NZ749214B2 - Multisubstituted aromatic compounds as serine protease inhibitors - Google Patents

Multisubstituted aromatic compounds as serine protease inhibitors Download PDF

Info

Publication number
NZ749214B2
NZ749214B2 NZ749214A NZ74921414A NZ749214B2 NZ 749214 B2 NZ749214 B2 NZ 749214B2 NZ 749214 A NZ749214 A NZ 749214A NZ 74921414 A NZ74921414 A NZ 74921414A NZ 749214 B2 NZ749214 B2 NZ 749214B2
Authority
NZ
New Zealand
Prior art keywords
unsubstituted
substituted
amino
compound
methyl
Prior art date
Application number
NZ749214A
Other versions
NZ749214A (en
Inventor
David Ben Kita
Son Minh Pham
Kevin Michael Short
David Charles Williams
Original Assignee
Verseon Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Verseon Corporation filed Critical Verseon Corporation
Priority claimed from NZ711064A external-priority patent/NZ711064A/en
Publication of NZ749214A publication Critical patent/NZ749214A/en
Publication of NZ749214B2 publication Critical patent/NZ749214B2/en

Links

Abstract

There are provided uses of compounds having a structure according to Formula (V) or pharmaceutically acceptable salt, ester, or solvate thereof, or a pharmaceutical composition comprising said compound, for the manufacture of a medicament for the treatment of a thrombotic disorder, a kallikrein-related disorder, or a disease or disorder responsive to inhibition of thrombin and/or kallikrein in a subject, wherein the kallikrein-related disorder comprises at least one of an ophthalmic disease, a thrombotic disease, a fibrinolytic disease, a type of cancer, an inflammatory condition, or a dermatological condition. Representative compounds of formula (V) include compounds such as 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(furan-3-yl)carbonyl]-1H-pyrazol-3-yl)-1,2-dihydropyridin-2-one, 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(furan-3-yl)carbonyl]-1H-pyrazol-3-yl)-1-methyl-1,2-dihydropyridin-2-one, 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(furan-3-yl)carbonyl]-1H-pyrazol-3-yl)-1-(pyridin-2-ylmethyl)-1,2-dihydropyridin-2-one, 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(furan-3-yl)carbonyl]-1H-pyrazol-3-yl)-1-(furan-2-ylmethyl)-1,2-dihydropyridin-2-one, 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(2-methoxyphenyl)carbonyl]-1H-pyrazol-3-yl)-1,2-dihydropyridin-2-one and the like. ted disorder, or a disease or disorder responsive to inhibition of thrombin and/or kallikrein in a subject, wherein the kallikrein-related disorder comprises at least one of an ophthalmic disease, a thrombotic disease, a fibrinolytic disease, a type of cancer, an inflammatory condition, or a dermatological condition. Representative compounds of formula (V) include compounds such as 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(furan-3-yl)carbonyl]-1H-pyrazol-3-yl)-1,2-dihydropyridin-2-one, 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(furan-3-yl)carbonyl]-1H-pyrazol-3-yl)-1-methyl-1,2-dihydropyridin-2-one, 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(furan-3-yl)carbonyl]-1H-pyrazol-3-yl)-1-(pyridin-2-ylmethyl)-1,2-dihydropyridin-2-one, 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(furan-3-yl)carbonyl]-1H-pyrazol-3-yl)-1-(furan-2-ylmethyl)-1,2-dihydropyridin-2-one, 3-(5-{[(5-chlorothiophen-2-yl)methyl]amino}-1-[(2-methoxyphenyl)carbonyl]-1H-pyrazol-3-yl)-1,2-dihydropyridin-2-one and the like.

Description

UBSTITUTED AROMATIC COMPOUNDS AS SERINE PROTEASE INHIBITORS b BACKGROUND OF THE INVENTION The present disclosure relates to compounds, e.g., ubstituted aromatic compounds, which exhibit biological ty, e. g., inhibitory action, against serine proteases, including thrombin and various kallikreins.
OO [0002] Kallikreins are a subgroup of serine proteases, diVided into plasma kallikrein and tissue kallikreins. Plasma kallikrein ) liberates kinins (bradykinin and kallidin) from the kininogens, peptides responsible for the regulation of blood pressure and activation of 11 inflammation. In the contact activation pathway of the coagulation cascade, plasma kallikrein 12 assists in the conversion of factor XII to factor XIIa (Keel, M.; Trentz, O. Injury 2005, 36, 13 691—709). Factor XIIa converts FXI into FXIa, which in turn activates FIX, which with its 14 co-factor FVIIIa forms the tenase complex, which finally activates FX to FXa. In the f1brinolysis part of the ation cascade, plasma kallikrein serves to convert plasminogen 16 to plasmin. Thus, it has been proposed that plasma rein inhibitors can be useful in the 17 treatment of thrombotic and fibrinolytic diseases and disease conditions (US Patent # 18 7,625,944; Bird et al. osis and Hemostasis 2012, 107, 1141). 19 [0003] In rodent models, it has been shown that activation of plasma kallikrein in the eye increases retinal vascular permeability; whereas inhibition of the rein-kinin system 21 reduces retinal leakage induced by diabetes and hypertension. These s suggest that 22 intraocular activation of the plasma kallikrein pathway can contribute to excessive retinal 23 vascular permeability that can lead to diabetic macular edema (DME). Thus, ce 24 suggests that plasma kallikrein inhibitors can provide a new therapeutic opportunity to reduce retinal vascular permeability (Feener, E. P. Curr Diab Rep 2010, 10, 270). 26 [0004] The Kallikrein-kinin system is involved in the regulation of vascular elial 27 growth factor (VEGF), endothelial NO synthase, and fibroblast growth factor 2, all of which 28 are involved in angiogenesis (Bader M. 2009, Arteriosclerosis, Thrombosis, and Vascular 29 Biology, 29: 617). Tissue kallikrein (KLKl) has been linked to blood vessel growth (Miura S., 2003, Hypertension, 41, l l 18). ies that moderate angiogenesis have been proposed 31 for the treatment of both diabetic macular edema (DME) and age-related macular 32 degeneration (AMD) (Syed, B.A.; Evans, J.B.; Bielory, L., 2012, Nature Reviews Drug 33 Discovery, 11, 827). Without filrther wishing to be bound by any theory, it is therefore 34 reasonable to conclude that KLKlinhibitors can be useful in the treatment of diabetic retinopathy, DME, and AMD. 36 [0005] Studies have shown that inflammation plays an important role in the origin and 37 development ofAMD, and treatment often es anti-inflammatories such as 38 osteroid (Telander, D., 2011, Seminars in Ophthalmology, 26(3), 192). The connection 39 between the kallikrein-kinin system and inflammation is also well established (Duchene, 40 2011, "Kallikrein-kinin kystem in inflammatory diseases". Kinins. De Gruyter. 261). Without 41 further wishing to be bound by any , it is reasonable to conclude that the anti- 42 inflammatory nature of kallikrein (e.g. KLKl and KLKB 1) inhibitors can be useful in the 43 treatment of AMD. 44 [0006] Ecallantide (Kalbitor) is a 60-amino acid recombinant n that acts as a potent 45 reversible inhibitor of plasma kallikrein ider L, et al., J Allergy Clin Immunol 2007, 46 120, 416). Ecallantide has been approvied by the FDA for the ent of acute attacks of 47 hereditary angioedema (HAE). Without fiarther wishing to be bound by any theory, it is 48 reasonable to believe that plasma kallikrein inhibition in general can be a useful treatment for 49 HAE, and thus there is strong interest in the development of plasma kallikrein inhibitors as a 50 therapy for HAE. 51 [0007] Tissue kallikreins (KLKs, for example, KLKl) are subdivided into various types, 52 and have been extensively investigated in cancer and inflammation biology. Various 53 kallikrein KLKs have been found to be up- or down-regulated in various cancer types, such 54 as al-, ular-, and non-small-cell lung adenocarcinoma (Caliendo et al. J. Med. 55 Chem., 2012, 55, 6669). Furthermore, overexpression of various KLKs in the skin has led to 56 the recognition that certain kallikrein inhibitors can be useful for certain dermatological 57 conditions, including atopic dermatitis, sis and rare skin diseases such as Netherton 58 me (Freitas et al. Bioorganic & Medicinal Chemistry Letters 2012, 22, 6072—6075). A 59 thorough sion of tissue kallikrein, plasma kallikreins, their fianctions and potential roles 60 in various diseases can be found in a variety of references, including the following which are 61 orated herein by reference in their entireties and for all purposes: Renne, T.; Gruber, A. 62 Thromb Haemost 2012, 107, 1012-3; Sotiropoulou, G.; Pampalakis, G. Trends in 63 Pharmacological Sciences 2012, 33, 623-634; akis, G.; Sotiropoulou, G. Chapter 9 64 Pharmacological Targeting n Tissue Kallikrein-Related Peptidases. In Proteinases 65 as Drug Targets, Dunn, 3., Ed. The Royal Society istry: 2012; pp 199-228; Caliendo, 66 G.; Santagada, V.; Perissutti, E.; Severino, B.; Fiorino, F.; tese, F.; Juliano, L. JMed 67 Chem 2012, 55, 6669-86. 68 [0008] In mammalian systems, blood vessel injuries result in bleeding events, which are 69 dealt with by the blood coagulation e. The cascade includes the extrinsic and intrinsic 70 pathways, ing the activation of at least 13 interconnected factors and a variety of co- 71 factors and other tory proteins. Upon vascular injury, plasma factor VII cts with 72 exposed Tissue Factor (TF), and the resultant TF-fVIIa x initiates a complex series of 73 . Factor fXa is produced directly ‘downstream’ from the TF-fVIIa complex, and 74 amplified manifold via the intrinsic y. FXa then serves as the catalyst for formation of 75 thrombin (fIIa), which in turn is the direct precursor to fibrinolysis. The outcome is a 76 fibrinolytic clot, which stops the bleeding. Fibrinolysis of the polymeric clot into fibrin 77 monomers leads to dissolution and a return of the system to the pre-clot state. The cascade is 78 a x balance of s and co-factors and is tightly regulated. In disease states, 79 red up- or down-regulation of any factor leads to conditions such as bleeding or 80 thrombosis. Historically, anticoagulants have been used in patients at risk of suffering from 81 thrombotic complications, such as angina, stroke and heart attack. Warfarin has d 82 dominance as a first-in-line anticoagulant therapeutic. Developed in the 1940s, it is a Vitamin 83 K nist and inhibits factors II, VII, IX and X, amongst others. It is administered orally, 84 but its ease of use is tempered by other s: it has a very long half-life (>2 days) and has 85 s drug-drug interactions. Importantly, since Vitamin K is a ubiquitous cofactor within 86 the ation cascade, antagonism results in the simultaneous inhibition of many clotting 87 factors and thus can lead to significant bleeding complications. 88 [0009] Much attention has been focused on heparin, the naturally-occurring polysaccharide 89 that activates AT III, the endogenous inhibitor ofmany of the factors in the coagulation 90 cascade. The need for parenteral administration for the heparin-derived therapeutics, and the 91 inconvenient requirements for close supervision for the orally available in, has resulted 92 in a drive to discover and develop orally available drugs with wide therapeutic windows for 93 safety and efficacy. Indeed, the position of thrombin in the coagulation cascade has made it a 94 popular target for drug discovery. Without wishing to be bound by any theory, it is believed 95 that the ultimate development of direct thrombin inhibitors (DTIs) is usefillly based upon the 96 classical D-Phe-Pro-Arg motif, a sequence that mimics fibrinogen, which is a natural 97 substrate of thrombin. Without further wishing to be bound by any theory, it is believed that 98 the use of DTIs is very well precedented, such as with the hirudin-based anticoagulants, and 99 thus there is strong st in the discovery and development of novel DTIs. 100 [0010] A gh discussion of thrombin and its roles in the coagulation process can be 101 found in a variety of references, ing the following which are incorporated herein by 102 nce in their entireties and for all purposes: Wieland, H. A., et al., 2003, Curr Opin 103 Investig Drugs, 4:264-71; Gross, P. L. & Weitz, J. 1., 2008, Arterioscler Thromb Vasc Biol, 104 28:380-6; Hirsh, J., et al., 2005, Blood, 105:453-63; Prezelj, A., et al., 2007, Curr Pharm 105 Des, 13:287-312. 106 BRIEF SUMMARY OF THE INVENTION 107 [0011] Embodiments of the invention encompass methods for treating or preventing a 108 kallikrein-related disease or disorder in a subject, the methods including administering a 109 compound of Formula (Ia): 110 R2 (Ia) 111 or a pharmaceutical composition including the compound and a pharmaceutically acceptable 112 excipient, to a subject in need thereof in an amount effective to treat or prevent said disease 113 or er, wherein: Ring A can be substituted or unsubstituted pyrazolyl, or tuted or 114 unsubstituted triazolyl; L1, L2, and L4 can be independently absent, a bond, substituted or 115 unsubstituted alkylene, substituted or unsubstituted heteroalkylene, -S-, -SO-, -SOz-, -O-, 116 -NHSOz-, or —NR7-; L3 can be a bond, substituted or unsubstituted alkylene, substituted or 117 unsubstituted heteroalkylene, -S-, -SO-, -SOz-, -O-, -, or —NR7-; R1, R2, and R4 can 118 be independently absent, en, halogen, substituted or unsubstituted alkyl, tuted or 119 unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted 120 cycloalkenyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted 121 heterocycloalkenyl, tuted or unsubstituted aryl, substituted or unsubstituted fused ring 122 aryl, or substituted or unsubstituted heteroaryl, provided that R1 can be absent when L1 can be 123 absent, R2 can be absent when L2 can be absent, and R4 can be absent when L4 can be absent; 124 R3 can be hydrogen, halogen, substituted or unsubstituted alkyl, substituted or unsubstituted 125 heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or tituted cycloalkenyl, 126 substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkenyl, 127 substituted or unsubstituted aryl, substituted or unsubstituted fused ring aryl, or tuted or 128 unsubstituted heteroaryl, provided that R2 can be absent when L2 can be absent, R3 can be WO 45986 129 absent when L3 can be , and R4 can be absent when L4 can be absent; and R7 can be 130 hydrogen, substituted or tituted a1ky1, substituted or unsubstituted heteroalkyl, 131 tuted or unsubstituted a1ky1ene, substituted or unsubstituted heteroalkylene, substituted 132 or unsubstituted cycloalkyl, substituted or unsubstituted lkenyl, tuted or 133 tituted heterocycloalkyl, substituted or unsubstituted heterocycloalkenyl, substituted or 134 unsubstituted aryl, substituted or unsubstituted fused ring aryl, or substituted or unsubstituted 135 heteroaryl. In some embodiments of the methods L4 and R4 can be absent. 136 [0012] In some embodiments, the compound can have the structure of Formula (Ila): R3’L3\«NY ['1‘ R1 N—N\ 137 R2 (IIa). 138 In some embodiments, L3 can be a bond, and R3 can be substituted or unsubstituted 139 heterocycloalkyl, tuted or unsubstituted aryl, substituted or unsubstituted fused ring 140 aryl, or substituted or unsubstituted heteroary1. In some embodiments, the heteroaryl can be 141 1, pyridazinyl, pyrimidiny1, thieny1, or fury1. In some embodiments, R3 can be 142 substituted or unsubstituted , substituted or unsubstituted morpholinyl, substituted or 143 unsubstituted oxanyl, tuted or unsubstituted oxetanyl, substituted or unsubstituted 144 naphthyl or tuted or tituted benzodioxinyl. In some embodiments, L3 can be a 145 bond or substituted or unsubstituted a1ky1ene, and R3 can be substituted or unsubstituted aryl, 146 or substituted or unsubstituted heterocycloalkyl. In some embodiments, L3 can be -C(O)O-, 147 and R3 can be substituted or unsubstituted a1ky1. In some embodiments, L3 can be -C(O)NR5-, 148 R5 can be hydrogen or alkyl, and R3 can be substituted or unsubstituted alkyl, or substituted 149 or unsubstituted aryl. In some embodiments, L1 can be -S-, -NR4-, tuted or 150 unsubstituted a1ky1ene, or substituted or unsubstituted heteroalkylene, and R1 can be 151 hydrogen, substituted or unsubstituted a1ky1, substituted or unsubstituted aryl, substituted or 152 unsubstituted fused ring ary1, substituted or unsubstituted aryl, or substituted or 153 unsubstituted heterocycloalkyl. In some embodiments, the heteroaryl can be pyridy1, 154 pyridazinyl, pyrimidiny1, thieny1, or fury1. In some embodiments, R1 can be chloro- 155 substituted thieny1. In some embodiments, R1 can be substituted or unsubstituted , 156 substituted or unsubstituted morpholinyl, substituted or unsubstituted oxanyl, substituted or 157 unsubstituted oxetanyl, substituted or unsubstituted naphthyl or substituted or unsubstituted 158 benzodioxinyl. In some embodiments, L2 and R2 can be absent. In some embodiments, L3 and 159 R3 can be absent. In some embodiments, L2 can be substituted or unsubstituted a1ky1ene or 160 -C(O)—, and R2 can be hydrogen, substituted or unsubstituted a1ky1, substituted or 161 unsubstituted aryl, substituted or unsubstituted fused ring aryl, substituted or unsubstituted 162 heteroaryl, or substituted or unsubstituted heterocycloalkyl. In some embodiments, the 163 heteroaryl can be l, zinyl, pyrimidinyl, thieny1, or fury1. In some embodiments, 164 R2 can be substituted or unsubstituted phenyl, substituted or unsubstituted linyl, 165 substituted or unsubstituted oxanyl, substituted or unsubstituted oxetanyl, substituted or 166 unsubstituted yl or substituted or unsubstituted benzodioxinyl. 167 [0013] In some embodiments, the compound can have the structure of Formula (III): R3J-3N\N’ N \ l1-2 168 R2 (111). 169 In some embodiments, L3 can be a bond, or substituted or unsubstituted ne, and R3 can 170 be substituted or unsubstituted aryl, substituted or tituted fused ring aryl, substituted or 171 unsubstituted heteroaryl, substituted or tituted cycloalkyl, or substituted or 172 unsubstituted heterocycloalkyl. In some embodiments, R3 can be phenyl, naphthyl, pyridyl, 173 pyridazinyl, pyrimidinyl, 1, filryl, morpholinyl, oxanyl, oxetanyl, or benzodioxinyl. In 174 some embodiments, L3 can be -C(O)O-, and R3 can be substituted or unsubstituted alkyl. In 175 some embodiments, L3 can be -C(O)NR’-, R, can be en or alkyl, and R3 can be 176 substituted or unsubstituted alkyl, or substituted or unsubstituted aryl. In some embodiments, 177 L1 can be -S-, -NR7-, substituted or unsubstituted alkylene, or substituted or unsubstituted 178 heteroalkylene, R7 can be a hydrogen or alkyl, and R1 can be hydrogen, substituted or 179 unsubstituted alkyl, substituted or unsubstituted aryl, substituted or unsubstituted fused ring 180 aryl, substituted or unsubstituted heteroaryl, or substituted or unsubstituted heterocycloalkyl. 181 In some embodiments, the heteroaryl can be pyridyl, pyridazinyl, pyrimidinyl, thieny1, or 182 fury1. In some embodiments, R1 can be chloro-substituted thieny1. In some embodiments, R1 183 can be substituted or unsubstituted phenyl, substituted or tituted morpholinyl, 184 substituted or unsubstituted oxanyl, substituted or unsubstituted oxetanyl, substituted or 185 unsubstituted naphthyl or substituted or unsubstituted benzodioxinyl. In some embodiments, 186 L2 can be a bond and R2 can be hydrogen. In some ments, L2 can be substituted or 187 unsubstituted alkylene or -C(O)-, and R2 can be hydrogen, substituted or unsubstituted alkyl, 188 substituted or unsubstituted aryl, tuted or unsubstituted fused ring aryl, substituted or 189 tituted heteroaryl, or substituted or unsubstituted heterocycloalkyl. In some 190 embodiments, the heteroaryl can be l, pyridazinyl, pyrimidinyl, thieny1, or fury1. In 191 some embodiments, R2 can be substituted or unsubstituted phenyl, substituted or 192 tituted morpholinyl, substituted or tituted oxanyl, substituted or unsubstituted 193 yl, substituted or tituted naphthyl or substituted or unsubstituted ioxinyl. 194 [0014] In some embodiments, the compound has the following structure of Formula (IV): 195 R2 (IV). 196 In some embodiments, L4 can be a bond, and R4 can be hydrogen, halogen, substituted or 197 unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, 198 or substituted or unsubstituted heteroaryl. In some ments, R4 can be halogen. In some 199 embodiments, R4 can be unsubstituted alkyl. In some ments, R4 can be phenyl, 200 naphthyl, pyridyl, pyridazinyl, dinyl, thienyl, or filryl. 201 [0015] In some embodiments, the compound can be selected from any of Tables B or C. In 202 some embodiments, the kallikrein-related disorder can be a thrombotic e, a fibrinolytic 203 disease, a type of cancer, an inflammatory condition, or a dermatological condition. In some 204 ments, the kallikrein-related disorder can be an ophthalmic disease. In some 205 embodiments, the ophthalmic disease can be diabetic macular edema, age-related macular 206 degeneration, or diabetic retinopathy. In some embodiments, the type of cancer can be 207 cervical-, testicular-, or all-cell lung adenocarcinoma. In some embodiments, the 208 inflammatory condition can be , inflammatory bowel disease, systemic inflammatory 209 response syndrome, or rheumatoid arthritis. In some embodiments, the dermatological 210 condition can be atopic dermatitis, psoriasis, or Netherton me. In some embodiments, 211 the compound acts by inhibiting kallikrein. In some embodiments, the compound acts by 212 inhibiting tissue kallikrein. In some embodiments, the compound acts by inhibiting plasma 213 kallikrein. In some embodiments, the compound or pharmaceutical composition can be 214 administered in the form of an ophthalmic composition applied topically to the eye. In some 215 embodiments, the ophthalmic composition can be in the form of eye drops. In some 216 embodiments, the compound or pharmaceutical composition can be administered in the form 217 of an ophthalmic composition Via intraVitreal injection. Embodiments of the invention also 218 encompass methods for treating or preventing a kallikrein-related disease or disorder in a 219 subject, including administering a compound from Table D or a pharmaceutical composition 220 including compound and a ceutically acceptable excipient, to a subject in need thereof 221 in an amount effective to treat or prevent the disease or disorder. 222 [0016] Embodiments of the invention also encompass compounds with structure of 223 a (V): R5—L5\ o W“\R‘\ / \N , N \ 224 R2 (V) 225 or pharmaceutically acceptable salt, ester, solvate, or prodrug thereof; wherein L1 can be a 226 bond, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, -S-, 227 -SO-, -S02-, -O-, -NHSOz-, or -NR7-; L2 and L5 can be independently absent, a bond, 228 substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, -S-, -SO-, 229 -S02-, -O-, -NHSOz-, or -NR7-; R1 can be hydrogen, halogen, substituted or unsubstituted 230 alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, 231 substituted or unsubstituted cycloalkenyl, ubstituted or unsubstituted heterocycloalkyl, 232 substituted or unsubstituted heterocycloalkenyl, substituted or unsubstituted aryl, substituted 233 or unsubstituted fused ring aryl, or substituted or unsubstituted heteroaryl; R2 and R5 can be 234 independently absent, hydrogen, halogen, substituted or unsubstituted alkyl, substituted or 235 unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or tituted 236 cycloalkenyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted 237 heterocycloalkenyl, substituted or unsubstituted aryl, substituted or unsubstituted fused ring 238 aryl, or substituted or unsubstituted heteroaryl; and R7 can be hydrogen, substituted or 239 unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted 240 alkylene, substituted or tituted heteroalkylene, substituted or unsubstituted lkyl, 241 substituted or tituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted 242 or unsubstituted heteroaryl. In some embodiments, L5 and R5 can be . In some 243 embodiments, L2 and R2 can be absent. In some embodiments, L2 can be , and R2 can 244 be substituted or unsubstituted alkyl, substituted or tituted cycloalkyl, substituted or 245 unsubstituted cycloalkenyl, substituted or unsubstituted heteroalkyl, tuted or 246 unsubstituted heterocycloalkyl, substituted or unsubstituted heterocycloalkenyl, substituted or 247 unsubstituted aryl, substituted or tituted fused ring aryl, or substituted or tituted 248 aryl. In some embodiments, R2 can be tuted or unsubstituted aryl or tuted or 249 unsubstituted heteroaryl. In some embodiments, the heteroaryl can be pyridyl, pyridazinyl, 2014/030853 250 pyrimidinyl, thienyl, or furyl. In some ments, R2 can be substituted or unsubstituted 251 alkyl, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted 252 heterocycloalkyl. In some embodiments, the heterocycloalkyl can be oxanyl, oxetanyl, or 253 morpholinyl. In some embodiments, the fiased ring aryl can be benzodioxinyl or naphthyl. In 254 some embodiments, L1 can be bond, -S-, -NR7-, substituted or unsubstituted alkylene, or 255 substituted or tituted alkylene, and R1 can be hydrogen, substituted or 256 unsubstituted alkyl, substituted or unsubstituted aryl, substituted or unsubstituted fused ring 257 aryl, substituted or unsubstituted heteroaryl, or substituted or unsubstituted heterocycloalkyl. 258 In some embodiments, the heteroaryl can be pyridyl, pyridazinyl, pyrimidinyl, thienyl, or 259 furyl. In some embodiments, R1 can be chloro-substituted thienyl. In some ments, the 260 heterocycloalkyl can be morpholinyl, oxanyl, or oxetanyl. In some ments, the fused 261 ring aryl can be benzodioxinyl or naphthyl. In some embodiments, L5 can be a bond or 262 tuted or unsubstituted alkylene, and R5 can be substituted or unsubstituted aryl, 263 substituted or unsubstituted fused ring aryl, or substituted or unsubstituted heteroaryl. In 264 some embodiments, the aryl can be pyridyl, zinyl, pyrimidinyl, thienyl, or furyl. 265 In some embodiments, the fused ring aryl can be benzodioxinyl or naphthyl. In some 266 embodiments, L5 can be substituted or unsubstituted alkylene, and R5 can be substituted or 267 unsubstituted heterocycloalkyl. In some embodiments, the heterocycloalkyl can be 268 morpholinyl, oxanyl, or oxetanyl. 269 [0017] Embodiments of the invention also encompass pharmaceutical compositions 270 including such compounds, or a compound as set forth in Table A, and a pharmaceutically 271 acceptable excipient. Embodiments of the ion also encompass methods for treating a 272 disease or disorder in a subject, including administering such compounds or pharmaceutical 273 compositions to a subject in need thereof in an amount effective to treat or prevent said 274 disease or disorder. In some embodiments, the disease or disorder can be a thrombotic 275 disorder. In some embodiments, the thrombotic er can be acute ry syndrome, 276 venous oembolism, al thromboembolism, cardiogenic thromboembolism, 277 disseminated intravascular coagulation, or a blood clot thrombus. In some embodiments, the 278 disease or disorder can be is. In some embodiments, the disease or disorder can be 279 Alzheimer’s Disease. In some embodiments, the e or disorder can be multiple sclerosis. 280 In some embodiments, the disease or disorder can be pain. In some embodiments, the disease 281 or disorder can be cancer. In some embodiments, the compound acts by inhibiting thrombin. 