FXa inhibitors, including fondaparinux, idraparinux, DX9065a, and razaxaban (DPC906); active-site blocked FXa (FXai); inhibitors of FVa or
2017203999 14 Jun 2017
FVIlIa, including activated protein C (APC) and soluble thrombomodulin; thrombin inhibitors, including hirudin, bivalirudin, argatroban, and ximelagatran; and antibodies or antibody fragments that bind a clotting factor (e.g., FV, FVII, FVII1, FIX, FX, FXIII, FII, FXI, FXII, von Willebrand factor, prekallikrein, or high molecular weight kininogen (HMWK)).
[0090] As used herein, “treating” and “treatment” refers to any reduction in the severity and/or onset of symptoms associated with a blood coagulation disorder. Accordingly, “treating” and “treatment” includes therapeutic and prophylactic measures. One of ordinary skill in the art will appreciate that any degree of protection from, or amelioration of, a blood coagulation disorder or symptom associated therewith is beneficial to a subject, such as a human patient. The quality of life of a patient is improved by reducing to any degree the severity of symptoms in a subject and/or delaying the appearance of symptoms. Accordingly, the method in one aspect is performed as soon as possible after it has been determined that a subject is at risk for developing a blood coagulation disorder (e.g,, a deficiency in a clotting factor (e.g., FV1II, FIX, or FXI) is detected) or as soon as possible after a blood coagulation disorder (e.g., hemophilia A, hemophilia B, or hemophilia C) is detected. In an additional aspect, the peptide is administered to protect, in whole or in part, against excessive blood loss during injury or surgery.
[0091] In view of the above, the invention provides a peptide for use in a method for the treatment of a subject, such as a method for the treatment of a disease where the inhibition of TFPI is beneficial. In one aspect, the disease or disorder is a blood coagulation disorder. The subject is suffering from a disease or disorder or is at risk from suffering from a disease or disorder (or adverse biological event, such as excessive blood loss). The method comprises administering to the subject the peptide of the invention in an amount and under conditions effective to treat or prevent, in whole or in part, the disease or disorder. The invention further provides a peptide for use in the manufacture of a medicament. For example, the peptide can be used in the manufacture of a medicament for the treatment of a blood coagulation disorder, as described in detail herein.
[0092] In some embodiments, it is advantageous to administer to a subject a nucleic acid comprising a nucleic acid sequence encoding a TFPI-binding peptide (e.g., TFPJ-inhibitory peptide) of the invention. Such a nucleic acid, in one aspect, is provided instead of, or in addition to, a TFPI-inhibitory peptide. Expression vectors, nucleic acid regulatory sequences, administration methods, and the like, are further described herein and in U.S. Patent Publication No. 20030045498.
2017203999 14 Jun 2017 [0093] A particular administration regimen for a particular subject will depend, in part, upon the TFPI- inhibitory peptide of the invention used, the amount of TFPI-binding peptide (e.g., TFPI-inhibitory peptide) administered, the route of administration, the particular ailment being treated, considerations relevant to the recipient, and the cause and extent of any side effects. The amount of peptide administered to a subject (e.g., a mammal, such as a human) and the conditions of administration (e.g., timing of administration, route of administration, dosage regimen) are sufficient to effect the desired biological response over a reasonable time frame. Dosage typically depends upon a variety of factors, including the particular TFPI-inhibitory peptide employed, the age and body weight of the subject, as well as the existence and severity of any disease or disorder in the subject. The size of the dose also will be determined by the route, timing, and frequency of administration. Accordingly, the clinician may titer the dosage and modify the route of administration to obtain the optimal therapeutic effect, and conventional range-finding techniques are known to those of ordinary skill in the art. Purely by way of illustr ation, in one aspect, the method comprises administering, e.g., from about 0.1 pg/kg to about 100 mg/kg or more, depending on the factors mentioned above. In other embodiments, the dosage may range from 1 pg/kg up to about 75 mg/kg; or 5 pg/kg up to about 50 mg/kg; or 10 pg/kg up to about 20 mg/kg. In certain embodiments, the dose comprises about 0.5 mg/kg to about 20 mg/kg (e.g., about 1 mg/kg, 1.5 mg/kg, 2 mg/kg, 2,3 mg/kg, 2.5 mg/kg, 3 mg/kg, 3.5 mg/kg, 4 mg/kg, 4.5 mg/kg, mg/kg, 5.5 mg/kg, 6 mg/kg, 6.5 mg/kg, 7 mg/kg, 8 mg/kg, 9 mg/kg, or 10 mg/kg) of peptide. Given the chronic nature of many blood coagulation disorders, it is envisioned that a subject will receive the TFPI-inhibitory peptide over a treatment course lasting weeks, months, or years, and may require one or more doses daily or weekly. In other embodiments, the TFPI-inhibitory peptide is administered to treat an acute condition (e.g., bleeding caused by surgery or trauma, or factor inhibitor/autoimmune episodes in subjects receiving coagulation replacement therapy) for a relatively short treatment period, e.g., one to 34 days, [0094] Suitable methods of administering a physiologically-acceptable composition, such as a pharmaceutical composition comprising a peptide described herein, are well known in the art. Although more than one route can be used to administer a peptide, a particular route can provide a more immediate and more effective reaction than another route. Depending on the circumstances, a pharmaceutical composition is applied or instilled into body cavities, absorbed through the skin or mucous membranes, ingested, inhaled, and/or introduced into circulation. In one aspect, a composition comprising a TFPI-inhibitory peptide is administered intravenously, intraarterially, or intraperitoneally to introduce the peptide of the
