Title of Invention

NOVEL FACTOR VIIa INHIBITORS

Abstract The present invention relates to novel compounds, their preparation, their use and pharmaceutical compositions containing the compounds which have a strong antithrombotic effect through reversible inhibition of activated blood coagulation factor VIIa (FVIIa).
Full Text Field of the Invention
The present invention relates to novel compounds, their preparation, their use and pharmaceutical compositions containing the compounds which have a strong antithrombotic effect through reversible inhibition of activated blood coagulation factor Vila (FVIIa).
Background of the Invention
Thrombus formation is normally the result of tissue injury which initiates the coagulation cascade and has the effect of slowing or preventing blood flow in wound healing. Other factors which are not directly related to tissue injury like atherosclerosis and inflammation may also initiate the coagulation cascade and may lead to pathological consequences.
Biood coagulation is a complex process involving a progressively amplified series of enzyme activation reactions in which plasma zymogens are sequentially activated by limited proteolysis. Mechanistically the blood coagulation cascade has been divided into intrinsic and extrinsic pathways, which converge at the activation of factor X; subsequent generation of the thrombin proceeds through a single common pathway (see Scheme 1).


Scheme 1: Blood coagulation cascade
Present evidence suggests that the intrinsic pathway plays an important role in the maintenance and growth of fibrin formation, while the extrinsic pathway is critical in the initiation phase of blood coagulation (H. Cole, Aust. J. Med. Sci. 16 (1995) 87; G.J. Braze, Blood Coagutation and Fibrinolysis 6, Suppl.1 (1995) S7-S13). It is generally accepted that blood coagulation is physically initiated upon formation of a tissue factor(TF)/factor Vila complex. Once formed, this complex rapidly initiates coagulation by activating factors IX and X. The newly generated activated factor X, i. e. factor Xa, then forms a one-to-one complex with factor Va and phospholipids to form a prothrombinase complex, which is responsible for converting soluble fibrinogen to insoluble fibrin via the activation of thrombin from its precursor prothrombin. As time progresses, the activity of the factor Vila/tissue factor complex (extrinsic pathway) is suppressed by a Kunitz-type protease inhibitor protein, TFPI, which, when complexed to factor Xa, can directly inhibit the proteolytic activity of factor Vila/tissue factor. In order to maintain the coagulation process in the presence

of an inhibited extrinsic system, additional factor Xa is produced via the thrombin-mediated activity of the intrinsic pathway. Thus, thrombin piays a dual autocatalytic role, mediating its own production and the conversion of fibrinogen to fibrin.
The autocatalytic nature of thrombin generation is an important safeguard against uncontrolled bleeding and it ensures that, once a given threshold level of prothrombinase is present, blood coagulation will proceed to completion. The ability to form biood clots is vital to survival. In certain disease states, however, the formation of blood clots within the circulatory system is itself a source of morbidity. It is nevertheless not desirable in such disease states to completely inhibit the clotting system because life threatening hemorrhage would ensue. Thus, it is most desirable to develop agents that inhibit coagulation by inhibition of factor Vila without directly inhibiting thrombin.
in many clinical applications there is a great need for the prevention of intravascular blood clots or for some anti-coagulant treatment. The currently available drugs are not satisfactory in many specific clinical applications. For example, nearly 50 % of patients who have undergone a total hip replacement develop deep vein thrombosis (DVT). The currently approved therapies are fixed dose low molecular weight heparin (LMWH) and variable dose heparin. Even with these drug regimes 10 % to 20 % of patients develop DVT and 5 % to 10 % develop bleeding complications.
Another clinical situation for which better anticoagulants are needed concerns subjects undergoing transluminal coronary angioplasty and subjects at risk for myocardial infarction or suffering from crescendo angina. The present, conventionally accepted therapy, which consists of administering heparin and aspirin, is associated with a 6 % to 8 % abrupt vessel closure rate within 24 hours of the procedure. The rate of bleeding complications requiring transfusion therapy due to the use of heparin also is approximately 7 %. Moreover, even though delayed

closures are significant, administration of heparin after termination of the procedures is of little value and can be detrimental.
The most widely used blood-clotting inhibitors are heparin and the related sulfated polysaccharides, LMWH and heparin sulfate. These molecules exert their anti-clotting effects by promoting the binding of a natural regulator of the clotting process, anti-thrombin ill, to thrombin and to factor Xa. The inhibitory activity of heparin primarily is directed toward thrombin, which is inactivated approximately 100 times faster than factor Xa. Hirudin and hirulog are two additional thrombin^-specific anticoagulants presently in clinical trials. However, these anticoagulants, which inhibit thrombin, also are associated with bleeding complications. Preclinical studies in baboons and dogs have shown that targeting enzymes involved at earlier stages of the coagulation cascade, such as factor Xa or factor Vila, prevents clot formation without producing the bleeding side effects observed with direct thrombin inhibitors (T. Yokoyama, A.B. Kelly, U.M. Marzec, S.R. Hanson, S. Kunitada, LA. Harker, Circulation 92 (1995) 485-491; L.A Harker, S.R. Hanson, A.B. Kelly, Thromb. Hemostas. 74 (1995) 464-472; C.R. Benedict, J. Ryan, J. Todd, K. Kuwabara, P. Tyburg, Jr., J. Cartwright, D. Stern, Blood 81 (1993) 2059-2066).
Specific inhibition of the factor Vlla/TF catalytic complex using monoclonal antibody [International Patent Application No. WO92/06711) and a protein such as Dhloromethyi ketone inactivated FVIIa (International Patent Application No. WO96/12800 and W097M7651) is an extremely effective means of controlling tirombus formation caused by acute arterial injury or the thrombotic complications •elated to bacterial septicemia. There is also experimental evidence suggesting that nhibition of factor Vlla/TF activity inhibits restenosis following ballon angioplasty ;t.A. Harker, S.R. Hanson, J.N. Wilcox, A.B. Kelly, Haernostasis 26 (1996) S1:76-32). Bleeding studies have been conducted in baboons and indicate that inhibition tf the factor Vlla/TF complex has the widest safety window with respect to herapeutic effectiveness and bleeding risk of any anticoagulant approach tested

including thrombin, plateiet and factor Xa inhibition (L.A. Marker, S.R. Hanson, A.B. Kelly, Thromb. Hemostas. 74 (1995) 464-472).
A specific inhibitor of factor Vila would have substantial practical value in the practice of medicine. In particular, a factor Vila inhibitor would be effective under circumstances where the present drugs of choice, heparin and related sulfated polysaccharides, are ineffective or only marginally effective. Thus, there exists a need for a low molecular weight, factor Vita-specific blood clotting inhibitor that is effective, but does not cause unwanted side effects. The present invention satisfies this need by providing factor Vila activity inhibiting derivatives of formula I and by providing related advantages as well.
The compounds of formula I are inhibitors of the blood clotting enzyme factor Vila. The invention also relates to processes for the preparation of the compounds of formula I, to methods of inhibiting factor Vila activity and of inhibiting blood clotting, to the use of the compounds of formula I in the treatment and prophylaxis of diseases which can be cured or prevented by the inhibition of factor Vila activity such as thromboembolic diseases including thrombosis, restenosis, infarction and angina, and the use of the compounds of formula. I in the preparation of medicaments to be applied in such diseases. The invention further relates to compositions containing a compound of formula I in a mixture or otherwise in association with an inert carrier, in particular pharmaceutical compositions containing a compound of formula I together with pharmaceutical^ acceptable carrier substances or excipients and/or auxiliary substances or additives.
Summary of the Invention
The present invention provides compounds that specifically inhibit factor Vila activity. In particular, a subject of the present invention are compounds of the formula I

R1-A-B-D-En-R2 (I)
wherein R1 represents
R13,
R12C(0)or
1 to 3 amino acids the N-terminus of which can be substituted with a
substituent selected from the series consisting of R14C(0), R15S(0)2 and an
amino protecting group,
wherein
R12 is selected from the series consisting of alkyl, aikenyl, alkynyl, alkyloxy, alkyiamino, alkenylamino, alkynylamino, alkenyloxy, alkynyloxy, aryl, heteroaryl, heterocycloalkyl, arylalkyl, heteroarylalkyl, heterocycloalkylalkyl, heteroalkyl, heteroalkenyl and heteroalkynyl, which residues can all be substituted,
R13 is selected from the series consisting of an amino protecting group, hydrogen, alkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl and heterocycloalkylalkyl,
R14 and R15 are independently selected from the series consisting of alkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, heterocycloalkyl and heterocycloalkylalkyl,
A is the group A1-A2-A3, wherein
A1 is NH,
A2 is CHR93, wherein R93 is 4-amidinophenylmethyl,
A3 is C(O),
B is the group B1-B2-B3, wherein

B1 is NR95, wherein R95 is selected from the series consisting of
hydrogen and alkyl, B2 is CHR97, wherein R97 is ethyl which is substituted in the 2-position by
a substituent selected from the series consisting of hydroxycarbonyl,
alkyloxycarbony! and aryialkyloxycarbonyl, B3 is C(O),
D is the group D1-D2-D3, wherein
D1 isNH,
D2 is CR81R82, wherein R81 and R82 are independently selected from the series consisting of hydrogen and the unsubstituted or substituted residues aiky), aryl, arylalkyi, heteroaryl, heteroarylalkyi, heterocycloalkyl and heterocycloalkylalkyl,
D3 isC(O),
En is (E1-E2-E3)n, wherein
n is zero, one, two or three,
E1 is NR70, wherein R70 is selected from the series consisting of hydrogen, alkyl, aryl, arylalkyi, heteroaryl, heteroarylalkyi, heterocycloalkyl and heterocycloalkylalkyl,
E2 is CR71R72, wherein R71 and R72 are independently selected from the series consisting of hydrogen and the unsubstituted or substituted residues alkyl, aryl, arylalkyi, heteroaryl, heteroarylalkyi, heterocycloalkyl and heterocycloalkylalkyl,
E3 is C(O),

R2 is selected from the series consisting of NR21R22, OR23 and R24, wherein R21, R22, R23 and R24 are independently selected from the series consisting of hydrogen and the unsubstituted or substituted residues aikyl, aryi, arylalkyl, heteroaryl, heteroaryialkyl, heterocycfoalkyl and heterocycloalkylalkyl,
alky! and heteroaikyi contain 1 to 13 carbon atoms where in a heteroalkyl residue
one or more carbon atoms are replaced with heteroatoms selected from the series
consisting of N, O and S;
alkenyl, aikynyl, heteroalkenyl and heteroalkynyl contain 2 to T3 carbon atoms
where in a heieroa&enyf and heteroalkynyl residue one or more carbon atoms are
replaced with heteroatoms selected from the series consisting of N; 0 and S;
aryl and heteroaryl contain 5 to 13 ring carbon atoms where in a heteroary! residue
one or more carbon atoms are replaced with heteroatoms selected from the series
consisting of N, O and S;
heterocycloalkyl contains 3 to 8 ring carbon atoms of which one to three carbon
atoms are replaced with heteroatoms selected from the series consisting of N, 0 and
S;
in any of their stereoisomer^ forms and mixtures thereof in any ratio, and the
pharmaceutically acceptable salts thereof.
Detailed Description of the Invention
The present invention provides peptides of the formula I
R1-A-B-D-En-R2 (I)
wherein R1, R2, A, B, D, E and n are defined as above, which are compounds that inhibit factor Vl/a activity but do not substantially inhibit the activity of other proteases involved in the blood coagulation pathway, in the compounds of the formula ! structural units are contained, for example in the groups A, B, D or E or in the group R1 in case R1 represents one, two or three amino acids, which are amino

acids or derivatives thereof or amino acid analogs or mimetic structures, and which in peptide fashion are linked to adjacent groups via amide bonds C(0)-N formed between a carboxy group of one such amino acid etc. and an amino group of another amino acid etc. As common in peptide chemistry, a divalent residue of an amino acid or of a group like A, B, D or E as present in formula I is obtained from the respective amino acid by formally removing a hydrogen atom from an amino group and a hydroxy group from a carboxy group.
As used herein, the term "amino acid" is used in its broadest sense to mean the twenty naturally occurring amino acids, which are translated from the genetic code and comprise the building blocks of proteins, including, unless specifically stated otherwise, L-amino acids and D-amino acids, as well as chemically modified amino acids such as amino acid analogs, naturally-occurring amino acids that are not usually incorporated into proteins such as norleucine, and chemically synthesized compounds having properties known in the art to be characteristic of an amino acid. For example, analogs or rnimetics of phenylalanine or proline, which allow the same conformational restriction of the peptide compounds as natural Phe or Pro, are included within the definition of "amino acids" and are known to those skilled in the art. Such analogs and rnimetics are referred to herein as "functional equivalents" of an amino acid. Other examples of amino acids and amino acids analogs are listed by Roberts and Vellaccio (The Peptides: Analysis, Synthesis, Biology, eds. Gross and Meienhofer, vol. 5, p. 341, Academic Press, Inc., New York 1983, which is incorporated herein by reference). Abbreviations of amino acids, amino acid analogs and mimetic structures as well as other abbreviations used in the application are listed in Table 1.
Table 1: Abbreviations used in the application
Compound/Residue Abbreviation
Acetic acid AcOH