282 In some embodiments, the disease or disorder can be a kallikrein-related disorder. In some 283 embodiments, the kallikrein-related disorder can be a thrombotic disease, a fibrinolytic 284 disease, a type of cancer, an inflammatory condition, or a ological condition. In some 285 embodiments, the kallikrein-related disorder can be an ophthalmic disease. In some 286 embodiments, the ophthalmic disease can be diabetic macular edema, age-related macular 287 degeneration, or ic retinopathy. In some embodiments, the type of cancer can be 288 al-, testicular-, or non-small-cell lung adenocarcinoma. In some embodiments, the 289 inflammatory ion can be sepsis, inflammatory bowel disease, ic inflammatory 290 response syndrome, or rheumatoid arthritis. In some embodiments, the dermatological 291 condition can be atopic dermatitis, psoriasis, or Netherton Syndrome. In some ments, 292 the compound acts by inhibiting kallikrein. In some embodiments, the compound acts by 293 inhibiting tissue kallikrein. In some embodiments, the compound acts by ting plasma 294 kallikrein. In some embodiments, the compound or pharmaceutical composition can be 295 administered in the form of an ophthalmic composition applied topically to the eye. In some 296 ments, the ophthalmic composition can be in the form of eye drops. In some 297 embodiments, the compound or pharmaceutical composition can be administered in the form 298 of an ophthalmic composition via intravitreal ion. 300 BRIEF DESCRIPTION OF THE DRAWINGS 301 [0018] Not applicable. 302 DETAILED DESCRIPTION OF THE INVENTION 303 1. Definitions 304 [0019] The abbreviations used herein have their conventional g within the al 305 and biological arts. The chemical structures and ae set forth herein are constructed 306 according to the standard rules of chemical valency known in the chemical arts. 307 [0020] Where substituent groups are specified by their conventional chemical formulae, 308 written from left to right, they equally encompass the chemically identical substituents that 309 would result from writing the structure from right to left, e.g., -CHZO- is equivalent to 310 -OCH2-. 311 [0021] As used herein, the term “attached” ies a stable covalent bond, certain 312 preferred points of attachment being apparent to those of ordinary skill in the art. 313 [0022] The terms “halogen” or “halo” include fluorine, chlorine, bromine, and iodine. 314 Additionally, terms such as “haloalkyl” are meant to include monohaloalkyl and 315 polyhaloalkyl. For example, the term “halo(C1-C4)alkyl” includes, but is not limited to, 316 fluoromethyl, difluoromethyl, trifluoromethyl, 2,2,2-trifluoroethyl, robutyl, 3- 317 bromopropyl, and the like. 318 [0023] The term “alkyl,” by itself or as part of another substituent, means, unless otherwise 319 stated, a straight (i.e., unbranched) or branched chain, or combination thereof, which can be 320 fully saturated, mono- or polyunsaturated and can include di- and multivalent radicals, having 321 the number of carbon atoms designated (i.e., C1-C10 means one to ten carbons). Examples of 322 saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n- 323 propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, (cyclohexyl)methyl, homologs and 324 isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. An unsaturated 325 alkyl group is one having one or more double bonds or triple bonds. Examples of unsaturated 326 alkyl groups include, but are not limited to, Vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2- 327 (butadienyl), 2,4-pentadienyl, -pentadienyl), ethynyl, 1- and 3-propynyl, nyl, and 328 the higher homologs and isomers. ingly, the term "alkyl" can refer to C1-C16 straight 329 chain saturated, C1-C16 branched saturated, C3-C8 cyclic saturated and C1-C16 straight chain or 330 branched ted aliphatic hydrocarbon groups substituted with C3-C8 cyclic saturated 331 aliphatic hydrocarbon groups haVing the specified number of carbon atoms. For example, this 332 tion shall include but is not limited to methyl (Me), ethyl (Et), propyl (Pr), butyl (Bu), 333 pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, isopropyl (i-Pr), isobutyl (i-Bu), tert-butyl 334 (t-Bu), sec-butyl (s-Bu), isopentyl, neopentyl, ropyl, cyclobutyl, cyclopentyl, 335 cyclohexyl, cycloheptyl, cyclooctyl, ropylmethyl, and the like. 336 [0024] The term “alkylene,” by itself or as part of another substituent, means, unless 337 ise stated, a nt radical derived from an alkyl, as exemplified, but not limited by, 338 -CH2CH2CH2CH2-. Typically, an alkyl (or alkylene) group will have from 1 to 24 carbon 339 atoms, with those groups having 10 or fewer carbon atoms being preferred in the compounds 340 disclosed herein. A “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene 341 group, generally haVing eight or fewer carbon atoms. 342 [0025] The term “heteroalkyl,” by itself or in combination with another term, means, unless 343 ise stated, a stable straight or branched chain, or combinations thereof, consisting of at 344 least one carbon atom and at least one heteroatom selected from the group consisting of O, N, 345 P, Si, and S, and wherein the nitrogen and sulfur atoms can optionally be oxidized, and the 346 nitrogen heteroatom can optionally be quatemized. The atom(s) O, N, P, S, and Si can 347 be placed at any or position of the alkyl group or at the position at which the alkyl 348 group is attached to the remainder of the molecule. Examples include, but are not limited to: WO 45986 349 -CH2-CH2-O-CH3, -CH2-CH2-NH-CH3, -CH2-CH2-N(CH3)-CH3, -CH2-S-CH2-CH3, 350 -CH2-CH2, -S(O)—CH3, -CH2-CH2-S(O)2-CH3, -CH=CH-O-CH3, -Si(CH3)3, 351 -CH2-CH=N-OCH3, -CH=CH-N(CH3)-CH3, -O-CH3, -O-CH2-CH3, and -CN. Up to two 352 heteroatoms can be consecutive, such as, for example, -CH2-NH-OCH3. 353 [0026] Similarly, the term “heteroalkylene,” by itself or as part of another substituent, means, 354 unless otherwise stated, a divalent radical derived from heteroalkyl, as exemplified, but not 355 limited by, -CH2-CH2-S-CH2-CH2- and -CH2-S-CH2-CH2-NH-CH2-. For heteroalkylene 356 groups, heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, 357 alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Still fiarther, for alkylene and 358 heteroalkylene linking groups, no orientation of the g group is implied by the direction 359 in which the formula of the linking group is written. For example, the formula -C(O)2R'- 360 represents both -C(O)2R'- and )2-. As described above, heteroalkyl groups, as used 361 herein, include those groups that are attached to the remainder of the molecule through a 362 heteroatom, such as -C(O)R', -C(O)NR', -NR'R", -OR', -SR', and/or -S02R'. Where 363 “heteroalkyl” is recited, followed by recitations of specific heteroalkyl groups, such as 364 -NR'R" or the like, it will be understood that the terms heteroalkyl and -NR'R" are not 365 redundant or mutually exclusive. Rather, the specific heteroalkyl groups are recited to add 366 clarity. Thus, the term “heteroalkyl” should not be interpreted herein as ing specific 367 heteroalkyl groups, such as -NR'R" or the like. 368 [0027] The terms “cycloalkyl” and “heterocycloalkyl,” by themselves or in combination with 369 other terms, mean, unless otherwise stated, cyclic versions of “alkyl” and “heteroalkyl,” 370 tively. onally, for heterocycloalkyl, a heteroatom can occupy the position at 371 which the cycle is attached to the remainder of the molecule. Examples of cycloalkyl 372 include, but are not limited to, cyclopropyl, utyl, cyclopentyl, cyclohexyl, l- 373 cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like. Examples of heterocycloalkyl 374 include, but are not limited to, 1-(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3- 375 dinyl, holinyl, holinyl, tetrahydrofuranyl, tetrahydrofuranyl, 376 ydrothienyl, tetrahydrothienyl, 1-piperazinyl, 2-piperazinyl, and the like. A 377 “cycloalkylene” and a “heterocycloalkylene,” alone or as part of another substituent, means a 378 divalent radical d from a cycloalkyl and cycloalkyl, respectively. 379 [0028] The term "alkenyl" includes C2-C16 straight chain unsaturated, C2-C11 ed 380 unsaturated, C5-Cg unsaturated cyclic, and C2-C16 straight chain or branched unsaturated 381 aliphatic hydrocarbon groups substituted with C3-C8 cyclic saturated and unsaturated 382 aliphatic hydrocarbon groups having the specified number of carbon atoms. Double bonds 383 can occur in any stable point along the chain and the carbon-carbon double bonds can have 384 either the 02's or trans configuration. For example, this definition shall include but is not 385 limited to ethenyl, propenyl, butenyl, pentenyl, hexenyl, heptenyl, l, nonenyl, decenyl, 386 undecenyl, 1,5-octadienyl, 1,4,7-nonatrienyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, 387 ctenyl, ethylcyclohexenyl, butenylcyclopentyl, l-pentenylcyclohexenyl, and the like. 388 Similarly, “heteroalkenyl” refers to heteroalkyl having one or more double bonds. 389 [0029] The term “alkynyl” refers in the customary sense to alkyl additionally having one or 390 more triple bonds. The term “cycloalkenyl” refers to cycloalkyl additionally having one or 391 more double bonds. The term “heterocycloalkenyl” refers to heterocycloalkyl additionally 392 having one or more double bonds. 393 [0030] The term “acyl” means, unless otherwise stated, -C(O)R Where R is a substituted or 394 unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted 395 alkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or 396 substituted or unsubstituted heteroaryl. 397 [0031] Each of the above terms (e. g., “alkyl,” “heteroalkyl,” “aryl,” and “heteroaryl”) 398 includes both substituted and tituted forms of the indicated radical. Preferred 399 substituents for each type of radical are provided herein. 400 [0032] Substituents for the alkyl and alkyl radicals (including those groups often 401 referred to as alkylene, alkenyl, heteroalkylene, alkenyl, alkynyl, cycloalkyl, 402 heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) can be one or more of a variety of 403 groups selected from, but not limited to, -OR', =0, =NR', =N—OR', -NR'R", -SR', -halogen, 404 -SiR'R”R'", -OC(O)R', -C(O)R', -C02R', -CONR'R", -OC(O)NR'R", -NR"C(O)R', 405 -NR'-C(O)NR"R'", -NR"C(O)2R', -NR-C(NR'R”)=NR'", -S(O)R', R', -S(O)2NR'R", 406 -NRS02R', -CN, and -N02 in a number ranging from zero to (2m'+1), Where m' is the total 407 number of carbon atoms in such radical. R', R", and R'" each preferably independently refer 408 to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, 409 substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl 410 tuted with 1-3 halogens), substituted or unsubstituted alkyl, , or thioalkoxy 411 groups, or arylalkyl groups. When a compound disclosed herein includes more than one R 412 group, for example, each of the R groups is independently selected as are each R', R", and R'" 413 group When more than one of these groups is present. When R' and R" are attached to the 414 same nitrogen atom, they can be combined with the en atom to form a 4-, 5-, 6-, or 7- 415 ed ring. For example, -NR'R" includes, but is not d to, l-pyrrolidinyl and 4- 416 morpholinyl. From the above discussion of substituents, one of skill in the art will understand 417 that the term “alkyl” is meant to include groups ing carbon atoms bound to groups 418 other than hydrogen groups, such as kyl (e.g., -CF3 and -CH2CF3) and acyl (e.g., 419 H3, -C(O)CF3, -C(O)CH20CH3, and the like). 420 [0033] Similar to the tuents described for the alkyl radical, substituents for the aryl 421 and heteroaryl groups are varied and are selected from, for example: -OR', -NR'R", -SR', 422 -halogen, -SiR'R”R"', -OC(O)R', -C(O)R', -C02R', R", -OC(O)NR'R", -NR"C(O)R', 423 -NR'-C(O)NR"R'", -NR"C(O)2R', -NR-C(NR'R”)=NR'", -S(O)R', -S(O)2R', -S(O)2NR'R”, 424 -NRS02R', -CN, -N02, -R', -N3, -CH(Ph)2, fluoro(C1-C4)alkoxy, and fluoro(C1-C4)alkyl, in a 425 number ranging from zero to the total number of open es on the aromatic ring system; 426 and where R', R", and R'" are preferably independently selected from hydrogen, substituted or 427 unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted 428 cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, 429 and substituted or unsubstituted heteroaryl. When a compound disclosed herein includes 430 more than one R group, for example, each of the R groups is independently selected as are 431 each R', R", and R'" groups when more than one of these groups is present. 432 [0034] Two or more substituents can optionally be joined to form aryl, heteroaryl, 433 cycloalkyl, or heterocycloalkyl groups. Such so-called ring-forming substituents are lly, 434 though not arily, found attached to a cyclic base ure. In one embodiment, the 435 ring-forming substituents are attached to adjacent members of the base structure. For 436 example, two ring-forming substituents attached to adjacent members of a cyclic base 437 structure create a fused ring structure. In another ment, the ring-forming substituents 438 are attached to a single member of the base structure. For example, two ring-forming 439 substituents attached to a single member of a cyclic base structure create a spirocyclic 440 structure. In yet another embodiment, the ring-forming substituents are attached to non- 441 adjacent members of the base structure. 442 [0035] Two of the substituents on adjacent atoms of the aryl or heteroaryl ring can 443 optionally form a ring of the formula -T-C(O)-(CRR')q-U-, wherein T and U are 444 independently -NR-, -O-, -CRR'-, or a single bond, and q is an integer of from 0 to 3. 445 Alternatively, two of the substituents on adjacent atoms of the aryl or heteroaryl ring can 446 optionally be ed with a substituent of the formula -A-(CH2)r-B-, n A and B are 447 independently -CRR'-, -O-, -NR-, -S-, -S(O) -, -S(O)2-, -S(O)2NR'-, or a single bond, and r is 448 an r of from 1 to 4. One of the single bonds of the new ring so formed can ally be 449 replaced with a double bond. Alternatively, two of the substituents on adjacent atoms of the WO 45986 450 aryl or heteroaryl ring can optionally be replaced with a substituent of the formula 451 -(CRR')S-X'- (C"R”')d-, where s and d are independently integers of from 0 to 3, and X' is -O-, 452 -NR'-, -S-, -S(O)—, -, or -S(O)2NR'-. The substituents R, R', R", and R'" are preferably 453 independently selected from en, substituted or unsubstituted alkyl, tuted or 454 unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or 455 unsubstituted aryl, and substituted or unsubstituted heteroaryl. 456 [0036] As used herein, the terms “heteroatom” or “ring atom” are meant to include 457 oxygen (0), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si). 458 [0037] The term "alkyloxy" (e. g. methoxy, ethoxy, propyloxy, allyloxy, cyclohexyloxy) 459 represents an alkyl group as defined above having the indicated number of carbon atoms 460 attached through an oxygen bridge (-O-). 461 [0038] The term "alkylthio" (e. g. methylthio, hio, propylthio, cyclohexylthio and the 462 like) ents an alkyl group as defined above having the indicated number of carbon atoms 463 attached through a sulfur bridge (-S-). 464 [0039] The term "alkylamino" represents one or two alkyl groups as defined above having 465 the indicated number of carbon atoms attached through an amine bridge. The two alkyl 466 groups can be taken together with the en to which they are attached forming a cyclic 467 system containing 3 to 8 carbon atoms with or without one C1-C16alkyl, arleo-C16alkyl, or 468 C0-C16alkylaryl substituent. 469 [0040] The term "alkylaminoalkyl" represents an alkylamino group attached through an 470 alkyl group as defined above having the indicated number of carbon atoms. 471 [0041] The term "alkyloxy(alkyl)amino" (e. g. methoxy(methyl)amine, 472 ethoxy(propyl)amine) represents an alkyloxy group as defined above attached through an 473 amino group, the amino group itself having an alkyl substituent. 474 [0042] The term "alkylcarbonyl" (e.g. cyclooctylcarbonyl, pentylcarbonyl, 3- 475 hexylcarbonyl) represents an alkyl group as d above having the indicated number of 476 carbon atoms attached through a carbonyl group. 477 [0043] The term "alkylcarboxy" (e.g. heptylcarboxy, cyclopropylcarboxy, 3- 478 pentenylcarboxy) represents an alkylcarbonyl group as defined above wherein the carbonyl is 479 in turn attached h an oxygen. 480 [0044] The term "alkylcarboxyalkyl" represents an arboxy group ed through an 481 alkyl group as defined above having the indicated number of carbon atoms. 482 [0045] The term "alkylcarbonylamino" (e.g. hexylcarbonylamino, 483 cyclopentylcarbonylaminomethyl, methylcarbonylaminophenyl) ents an alkylcarbonyl 484 group as defined above wherein the carbonyl is in turn attached through the nitrogen atom of 485 an amino group. 486 [0046] The nitrogen group can itself be substituted with an alkyl or aryl group. 487 [0047] The term “aryl” means, unless otherwise stated, a polyunsaturated, aromatic, 488 hydrocarbon substituent, which can be a single ring or multiple rings rably from 1 to 3 489 rings) that are fused together (i.e., a fused ring aryl) or linked covalently. A fused ring aryl 490 refers to multiple rings fused together n at least one of the fused rings is an aryl ring. 491 The term “heteroaryl” refers to aryl groups (or rings) that contain from one to four 492 heteroatoms selected from N, O, and S, wherein the nitrogen and sulfur atoms are optionally 493 oxidized, and the nitrogen atom(s) are optionally quatemized. Thus, the term “heteroaryl” 494 includes fused ring heteroaryl groups (i.e., multiple rings fused together wherein at least one 495 of the fiJsed rings is a heteroaromatic ring). A 5,6-fused ring heteroarylene refers to two rings 496 fused together, wherein one ring has 5 members and the other ring has 6 members, and 497 wherein at least one ring is a heteroaryl ring. Likewise, a 6,6-fi1sed ring heteroarylene refers 498 to two rings fused together, wherein one ring has 6 members and the other ring has 6 499 members, and wherein at least one ring is a heteroaryl ring. And a 6,5-fi1sed ring 500 heteroarylene refers to two rings fused together, n one ring has 6 s and the 501 other ring has 5 members, and wherein at least one ring is a heteroaryl ring. A heteroaryl 502 group can be attached to the remainder of the molecule h a carbon or heteroatom. Non- 503 limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4- 504 biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, azolyl, 4-imidazolyl, 505 pyrazinyl, 2-oxazolyl, 4-oxazolyl, yloxazolyl, olyl, 3-isoxazolyl, 4- 506 isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, l, 3-fi1ryl, 2-thienyl, 507 3-thienyl, dyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, 508 purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinolyl, 5-isoquinolyl, 2-quinoxalinyl, 5- 509 quinoxalinyl, 3-quinolyl, and 6-quinolyl. Substituents for each of the above noted aryl and 510 aryl ring systems are selected from the group of acceptable substituents described 511 below. An “arylene” and a “heteroarylene,” alone or as part of another substituent, mean a 512 divalent radical derived from an aryl and heteroaryl, respectively. Accordingly, the term 513 "aryl" can represent an unsubstituted, mono-, di- or trisubstituted monocyclic, polycyclic, 514 biaryl and cyclic aromatic groups ntly ed at any ring position capable of 515 forming a stable covalent bond, certain preferred points of attachment being apparent to those WO 45986 516 skilled in the art (e. g. lyl, 4-imidazolyl). The aryl substituents are ndently 517 selected from the group consisting of halo, nitro, cyano, trihalomethyl, C1_16alkyl, arle1_ 518 16alkyl, C0-16alkyloxyC0-16alkyl, arleo-16alkyloxyC0-16alkyl, C0-16alkylthioC0-16alkyl, 519 arleo-16alkylthioC0-16alkyl, C0-16alkylaminoC0-16alkyl, arle0-16alkylaminoC0-16alkyl, 520 di(arle1-16alkyl)aminoC0-16alkyl, C1-16alkylcarbonle0-16alkyl, arle1_16alkylcarbonle0- 521 16alkyl, C1-16alkylcarboxyC0-16alkyl, arle1-16alkylcarboxyC0-16alkyl, C1_ 522 16alkylcarbonylaminoC0-16alkyl, arle1-16alkylcarbonylaminoC0-16alkyl,-C0-16alkleOOR4, - 523 C0-16alkleONR5R6 wherein R4, R5 and R6 are independently selected from hydrogen, C1- 524 C11alkyl, arleo-Cllalkyl, or R5 and R6 are taken together with the nitrogen to which they are 525 attached forming a cyclic system containing 3 to 8 carbon atoms with or without one C1- 526 l, arleo-C16alkyl, or C0-C116alkylaryl substituent. Aryl includes but is not limited to 527 pyrazolyl and triazolyl. 528 [0048] For brevity, the term “aryl” when used in combination with other terms (e.g., 529 aryloxy, ioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above. Thus, 530 the terms “arylalkyl,” “aralkyl” and the like are meant to e those radicals in which an 531 aryl group is attached to an alkyl group (e.g., benzyl, hyl, pyridylmethyl, and the like) 532 including those alkyl groups in which a carbon atom (e.g., a methylene group) has been 533 replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(1- 534 naphthyloxy)propyl, and the like), or a sulfur atom. Accordingly, the terms "arylalkyl" and 535 the like (e. g. (4-hydroxyphenyl)ethyl, (2-aminonaphthyl)hexyl, pyridylcyclopentyl) 536 represents an aryl group as defined above attached through an alkyl group as defined above 537 having the indicated number of carbon atoms. 538 [0049] The term “oxo,” as used herein, means an oxygen that is double bonded to a carbon 539 atom. 540 [0050] The term “alkylsulfonyl,” as used herein, means a moiety having the formula 541 -S(02)-R', where R' is an alkyl group as defined above. R' can have a ed number of 542 carbons (e.g., “C1-C4 alkylsulfonyl”). 543 [0051] The term "carbonyloxy" represents a carbonyl group attached through an oxygen 544 bridge. 545 [0052] In the above definitions, the terms "alkyl" and yl" can be used 546 interchangeably in so far as a stable chemical entity is formed, as would be apparent to those 547 skilled in the art. 2014/030853 548 [0053] The term “linker” refers to attachment groups interposed n substituents, e.g., 549 R1, R2, R3 or R4 bed herein, e.g., a (Ia) and generically referred to as R“, and the 550 group which is substituted, e.g., “ring A” group of e.g., Formula (Ia). In some embodiments, 551 the linker includes amido (-CONH-RIl or -NHCO-R“), thioamido (-CSNH-RIl or Rn), 552 carboxyl (-COz-RIl or -OCOR“), carbonyl (-CO-Rn), urea (-NHCONH-Rn), thiourea 553 (-NHCSNH-Rn), sulfonamido (-NHSOz-RIl or -S02NH-Rn), ether (-O-R“), sulfonyl 554 (-SOz-Rn), sulfoxyl (-SO-Rn), carbamoyl (-NHC02-RIl or -OCONH-Rn), or amino (-NHR“) 555 linking moieties. 556 [0054] A “substituent group,” as used , means a group selected from the following 557 moieties: 558 (A) -OH, -NH2, -SH, -CN, -CF3, -N02, oxo, halogen, -COOH, unsubstituted alkyl, 559 unsubstituted heteroalkyl, unsubstituted cycloalkyl, tituted heterocycloalkyl, 560 unsubstituted aryl, unsubstituted heteroaryl, and 561 (B) alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, and aryl, substituted with 562 at least one substituent selected from: 563 (i) oxo, -OH, -NH2, -SH, -CN, -CF3, -N02, halogen, -COOH, unsubstituted alkyl, 564 unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted cycloalkyl, 565 unsubstituted aryl, unsubstituted heteroaryl, and 566 (ii) alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, substituted 567 with at least one substituent selected from: 568 (a) oxo, -OH, -NH2, -SH, -CN, -CF3, -N02, halogen, -COOH, unsubstituted alkyl, 569 unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted 570 heterocycloalkyl, unsubstituted aryl, unsubstituted heteroaryl, and 571 (b) alkyl, heteroalkyl, cycloalkyl, cycloalkyl, aryl, or aryl, substituted 572 with at least one tuent selected from: oxo, -OH, -NH2, -SH, -CN, -CF3, 573 -N02, halogen, -COOH, unsubstituted alkyl, unsubstituted alkyl, 574 unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, and 575 unsubstituted heteroaryl. 576 [0055] A “size-limited substituent” or “ size-limited substituent group,” as used herein, 577 means a group selected from all of the substituents described above for a “substituent group,” 578 wherein each substituted or unsubstituted alkyl is a substituted or unsubstituted C1-C20 alkyl, 579 each substituted or unsubstituted heteroalkyl is a substituted or unsubstituted 2membered 580 heteroalkyl, each substituted or unsubstituted cycloalkyl is a substituted or unsubstituted C4- 581 Cg cycloalkyl, and each substituted or unsubstituted heterocycloalkyl is a substituted or 582 unsubstituted 4membered heterocycloalkyl. 583 [0056] A “lower substituent” or “lower substituent group,” as used herein, means a group 584 selected from all of the tuents described above for a “substituent group,” wherein each 585 tuted or unsubstituted alkyl is a substituted or unsubstituted C1-C8 alkyl, each 586 substituted or unsubstituted alkyl is a tuted or unsubstituted 2membered 587 heteroalkyl, each substituted or unsubstituted cycloalkyl is a tuted or unsubstituted C5- 588 C7 cycloalkyl, and each substituted or unsubstituted heterocycloalkyl is a tuted or 589 unsubstituted 5membered heterocycloalkyl. 590 [0057] The term ” used in the context of a numeric value indicates a range of +/- 591 10% of the numeric value, unless expressly indicated otherwise. 592 11. Compounds 593 [0058] In one aspect, there is provided a compound with structure of Formula (Ia): 594 R2 (Ia) 595 or pharmaceutically acceptable salt, ester, solvate, or prodrug thereof. Ring A is substituted 596 or unsubstituted pyrazolyl, or substituted or unsubstituted triazolyl. L1, L2 and L3 are 597 independently absent, a bond, substituted or unsubstituted alkylene, substituted or 598 unsubstituted heteroalkylene, -S-, -SO-, -S02-, -O-, -NHS02-, or —NR7-. L4 is absent, a bond, 599 substituted or unsubstituted alkylene, tuted or unsubstituted heteroalkylene, -S-, -SO-, 600 -SOz-, -O-, -NHSOz-, or —NR7-. R1, R2, and R4 are independently absent, hydrogen, halogen, 601 substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or 602 unsubstituted cycloalkyl, substituted or unsubstituted cycloalkenyl, tuted or 603 unsubstituted heterocycloalkyl, tuted or unsubstituted heterocycloalkenyl, substituted or 604 unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted 605 cycloalkenyl, substituted or unsubstituted heterocycloalkenyl, or substituted or unsubstituted 606 fused ring aryl. In some embodiments, R1 can be absent provided L1 is also absent. In some 607 embodiments, R2 can be absent provided L2 is also absent. In some embodiments, R4 can be 608 absent ed L4 is also . R3 is hydrogen, halogen, substituted or tituted alkyl, 609 substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or 610 unsubstituted cycloalkenyl, substituted or unsubstituted heterocycloalkyl, substituted or 611 unsubstituted heterocycloalkenyl, tuted or unsubstituted aryl, substituted or 612 unsubstituted fused ring aryl, or substituted or unsubstituted heteroaryl. R7 is hydrogen, 613 tuted or tituted alkyl, substituted or unsubstituted heteroalkyl, substituted or 614 unsubstituted alkylene, substituted or unsubstituted heteroalkylene, substituted or 615 unsubstituted cycloalkyl, substituted or unsubstituted cycloalkenyl, substituted or 616 unsubstituted heterocycloalkyl, substituted or unsubstituted heterocycloalkenyl, substituted or 617 unsubstituted aryl, tuted or unsubstituted fused ring aryl, or substituted or unsubstituted 618 heteroaryl. 619 [0059] In some embodiments, the compound is a pharmaceutically acceptable salt, ester, 620 solvate, or prodrug of a compound of Formula (Ia). In some embodiments, the compound is 621 not an ester, not a solvate, and not a prodrug. 622 [0060] In some embodiments, L4 and R4 are absent, providing a compound with structure 623 of Formula (Ib) ing.