2017203999 14 Jun 2017 invention into circulation. Non-intravenous administration also is appropriate, particularly with respect to low molecular weight therapeutics. In certain circumstances, it is desirable to deliver a pharmaceutical composition comprising the TFPI-inhibitory peptide orally, topically, sublingually, vaginally, rectally; through injection by intracerebral (intraparenchymal), intracerebroventricular, intramuscular, intra-ocular, intraportal, intralesional, intramedullary, intrathecal, intraventricular, transdermal, subcutaneous, intranasal, urethral, or enteral means; by sustained release systems; or by implantation devices. If desired, the TFPI-inhibitory peptide is administered regionally via intraarterial or intravenous administration feeding a region of interest, e.g,, via the femoral artery for delivery to the leg. In one embodiment, the peptide is incorporated into a microparticle as described in, for example, U.S. Patents 5,439,686 and 5,498,421, and U.S. Patent Publications 2003/0059474, 2003/0064033, 2004/0043077, 2005/0048127, 2005/0170005, 2005/0142205, 2005/142201, 2005/0233945, 2005/0147689. 2005/0142206, 2006/0024379, 2006/0260777, 2007/0207210, 2007/0092452, 2007/0281031, and 2008/0026068. Alternatively, the composition is administered via implantation of a membrane, sponge, or another appropriate material on to which the desired molecule has been absorbed or encapsulated. Where an implantation device is used, the device in one aspect is implanted into any suitable tissue, and delivery of the desired molecule is in various aspects via diffusion, timed-release bolus, or continuous administration. In other aspects, the TFPI- inhibitory peptide is administered directly to exposed tissue during surgical procedures or treatment of injury, or is administered via transfusion of blood procedures. Therapeutic delivery approaches are well known to the skilled artisan, some of which are further described, for example, in U.S. Patent No. 5,399,363.
[0095] To facilitate administration, the TFPI-binding peptide (e.g., TFPI-inhibitory peptide) in one embodiment is formulated into a physiologically-acceptable composition comprising a carrier (i.e., vehicle, adjuvant, buffer, or diluent). The particular carrier employed is limited only by chemico-physical considerations, such as solubility and lack of reactivity with the peptide, and by the route of administration. Physiologically-acceptable earners are well known in the art. Illustrative pharmaceutical forms suitable for injectable use include without limitation sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (for example, see
U.S. Patent No. 5,466,468). Injectable formulations are further described in, e.g.. Pharmaceutics and Pharmacy Practice, J. B. Lippincott Co., Philadelphia. Pa., Banker and Chalmers, eds., pages 238-250 (1982), and ASHP Handbook on Injectable Drugs, Toissel,
2017203999 14 Jun 2017
4th ed,, pages 622-630 (1986)). A pharmaceutical composition comprising a peptide provided herein is optionally placed within containers, along with packaging material that provides instructions regarding the use of such pharmaceutical compositions. Generally, such instructions include a tangible expression describing the reagent concentration, as well as, in certain embodiments, relative amounts of excipient ingredients or diluents that may be necessary to reconstitute the pharmaceutical composition.
[0096] When appropriate, the TFPI-binding peptide (e.g., TFPI-inhibitory peptide) of the invention is administered in combination with other substances and/or other therapeutic modalities to achieve an additional or augmented biological effect. Co-treatments include, but are not limited to, plasma-derived or recombinant coagulation factors, hemophilia prophylaxis treatments, immunosuppressants, plasma factor-inhibiting antibody antagonists (i.e., anti-inhibitors), antifibrinolytics, antibiotics, hormone therapy, anti-inflammatory agents (e.g., Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) or steroidal anti-inflammatory substances), procoagulants, and pain relievers. In one aspect, the method is an adjunct therapy to traditional replacement factor treatment regimens involving administration of, e.g., FXIII, FXII, FXI (e.g., HEMOLEVEN® (Laboratoire francais du Fractionnement et des Biotechnologies, Les Ulis, France) and FXI concentrate (BioProducts Laboratory, Elstree, Hertfordshire, UK)). FX, FIX (e.g., BENEFIX® Coagulation Factor IX (Wyeth, Madison,
NJ); ALPHANINE® SD (Grifols, Los Angeles, CA); MONONINE® (CSL Behring, King of Prussia, PA); BEBULIN-VH™ (Baxter, Deerfield, IL); PROFILNINE® SD (Grifols, Los Angeles, CA); or PROPLEX T™ (Baxter, Deerfield, IL)), FVIII (e.g., ADVATE™ (Baxter, Deerfield, IL); HELIXATE® FS (CSL Behring, King of Prussia, PA); REFACTO® (Wyeth, Madison, NJ), ΧΥΝΤΉΑ™ (Wyeth, Madison, NJ), KOGENATE® and KOGENATE® FS (Bayer, Pittsburgh, PA); ALPHANATE® (Grifols, Los Angeles, CA); HEMOPHIL M™ (Baxter, Deerfield, IL); KOATE®-DVI (Talecris Biotherapeutics-USA, Research Triangle Park, NC); or MONARC-M™ (Baxter, Deerfield, IL)), FVIIa (e.g., NOVOSEVEN® FVSIa (Novo Nordisk. Princeton, NJ) and FVI1 concentrate (Baxter Bioscience, Vienna, Austria, or BioProducts Laboratory, Elstree, Hertfordshire, UK)), FV, FVa, FII, and/or Fill, to a subject.
In some instances, the subject also receives FEIBA VH Immuno™ (Baxter BioScience, Vienna, Austria), which is a freeze-dried sterile human plasma fraction with Factor VIII inhibitor bypassing activity. FEIBA VH Immuno™ contains approximately equal units of Factor VIII inhibitor bypassing activity and Prothrombin Complex Factors (Factors II, VII,
IX, and X and protein C). Other exemplary co-treatments include, but are not limited to, prekallikrein, high molecular weight kininogen (HMWK), Von Willebrand’s factor, Tissue
2017203999 14 Jun 2017
Factor, and thrombin. Alternatively or in addition, the TFPI-inhibitory peptide is coformulated with one or more different TFPFinhibitory peptides.
[0097] The invention thus includes administering to a subject a TFPl-binding peptide (e.g., TFPI-inhibitory peptide) of the invention (or multiple TFPI-inhibitory peptides), in combination with one or more additionally suitable substances(s), each being administered according to a regimen suitable for that medicament. Administration strategies include concurrent administration (i.e,, substantially simultaneous administration) and non-concurrent administration (i.e., administration at different times, in any order, whether overlapping or not) of the TFPI-inhibitory peptide and one or more additionally suitable agents(s). It will be appreciated that different components are optionally administered in the same or in separate compositions, and by the same or different routes of administration.
[0098] In some embodiments, the peptide of the invention is conjugated to a moiety, e.g., a therapeutic or diagnostic moiety, such as the detection moieties and co-treatments described above. Alternatively or in addition, the peptide is administered in combination with an interaction partner (e.g., an antibody, antibody fragment, anticalin, aptamer, or spiegelmer) that (a) binds the peptide and (b) is therapeutically active and/or is linked to a moiety that provides additional functionality to the interaction partner (e.g,, a therapeutic, diagnostic, or detection agent). Suitable moieties include, but are not limited to, dyes, radionuclides, radionuclide-containing complexes, enzymes, toxins, antibodies, antibody fragments, and cytotoxic agents, and, in some instances, the moiety possesses therapeutic activity (i.e., achieves an advantageous or desired biological effect), The peptide conjugates or peptideinteraction partner pair is suitable for use in any of the methods described herein, such as methods of treating a subject suffering from a disease or disorder or at risk of suffering from a disease or disorder.