Acetylaminomethyl Acrn
Aianine Ala
Allyloxycarbonyl Alloc
p-Amidinophenylalanine pAph
2-Ami no butyric acid 2-Abu
Arginine Arg
Asparagine Asn
Aspartic acid Asp
Benzyl Bzi
i-Butyloxycarbony! Boc
t-Butyl tBu
Cyciohexylgiycine Chg
Cyclohexyl Chx
Cyciohexylaianine Cha
Cysteine Cys
2,4-Diaminobutyric acid Dab
2,3-Diaminopropionic acid Dap
Dichloromeihane DCM
Diisopropylcarbodiimide DIC
Diisopropyethylamine DIEA
N,N-DimethyIformamide DMF
Dimethylsulfoxide DMSO
9-Fluorenylmethyloxycarbonyl Fmoc
Glutamic acid Glu
Glutamine Gin
Glycine Gly
Histidine His
N-Hydroxybenzotriazole HOBt
4-Hydroxymethylphenoxy-
acetic acid HMPA
Isoleucine He

Leucine Leu
Lysine Lys
Methyl Me
N-Methylimidazoie NMI
N-Methylmorpholine NMM
2,2,5,7.8-Pentamethyl-
chroman-6-suffonyl Pmc
Ornithine Orn
Phenyl Ph
Phenylalanine Phe
Phenylglyctne Phg
Proline Pro
Serine Ser
Tetrahydrofuran THF
Tetramethylfluoroformamidino-
hexafluorophosphate TFFH
Threonine Thr
Trifluoroacetic acid TFA
Trityl Trt
Tryptophan Trp
Valine Val
Unless specified otherwise, amino acids abbreviated as specified above have L configuration. Amino acids of D configuration are denoted by D-prefix using three-letter code (for example D-Ala, D-Cys, D-Asp, D-Trp, D-pAph). Abbreviations like, for example, Phe{4-CN) and Phe[4-C(-S-CH2-CH2-S-)-Ph] denote the residue of the amino acid phenylalanine which in the 4-position of the phenyl group carries a cyano substituent or a 2-phenyl-1,3-dithiolan-2-yl substituent, respectively. An abbreviation like, for example, Dap[-C(=NH)-NH2] denotes the residue of the amino acid 2,3-diaminopropionic acid in which the amino group in the side chain, i. e. the amino group in the 3-position, is substituted with an amidino group -C(-NH)-NH2

(carbamimidoy! group) whereby a guanidino group -NH-C(=NH)-NH2 attached to the 3-position of the propionic acid unit results. Abbreviations like, for example, Orn[-C(=NH)-NH2] or Cys(Me) denote the residue of the amino acid ornithine in which the amino group in the side chain carries an amidino group, or the residue of the amino acid cysteine in which the mercapto group carries a methyi group, respectively.
The terms TOTU, HATU and BOP mean 0-[cyan(ethoxycarbonyl)methylenamino]-1,1,3,3-tetramethyluronium tetrafluoroborate, 0-(7-azabenzotriazo!-1 -yl)-1,1,3,3-tetramethyluronium hexafluorophosphate, and 1-benzotriazolyloxy-tris-(dimethylamino)-phosphonium hexafiuorophosphate, respectively.
As used herein, the term "specific" when used in reference to the inhibition of factor Vila activity means that a compound of the formula I can inhibit factor Vila activity without substantially inhibiting the activity of other specified proteases, including plasmin and thrombin (using the same concentration of the inhibitor). Such proteases are involved in the biood coagulation and fibrinolysis cascade.
As used herein, the term "substituent" refers to any of various chemical groups that is substituted onto the peptide backbone or side chain of a peptide, peptide analogue, mimetic or organic compound disclosed herein. A substituent can include any of a variety of different moieties known to those skilled in the art (see, for example, Giannis and Kolter, Angew. Chem. int. Ed. Engl. 32 (1993) 1244-1267, which is incorporated herein by reference).
As used herein, the term "alkyl" is used in the broadest sense to mean saturated or unsaturated, linear, branched or cyclic chains of about 1 to 13 carbon atoms where, of course, an unsaturated alkyl group contains at least 2 carbon atoms and a cyclic alkyl group at least 3 carbon atoms. An unsaturated group can contain one or more double bonds and/or triple bonds. Thus, the term "alkyl" includes, for example, methyl, ethyl, n-propyf, isopropyl, n-butyl, isobuty!, sec-butyl, tert-butyf,

1-methylbutyf, 2,2-dimethylbutyi, 2-methyipentyl, 2,2-dimethyipropyl, n-pentyl and n-hexyl groups, alkylene groups, cyclic chains of carbon atoms such as cyclopropyl, cyciobutyl, cyciopentyl, cyciohexyl and cycloheptyl groups, as well as combinations of linear or branched chains and cyclic chains of carbon atoms such as a methylcyclohexyh cyclohexylmethyl-, 1-cyclohexylethyl-, 2-cyclohexylethyl-, cyclopeniyimethyl-, 1-cyclopentylethyf-, 2-cyclopentylethyl-, cyclopropylmethyl-, 1-cyclopropylethyl- 2-cy do propyl ethyl or cyclopropylmethylene group. Thus, alkyl also comprises cycliG alkyl groups which carry one or more alkyl substituents. Further examples of alkyl are the below-mentioned specific unsaturated groups. In addition, it should be recognized that an alkyl as defined herein can be substituted with one or more identical or different substituents, for example one, two, three or four substituents, which can be present in any desired suitable position,
Preferably, the term "alkyl" means saturated, linear or branched chains of from 1 to 6 carbon atoms, unsaturated linear or branched chains of from 2 to 6 carbon atoms, or cyclic alkyl groups of from 3 to 8 carbon atoms, in particular of from 3 to 6 or of from 4 to 6 ring carbon atoms. With respect to unsaturated alkyl chains, (C2-Cs)-alkenyl and (C2-C6)~aikynyl are preferred. Examples of unsaturated alkyl groups are alkenyl and alkynyl groups such as vinyl, prop-1-enyl, prop-2-enyf (= ally!), but-2-enyl, buten-3-yl, 3-methylbut-2-enyl, ethinyl, prop-2-ynyi, but-2-ynyi and the like.
Similarly, the term "acyl" is used in its broadest sense to mean saturated or unsaturated, linear, branched or cyclic chains of about 1 to 13 carbon atoms or aryl groups having 5 to 13 ring carbon atoms which are attached to a carbony! moiety -C(O)- and are bonded via said carbonyl group. An acyl group can be considered as derived from the respective compound containing a carboxyl group C(0)-OH by formal removal of the hydroxy group. Thus, the term "acyl" encompasses, for example, groups such as formyl, acetyl, benzoyl and the like. A preferred group of acyl groups encompasses the hereinbefore mentioned saturated or unsaturated, i linear, branched or cyclic chains having the preferred range of carbon atoms, which in addition contain a carbony] group via which they are bonded.

The term "ary!" refers to aromatic groups containing about 5 to 13 ring carbon atoms and at least one "ring" group having a conjugated pi electron system. Preferable, the term "ary!" refers to aromatic groups having 6 to 10 ring carbon atoms. Examples of aryi include, for example, phenyl, naphthyl such as 1-naphthyl and 2-naphthylf fluorenyl, biphenylyi groups, and analogues and derivatives thereof, ail of which optionally can be substituted with one or more, for example one, two, three or four, identical or different substituents which can be present in any desired suitable position. For example, a monosubstituted phenyl group can be substituted in 2-, 3-or 4-position, a disubstituted phenyl group in 2,3-, 2,4-, 2,5- 2,6-, 3,4- or 3,5-position.
The term "arylaikyl" refers to an aikyl as defined above substituted with one or more, for example one or two, identical or different aryl groups. Suitable arylalkyl groups include benzyl, phenylethyl such as 1-phenylethyi and 2-phenyiethyl, diphenyimethyi, diphenylethyl such as 1,2-diphenylethyl and 2,2-diphenylethyl, phenylpropyl such as 1-phenylpropyI, 2-phenylpropyl and 3-phenylpropyl, diphenylpropyi such as 2,3-diphenylpropyl and 3,3-diphenylpropyl, naphthyl methyl, naphthylethyl such as 1-naphthylethyl and 2-naphthylethyl, naphthylpropyl such as 1-naphthy!prapyl, 2-naphthylpropyl and 3-naphthylpropyl, 1,2,3,4-tetrahydro-1-naphthyl, 1h2,3,4-tetrahydro-2-naphthyl, and the like, ail of which can optionally be substituted.
The terms "heteroalkyl", "heteroalkenyi, "heteroalkynyl, "heteroarylalkyl" and "heteroaryl" as used herein refer to an aikyl, arylalkyl and aryi group, respectively, wherein one or more carbon atoms, for example one, two or three carbon atoms, are replaced with identical or different heteroatoms such as N, O or S. In addition, the term "heterocycioaikyl" is used in reference to a cyclic aikyl group in which one or more ring carbon atoms are replaced with heteroatoms, Preferably, the term "heterocycioaikyl" means a cyclic aikyl group having 3 to 8 ring carbon atoms, of which 1, 2 or 3 are replaced with identical or different hetero atoms such a N, O or S.

All these groups can be bonded via any desired suitable position including suitable ring nitrogen atoms in the case of nitrogen heterocylces. Suitable heteroaryl groups, heteroarylalkyl groups, heteroaikyl groups and heterocycloalkyl groups include, for example, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-thienyl, 3-thienyl, indolyl, imidazolyl, furyl, piperonyl 2-pyridyimethyl, 3-pyridylmethyL 4-pyridylmethyl, 1-(2-pyridyl)ethyl, 1-(3-pyridyljethyl, 1-(4-pyridyl)ethyl, 2-(2-pyridyl)ethyl, 2-{3-pyridyl)ethyi, 2-(4-pyridy))ethyl, picolyl, pyrrolidinyl, piperidinyl, tetrahydrofuryl, tetrahydrofuran-2-yimethyi, morpholinyl, 4-morphoiinyl, 2-(4-morpholiny!)ethyl, piperazinyl, 2-(4~ methyipiperazin-1-yl)ethyl, and the like, all of which can optionally be substituted with one or more, for example one, two, three or four, identical or different substituents.
The peptides of the invention can be modified at the N-terminus and/or the C-terrninus by reaction with suitable reagents or by introduction (or by the presence of) an amino-protecting group or carboxy-protecting group, respectively. The N-terminus of a peptide or peptide analog can be chemically modified such that the N-terminus amino group is substituted, for example, by an acyl group (for example acetyl, cyclopentylcarbonyl isoquinolylcarbonyl, furoyS, tosyl, benzoyl, pyrazinecarbonyl or other such groups), by reaction with an isocyanate, chloroformate, alkylating agent or by introducing other such group, a)l of which can be substituted by a substituent as described above: It should be recognized that the term "amino group" is used broadly herein to refer to any free amino group, including a primary, secondary or tertiary amino group, present in a peptide. In comparison, the term "N-terminus" refers to the ct-amino group of the first amino acid present in a peptide written in the conventional manner.
The N-terminus of a peptide of the invention can be protected by linking thereto an amino-protecting group. The term "amino-protecting group" is used broadly herein to refer to a chemical group that can react with a free amino group, including, for example, the a-amino group present at the N-terminus of a peptide of the invention. By virtue of reacting therewith, an ammo-protecting group protects the otherwise

reactive amino group against undesirable reactions as can occur, for example, during a synthetic procedure or due to exopeptidase activity on a final compound. Modification of an amino group also can provide additional advantages, including, for example, increasing the solubility or the activity of the compound. Various amino-protecting groups are disclosed herein or otherwise known in the art and include, for example, acyi groups such as an acetyl, tert-butyioxycarbonyf, aNyloxycarbonyl, benzyloxycarbonyl group or benzoyl groups, as well as an aminoacyl residue, which itself can be modified by an amino-protecting group. Other amino-protecting groups are described, for example, in The Peptides, eds. Gross and Meienhofer, vol. 3 (Academic Press, Inc., New York, 1981); and by Greene and Wuts, in Protective Groups in Organic Synthesis, 2d ed., pages 309-405 (John Wiley & Sons, New York, 1991), each of which is incorporated herein by reference. The product of any such modification of the N-terrninus amino group of a peptide or peptide analog of the invention is referred to herein as an "N-termina) derivative."
Similarly, a carboxy group such as the carboxy group present at the C-terminus of a peptide can be chemically modified using a carboxy-protecting group. The terms "carboxy group" and "C-terminus" are used in a manner consistent with the terms "amino group" and "N-terminus" as defined above. A carboxy group such as that present at the C-terminus of a peptide can be modified by reduction of the C-terminus carboxy group to an alcohol or aldehyde or by formation of an oral ester or by substitution of the carboxy group with a substituent such as a thiazolyl, cyclohexyi or other group. Oral esters are well known in the art and include, for example, aikyloxymethyl groups such as methoxymethyl, ethoxymethyl, isopropoxymethyl and the like; the 1-((Ci-C4)-alkyloxy)ethyl groups such as methoxyethyl, ethoxyethyl, propoxyethyl, isopropoxyethyi and the like; the 2-oxo-1,3-dioxo!en-4~ylmethyi groups such as 5-methyl-2-oxo-1,3-dioxolen-4-ylmethyiT 5-phenyi-2-oxo-1,3-dioxolen-4-ylmethyl and the like; the ((CrC3)-alkylthio)methyl groups such as methylthiomethyl, ethyithiomethyl, isopropyithiomethy! and the like; the acyloxymethyl groups such as pivaloyloxymethyl, acetoxymethyl and the like; the ethoxycarbonylmethyl group; the 1-acyloxy-1-substituted methyl groups such as 1-acetoxyethyl; the 3-phthalidyl or