LKRZ 624 (Ib) 625 [0061] In some embodiments, there is provided a compound according to Formula (Ib) with 626 structure of Formula (IIa) following.
/L3 N L1 :22 627 (11a) 628 [0062] In some embodiments, the compound has the ure of Formula (IIa), wherein L3 629 is a bond, and R3 is substituted or unsubstituted aryl, substituted or tituted fused ring 630 aryl, substituted or tituted heterocycloalkyl, or substituted or unsubstituted heteroaryl. 631 In some ments, R3 is unsubstituted aryl, preferably phenyl. In some embodiments, R3 632 is substituted aryl, preferably halogen-substituted phenyl. In some embodiments, R3 is 633 substituted or unsubstituted , or substituted or unsubstituted thienyl. In some 634 embodiments, R3 is unsubstituted thienyl. In some embodiments, R3 is a chloro-substituted 635 thienyl. In some embodiments, R3 is substituted or unsubstituted l, or substituted or 636 unsubstituted pyridazinyl. In some embodiments, R3 is unsubstituted pyridyl. In some 637 embodiments, R3 is unsubstituted pyridazinyl. In some embodiments, R3 is substituted or 638 unsubstituted pyrimidinyl, or substituted or unsubstituted furyl. In some embodiments, R3 is 639 unsubstituted pyrimidinyl. In some ments, R3 is unsubstituted fierl. In some 640 embodiments, R3 is substituted or unsubstituted linyl, or substituted or unsubstituted 641 oxanyl, or substituted or unsubstituted oxetanyl. In some embodiments, R3 is unsubstituted 642 linyl. In some embodiments, R3 is unsubstituted oxanyl. In some embodiments, R3 is 643 unsubstituted oxetanyl. In some embodiments, R3 is substituted or unsubstituted 644 benzodioxinyl, or substituted or unsubstituted naphthyl. In some embodiments, R3 is 645 unsubstituted benzodioxinyl. In some ments, R3 is unsubstituted naphthyl. 646 [0063] In some embodiments, a compound is provided with structure of Formula (IIa), 647 wherein L3 is a bond, substituted or unsubstituted alkylene, and R3 is substituted or 648 unsubstituted aryl, or substituted or unsubstituted heterocycloalkyl. 649 [0064] In some embodiments, the compound has the ure of Formula (IIa), wherein L3 650 is -C(O)O-, and R3 is substituted or tituted alkyl, preferably unsubstituted alkyl, more 651 preferably unsubstituted lower alkyl. 652 [0065] In some embodiments, the compound has the structure of Formula (IIa), wherein L3 653 is -C(O)NR5-, R5 is hydrogen or alkyl, and R3 is tuted or unsubstituted alkyl, or 654 substituted or tituted aryl. 655 [0066] Further to any ment above wherein the compound has the structure of 656 Formula (IIa),in some ments L1 is -S-, -NR7-, substituted or unsubstituted alkylene, or 657 substituted or unsubstituted alkylene, where R7 is as described in formula Ia, and R1 is 658 hydrogen, substituted or unsubstituted alkyl, substituted or tituted aryl, substituted or 659 unsubstituted fused ring aryl, substituted or unsubstituted heteroaryl, or substituted or 660 unsubstituted cycloalkyl. In some embodiments, R1 is substituted or unsubstituted 661 phenyl. In some embodiments is an R1 is unsubstituted phenyl. In some embodiments, R1 is a 662 substituted or unsubsituted pyridyl. In some embodiments, R1 is a substituted or unsubsituted 663 pyridazinyl. In some embodiments, R1 is a substituted or unsubsituted pyrimidinyl. In some 664 embodiments, R1 is a substituted or ituted thienyl. In some embodiments, R1 is a 665 substituted or ituted fierl. In some ments, R1 is an unsubsituted pyridyl. In 666 some embodiments, R1 is an unsubsituted pyridazinyl. In some embodiments, R1 is an 667 unsubsituted pyrimidinyl. In some embodiments, R1 is an unsubsituted thienyl. In some 668 embodiments, R1 is a chloro-substituted thienyl. In some embodiments, R1 is an unsubsituted 669 furyl. In some embodiments, R1 is a substituted or ituted morpholinyl. In some 670 embodiments, R1 is a substituted or unsubsituted oxanyl. In some embodiments, R1 is a 671 substituted or unsubsituted oxetanyl. In some embodiments, R1 is an unsubsituted 672 morpholinyl. In some embodiments, R1 is an unsubsituted oxanyl. In some ments, R1 673 is an unsubsituted oxetanyl. In some embodiments, R1 is substituted or unsubstituted 674 ioxinyl. In some ments, R1 is substituted or unsubstituted naphthy1. In some 675 embodiments, R1 is unsubstituted benzodioxinyl. In some embodiments, R1 is unsubstituted 676 naphthy1. In some embodiments, R3 is substituted or unsubstituted aryl. In some 677 embodiments, R3 is unsubstituted aryl. In some ments, L3 and R3 are absent. In some 678 embodiments, L2 and R2 are absent. In some embodiments, L2 is a bond. In some 679 embodiments, L2 is a bond and R2 is hydrogen. 680 [0067] Further to any embodiment above wherein the compound has the structure of 681 Formula (IIa), R2 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted 682 heteroalkyl, substituted or unsubstituted lkyl, substituted or tituted cycloalkenyl, 683 substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted cycloalkenyl, 684 substituted or unsubstituted aryl, substituted or unsubstituted filSGd ring aryl, or substituted or 685 unsubstituted heteroaryl. In some embodiments, R2 is substituted or unsubstituted . In 686 some embodiments is an R2 is unsubstituted phenyl. In some embodiments, R2 is a substituted 687 or unsubsituted pyridyl. In some embodiments, R2 is a substituted or unsubsituted 688 pyridazinyl. In some embodiments, R2 is a substituted or unsubsituted pyrimidinyl. In some 689 embodiments, R2 is a substituted or unsubsituted thieny1. In some embodiments, R2 is a 690 tuted or unsubsituted fierl. In some embodiments, R2 is an unsubsituted pyridyl. In 691 some embodiments, R2 is an ituted pyridazinyl. In some embodiments, R2 is an 692 unsubsituted pyrimidinyl. In some embodiments, R2 is an unsubsituted thieny1. In some 693 embodiments, R2 is a chloro-substituted thieny1. In some embodiments, R2 is an unsubsituted 694 furyl. In some embodiments, R2 is a substituted or unsubsituted morpholinyl. In some 695 embodiments, R2 is a substituted or unsubsituted oxanyl. In some ments, R2 is a 696 tuted or unsubsituted oxetanyl. In some embodiments, R2 is an unsubsituted 697 morpholinyl. In some ments, R2 is an unsubsituted oxanyl. In some embodiments, R2 698 is an unsubsituted oxetanyl. In some embodiments, R2 is substituted or unsubstituted 699 benzodioxinyl. In some embodiments, R2 is substituted or unsubstituted naphthy1. In some 700 embodiments, R2 is unsubstituted ioxinyl. In some embodiments, R2 is unsubstituted 701 naphthyl. 702 [0068] In some embodiments, the compound of Formula (Ila) has the structure of Formula 703 (IIb) ing, wherein L1 is -NH-(CH2)n-, n is 0 to 6, preferably 1, and R1 is en, 704 substituted or unsubstituted alkyl, substituted or unsubstituted aryl, substituted or 705 unsubstituted heteroaryl, or substituted or unsubstituted cycloalkyl.
L3 N R R1 R3 \6 \7’ M; N—N\ 706 R2 (11b) 707 [0069] In some embodiments of the compound of Formula (IIb), L1 is -NHCH2- or 708 -NH(CH2)2-, and R1 is substituted or unsubstituted aryl. In some embodiments, R1 is 709 unsubstituted aryl. In some embodiments, R1 is aryl, preferably phenyl, substituted with 710 halogen, -CN or alkyloxy, preferably methoxy. In some embodiments, R1 is unsubstituted 711 alkyl, preferably lower alkyl, more preferably methyl or ethyl. In some embodiments, n is 0, 712 and R1 is hydrogen. 713 [0070] In some embodiments, the compound of a (IIa) has the structure of Formula 714 (IIc) following, wherein L1 is a bond, and R1 is unsubstituted alkyl, or substituted or 715 unsubstituted aryl. In some embodiments, R1 is tituted alkyl, preferably lower alkyl. In 716 some embodiments, R1 is substituted aryl, preferably halogen-substituted phenyl.
/L3 N S R1 R3 \6 \Y W N—N\ 717 R2 (IIc) 718 [0071] In some embodiments, there is ed a compound according to a (Ib) with 719 structure of Formula (III) following. 720 R2 (111) 721 [0072] In some embodiments, the compound has the structure of a (III). In some 722 embodiments, L3 is a bond, or substituted or tituted alkylene, and R3 is substituted or 723 unsubstituted aryl, substituted or unsubstituted fused ring aryl, substituted or unsubstituted 724 heterocycloalkyl, or substituted or unsubstituted heterocycloalkyl. In some embodiments, R3 725 is substituted or unsubstituted phenyl, or substituted or unsubstituted thienyl. In some 726 embodiments, R3 is unsubstituted phenyl. In some embodiments, R3 is unsubstituted thienyl. 727 In some embodiments, R3 is a chloro-substituted thienyl. In some embodiments, R3 is 728 substituted or unsubstituted pyridyl, or substituted or tituted pyridazinyl. In some 729 embodiments, R3 is unsubstituted pyridyl. In some embodiments, R3 is unsubstituted 730 pyridazinyl. In some embodiments, R3 is substituted or unsubstituted pyrimidinyl, or 731 substituted or tituted fierl. In some embodiments, R3 is unsubstituted pyrimidinyl. In 732 some embodiments, R3 is unsubstituted furyl. In some embodiments, R3 is substituted or 733 unsubstituted morpholinyl, or tuted or unsubstituted oxanyl, or substituted or 734 unsubstituted yl. In some embodiments, R3 is tituted morpholinyl. In some 735 embodiments, R3 is unsubstituted oxanyl. In some ments, R3 is unsubstituted 736 oxetanyl. In some embodiments, R3 is substituted or unsubstituted ioxinyl, or 737 substituted or unsubstituted naphthyl. In some embodiments, R3 is unsubstituted 738 benzodioxinyl. In some ments, R3 is unsubstituted naphthyl. 739 [0073] In some embodiments, the compound has the structure of Formula (III) wherein L3 740 is -, and R3 is substituted or unsubstituted alkyl. In some embodiments, the compound 741 has the structure of Formula (III) wherein L3 is -C(O)NR7, R7 is hydrogen or alkyl, and R3 is 742 substituted or unsubstituted alkyl, or substituted or unsubstituted aryl. 743 [0074] Further to any ment above wherein the compound has the structure of 744 Formula (III), in some ments, L1 is -S-, -NR7-, substituted or unsubstituted alkylene, 745 or substituted or tituted heteroalkylene, where R7 is as described in Formula (Ia), and 746 R1 is hydrogen, tuted or unsubstituted alkyl, substituted or unsubstituted aryl, 747 substituted or tituted fused ring aryl, substituted or unsubstituted heteroaryl, or 748 substituted or unsubstituted heterocycloalkyl. In some embodiments, R1 is substituted or 749 unsubstituted phenyl. In some embodiments is an R1 is unsubstituted phenyl. In some 750 embodiments, R1 is a substituted or unsubsituted pyridyl. In some embodiments, R1 is a 751 substituted or unsubsituted pyridazinyl. In some embodiments, R1 is a substituted or 752 unsubsituted pyrimidinyl. In some embodiments, R1 is a substituted or unsubsituted thienyl. 753 In some embodiments, R1 is a substituted or unsubsituted furyl. In some embodiments, R1 is 754 an unsubsituted pyridyl. In some ments, R1 is an unsubsituted pyridazinyl. In some 755 embodiments, R1 is an unsubsituted pyrimidinyl. In some embodiments, R1 is an unsubsituted 756 thienyl. In some embodiments, R1 is a chloro-substituted thienyl. In some embodiments, R1 is 757 an unsubsituted furyl. In some embodiments, R1 is a substituted or unsubsituted morpholinyl. 758 In some embodiments, R1 is a tuted or unsubsituted oxanyl. In some embodiments, R1 759 is a substituted or unsubsituted oxetanyl. In some embodiments, R1 is an unsubsituted 760 linyl. In some embodiments, R1 is an unsubsituted oxanyl. In some embodiments, R1 761 is an unsubsituted oxetanyl. In some embodiments, R1 is substituted or unsubstituted 762 benzodioxinyl. In some embodiments, R1 is substituted or unsubstituted naphthyl. In some 763 embodiments, R1 is unsubstituted benzodioxinyl. In some embodiments, R1 is unsubstituted 764 naphthyl. 765 [0075] r to any embodiment above wherein the compound has the structure of 766 a (III), in some embodiments, L2 is a bond. In some ments, R2 is en. In 767 some embodiments, L2 is substituted or unsubstituted alkylene or -C(O)-, and R2 is hydrogen, 768 substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or 769 unsubstituted cycloalkyl, substituted or unsubstituted lkenyl, substituted or 770 unsubstituted heterocycloalkyl, substituted or unsubstituted heterocycloalkenyl, substituted or 771 unsubstituted aryl, substituted or unsubstituted fused ring aryl, or substituted or unsubstituted 772 aryl. In some embodiments, R2 is substituted or unsubstituted phenyl. In some 773 embodiments is an R2 is unsubstituted phenyl. In some embodiments, R2 is a substituted or 774 unsubsituted pyridyl. In some embodiments, R2 is a substituted or unsubsituted pyridazinyl. 775 In some embodiments, R2 is a substituted or unsubsituted pyrimidinyl. In some embodiments, 776 R2 is a tuted or unsubsituted thienyl. In some embodiments, R2 is a substituted or 777 unsubsituted furyl. In some embodiments, R2 is an unsubsituted pyridyl. In some 778 embodiments, R2 is an unsubsituted pyridazinyl. In some embodiments, R2 is an unsubsituted 779 pyrimidinyl. In some embodiments, R2 is an ituted thienyl. In some embodiments, R2 780 is a chloro-substituted thienyl. In some embodiments, R2 is an unsubsituted fierl. In some 781 embodiments, R2 is a substituted or ituted linyl. In some ments, R2 is a 782 substituted or unsubsituted oxanyl. In some embodiments, R2 is a substituted or unsubsituted 783 oxetanyl. In some embodiments, R2 is an ituted morpholinyl. In some embodiments, 784 R2 is an unsubsituted oxanyl. In some embodiments, R2 is an unsubsituted oxetanyl. In some 785 embodiments, R2 is substituted or unsubstituted benzodioxinyl. In some embodiments, R2 is 786 substituted or unsubstituted naphthyl. In some embodiments, R2 is unsubstituted 787 benzodioxinyl. In some embodiments, R2 is unsubstituted naphthyl. 788 [0076] In some embodiments, there is ed a compound ing to Formula (Ia) with 789 structure of Formula (IV) following. 790 R2 (IV) 791 [0077] In some ments, there is provided a compound according to Formula (IV) 792 wherein L4 is a bond; and R4 is en, halogen, substituted or unsubstituted alkyl, or 793 substituted or tituted heteroalkyl, substituted or unsubstituted aryl, or substituted or 794 unsubstituted heteroaryl. In some embodiments, R4 is halogen. In some embodiments, R4 is 795 tituted alkyl. In some embodiments, R4 is substituted or unsubstituted aryl or 796 substituted or unsubstituted heteroaryl. In some embodiments, R4 is phenyl, naphthyl, 797 pyridyl, pyridazinyl, pyrimidinyl, thienyl, or furyl. 798 [0078] In some embodiments, there is provided a compound ing to Formula (III) 799 with structure of Formula (V) following.
R5—L5\ o WRW1\/ \ N,N\ 800 R2 (V) 801 [0079] In some embodiments, the compound is a ceutically acceptable salt, ester, 802 solvate, or g of a compound of Formula (V). In some embodiments, the compound is 803 not an ester, not a e, and not a prodrug. In some embodiments, there is provided a 804 compound according to Formula (V) wherein L1 is a bond, substituted or unsubstituted 805 alkylene, substituted or unsubstituted heteroalkylene, -S-, -SO-, -S02-, -O-, -NHSOz-, or - 806 NR7-. R1 is hydrogen, a n, substituted or unsubstituted alkyl, substituted or 807 unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted 808 heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted cycloalkenyl, 809 substituted or unsubstituted heterocycloalkenyl, substituted or unsubstituted fused ring aryl, 810 or substituted or unsubstituted heteroaryl. L2 is absent, a bond, substituted or tituted 811 alkylene, substituted or tituted heteroalkylene, -S-, -SO-, -S02-, -O-, -NHSOz-, or — 812 NR7. L5 is absent, a bond, substituted or unsubstituted alkylene, substituted or tituted 813 heteroalkylene, -S-, -SO-, -SOz-, -O-, -NHSOz-, or —NR7. R2 is absent, hydrogen, a halogen, 814 substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or 2014/030853 815 unsubstituted cycloalkyl, substituted or unsubstituted cycloalkyl, substituted or 816 unsubstituted aryl, tuted or unsubstituted cycloalkenyl, tuted or unsubstituted 817 heterocycloalkenyl, substituted or unsubstituted fused ring aryl, or substituted or 818 unsubstituted heteroaryl. In some embodiments, R2 is absent provided L2 is also absent. R5 is 819 absent, hydrogen, a halogen, tuted or unsubstituted alkyl, tuted or unsubstituted 820 heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted 821 heterocycloalkyl, substituted or unsubstituted aryl, substituted or tituted cycloalkenyl, 822 substituted or unsubstituted heterocycloalkenyl, substituted or unsubstituted fused ring aryl, 823 or substituted or unsubstituted heteroaryl. In some embodiments, R5 is absent provided L5 is 824 also absent. R7 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted 825 heteroalkyl, substituted or unsubstituted alkylene, substituted or unsubstituted heteroalkylene, 826 substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, 827 tuted or unsubstituted aryl, substituted or unsubstituted cycloalkenyl, substituted or 828 unsubstituted heterocycloalkenyl, substituted or unsubstituted fused ring aryl, or substituted 829 or unsubstituted heteroaryl. 830 [0080] Further to any embodiment above wherein the compound has the structure of 831 Formula (V), in some embodiments, L2 is a bond. In some embodiments, R2 is hydrogen. In 832 some ments, L2 is substituted or tituted alkylene or , and R2 is hydrogen, 833 substituted or tituted alkyl, substituted or unsubstituted heteroalkyl, substituted or 834 unsubstituted cycloalkyl, substituted or unsubstituted cycloalkenyl, substituted or 835 unsubstituted heterocycloalkyl, tuted or unsubstituted heterocycloalkenyl, substituted or 836 unsubstituted aryl, substituted or unsubstituted fused ring aryl, or substituted or tituted 837 heteroaryl. In some embodiments, R2 is substituted or unsubstituted phenyl. In some 838 ments is an R2 is unsubstituted phenyl. In some embodiments, R2 is a substituted or 839 ituted pyridyl. In some embodiments, R2 is a substituted or ituted pyridazinyl. 840 In some embodiments, R2 is a substituted or unsubsituted pyrimidinyl. In some embodiments, 841 R2 is a tuted or unsubsituted thienyl. In some embodiments, R2 is a substituted or 842 unsubsituted furyl. In some embodiments, R2 is an unsubsituted pyridyl. In some 843 embodiments, R2 is an unsubsituted pyridazinyl. In some embodiments, R2 is an unsubsituted 844 pyrimidinyl. In some embodiments, R2 is an unsubsituted thienyl. In some embodiments, R2 845 is a chloro-substituted thienyl. In some embodiments, R2 is an unsubsituted fierl. In some 846 embodiments, R2 is a substituted or unsubsituted morpholinyl. In some embodiments, R2 is a 847 substituted or unsubsituted oxanyl. In some embodiments, R2 is a substituted or unsubsituted 848 oxetanyl. In some embodiments, R2 is an unsubsituted morpholinyl. In some embodiments, 849 R2 is an unsubsituted oxanyl. In some embodiments, R2 is an unsubsituted oxetanyl. In some 850 embodiments, R2 is substituted or unsubstituted benzodioxinyl. In some embodiments, R2 is 851 substituted or unsubstituted yl. In some embodiments, R2 is unsubstituted 852 benzodioxinyl. In some embodiments, R2 is unsubstituted naphthyl. 853 [0081] Further to any embodiment above wherein the compound has the ure of 854 Formula (V), in some embodiments, L5 is a bond, or substituted or unsubstituted alkylene, 855 and R5 is substituted or unsubstituted aryl, tuted or unsubstituted fused ring aryl, 856 substituted or unsubstituted heterocycloalkyl, or tuted or unsubstituted 857 heterocycloalkyl. In some embodiments, R5 is substituted or unsubstituted phenyl, or 858 substituted or unsubstituted thienyl. In some embodiments, R5 is tituted phenyl. In 859 some embodiments, R5 is tituted thienyl. In some embodiments, R5 is a chloro- 860 substituted thienyl. In some embodiments, R5 is tuted or unsubstituted pyridyl, or 861 substituted or unsubstituted pyridazinyl. In some embodiments, R5 is unsubstituted pyridyl. In 862 some embodiments, R5 is unsubstituted pyridazinyl. In some embodiments, R5 is substituted 863 or unsubstituted pyrimidinyl, or substituted or unsubstituted furyl. In some embodiments, R5 864 is tituted pyrimidinyl. In some embodiments, R5 is unsubstituted furyl. In some 865 embodiments, R5 is substituted or unsubstituted morpholinyl, or substituted or unsubstituted 866 oxanyl, or substituted or unsubstituted yl. In some embodiments, R5 is unsubstituted 867 morpholinyl. In some embodiments, R5 is unsubstituted oxanyl. In some embodiments, R5 is 868 unsubstituted oxetanyl. In some ments, R5 is substituted or unsubstituted 869 benzodioxinyl, or substituted or unsubstituted yl. In some embodiments, R5 is 870 unsubstituted benzodioxinyl. In some embodiments, R5 is unsubstituted naphthyl. 871 [0082] Further to any embodiment above wherein the compound has the structure of 872 Formula (V), in some ments, L1 is -S-, -NR7-, substituted or unsubstituted alkylene, or 873 substituted or unsubstituted heteroalkylene, where R7 is as bed in formula Ia, and R1 is 874 hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted aryl, substituted or 875 unsubstituted fused ring aryl, substituted or unsubstituted heteroaryl, or substituted or 876 unsubstituted heterocycloalkyl. In some embodiments, R1 is substituted or unsubstituted 877 phenyl. In some embodiments is an R1 is tituted phenyl. In some embodiments, R1 is a 878 substituted or unsubsituted pyridyl. In some embodiments, R1 is a substituted or unsubsituted 879 pyridazinyl. In some embodiments, R1 is a substituted or unsubsituted pyrimidinyl. In some 880 embodiments, R1 is a substituted or unsubsituted thienyl. In some embodiments, R1 is a 881 substituted or unsubsituted fierl. In some embodiments, R1 is an ituted pyridyl. In 882 some embodiments, R1 is an unsubsituted pyridazinyl. In some embodiments, R1 is an WO 45986 883 ituted pyrimidinyl. In some embodiments, R1 is an unsubsituted thienyl. In some 884 embodiments, R1 is a chloro-substituted thienyl. In some embodiments, R1 is an unsubsituted 885 furyl. In some embodiments, R1 is a substituted or unsubsituted morpholinyl. In some 886 embodiments, R1 is a substituted or unsubsituted oxanyl. In some embodiments, R1 is a 887 substituted or unsubsituted oxetanyl. In some embodiments, R1 is an unsubsituted 888 morpholinyl. In some embodiments, R1 is an unsubsituted oxanyl. In some embodiments, R1 889 is an unsubsituted oxetanyl. In some embodiments, R1 is substituted or unsubstituted 890 benzodioxinyl. In some embodiments, R1 is substituted or unsubstituted naphthyl. In some 891 embodiments, R1 is unsubstituted benzodioxinyl. In some ments, R1 is tituted 892 naphthyl. 893 [0083] In some embodiments, there is provided a compound according to Formula (V) and 894 its listed embodiments, wherein L2 and R2 are absent, providing a compound with structure of 895 Formula (VI) following.