[0099] All publications, patents and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. In addition, the entire document is intended to be related as a unified disclosure, and it should be understood that all combinations of features described herein are contemplated, even if the combination of features are not found together in the same sentence, or paragraph, or section of this document. For example, where protein therapy is described, embodiments involving polynucleotide therapy (using polynucleotides/vectors that encode the protein) are specifically contemplated, and the reverse also is troe. Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of
2017203999 14 Jun 2017 understanding, it will be readily apparent to those of ordinary skill in the art in light of the teachings of this invention that certain changes and modifications may be made thereto without departing from the spirit or scope of the appended claims. The invention includes, for instance, all embodiments of the invention narrower in scope in any way than the variations specifically mentioned above. With respect to aspects of the invention described as a genus, all individual species are individually considered separate aspects of the invention. With respect to aspects of the invention described or claimed with “a” or “an,” it should be understood that these terms mean “one or more” unless context unambiguously requires a more restricted meaning. With respect to elements described as one or more within a set, it should be understood that all combinations within the set are contemplated.
EXAMPLES
Example 1 [00100] The following example describes production, identification, and screening of peptides for binding to TFPI.
[00101] Peptides candidates were obtained from commercial suppliers (e.g., PolyPeptide Laboratories SAS (Strasbourg, France) and JPT Peptide Technologies GmbH (Berlin, Germany)). Methods for synthesizing candidate peptides are provided above. Candidate peptides were synthesized as trifluoroacetate (TEA) salts with a purity >90% or >60%, All peptides were solved in DMSO to a stock concentration of 10 mM. TFPI-binding peptide sequences were identified using an mRNA display library. The rnRNA display technology is superior to other library screening techniques for allowing for a diversity of 1014 different sequences within a starting pool and avoiding, e.g,, the in vivo steps required for phage display. In brief, the technology involves directly linking mRNA to its encoded candidate peptide through a puromycin molecule (Figure 5). The mRNA display method is further described in International Patent Publication No. WO 2005/051985 and Liu et ah, Methods in Enzymology, 3J8, 268-293 (2000). TFPI was immobilized to a solid support via biotin and exposed to candidate peptide-RNA complexes. TFPI-bound candidate peptide-RNA complexes were isolated, and the RNA reverse transcribed to obtain coding DNA. High affinity binders were obtained following six to ten selection rounds using a competitive elusion strategy. Many of the candidate peptides were 31 amino acids in length (27 randomized amino acids and 2 amino acids flanking both termini).
2017203999 14 Jun 2017 [00102] Selected peptides were synthesized and subjected to peptide optimization using a micro array-based scan analysis to identify peptide fragments retaining TFPI-binding affinity. For example, a microarray-based scan of JBT0047 was performed using a series of 20 amino acid fragments of the peptide, the sequences of which overlapped by 19 amino acids. N- and C-terminal truncation analysis supplemented the scan analysis. The microarray scan results demonstrated that that peptide JBT0293 bound TFPI with the highest affinity. A series of substitution mutants based on the amino acid sequence of JBT0293 was generated and tested for TFPI binding properties.
[00103] The affinity of a subset of peptides for TFPI was demonstrated via an enzymelinked immunosorbent assay (ELISA)-like assay (binding (EC50) ELISA) performed with biotinylated peptides. Ninety-six well MaxiSorp plates (Nunc) were coated with 3 pg/mL TFPI in coating buffer (15 mM NajCOs, 35 mM NaHCCE. pH 9.6) over night. Plates were washed three times with 350 pi wash buffer (HNaT: 175 mM NaCl, 25 mM HEPES, 5 mM CaCE, 0.1% Tween 80, pH 7.35), and subsequently blocked with 200 pi 2% yeast extract in HNaT for 2 hours. Plates were then washed three times with 350 pi HNaT. Biotinylated candidate peptides were diluted from a DMSO stock 1/200 in HNaT. The initial peptide concentration was 50 pM if no precipitate appeared during the 1/200 dilution of the 10 mM peptide stock solution. Pre-dilutions of the peptide stock in DMSO were conducted if precipitates formed. The diluted peptides were applied to the Maxisorp plates, serial dilutions (1/3) were generated, and the dilutions were incubated for 1.5 hours at room temperature. Incubation was followed by three wash steps (350 pi HNaT). Bound peptide was detected by incubation with horseradish peroxidase-conjugated streptavidin (1 hour), followed by three wash steps with HNaT and a subsequent chromogenic conversion of added TMB (3,3’5,5'-Tetramethy]benzidin). The assay is illustrated in Figure 6A.
[00104] Generally, peptide binding to immobilized TFPI was significantly above background. EC50 values for biotinylated peptides are given in Figures 32-39. The binding curve of one TFPI-binding peptide, JBT0132, is depicted in Figure 7. The EC50 of JBT0132 was calculated to be about 2.2 nM.
[00105] In addition, a competition (IC50) ELISA was performed using biotinylated TFPIbinding peptides as “tracers” to compete for TFPI-binding with non-biotinvlated candidate peptides. The assay principle is depicted in Figure 6B. Ninety-six well MaxiSorp plates (Nunc) were coated with 3 pg/mL TFPI in coating buffer (15 mM Na^CCE, 35 mM NaHCCE, pH 9,6) over night. Plates were washed three times with 350 pi wash buffer (HNaT: 175 mM NaCl, 25 mM HEPES, 5 mM CaCE, 0.1% Tween 80, pH 7.35), and blocked with 200 pi 2%
2017203999 14 Jun 2017 yeast extract in HNaT for 2 hours. Plates were then washed three times with 350 μΐ HNaT. Biotinylated tracer peptides were applied at a concentration corresponding to their respective EC90 values determined in the binding ELISA (median if n > 2). A competitor stock solution of peptide (10 mM) was diluted 1/33.3 in HNaT without HSA, and a serial 1/3 dilution was prepared with HNaT with 3% DMSO. The dilution was further diluted with the biotinylated tracer peptide in a ratio of 1:6 (20 pi competitor dilution and 100 pi tracer peptide). The mixture of competitor and tracer peptide was applied to the TFPl-coated microtiter plate and incubated for 1.5 hours. The plates were washed three times with 350 μΐ HNaT. PeptideTFPI binding was detected by applying HRP-conjugated streptavidin to the microtiter plate, incubating the mixture for one hour, washing the plate three times with 350 μΐ HNaT, applying TMB (3,3'5,5’-Tetramethylbenzidin), and detecting the subsequent chromogenic conversion of TMB by HRP. IC50 graphs for representative non-biotinylated peptides are provided in Figures 8A-8D. ICjo measurements of peptides JBT0303, JBT0120, and JBT0224 are set forth in Table 3.