5,6-dirnethylphthalidyl groups; the 1-({(Ci-C4)-alkylaxy}carbonyloxy)ethyl groups such as the 1-(ethoxycarbonyloxy)ethyl group; and the 1-(((Ci-C4)-alkylamino)carbonyloxy)ethyl group such as the 1-(methylaminocarbonyfoxy)ethyl group.
A peptide of the invention can be modified by linking thereto a carboxy-protecting group. Carboxy-protecting groups are well known in the art and, by virtue of being bound to a peptide, protect a carboxy group against undesirable reactions (see, for example, Greene and Wuts, supra, pages 224-276 (1991), which is incorporated herein by reference). The skilled artisan would recognize that such modifications as described above, which can be effected upon the N-terminus amino group or C-terminus carboxy group of a peptide, similarly can be effected upon any reactive amino group or carboxy group present, for example, on a side chain of an amino acid or amino acid analog in a peptide of the invention. Methods for performing such modifications are disclosed herein or otherwise known in the art.
The choice of including an L- or a D-amino acid in a compound of the present invention can depend, in part, on the desired characteristics of the peptide. For example, the incorporation of one or more D-amino acids can confer increased stability on the compound in vitro or in vivo. The incorporation of one or more D-amino acids also can increase or decrease the pharmacological activity of the compound. In some cases it can be desirable to allow the compound to remain active for only a short period of time. In such cases, the incorporation of one or more L-amino acids in the compound can allow endogenous peptidases in an individual to digest the compound in vivo, thereby limiting the individual"s exposure to the active compound. The skilled artisan can determine the desirable characteristics required of compound of the invention by taking into consideration, for example, the age and general health of an individual. In general, the present invention relates to the compounds of the formula ! in all their stereoisomeric forms and mixtures of two or more stereosiomers in all ratios, for example to pure enantiomers, pure diastereomers, mixtures of two enantiomers in all ratios including racemates,

mixtures of diastereomers, cis isomers, trans isomers, E isomers or Z isomers. The invention also relates to the compounds of the formula I in ail their tautomeric forms. Further, the invention relates to prodrugs of the compounds of the formula 1, for example esters, amides, aldehydes or alcohols obtainable from carboxy groups as already mentioned, or acyl derivatives like (Ci-Cs)-alkylcarbonyl, (Ci-Cs)-alkyioxycarbonyl or aryl-(Ci-C4)-alkyloxycarbonyl derivatives obtainable from acylatable groups including amino groups, imino groups, guanidino groups and amidino groups, and the invention also relates to active metabolites of the compounds of the formula I.
In the compounds of the formula I the group R1 preferably is R12C(0). A specific series of denotations of R12 is formed by the groups alky!, alkenyl, alkynyl, alkyioxy, alkytamino, alkenyiamino, alkynyiamino, alkenyloxy, alkynyloxy, aryl, heteroaryl, heterocycloaikyl, heteroaryJaikyl, heterocycloalkylalkyl, heteroaikyl, heteroaikenyl and heteroalkynyi, which residues can all be unsubstituted or substituted. R12 preferably is atkyl, alkenyl, alkynyl, alkyioxy, alkenyloxy, alkynyloxy, alkylamino, alkenyiamino, alkynyiamino, aryl, heteroaryl, arylalkyl or heteroarylalkyl, more preferably alkenyloxy, alkenyiamino or aryl, which residues can all be unsubstituted or substituted. Particularly preferably R12 is alkenyloxy or alkenyiamino like (Cz-CB)-alkenyloxy or (C2-C6)-alkenylamino each containing one double bond, for example allyloxy or allyiamino. Moreover preferably R12 is {C2-Cs)-alkenyioxy. The residues representing R12 can be unsubstituted or substituted. In substituted residues R12 the residues preferably are substituted with one or more identical substituents selected from the series consisting of halogen, i. e. fluorine, chlorine, bromine or iodine, trifluoromethyl, hydroxy, nitro, amino, cyano, carboxy, aminocarbonyi, alkylsulfonyl, aminosuffony], alkyioxy, afkyicarbonylamino and mono-or dialkylamino. Similarly, the residues representing R13, R14 and R15 can be unsubstituted or substituted, for example by the substituents that can be present in R12, where R14 and R15 are independent of each other and can be identical or i different.

The group A in the compounds of the formula I which is the divafent 4-amidinophenylaianine residue-NH-CH[-CH2-C6H4-(4-C(=NH)-NH2)]-C(0)-, preferably is an (L)-4-amidinophenyialanine residue {= (S)-4-amidinophenyialanine residue). The group B which is the divalent glutamic acid residue -NH-CH[-CH2-CHrC(0)OH]-C{0)- or a pharmaceutical acceptable salt or ester thereof, preferably is an (L)-glutamic acid residue (= (S)-giutamic acid residue) or a pharmaceutical^ acceptable salt or ester thereof.
R95 preferably is hydrogen or (Ci-C4)-alkyl, more preferably hydrogen or methyl, particularly preferably hydrogen.
Substituted residues R81 and RS2 can independently carry one or more, for example one, two, three or four, identical or different residues which preferably are selected from the series consisting of amino, aminocarbonyl, amidino, guanidino, aminoalkyt, hydroxy, mercapto, which can all be substituted with a protecting group, and acetimido (-C(=NH)-CH3), nitro and cyano. As regards nitro groups, in the compounds of the formula I according to the invention preferably only up to two nitro groups are present. Suitable protecting groups for the listed groups are known to one skilled in the art and can be found in the abovementioned references like Greene and Wuts, Protective Groups in Organic Synthesis, 2d ed., (John Wiley & Sons, New York, 1991), which is incorporated herein by reference. Examples of protecting groups are the abovementioned amino protecting groups like tert-butyloxycarbonyl, benzyJoxycarbonyl and aUyloxycarbonyl which can also be protective groups on amidino groups and guanidino groups, the nitro group which can be used to protect a guanidino group, or groups like benzyl, methyl, trityl or acetylaminomethyl which can be used to protect groups like hydroxy, mercapto and others. Preferably R81 and R82 are selected from the series consisting of hydrogen, alkyl like {CrC^-alky!, aryl like phenyl, arylalkyl like phenyl-(Ci-C2)-alkyl and heteroarylalkyl like heteroaryHCi-C2)-alkyl, which can all be unsubstituted or substituted and in which heteroaryl preferably is the residue of a monocyclic or bicyclic aromatic ring system containing one or two identical or different ring

heteroatcms such as N, 0 or S. Mare preferably RS1 is hydrogen and R32 is an unsubstituted or substituted residue as defined.
Particularly preferably the group D represents a residue selected from the series consisting of Arg. Dap, Dab, Orn, Lys, Dap[-C(~NH)-NH2], Dab[-C(=NH)-NH2], Lys[-C(=NH)-NH2], Lys[-C(=NH)-CH3], Orn[-C(=NH)-CH3], Dab[-C(=NH)-CH3], Dap[-C(=NH)-CH3], Dab(Alloc), Asn, Gin, Met, Ser, Thr, Ser(Bzt), Thr(BzI), Cys(Me), Cys(Bz!}, Cys(Acm), Arg(N02}1 His, Trp, Phg, Giy, Ala, Val, lie, Leu, Phe, Phe(4-N02), Phe(4-NH-C(=NH)-NH2), 2-Abu, Ala The number n preferably is zero, one or two, more preferably zero or one. If n is zero the group R2 is directly bonded to the carbonyl group representing D3. If n is different from zero the group R2 is bonded to the carbonyl group representing the terminal group E3. If n is two or three the groups E can all be identical or different.
Substituted residues R71 and R72 can independently carry one or more, for example one, two, three or four, identical or different residues which preferably are selected from the series consisting of alkyl, aikyloxy, halogen, trifluoromethyl, nitro, cyano, alkylsulfonyl, alkylcarbonyl, phenylcarbonyl and2-phenyl-1,3-dithiolan-2-yl, which can be further substituted. A subgroup of substituents which can be present in R71 and R72 is formed by the series consisting of alkyl, aikyloxy, halogen, trifluoromethyl, nitro, cyano, alkylsulfonyl and alkylcarbonyl, which can be further substituted. Preferably R71 is hydrogen and R72 is an unsubstituted or substituted residue as defined. R72 preferably is alkyl, in particular (C3-Ca)-aIkyl, including cyclic alkyl like cycioaikylalkyi such as cyciaa!kyl-(Ci-C2)~a!kyi, or aryL in particular phenyl, or aryfalkyl, in particular phenyl-(Ci-C2}-a(ky(, or heteroarylalky/, in particular

heteroaryi-(Ci~C2)-alkyf, where all these residues can be unsubstituted or substituted and where heteroaryl preferably is a monocylic 5-membered or 6-membered aromatic ring containing one or two identical or different ring heteroatoms such as N, O and S. The group or the groups E, in particular in case the number n is one, is preferably selected from the series consisting of Phe which is unsubstituted or substituted in the phenyl group, Cha and Chg. Particularly preferably E is selected from the series consisting of Cha, Chg and Phe[4-C{-S-CH2-CH2~S-)-Ph]. The group R70 present in the group E is preferably hydrogen, alky!, in particular (C,-C4)-alkyl including methyl, or arylalkyl, in particular phenyl-(Ci-C4}-alkyl including benzyl and 2-phenylethyl which can be unsubstituted or substituted in the phenyl group. Particularly preferably R70 is hydrogen.
Substituted residues R21, R22, R23 and R24 can independently carry one or more, for example one, two, three or four, identical or different residues which preferably are selected from the series consisting of halogen, in particular F, CI, Br, hydroxy, trifluoromethyl, nitro, cyano, dialkylamino, alkyloxy like metbyloxy, aikylenedioxy, alkylsuffonyl, aminosulfonyi and oxo (=0)., which can be further substituted. Examples of alkylenedioxy are methyl enedioxy (0-CH2-0) or 1,2-ethylenedioxy. Examples of dialkylamino are dirnethylamino, diethyiarnino or dibutylamino, examples of alkylsulfonyl are methylsulfonyl, ethylsulfonyl or butylsulfonyl. R2 preferably is NR21R22 wherein R21 and R22 are as defined. R21 preferably is hydrogen, (C-,-C4}-alkyl or phenyi-(C-rC4)-alkyi which is unsubstituted or substituted in the phenyl group. Particularly preferably R21 is hydrogen, i. e. particularly preferably NR21R22 is NHR22, and thus particularly preferably R2 is NHR22. R22 preferably is a residue selected from the series consisting of hydrogen, alkyl, in particular (Ci-Ci2)-aikyl, including cyclic alkyl like cycloalkylalkyl such as cycloalkyl-(C-rC4)-alkyl, aryl, in particular (Ce-C^-aryl, arylalkyl, in particular (Ci-C4)-alkyl substituted with one or two (Cs-Ct2)-aryl residues, heteroarylalky!, in particular (Ci-C4)-alkyi substituted with a monocyclic or bicyclic heteroaryl residue containing one or two identical or different heteroatoms such as N, 0 or S, and heterocycloaikylatkyl, in particular (Ci-C4)-alkyl substituted with a monocyclic 4-, 5-,

6- or 7-membered heterocycloalkyl group containing one or two identical or different heteroatorns such as N, O or S, which residues can al! be unsubstituted or substituted as indicated before.
Particularly preferably R22 is a residue selected from the series consisting of hydrogen, benzyl, naphthyimethyl, pyridylmethyl, phenylethyl, naphthylethyl, pyridylethyl, phenylpropyl, naphthylpropyl, pyridylpropy!, fluorenyl, diphenylmethyl, diphenylethyl and diphenyipropyl, which residues are unsubstituted or substituted with one or more, for example one, two; three or four, identical or different substituents,which are preferably selected from the series consisting of F, C\, Br, hydroxy, methoxy, methylenedioxy, nitro, cyano, dialkylamino, alkylsulfonyl, amincsuifonyl and trifluoromethyl, which can be further substituted. A series of particularly preferred compounds of the formula I is formed by compounds in which simultaneously the number n is different from zero, R2 is NHR22 and R22 is hydrogen. Another series of particularly preferred compounds is formed by compounds in which simultaneously n is zero, R2 is NHR22 and R22 is different from hydrogen, where in this series of compounds a preferred denotation of the group D is Asn.
Preferred compounds of the formula I are those compounds in which one or more of the groups or residues have preferred denotations, all combinations of preferred denotations being a subject of the present invention.
A group of preferred compounds of the invention is formed by compounds of the
formula I wherein
R1 is R12C{0) wherein R12 is as defined,
A is as defined,
B is as defined, and preferably B is NH-CHR97-C(0) wherein R97 is ethyl which is
substituted in the 2-position by hydroxycarbonyl or a salt thereof or afkyloxycarbonyl
like (Ci-C4}-alkyfoxycarbonyl,
D is NH-CHR82-C(0), wherein R82 is as defined,