R5_L\5 O N \ R1 \ / \ 896 N,NH (VD 897 In some embodiments, the compound is a pharmaceutically acceptable salt, ester, solvate, or 898 g of a compound of Formula (VI). In some embodiments, the compound is not an 899 ester, not a solvate, and not a prodrug. 900 [0084] In some embodiments, there is provided a compound according to Formula (V) and 901 its listed embodiments, wherein L5 and R5 are absent, ing a compound with structure of 902 Formula (VII) following. 903 (v11) 904 [0085] In some embodiments, the compound is a ceutically acceptable salt, ester, 905 solvate, or prodrug of a nd of Formula (VII). In some embodiments, the compound is 906 not an ester, not a e, and not a prodrug. 907 [0086] Exemplary compounds, e.g., ubstituted aromatic compounds, in accordance 908 with the present disclosure are provided herein. In Table A, B, C, and D following, 909 compound (Cmpd) number, chemical name (i.e., International Union of Pure and Applied 910 Chemistry [IUPAC] name), calculated molecular weight (MW) and biological ty (i.e., 911 tion activity in thrombin, KLKl and KLKBl assays) are disclosed. 912 [0087] For Table A following, the disclosed compounds were assayed for inhibition of the 913 protease activity of thrombin as described herein. In Table A, the level of inhibition in the 914 thrombin assay is indicated as follows: a IC50 S 0.1 11M; b: 0.1 11M < IC50 < 1 11M; c: 1 11M < 915 IC50 < 10 11M; d: 10 11M < IC50 < 100 11M; e: IC50 2 100 11M. Accordingly, in some 916 ments, there is provided a compound as expressly set forth in Table A following. 917 Table A IUPAC name Activity 3-(5-[(5-chlorothiophenyl)methyl]amino(2,2- dimethylpropanoyl)- 11pyrazol-3 -y-l) 1 ,2-dihydropyridin 3-(5- [(5-chlorothiophenyl)methyl]amino [(furan yl)carbonyl]— l-Hpyrazol-3 -y-l) 1-(thiophenylmethyl)- 1 ,2- 3-(5- [(5-chlorothiophenyl)methyl]a]mino [(furan yl)carbonyl]— l-Hpyrazol-3 -y-l) idinylmethyl)- 1 ,2- 3 5-chlorothiophenyl)methyl]amino[(furan-3 - bonyl]—1H-pyrazol-3 -yl)methyl- 1,2-dihydropyridin- 3-(5- [(5-chlorothiophenyl)methyl]a]mino [(furan yl)carbonyl]— l-Hpyrazol-3 -y-l) 1-(pyridinylmethyl)- 1 ,2- 3-(5- [(5-chlorothiophenyl)methyl]a]mino [(furan yl)carbonyl]— azolyl)- 1-(furanylmethyl)- 1 ,2- 1- [(2-chlorophenyl)methyl]]—3-(5- [(5-chlorothiophen yl)methyl]amino(2,2-dimethylpropanoyl)- 11pyrazol 1- [(2-chlorophenyl)methyl]]—3-(5- [(5-chlorothiophen yl)methyl]amino [(-2methoxyphenyl)carbonyl]— 1H- 1- [(2-chlorophenyl)methyl]]—3 -(5 - [(5-chlorothiophen yl)methyl] amino- 1- [(furan-3 -yl)carbonyl]—1H-pyrazol-3 -yl) - 1,2-dih dro o idinone 1- [(2-chlorophenyl)methyl] -3 -(5 - [(5-chlorothiophen yl)methyl] amino- 1H-pyrazol-3 -yl)- 1 ,2-dihydropyridin 1 -1 ,2-dih dro. ridin-one 32 1- [(3-chloropheny1)methyl]]--3(5- [(5-chlorothiophen 5_— y1)methy1] amino- 1- thoxyphenyl)carbony1]— 1H- 33 1- [(3 -chloropheny1)methy1]—]3 -(5 - [(5-chlorothiophen y1)methy1] amino- 1- [(furan-3 -y1)carbony1]—1H-pyrazol-3 -y1)- 1,2-dih dro. ridinone 34 1- [(3 -chloropheny1)methy1]—3 -(5 - [(5-chlorothiophen hy1] amino- 1H-pyrazol-3 -y1)- 1 ,2-dihydropyridin 1- [(4-chloropheny1)methyl] -3 -(5 - lorothiophen yl)methy1]amino(2,2-dimethy1propanoy1)-1H-pyrazol-3 1,2-dih dro. ridinone 36 1- [(4-chloropheny1)methyl]]—3-(5- [(5-chlorothiophen y1)methy1] amino- 1- [(2-methoxyphenyl)carbony1]— 1H- 37 1- [(4-chloropheny1)methyl]]—3 -(5 - [(5-chlorothiophen y1)methy1] amino- 1- n-3 -y1)carbony1]—1H-pyrazol-3 -y1)- 1,2-dih dro. ridinone 3 8 1- [(4-chloropheny1)methyl] -3 -(5 - [(5-chlorothiophen y1)methy1] amino- 1H-pyrazol-3 -y1)- 1 ,2-dihydropyridin 39 1- [(5-chlorothiopheny1)methy1]]—3-(5- [(5-chlorothiophen- 2-y1)methy1] amino- 1-(22-dimethylpropanoyl)- 1H-pyrazol- 40 1- lorothiopheny1)methy1]]—3-(5- [(5-chlorothiophen- 2-y1)methy1] amino [(2-methoxyphenyl)carbony1]— 1H- 41 1- [(5 -chlorothiopheny1)methy1]]—3 -(5 - [(5-chlorothiophen- 2-y1)methy1] amino- 1-[(furan-3 -y1)carbony1]—1H-pyrazol-3 1,2-dih dro. ridinone 42 1- [(5 -chlorothiopheny1)methy1]-3 -(5 - [(5-chlorothiophen- ethy1] amino- 1H-pyrazol-3 -y1)- 1 ydropyridin 43 l-benzy1-3 -(5-[(5-chlorothiopheny1)methy1]amino(2,2- dimethylpropanoyl)- 1H-pyrazol-3 -y1)- 1,2-dihydropyridin 44 1 -b-enzy13-(5- lorothiopheny1)methy1]]amino- 1-(3- methoxy-2,2-dimethylpropanoy1)- 1H-pyrazol-3 -y-1) 1 2,- 45 l-benzy1-3 -(5- [(5-chlorothiopheny1)methy1] amino- 1- [(2,3 -dihydro-1,4-benzodioxiny1)carbony1]-1H-pyrazol-3 - 46 1 -b-enzy13-(5- [(5-chlorothiopheny1)methy1]]amino- 1- [(2,4-dimethoxyphenyl)carbony1]- 1H-pyrazol-3 -y-1) 1 ,2- 47 1 -b-enzy13-(5- [(5-chlorothiopheny1)methy1] amino[(2- methoxyphenyl)carbony1]— 1H-pyrazoly1)- 1 ,2- 48 1 -b-enzy13-(5- [(5-chlorothiopheny1)methy1] amino[(4- methyloxany1)carbony1]— 1H-pyrazol-3 -y-1) 1 ,2- 49 l-benzy1-3 -(5- [(5-chlorothiopheny1)methy1] amino- 1- [(fi11any1)carbony1]— 1H-pyrazol-3 -y1)-1,2-dihydropyridin- 2-one WO 45986 50 y1—3 -(5- [(5-chlorothiopheny1)methy1] amino [(furan-3 -y1)carbony1]—1H-pyrazol-3 -y1)-1,2-dihydropyridin- 51 1 -b-enzy13-(5- [(5-chlorothiopheny1)methy1]]amino [(thiopheny1)carbony1]- 11pyrazoly1)- 1 ,2- 52 1-benzy1—3 -(5- [(5-chlorothiopheny1)methy1] amino [(thiophen-3 -y1)carbony1]—1H-pyrazol-3 -y1)-1,2- dihydropyridinone 53 1 -b-enzy13-(5- [(5-chlorothiopheny1)methy1]]amino [4- (morpholiny1)pheny1]carbony1- 11pyrazoly1)- 1 ,2- 55 ] -b-enzy13 [(2-chloropheny1)carbonyl]]—5- [(5- chlorothiopheny1)methy1] amino- 11pyrazol-3 --y1 1 2,- 56 2- [3- (5- [(5-chlorothiopheny1)methy1]]amino(2,2- dimethylpropanoy1)- 11pyrazol-3 -y-1)2---oxo 1 ,2- 57 2- [3 5-chlorothiopheny1)methy1]amino[(2- methoxypheny1)carbony1] - 1H-pyrazol-3 -y1)oxo-1,2- dih dro o ridin 1]acetonitri1e 58 2- [3 -(5 - [(5 -chlorothiopheny1)methy1] amino [(furan—3 - y1)carbony1]—1H-pyrazol-3 -oxo-1,2-dihydropyridin 1]acetonitri1e 59 2- [3 -(5 - [(5 othiopheny1)methy1] amino- 1H-pyrazol- 3- 1oxo-1,2-dih dro. ridin-l- 1]acetic acid 6O 2- [3 -(5 - [(5 -chlorothiopheny1)methy1] amino- azol- 3-1 --oxo 1,2dih dro. ridin- 1- 1]acetonitri1e 61 3-(1 -b-enzoy15- [(5-chlorothiopheny1)methy1]amino-1H- 63 3-(1 -b-enzoy15- [(5-chlorothiopheny1)methy1]amino-1H- pyrazol-3 -y-1) 1-(furany1methy1)- 1 ,2dihydropyridin 64 3 -(1-benzoy1—5-[(5-chlorothiopheny1)methy1]amino-1H- pyrazol-3 -y1)(furan-3 hy1)- 1 ,2-dihydropyridin 65 3 -(1-benzoy1—5-[(5-chlorothiopheny1)methy1]amino-1H- pyrazol-3 -y1)(pyridazin-3 -y1methy1)- 1,2-dihydropyridin- 2-one 66 3 -(1-benzoy1—5-[(5-chlorothiopheny1)methy1]amino-1H- pyrazol-3 -y1)(pyridiny1methy1)-1,2-dihydropyridin 67 3 -(1-benzoy1—5-[(5-chlorothiopheny1)methy1]amino-1H- pyrazol-3 -y1)(pyridin-3 -y1methy1)-1,2-dihydropyridin 68 3 -(1-benzoy1—5-[(5-chlorothiopheny1)methy1]amino-1H- pyrazol-3 -y1)(pyridiny1methy1)-1,2-dihydropyridin 69 3-(1 -b-enzoy15- [(5-chlorothiopheny1)methy1]amino-1H- . razol1 o rimidin lmeth 1 -1 ,2dih dro. 3 -(1-benzoy1—5 - [(5-ch10r0thiopheny1)methy1]amino-1H- l-3 -y1)- 1-(thiopheny1methyl)-1,2-dihydropyridin(1-benzoy1—5 - [(5-ch10r0thiopheny1)methy1]amino-1H- pyrazol-3 -y1)- 1-(thiopheny1methy1)-1,2-dihydropyridin- 72 3-(1 -b-enz0y15- [(5-ch10r0thiopheny1)methy1]amino-1H- 74 3-(5- [(4-fluorophenyl)methyl] amino- 1- [(furan y1)carb0ny1]- 1-Hpyrazol-3 --y1) 1-(pyridinylmethy1)- 1 ,2- 75 3 -(5-[(5-ch10r0thiopheny1)methy1]amino(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y1)(2-meth0xyethyl)- 1,2-dih dro o idin0ne 76 3 -(5-[(5-ch10r0thiopheny1)methy1]amino(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y1)- 1 enylethy1)- 1,2-dih dro o idin0ne 77 3 -(5-[(5-ch10r0thiopheny1)methy1]amino(2,2- dimethylpropan0y1)- 1 H-pyrazol-3 -y1)(furany1methyl)- 1,2-dih dro. -0ne 78 3-(5- [(5-chlorothiopheny1)methy1]]amin0(22- dimethylpropanoyl} 1H-pyrazol-3 -y-1) 1-(furan-3 -y-1methy1) 1 ,2-dih droo ' 79 3-(5- [(5-chlorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- azol-3 -y-1) 1-(naphthalen- 1- 8O 3-(5- [(5-chlorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y-1) 1-(naphthalen 81 3-(5- [(5-chlorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y-1) 1-(pyridazin 82 3-(5- [(5-chlorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y-1) 1-(pyridin 83 3-(5- [(5-chlorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y-1) 1-(pyridin 84 3-(5- lorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y-1) 1-(pyridin 85 3-(5- lorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y-1) 1-(pyrimidin 86 3-(5- [(5-chlorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y-1) ophen—2- 87 3-(5- [(5-chlorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- 1H-pyrazol-3 -y-1) 1-(thiophen—3- 88 3-(5- [(5-chlorothiopheny1)methy1]]amin0(2,2- dimethylpropanoyl} azol-3 -y-1) 1- [2-(m0rph011n 89 3-(5-[(5-ch1010thi0pheny1)methy1]]amino(22- dimethylpropan0y1)- 1H-py1a201-y1) 1- [2-(m01ph01in 90 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(22- dimethylprpp-anoyl) 1H-py1azol-3 -y-1) 1--methy1 1 2,- 91 3 -(5 - [(5 -ch1010thi0pheny1)methy1]amino(3 -hyd10xy- methy1p10pan0y1)-1H-py1azol-3 -y1)-1,2- dihyd10py1idinone 92 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3-hyd10xy- 2,2-dimethylpropan0y1)- 1H-py1azol-3 -y-1) 1-(2- 93 3-(5- [(5-ch1010thi0pheny1)methy1] amino- yd10xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) 1-(furan—2- 94 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3-hyd10xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) 1-(py1idin 95 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3-hyd10xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) 1-(thiophen 96 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3-meth0xy- 2,2-dimethylpropan0y1)- azol-3 -y-1) 1 ,2- 97 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1H-py1azol-3 -y-1) 1-(2- 98 3-(5- [(5-ch1010thi0pheny1)methy1] amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) 1-(furan—2- 99 3-(5- 1010thi0pheny1)methy1]]amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1azol3 -y-1) 1-(furan—3- 100 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1azol3 -y-1) 1-(py1idazin 101 3-(5- 1010thi0pheny1)methy1]]amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) 1-(py1idin 102 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) 1-(py1idin 103 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) 1-(py1idin 104 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) 1-(py1imidin 105 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) ophen 106 3-(5- [(5-ch1010thi0pheny1)methy1]]amino- 1-(3 -meth0xy- 2,2-dimethylpropan0y1)- 1H-p-y1azol3 -y-1) 1-(thiophen 107 3-(5-[(5-chlorothiopheny1)methy1]]amino(3 -meth0xy- 22-dimethylpropan0y1)- 1H-pyrazol-3 --y1) 1- [2-(morpholin- 108 3-(5- lorothiopheny1)methy1]]amin0 [(2,3-dihydro- 1 ,4-benzodioxiny1)carb0ny1]- 1H-pyrazoly1)- 1 ,2- 109 3 -(5-[(5-chlorothiopheny1)methy1]amino[(2,3 -dihydro- 1,4-benzodi0xiny1)carb0ny1]-1H-pyrazoly1)(2- methoxyethy1)- 1 ,2-dihydropyridin0ne 110 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(2,3-dihydro- 1 ,4benzodioxiny1)carb0ny1]- 1H-pyrazol-3 -y-1) 1-(furan- 111 3-(5- lorothiopheny1)methy1]]amin0 [(2,3-dihydro- 1 ,4benzodioxiny1)carb0ny1]- 1H-pyrazol-3 -y-1) 1-(furan- 112 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(2,3-dihydro- 1 ,4benzodioxiny1)carb0ny1]- 1H-pyrazoly1)- 1- 113 3-(5-[(5-chlorothiopheny1)methy1]amino dihydro- 1 ,4benzodioxiny1)carb0ny1]- 1H-pyrazoly1)- 1- . ridin lmeth 1 -1,2-dih dro. ridin0ne 1 14 3 -(5-[(5-chlorothiopheny1)methy1]amino[(2,3 -dihydro- nzodi0xiny1)carb0ny1]-1H-pyrazoly1) o ridin lmeth 1 -1,2-dih dro. ridin0ne 1 15 3 -(5-[(5-chlorothiopheny1)methy1]amino[(2,3 -dihydro- 1,4-benzodi0xiny1)carb0ny1]-1H-pyrazoly1) o ridin lmeth 1 -1,2-dih dro. 2-0ne 1 16 3 -(5-[(5-chlorothiopheny1)methy1]amino[(2,3 -dihydro- 1,4-benzodi0xiny1)carb0ny1]-1H-pyrazoly1) o rimidin lmeth 1 -1,2-dih dro. 2-0ne 117 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(2,3-dihydro- 1 odioxiny1)carb0ny1]- 1H-pyrazoly1)- 1- 118 3-(5- 1010thi0pheny1)methy1]amin0 [(2,3-dihydro- 1 ,4benzodioxiny1)carb0ny1]- 1H-pyrazoly1)- 1- 119 3-(5- [(5-chlorothiopheny1)methy1]amin0 [(2,3-dihydro- 1 ,4benzodioxiny1)carb0ny1]- 1H-pyrazoly1)- 1- [2- 120 3 -(5-[(5-chlorothiopheny1)methy1]amino[(2,3 -dihydro- 1,4-benzodi0xiny1)carb0ny1]-1H-pyrazol-3 -y1)methy1— 1,2-dih dro. ridin0ne 121 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(2,4- dimethoxypheny1)carbony1]- 1H-pyrazol-3 -y-1) 1 ,2- 122 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(2,4- dimethoxypheny1)carbony1]- 1H-pyrazol-3 -y-1) 1-(2- 123 3-(5- [(5-chlorothiopheny1)methy1]amin0 [(2,4- dimethoxyphenyl)carb0ny1]— 1H-pyrazol-3 -y-1) 1-(furan 124 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(2,4- dimethoxyphenyl)carb0ny1]— 1H-pyrazol-3 -y-1) 1-(pyridazin- 125 3-(5- [(5-chlorothiopheny11methy1]1amin0- 1- [(2,4- oxypheny11carbonyl1— 1H-pyrazol-3 -y-11 idin 126 3-(5- [(5-chlorothiopheny11methy11amin0 [(2,4- dimethoxypheny11carbony11- 1H-pyrazol-3 -y-11 1-(pyridin 127 3 -(5-[(5-chlorothiopheny11methy1]amino[(2,4- dimethoxypheny11carbonyl] - 1H-pyrazol-3 -y11(pyridin ylmethy11- 1 ,2-dihydropyridin0ne 128 3-(5- [(5-chlorothiopheny11methy11amin0 [(2,4- oxypheny11carbony11- lH-pyrazol-3 -y-11 1-(pyrimidin- 129 3-(5- [(5-chlorothiopheny11methy11amin0 [(2,4- dimethoxypheny11carb0ny11- 1H-pyrazoly11- 1-(thiophen- 130 3-(5- [(5-chlorothiopheny11methy11amin0 [(2,4- dimethoxypheny11carb0ny11- 1H-pyrazoly11- 1-(thiophen- 13 1 3-(5- [(5-chlorothiopheny11methy11amin0[(2,4- dimethoxypheny11carbonyl]-1H-pyrazol-3 -y11- 1-[2- 132 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxypheny11carb0ny11- 1H-pyrazoly11- 1 ,2- 133 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxypheny11carb0ny11- 1H-pyrazoly11- 1-(2- 134 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxypheny11carb0ny11- 1H-pyrazoly11- 1-(2- 135 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxyphenyl1carb0ny11— 1H-pyrazoly11- 1-(furan 136 3-(5- lorothiopheny11methy11amin0 [(2- methoxyphenyl1carb0ny11— azoly11- an 137 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxypheny11carb0ny11- 1H-p-yrazol3-y11- 1-(naphthalen- 138 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxyphenyl1carb0ny11— 1H-pyrazoly11- 1-(pyridazin 139 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxyphenyl1carb0ny11— 1H-pyrazoly11- 1-(pyridin 140 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxyphenyl1carb0ny11— 1H-pyrazoly11- idin 141 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxyphenyl1carb0ny11— 1H-pyrazoly11- 1-(pyridin 142 3-(5- [(5-chlorothiopheny11methy11amin0 [(2- methoxypheny11carb0ny11- 1H-pyrazoly11- 1-(pyrimidin 143 3-(5-[(5-ch1010thi0pheny1)methy1]]amino [(2- methoxyphenyl)ca1b0ny1]— 1H-py1azoly1)- ophen 144 3-(5-[(5-ch1010thi0pheny1)methy1]]amino [(2- methoxyphenyl)ca1b0ny1]— 1H-py1azoly1)- 1-(thiophen 145 3 -(5-[(5-ch1010thi0pheny1)methy1]]amino[(2- methoxypheny1)ca1b0ny1] - 1 H-py1azol-3 -y1)[2- (m01ph01iny1)0xoethy1]-1,2-dihyd10py1idin—2-0ne 146 3 5-ch1010thi0pheny1)methy1]amino[(2- methoxypheny1)ca1b0ny1] - 1 H-py1azol-3 -y1)[2- 147 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(2- yphenyl)ca1b0ny1]— 1H-py1azoly1)- 1--methy1 1 2- 148 3 -(5-[(5-ch1010thi0pheny1)methy1]amino[(3 - methyloxetan-3 -y1)ca1b0ny1]— 1H-py1azol-3 -y1)methy1— 1,2-dih d10o1idin0ne 149 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(4- methyloxany1)ca1b0ny1]- 1H-py1azol-3 y-1)- 1-(2- 150 3-(5- [(5-ch1010thi0pheny1)methy1]amin0 [(4- methyloxany1)ca1b0ny1]— 1-Hpy1azol-3 y-1)- 1-(furan 151 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(4- methyloxany1)ca1b0ny1]— 1-Hpy1azol-3 -y-1) 1-(furan 152 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(4- methyloxany1)ca1b0ny1]— 1H-py1azol-3 -y-1) 1-(pyridazin- 153 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(4- methyloxany1)ca1b0ny1]— 1-Hpy1azol-3 -y-1) 1-(py1idin 154 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(4- methyloxany1)ca1b0ny1]— 1-Hpy1azol-3 -y-1) 1-(py1idin 155 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(4- methyloxany1)ca1b0ny1]— 1-Hpy1azol-3 -y-1) 1-(py1idin 156 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(4- methyloxany1)ca1b0ny1]— 1H-py1azol-3 -y-1) 1-(pyrimidin- 157 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(4- oxany1)ca1b0ny1]— 1-Hpy1azol-3 -y-1) 1-(thiophen 158 3-(5- 1010thi0pheny1)methy1]]amin0 [(4- methyloxany1)ca1b0ny1]— 1-Hpy1azol-3 -y-1) ophen 159 3 -(5 - [(5 -ch1010thi0pheny1)methy1]]amino[(4- methyloxany1)ca1b0ny1]-1H-py1az01—3 -y1)[2- 160 3-(5- [(5-ch1010thi0pheny1)methy1]]amin0 [(fu1an y1)ca1b0ny1]— azol-3 -y-1) 1-(2-methoxyethy1)- 1 2- 161 (5-chlorothiopheny1)methy1]]amino [(furan b0ny1]— 1-Hpyrazoly1)- 1-(furany1methy1)- 1 ,2- 162 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(furan y1)carb0ny1]— 1-Hpyrazoly1)- an-3 thy1) 1 ,2- 163 3 -(5 - [(5 oth10pheny1)methy1] amino [(furan y1)carb0ny1]—1H-pyrazol-3 -y1)(pyridazin-3 -y1methy1)-1,2- dihydropyridinone 164 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(furan y1)carb0ny1]- 1-Hpyrazol-3 y-1)- 1-(pyridiny1methy1)- 1 ,2- 165 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(furan y1)carb0ny1]- 1-Hpyrazol-3 -y-1) 1-(pyridin-3 -y-1methy1) 1 ,2- 166 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(furan y1)carb0ny1]- 1-Hpyrazol-3 -y-1) 1-(pyridiny1methy1)- 1 ,2- 167 3 -(5 - [(5 -chloroth10pheny1)methy1] amino n y1)carb0ny1] - 1 H-pyrazol-3 -y1)(pyrimidiny1methy1)- 1,2-dih dro. ridin0ne 168 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(furan b0ny1]— 1-Hpyrazol-3 -y-1) 1-(thiopheny1methy1)- 1 ,2- 169 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(furan y1)carb0ny1]— 1-Hpyrazol-3 -y-1) 1-(thiopheny1methy1)- 1 ,2- 170 3 -(5 - [(5 oth10pheny1)methy1] amino [(furan y1)carb0ny1]—1H-pyrazol-3 -y1)[2-(m0rpholiny1)ethy1]- 1,2-dih dro. ridin0ne 171 3-(5-[(5-chloroth10pheny1)methy1]]amin0 [(furan y1)carb0ny1]— 1-Hpyrazol-3 -y-1) 1-(2-methoxyethy1)- 1 ,2- 172 3-(5-[(5-chloroth10pheny1)methy1]]amin0 [(furan y1)carb0ny1]— azol-3 -y-1) 1-(2-phenylethy1)- 1 ,2- 173 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(furan y1)carb0ny1]— 1-Hpyrazoly1)- 1-(furan-3 -y-1methy1) 1 ,2- 174 3 -(5 - [(5 -chloroth10pheny1)methy1] amino [(furan-3 - y1)carb0ny1]—1H-pyrazol-3 -y1)(naphthaleny1methy1)- 1,2-dih dro. ridin0ne 175 3 -(5 - [(5 -chloroth10pheny1)methy1] amino [(furan-3 - y1)carb0ny1]—1H-pyrazol-3 -y1)(naphthaleny1methy1)- 1,2-dih dro. ridin0ne 176 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(furan-3 - y1)carb0ny1]— 1Hpyrazol-3 -y-1) 1-(pyridazin-3 -y-1methy1) 1 ,2- 177 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(furan y1)carb0ny1]- 1-Hpyrazol-3 -y-1) 1-(pyridin-3 -y-1methy1) 1 ,2- 178 3 -(5 - [(5 -chloroth10pheny1)methy1] amino [(furan-3 - y1)carb0ny1] - 1 H-pyrazol-3 -y1) - 1 midiny1methy1)- 1,2-dih dro o ridin0ne 179 3-(5-[(5-chlorothiopheny1)methy1]]amino [(furan y1)carb0ny1]— 1-Hpyrazol-3 --y1) 1-(thiopheny1methy1)- 1 ,2- 180 3-(5-[(5-ch101‘0th10pheny1)methy1]amino[(furan b0ny1]—1H-pyrazol-3 -y1)[2-(m0rph01in—4-y1) oxoeth 1]-1,2-dih dro o ridin0ne 181 (5-chlorothiopheny1)methy1]amino[(furan y1)carb0ny1]—1H-pyrazol-3 -y1)[2-(m0rph01in—4-y1)ethy1]- 1 ydropyridin0ne 182 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen y1)carb0ny1]— 1-Hpyrazol-3 y-1)- 1-(2-methoxyethy1)- 1 ,2- 183 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen y1)carb0ny1]— azoly1)- 1-(furany1methy1)- 1 ,2- 184 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen y1)carb0ny1]— 1-Hpyrazoly1)- 1-(furan-3 -y-1methy1) 1 ,2- 185 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen y1)carb0ny1]— 1-Hpyrazol-3 -y-1) 1-(pyridazin-3 -y-1methy1) 1 ,2- 186 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen y1)carb0ny1]- azol-3 -y-1) 1-(pyridiny1methy1)- 1 ,2- 187 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen y1)carb0ny1]- 1-Hpyrazol-3 -y-1) 1-(pyridin-3 -y-1methy1) 1 ,2- 188 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen b0ny1]- 1-Hpyrazol-3 -y-1) 1-(pyridiny1methy1)- 1 ,2- 189 3 -(5-[(5-chlorothiopheny1)methy1]amino[(thi0phen y1)carb0ny1] - 1 H-pyrazol-3 -y1)(pyrimidiny1methy1)- 1,2-dih dro. ridin0ne 190 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen y1)carb0ny1]— 1-Hpyrazol-3 -y-1) 1-(thiopheny1methy1)- 1 ,2- 191 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen y1)carb0ny1]— 1-Hpyrazol-3 -y-1) 1-(thiopheny1methy1)- 1 ,2- 192 3 -(5-[(5-chlorothiopheny1)methy1]amino[(thi0phen y1)carb0ny1]—1H-pyrazol-3 -y1)[2-(m0rph01in—4-y1)ethy1]- 1 ,2dih dro o ridin-0ne 194 3-(5- [(5-chlorothiopheny1)methy1]amin0 [(thi0phen b0ny1]— 1-Hpyrazol-3 -y-1) 1-(2-methoxyethy1)- 1 ,2- 195 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen y1)carb0ny1]— 1-Hpyrazoly1)- any1methy1)- 1 ,2- 196 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thi0phen b0ny1]— 1-Hpyrazoly1)- 1-(furan-3 -y-1methy1) 1 ,2- 198 3-(5-[(5-chlorothiopheny1)methy1]]amino [(thiophen y1)carb0ny1]- 1-Hpyrazol-3 --y1) 1-(pyridiny1methy1)- 1 ,2- 199 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thiophen y1)carb0ny1]- 1-Hpyrazol-3 --y1) 1-(pyridin-3 -y-1methy1) 1 ,2- 200 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thiophen y1)carb0ny1]- 1-Hpyrazol-3 -y-1) 1-(pyridiny1methy1)- 1 ,2- 201 3 -(5-[(5-chlorothiopheny1)methy1]amino[(thiophen-3 - y1)carb0ny1] - 1 H-pyrazol-3 -y1)(pyrimidiny1methy1)- h dro. ridin0ne 202 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(thiophen y1)carb0ny1]— 1-Hpyrazol-3 -y-1) 1-(thiopheny1methy1)- 1 ,2- 203 3-(5- lorothiopheny1)methy1]]amin0 [(thiophen b0ny1]— 1-Hpyrazol-3 -y-1) 1-(thiopheny1methy1)- 1 ,2- 204 3 -(5-[(5-chlorothiopheny1)methy1]amino[(thiophen-3 - y1)carb0ny1]—1H-pyrazol-3 -y1)[2-(morpholiny1)ethy1]- h dro. ridin0ne 205 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [2- (morpholiny1)pheny1]carbonyl- 1H-pyrazol-3 -y-1) 1 ,2- 206 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [4-(2- methoxyethoxy)pheny1]carbonyl1-pyrazol-3 -y-1) 1 ,2- 207 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [4- (morpholiny1)pheny1]carbonyl- 1H-pyrazol-3 -y-1) 1 ,2- 208 3-(5- [(5-chlorothiopheny1)methy1]]am-ino 1- [4- oliny1)pheny1]carbonyl- 1H-pyrazoly1)- 1-(2- 209 3-(5- [(5-chlorothiopheny1)methy1]amin0 [4- (morpholiny1)pheny1]carbonyl- 1H-pyrazoly1)- 1- furan lmeth 1 -1,2-dih dro. ridin0ne 210 3 -(5-[(5-chlorothiopheny1)methy1]amino[4- (morpholiny1)pheny1]carbonyl-1H-pyrazol-3 -y1) furan lmeth 1 -1,2-dih dro. 2-0ne 211 3-(5- [(5-chlorothiopheny1)methy1]]am-ino 1- [4- (morpholiny1)pheny1]carbonyl- 1H-pyrazoly1)- 1- 212 3-(5-[(5-chlorothiopheny1)methy1]amino [4- olinyl)pheny1]carb0ny1— 1Hpyrazoly1)- 1- . ridin lmeth 1 -1,2-dih dro o ridin0ne 213 3 -(5-[(5-chlorothiopheny1)methy1]amino[4- (morpholiny1)pheny1]carbonyl-1H-pyrazol-3 -y1) o ridin-3 - lmeth 1 -1,2-dih dro o ridin0ne 214 3 -(5-[(5-chlorothiopheny1)methy1]amino[4- (morpholiny1)pheny1]carbonyl-1H-pyrazol-3 -y1) o ridin lmeth 1 -1,2-dih dro o ridin0ne 215 3 -(5-[(5-chlorothiopheny1)methy1]amino[4- (morpholiny1)pheny1]carbonyl-1H-pyrazol-3 -y1) o rimidin lmeth 1 ih dro o ridin0ne 2014/030853 216 3-(5-[(5-chlorothiopheny1)methy1]]amino [4- (morpholinyl)pheny1]carb0ny1— 1H-pyrazoly1)- 1- 217 3-(5- [(5-chlorothiopheny]1)methy1 amino [4- oliny1)pheny1]carbonyl- 1H-pyrazoly1)- 1- 218 3-(5- [(5-chlorothiopheny1)methy1] amino- 1- [4- (morpholiny1)pheny1]carbonyl- 1H-pyrazoly1)- 1-[2- (morpholinyl)ethyl]- 1 ,2-dihydropyridin0ne 219 3-(5-[(5-chlorothiopheny1)methy1]amino-1H-pyrazol y1)(1H-1,2,3 ,4-tetrazoly1methy1)- 1,2-dihydropyridin- 220 3-(5- [(5-chlorothiopheny1)methy1] amino- 1H-pyrazol 233 3-(5-[(5-ch101‘0th10pheny1)methy1amino- 1H-pyrazol y1)[2-(m0rpholiny1)0xoethy1]-1,2-dihydropyridin 23 7 3 -[5-(benzy1amin0)[(fi11anyl)carb0nyl]-1H-pyrazol-3 - 1]- 1 2-dih dro. ridinone 239 3-[5-(dimethylamin0)- 1- [(furanyl)carb0ny1]—1H-pyrazol- 3- l]—1,2-dih dro. ridin—2-0ne 240 3- [5-(dimethy1amin0)- l- nyl)carb0nyl]— 1H-pyrazol- 242 3 [(2-chlorophenyl)carb0nyl] [(5-chlorothiophen y1)methyl]amin0-1H-pyrazol-3 -y-l l-(2-methoxyethyl)- l ,2- 243 3 - l - [(2-chlorophenyl)carb0nyl] [(5-chlorothiophen yl)methyl] amino- 1 zol-3 -yl-l-(fi11anylmethyl)-l,2- dihydropyridinone 244 3 lorophenyl)carb0nyl] [(5-chlorothiophen y1)methyl]amin0-1H-pyrazol-3 -y-l an-3 -y-lmethyl) l ,2- 245 3 - l - [(2-chlorophenyl)carb0nyl] [(5-chlorothiophen yl)methyl] amino- 1 H-pyrazol-3 -yl- l -(pyridazin-3 -ylmethyl) 1,2-dih dro. -0ne 246 3 - l - [(2-chlorophenyl)carb0nyl] lorothiophen yl)methyl] amino- lH-pyrazol-3 -yl- l -(pyridinylmethyl)- 1,2-dih dro. idin0ne 247 3 - l - [(2-chlorophenyl)carb0nyl] [(5-chlorothiophen yl)methyl] amino- azol-3 -yl- l -(pyridin-3 -ylmethyl)- 1,2-dih dro. idin0ne 248 3 [(2-chlorophenyl)carb0nyl] [(5-chlorothiophen yl)methyl]amin0-1H-pyrazol-3 -y-l l-(pyridinylmethyl)- l ,2-dih droo ' 249 3 [(2-chlorophenyl)carb0nyl] [(5-chlorothiophen yl)methyl]amin0-1H-pyrazol-3 -y-l l-(pyrimidin 250 3 - l - [(2-chlorophenyl)carb0nyl]]—5- [(5-chlorothiophen yl)methyl] amino- lH-pyrazol-3 -yl- l -(th10phenylmethyl)- 1,2-dih dro. idin0ne 251 3 [(2-chlorophenyl)carb0nyl] [(5-chlorothiophen yl)methyl]amin0-1H-pyrazol-3 -y-l l-(thiophen-3 -y-lmethyl) 252 3 [(2-chlorophenyl)carb0nyl] [(5-chlorothiophen yl)methyl]amin0-1H-pyrazol-3 -y-l l- rpholin 254 3 [(4-tert-butylphenyl)carb0nyl]]—5- [(5-chlorothiophen y1)methyl]amin0-1H-pyrazol-3 -y-l 1 --methyl 1 ,2- . ridin lmeth l -l,-2dih dro. 257 ethyl 2- [3-(5- [(5-chlorothiophenyl)methyl] amino-l-(2,2- dimethylpropanoyl)- l H-pyrazol-3 -yl)0x0- l ,2- dih dro. 'din-l- l]acetate 258 ethyl 2-[3-(5- [(5-chlorothiophenyl)methyl]amino- l -[(2- methoxyphenyl)carb0nyl] - lH-pyrazol-3 -0x0- l ,2- dih dro. 'din-l- l]acetate 259 ethyl 2-[3-(5- [(5-chlorothiophenyl)methyl]amino- l - [furan lcarbon l]—lH- azol l0x0-l,2- dih dro. ridin-l- l]acetate 261 tert—butyl 2- [3-(5- [(5-chlorothiophenyl)methyl]]amino-l- methylpropanoyl)- 1H-pyrazol-3 --yl)2---oxo l ,2- ridin- l— l]acetate 262 tert—butyl 2- [3-(5- lorothiophenyl)methyl]a]mino- l- [(2-methoxyphenyl)carbonyl]— 1H-pyrazol-3 --yl)2---oxo l 2,- ridin- l— l]acetate 263 tert—butyl 2- [3-(5- [(5-chlorothiophenyl)methyl]]amino-l- [(furan-3 -yl)carbonyl]— azol-3 -y-l)2-0x0- 1 ,2- 265 l- [(5 - [(5 -chlorothiophenyl)methyl] amino-3 -(l -methyl 0x0- 1 ,2-dihydropyridin-3 -yl)- l H-pyrazol- l - yl)carbonyl]cyclopropylmethyl l- h drox meth l c clo o ro o ane- l -carbox late 919 [0088] For Table B following, the disclosed compounds were assayed for inhibition of the 920 protease activity of KLKl and KLKBl as described herein. In Table B, C, and D, the level of 921 inhibition in the KLKl and KLKBl assays are ted as follows: a: IC50 S 0.1 11M; b: 0.1 922 11M < IC50 <1 11M;c: 111M < IC50 <10 11M; d: 10 11M < IC50 < 100 11M; e: IC50 2100 11M. 923 Accordingly, in some embodiments, there is provided a nd as expressly set forth in 924 Table B following. 3-(5- [(5-chlorothiophenyl)methyl] amino- 1- [(furan yl)carbonyl]— 1H-pyrazol-3 -y-l) l-(thiophenylmethyl)- l,2-dih dro. ridinone 3-(5- [(5-chlorothiophenyl)methyl]a-]mino l- [(furan yl)carbonyl]— 1-Hpyrazol-3 -y-l) l -m-ethyl l ,2- l-(5 - [(5 othiophenyl)methyl] aminofluoro-3 - (piperidinyl)- l H-pyrazol- l -yl)-2,2-dimethylpropanone 3 -(5 - [(5 -chlorothiophenyl)methyl] amino [(furan-3 - yl)carbonyl] - l H-pyrazol-3 -yl)- l -(pyridinylmethyl)- 1,2-dih dro. ridinone 3 -(5 - [(5 -chlorothiophenyl)methyl] amino [(furan-3 - o razol l - l - furan lmeth l - l ,2- dih dro. ridin0ne -_ 46 1 -b-enzy13-(5- 1010thi0pheny1)methy1]]amino- 1- 561 d [(2,4-dimethoxypheny1)carbony1]- 11pyrazol-3 --y1) 1 ,2- 48 ] -b-enzy13-(5-[(5-ch1010thi0pheny1)methy1]]amin0 523 e [(4-methyloxany1)carb0ny1]- 11pyrazol-3 --y1) 1 ,2- 49 1-benzy1—3 -(5- [(5-ch1010thi0pheny1)methy1] amino 491 b [(fiirany1)carb0ny1]-1H-pyrazol-3 -y1)-1,2- dihydropyridinone 50 1 -b-enzy13-(5- [(5-chlorothiopheny1)methy1]]amin0 49 1 d [(furan-3 -y1)carb0ny1]— 11pyrazol-3 -y-1) 1 2,- 51 1 -b-enzy13-(5-[(5-ch1010thi0pheny1)methy1]]amin0 507 e [(thiopheny1)carb0ny1]- 11pyrazoly1)- 1 ,2- 52 ] -b-enzy13-(5- [(5-ch1010thi0pheny1)methy1]]amin0 507 b phen-3 -y1)carb0ny1]— 11pyrazoly1)- 1 ,2- 53 1-benzy1—3 -(5- [(5-ch1010thi0pheny1)methy1] amino 5 86 [4-(m01ph01iny1)pheny1]carbonyl- 1H-pyrazol-3 -y1)- 1,2-dih dro. ridin0ne 59 ' ' —- I- 62 3-(1 0y15- [(5-ch1010thi0pheny1)methy1]amin0- 469 0 azoly1)- 1-(2-methoxyethy1)- 1 ,2- 73 I-e 77 3-(5- [(5-chlorothiopheny1)methy1]amin0(2,2- dimethylpropan0y1)- 11pyrazol-3 -y-1) 1-(furan 81 3-(5- [(5-chlorothiopheny1)methy1]]amin0(2,2- dimethylpropan0y1)- 11pyrazol-3 -y-1) 1-(pyridazin 92 3-(5- [(5-chlorothiopheny1)methy1]]amin0(3- hydroxy-2,2-dimethylpropanoyl} 11pyrazol-3 -y-1) 1-(2- 93 3-(5- lorothiopheny1)methy1]amin0(3- hydroxy-2,2-dimethylpropan0y1)- 11pyrazol-3 -y-1) 1- 94 3 -(5 - [(5 othiopheny1)methy1]amino(3 - hydroxy-2,2-dimethy1propan0y1)- 1 H-pyrazol-3 -y1) 97 3-(5- [(5-chlorothiopheny1)methy1]]amin0(3- methoxy-2,2dimethylpropan0y1)- 1H-pyrazol-3 -y-1) 1- 98 3 -(5-[(5-chlorothiopheny1)methy1]amino(3 - methoxy-2,2-dimethy1propan0y1)-1H-pyrazol-3 -y1) 100 3-(5- [(5-chlorothiopheny1)methy1]]amin0(3- methoxy-2,2dimethylpropan0y1)- 11pyrazol-3 -y-1) 1- 101 3- 5- [5-chlorothiOohen lmeth 1]am4in0 3- -_ d methoxy-2,2-dimethy1propan0y1)-1H-pyrazol-3 -y1) o 2- lmeth 1 -1,2-dih dro o ridin0ne 102 3 -(5-[(5-chlorothiopheny1)methy1]amino(3 - methoxy-2,2-dimethy1propan0y1)-1H-pyrazol-3 -y1) o ridin-3 - lmeth 1 -1,2-dih dro o ridin0ne 103 3 -(5-[(5-chlorothiopheny1)methy1]amino(3 - y-2,2-dimethy1propan0y1)-1H-pyrazol-3 -y1) o 4- lmeth 1 -1,2-dih dro o ridin0ne 104 3 5-chlorothiopheny1)methy1]amino(3 - methoxy-2,2-dimethy1propan0y1)-1H-pyrazol-3 -y1) (pyrimidiny1methy1)- 1 ydropyridin0ne 105 3-(5-[(5-chlorothiopheny1)methy1]]amino(3- methoxy-2,2-dimethylpropanoyl} 1H-pyrazol-3 -y-1) 1- 106 3-(5- [(5-chlorothiophehy1)methy1]amin0(3- y-2,2-dimethylpropan0y1)- 1H-pyrazol-3 -y-1) 1- 109 3-(5- [(5-chlorothiopheny1)methy1]amin0 [(2,3- dihydro- 1 ,4-benzodioxiny1)carb0ny1]- 1H-pyrazol 113 3-(5- [(5-chlorothiopheny1)methy1]amin0 [(2,3- dihydro- 1 ,4-benzodioxiny1)carb0ny1]- 1H-p-yrazol3- 116 3-(5- [(5-chlorothiopheny1)methy1]amin0 [(2,3- dihydro- 1 ,4-benzodioxiny1)carb0ny1]- 1H-pyrazol 117 3-(5- [(5-chlorothiopheny1)methy1]amin0[(2,3- dihydro-1,4-benzodioxiny1)carb0ny1]—1H-pyrazol-3 - 122 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(2,4- dimethoxypheny1)carbony1]- 1H-pyrazol-3 -y-1) 1-(2- 123 3-(5- [(5-chlorothiopheny1)methy1]amin0 [(2,4- dimethoxypheny1)carbony1]- 1H-pyrazol-3 -y-1) 1-(furan— 125 3-(5- [(5-chlorothiopheny1)methy1]]amin0[(2,4- dimethoxypheny1)carbonyl] - 1 H-pyrazol-3 -y1) o ridin lmeth 1 -1,2-dih dro o ridin0ne 126 3 -(5-[(5-chlorothiopheny1)methy1]amino[(2,4- dimethoxypheny1)carbonyl] - 1 H-pyrazol-3 -y1) o ridin-3 - lmeth 1 -1,2-dih dro o ridin0ne 13 1 3 -(5-[(5-chlorothiopheny1)methy1]amino[(2,4- dimethoxypheny1)carbonyl]-1H-pyrazol-3 -y1) [2- 132 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(2- methoxyphenyl)carb0ny1]— 1H-pyrazoly1)- 1 ,2- 138 3-(5- [(5-chlorothiopheny1)methy1]]amin0 [(2- yphenyl)carb0ny1]— 1H-pyrazoly1)- 1- 140 3-(5-[(5-chlorothiopheny1)methy1]amino [(2- methoxyphenyl)carb0ny1]— 1H-p-yrazol3-y1)- 1-(pyridin- 149 3-(5-[(5-chlorothiopheny1)methy1]]amino [(4- methyloxanyl)carb0ny1]— lH-pyrazol-3 --y1) 1-(2- 157 3-(5-[(5-chlorothiopheny1)methy1]amino [(4- methyloxanyl)carb0ny1]— 1H-pyrazol-3 --y1) 1- 160 3-(5- [(5-chlorothiophehy]1)methy1 amino[(furan y1)carb0ny1] - 1 H-pyrazol-3 -y1)(2-meth0xyethyl)-l,2- dihydropyridinone 179 3 -(5-[(5-chlorothiopheny1)methy1]amino[(furan-3 - b0ny1] - 1 zol-3 -y1)(thiophen-3 -y1methy1)- 1,2-dih dro. ridin0ne 182 3-(5- [(5-chlorothiopheny1)methy1]]amino- 1- [(thiophenyl)carb0ny1]— 1H-pyrazoly1)- 1-(2- 184 3-(5- lorothiopheny1)methy1] amino- 1- [(thiophenyl)carb0ny1]— lH-pyrazol-3 -y-1) 1-(furan 194 3-(5- [(5-chlorothiopheny1)methy1]]amino- 1- [(thiophen-3 -y1)carb0ny1]— 1H-pyrazoly1)- 1-(2- 196 3-(5- [(5-chlorothiopheny1)methy1] amino- 1- [(thiophen-3 -y1)carb0ny1]— lH-pyrazol-3 -y-1) an 198 3-(5- [(5-chlorothiopheny1)methy1]]amino- 1- [(thiophen-3 -y1)carb0ny1]— 1H-p-yrazol3 -y-1) 1-(pyridin- 201 3-(5- [(5-chlorothiopheny1)methy1]]amino [(thiophen-3 -y1)carb0ny1]—1H-pyrazol-3 -y1) 203 3-(5- [(5-chlorothiopheny1)methy1]]amino- 1- [(thiophen-3 -y-1)carb0ny1] lH-pyrazoly-1) 1- 208 3-(5- [(5-chlorothiopheny1)methy1] amino- 1- [4- (morpholinyl)pheny1]carb0ny1-1H-pyraz01—3-y1)- 1- 210 3 -(5-[(5-chlorothiopheny1)methy1]amino[4- (morpholinyl)phenyl]carbonyl-1H-pyrazol-3 -y1) furan-3 - lmeth 1 -1,2-dih dro o ridin0ne 213 3 -(5-[(5-chlorothiopheny1)methy1]amino[4- (morpholinyl)phenyl]carbonyl-1H-pyrazol-3 -y1) o ridin-3 - lmeth 1 -1,2-dih dro o ridin0ne 214 3 -(5-[(5-chlorothiopheny1)methy1]amino[4- (morpholinyl)phenyl]carbonyl-1H-pyrazol-3 -y1) o ridin lmeth 1 -1,2-dih dro o ridin0ne 21 8 3-(5- [(5-chlorothiopheny1)methy1]]amino- 1- [4- oliny1)pheny1]carbonyl- 1H-pyrazoly1)- 1- 235 3- 5-[5-ch101‘0th10oh-en2- lmeth l]am41n0-1H-o razol- -_ d 238 3- [5-(benzy1amin0)- 1- [(furanyl)carb0ny1]—1H- pyrazol-3 -y1]](pyridinylmethy1)- 1 2- 242 3 [(2-chlorophenyl)carb0ny1][(5-chlorothiophen y1)methy1] amino- 1H-pyrazol-3 -y1—1-(2-meth0xyethy1)- 1,2-dih dro. ridin0ne 243 3 [(2-chlorophenyl)carb0ny1][(5-chlorothiophen y1)methy1] amino- 1H-pyrazol-3 -y1—1-(filranylmethyl)- 1,2-dih dro. 2-0ne 244 3 [(2-chlorophenyl)carb0ny1][(5-chlorothiophen y1)methy1] amino- azol-3 -y1—1-(furan-3 -y1methy1)- 1,2-dih dro. ridin0ne 247 3 lorophenyl)carb0ny1]]—5- [(5-chlorothiophen y1)methy1] amino- 1H-pyrazol-3 -y-1 idin-3 - 248 3 [(2-chlorophenyl)carb0ny1]]—5- lorothiophen y1)methy1] amino- 1H-pyrazol-3 -y-1 1-(pyridin 265 1- [(5- [(5-chlorothiopheny1)methy1] amino-(1- methyl--0x0 1 2-dihydropyridin-3 -y-1) 1H-pyrazol- 1- y1)carbonyl]cyclopropylmethyl 1- 269 1-(5 - [(5 -chlorothiopheny1)methy1] amino-3 -(3 - fluoropyridin—2-yl)-1H-pyrazoly1)-2,2- dimeth 1.10 u an0ne 272 1-(5 - [(5 -chlorothiopheny1)methy1] amino-3 -(pyridin- 2-y1)- 1H-pyrazolmeth0xy-2,2-dimethylpropan- ] -0ne 273 1-(5 - [(5 -chlorothiopheny1)methy1] aminofluoro-3 - (0xany1)-1H-pyrazoly1)-3 -hydroxy-2,2- dimeth 1.10 u an0ne 274 1-(5 - [(5 -chlorothiopheny1)methy1] aminofluoro-3 - (piperidiny1)-1H-pyrazolyl)-3 -hydroxy-2,2- dimeth 1.10 u an0ne 275 1-(5 - [(5 -chlorothiopheny1)methy1] aminofluoro-3 - phenyl-lH-pyrazoly1)-3 -hydroxy-2,2- dimeth 1.10 u an0ne 276 1- [(2-aminopheny1)carb0nyl]-N- [(5-chlorothiophen 1 meth 1] -3 fluoro o ridin 1 -1H- o raz01amine 277 1- [(2-aminopheny1)carb0nyl]-N- [(5-chlorothiophen o raz01amine 278 1- [(2-meth0xyphenyl)carbonyl] -3 -(pyridiny1)-N— thio o hen lmeth 1 -1H- o razolamine 279 l- [5 -(benzylamino)fluoro-3 -(pyridinyl)-1H- azol- l - l]-2,2-dimeth 1.10 u anone 280 1-5 - [(furanylmethyl)amino] -3 -(pyridinyl)-1H- azol- 1- 1dimeth loro oan- 1--one 28 l 282 2-(5 - [(5 -chlorothiophenyl)methyl] amino [(2- methoxyphenyl)carbonyl] - l H-pyrazol-3 -yl)cyclohexan- 1-01 283 \ -(furanylmethyl) - l - [(2-methoxyphenyl)carbonyl] (pyridinyl) - l zol-S-amine 284 \ - [(5 -chlorothiophen—2-yl)methyl] - l - [(2- methoxyphenyl)carbonyl] (oxanyl)- lH-pyrazol-S - amine 285 \ - [(5 -chlorothiophen—2-yl)methyl] - l - [(2- methoxyphenyl)carbonyl] -3 -(pyridinyl) - l H-pyrazol amine 286 \ - [(5 othiophen—2-yl)methyl] - l - [(2- phenoxyphenyl)carbonyl] -3 -(pyridinyl)- l H-pyrazol a[mine 290 \ - [(5 -chlorothiophenyl)methyl] -]3 -(3 -fluoropyridin- 2-yl)- l - [(2-methoxyphenyl)carbonyl] - l zol-S - amine 29 l 296 \-benzylfluoro- l - [(2-methoxyphenyl)carbonyl] o hen l- l H- o razol-S-amine 297 tert-butyl 4-(5 - [(5-chlorothiophenyl)methyl] amino imethylpropanoyl)- l H-pyrazol-3 -yl)piperidine- l - carbox late 298 tert-butyl 4-(5 - [(5-chlorothiophenyl)methyl] amino [(thiophen-3 -yl)carbonyl] - l H-pyrazol-3 -yl)piperidine- l - carbox late 927 [0089] In some ments, there is provided a compound as expressly set forth in Table 928 C following. 929 Table C KLKl KLKB 1 Cmpd IUPAC name MW Activity Activity 1-(5- [(4-flu010pheny1)methy1] amin0-(py1idin-3 --y1) --- 1112,4-t1iazol 1 N—benzyl[(fi11any1)ca1b0ny1](py1idiny1)-1H---—1,2,4-t1iaz01—5-amine 1-[5-(benzylamin0)(py1idin—2-y1)-1H-1,2,4-t1ia201--—1] .hen n0ne N— [(4-flu010pheny1)methy1]-3 -(py1idiny1) --- [ thi0 .hen 1 carbon 1]-1H-1,2,4-t1iazolamine 11 N— [(4-flu010pheny1)methy1]-3 diny1) --- [ thi0 . hen 1 carbon 1]- 1 H- 1,2,4-t1iaz01—5-amine 12 N—[(4-f1u010pheny1)methy1]—1-[(m01ph01in --— 14 3-(5- [(4-flu010pheny1)methy1]sulfanyl- 1- [(2- methoxyphenyl)ca1b0ny1]— 1H- 1 ,2,4-t1iazol l.-420 1- [(2-meth0xypheny1)ca1b0ny1] (methylsulfany1)-3 - thi0 . hen 1 -1H-1,2,4-t1iazole 16 N—benzyl[(2-meth0xypheny1)ca1b0ny1]-3 -(py1imidin- --— 41H-1,2,4-t1iazolamine 17 N—benzyl[(2-meth0xypheny1)ca1b0ny1]-3 -(py1imidin- --— 51H-1,2,4-t1iazolamine 18 N—benzyl[(2-meth0xypheny1)ca1b0ny1]-3 -(py1imidin- I"— 21H-1,2,4-t1iazolamine 22 N-[(5-ch1010thi0phen—2-y1)methy1][(2- ypheny1)ca1b0ny1]—3 -(py1idiny1)-1H-1,2,4- t1iazolamine l.- 1-(1,3-benz0thiazoly1)-N—[(4-flu010pheny1)methy1]—--—'din 1 -1H-1,2,4-t1iazolamine 300 1-(5-[(4-f1u010pheny1)methy1]amino(fu1any1)-1H- --- 1,2,4-t1iazol 1 -2,2-dimeth 1.10 . an0ne 301 1-(5-[(4-f1u010pheny1)methy1]amino-3 -(py1idiny1)- --- 1H-1,2,4-t1ia201 1 -2,2-dimeth 1.10 . an0ne 302 1-(5-[(4-f1u010pheny1)methy1]amino-3 -(py1idiny1)- --- 1H-1,2,4-t1ia201 1 meth 1.10 . an0ne 303 (4-f1u010pheny1)methy1]amino-3 -(py1idiny1)- --- 1H-1,2,4-t1ia201 1 . hen lethan0ne 304 1-(5-[(4-f1u010pheny1)methy1]amino-3 -(py1idiny1)- --- 1H-1,2,4-t1ia201 1 meth 1butan0ne 305 1-(5-[(4-f1u010pheny1)methy1]amino-3 -(py1idiny1)- --- 1H-1,2,4-t1ia201 1 . hen 1.10 . an0ne 306 1-(5-[(4-f1u010pheny1)methy1]amino-3 -(py1idiny1)- --- 1H-1,2,4-t1ia201 1 1-0ne 307 1-(5-[(4-f1u010pheny1)methy1]amino-3 -(py1idiny1)- --- 1H-1,2,4-t1ia201 1 308 1-(5-[(4-f1u010pheny1)methy1]amino-3 -(py1idin-3 -y1)- --- 1H-1,2,4-t1ia201 1 -2,2-dimeth 1.10 . an0ne 309 (4-f1u010pheny1)methy1]amino-3 -(py1idin-3 -y1)- --— ,4-t1ia201 1 meth0x ethan—1-0ne 310 1-(5 - 010pheny1)methy1]amino-3 -(py1idin-3 -y1)- -_n _I-II_-__ 311 1-(5-[(4-flu0r0phenyl)methyl]amino(pyridinyl)- --- 1H- 1,2,4-triaz01— 1 2- o hen lethan0ne 312 1-(5-[(4-flu0r0phenyl)methyl]amino(pyridinyl)- --- 1H- 1,2,4-triaz01— 1 3-meth lbutan0ne 313 1-(5-[(4-flu0r0phenyl)methyl]amino(pyridinyl)- --- 1H- 1,2,4-triaz01— 1 3- o hen 1. r0 o an0ne 314 (4-flu0r0phenyl)methyl]amino(pyridinyl)- --- 1H-1,2,4-triazol 1 butan0ne 315 1-(5-[(4-flu0r0phenyl)methyl]amino(pyridinyl)- --- 1H-1,2,4-triazol 1 h 1. r0 o an0ne 316 (4-flu0r0phenyl)methyl]amino(pyridinyl)- --— 1H- 1,2,4-triaz01— 1 3-meth lbutan0ne 317 1-(5-[(4-flu0r0phenyl)methyl]amino(pyridinyl)- --— 1H-1,2,4-triazol 1 butan0ne 318 1-(5-[(4-flu0r0phenyl)methyl]amino(pyridinyl)- --— 1H-1,2,4-triazol 1 319 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—2-yl)---- 1H- 1,2,4-triaz01— 12,2-dimeth 1. r0 o an0ne 320 (4-meth0xyphenyl)methyl]amino(pyridin—2-yl)---— 1H-1,2,4-triazol 1 meth 1. r0 o an0ne 321 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—2-yl)---- 1H- 1,2,4-triaz01— 1 2- o hen lethan0ne 322 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—2-yl)---- 1H- 1,2,4-triaz01— 1 3-meth lbutan0ne 323 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—2-yl)-I"- 1H- 1,2,4-triaz01— 1 3- o hen 1. r0 o an0ne 324 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—2-yl)---- 1H-1,2,4-triazol 1 butan0ne 325 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—2-yl)---- 1H-1,2,4-triazol 1 326 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—3-yl)---— 1H- 1,2,4-triaz01— 1- 1 imeth 1. r0 o an0ne 327 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—3-yl)---- 1H- 1,2,4-triaz01— 1 2- o hen lethan0ne 328 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—3-yl)---— 1H- 1,2,4-triaz01— 1 3-meth lbutan0ne 329 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—3-yl)---— 1H- 1,2,4-triaz01— 1 3- o hen 1. r0 o an0ne 330 1-(5-[(4-meth0xyphenyl)methyl]amino(pyridin—3-yl)---- 1H-1,2,4-triazol 1 butan0ne 331 1-(5-[(5-ch10r0thi0pheny1)methy1]amino(0x01an- I“ 21H-1,2,4-triazol 1 -2,2-dimeth 1. r0 o an0ne 332 meth0xyphenyl)carbonyl](pyridinyl)-N-(1,3-In- thiazol lmeth 1 -1H-1,2,4-triazolamine 333 1-[(2-meth0xyphenyl)carbonyl]pheny1—N-(1,3- --— thiazol lmeth 1 2,4-triazolamine 334 1- [(2-meth0xyphenyl)carbonyl] -3 -pheny1-N—(thi0phen- --- 2- lmeth 1 -1H-1,2,4-triazolamine 335 1- [(2-meth0xyphenyl)carbonyl] -3 -pheny1-N—(thi0phen- --- 3- lmeth 1 -1H-1,2,4-triazolamine 3[(fi1ranyl)carb0nyl]-N-[(4-meth0xyphenyl)methy1]----o ridin 1 -1H-1,2,4-triazolamine 3[(fi1ranyl)carb0nyl]-N-[(4-meth0xyphenyl)methy1]---—o 41H-1,2,4-triazolamine 33 8 1- [(furan-3 -y1)carb0ny1] -N- [(4-meth0xypheny1)methy1] - 3- o ridin 1 -1H-1,2,4-triazolamine 33 9 1- [(furan-3 -y1)carb0ny1] -N- [(4-meth0xypheny1)methy1] - 3- o ridin 1 -1H-1,2,4-triazolamine 340 1- [3 - in-3 -y1)-5 - [(thiopheny1methy1)amino ] - 1 H- 1,2,4-triazol 1] o r0 o an0ne 341 1-[4-(5-[(5-ch10r0thi0pheny1)methy1]amino[(2- methoxyphenyl)carb0ny1]—1H-1,2,4-triazol o i o eridin 1]—2,2-dimeth 1. r0 o an0ne 342 1-[5-(benzylamin0)(furany1)-1H-1,2,4-triazol 1]—2,2-dimeth 1. r0 o an0ne 343 1-[5-(benzylamin0)(furany1)-1H-1,2,4-triazol 1] o hen lethan0ne 344 1- [5 -(benzy1amin0)-3 -(pyridin—2-y1)-1H-1,2,4-triazol yl] -2,2-dimethy1pr0pan0ne 345 1- [5 y1amin0)-3 din—2-y1)-1H-1,2,4-triazol 1]—2-meth 1. r0 o an0ne 346 1- [5 y1amin0)-3 -(pyridin—2-y1)-1H-1,2,4-triazol 1] -3 -meth lbutan0ne 347 1-[5-(benzy1amin0)(pyridin—2-y1)-1H-1,2,4-triazol y1]butan0ne 348 1- [5 -(benzy1amin0)-3 -(pyridin—2-y1)-1H-1,2,4-triazol 1] oro u an— 1 -0ne 349 1- [5 -(benzy1amin0)-3 -(pyridin—3 -y1)-1H-1,2,4-triazol 1] oro u an— 1 -0ne 350 1- [5 -(benzy1amin0)-3 -(pyridin—4-y1)-1H-1,2,4-triazol 1]—2,2-dimeth 1. r0 o an0ne 351 1- [5 -(benzy1amin0)-3 -(pyridin—4-y1)-1H-1,2,4-triazol eth 1. r0 o an0ne 352 1- [5 -(benzy1amin0)-3 -(pyridin—4-y1)-1H-1,2,4-triazol 1] o hen lethan0ne 353 1- [5 -(benzy1amin0)-3 -(pyridin—4-y1)-1H-1,2,4-triazol 1] -3 -meth lbutan0ne 354 1- [5 -(benzy1amin0)-3 -(pyridin—4-y1)-1H-1,2,4-triazol 1]—3- ohen n-l-one 355 benzy1amin0)(pyridin—4-y1)-1H-1,2,4-triazol y1]butan0ne 356 1- [5 -(benzy1amin0)-3 -(pyridin—4-y1)-1H-1,2,4-triazol 1] oro u an— 1 -0ne 357 1-[5-(benzy1amin0)(thiopheny1)-1H-1,2,4-triazol- 1- 1]—2,2-dimeth 1. r0 o an0ne 8 1-benzoy1-\ -[(4-flu0r0pheny1)methy1]-3 -(pyridiny1)- 1H- 1 ,2,4-triaz01amine 359 1-benzoy1-\ -[(4-flu0r0pheny1)methy1]-3 -(pyridin-3 -y1)- 1H- 1 ,2,4-triaz01amine 4-triazolamine 362 1-benzoy1-\ -benzy1-3 -(pyridiny1)- 1 H- 1,2,4-triaz01-5 - amine 363 1-benzoy1-\ -benzy1-3 diny1)- 1 H- 1,2,4-triaz01-5 - amine 364 3 5-chlorothiopheny1)methyl]amino[(2- 5 1 1 yphenyl)carbony1]—lH- 1,2,4-triazol-3 -y1)-N,N— dimeth dinesulfonamide 365 3-(pyridiny1)[(pyridiny1)carbony1]-N-(thiophen---- 2- lmeth 1 -1H-1,2,4-triazolamine 366 3 [(2-methoxypheny1)carbony1]-5 -(methylsulfany1)- 1H-1,2,4-triazol 1o ridine 367 \-[(4-fluoropheny1)methy1]—1-[(2- 403 c methoxyphenyl)carbony1]—3 -(pyridiny1)-1H-1,2,4- l.