TABLE 3
Peptide IC5o [pM] n SD Tracer Peptide Tracer Concentration fpMI
JBT0303 0.119 2 0.064 JBT0131 0.0409
JBT0120 0.0189 3 0.0044 JBT0124 0.0718
JBT0224 n. a. 1 JBT0126 0.240
[00106] In addition to the competition ELISA (1C5O) assay, a screening assay was employed to measure higher numbers of peptides in parallel. The screening ELISA is similar to the competition IC50 ELISA with the exception that only three different concentrations of the competitor were employed (300 nM, 100 nM and 33.3 nM for the JBT0047 class, and 50000 nM, 16667 nM and 5556 nM for the JBT0122 class). In some instances, screening results were expressed as percent inhibition of the tracer signal in relation to a competitive peptide (competitive peptide JBT0477 for the JBT0047 family, and competitive peptide JBT1697 for the JBT0122 family). The competition ICA, assay results and the screening assay results of peptides prepared and screened in accordance with the methods set forth herein are provided in Figures 32-39. The mean IC50 values presented in Figures 32-39 are based on a greater number of assays than die values presented in Table 3 and, therefore, the values may differ slightly. The results of the screening ELISA are presented as percent inhibition of tracer peptide JBT0131 binding. Several peptides that were analyzed using the
2017203999 14 Jun 2017
IC5o ELISA are classified in Figures 32-39 according to their binding affinity as set forth in Table 4.
TABLE 4
TFPI competition ELISA IC50 [nM] Group
<50 nM A
50 < x <100 nM B
100<x<250nM C
250 < x < 1000 nM D
1000 < x < 5000 nM E
5000 < x < 10000 nM F
10000 < x < 50000 nM G
[00107] Exemplary TFPI-binding peptides identified using the methods described herein are presented in Table 5. Some peptides were biotinylated, and many comprise N- and Cterminal lysines to promote solubility. Several peptides exhibited TFPl-inhibitorv activity in model and/or plasmatic assay systems, as described below.
TABLE 5
Peptide | Parent Sequence SEQ 1I>
JBT0047 QSKKN VFVFG YFERLRAK 1
JB’IO047 JBT0047 Ac-SGVGRLQVAFQSKKN VFVFG YFERLRAKLTS-NH2 253
JBT0051 JBT0047 Biotinyl-Ttds-SGVGRLQVAFQSKKNVF\'FGYEERLRAKLTS-NH2 962
JBT0055 JBT0047 Ac-SGVGRLQVAFQSKKNVFVFGYFERLRAKLTS-Ttds-Lys(Biolinyl)- NII2 963
JBT0131 JBT0047 Biotinyl-Ttds-AFQSKKNVFVFGYFERLRAK-NH2 964
JBT0132 JBT0047 Biotinyl-Ttds-FQSKKNVFVFGYFERLRAKL-NH2 965
JBT0133 JBT0047 Biolinyl-Tlds-QSKKNVFVFGYFERLRAKLT-NH2 966
JBTO155 JBT0047 Ac-KKSGVGRIJQVAFQSKKNVIiVFGYFERLRAKJ,TSKK-NH2 8
JBTO158 JBT0047 Ac~KKSGVGRLQVAFQSKKNVFVFGYFERLRARKK-N,H2 9
JBT0162 JBT0047 Ac-KKGRLQVAFQSKKNVFVFGYFERLRAKLTSKK-NH2 10
JBT0163 JBT0047 Ac-KKQVAFQSKKN VFVFG YFERLRAK1.TSKK-NH2 11
JBT0164 JBT0047 Ac-KKFQSKKNVFVFGYFERLRAKLTSKK-NH2 12
JBT0166 JBT0047 Biotiny]-Ttds-KKFQSKKNVFVFGYFERLRAKLKK-NH2 968
JBT0169 JBT0047 Ac-KKAFQSKKNVFVFGYFERLRAKKK-NH2 254
JBT0170 JBT0047 Ac-KKFQSKKN VFVFG YFTERLRAKI.KK-N'H2 13
JBT017I JB7O047 Ac-KKQSKKNVFVFGYFERIJRAKLTKK-NH2 255
JBT0174 JBT0047 AC-KKAFQSKKNVFVFGYFERLRAKLKK-NH2 34
JBT0175 JBT0047 Ac-KKAFQSKKN VFVFG YFERLRAKLTKK-NJI2 182
JBT0293 JBT0047 Ac-FQSKKNVFVFGYFERLRAKL-NH2 256
X,X4X;KX7XVFXJ1X12GYX,3X,6RLRAKX22 2
JBT0294 JBT0047 Ac-YQSKKNVFVFGYFERLRAKL-NH2 257
JBT0295 JBT0047 AC-FSSKKNVFVFGYFERLRAKL-NH2 713
JBT0296 JBT0047 AC-FQNKKNVFVFGYFERLRAKL-NH2 407
JBT0297 JBT0047 Ac-FQSKNXVFVFGYFERLRAKL-NI-12 183
2017203999 14 Jun 2017
JBT0298 JBTOO47 Ac-FQSKQNVFVFGYH3RLRAKL-NH2 747
JBT0299 JBT0047 Ac-FQSKKN VFAFGYFERLRAKL-ΝΊΙ2 408
ΪΒΊΌ300 JBT0047 Ac-FQSKKN VFSFGYEERLRAKL-NH2 409
JBT0301 JBT0047 Ac-FQSKKNVFrFGYIFRI.RAKL-NH2 470
JBT03O2 JBT0047 Ac-FQSKKN VFVAGYFERJ.RAKL-NH2 258
JBT0303 JBT0047 AC-FQSKKNVFVDGYFERLRAKL-NH2 184
JBT0304 JBT0047 Ac-FQSKKN VFVLGYFERLRAKL-NH2 259
JBT0305 JBT0047 AC-FQSKKNVFVQGYFERLRAKI.-NH2 260
JBTO3O6 JBT0047 Ac-FQSKKNVFVSGYFERLRAKL-NH2 185
JBTO3O7 JBT0047 Ac-FQSKKNWVYGYFERI.RAKI.-NH2 261
JBT0308 JBT0047 Ac-FQSKKNVFVFGYKF.RLRAKI.-NH2 411
JBT0309 JBT0047 Ac-FQSKKN VFVFGYYERLRAKL-NH2 412
JBT03I0 JBT0047 Ac-FQSKKN VFVFGYFDRLRAKL-NH2 262
JBT03U JBT0047 Ac-FQSKKN VI· VFGYFERLR AKN -N112 748
ri\oeri.iyiij ignmgmyaaqlki 3
JBT0049 JBT0049 Ac-SGNTFVDERLLYFLT1GNMGMYAAQI.KFRTS-NH2 3025
JBT0053 JBT0049 BiotinyJ-Tlds-SGNTFVDERI±,YFLTIGNMGMYAAQLKFRTS-NH2 3006
JBT0057 JBT0049 Ac-SGNTFVDERLLYFLTJGNMGMYAAQLKFRTS-Ttds-Lysjn(biodn)- NH2 3018
JBT0190 JBT0049 Ac-KKSGNTFVDERIiYFLTJGNMGMYAAQLKFRTSKK-NH2 3031
JBT0193 JBT0049 AC-KKSGNTFVDERLLYFLTIGNMGMYAAQLKFKK-NH2 3073
JBT0197 JBT0O49 AC-KKTFVDERLLYFLT1GNMGMYAAQLKFRTSKK-NH2 3076
VIVF1FRHNKIJGYERRY 4
JBT005O JBT0O5O Ac-SGRGCTKVIVFTFRHNKL1GYERRYNCTS-NH2 3047
JBTO054 JBT0050 Biotinyl-Ttds-SGRC5CTKVIVFTFRHNKLIGYERRYNCTS-NH2 3002
JBT0058 JBTOO50 Ac-SGRGCTKVIVFTFRHNKL1GYERRYNCTS-I'1ds-Lvsin(biotin)-NH2 3003
JBT0129 JBT0O50 Ac-SGRGlCi'KVJVi’’j'FRHNKLJCiYERRYNGJ'J'S-NH2 3026
JBTO13O JBT0050 Biotjnyl-Ttds-SGRG|CTKVlVFlT'RHNKLlGYJ;RRYNCrJ'S-NH2 3001
JBT0205 JBT0050 Ac-KKSGRGCTKVlVFrFRHNKLJGYERRYNCr$KK-NH2 3029
JB’J'0208 JBT0050 Ac-kk,SGRGClKVIVFrFRHNKLIGYERRYNKK-NJI2 3027
JBT0211 JBT0050 Ac-KKGCTK VIVFIFRI INK L1G Y ERR YNCTSKK-N112 3032
JBT0212 JBT0050 Ac-KKKVIVF]HUINKLIGYERRYNCTSKK-NH2 3033
JBTO2I7 JBT0050 Ac-KK'i'KVI VFi'FRI INKIJGYERR YKK-N112 3062
JBTO2J8 JBT0050 Ac-KKKVTVFTFRHNKIJGYERRYNKK-NH2 3063
JBT0219 JBT0050 AC-KKVIVFTFRHNKLIGYERRYNCKK-NH2 3030
GVWQTIIPRYFWTMWPDIKGEVIVTFGT 5
JBTOIOI JBTO10! Ac-KKSGVWQTHPRYFWTMWPOTKGFV1VEFGTSKK~NH2 3036
ΙΒΊΌ052 JBTOIOI Biot!r>yI-Ttds-KKSGVWQTHPRYFWTMWPDIKGEVTVEFGTSKK-NH2 3004
JBT0W3 JBTOIOI Ac-KKSGVWQTHPRYFWTMWPDIKGEVrVEFGTS-Ttds-KK- Lysin(biotiny])-NH2 3005
JBT0178 JBTOIOI Ac-KKSGVWQTHPRYFWTMWPDIKGEViVLFGTKR-NH2 3028
JBT0182 JBTOIOI Ac-KKGVWQTHPRYFWTMWPDIKGEVIVLFGTSKK-NH2 3037
KWCGMRD.MKGTMSCVWVKF 6
JBT0120 JBT0120 Ac-SGASRYKWF|CGMRDMKGTMSC]VWVKFRYDTS-NH2 1047
JBT0124 Biotinyl-Ttds-SGASRYKWF{CGMRDMKGTMSC]VWVKFRYDTS-NH2 1290
JBT0247 JBT0120 Ac-SGASRYKWFCGMRDMKGT’MSCVWVKFRYDTS-NH2 1213
JBT0248 JBT0120 Ac-KKSGASRYKWF[CGMRDMKGTMSC]VWVKFRYDTSKK-NH2 1001
JBT025I JBT0I2O Ac-KKKWFCGMRDMKGTMSCVWVKFKK-NH2 1202
JBT0252 JBT0120 Ac-KKCGMRDMKGTMSCVWVKFRYDKK-NH2 1215
ASFPLAVQLHVSKRSKEMA 7
JBTOI22 JBT0122 Ac-StiYASl PJ AVQI.I1VSKRSK1:.V1AI .'\Ri.YYK lS-NH2 2002
JBT0126 JBT0322 Bioiinyi-fids-SGYASH’LAVQLHVSKRSKEMALARLYYKTS-NH2 2498
JBT0221 JBT0122 Ac-KKSGYASFPLAVQLHVSKRSKEMALARLYYKTSKK-NH2 2003
JBT0224 JBT0122 Ac-KKSGYASFPLAVQLHVSKRSKEMALARLYYKK-NH2 2298
JBT0225 JBT0122 Ac-KKSGYASFPLAVQLHVSKRSKEMALARKK-NH2 2128
JBT0226 JBT0122 Ac-KKSGYASFPLAVQLHVSKRSKEMAKK-NH2 2299
JBTO228 JBT0122 AC-KKASFPLAVQLHVSKRSKEMALARLYYKTSKK-NH2 2016
JBT0232 JBT0122 Ac-KKGYASFPLAVQLHVSKRSKEMKK-NI-12 2303
JBT0233 JBT0122 AC-KKYASFPLAVQLHVSKRSKEMAKK-NH2 2304