En is (E1-E2-E3)ni wherein
n is zero, one or two,
E1 is NH,
E2 is CHR72, wherein the residues R72 which are independent of each other
and are identical or different, are as defined,
E3 is C(O), and R2 is as defined.,
in any of their stereoisomer^ forms or a mixture thereof in any ratio, and the pharmaceutical^ acceptable salts, amides and esters thereof.
A group of particularly preferred compounds is formed by the compounds of the
formula i wherein
R1 is allyloxycarbonyl or allylaminocarbonyl,
A is the residue of (L)-4-amidinophenylalanine,
B is the residue of (L)-giutamic acid or a pharmaceutically aceptable salt or ester of (L)-giutamic acid,
D is a residue selected from the series consisting of Arg, Dap, Dab, Orn, Lys, Dap[-C(=NH)-NH2], Dab[-C(=NH)-NH2], Lys[-C(=NH)-NH2], Lys[-C(=NH)-CH33, Orn[-C(=NH)-CH3], Dab[-C(=NH)-CH3], Dap[-C(=NH)-CH3], Dab(Alloc), Asn, Gin, Met, Ser, Thr, Ser(Bzf), Thr(Bzl), Cys(Me), Cys(Bzl), Cys(Acrn), Arg(N02), His, Trp, Phg, Gly, Ala, Val, lie, Leu, Phe, Phe(4-N02), Phe(4-NH-C(=NH)-NH2), 2-Abu, Ala{3-CN), Ala[3-C(=NH)-NH2], 2-Abu(4-CN) and 2-Abu[4-C(=NH)-NH23,
n is zero or one,
E is a residue selected from the series consisting of Cha, Chg and Phe[4-C(-S~CH2-CH2-S-)-Ph],
R2 is NHR22,
R22 is hydrogen or a residue selected from the series consisting of benzyl, naphthylmethyi, pyridylmethyl, phenyfethyl, naphthylethyl, pyridylethyi, phenylpropyl, naphthylpropyl, pyridylpropyl, fluorenyJ, diphenyimethyl, diphenylethyl and diphenylpropyl, which residues are unsubstituted or

substituted with one or more identical or different substituents selected from the series consisting of F, Ci, Br, hydroxy, methoxy, methylenedioxy; nitro, cyano, dialkyiamino, alkylsulfonyl, aminosulfonyl and trifluoromethyi,
in any of their stereoisomeric forms era mixture thereof in any ratio, and the
pharmaceutically acceptable salts, amides and esters thereof.
Specific examples of the compounds of the invention include, for example, the compounds listed in Table 2 below and in the example section, and their pharmaceutically acceptable salts, amides and esters.
The compounds of the formula i can be prepared, for example, according to the methods of solid phase chemistry by a process which comprises a1) coupling a compound of the formula Fmoc-En-OH wherein n is one, two or three, to an acid sensitive iinker attached to a resin or in general a solid support, cleaving off the protecting group Fmoc, coupling a compound of the formula Fmoc-D1-D2-C(0)OH to the free amino group obtained and again cleaving off the protecting group Fmoc,
or for the preparation of a compound of the formula I in which n is zero, coupling a compound of the formula Fmoc-D1-D2-C(0)OH to an acid sensitive linker attached to a resin or in general a solid support, and cleaving off the protecting group Fmoc, a2) coupling a compound of the formula Fmoc-B1-B2-C(0)OH to the free amino
group obtained in step a1) and cleaving off the protecting group Fmoc, a3) coupling a compound of the formula R1-A1-A2-C(0)OH to the free amino group
obtained in step a2), and a4) cleaving off the compound obtained according to steps a1) through a3) from the resin by means of trifluoroacetic acid.
The resin or the linker used in this process may be of a type such that the carboxy group in the compound which is coupled to the restn or the linker, respectively, is transformed into an amide group C{0)-NH2, for.example, a Knorr Linker or a Rink

amide resin. The preparation of a compound in which the number n is two or three can also be carried by stepwise assembling the unit ER as follows. In step a1) instead of a compound of the formula Fmoc-En-OH wherein n is two or three first a compound of the formula Fmoc-En-OH wherein n is one is coupled to an acid sensitive linker attached to a resin, then the protecting group Fmoc is cleaved off and a second compound of the formula Fmoc-En-OH wherein n is one is coupled to the free amino group obtained. For the preparation of a compound in which n is three then the protecting group Fmoc is cleaved off and a third compound of the formula Fmoc-En-OH wherein n is one is coupled to the free amino group obtained. Finally, the protecting group Fmoc is cleaved off and steps a2) through a4) follow.
Another process for the preparation of the compounds of the formula I comprises b1) coupling the side chain carboxylic acid of a compound of the formula Fmoc~B1-
CHR97-C(0)OPG, wherein R97 is 2-hydroxycarbonylethy! and PG is a
protecting group, to an acid sensitive benzylalcohol type of linker attached to an
amino functionaiized resin, b2) cleaving off the protecting group PG, b3) coupling a compound of the formula H2N-D2-D3-En-R2, wherein n is zero, one,
two or three, to the free carboxylic acid obtained in step b2), b4) cleaving off the protecting group Fmoc, t>5) coupling a compound of the compound R1-A1~A2-C(0)OH to the free amino
group obtained in step b4), and b6) cleaving off the compound obtained according to steps b1) through b5) from the
resin by means of trifluoroacetic acid.
Similarly to the modification of the first process described above, in this process the structural unit H2N-D2-D3-En-R2 may be assembled stepwise on the resin. According to a further process similar to this process, the compounds of the formula I can also be prepared by first coupling a carboxylic acid group which is present in a side chain in the group D2 of the group D, t. e. which is present in one of the groups R81 and R82, to a linker attached to a resin. Analogously to the above compound of the

formula Fmoc-B1 -CHRS7-C(0)OPG, such a compound may for example have the formula Fmoc-NH-CR8tR82-C(0)OPG wherein R82 is as defined with the proviso that it contains a group C(0)OH, and R81 is as defined. For example, R81 can be hydrogen and R82 can be hydroxycarbonylmethyl and the compound of the formula Fmoc-NH-CR81R82-C(0)OPG can thus be a protected aspartic acid derivative. After deprotecting the group C(0)OPG the carbonyl group of which is the group D3 in formula I, the free carboxylic acid group obtained is coupled with a compound like H2N-En-R2 or H-R2. Then, after deprotecting the protecting group Fmoc, the amino group obtained is coupled with a compound of the formula Fmoc-B1-B2-C(0)OH and, after deprotecting the amino group, the product is coupled with a compound of the formula R1-A1-A2-C(0)OH. Again the resin or the linker used maybe of a type such that the carboxy group in the compound which is coupled to the resin or the linker, is transformed into an amide group C(0)-NH2. For example, by using an amide resin an aspartic acid unit attached to the resin can be converted into an asparagine unit in the final compound.
A compound of the invention can be chemically synthesized using, for example, an automated synthesizer (see Example I). Selective modification of a reactive group such as a group present on an amino acid side chain or an N-terminus or a C-terminus reactive group in a peptide can impart desirable characteristics such as increased solubility or enhanced inhibitory function to a compound of the invention. Where solid phase synthesis methods are employed, the chemical composition of a compound can be manipulated while the nascent peptide is attached to the resin or after the peptide has been cleaved from the resin to obtain, for example, an N-terminal derivative such as an N-terminus acylated, e. g. acetylated, compound. Similar modifications also can be made to a carboxy group of a compound, including a C-terminus carboxy group, which, for example, can be amidated.
The compounds can also be prepared by coupling of protected amino acids according to the methods of traditional medicinal chemistry, or solution phase organic chemistry, and deprotecting to the target molecule, by standard procedures

known in the art. In general, suitable reactions for the synthesis of the compounds of the formula I by solid phase methods or solution phase methods as well as experimental details like suitable coupling agents such as carbodiimides, TOTU or HATU, or solvents and reaction temperatures, are well known to one skilled in the art and can also be found in standard references including the references mentioned herein as weil as are exemplified below.
A synthesized compound can be purified using well known methods such as reverse phase-high performance liquid chromatography (RP-HPLC; see Example I) or other methods of separation based, for example, on the size, charge or hydrophobicity of the compound. Similarly, well known methods such as amino acid sequence analysis or mass spectrometry (MS) can be used for characterizing the structure of a compound of the invention (see Example I).
Various compounds containing different arrangements of the substituents exhibit different levels of inhibitory activity for factor Vila. For example, the choice of the substituents influences the binding affinity of the compounds. These compounds were synthesized according to the procedures described in the Examples. Testing the peptides for inhibitory activity was accomplished using the assay described in Example 22. Using such methods, one skilled in the art can synthesize a compound as disclosed herein, including a modification thereof, and determine the factor Vila inhibitory activity of the compound. A composition of the present invention can be provided as a homogenous composition or as a mixture of compounds containing various combinations of substituents. The flexibility permitted by the choice of substituents permits a great deal of control over the biological and physico-chemical properties of the compounds and compositions of the invention.
The invention provides compounds that specifically inhibit factor Vlf activity. Such compounds preferably have a Ki
same concentration of the inhibitor). Such other proteases include, for example, factor Xa, thrombin and piasmin.
The following Table 2 shows the factor Vila inhibitory activities (see Example 22 for
the method of determining Ki) of selected compounds of the formula I which also
exemplify the invention.
Table 2: Factor Vila inhibitory activities of selected compounds of the formula I
Ki (pM)
AIIoc-pAph-Glu-Arg-Cha-NH2 0.046
AIIylaminocarbonyf-pAph-Glu-Arg-Cha-NH2 0.042
Alloc-pAph-Glu-Arg-Chg-NHz 0.238
A!loc-pAph-Glu-Dap[-C(=NH)-NH2]-Cha-NH2 0.012
Alloc-pAph-Giu-Ala[3-C(=NH)-NH2]-Cha-NH2 0.03
A!toc-pAph-Glu-Asn-Cha-NH2 0.021
Alloc-pAph-GIu-Dab-Cha-NH2 0.055 "
Alloc-pAph-Glu-Dap[-C{=NH)-NH2]-NH2 0.26
Atioc-pAph-Glu-Giy-Cha-NH2 0.12
Alloc-pAph-Glu-Thr(B2l)-NH-(CH2)2-CH{Ph)2 0.17
Alloc-pAph-Glu-Dab-NH-(CH2)2-Ph 0.38
Ailoc-pAph-Glu-Asn-NH-CH2-Chx 0.15
Alioc-pAph-G!u-Dap[-C(=NH)-CH33-Cha-NH2 0.11
Alloc-pAph-Glu-Dab[-C(=NH)-NH2]-Cha-NH2 0.012
Alloc-pAph-Glu-2-Abu(4-CN)-Cha-NH2 0.063
Alloc-pAph-Giu-A!a(3-CN)-Cha-NH2 0.12
AUoc-pAph-Glu-Asn-1 -naphthyirnethylamide 0.031
Alioc-pAph-Glu-Asn-1-(1-naphthy!)ethylamide 0.021
Alloc-pAph-Glu-Asn-2~naphthylrnethylamide 0.027
A!loc-pAph-Glu-Asn-3,4-dich!orobenzylamide 0.026

Ailoc-pAph-G Alloc-pAph-G Ailoc-pAph-G Alloc-pAph-G Alloc-pAph-G AiJoc-pAph-G Alloc-pAph-G Alloc-pAph-G AHoc-pAph-G Alloc-pAph-G Alloc-pAph-G AlloopAph-G Alioc-pAph-G Alioc-pAph-G AlioopAph-G Alloc-pAph-G Alloc-pAph-G AHoc-pAph-G A!loc-pAph-G Alloc-pAph-G Alloc-pAph-G Alloc-pAph-G Alloc-pAph-G Alioc-pAph-G Aiioc~pAph-G Alloc-pAph-G Alloc-pAph-G Alloc-pAph-G AlloopAph-G Alloc-pAph-G Alloc-pAph-G
iu-Asn-2-{3-chioropheny!}ethy!amide 0.023
lu-Arg(N02)-Cha-NH2 0.014
lu-Cys(Bz!)-Cha-NH2 0.026
iu-Trp-Cha-NH3 0.017
iu-Phg-Cha-NH2 0.017
iu-Asn-9-f)uoreny!amide 0.023
iu-Asn-3,5-bistrifluoromethylbenzylamide 0.033
lu-Pbe(4-guanidino)-Cha-NH2 0.12
iu-D-Phe(4-guanidino)-Cha-NH2 11.3
iu-Orn[~C(=NH)-CH3]-Cha-NH2 0.13
lu-Dab[-C(=NH)-CH3]-Cha-NH2 0.19
lu-Dap[-C(=NH)-NH2]-Phe[4-C(-S-(CH2}2-S-}-Ph]-NH2 0.015
lu-Gln-NH2 1.5
lu-Orn-NH2 6.2
iu-Gly-Cha-NH2 0.12
lu-Cys(Acm)-Cha-NH2 0.12
lu-Cys(Me)-Cha-NH2 0.20
[u-Cys(Bzl)-Cha-NH2 0.026
lu-Thr(Bzl)-Cha-NH2 0.019
iu-Dab(Alloc)-Cha-NH2 0.15
iu~His-Cha~NH2 0.14
lu-Met-Cha-NH2 0.11
lu-Phe(4-N02)-Cha-NH2 0.046
lu-D-Lys[-C(=NH)-NHa]-Cha-NHa 22
!u-D-Arg-Cha-NH2 12
lu-Asn-3,4-methylenedioxybenzylamide 0.12
iu-Asn-2-{4-morpholinyl)ethyfamide 0.41
lu-Asn-2-(2-naphthyl)ethylamide 0.052
lu-Asn-2-(1-naphthyl)ethylamide 0.022
lu-Asn-2-tetrahydrofuranylmethylamide 0.17
lu-Asn-3-methylbutylamide 0.11