- triazolamine 368 \-[(4-fluoropheny1)methy1]—1-[(2- 403 c methoxyphenyl)carbony1]—3 -(pyridin-3 H-1,2,4- triazolamine 369 \-[(4-fluoropheny1)methy1]—1-[(furany1)carbony1]—3 -I“ o ridin 1 -1H-1,2,4-triazolamine 370 \-[(4-fluoropheny1)methy1]—1-[(furany1)carbony1]—3 -I“ o ridin 1 -1H-1,2,4-triazolamine 371 \-[(4-fluoropheny1)methy1]—1-[(furan-3 -y1)carbony1] -3 - I“ o 21H-1,2,4-triazolamine 372 \-[(4-fluoropheny1)methy1]—1-[(furan-3 -y1)carbony1] -3 - I“ o ridin 1 -1H-1,2,4-triazolamine 373 \-[(4-fluoropheny1)methy1]—1-[(furan-3 -y1)carbony1] -3 - --— o ridin 1 -1H-1,2,4-triazolamine 374 \-[(4-fluorophenyl)methyl]propy1(pyridinyl)- --— 1H-1,2,4-triazolamine 375 \ — [(4-fluorophenyl)methy1](pyridiny1)--— thio o hen 1 carbon l]—1H-1,2,4-triazolamine 376 \ — [(4-fluorophenyl)methy1]-3 -(pyridin-3 -yl) --- thio o hen 1 carbon 1,2,4-triazolamine 377 I-II 378 I-— 379 I-— 380 I- thio o hen 1 carbon l]—1H-1,2,4-triazolamine I-— 382 \ — [(4-methoxyphenyl)methyl] —3 -(pyridin-3 -yl) --- thio o hen 1 carbon l]—1H-1,2,4-triazolamine 383 \ -[(5-chlorofi1ranyl)methyl][(2- 409 methoxyphenyl)carbony1]pheny1—1H-1,2,4-triazol amine 384 \-[(5-chlorothiopheny1)methyl][(2,4- 463 dimethoxypheny1)carbony1]-3 -(oxany1)-1H-1,2,4- triazolamine 385 \-[(5-chlorothiopheny1)methyl][(2- 43 3 methoxyphenyl)carbony1]—3 -(oxany1)-1H-1,2,4- triazolamine 386 \-[(5-chlorothiopheny1)methyl][(2- 419 methoxyphenyl)carbony1]—3 -(oxolany1)-1H-1,2,4- triazolamine 387 \-[(5-chlorothiopheny1)methyl][(2- 432 methoxyphenyl)carbony1]—3 -(piperidinyl)-1H-1,2,4- triazolamine 388 chlorothiophen—2-yl)methyl][(2- yphenyl)carbonyl][1-(pyrrolidine sulfon l azetidin—3- l]-1H-1,2,4-triazolamine 389 \-[(5-chlorothiophen—2-yl)methyl][(2- methoxyphenyl)carbonyl]—3 - [4-(morpholin o hen l]-1H-1,2,4-triazolamine 390 \-[(5-chlorothiophen—2-yl)methyl][(2- methoxyphenyl)carbonyl]phenyl-1H-1,2,4-triazol amine 391 \-[(5-chlorothiophen—2-yl)methyl][(2- methylphenyl)carbonyl](pyridinyl)-1H-1,2,4- lamine 392 \-[(5-chlorothiophenyl)methyl][(furan-3 - 1 carbon l]—3- oxolan—2- l -1H-1,2,4-triazolamine 393 \ - [(5-chlorothiophenyl)methyl][4- (dimethylamino)phenyl]— 1 - [(2- ethoxp hen 1 carbon l]-1H-1,2,4-triazolamine 394 \-benzyl[(2,6-difluorophenyl)carbonyl]-3 -(pyridin- 2- l -1H-1,2,4-triazolamine 395 \-benzyl[(2-chlorophenyl)carbonyl]—3 -(pyridin --— l -1H-1,2,4-triazolamine 396 \-benzyl[(2-chlorophenyl)carbonyl]—3 -(thiophen --— l -1H-1,2,4-triazolamine 397 \-benzyl[(2-methoxyphenyl)carbonyl]-3 -(pyridin I“ l -1H-1,2,4-triazolamine 398 \-benzyl[(2-methoxyphenyl)carbonyl]-3 -(thiophen- I“ 2- l -1H-1,2,4-triazolamine 399 \-benzyl[(4-chlorophenyl)carbonyl]—3 din --— l -1H-1,2,4-triazolamine 400 yl[(filranyl)carbonyl](pyridinyl)-1H----1,2,4-triazolamine 401 \-benzyl[(furanyl)carbonyl]—3-(pyridinyl)-1H---—1,2,4-triazolamine 402 \-benzyl[(furanyl)carbonyl]—3-(pyridinyl)-1H----1,2,4-triazolamine \-benzyl(fi1ranyl)[(2----ethoxphen 1 carbon l]-1H-1,2,4-triazolamine 404 \-benzyl(pyridin—2-yl)[(thiophenyl)carbonyl]- --- 1H-1,2,4-triazolamine 405 \-benzyl(pyridin—2-yl)[(thiophenyl)carbonyl]— --- 1H-1,2,4-triazolamine 406 \-benzyl(pyridin—4-yl)[(thiophenyl)carbonyl]- --— 1H-1,2,4-triazolamine 407 \-benzyl(pyridin—4-yl)[(thiophenyl)carbonyl]— I-— 1H-1,2,4-triazolamine 408 methyl 5-[(4-methylbenzene)amido]—1H-1,2,4-triazole- --— ox late 409 phenyl 5 - [(4-fluorophenyl)methyl] amino-3 -(pyridin-3 - --— l -1H-1,2,4-triazolecarbox late 410 propanyl 5-[(4-fluorophenyl)methyl]amino --— o ridin l -1H-1,2,4-triazolecarbox late 411 tert-butyl 3 -(5-[(5-chlorothiophenyl)methyl]amino 504 [(2-methoxyphenyl)carbonyl]-1H-1,2,4-triazol-3 - l azetidine-l-carbox late 412 tert-butyl 4-(5-[(5-chlorothiophenyl)methyl]amino -_n 3- l-lH-l,2,4-triazole-l-carbox late 932 [0090] In some embodiments, there is provided a compound as expressly set forth in Table 933 D following. 934 Table D KLKBl Cmpd IUPAC name MW No. Activity 414 (3R)-N— [(3-chloro- 11indolyl)methyl]—]l- [(4- 416 chlor0ohen lmeth l] -ox0o olidinecarboxamide 415 (38)- l -b-enzyl-N [(3-chloro- 11indolyl)methyl]]5—- 382 olidinecarboxamide amide(3S)-N-[(3-chloro-l-methyl-1H-indolyl)methyl]—1-.-[(4-chlorophenyl)methyl]oxopyrrolidine 417 -[(3-chloro-1H-indolyl)methyl]—1-[(2- .- ohen lmeth l]—5-ox0o olidinecarboxamide 418 (3 S)-N- [(3 -chloro- 1 H-indolyl)methyl] — 1— [(3 — .- chloro o hen l meth l] -5 -oxo o olidine-3 -carboxamide 419 2-\ - [(2R)amino- 1 ,2,3 ,4-tetrahydronaphthalenyl] - 4-\ -(2-fluorophenyl)N,6-dimethylpyrimidine-2,4- diamine 420 2-\ - [(2R)amino- 1 ,2,3 ,4-tetrahydronaphthalenyl] - 4-\ -(3 -fluorophenyl)N,6-dimethylpyrimidine-2,4- diamine 421 2-\ - [(2R)amino- 1 ,2,3 ,4-tetrahydronaphthalenyl] - 4-\ -(4-fluorophenyl)N,6-dimethylpyrimidine-2,4- diamine 423 2-\ - [(2S)amino- 1 ,2,3 ,4-tetrahydronaphthalenyl] - 4-\ -(2-chlorophenyl)N,6-dimethylpyrimidine-2,4- diamine 424 2-\ - [(2S)amino- 1 ,2,3 ,4-tetrahydronaphthalenyl] - 4-\ -(3 -chlorophenyl)N,6-dimethylpyrimidine-2,4- diamine 425 2-\ - [(2S)amino- 1 ,2,3 ,4-tetrahydronaphthalenyl] - 4-\ -(4-chlorophenyl)N,6-dimethylpyrimidine-2,4- diamine 936 [0091] Compounds disclosed herein also e racemic mixtures, stereoisomers and 937 mixtures of the nds, including ically-labeled and radio-labeled compounds. See 938 e.g., Goding, 1986, MONOCLONAL ANTIBODIES PRINCIPLES AND PRACTICE; Academic Press, 939 p. 104. Such s can be isolated by standard resolution techniques, including e. g., 940 fractional crystallization, chiral chromatography, and the like. See e.g., Eliel, E. L. & Wilen 941 S. H., 1993, STEREOCHEMISTRY IN ORGANIC COMPOUNDS ; John Wiley & Sons, New York. 942 [0092] In some embodiments, compounds disclosed herein have asymmetric s and 943 can occur as racemates, racemic mixtures, and as individual enantiomers or diastereoisomers, 944 with all isomeric forms as well as mixtures thereof being contemplated for use in the 945 compounds and methods described herein. The nds contemplated for use in the 946 compounds and methods described herein do not include those that are known in the art to be 947 too unstable to synthesize and/or isolate. 948 [0093] The compounds disclosed herein can also contain unnatural tions of atomic 949 isotopes at one or more of the atoms that constitute such compounds. For example, the 950 compounds can be radiolabeled with radioactive isotopes, such as for e tritium (3H), 951 iodine-125 (125I), or carbon-14 (14C). All isotopic variations of the compounds disclosed 952 , whether radioactive or not, are encompassed within the contemplated scope. 953 [0094] In some embodiments, lites of the compounds sed herein are useful for 954 the methods sed herein. 955 [0095] In some embodiments, compounds contemplated herein are provided in the form of 956 a prodrug. The term "prodrug" refers to a compound that can be converted into a compound 957 (e.g., a biologically active compound) bed herein in viva. Prodrugs can be useful for a 958 variety of reason known in the art, including e. g., ease of administration due e. g., to enhanced 959 bioavailability in oral administration, and the like. The prodrug can also have improved 960 lity in pharmaceutical itions over the biologically active compounds. An 961 example, without limitation, of a prodrug is a compound which is administered as an ester 962 (i.e., the "prodrug") to facilitate transmittal across a cell membrane where water solubility is 963 detrimental to mobility but which then is metabolically hydrolyzed to the carboxylic acid, the 964 active entity, once inside the cell where water solubility is beneficial. Conventional 965 procedures for the selection and preparation of suitable prodrug derivatives are described, for 966 example, in DESIGN OF PRODRUGS, (ed. H. Bundgaard, Elsevier, 1985), which is hereby 967 incorporated herein by nce for the limited e describing procedures and 968 preparation of suitable prodrug tives. 969 [0096] ingly, in some embodiments, compounds contemplated herein are provided 970 in the form of a prodrug ester. The term "prodrug ester" refers to derivatives of the 971 compounds disclosed herein formed by the addition of any of a variety of ester-forming 972 groups, e.g., groups known in the art, that are hydrolyzed under physiological conditions. 973 es of prodrug ester groups include pivaloyloxymethyl, acetoxymethyl, phthalidyl, 974 l and ymethyl, as well as other such groups known in the art, including a (5-R— 975 2-oxo-1,3-dioxolenyl)methyl group. Other examples of prodrug ester groups can be found 976 in, for example, T. i and V. Stella, in "Pro-drugs as Novel Delivery Systems", Vol. 14, 977 A.C.S. Symposium , American Chemical Society (1975); and BIOREVERSIBLE 978 CARRIERS IN DRUG DESIGN: THEORY AND APPLICATION, edited by E. B. Roche, Pergamon 979 Press: New York, 14-21 (1987) (providing examples of esters useful as prodrugs for 980 compounds containing carboxyl groups). Each of the above-mentioned references is herein 981 incorporated by reference for the d purpose of disclosing ester-forming groups that can 982 form prodrug esters. 983 [0097] In some embodiments, prodrugs can be slowly converted to the compounds 984 described herein useful for the methods described herein when placed in a transdermal patch 985 oir with a suitable enzyme or chemical reagent. 986 [0098] Certain nds disclosed herein can exist in ated forms as well as 987 solvated forms, including hydrated forms. In general, the solvated forms are equivalent to 988 unsolvated forms and are encompassed within the scope of contemplated compounds. Certain 989 compounds of the present invention can exist in multiple crystalline or amorphous forms. In 990 general, all physical forms are equivalent for the compounds and methods contemplated 991 herein and are intended to be within the scope disclosed herein. 993 III. Biological Activities 994 [0099] In some embodiments, compounds described herein exhibit inhibitory activity 995 against thrombin with activities 2 1 11M, e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 996 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 997 nM, or even greater. In some embodiments, the compounds exhibit inhibitory activity against 998 thrombin with activities between 0.1 11M and 1 11M, e.g., about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 999 0.8, 0.9 or 1.0 uM. In some embodiments, compounds described herein exhibit inhibitory 1000 activity against thrombin with activities S 0.1 11M, e.g., about 1, 2, 5, 10, 15, 20, 30, 40, 50, 1001 60, 70, 80, 90, or 100 nM. Ranges of values using a combination of any of the values recited 1002 herein as upper and/or lower limits are also contemplated, for example, but not d to, 1003 1—10 nM, 10—100 nM, 0.1—1 uM, 1—10 uM, 10—100 11M, 100—200 11M, 200—500 11M, or 1004 even 00 uM. In some ments, the inhibitory activity is in the range of about 1005 1—10 nM, 10—100 nM, 0.1—1 uM, 1—10 uM, 10—100 uM, 100—200 uM, 200—500 uM, or 1006 even 500—1000 uM. It is understood that for purposes of quantification, the terms “activity,” 1007 “inhibitory activity,” “biological activity,3, “thrombin activity and the like in the context of an 1008 inhibitory compound disclosed herein can be quantified in a variety of ways known in the art. 1009 Unless indicated otherwise, as used herein such terms refer to IC50 in the customary sense 1010 (i.e., concentration to achieve half-maximal inhibition). 1011 [0100] tory activity against thrombin in turn inhibits the blood coagulation process. 1012 ingly, compounds disclosed herein are indicated in the treatment or management of 1013 thrombotic disorders. In some embodiments, a dose or a therapeutically effective dose of a 1014 nd disclosed herein will be that which is sufficient to achieve a plasma concentration 1015 of the compound or its active metabolite(s) within a range set forth herein, e.g., about 1—10 1016 nM, 10—100 nM, 0.1—1 uM, 1—10 uM, 10—100 uM, 100—200 uM, 200—500 uM, or even 1017 500-1000 uM, preferably about 1—10 nM, 10—100 nM, or 0.1—1 uM. Without g to be 1018 bound by any theory, it is believe that such compounds are indicated in the treatment or 1019 management of thrombotic disorders. 1020 [0101] In some embodiments, compounds described herein t inhibitory activity 1021 against KLKl and KLKBl with activities between 1 uM and 10 0M, e.g., about 1, 2, 3, 4, 5, 1022 6, 7, 8, 9 or 10 uM. In some embodiments, compounds described herein exhibit inhibitory 1023 activity against KLKl and KLKBl with activities 2 10 uM, e.g., about 10, 20, 50, 100, 150, 1024 200, 300, 400, 500, 600, 700, 800, 900, 1000 uM or even greater. In some ments, 1025 compounds described herein exhibit inhibitory activity against KLKl and KLKBl with 1026 ties S 1 uM, e.g., about 900, 800, 700, 600, 500, 400, 300, 200, 100, 50 nM or even 1027 lower. Ranges of values using a combination of any of the values recited herein as upper 1028 and/or lower limits are also contemplated, for example, but not limited to, 1—10 nM, 10—100 1029 nM, 0.1—1 uM, 1—10 uM, 10—100 uM, 100—200 uM, 0 uM, or even 500—1000 uM. 1030 In some ments, the inhibitory activity is in the range of about 1—10 nM, 10—100 nM, 1031 0.1—1 uM, 1—10 uM, 10—100 uM, 100—200 uM, 200—500 uM, or even 500—1000 uM. It is 1032 understood that for purposes of quantification, the terms “activity,” “inhibitory activity,” 1033 gical activity,” “KLKlactivity,” “KLKBl activity” and the like in the context of an 1034 inhibitory compound disclosed herein can be quantified in a variety of ways known in the art. 1035 Unless indicated otherwise, as used herein such terms refer to IC50 in the ary sense 1036 (i.e., concentration to achieve aximal inhibition).
WO 45986 2014/030853 1037 [0102] Inhibitory activity t KLKBl has an effect on the coagulation cascade and the 1038 inflammatory response. Thus, it has been proposed that KLKBl inhibitors can be useful in 1039 the treatment of thrombotic and fibrinolytic diseases and disease conditions. 1040 [0103] Accordingly, compounds disclosed herein are indicated in the treatment or 1041 management of a variety of diseases or disorders. In some embodiments, a dose or a 1042 eutically effective dose of a compound disclosed herein will be that which is sufficient 1043 to achieve a plasma concentration of the compound or its active metabolite(s) within a range 1044 set forth herein, e.g., about 1—10 nM, 10—100 nM, 0.1—1 uM, 1—10 uM, 10—100 uM, 1045 100—200 uM, 200—500 uM, or even 500—1000 uM, ably about 1—10 nM, 10—100 nM, 1046 or 0.1—1 uM. Without wishing to be bound by any theory, it is e that such compounds 1047 are indicated in the ent or management of es associated with thrombin or 1048 kallikrein. 1049 IV. Methods of Treating and Preventing Disease 1050 [0104] Kallikrein-related diseases or disorders are biological conditions associated with or 1051 moderated by kallikrein. They include, but are not limited by, those conditions associated 1052 with biological pathways that are moderated by tissue and plasma kallikrein. An example of 1053 such a pathway is the kallikrein-kinin system (Moreau, ME. 2005, Journal of 1054 Pharmacological Sciences, 99, 6). Kallikrein-related diseases or disorders include, but are not 1055 limited to, fibrosis, ation, thrombosis, hereditary angioedema, skin disorders, cancer, 1056 and ophthalmic diseases. Ophthalmic diseases include, but are not limited to, diabetic 1057 macular edema, diabetic retinopathy, and age-related macular degeneration. 1058 [0105] Diabetic Macular Edema. In rodent models, it has been shown that activation of 1059 KLKBl in the eye increases retinal vascular permeability; whereas inhibition of the 1060 kallikrein-kinin system reduces retinal leakage induced by diabetes and hypertension. These 1061 s suggest that intraocular activation of the KLKBl pathway can contribute to 1062 ive retinal vascular permeability that can lead to diabetic macular edema. Thus, 1063 ce ts that KLKBl inhibitors can provide a new therapeutic opportunity to reduce 1064 l vascular permeability (Feener, E. P. 2010, Carr Diab Rep 10, 270). 1065 [0106] Hereditary Angioedema. Ecallantide (Kalbitor) is a 60-amino acid recombinant 1066 protein that acts as a potent ible inhibitor of KLKBl (Schneider L, et al. 2007, J 1067 Allergy Clin Immunol, 120, 416) and has been ed by the FDA for the treatment of 1068 acute attacks of hereditary angioedema (HAE). Thus plasma kallikrein inhibition can be a 1069 useful treatment for HAE, and there is strong interest in the development of plasma kallikrein 1070 inhibitors as a therapy for HAE. 1071 [0107] Skin. Overexpression of various KLKs in the skin has led to the recognition that 1072 certain kallikrein inhibitors can be useful for certain dermatological conditions, including 1073 atopic dermatitis, psoriasis and rare skin diseases such as Netherton Syndrome (Freitas et al. 1074 Bioorganic & Medicinal Chemistry Letters 2012, 22, 6072—6075). 1075 [0108] Thrombosis. Thrombotic diseases are the y indications for thrombin 1076 tion, because of thrombin’s location in the coagulation cascade and, in turn, the 1077 importance of the coagulation cascade in the ssion of blood clotting processes. 1078 However, without wishing to be bound by any theory, it is believed the coagulation cascade 1079 in l, and thrombin in ular, is important in a variety other disease states. 