2017203999 14 Jun 2017 [00108] This example provides exemplary methods of generating and characterizing TFPI inhibitory peptides. All peptides in Table 5 were found to bind human TFPI-la. Mutation analysis demonstrated that at least one amino acid in a TFPI-binding peptide may be substituted while retaining affinity for TFPI. The peptides of Table 5 tested in ELISA assays bound TFPI-la with an EC50 of less than 10 μΜ (1 x 10‘5 M) and an IC50 of less than 50 μΜ
Example 2 [00109] Selected TFPI-binding peptides were further characterized in terms of “antitarget” binding. This example demonstrates that TFPl-inhibitory peptides exhibit reduced affinity for non-TFPI-1 proteins, [00110] TFPI-2 was selected as an anti-target because of its similarity to TFP1-L The binding kinetics of TFPI-binding peptides to human TFPI-1 (residues 29-282 fused at theCterminus to a 10 His-tag; MW 41 kDa (R&D Systems, Minneapolis, MN; catalog number 2974-PI)) murine TFPI-1 (residues 29-289 fused at the C-terminus to a 10 His-tag; MW 41 kDa (R&D Systems; catalogue number 2975-PI)), and TFPI-2 (R&D Systems, Minneapolis, MN) were studied using a BIAcore 3000™ surface plasmon resonance assay (GE Healthcare, Chalfont St. Giles, UK). TFPI proteins were immobilized on a Cl chip (GE Healthcare, Order Code: BR-1005-40) by amine coupling chemistry aiming for 500 RU. Several TFPI-binding peptides were employed as analytes for interacting with the immobilized TFPI proteins. A flow rate of 30 μΐ/min was utilized. After 180 seconds, 180 μΐ of peptide solution was injected at six different concentrations ranging from 3.84 nM to 656.25 11M, followed by a dissociation time of 480 seconds. The chip was regenerated with 45 μΐ 10 mM NaOH. Each binding experiment was preceded and followed by four measurements with HBS-P buffer (10 mM HEPES, pH 7.4, 150 mM NaCl, 0.005% P20) plus 1% DMSO and 0.8% P80. BlAevaluation® Version 4.1 software (GE Healthcare) was employed to analyze the data. Sensorgrams were fitted to a 1:1 Langmuir binding curve to determine kO[1 and koff and calculate [00111] Certain tested peptides, e.g., JBT00S0, JBT0121, JBT0205 and JBT0211. bound to the blank cell and binding constants from those sensorgrams could not be determined. JBT0133 showed weak binding to TFPI-L Sensorgrams from other peptides gave reliable binding constants. Results from BIAcore analysis of several TFPl-inhibitory peptides is
2017203999 14 Jun 2017 provided in Table 6 and Figures 19-21. Each of the peptides listed in Table 6 presented a KD of less than 10 μΜ. Sensorgrams of two of the peptides are provided as Figures 9A and 9B,
TABLE 6
Peptide kUl/Ms) Liril/s) K»(M)
JBT0047 4.0 x 105 1.9 x IO 4.7 x 10's
JBT0120 3;Ϊ7Χΐθ6 4.78 x 10' 4.08 x 10's
JBT0131 .....ϊ.4χ ϊο* 6.0 x 10'- 4.31 x 10'y
JBT0132 3.55 x 104 3.26 x 10'J 9.17x 10'7
JBT0224 6.39 x 104 1,95 x JO'2 ΓΤθ5χΊ0'7
JBT0293 6.0 x 105 5,6 x IO'4 9.5 x 10‘K
JBT0297 5.0 x 101 1.4 x IO' 2.9 x 10'*
JBT0303 8.13 x JO5 2.75 x 10 3Axl(P
JBT0305 7.5 x 105 3.1 x JO'2 6.1 x 10'*
[00112] Interaction with the TFPI-2 anti-target also was examined. The maximum signal generated from candidate peptide interaction with human TFPI-2 was much lower than the signals obtained with TFPI-1 as an interaction partner. Kinetic analysis of the low TFPI-2 binding signals was prone to error; therefore, visual comparison of sensorgrams was used to estimate binding affinity. A sensorgram illustrating JBT0120 binding to TFP1-1 and TFPI-2 is provided as Figures 10A and 10B. JBT0120 binds TFPI-2 with 10-fold lower affinity compared to its binding affinity for TFPM. JBT0132 also was found to exhibit at least 10fold greater affinity for TFPI-l than TFPI-2.
[00113] The data provided by this example confirm that TFPI-inhibitory peptides specifically bind TFPI-1.
Example 3 [00114] The following example describes the characterization of TFPI-inhibitory activity of select peptides identified in Example 1 using a FXa inhibition and extrinsic tenase inhibition assay. Both assays are predictive of activity in plasmatic systems. The extrinsic tenase assay gives insight into the influence of the peptides on (a) the interaction of FXa and TFPI and (b) the interaction of the FXa-TFPl complex with the TF-FVDa complex. The FXa inhibition assay measures a peptide’s influence on (he interaction of FXa and TFPI only.
[00115] The extrinsic tenase complex is responsible for FX and FIX activation upon initiation of the coagulation process. The extrinsic complex is composed of FVlIa, Tissue Factor (TF), and FX substrate. To determine the influence of peptides on the TFPl-mediated
2017203999 14 Jun2017 inhibition of the extrinsic tenase complex, a coupled enzyme assay was established. Peptides were diluted 1/6.25 from 10 mM stocks (in DMSO) and further diluted by serial 1/4 dilutions in DMSO to prevent unwanted precipitation. TFPI was diluted in HNaCa-HSA (25mM HEPES; 175mM NaCl; 5mM CaCE; 0,1% HSA; pH 7.35). FVIIa, lipidated TF, phospholipid vesicles (DOPC/ POPS 80/20). and chromogenic substrate specific for FXa (S2222 (available from DiaPharma, West Chester. OH)), all diluted in HNaCa-HSA, were added to 96-well plates. After an incubation period, TFPI and peptide dilutions were added, resulting in a final concentration of 2.5% DMSO. FX activation was initiated by adding FX to the wells. FXa-mediated chromogenic substrate conversion was determined by observing an increase in absorbance using a micro-plate reader. The amount of FXa generated at certain time points was calculated from the OD readings. FXa generated at 20 minutes after start of the reaction was considered for calculation of EC50 from plots of peptide concentration versus the inhibition of TFPI (%).
[00116] The functional inhibition of TFPI also was examined using a FXa inhibition assay. A FXa-specific chromogenic .substrate (S-2222) and TFPI, both diluted in HNaCa-HSA, were added to 96 well plates. Peptides were diluted 1/6.25 from 10 mM stocks (in DMSO) and further diluted by serial 1/4 dilutions in DMSO to prevent unwanted precipitation. The peptide dilutions (2.5 μΐ) were added to the 96 well plates, resulting in a final concentration of 2.5% DMSO. The conversion of chromogenic substrate was triggered by the addition of FXa, and the kinetics of the conversion were measured in a micro-plate reader. Because TFPI inhibits FXa slowly, OD readings after 115 minutes were considered for calculation of the EC50 from plots of peptide concentration versus the inhibition of TFPI (%).
[00117] Results from the extrinsic tenase assay and FXa inhibition assay are provided in Table 7 and Figures 22-27.
TABLE 7
FXa Inhibition Assay Extrinsic Tenase Assay
ECsHpM] % inhibition @ 2,5 pM ECS0 [pMJ % inhibition @ 2.5 pM
JBT0J20 0.9 45 0.9 45
JBT0132 1.2 36 0.1 10
JBT0224 n.a. 26 3.5 18
JBT0303 1.2 61 n. a. 8
[00118] Referring to Table 7, JBT0120, JBT0132, and JBT0224 restored extrinsic complex-mediated FX activation in the presence of TFPM with an EC50 of < 2 pM, resulting in between about 20% to about 60% inhibition of TFPI activity. JBT0047 (EC5o=E4 pM),
2017203999 14 Jun 2017