AIIoc-pAph-Giu-Asn-2-(2-pyridyl)ethy!amide 0.071
Alloc-pAph-G!u-Asn-1,2,3,4-tetrahydro-1 -naphthylamide 0.045
Alloc-pAph-Giu-Asn-N,N-dibenzylarnide 0.41
Alloc-pAph-Glu-Asn-N-rnethyl-N-(1 -naphthySmethyl)arnide 1.7
Alloc-pAph~Glu-Asn-2,2-diphenylethyfamide 0.049
Alfoc-pAph-Gfu-Asn-2,4~difiuorobenzylamide 0.051
Alloc-pAph-G(u-Asn-2-{4-sulfamoylpheny!)ethylamide 0.35
Alioc-pAph-Giu-Asn-4-dimethy laminobenzyiamide 0.11
AJIac-pAph-Glu-Asn-(CH3-)Cha-NH2 0.062
Alloc-pAph-Glu-Asn-3-phenyipropyiamide 0.026
AIIoc-pAph-Glu-Asn-3,3-diphenylpropylamide 0.024
Alloc-pAph-Giu-Asn-4-methoxybenzylamide 0.083
A!loc-pAph-Glu-Asn-3,4-dichlorobenzy!amide 0.026
The thrombin inhibitory activities of the above compounds can be expressed in Ki values which generally are considerably higher than the above indicated factor Vila inhibitory activities, for example about 200 or about 500 or about 1000 times as high as the factor Vila inhibitory activities. Also, the factor Xa inhibitory activities of the above compounds as determined can be expressed in Ki values which generally are considerably higher than the above indicated factor Vila inhibitory activities, for example about 100 times as high as the factor Vila inhibitory activities.
These results demonstrate that the compounds of the formula I are useful as inhibitors of factor Vila, but do not substantially inhibit the activity of factor Xa or serine proteases such as thrombine, which are involved in the process of blood coagulation and fibrinolysis.
A compound of the invention can be used advantageously as an anticoagulant, which can be contacted with a blood sample to prevent coagulation. For example, an effective amount of a compound of the invention can be contacted with a freshly drawn blood sample to prevent coagulation of the blood sample. As used herein, the

term "effective amount" when used in reference to a compound of the invention means an amount of a compound that inhibits factor Vila activity. The skilled artisan would recognize that an effective amount of a compound of the invention can be determined using the methods disclosed herein (see Example 22) or otherwise known in the art. )n view of the disclosed utility of a compound of the invention, the skilled artisan also would recognize that an agent such as heparin can be replaced with a compound of the invention. Such a use of a compound of the invention can result, for example, in a cost saving as compared to other anticoagulants.
In addition, a compound of the invention can be administered to an individual for the treatment of a variety of clinical conditions, including, for example, the treatment of a cardiovascular disorder or a complication associated, for example, with infection or surgery. Examples of cardiovascular disorders include restenosis following angioplasty, aduit respiratory disstress syndrome, multi-organ failure, stroke and disseminated intravascular coagulation clotting disorder. Examples of related complications associated with surgery include, for example, deep vein and proximal vein thrombosis, which can occur following surgery. Thus, a compound of the invention is useful as a medicament for reducing or inhibiting unwanted coagulation in an individual.
Since a compound of the invention can inhibit factor Vila activity, such a compound can in general be useful for reducing or inhibiting blood clotting in an individual. As used herein, the term "individual" means a vertebrate, including a mammal such as a human, in which factor Vila is involved in the clotting cascade.
Blood clotting in an individual can be reduced or inhibited by administering to the individual a therapeutically effective amount of a compound of the invention. As used herein, the term "therapeutically effective amount" means the dose of a compound that must be administered to an individual in order to inhibit factor Vila activity in the individual. More specifically, a therapeutically effective amount of a compound of the invention inhibits factor Vila catalytic activity either directly, within the

prothrombinase complex or as a soluble subunit, or indirectly, by inhibiting the assembly of factor Vila into the prothrombinase complex. Preferred compounds can inhibit factor Vila activity with aKi in the practice of a therapeutic method of the invention, the particular dosage to obtain a therapeutically effective amount of a pharmaceutical composition to be administered to the individual will depend on a variety of considerations, including, for example, the nature or severity of the disease, the schedule of administration and the age and physical characteristics of the individual. An appropriate dosage can be established using clinical approaches well known in the medical art. Thus, the invention provides a method of specifically inhibiting factor Vila activity by contacting factor Vila with a compound having the formula R1~A-B-D-En-R2. The invention further provides a method of reducing or inhibiting the formation of a blood clot in an individual by administering a therapeutically effective amount of a compound of the invention.
A compound of the invention generally will be administered to an individual as a composition containing one or more compounds of the formula I and a pharmaceutically acceptable carrier. The term "pharmaceutically acceptable carrier" refers to a medium or compositton that is non-toxic to an individual or has acceptable toxicity as determined by the appropriate regulatory agency. As used herein, the term pharmaceutically acceptable carrier encompasses any of the standard pharmaceutical carriers comprising solid carrier substances like corn starch, lactose, fats, waxes, etc., or liquids such as, for example, phosphate buffered saline, water, emulsions such as oil/water or water/oil emulsions, and/or usual additives, for example any of various types of wetting agents. Suitable pharmaceutical carriers and their formulations are described by Martin in Remington"s Pharmaceutical i Sciences, 15th ed. (Mack Publishing Co., Easton, 1975) which is incorporated herein by reference. Such compositions will, in general, contain a therapeutically effective

amount of a compound of the invention together with a suitable amount of carrier so as to comprise the proper dosage for administration to an individual. Thus, the claimed compounds can be useful as medicaments for inhibiting factor Vila activity and blood clotting in an individual.
The pharmaceutical compositions or medicaments of the invention can be administered orally, for example in the form of pills, tablets, lacquered tablets, coated tablets, granules, hard and soft gelatin capsules, solutions, syrups, emulsions, suspensions or aerosol mixtures. Administration, however, can also be carried out rectally, for example in the form of suppositories, or parenterally, for example intravenously, intramuscularly or subcutaneously, in the form of injection solutions or infusion solutions, microcapsules, implants or rods, or percutaneously or topically, for example in the form of ointments, solutions or tinctures, or in other ways, for example in the form of aerosols or nasal sprays. The amount of the active ngredient of the formula I or its pharmaceuticafly acceptable salt or derivative in a unit dose of a pharmaceutical composition usually is from about 0.5 mg to about 1000 mg, preferably from about 1 mg to about 500 mg, but depending on the type of the pharmaceutical composition the amount may also be higher. The daily dose of the compounds of the formula I that is to be administered can be a single daily dose or can be divided into several, for example, two, three or four, part administrations.
Pharmaceutically acceptable carriers also can include, for example, other mediums, compounds or modifications to a factor Vila inhibitor compound of the formula I that enhances its pharmacological function. A pharmaceutically acceptable medium can include, for example, a pharmaceutically acceptable salt. An acid addition salt of a compound of the formula I can be formed, for example, with an inorganic acid such as hydrochforic acid, hydrobromic acid, phosphoric acid, sulfuric acid or perchloric acid, or with an organic carboxylic acid such as acetic acid, oxalic acid, maleic acid, malic acid, formic acid, lactic acid, tartaric acid, citric acid, succinic acid or malonic acid, or a organic sulfonic acid such as methanesulfonic acid or p-to!uenesulfonic acid. An acid group in a compound of the formula I, for example a carboxylic acid

group, can be present as a metal salt the cation of which is based on the alkali and alkaline earth metals such as sodium, lithium, potassium, calcium or magnesium, or as well as a non-toxic ammonium salt including quaternary ammonium sa\ts and acid addition salts with amines, for example as ammonium, methylammonium, dimeihylarnmoniurn, trimethylammonium, tetramethylammonium, ethylammonium, thethylammonium ortetraethylammonium salt.
Examples of modifications that enhance the pharmacological function of the compound include, for example, esterification such as the formation of (Ci-C6)-a!kyl esters, preferably (Ci-C^)-alkyl esters, wherein the alkyl group is a straight or branched chain. Other acceptable esters include, for example, (C5-C7)-cycloalkyl esters and arylalkyl esters such as benzyl esters. Such esters can be prepared from the compounds described herein using conventional methods well known in the art of peptide chemistry.
Pharmaceutical^ acceptable modifications also can include, for example, the formation of peptide amides. Such amide modifications, which can be effected upon the compounds of the invention, include, for example, those derived from ammonia, primary (Ci-C6)-alkylamines and secondary di-{Ci-C6)-alkylamines, where the alkyl groups are straight or branched chain, or arylamines having various substitutions. In the case of secondary amines, the amine also can be in the form of a 5- or 6-membered heterocycie which can contain an unsubstituted or substituted nitrogen atom, an oxygen atom or a sulfur atom in addition to the amide nitrogen atom. Methods for preparing such amides are well known in the art.
In another embodiment of the invention, a compound of the invention can be used in an assay to identify the presence of factor Vila or to isolate factor Vila in a substantially purified form. Preferably, the compound of the invention is labeled with, for example, a radioisotope, and the labeled compound is detected using a routine method useful for detecting the particular label. In addition, a compound of the

invention can be used advantageously as a probe to detect the iocation or amount of factor Vila activity in vivo, in vitro or ex vivo.
It is understood that modifications that do not substantially affect the activity of the various embodiments of this invention are included within the invention disclosed herein. Accordingly, the following examples are intended to illustrate but not limit the present invention.
Example 1; Peptide synthesis procedures and general synthesis procedures
Starting materials used in the synthesis were obtained from chemical vendors such as Aldrich, Sigma, Fluka, Nova Biochem and Advanced Cherntech. During the synthesis, the functional groups of the amino acid derivatives used were protected by blocking groups to prevent side reaction during the coupling steps. Examples of suitable protecting groups and their use are described in The Peptides, supra, 1981, and in vol. 9, Udenfriend and Meienhofer (eds.), 1987, which is incorporated herein by reference.
General solid-phase peptide synthesis was used to produce the compounds of the invention. Such methods are described, for example, by Steward and Young, Solid Phase Peptide Synthesis (Freeman & Co., San Francisco, 1969), which is incorporated herein by reference.
Unless indicated otherwise, peptides were synthesized on TentaGel S NH2 Resin (Rapp Polymere, Tubingen, Germany). An acid sensitive linker p-[(R,S)-a-[H9H-fluoren-9-yl)methoxyformamido]-2,4-dimethoxybenzyllphenoxyacetic acid (Knorr Linker) was coupled to the solid support (Bematowicz et. al, Tetr. Lett. 30 (1989) 4645, which is incorporated herein by reference). Alternatively, peptides were synthesized on polystyrene resin cross-linked with 1 % divinylbenzene modified with an acid sensitive tinker (Rink resin) (Rink, Tetr. Lett. 28 (1987) 3787; Sieber, Tetr.

Lett. 28 (1987) 2107, each of which is incoporated herein by reference). When peptides were synthesized by first coupling the side chain carboxylic acid of a compound of the formula Fmoc-B1-CHR97-C(0)OPG to the resin, TeniaGe! S NH2 resin modified by attachment cf the HMPA linker was employed. Coupling was performed using N^"-diisopropyicarbodnmide (DlC) in the presence of an equivalent amount of HOBt, with the exception of AI!oc-pAph-OK where 2 equivalents (eq.) of HOBt were used. All couplings were done in either N,N-dimethylformamide (DMF) or DMF/DMSO (1/1 mixture) at room temperature (RT). Completion of coupling was monitored by ninhydrin test. A second (double) coupling was performed where coupling in the first instance was incomplete.
Deprotection of the Fmoc group was accomplished using 50% pipehdine in DMF for 2+10 min. The amount of Fmoc released was determined from the absorbance at 300 nm of the solution after deprotection, volume of washes and weight of the resin jsed in the synthesis.
The cycle of each coupling was as follows;

Step Action/Reagent Solvent
1. 0.5 g of functionalized peptide resin
2. 3 fold-excess of amino acid
derivative/HOBt/DIC 4 ml DMF
3. Couple (min. 1 h)
4. Wash (3x5 ml) DMF
5. Ninhydrin test
6. Deprotection (2+10 min)
Piperidine/DMF 5 ml 50%
7. Wash (6x5 ml) DMF
8. Repeat starting at step 2