1080 [0109] It has been ered that compounds described herein, e.g., multisubstituted 1081 aromatic compounds, exhibit inhibitory action against thrombin (activated coagulation 1082 factor 11; EC 3.4.21.5). This, in turn inhibits the blood coagulation process. 1083 [0110] This inhibitory action is useful in the treatment of a variety of thrombotic disorders, 1084 such as, but not limited to, acute vascular diseases such as acute coronary mes; 1085 venous-, arterial- and genic thromboembolisms; the prevention of other states such as 1086 disseminated ascular coagulation, or other conditions that involve the presence or the 1087 potential formation of a blood clot thrombus. Other indications for methods described herein 1088 include the following. 1089 [0111] Cancer. Tissue kallikreins (KLKs) are subdivided into various types, and have been 1090 extensively investigated in cancer and inflammation biology. Various kallikrein KLKs have 1091 been found to be up- or down-regulated in various cancer types, such as cervical-, testicular-, 1092 and non-small-cell lung arcinoma (Caliendo et al. J. Med. Chem, 2012, 55, 6669). It 1093 has been proposed that KLKl inhibitors will be useful in certain s. 1094 [0112] It has long been recognized that cancer progression is accompanied by venous 1095 thrombosis, but it has not been understood how each disease is related. From several clinical 1096 trials studying the treatment of VTE, meta-analyses have shown that low molecular weight 1097 heparins (LMWHs) e overall survival in ups of cancer patients. See e.g., 1098 Zacharski, L. R. & Lee, A. Y., 2008, Expert Opin Investig Drugs, 17: 1029-1037; Falanga, A. 1099 & Piccioli, A., 2005, t Opinion in Pulmonary Medicine, 11:403-407; Smorenburg, S. 1100 M., et al., 1999, Thromb Haemost, 82:1600-1604; Hettiarachchi, R. J., et al., 1999, Thromb 1101 Haemost, -952. This finding was substantiated in later clinical trials that measured 2014/030853 1102 specifically the al of cancer patients. See e.g., Lee, A. Y.et al., 2005, J Clin Oncol, 1103 23:2123-2129; Klerk, C. P.et al., J Clin Oncol 2005, 23:2130-2135; , A. K., et al., 1104 2004, J Clin Oncol, 22:1944-1948; Altinbas, M., et al., 2004, J Thromb Haemost, 2:1266- 1105 1271. 1106 [0113] More recently, researchers have d on the specific anticancer effect of DTIs. 1107 For example, it was shown that heparin significantly ged the al of patients with 1108 limited small cell lung cancer. See e.g., Akl, E. A., et al., 2008, JExp Clin Cancer Res, 27:4. 1109 Other investigators found that systemic use of argatroban reduced tumor mass and prolonged 1110 survival time in rat glioma models leading to the conclusion that argatroban should be 1111 considered as a novel therapeutic for glioma, a notoriously difficult to treat cancer type. See 1112 e.g., Hua, Y., et al., 2005, Acta Nearochz'r, Suppl 2005, 95:403-406; Hua, Y., et al., 2005, J 1113 Thromb Haemost, 3: 1917-1923. Very recently, it was demonstrated that dabigatran etexilate, 1114 a DTI recently FDA-approved (see e.g., Hughes, B., 2010, Nat Rev Drag Discov, 9:903-906) 1115 for DVT tions, inhibited both the invasion and metastasis of malignant breast tumors. 1116 See e.g., DeFeo, K.et al., 2010, Thrombosis Research, 125 (Supplement 2): S188-S188; 1117 Defeo, K., et al., 2010, Cancer Biol Ther, 10: 1001-1008. Thus, dabigatran etexilate treatment 1118 led to a 50% reduction in tumor volume at 4 weeks with no weight loss in treated mice. 1119 Dabigatran etexilate also reduced tumor cells in the blood and liver micrometastases by 50- 1120 60%. These investigators concluded that dabigatran etexilate can be beneficial in not only 1121 preventing thrombotic events in cancer patients, but also as adjunct therapy to treat malignant 1122 tumors . 1123 [0114] Further, n and the LMWH nadroparin dramatically reduced the number of 1124 lung metastases when administered prior to cancer cell inoculation. See e.g., Hu, L., et al., 1125 2004, Blood, 104:2746-51. 1126 [0115] The de novo thrombin inhibitor Oic-Pro-d-Ala-Phe(p-Me) has been found to 1127 block thrombin-stimulated invasion of te cancer cell line PC-3 in a concentration 1128 dependent manner. See e.g., Nieman, M. T., et al., 2008, J Thromb Haemost, 6:837-845. A 1129 d rate of tumor growth was observed in mice dosed with the pentapeptide through their 1130 drinking water. The mice also showed reduced fold rate in tumor size and reduced overall 1131 tumor weight compared to untreated mice. Microscopic examination of treated tumors 1132 showed reduced number of large blood vessels thus concluding that the pentapeptide 1133 ered with tumor angiogenesis. Nieman, M. T., et al. Thromb Haemost, 104: 1044-8. 1134 [0116] In view of these and related studies, it is suggested that anticoagulants affect tumor 1135 metastasis; that is, angiogenesis, cancer cell adhesion, migration and invasion processes. See 1136 e.g., Van Noorden, C. J., et al., 2010, Thromb Res, 125 Suppl 2:S77-79. 1137 [0117] Fibrosis. Kallikreins are a subgroup of serine proteases, divided into plasma 1138 kallikrein (KLKB 1) and tissue kallikreins. KLKBl liberates kinins (bradykinin and kallidin) 1139 from the gens, peptides responsible for the regulation of blood pressure and activation 1140 of inflammation. In the Contact Activation Pathway of the coagulation cascade, KLKBl 1141 assists in the conversion of factor XII to factor XIIa (Keel, M.; Trentz, O. Injury 2005, 36, 1142 691—709). Factor XIIa converts FXI into FXIa, which in turn activates FIX, which with its 1143 co-factor FVIIIa forms the tenase complex, which finally activates FX to FXa. In the 1144 fibrinolysis part of the coagulation cascade, KLKBl serves to convert plasminogen to 1145 plasmin. Thus, it has been proposed that KLKBl inhibitors can be useful in the ent of 1146 thrombotic and fibrinolytic diseases and disease conditions (US Patent # 7,625,944; Bird et 1147 al. Thrombosis and Hemostasis 2012, 107, 1141). 1148 [0118] Several studies have shown the utility of anticoagulant therapy in fibrotic disorders. 1149 For example, in a rat model of CCl4-induced chronic liver injury, the DTI SSR182289 1150 decreased liver fibrogenesis significantly after 7 weeks of administration. Similar 1151 observations were made in other studies using the LMWHs nadroparin, tinzaparin, 1152 enoxaparin, and dalteparin sodium. See e.g., Duplantier, J. G., et al., 2004, Gut, 53:1682- 1153 1687; Abdel-Salam, O. M., et al., 2005, col Res, 51 :59-67; Assy, N., et al., 2007, Dig 1154 Dis Sci, 52: 1 187-1 193; Abe, W., et al., 2007, JHepatol, 46:286-294. Thus a thrombin 1155 inhibitor as an anticoagulant can be useful in the treatment of fibrinolytic diseases. 1156 [0119] In another e, the DTI tran greatly reduced ischemia reperfusion injury 1157 in a kidney transplant model in the large white pig. This led to a drastically improved kidney 1158 graft survival at 3 months. See e.g., Favreau, F., et al., 2010, Am J Transplant, 10:30-39. 1159 [0120] Recent studies have shown that in a bleomycin-induced mouse model of pulmonary 1160 fibrosis, dabigatran etexilate treatment d important profibrotic events in lung 1161 fibroblasts, ing the production of collagen and connective tissue growth factor. See 1162 e.g., Silver, R. M., et al., 2010, Am. J. Respir. Crit. Care Med, 181 :A6780; Bogatkevich, G. 1163 S., et al., 2009, Arthritis Rheum, 5-3464. 1164 [0121] The above mental evidence points to a close relationship between in 1165 and s and ts novel therapeutic opportunities for s using thrombin 1166 inhibitors. See e.g., uso, V., et al., 2008, Gut, 57:1722-1727; Chambers, R. C., 2008, 1167 Br JPharmacol, 153 Suppl 1:S367-378; Chambers, R. C. & Laurent, G. J m ., 2002, 1168 Soc Trans, -200; Howell, D. C., et al., 2001, Am JPathol, 159:1383-1395. 1169 [0122] Inflammation. Kallikrein has long been ated in inflammation (Clements, J.A. 1170 The lar Biology ofthe Kallikreins and Their Roles in Inflammation, Academic Press: 1171 San Diego, CA, 1997; Vol. 5). There is experimental evidence that KLKBl is associated with 1172 sepsis and inflammatory arthritis (Colman, R.W., 1998, al Reviews in Allergy and 1173 Immunology, 16: 365). Thus a KLKBl inhibitor can be useful in the treatment of 1174 inflammatory conditions ated with the kallikrein-kinin system, such as systemic 1175 inflammatory response me, sepsis, rheumatoid arthritis, and inflammatory bowel 1176 disease. 1177 [0123] Age-Related Macular Degeneration. KLKl has been linked to blood vessel 1178 growth moderated by the VEGF pathway (Miura S., 2003, Hypertension, 41, 1118). Age- 1179 related macular degeneration (AMD) is associated with the proliferation of abnormal blood 1180 s and VEGF expression (Lopez, P.F., 1996, Investigative Ophthalmology & Visual 1181 Science, 37, 855). Thus, KLKl inhibitors have been proposed for the treatment ofAMD (US 1182 Patent #20120264798; Ferrara, N., 2000, Current Opinion in Biotechnology, 11, 617). 1183 [0124] Alzheimer’s e. Very recent experiments conflrm higher in levels in 1184 brain endothelial cells of patients with Alzheimer’s disease. While ‘normal’ thrombin levels 1185 are connected to regulatory CNS fianctions, thrombin accumulation in the brain is toxic. It has 1186 also been found that the neural thrombin inhibitor Protease Nexin 1 (PN-l) is significantly 1187 reduced in the Alzheimer’s disease brain, despite the fact that PN—l mRNA levels are 1188 unchanged. These observations have led some investigators to suggest that reduction of CNS- 1189 resident thrombin will prove useful in Alzheimer's Disease (AD) ent. See e.g., 1190 Vaughan, P. J., et al., 1994, Brain Res, 668: 160-170; Yin, X., et al., 2010, Am JPathol, 1191 176: 1600-1606; Akiyama, H., et al., 1992, Neurosci Lett, 146: 152-154. 1192 [0125] Multiple Sclerosis. igators found that hirudin treatment in an animal model of 1193 Multiple Sclerosis (MS) showed a dramatic improvement in disease severity. See e. g., Han, 1194 M. H., et al., 2008, Nature, 451 :1076-1081. Similar results were obtained following treatment 1195 with heparin (a DTI) and dermatan sulfate, another coagulation inhibitor. See e.g., 1196 Chelmicka-Szorc, E. & , B. G., 1972, Arch Neurol, 27: 153-158; Inaba, Y., et al., 1197 1999, Cell Immunol, 198:96-102. Other evidence shows that naturally occurring rombin 1198 III has anti-inflammatory effects in diseases such as endotoxemia and other sepsis-related 1199 conditions. See e.g., Wiedermann, C. J. & Romisch, J Acta Med Austriaca, 29:89-92. ., 2002, 1200 Naturally occurring thrombin tors are presumably synthesized in situ and have 1201 protective roles in CNS inflammation. Therefore, therapeutic thrombin inhibition has been 1202 proposed as a potential MS ent. See e.g., Luo, W., et al., 2009, In: THROMBIN, 1203 Maragoudakis, M. E.; Tsopanoglou, N. E., Eds. Springer New York: 2009; pp 133-159. 1204 [0126] Pain. In a rat pain model with partial lesion of the sciatic nerve, intrathecal hirudin 1205 prevented the development of neuropathic pain and curbed pain responses for 7 days. The 1206 investigators found that following injury, neuropathic pain was ed by thrombin 1207 generation, which in turn activated PAR-1 receptor in the spinal cord. Hirudin inhibited 1208 thrombin generation and ultimately led to pain relief See e. g., Garcia, P. S., et al., 2010, 1209 Thromb Haemost, 103:1145-1151; Narita, M., et al. 25: 10000-10009. , 2005, JNeuroscz', 1210 chers hypothesize that in and the PARs are involved not just as part of the 1211 coagulation cascade, but in inflammation, nociception and neurodevelopment. Development 1212 of a DTI to intersect an unexploited pharmacology will lead to pain therapeutics distinct from 1213 opioids and NSAIDs, whose shortcomings are well documented. See e. g., Garcia 2010, Id. 1214 [0127] Accordingly, in a fiarther , there is ed a method for treating a disease or 1215 disorder in a subject in need thereof. The method includes administering a compound of any 1216 of Formulae (Ia), (Ib), (11a), (11b), (11c), (III), (IV), (V), (VI) or (VII) as disclosed herein, a 1217 compound as set forth in Table A, B, C, or D, pharmaceutically acceptable salt, ester, solvate, 1218 or prodrug thereof, or pharmaceutical composition thereof, to a subject in need thereof in an 1219 amount effective to treat the disease or disorder. The terms “therapeutically effective 1220 amount,3, nt effective to treat,3) “amount effective to prevent” and the like refer to that 1221 amount of drug or pharmaceutical agent (e.g., compound or pharmaceutical composition 1222 disclosed herein) that will elicit the biological or medical se of a tissue, system, 1223 animal, or human that is being sought by a cher, veterinarian, medical doctor or other 1224 clinician. 1225 [0128] Compounds useful for s disclosed herein include the compounds set forth for 1226 Formulae (Ia), (Ib), (11a), (11b), (11c), (III), (IV), (V), (VI) or (VII)and for the compounds set 1227 forth in Table A, B, C, or D above. 1228 [0129] In some embodiments, the diseases or ers are fibrinolytic diseases. In some 1229 embodiments the disease is a fibrotic disorder. In some embodiments, the disease is cancer. 1230 In some embodiments, the diseases are inflammatory diseases. In some embodiments the 1231 e is sepsis. In some embodiments the disease is inflammatory arthritis. In some 1232 embodiments, the disease is ic macular edema. In some ments, the disease is 1233 hereditary angioedema. In some embodiments, the disease is diabetic retinopathy. In some 1234 embodiments, the disease is age-related macular degeneration. In some embodiments, the 1235 diseases are various skin diseases which include but are not limited to atopic dermatitis, 1236 psoriasis and rare skin diseases such as Netherton Syndrome. In some embodiments, the 1237 diseases or er is Alzheimer’s disease. In some embodiments, the disease is multiple 1238 sclerosis. In some embodiments, the disease is pain. 1239 [0130] In some ments, the disease or disorder is cancer. In some embodiments, the 1240 cancer is limited small cell lung cancer. In some embodiments, the cancer is a glioma. In 1241 some embodiments, the cancer is malignant breast cancer. In some embodiments, the cancer 1242 is a etastasis. In some embodiments, the micrometastasis is of the blood or liver. In 1243 some embodiments, the cancer is a lung metastasis. In some ments, the cancer is 1244 prostatic cancer. 1245 [0131] In another aspect, there is provided a method for preventing a disease or disorder in 1246 a subject. The method es administering a compound of any of ae (Ia), (Ib), (11a), 1247 (11b), (11c), (III), (IV), (V), (VI) or (VII)as disclosed herein, compound as set forth in any of 1248 Table A, B, C, or D herein, pharmaceutically acceptable salt, ester, solvate, or prodrug 1249 thereof, or pharmaceutical composition thereof, to a subject in need f in an amount 1250 effective to prevent the disease or disorder. 1251 V. Assays 1252 [0132] Compounds described herein can be d, by a variety of methods known in the 1253 art and bed herein, for inhibition of ical activity, e.g., protease activity, of a 1254 variety of proteins, e.g.,thrombin, KLKBl and KLK1. 1255 [0133] The KLKBl kallikrein activity reported herein (e. g., Tables B, C, and D) was 1256 obtained as follows. Human KLKBl protein was obtained from Enzyme Research Labs. The 1257 chromogenic substrate S-2302 was obtained from DiaPharma. KLKBl was assayed in buffer 1258 containing 0.05 M Tris (pH 7.4), 0.01 M NaCl and 0.2% w/v PEG-8000. The final 1259 tration of enzyme used was 3 nM KLKB 1. The final concentration of substrate used 1260 was 250 uM S-2302 for KLKBl. All assays were performed in 96-well microtiter plates at 1261 room temperature (RT). The enzyme and tor were pre-incubated for 10 minutes then 1262 substrate was added and read at 405 nm in a SpectraMaX Plus Spectrophotometer (Molecular 1263 Devices). Inhibitor IC50 values were determined by adding test compound as ten point, three- 1264 fold serial dilutions in buffer solution, as known in the art. The plate was read at 10 s 1265 after substrate addition. The IC50 was calculated by plotting the percent (%) inhibition against 1266 compound concentration and fitting the data to a constrained four parameter sigmoidal curve, 1267 as known in the art. 1268 [0134] The KLKl kallikrein ty reported herein (e.g., Tables B and C) was obtained as 1269 follows. Recombinant human tissue kallikrein (KLKl) was obtained from R&D s. 1270 Pro-Phe-Arg-AMC (I-1295) substrate was obtained from Bachem. KLKl enzyme is activated 1271 by incubating 0.5 mg/ml KLKl combined with 0.1 ug/ml thermolysin in a buffer of 0.05 M 1272 Tres (pH 7.5), 0.15 M NaCl, and 0.01 M CaClz for one hour at 37°C. The thermolysin is then 1273 deactivated by the addition of equal parts 20 mM 1, 10 phenanthroline solution in water. The 1274 activated KLKl solution is then added to CHES buffer (0.05 M CHES, 0.15 M NaCl, 0.01 M 1275 CaClz, pH 10) for a final concentration of 5 nM along with the test article and incubated for 1276 10 minutes. Substrate is then added at a concentration of 2.75 uM. Substrate activation is 1277 read 10 minutes after substrate addition using a y H1 multifilnction plate reader 1278 (Biotek) programmed with a 360 nm excitation wavelength and a 480 nm emission 1279 ngth. Inhibitor response was established by adding test compound as ten point, three- 1280 fold serial dilutions, as known in the art. The IC50 was calculated by plotting the percent (%) 1281 inhibition against compound concentration and fitting the data to a constrained four ter 1282 sigmoidal curve, as known in the art. 1283 [0135] The in activity reported herein (e.g., Table A) was ed as follows. 1284 Human thrombin was obtained from Haematologic logies Inc. The chromogenic 1285 substrate S-2238 was obtained from DiaPharma. Thrombin was assayed in buffer containing 1286 0.05 M Tris (pH 7.4), 0.015 M NaCl and 0.01% PEG-8000. The final tration of 1287 enzyme used was 3 nM thrombin. The final concentration of substrate used was 125 uM S- 1288 2238 for thrombin. All assays were med in 96-well iter plates at room 1289 temperature (RT). The enzyme and inhibitor were pre-incubated for 10 minutes then substrate 1290 was added and read at 405 nm in a SpectraMax Plus Spectrophotometer (Molecular Devices). 1291 Inhibitor IC50 values were determined by adding test compound as ten point, three-fold serial 1292 dilutions in buffer on, as known in the art. The plate was read at 10 minutes after 1293 substrate addition. The IC50 was calculated by plotting the percent (%) inhibition against 1294 compound concentration and fitting the data to a constrained four parameter sigmoidal curve, 1295 as known in the art. 1296 VI. Pharmaceutical Compositions 1297 [0136] In another , there is provided a pharmaceutical composition comprising a 1298 compound disclosed herein and a pharmaceutically acceptable excipient. The compound is a 1299 compound of any of Formulae (Ia), (1b), (Ila), (11b), (11c), (III), (IV), (V), (VI) or (VII) as 1300 disclosed , a compound as set forth in Table A, B, C, or D herein, or pharmaceutically 1301 acceptable salt, ester, solvate, or prodrug thereof. In some ments, the compound is set 1302 forth in Table A, B, C, or D herein. 