JBTO131 (EC5o=2.2 μΜ), and JBT0293 (EC50=2.9 μΜ) also restored extrinsic complex activity in the presence of TFP1-1. In addition, JBTO32O, JBT0132, JBT0224, and JBT0303 restored FXa activity in the presence of TFPI-1 with an EC50 of < 5 μΜ, resulting in between about 5% to about 50% inhibition of TFPJ activity, in the FXa inhibition assay, JBT0047 (EC50=0.7 μΜ), JBT0131 (EC5O=8.2 μΜ), JBT0293 (EC5o=1.3 μΜ), JBT0297 (EC,0=0.6 μΜ), and JBT0305 (ECso=2.3 μΜ) also restored activity of FXa in the presence of TFPI-1 in the FXa inhibition assay. This example confirms that peptides of the invention are TFPI antagonists.
Example 4 [00119] In this example, the TFPI inhibitory activity of peptides is established using a plasma-based assay.
[00120] The influence of peptides on thrombin generation was measured in duplicate via calibrated automated thrombography in a Fluoroskan Ascent® reader (Thermo Labsystems, Helsinki, Finland; filters 390 nm excitation and 460 nm emission) following the slow cleavage of the thrombin-specific fluorogenic substrate Z-G3y-Gly-Arg-AMC (Hemker, Pathophysiol. Haemost. Thromb., 33, 4-15 (2003)). Plasma from patients with FVIII or FIX deficiency (George King Bio-Medical Inc., Overland Park, KN) was obtained for testing.
The residual coagulation factor acti vity for each of the plasmas was lower than 1%. As a model for antibody-mediated FVIII deficiency, frozen pooled normal plasma (George King Bio-Medical Inc., Overland Park, KN) was incubated with high titer, heat inactivated, antihuman FVIII plasma raised in goat (4490 BU/ml; Baxter BioScience, Vienna, Austria) giving rise to 50 BU/rnL. The plasmas were mixed with com trypsin inhibitor (CTI) (Hematologic Technologies, Inc., Essex Junction, VT) to inhibit Factor Xlla contamination, resulting in a final concentration of 40 pg/mL.
[00121] Pre-wanned (37°C) plasma (80 p.L) was added to each well of a 96 well microplate (Immulon 2HB, clear U-bottom; Thermo Electron, Waltham, M A). To trigger thrombin generation by Tissue Factor, 10 μ L of PPP low reagent containing low amounts (12 pM) of recombinant human Tissue Factor and phospholipid vesicles composed of phosphatidylserine, phosphatidylcholine and phosphatidyl ethanol amine (48 μΜ) (Thrombinoscope BV, Maastricht, The Netherlands) were added. Peptides were diluted 1/7.5 from 10 mM stocks with DMSO, and further diluted 1/8,33 with Aqua-Dest resulting in a DMSO concentration of 12%. providing a 0.5% DMSO concentration in the final assay mix.
2017203999 14 Jun 2017
Just prior putting the plate into the pre-warmed (37°C) reader, 5 μ L of HEPES buffered saline with 5 mg/mL human serum albumin (Sigma-Aldrich Corporation, St. Louis, Missouri, USA) or 12% DMSO in Aqua-Dest was added, followed by addition of the peptide dilutions or reference proteins (FVIII Immunate reference standard (Baxter BioScience, Vienna, Austria); Factor V1IJ Inhibitor By-Passing Activity (FEIBA) reference standard (Baxter BioScience, Vienna, Austria); NovoSeven (Novo Nordisk, Denmark); and purified human plasma FIX (Enzyme Research Laboratories, South Bend, IL)). Thrombin generation was initiated by dispensing into each well 20 μ L of FluCa reagent (Thrombinoscope BV, Maastricht, The Netherlands) containing a fluorogenic substrate and HEPES-buffered CaCl2 (100 mM). Fluorescence intensity was recorded at 37°C.
[00122] The parameters of the resulting thrombin generation curves were calculated using Thrombinoscope™ software (Thrombinoscope BV, Maastricht, The Netherlands) and thrombin calibrator to correct for inner filter and substrate consumption effects (Hemker, Pathophysiol. Haemost. Thromb.. 33, 4-15 (2003)). For calculating the thrombin generating activity of certain peptide concentrations equivalent to the reference proteins (e.g., FVIII Immunate® reference standard, FEIBA reference standard), the thrombin amounts at the peak of each thrombin generation curve (peak thrombin, nM) were plotted against the standard concentrations, and fitted by a non-linear algorithm. Based on this calibration, FVIII Immunate, FIX, FEIBA or NovoSeven equivalent activities were calculated. Results for various peptides are provided in Figures 12-18 and 28-30. Representative results are provided in Table 8. (* denotes that FVIII deficient plasma was obtained from a different donor.)
TABLE 8
% FVIII-equivalent activity in FVIII deficient plasma @ 10 μΜ peptide FEIBA-equivalent activity in FVIII inhibited plasma @ 10 μΜ peptide [mU/ml]
JBT0120 37.4* 298
JBT0I32 5.3 4.1
JBT0224 16.2 191
JBT0303 20.8 253
[00123] With reference to Table 8, JBT0120, JBT0132, JBT0224, and JBT0303 improved TFPI-dependent thrombin generation in FVlIJ-depleted plasma to levels exceeding 1% of the level of thrombin generation in plasma containing FVIII (% FVIII-equivalent activity). The tested peptides exhibited approximately 5%-40% FVIII-equivalent activity in FVlIl-deficient
2017203999 14 Jun 2017 plasma. JBT0120 and JBT0132 improved peak thrombin and peak time, dose dependently, as illustrated in Figures 31A and 1 IB.
Example 5 [00124] The following example demonstrates that the peptides of the invention can be modified by the addition of moieties that enhance physicochemical or pharmacokinetic properties of the peptides. As illustrated below, the addition of 40 kDa PEG to peptides described herein dramatically improved the pharmacokinetic behavior of the peptides.
[00125] Methods of conjugating chemical or biological moieties to peptides are known in the art. To add PEG (polyethylene giycoi) to the peptides describe herein, a functional group (AOA = aminooxy acetate) was added to the N-terminus of the peptides for coupling to aldehydes and ketones. Alternatively, a cysteine was added to the C-terminal part of the peptide for coupling with maleimid (Hermanson, Bioconjugaie Techniques, Academic Press (1996)). The peptides (JBT1586) AOA-FQSKGNVFVDGYFERL-Aib-AKL-NH2 (SEQ ID NO: 166) and (JBT1587) Ac-FQSKGNVFVDGYFERL-Aib-AKLC-NH2 (SEQ ID NO: 167) were used for N-terminal and C-terminai modification with PEG, respectively. AOAFQSKGNVFVDGYFERL-Aib-AKL-NH2 (SEQ ID NO: 166) and AcFQSKGNVFVDGYFERL-Aib-AKLC-NH2 (SEQ ID NO: 167) were incubated with excess 40 kDa mPEG-Propionaldehyde (SUNBRIGHT ME-400AL2, NOF, Japan) and 40 kDa mPEG-maieimide (SUNBRIGHT ME-400MA, NOF, Japan), respectively. The resulting PEGylated peptides, JBT1S53 and JBT1855, show similar affinities compared to the starting structure Ac-FQSKGNVFVDGYFERL-Aib-AKL-NH2 (JBT0740) (SEQ ID NO: 66).
[00126] The resulting PEGylated peptides demonstrated significantly increased plasma stability and prolonged plasma half-life in mice. Figure 31 illustrates the results from a pharmacokinetic analysis of the free peptide JBT0740 (Ac-FQSKGNVFVDGYFERL-AibAKL-NH2) (SEQ ID NO: 66) compared to the C-terminally PEGylated peptide JBT1855 (Ac-FQSKGNVFVDGYFERL-Aib-AKLC(PEG(40kD))-NH2) (SEQ ID NO: 252). In contrast to the unPEGyiated peptide, the PEGylated peptide is present at high concentrations in mouse plasma at 100 minutes post-administration. The unPEGyiated peptide is rapidly cleared from the plasma.