After completion of peptide assembly on the resin, the final Fmoc deprotection, if necessary, was performed. The peptide resin was then washed successively with DMF and DCM and the peptide was then cleaved and deprotected by a mixture TFA/thioanisole (95/5) for 1.5 hours, unless specified otherwise. The resin was washed with DCM and the DCM wash combined with the TFA releasate. The solution was evaporated, the product precipitated by anhydrous diethyl ether and the solid precipitate was isolated by filtration or centrifugation and dried in vacuo over pellets of solid KOH. The solid was redissoived in a mixture of water and acetonitriie and lyophylized.
The dried peptide was subjected to HPLC purification using an appropriate gradient of 0.1 % TFA Jn water and acetonitriie (ACN). After collecting the peak containing the intended synthetic product, the peptide solution was lyophilized and the peptide was subjected to an identification process, which included electrospray mass spectrum (MS) and/or NMR and/or amino acid analysis to confirm that the correct compound was synthesized:
For HPLC analysis, a sample of the product was analyzed using Beckman HPLC system (consisting of 126 Solvent Deliver System, 166 Programmable Detector Module 507e Autosampier, controlled by Data Station with Gold Nouveau software) and YMC ODS-AM 4.6x250 mm column at 230 nm and flow rate 1ml/min.
For product purification, a sample of crude lyophilized peptide was dissolved in a mixture of 0.1% aqueous TFA containing 10% to 50% ACN. The peptide solution usually was filtered through a syringe connected to a 0.45 jxm "ACRODISC" 13 CR PTFE (Gelman Sciences; Ann Arbor Ml) filter. A proper volume of filtered peptide solution was injected into a semi-preparative C18 column {Vydac Protein and Peptide C18, 218TP1022 (22x250 mm); The Separation Group; Hesperia CA, or YMC ODS-A column {20x250 mm), YMC, Inc., Wilmington, NC). The flow rate of a 1 gradient or isocratic mixture of 0.1% TFA buffer and ACN (HPLC grade) as an eluent was maintained using a Beckman "SYSTEM GOLD" HPLC (Beckman, System Gold,

Programmable Solvent Module 126 and Programmable Detector Module 166 controlled by "SYSTEM GOLD" software), Elution of the peptide was monitored by UV detection at 230 nm. After identifying the peak corresponding to the compound under synthesis using MS, the compound was collected, lyophiiized and biologically tested. MS was performed using a VG Platform (Fisons Instruments) instrument in ES+ mode. For NMR, typically samples were measured in DMSO-d6 (Aldrich) using a Bruker Avance DPX 300 instrument.
Example 2: Synthesis of Ailoc-pAph-OH
The same procedure is applicable to Aiioc-D-pAph-OH.
Alloc-Phe(4-CN)-OH
5.7 g (30 mmol) of H-Phe(4-CN)-OH were dissolved in 100 ml of 1M NaOH with addition of 2M NaOH to pH=10 with ice cooling. With vigorous stirring, ailyl chloroformate (7.5 ml) was slowly added (pH kept at 10 by 2M NaOH). The reaction mixture was stirred at 0°C for 15 min and at RT for 30 min, acidified with HC! to pH = 2, extracted with ethyl acetate (3 times), dried with MgS04 and evaporated. The residue was recrystallized from ethyl acetate/hexane to give a white solid. Yield; 7.0 g (85%).
AI(oc-Phe[4-C(=S)-NH2]-OH
2.74 g of Alioc-Phe(4-CN)-OH was dissolved in mixture a of pyridine (50 ml) and triethylamine (20 ml) and H2S was passed through for 30 min. Reaction mixture was kept overnight at RT and evaporated. Drying on high vacuum gave 3.21 g of a solid foam of the crude thioamide, which is directly converted to the methylthioimidate.
Alloc-Phe[4-C(=NH)-SCH3l-OH" HI

1 g of Alloc-Phe[4-C(=S)-NH2]-OH was dissolved in acetone (50 ml) and methyl iodide (5 ml) was added. The reaction mixture was kept overnight at RT, volatile solvents evaporated (fast, 35°C max.) and the residue treated with diethyl ether. After 1 hour at 0°C, the ether was decanted, the product washed with diethyl ether and dried in vacuo. A yellow solid foam was obtained which was directly converted into the amidine.
Alloc~pAph-OH
All of the Alioc-Phe(4-C(=NH)-SCH3)-OH ■ HI above was dissolved in 50 ml methanol with 300 pi of acetic acid, and 0.5 g of ammonium acetate were added. The mixture was heated for 3 hours to 55*C, evaporated and 10 ml of acetone was added. After 2 hours at 0°C, the solid product was filtered, washed with a little cold acetone, a little cold methanol and diethyl ether and dried in vacuo to give a yellowish solid. Yield; 0.53 g.
Example 3: Synthesis of AI!oc-pAph-Glu-Arg-Cha-NH2
To 1 g of TentaGel S NH2 resin (substitution 0.26 mmol/g), Knorr amide linker was attached. According to the general procedures outlined in Example 1, the following protected amino acids were coupled: Fmoc-Cha-OH, Fmoc-Arg(Pmc)-QH, Fmoc-Glu(OtBu)-OH and Alloc-pAph. The peptide was cleaved and deprotected by TFA/thioanisole (95/5) for 3 hours and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+: found 729.1, calc. 729.4.
Example 4: Synthesis of allyl-NH-C(0)-pApn-Glu-Arg-Cha-NH2
To 0.5 g of TentaGel S NH2 resin (substitution 0.26 mmol/g), Knorr amide linker was attached. According to the general procedures in Example 1, the following protected amino acids were coupled: Fmoc-Cha-OH, Fmoc-Arg(Pmc)-OH,

Fmoc-Glu(OtBu)-OH and Fmoc-Phe(4-CN). After N-terminal Fmoc deprotection, the resin was treated with a solution of 1 mmo! of allyl isocyanate in 3 ml of DMF for 2 hours. The resin was then washed with DMF and triethylarnine/pyridine (1/2) and treated with a saturated solution of H2S in pyridine/triethylamine overnight. The resin was washed with acetone and the thioamide resin was reacted with methyl iodide [3 ml of 10% solution of methyl iodide in acetone) for 6 hours. The methylthioimidate resin was washed with acetone, methanol and treated with solution of 0.2 g ammonium acetate, 100 pi acetic acid in 3 ml of methanol at 55°C for 3 hours. The resin was washed with methanol, DMF and DCM and the peptide was cleaved and deprotected by TFA/thioanisole (95/5) for 3 hours and processed as described in Example 1. The crude material was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+; found 728.3, calc. 728.4.
Example 5: Synthesis of AIIoc-pAph-Glu-Arg-Chg-NH2
To 1 g of TentaGef S NH2 resin (substitution 0.26 mmol/g), Knorr amide linker was attached. According to the general procedures in Example 1, the following protected amino acids were coupled: Frnoc-Chg-OH, Fmoc-Arg(Pmc)-OH, Fmoc-Glu(OtBu)-OH and Alloc-pAph, The peptide was cleaved and deprotected by TFA/thioanisole (95/5) for 3 hours and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1"and characterized by MS. (M+H)+: found 715.8, calc. 715.4.
Example 6: Synthesis of Alloc-D-pAph-Giu-Arg-Cha-NH2
To 1 g of TentaGel S NH2 resin (substitution 0.26 mmol/g), Knorr amide linker was attached. According to the general procedures in Example 1, the following protected amino acids were coupled: Fmoc-Cha-GH, Fmoc-Arg(Pmc)-OH, Fmoc-Glu{OtBu)-OH and Alloc-D-pAph-OH (synthesized, according to the same procedure as Alfoc-pAph-OH in Example 2). The peptide was cleaved and. • deprotected by TFA/thioanisole (95/5) for 3 hours and processed as described in

Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+Hf: found 729.2, calc. 729.4.
Example 7: Synthesis of Aiioc-pAph-GJu-Phe(4-guanidino)-Cha-NH2
To 0.25 g of TentaGel S NH2 resin (substitution 0.23 mmol/g), Knorr amide linker was attached. According to the general procedures in Example 1, the following protected amino acids were coupled: Fmoc-Cha-OH,
Fmoc-Phe{4-NH-C{=NBoc)-NH-Boc)-OH, Fmoc-Giu(OtBu)-OH and Alloc-pAph-OH. The peptide was cleaved and deprotected by TFMhioanisofe (95/5) for 1 hour and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+: found 777.1, calc. 777.4.
Example 8: Synthesis of Alioc-pAph-Glu-Dap[-C(=NH)-NH2]-Cha-NH2
To 0.25 g of TentaGel S NH2 resin (substitution 0.23 mmol/g), Knorr amide linker was attached. According to the general procedures in Example 1, the following protected amino acids were coupled; Fmoc-Cha-OH,
Fmoc-Dap[-C(=N~Boc)-NH-Boc]-OH, Fmoc-Glu(OtBu)-OH and AIloc-pAph-OH. The peptide was cleaved and deprotected by TFMhioanisole (95/5) for 1 hour and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+: found 729.1, calc. 729.4.
Example 9: Synthesis of Alloc-pAph-Glu-Dap[-C(=NH)-CH3]-Cha-NH2
To 0.25 g of TentaGel S NH2 resin (substitution 0.26 mmol/g), Knorr amide linker was attached. According to the general procedures in Example 1, the following protected amino acids were coupied: Fmoc-Cha-OH, Fmoc-Dap(AJ)oc)-OH and Fmoc-Glu(OtBu)~OH. With the N-terminal Fmoc-protecting group attached, the resin

was washed with a DMF/NMM/AcOH (5/0.5/1) mixture, and under constant mixing with a stream of argon, the Alloc group was deprotected by addition of 100 mg of Pd(P(Ph)3)4 over a period of 3 hours. The resin was washed with DMF and treated *ith solution of 150 mg of 2-methylnaphthyl acetthroimidate in 4 ml of ethanol/DMSO (3/1) for 1 hour. After washing with DMF, the Fmoc group was deprotected (1+5 min) and the N-terminal Ailoc-pAph-OH was coupled. The peptide was cleaved and deprotected by TFA/thioaniso!e (95/5) for 1 hour and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. {M+H)+: found 700.1, ca)c. 700.4.
Example 10: Synthesis of Ailoc-pAph-Giu-A!a[3-C(=NH)-NH2]-Cha-NH2
To 0.25 g of TentaGe! S NH2 resin (substitution 0.26 mmol/g), Knorr amide linker was attached. According to the general procedures in Example 1, the following protected amino acids were coupled: Fmoc-Cha-OH, Fmoc-AIa(3-CN)-OH, Fmoc-Glu(OtBu)-OH and Alloc-Phe(4-CN)-OH. A mixture of pyridine and triethylamine (2/1) was saturated with H2S (RT, 15-30 min) and this solution added to the resin prewashed with pyridine/thethylamine (2/1). After standing overnight, the resin was washed with acetone and treated with a solution of 20% methyl iodide in acetone overnight. The resin was then washed with acetone and methanol. The resin bound methylthioimidate was then converted into the amidine by heating {waterbaih, 55°C; 3 hours) of the resin with a solution of 10 eq. of ammonium acetate in methanol containing 5% acetic acid. After this final conversion, the resin was washed with methanol, DMF, DCM. The peptide was cleaved and deprotected by TFA/thioanisole (95/5) for 1 hour and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+: found 685.9, calc. 686.4.
Example 11: Synthesis of Ailoc-pAph-Glu-Asn-Cha-NH2

To 0.125 g of Rink resin (substitution 0.78 mmol/g}, after Frnoc-deprotection the following protected amino acids were coupled according to the general procedures described in Example 1: Fmoc-Cha-OH, Fmoc-Asn-OH, Fmoc-Glu(OtBu)-OH and AIJoc-pAph-OH, The peptide was cleaved and deprotected by TFA/thioanisole (95/5.) for 1 hour and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+: found 686.9, calc. 687.3
Example 12: Synthesis of Aiioc-pAph-GJu-Dab-Cha-NH2
To 0.25 g of TentaGel S NH2 resin (substitution 0.26 mmol/g), Knorr amide linker was attached. According to the general procedures in Example 1, the following protected amino acids were coupled: Fmoc-Cha~OH, Fmoc-Dab(Boc)-OH, Fmoc-Glu(OtBu)-OH and Alloc-pAph, The peptide was cleaved and deprotected by TFA/thioanisole (95/5) for 1 hour and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+: found 673.2, calc. 673.4.
Example 13: Synthesis of Allac-pAph-Glu-Ala[3-C(=NH}-NH23-NH2
To 0.25 g of TentaGel S NH2 resin (substitution 0.26 mmol/g), Knorr amide linker was attached. According to the general procedures in Example 1, the following protected amino acids were coupled: Fmoc-Ala(3-CN)-OH, Fmoc-Glu(OtBu)-OH and A(Soc-Phe(4-CN)-OH. A mixture of pyridine and tnethylamine (2/1) was saturated with H2S (RT, 15-30 min) and this solution added to the resin prewashed with pyridine/triethylamine (2/1). After standing overnight, the resin was washed with acetone and treated with a solution of 20% methyl iodide in acetone overnight. The resin was then washed with acetone and methanol. The resin bound methyithioimidate was then converted into the amidine by heating (55°C, waterbath, 3 hours) of the resin with solution of 10 eq. of ammonium acetate in methanol containing 5% acetic acid. After this final conversion, the resin was washed with

methanoi, DMF, DCM. The peptide was cleaved and deprotected by TFMhioanisole (95/5) for 1 hour and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+: found 533.3, caic. 533.2.
Example 14: Synthesis of Alloc-pAph-Glu-Gly-Cha~NH2
To 0.150 g of Rink resin (substitution 0.78 mmol/g), after Fmoc-deprotection, the following protected amino acids were coupled according to the genera) procedure described in Example 1: Fmoc-Cha-OH, Fmoc-Gly-OH, Fmoc-G!u(OtBu)-OH and Alloc-pAph-OH. The peptide v/as cleaved and deprotected by TFA/thioanisole (95/5) for 1 hour and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+: found 630.1, calc. 630.3.
Example 15: Synthesis of Alloc-pAph-Glu-Asn-(Ph-CH2-CH2-)Gly-NH2
For N-substituted glycines, the procedure of Zuckermann et al. (J. Am. Chem. Soc. 114 (1992) 10646, which is incorporated herein by reference) was used. To 0.1 g of Rink resin (substitution 0.78 mmol/g), after Fmoc-deprotection, bromoacetic acid was coupled via the symmetrical anhydride in DCM/DMF. After 10 minutes, the resin was washed with DCM and the coupling repeated once more. After washing with DCM and DMF, the resin was treated with a 1M solution of 2-phenylethylamine in DMSO overnight. After washing with DMF, the resin now carrying the residue (Ph-CH2-CH2-}NH-CH2-C(0) attached to the linker was reacted with the symmetrical anhydride of Fmoc-Asn(Trt)-OH in DCM/DMF. After Fmoc-deprotection, according to the the general procedures in Example 1, the following protected amino acids were coupled: Fmoc-Glu(OtBu)-OH and AJJoc-pAph-OH. The peptide was cleaved and deprotected by TFA/thioanisole (95/5) for 1 hour and processed as described in Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+; found 694.9, calc, 695.3

Example 16: Synthesis of Alloc-pAph-GIu-Thr(Bzl)-NH-CH2-CH2-CH{Ph)2 H-Thr(Bzl)-NH-CH2-CH2-CH(Ph)2" HCI
0.62 g (2 mmol) of Boc-Thr(Bzt)-OH were dissolved in 10 ml DCM, 2 mmol of tri ethyl amine were added and the solution was cooled to 0°C. With stirring, 2 mmo! of isobutyl chioroformate were slowly added. The cooling bath was removed, the solution was stirred for 15 minutes and 2.5 mmoi of 3,3-diphenylpropylamine in 2 ml of DMF were added and stirred at room temperature for 1 hour. The solution was evaporated, dissolved in ethyl acetate and extracted with 0.5M KHS04 solution, sat. NaHC03 solution and brine, dried with MgS04 and evaporated. The ojjy product was dissolved in 10 ml of DCM, and 10 ml of a 4M solution of hydrochloric acid in dioxane were added. After 10 minutes the solvents were evaporated, the product hydrochloride was precipitated with diethyl ether, filtered off, washed with diethyl ether and dried in vacuo to give a white solid. MS analysis: (M+H)+: found 403.1, calc. 403.2.
Afloc-pAph~Glu-Thr(Bz!)-NH-CH2-CH2-CH(Ph}2
To 0.5 g of TentaGel S NH2 resin {substitution 0.26 mmol/g), 4-hydroxymethylphenoxyacetic acid was attached (3 eq., activated with DIC/HOBt for 1.5 h). Fmoc-Glu(OH)-0-ally! was attached to the resin via side chain using DlC/HOBt/NMl in DMF overnight. The allyl protecting group was removed by shaking the resin with Pd(PPh3)4 in DMF/AcOH/NMM (10/2/1) for 4 h under argon. The deprotected carboxy group was activated with a solution of 0.5 mmol BOP, 0,5 mmoi HOBt, 1.5 mmol DIEA and 0.5 mmol of of H-Thr(Bzl)-NH-CH2-CH2-CH(Ph)2 ■ HCI in 1.5 ml of DMF for 2 hours. After Fmoc deprotection, Alloc-pAph-OH was coupled according to the general procedure in Example 1. The peptide was cleaved and deprotected by TFA/thioanisole (95/5) for 1.5 hours and processed as described in

Example 1. The crude compound was purified using HPLC as described in Example 1 and characterized by MS. (M+Hf: found 805.0, caic. 805.4.
Example 17: Synthesis of A!loc-pAph-Glu-Dab-NH-CH2-CH2-Ph
To 0.2 g of TentaGel S NH2 resin (substitution 0.26 mmoi/g), 4-hydroxymethylphenoxyacetic acid was attached (2.5 eq., activated with DIC/HOBt for 4 h). The hydroxy group was substituted with bromine by treatment of the resin with CBr4 (5 eq.)/PPh3 (5 eq.) in DCM for 4 h. The bromine derivatized resin was treated with a 2M solution of phenyl ethyl amine in DCM overnight. Fmoc~Dab(Boc)-OH was coupled to the resin using TFFH/DIEA (acyl fluoride generated in situ). According to the general procedure in Example 1, the following protected amino acids were coupled: Fmoc-Glu(OtBu)-OH and Alioc-pAph-OH. The peptide was cleaved and deprotected with TFA/triisopropyisiiane (99/1) for 2 h. TFAwas evaporated, the peptide was dissolved in H20/ACN and lyophilized. The crude material was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+ : found 624.2, catc. 624.3.
Example 18: Synthesis of Alloc-pAph-Glu-NH-CH2-CH2-CN
To 0.2 g of TentaGel S NH2 resin (substitution 0.26 mmol/g), 4-hydroxymethylphenoxyacetic acid was attached (3 eq., activated with DIC/HOBt for 1.5 h). Fmoc-G)u(OH)-0-allyI was attached to the resin via the side chain using DIC/HOBt/NMi in DMF overnight. The allyl protecting group was removed by shaking the resin with Pd(PPh3)4 in DMF/AcOH/NMM (1072/1) for 4 h under argon. The deprotected carboxy group was activated with DIG (3 eq.)/HOBt (3 eq.) for 10 min and 2-cyanoethylamine (3 eq.) in DMF was added to the resin for 3 h. After Fmoc deprotection, Alioc-pAph-OH was coupled according to the general procedure in Example 1. The peptide was cleaved and deprotected with TFA/triisopropylsiiane (99/1) for 2 h. TFA was evaporated, the peptide was dissolved in H20/ACN and

lyophiiized. The crude material was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+: found 473.1, calc. 473.2.
Example 19: Synthesis of Alioc-pAph-Giu-Asn-NH~CH2-Chx
To 0.1 g of TentaGel S NH2 (substitution 0.26 mmot/g) Knorr amide linker was attached. Fmoc-Asp(OH)-0-allyl was coupled to the linker via the side chain and the allyl protecting group was removed as in Example 18. The deprotected carboxy group was activated with DIC (5 eq.)/HOBt (5 eq.) and cyclohexylmethylamine (5 eq.) in DMF was added ior 2.5 h. After Fmoc deprotection, Frnoc-G!u(OtBu)-OH and Alloc-pAph-OH were coupled according to the general procedure in Example 1. The peptide was cleaved and deprotected with TFA/tn"isopropyls"tlane (99/1) for 2 h. TFA was evaporated, the peptide was dissolved in H20/ACN and lyophiiized. The crude material was purified using HPLC as described in Example 1 and characterized by MS. (M+H)+ : found 629.9, calc. 630.3.
Example 20: Synthesis of Alloc-pAph~Glu-Asn-NH~CH2-CH2-Ph
2-(SH2-(S)-Allyloxycarbonylamino-3-{4-carbamimidoyl-phenyl)-propionylamino]-pentanedioic acid 5-tert-butyl ester 1-methyl ester hydrochloride
To 2-(S)-allyloxycarbonylamino~3-(4-carbamimidoyl-phenyl)-propionicacid hydrochloride (3.48 g, 10.6 mmol) and 2-(S)-amino-pentanedioic acid 5-tert-butyl ester 1-methyl ester hydrochloride {2.7 g, 10.6 mmol) in 20 mi of DMF were added at -15°C TOTU {3.83 g, 11.67 mmol) and N-ethylmorpholine (2.7 ml, 21.2 mmol). The mixture was stirred for 1 hour and then allowed to warm to room temperature. After evaporation ethyl acetate was added to the residue and the organic layer was extracted with aqueous sodium hydrogen carbonate solution, potassium hydrogen sulfate solution and water. The organic layer was evaporated. Yield: 2.8 g (50%). MS: m/z = 491.3 (M+H)+.

2-(S)-[2-(S)-Allyloxycarbonylarriino-3-(4-carbarnimido"yl-phenyl)-proptonylamino]-
pentanedioic acid 5-tert-butyl ester
To2-(S)"[2-(S}-allyloxycarbonylamino-3-(4-carbarnirnidoyl-phenyl}-propionyiarnino]-pentanedioic acid 5-tert-butyl ester 1-methyl ester hydrochloride (3.06 g, 5.8 mmol) in 100 m! of water and 30 ml of THF was added lithium hydroxide hydrate (0.49 g, 11.6 mmoi). The solution was stirred at room temperature for 12 hours, evaporated and freeze-dried. The residue was purified by chromatography on Sephadex LH20 employing n-butanol/glacial acetic acid/water (17/1/2) as eiuent. Pure fractions were combined. The solvent was evaporated, the residue was taken up in water and the aqueous solution was freeze-dried. Yield: 2.7 g (97%). MS: m/z = 477.4 (M+H)+.
4-(S)-[2-(S)-Allyloxycarbonylamino-3-(4-carbamimidoyl-phenyl)-propionylamino]-4-(2-carbamoyl-1-(S)-(2-phenylethylcarbamoyl)-ethyicarbamoy|)-butyricacid hydrochloride (Alloc-pAph-G!u~Asn-NH-CH2-CH2-Ph)
To 2-(S)-[2-(S)-aliyioxycarbonylamino-3-(4-carbamimidoyl-phenyl)-propionylamino]-pentanedioic acid 5-tert-butyl ester (48 mg, 0.1 mmol) and2-(S)-amino-N1-phenyleihyl-succinamide hydrochloride (27 mg, 0.1 mmol) in 5 ml of DMF were added at Q°C HATU (39 mg, 0.1 mmol) and coUidme {24.2 mg, 0.2 mmol). The mixture was stirred for 1 hour and then allowed to warm to room temperature. After evaporation the residue was purified by chromatography on Sephadex LH20 employing n-butanol/glacial acetic acid/water (17/1/2) as eiuent. Pure fractions were combined. The solvent was evaporated, the residue was taken up in water and the ■ aequous solution was freeze-dried. Yield: 45 mg (66%). MS: m/z = 638.4 (M+H)+.
Example 21: Synthesis of Alloc-pAph-Glu-Asn-NH-(3-chlarobenzyl)
To 2-(S)-[2-(S)-atlyloxycarbonylamino-3-(4-carbamimidoyl-phenyl)-propionylamino]-) pentanedioic acid 5- tert-butyl ester (50 mg, 0.105 mmol) and 2-(S)-amino-N1-(3-
chlorobenzyi)succinamide trifluoroacetate (61 mg, 0.16 mmol) in 5 mi of DMF were

added at 0°C TOTU (36 mg, 0.11 mmol) and N-ethyimorpholine (57 pi, 0-4 mmol). The mixture was stirred for 1 hour and then allowed to warm to room temperature. After evaporation the residue was purified by chromatography on Sephadex LH20 employing n-butanol/glacial acetic acid/water (17/1/2) as eluent. Pure fractions were combined. The solvent was evaporated, the residue was taken up in water and the aqueous solution was freeze-dried. Yield of 4-(S)-[2-(S)-al!yloxycarbonylamino-3-(4-carbamimidoyl-pheny!)-propionylamino]-4-(2-carbamoy!-1-(SH3-chlorobenzylcarbamoyl)-ethylcarbamoyl)-butyricacid (Alloc-pAph-Giu~Asn-NH-(3-chlorobenzyl or Alloc-pAph-Glu-Asn-3-chlorobenzylamide): 28 mg (41%). MS:m/z = 558.3 (M+H)+.
Further example compounds prepared analogously to the above examples are listed in Table 2 above.
Example 22: Determination of Ki for FVIia inhibition
The inhibitory activity (Ki) of each compound towards factor Vita/tissue factor activity was determined using a chromogenic assay essentially as described previously (J.A. Ostrem, F. Al-Obeidi, P. Safar, A. Safarova, S.K. Stringer, M. Patek, M.T. Cross, J. Spoonamore, J.C. LoCascio, P. Kasireddy, D.S. Thorpe, N. Sepetov, M. Lebl, P. Wildgoose, P. Strop, Discovery of a novel, potent, and specific family of factor Xa inhibitors via combinatorial chemistry. Biochemistry 37 (1998) 1053-1059). Kinetic assays were conducted at 25 *C in naif-area microtiter plates (Costar Corp., Cambridge, MA) using a kinetic plate reader (Molecular Devices Spectramax 250). A typical assay consisted of 25 p! of human factor Vila and TF (5 nM and 10 nM, respective final concentration) combined with 40 pi of inhibitor dilutions in 10% DMSO/TBS-PEG buffer (50 mM Tris, 15 mM NaC!T 5 mM CaCI2; 0.05% PEG 8k, pH 8.15). Following a 15 minute preincubation period, the assay was initiated by the addition of 35 pi of the chromogenic substrate S-2288 (D-lle-Pro-Arg-pNA, Pharmacia Hepar Inc. 500 uM final concentration.). The apparent inhibition constants were calculated from the slope of the progress curves during the linear

part of the time course, typically between 1 and 5 min following addition of substrate to the assay. The true Ki was subsequently determined for each compound by correcting for substrate concentration (S) and the Km using the formula Ki= Ki app/ (1 + (S)/Km) (I.H. Segal, Enzyme Kinetics, pp 100-125 (John Wiiey & Sons, New York, 1975)).