Claims (36)

  1. THE CLAIMS DEFINING THE INVENTION ARE AS FOLLQWS:1. A peptide that binds TFPI, comprising an amino acid sequence of at feast 60 % identity
    Ac-GYASFPWFVQLHVHKRSWEMA-NH2 (X) (SEQ ID NO: 2023).
  2. 2. The peptide according to claim 1, wherein the amino acid sequence identity is at least 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100%.
  3. 3. A peptide consisting of the amino acid sequence selected from the group consisting of SEQ ID NOs: 2001-2126, 2128-2296, and 2298-2498.
  4. 4. The peptide according to any one of claims 1 to 3, wherein said peptide inhibits TFPI activity, whereby preferably the peptide binds to TFP11-alpha with a dissociation constant of less than 10 μΜ.
  5. 5. The peptide according to any one of claims 1 to 4, wherein the IC50 of the peptide is less than 50,000 nM, preferably less than 10,000 nM and more preferably less than 5,000 nM.
  6. 6. The peptide according to any one of claims 1 to 5, wherein the peptide is conjugated to a polyethylene glycol (PEG) moiety.
  7. 7. A peptide according to any one of claims 1 to 6 for use in a method for the treatment of a subject.
  8. 8. The peptide according to claim 7, wherein the method is for the treatment of a blood coagulation disorder.
  9. 9. Use of a peptide according to any one of claims 1 to 8 for the manufacture of a medicament.
    07/06/19
    2017203999 07 Jun 2019
  10. 10. Use of a peptide according to any one of claims 1 to 6 for the manufacture of a medicament for the treatment of a blood coagulation disorder.
  11. 11. A method for treating a blood coagulation disorder in a subject, the method comprising administering to the subject a peptide of any one of claims 1 to 10, wherein the peptide is administered in an amount sufficient to treat the blood coagulation disorder in the subject.
  12. 12. The method according to claim 11, wherein the subject is Factor VIII or Factor IX deficient.
  13. 13. The method according to claim 11 or 12, wherein the peptide binds to TFPI 1 alpha with a dissociation constant of less than 10 pm.
  14. 14. The method according to any one of claims 11 to 13, wherein the peptide is linked to an effector moiety.
  15. 15. A pharmaceutical composition comprising a peptide as defined in any one of claims 1 to 6 and a pharmaceutical acceptable carrier.
  16. 16. The pharmaceutical composition according to claim 15, wherein the composition comprises a further pharmaceutically effective agent.
  17. 17. The pharmaceutical composition according to claim 15 or 16, wherein the pharmaceutical composition is for use in a method as defined in any one of claims 11 to 14.
  18. 18. A method for targeting a cell displaying TFPI, wherein the method comprises contacting the ceil with a peptide as defined in any one of claims 1 to 6, with the proviso that, for a human celt, the method is performed ex vivo.
    07/06/19
    2017203999 07 Jun 2019
  19. 19. The method according to claim 18, wherein the method further comprises detecting peptide binding to TFPI displayed on the cell.
  20. 20. The method according to claim 18 or 19, wherein the peptide is conjugated to a moiety, wherein such moiety is preferably selected from the group consisting of a dye, a fluorescence dye, a radionuclide, a radionuclidecontaining complex, an enzyme, a toxin, an antibody, and a cytotoxic agent.
  21. 21. The method according to claim 20, wherein peptide-TFPI binding is detected by detecting an interaction partner complexed with the peptide or the moiety.
  22. 22. The method according to claim 21, wherein the interaction partner is selected from the group consisting of an antibody, an anticalin, an aptamer, and a spiegeimer.
  23. 23. The method according to claim 21 or 22, wherein the interaction partner comprises a detection moiety, wherein the detection moiety is preferably selected from the group consisting of a dye, a fluorescence dye, a radionuclide, a radionuclide-containing complex, and an enzyme.
  24. 24. A method for treating a subject suffering from a disease or being at risk of suffering from a disease, wherein the method comprises administering to the subject a peptide as defined in any one of claims 1 to 6, wherein the peptide is conjugated to a therapeutic agent and wherein the disease can be treated or the disease symptoms can be ameliorated by the inhibition of TFPI.
  25. 25. A method for treating a subject suffering from a disease or being at risk of suffering from a disease, wherein the method comprises administering to the subject a peptide as defined in any one of claims 1 or 6, and administering to the subject an interaction partner that (a) binds the peptide and (b) is a therapeutic agent or is conjugated to a therapeutic agent, wherein the disease
    07/06/19
    2017203999 07 Jun 2019 can be treated or the disease symptoms can be ameliorated by the inhibition of TFPi.
  26. 26. The method according to claim 25, wherein the interaction partner is selected from the group consisting of an antibody, an anticalin, an aptamer and a spiegelmer.
  27. 27. The method according to any one of claims 24 to 26, wherein the therapeutic agent is selected from the group consisting of a dye, a fluorescence dye, a radionuclide, a radionuclide-containing complex, an enzyme, a toxin, an antibody, and a cytotoxic agent.
  28. 28. The method according to any one of claims 24 to 27, wherein the disease is a blood coagulation disorder.
  29. 29. A method for diagnosing a subject suffering from a disease or being at risk of suffering from a disease, comprising (a) administering to the subject a peptide as defined in any one of claims 1 to 6 conjugated to a detectable moiety and (b) detecting the detectable moiety, wherein the disease is a disease which is associated with TFPI, which can be treated or the symptoms of which can be ameliorated by the inhibition of TFPI.
  30. 30. A method for diagnosing a subject suffering from a disease or being at risk of suffering from a disease, comprising (a) administering to the subject a peptide as defined in any one of claims 1 to 6, (b) administering to the subject an interaction partner conjugated to a detectable moiety, and (c) detecting the detectable moiety, wherein the disease is a disease which is associated with TFPI, which can be treated or the symptoms of which can be ameliorated by the inhibition of TFPi.
    07/06/19
    2017203999 07 Jun 2019
  31. 31. The method according to claim 30, wherein the interaction partner is selected from the group consisting of an antibody, an anticalin, an aptamer, and a spiegelmer.
  32. 32. The method according to claim 30 or 31, wherein the detectable moiety is selected from the group consisting of a dye, a fluorescence dye, a radionuclide, a radionuclide-containing complex, an enzyme, a toxin, an antibody, and a cytotoxic agent.
  33. 33. The method according to any one of claims 29 to 32, wherein the disease is a blood coagulation disorder.
  34. 34. A method for purifying TFPI, wherein the method comprises
    a) contacting a sample containing TFPI with a peptide as defined in any one of claims 1 to 6 under conditions appropriate to form a complex between TFPI and the peptide;
    b) removing the complex from the sample; and, optionally,
    c) dissociating the complex to release TFPI.
  35. 35. The method according to claim 34, wherein the peptide is immobilized to a support, preferably a solid support.
  36. 36. The method according to claim 34 or 35, wherein the peptide is immobilized to a chromatography stationary phase, and step (c) comprises eluting TFPI bound to the immobilized peptide.
AU2017203999A 2008-12-19 2017-06-14 Tfpi inhibitors and methods of use Active AU2017203999B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2017203999A AU2017203999B2 (en) 2008-12-19 2017-06-14 Tfpi inhibitors and methods of use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US61/139,272 2008-12-19
AU2009327338A AU2009327338B2 (en) 2008-12-19 2009-12-21 TFPI inhibitors and methods of use
AU2015234306A AU2015234306B2 (en) 2008-12-19 2015-09-30 Tfpi inhibitors and methods of use
AU2017203999A AU2017203999B2 (en) 2008-12-19 2017-06-14 Tfpi inhibitors and methods of use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2015234306A Division AU2015234306B2 (en) 2008-12-19 2015-09-30 Tfpi inhibitors and methods of use