WE CLAIM :
1. A compound of the formula I,
R1-A-B-D-En-R2 (!)
wherefn
R1 is aftyioxycarbonyl or allylaminocarbonyl,
A is the group At-A2-A3, wherein
A1 is NH,
A2 is CHR93r wherein R93 is 4-amidinophenylmethyi,
A3 is C(O),
B is the group B1-B2-B3, wherein
B1 isNH,
B2 is CHR97, wherein R97 is ethyi which is substituted in the 2-position by a substituent selected from the series consisting of hydroxycarbonyl, alkyloxycarbonyl and arylalkyloxycarbonyl,
B3 is C(O),
D is the group D1-D2-D3, wherein
D1 is NH,
D2 is CHR82, wherein R82 is selected from the series consisting of hydrogen and the unsubstituted or substituted residues (CrCe)-aikyl, phenyl, phenyl-(Ci-C2)-alkyl and heteroaryl-(C1-C2)-aikyi3 where the residues representing RB2 can be substituted with one, two or three identical or different substitue"nts selected from

the series consisting of amino, aminocarbonyl, amidino, gua-nidino, aminoalkyJ, hydroxy, mercapto, which can all be substi¬tuted with a protecting group, and acetimido, nitro and cyano, D3 is C(0)f
En is (E1-E2-E3)n, wherein
n is zero or one,
E1 is NR7Q, wherein R70 is selected from the series consisting of hydrogen, (d-GO-alkyl and phenyl-(Ci-C4)-alkyl,
E2 is CHR72, wherein R72 is selected from the series consisting of the unsubstituted or substituted residues alkyl, phenyl, phenyl-(Ci-C2)-alkyl and heteroaryl-(Ci-C2>all yi,
E3 is C(O),
R2 is NR21R22, wherein R21 is selected from the series consisting of hy¬drogen, (Ci-C4)-alkyl and phenyl-(Ci-C4)-alkyi which is unsubstituted or substituted in the phenyl group, and R22 is selected from the series consisting of hydrogen and the unsubstituted or substituted residues al¬kyl, aryl, arylalkyf and heteroarylalkyl, where the residues representing R21 and R22 can independently be substituted with one, two or three identical or different substituents selected from the series consisting of halogen, trifluoromethyl, hydroxy, nitro, cyano, alkyloxy, alkylenedioxy, dialkylamino, alkyjsulfonyl, aminosulfonyl and =0,

alkyl, unless stated otherwise, has 1 to 6 carbon atoms in the case of satu¬rated linear or branched alkyl chains, 2 to 6 carbon atoms m the case of un¬saturated linear or branched alkenyl and alkynyl chains, and 3 to 8 carbon at¬oms in the case of cyclic alkyl groups,
aryl and heteroaryl contain 5 to 13 ring carbon atoms where in a heteroaryl: re¬sidue one or two carbon atoms are replaced with heteroatoms selected from the series consisting of N, O an6 Sr;
in any of its stereoisomer^ forms or a mixture thereof in any ratio, or a phar-maceutically acceptabJe salt thereof.
2. The compound as claimed in claim 1, wherein n is zero or one and R2 is NHR22, wherein R22 is as defined.
3. The compound as claimed in claim 1 to 2, wherein A is the residue of (L)-4-amidinophenylalanine.
4. The compound as claimed in one or more of claims 1 to 3, wherein B is the residue of (L)-glutamic acid or a pharmaceutically acceptable salt or ester thereof
5. The compound as claimed in one or more of claims 1 to 4, wherein D is a residue selected from the series consisting of Arg, Dap, Dab, Orn, Lys, Dap[-C(^NH)-NH2], Dab[-C(=NH)-NH2], Lys[-C(=NH)-NH2], Lys[-C(=NH)-CH3], Orn[-C(=NH)-CH3], Dab[-C(=NH)-CH3], Dap[-C(=NH)-CH3], Dab(AHoc), Asn, Gin, Met, Ser, Thr, Ser(Bzl), Thr(Bzl), Cys(Me), Cys(Bzl), Cys(Acm), Arg(N02), His, Trp, Phg, Gly, Ala, Val, lie, Leu, Phe, Phe(4-NH-C(=NH)-NH2), Phe(4-N02), 2-Abu, Ala(3-CN), Ala[3-C(=NH>NH2], 2-Abu(4-CN) and 2-Abu[4-C(=NH)-NH2].
6. The compound as claimed in one or more of claims 1 to 5, wherein E is a residue selected from the series consisting of Cha, Chg and Phe(4-C(-S-CH2-CH2-S-)-Ph].

7, The compound of iheformula 1 as claimed in one or more of claims 1 to 6,
wherein
R1 is ailyloxycarbonyl or allyiaminocarbonyl,
A is the residue of (L)-4-amidinophenylaianine,
B is the residue of (L)-glutamic acid or a pharmaceuticaily acceptable salt or ester of (L)-glutamic acid,
D is a residue seiected from the series consisting of Arg, Dap: Dab, Qrn, Lys, Dap[-C{=NH)-NH2], Dab[-C(=NH)-NH2], Lys[-C{=NH}-NH23, Lys[~C(=NH)-CH3l, Om[-C(=NH)-CH3], Dab[-C(=NH)-CH3L Dap[-C(=NH)-CH3]: Dab(Alloc), Asn; Gin, Met, Ser, Thr, Ser(Bzl), Thr(Bzi), Cys(Me), Cys(Bzl), Cys(Acm), ArgfN02}, His, Trp, Phg, Gly, Ala, Val, He, Leu, Phe, Phe(4-N02), Phe(4-NH-C(=NH)-NH2), 2-Abu, Ala(3-CN), Aia[3-C(=NH)-NH2], 2-Abu(4-CN) and 2-Abu[4-C(=NH)-NH2],
n is zero or one..
E is a residue seiected from the series consisting of Cha, Chg and Phe[4-C(-S-CH2-CH2-S-)-Ph],
R2 is NHR22,
R22 is hydrogen or a residue selected from the series consisting of benzyl, napbthyimethyl, pyridylmethyl, phenyfethyl, naphthylethy), pyridylethyl, phenylpropyl, naphthyfpropyl, pyridyipropyi, fiuorenyt, diphenylmethyf, diphenylethyl and diphenylpropyl, which residues are unsubstituted or substituted with substituents seiected from the series consisting of F, CI. Br. hydroxy, methoxy, methylenedioxy, nitro, cyano: dialkyiamino, alkyisurfony!, eminosuifonyi and tnfiuorofnethyl,
in any of its stereoisomer^ forms or a mixture thereof in any ratio, or a
pharmaceuticaily acceptable salt thereof.

S# The compound of the formula I as claimed in one or more of claims 1 to £ which is
Ailoc-pAph~Glu-Arg-Cha-NH2l Allylaminocarbony!-pAph-Glu-Arg-Cha-NH2, Alloc-pAph-Glu-Arg-Chg-NH2l A!loc-pAph-Glu-Dap[-C(=NH)-NH2]-Cha-NH2, ANoc-pAph-Glu-Ala[3-C(=NH)-NH2}-Cha-NH2j Alioc-pAph-Glu-Asn-Cha-NH2, Alloc-pAph-Glu-Dab-Cha-NH2, Alloc-pAph-Glu-Dap[-C(=NH)-NH2]-NH2l Alloc-pAph-Glu-Gly-Cha-NH2, Alloc-pAph-Glu-Thr(Bzl)-NH-(CH2)2-CH(Ph)2l Alloc-pAph-Glu~Dab-NH-(CH2)2-Ph, Alfoc-pAph-GIu-Asn-NH-CH2-Chx, Alloc-pAph-Glu-Dap[-C(=NH)-CH3]-Cha-NH2] AI!oc-pAph-G!u-Dab[-C(=NH)-NH2]-Cha-NH2, Alloc-pAph-Glu-2-Abu(4-CN)-Cha-NH2, Alloc-pAph-Glu-Ala(3-CN)-Cha-NH2, Alloc-pAph-Glu-Asn-1-naphthylmethylamide, Alloc-pAph-Glu-Asn-1-(1-naphthyl)ethylamide, Alloc-pAph-Giu-Asn-2-naphthylmethylamide, Ailoc-pAph-Glu-Asn-3,4~dichiorobenzylamide, Alloc-pAph-Glu-Asn-2-(3-chlorophenyl)ethylamide, Alloc-pAph-Glu-Arg(N02)-Cha-NH2l Alloc-pAph-G!u-Cys(Bzl)-Cha-NH2, Alloc-pAph-Glu-Trp-Cha-NH2l Affoc-pAph-Gfu-Phg-Cha-NH2l Alloc-pAph-GIu-Asn-9-fluorenylamide, Alloc-pAph-Glu-Asn-3,5-bistrifluoromethylbenzylamide, Alioc-pAph-Glu-Dap[-C(=NH)-NH2]-Phe[4-C(-S-(CH2)2-S-)-Ph]-NH2l

A!loc-pAph-Glu-Cys(BzI)-Cha-NH2,
Ai!oc-pAph-Glu-Thr(Bzl)-Cha-NH2l
Alloc-pAph-Glu-Phe(4-N02)-Cha-NH2r
Alloc-pAph-Glu-Asn-3r4-methylenedioxybenzylamide,
Alloc-pAph-Glu-Asn-2-(2-naphthyl)ethylamide,
Alloc-pAph-Glu-Asn-2-(1-naphthyl)ethylarnide,
Alloc-pAph-Glu-Asn-2-(2-pyridyi)ethylamide,
Alloc-pAph-Glu-Asn-2,2-diphenylethylamider
Alloc-pAph-Glu-Asn^^-difluorobenzylamide, or
Alloc-pAph-Glu-Asn-4-dimethylaminobenzyiamide,
or a pharmaceutical^ acceptable salt, amide or ester thereof.
-9 • A process for the preparation of a compound as claimed in one or more of claims 1 to 8, which comprises a1) coupling a compound of the formula Fmoc-En-OH wherein n is one, two or
three, to an acid sensitive linker attached to a resin, cleaving off the
protecting group Fmoc, coupling a compound of the formula
Fmoc-D1-D2-C(O)0H to the free amino group obtained and again
cleaving off the protecting group Fmoc,
or for the preparation of a compound of the formula I in which n is zero,
coupling a compound of the formula Fmoc-Dl-D2-C(0)OH to an acid
sensitive linker attached to a resin and cleaving off the protecting group
Fmoc, a2) coupling a compound of the formula Fmoc-B1-B2-C(0)OH to the free
amino group obtained in step a1) and cleaving off the protecting group
Fmoc, a3) coupling a compound of the formula R1-A1 -A2-C(0)OH to the free amino
group obtained in step a2), and a4) cleaving off the compound obtained according to steps a1) through a3)
from the resin by means of trifluoroacetic acid,
or

b1) coupling the side chain carbaxyiic acid of a compound of the formula
Frnoc-B1-CHR97-C(0)OPG, wherein R97 is 2-hydroxycarbonylethyl and
PG is a protecting group, to an acid sensitive benzyialcohol type of linker
attached to an amino functionaiized resin, b2) cleaving off the protecting group PG, b3) coupling a compound of the formula H2N-D2~D3-En-R2, wherein n is zero,
one, two or three, to the free carbaxyiic acid obtained in step b2), b4) cleaving off the protecting group Fmoc, b5) coupling a compound of the compound R1-A1-A2-C(0)OH to the free
amino group obtained in step b4), and b6) cleaving off the compound obtained according to steps b1) through b5)
from the resin by means of trifluoroacetic acid, or d) coupling of protected amino acids by traditional medicinal chemistry and
deprotecting to the target molecule by standard procedures known in the
art, where R1, R2, A1, A2, B1, B2, Dl, D2, D3 and E are defined as in claims 1 to 8 and Fmoc is 9-fluorenyimethyioxycarbonyi.
A pharmaceutical composition comprising an effective amount of a compound as claimed in one or more of claims 1 to 8, or a pharmaceutical^ acceptable salt thereof, and a pharmaceutical^ acceptable carrier.

Documents:

in-pct-2001-0348-che abstract duplicate.pdf

in-pct-2001-0348-che abstract.pdf

in-pct-2001-0348-che claims duplicate.pdf

in-pct-2001-0348-che claims.pdf

in-pct-2001-0348-che correspondence others.pdf

in-pct-2001-0348-che correspondence po.pdf

in-pct-2001-0348-che description (complete) duplicate.tif

in-pct-2001-0348-che description (complete).pdf

in-pct-2001-0348-che form-1.pdf

in-pct-2001-0348-che form-13.pdf

in-pct-2001-0348-che form-19.pdf

in-pct-2001-0348-che form-26.pdf

in-pct-2001-0348-che form-3.pdf

in-pct-2001-0348-che form-5.pdf

in-pct-2001-0348-che pct.pdf

in-pct-2001-0348-che petition.pdf


Patent Number 219102
Indian Patent Application Number IN/PCT/2001/348/CHE
PG Journal Number 23/2008
Publication Date 06-Jun-2008
Grant Date 25-Apr-2008
Date of Filing 13-Mar-2001
Name of Patentee SANOFI-AVENTIS DEUTSCHLAND GMBH
Applicant Address
Inventors:
# Inventor's Name Inventor's Address
1 WILDGOOSE, Peter
2 SAFAR, Pavel
3 SAFAROVA, Alena
PCT International Classification Number A61K 38/00
PCT International Application Number PCT/EP1999/006449
PCT International Filing date 1999-09-02
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 EP 98117506.0 1998-09-15 U.K.