Publications (2)

Publication Number Publication Date
AU2017203999A1 AU2017203999A1 (en) 2017-07-06
AU2017203999B2 true AU2017203999B2 (en) 2019-07-04

Family

ID=54345667

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2017203999A Active AU2017203999B2 (en) 2008-12-19 2017-06-14 Tfpi inhibitors and methods of use

Country Status (1)

Country Link
AU (1) AU2017203999B2 (en)

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Gerhard, D. S. et al "The Status, Quality, and Expansion of the NIH Full-Length cDNA Project: The Mammalian Gene Collection (MGC)", Genome Res., 2004, Vol. 14, pp 2121-2127 *

Also Published As

Publication number Publication date
AU2017203999A1 (en) 2017-07-06
AU2015234306A1 (en) 2015-10-22

Similar Documents

Publication Publication Date Title
US11001613B2 (en) TFPI inhibitors and methods of use
AU2011227714B2 (en) TFPI inhibitors and methods of use
AU2013235741C1 (en) TFPI inhibitors and methods of use
AU2017203999B2 (en) Tfpi inhibitors and methods of use
US10124033B2 (en) Peptides and methods of use
EA041584B1 (en) PEPTIDES INHIBITING PROTEIN S ACTIVITY AND THEIR USE FOR THE TREATMENT OF BLOOD COAGULATION DISTURBANCES

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
PC Assignment registered

Owner name: TAKEDA PHARMACEUTICAL COMPANY LIMITED

Free format text: FORMER OWNER(S): BAXALTA INCORPORATED; BAXALTA GMBH