1303 [0137] The term “pharmaceutically acceptable salts” is meant to include salts of the active 1304 compounds that are prepared with relatively nontoxic acids or bases, depending on the 1305 particular substituents found on the compounds described herein. When compounds disclosed 1306 herein contain relatively acidic fianctionalities, base addition salts can be obtained by 1307 contacting the neutral form of such compounds with a sufficient amount of the desired base, 1308 either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable base 1309 addition salts include , potassium, calcium, um, organic amino, or magnesium 1310 salt, or a similar salt. When compounds disclosed herein n relatively basic 1311 functionalities, acid addition salts can be obtained by ting the neutral form of such 1312 compounds with a sufficient amount of the desired acid, either neat or in a suitable inert 1313 solvent. Examples of pharmaceutically acceptable acid on salts include those derived 1314 from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, 1315 monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, 1316 sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the 1317 salts derived from relatively ic organic acids like acetic, propionic, isobutyric, maleic, 1318 malonic, c, succinic, suberic, c, lactic, mandelic, phthalic, benzenesulfonic, p- 1319 tolylsulfonic, citric, tartaric, oxalic, methanesulfonic, and the like. Also included are salts of 1320 amino acids such as te and the like, and salts of organic acids like onic or 1321 uronic acids and the like (see, for example, Berge et (1]., “Pharmaceutical Salts”, 1322 Journal ofPharmaceutical Science, 1977, 66, 1-19). Certain specific compounds disclosed 1323 herein contain both basic and acidic functionalities that allow the compounds to be converted 1324 into either base or acid addition salts. 1325 [0138] Compounds disclosed herein can exist as salts, such as with pharmaceutically 1326 acceptable acids. ingly, the compounds contemplated herein e such salts. 1327 Examples of such salts include hydrochlorides, hydrobromides, sulfates, methanesulfonates, 1328 nitrates, maleates, acetates, citrates, fumarates, tartrates (e. g., (+)-tartrates, (-)—tartrates, or 1329 es thereof including racemic mixtures), succinates, benzoates, and salts with amino 1330 acids such as glutamic acid. These salts can be prepared by methods known to those skilled in 1331 the art. 1332 [0139] The neutral forms of the nds are preferably rated by contacting the 1333 salt with a base or acid and isolating the parent compound in the conventional manner. The 1334 parent form of the compound differs from the various salt forms in certain physical 1335 properties, such as solubility in polar solvents. 1336 [0140] Pharmaceutically acceptable salts of the compounds above, where a basic or acidic 1337 group is present in the structure, are also included within the scope of compounds 1338 contemplated herein. When an acidic substituent is present, such as -NHSO3H, -COOH and 1339 -P(O)(OH)2, there can be formed the ammonium, sodium, potassium, calcium salt, and the 1340 like, for use as the dosage form. Basic groups, such as amino or basic heteroaryl radicals, or 1341 pyridyl and acidic salts, such as hydrochloride, hydrobromide, acetate, maleate, palmoate, 1342 methanesulfonate, p-toluenesulfonate, and the like, can be used as the dosage form. 1343 [0141] Also, in the embodiments in which R-COOH is present, pharmaceutically 1344 acceptable esters can be employed, e. g. and , methyl, ethyl, tert-butyl, pivaloyloxymethyl, 1345 the like, and those esters known in the art for modifying solubility or hydrolysis 1346 characteristics for use as sustained e or prodrug formulations. 1347 A. Formulations 1348 [0142] The compounds disclosed herein can be prepared and stered in a wide variety 1349 of ophthalmic, oral, parenteral, and topical dosage forms. The compounds described herein 1350 can be administered by eye drop. Also, nds described herein can be stered by 1351 injection (e.g. intravenously, intramuscularly, intravitreally, intracutaneously, 1352 subcutaneously, intraduodenally, or intraperitoneally). As such, compounds described herein 1353 can also be administered by itreal injection. Also, the compounds described herein can 1354 be administered by inhalation, for example, asally. Additionally, the compounds 1355 disclosed herein can be administered transdermally. It is also envisioned that multiple routes 1356 of administration (e.g., intramuscular, oral, ) can be used to ster the compounds 1357 disclosed herein. 1358 [0143] In some embodiments, the compounds disclosed herein can be prepared in liquid 1359 pharmaceutical compositions for ocular stration. The composition for ocular use can 1360 n one or more agents selected from the group of buffering agents, solubilizing agents, 1361 coloring agents, viscosity enhancing , and preservation agents in order to produce 1362 pharmaceutically elegant and convenient preparations. 1363 [0144] In some embodiments, the composition for ocular use can n preservatives for 1364 protection against microbiological contamination, including but not limited to benzalkodium 1365 chloride and/or EDTA. Other possible preservatives include but are not limited to benzyl 1366 alcohol, methyl ns, propyl parabens, and chlorobutanol. Preferably, a preservative, or 1367 combination of vatives, will be employed to impart microbiological protection in 1368 addition to protection against oxidation of components. 1369 [0145] In some embodiments, the compounds disclosed herein can be administered orally 1370 as tablets, aqueous or oily suspensions, lozenges, troches, powders, granules, emulsions, 1371 capsules, syrups or elixirs. The composition for oral use can contain one or more agents 1372 selected from the group of sweetening agents, flavoring agents, ng agents and 1373 preserving agents in order to produce pharmaceutically elegant and palatable preparations. 1374 Accordingly, there are also provided pharmaceutical compositions sing a 1375 pharmaceutically acceptable carrier or excipient and one or more compounds disclosed 1376 herein. 1377 [0146] In some embodiments, tablets n the acting ingredient in ure with non- 1378 toxic pharmaceutically acceptable excipients that are suitable for the manufacture of tablets. 1379 These excipients can be, for example, (1) inert diluents, such as m carbonate, lactose, 1380 calcium phosphate, carboxymethylcellulose, or sodium phosphate; (2) granulating and 1381 disintegrating agents, such as corn starch or alginic acid; (3) binding agents, such as starch, 1382 gelatin or ; and (4) lubricating agents, such as magnesium stearate, stearic acid or talc. 1383 These s can be uncoated or coated by known techniques to delay disintegration and 1384 absorption in the intestinal tract and thereby provide a sustained action over a longer 1385 period. For example, a time delay material such as glyceryl monostearate or glyceryl 1386 distearate can be employed. 1387 [0147] For preparing pharmaceutical compositions from the compounds sed herein, 1388 pharmaceutically acceptable rs can be either solid or liquid. Solid form preparations 1389 include powders, tablets, pills, capsules, cachets, itories, and dispersible granules. A 1390 solid carrier can be one or more substance that can also act as diluents, flavoring agents, 1391 binders, vatives, tablet disintegrating agents, or an encapsulating material. 1392 [0148] A compound disclosed , in the form of a free nd or a 1393 pharmaceutically-acceptable pro-drug, metabolite, analogue, derivative, solvate or salt, can 1394 be administered, for in vivo application, parenterally by injection or by gradual perfusion 1395 over time. Administration can be intravenously, intraperitoneally, intramuscularly, 1396 subcutaneously, intracavity, or transdermally. For in vitro studies the compounds can be 1397 added or dissolved in an appropriate biologically acceptable buffer and added to a cell or 1398 tissue. 1399 [0149] In powders, the carrier is a finely divided solid in a e with the finely divided 1400 active component. In tablets, the active component is mixed with the carrier having the 1401 necessary binding properties in suitable proportions and compacted in the shape and size 1402 desired. 1403 [0150] The powders and tablets preferably contain from 5% to 70% of the active 1404 compound. le carriers are magnesium carbonate, magnesium stearate, talc, sugar, 1405 lactose, pectin, dextrin, starch, gelatin, tragacanth, methylcellulose, sodium 1406 carboxymethylcellulose, a low melting wax, cocoa butter, and the like. The term 1407 “preparation” is intended to include the formulation of the active compound with 1408 encapsulating material as a carrier ing a capsule in which the active component with or 1409 without other carriers, is surrounded by a carrier, which is thus in association with it. 1410 Similarly, cachets and lozenges are included. s, powders, capsules, pills, cachets, and 1411 es can be used as solid dosage forms suitable for oral administration. 1412 [0151] For preparing suppositories, a low g wax, such as a mixture of fatty acid 1413 glycerides or cocoa butter, is first melted and the active component is dispersed 1414 homogeneously therein, as by stirring. The molten homogeneous mixture is then poured into 1415 convenient sized molds, allowed to cool, and thereby to solidify. 1416 [0152] Liquid form preparations include solutions, suspensions, and emulsions, for 1417 example, water or water/propylene glycol solutions. For parenteral injection, liquid 1418 preparations can be formulated in solution in s polyethylene glycol solution. 1419 [0153] When parenteral ation is needed or d, particularly suitable admixtures 1420 for the compounds disclosed herein are injectable, sterile solutions, preferably oily or 1421 aqueous solutions, as well as suspensions, emulsions, or implants, including suppositories. In 1422 particular, carriers for parenteral administration include aqueous solutions of dextrose, , 1423 pure water, ethanol, glycerol, propylene glycol, peanut oil, sesame oil, polyoxyethylene-block 1424 rs, and the like. Ampoules are convenient unit s. The compounds disclosed 1425 herein can also be incorporated into liposomes or administered via transdermal pumps or 1426 patches. Pharmaceutical ures suitable for use in the pharmaceuticals compositions and 1427 methods disclosed herein include those described, for example, in CEUTICAL 1428 SCIENCES (17th Ed., Mack Pub. Co., Easton, PA) and WO 96/05309, the teachings of both of 1429 which are hereby incorporated by reference. 1430 [0154] In some embodiments, preparations for eral administration e sterile 1431 aqueous or non-aqueous ons, suspensions, and ons. Examples of non-aqueous 1432 solvents are propylene , polyethylene glycol, vegetable oils such as olive oil, and 1433 injectable organic esters such as ethyl oleate. Aqueous carriers include water, 1434 alcoholic/aqueous solutions, emulsions or suspensions, including saline and ed media. 1435 eral vehicles include sodium chloride solution, ’s dextrose, dextrose and sodium 1436 chloride, lactated Ringer’s intravenous vehicles include fluid and nutrient replenishers, 1437 electrolyte replenishers (such as those based on Ringer’s dextrose), and the like. Preservatives 1438 and other additives can also be present such as, for example, antimicrobials, anti-oxidants, 1439 ing agents, growth s and inert gases and the like. 1440 [0155] Aqueous solutions suitable for oral use can be prepared by dissolving the active 1441 component in water and adding suitable colorants, flavors, stabilizers, and thickening agents 1442 as desired. Aqueous suspensions suitable for oral use can be made by dispersing the finely 1443 divided active component in water with viscous al, such as natural or synthetic gums, 1444 , methylcellulose, sodium carboxymethylcellulose, and other well-known suspending 1445 agents. 1446 [0156] Also included are solid form preparations that are intended to be converted, shortly 1447 before use, to liquid form preparations for oral administration. Such liquid forms include 1448 solutions, suspensions, and emulsions. These preparations can contain, in addition to the 1449 active ent, colorants, flavors, stabilizers, buffers, artificial and natural sweeteners, 1450 dispersants, thickeners, solubilizing agents, and the like. 1451 [0157] The pharmaceutical preparation is preferably in unit dosage form. In such form the 1452 preparation is subdivided into unit doses containing riate quantities of the active 1453 component. The unit dosage form can be a packaged preparation, the package containing 1454 discrete quantities of preparation, such as packeted tablets, capsules, and powders in vials or 1455 ampoules. Also, the unit dosage form can be a capsule, tablet, cachet, or lozenge itself, or it 1456 can be the appropriate number of any of these in packaged form. 1457 [0158] The quantity of active component in a unit dose preparation can be varied or 1458 adjusted from 0.1 mg to 10000 mg, more typically 1.0 mg to 1000 mg, most typically 10 mg 1459 to 500 mg, according to the particular ation and the potency of the active component. 1460 The composition can, if desired, also contain other compatible therapeutic agents. 1461 [0159] Some compounds can have limited solubility in water and therefore can require a 1462 tant or other riate co-solvent in the composition. Such co-solvents include: 2014/030853 1463 Polysorbate 20, 60, and 80; Pluronic F-68, F-84, and P-103; extrin; and polyoxyl 35 1464 castor oil. Such co-solvents are typically employed at a level between about 0.01 % and about 1465 2% by weight. 1466 [0160] Viscosity greater than that of simple aqueous solutions can be desirable to decrease 1467 variability in sing the formulations, to decrease physical tion of ents of a 1468 suspension or emulsion of formulation, and/or otherwise to improve the formulation. Such 1469 ity building agents include, for example, polyvinyl alcohol, polyvinyl pyrrolidone, 1470 methyl cellulose, y propyl methylcellulose, yethyl cellulose, carboxymethyl 1471 cellulose, hydroxy propyl cellulose, chondroitin e and salts thereof, hyaluronic acid and 1472 salts thereof, and combinations of the foregoing. Such agents are typically employed at a 1473 level between about 0.01% and about 2% by weight. 1474 [0161] The compositions sed herein can additionally include components to provide 1475 sustained release and/or comfort. Such components include high molecular weight, anionic 1476 mucomimetic polymers, gelling polysaccharides, and finely-divided drug carrier substrates. 1477 These components are discussed in greater detail in US. Pat. Nos. 4,911,920; 5,403,841; 1478 5,212,162; and 4,861,760. The entire contents of these patents are incorporated herein by 1479 reference in their entirety for all es. 1480 [0162] By the present, there are provided methods for ameliorating wound healing and for 1481 mediating tissue repair ding but not limited to treatment of peripheral and coronary 1482 vascular disease). According to these methods, a subject having a wound or in need of tissue 1483 repair, is treated at the site of the wound or damaged tissue or treated systemically, with a 1484 compound disclosed herein in the form of a free compound or a pharmaceutically-acceptable 1485 prodrug, metabolite, ue, derivative, solvate or salt. 1486 [0163] Generally, the terms “treating3, CCtreatment” and the like are used herein to mean 1487 ing a subject, tissue or cell to obtain a desired pharmacologic and/or physiologic effect. 1488 The effect can be prophylactic in terms of completely or partially preventing a disease or 1489 disorder or sign or symptom thereof, and/or can be therapeutic in terms of a partial or 1490 complete cure for a disorder and/or adverse effect attributable to it. “Treating” as used herein 1491 covers any treatment of, or prevention of a disease or disorder in a vertebrate, a mammal, 1492 particularly a human, and includes: (a) preventing the disease or er from occurring in a 1493 subject that can be predisposed to the disease or disorder, but has not yet been diagnosed as 1494 having it; (b) inhibiting the disease or disorder, i.e. its development; or (c) , arresting WO 45986 1495 relieving or ameliorating the disease or disorder, i.e. cause regression of the disease or 1496 disorder. 1497 [0164] There are provided various pharmaceutical compositions useful for ameliorating 1498 certain diseases and disorders. The pharmaceutical compositions according to one 1499 embodiment are prepared by formulating a compound disclosed herein in the form of a free 1500 compound or a pharmaceutically-acceptable pro-drug, metabolite, analogue, derivative, 1501 solvate or salt, either alone or together with other ceutical agents, suitable for 1502 administration to a t using carriers, excipients and additives or auxiliaries. Frequently 1503 used rs or auxiliaries include magnesium carbonate, titanium dioxide, lactose, mannitol 1504 and other , talc, milk protein, gelatin, starch, vitamins, cellulose and its derivatives, 1505 animal and vegetable oils, polyethylene glycols and solvents, such as sterile water, alcohols, 1506 ol and polyhydric alcohols. Intravenous vehicles include fluid and nutrient replenishers. 1507 [0165] Preservatives include antimicrobial, anti-oxidants, ing agents and inert gases. 1508 Other pharmaceutically able carriers include aqueous solutions, non-toxic excipients, 1509 including salts, preservatives, buffers and the like, as described, for instance, in Remington’s 1510 Pharmaceutical Sciences, 15th ed. Easton: Mack Publishing Co. 1461-1487 , 412, 1511 (1975) and The National Formulary XIV., 14th ed. Washington: American Pharmaceutical 1512 Association (1975), the contents of which are hereby incorporated by reference. The pH and 1513 exact tration of the various components of the ceutical composition are 1514 adjusted according to routine skills in the art. See e. g., Goodman and Gilman (eds.), 1990, 1515 THE PHARMACOLOGICAL BASIS FOR THERAPEUTICS (7th ed.). 1516 [0166] The pharmaceutical compositions are preferably prepared and administered in dose 1517 units. Solid dose units are tablets, capsules and suppositories. For treatment of a subject, 1518 depending on activity of the compound, manner of administration, nature and severity of the 1519 disease or disorder, age and body weight of the subject, different daily doses can be used. 1520 [0167] Under certain stances, however, higher or lower daily doses can be 1521 appropriate. The administration of the daily dose can be carried out both by single 1522 administration in the form of an dual dose unit or else several smaller dose units and 1523 also by multiple administrations of subdivided doses at specific intervals. 1524 [0168] The pharmaceutical compositions contemplated herein can be administered locally 1525 or systemically in a therapeutically ive dose. Amounts effective for this use will, of 1526 course, depend on the severity of the e or er and the weight and general state of 1527 the subject. Typically, dosages used in vitro can provide useful ce in the amounts 2014/030853 1528 useful for in situ administration of the pharmaceutical composition, and animal models can be 1529 used to determine effective dosages for treatment of particular disorders. 1530 [0169] Various considerations are described, e. g. in Langer, 1990, Science, 249: 1527; 1531 Goodman and Gilman's (eds.), 1990, 161., each of which is herein incorporated by reference 1532 and for all purposes. Dosages for eral administration of active pharmaceutical agents 1533 can be ted into corresponding dosages for oral administration by multiplying 1534 parenteral dosages by appropriate conversion s. As to general applications, the 1535 parenteral dosage in mg/mL times 1.8 = the corresponding oral dosage in milligrams (“mg”). 1536 As to oncology applications, the parenteral dosage in mg/mL times 1.6 = the ponding 1537 oral dosage in mg. An average adult weighs about 70 kg. See e. g., Miller-Keane, 1992, 1538 ENCYCLOPEDIA & DICTIONARY OF MEDICINE, NURSING & ALLIED HEALTH, 5th Ed., (W. B. 1539 Saunders Co.), pp. 1708 and 1651. 1540 [0170] The method by which the compound disclosed herein can be administered for oral 1541 use would be, for example, in a hard gelatin e wherein the active ient is mixed 1542 with an inert solid diluent, or soft gelatin capsule, wherein the active ingredient is mixed with 1543 a co-solvent mixture, such as PEG 400 containing 20. A compound disclosed herein 1544 can also be stered in the form of a sterile injectable aqueous or oleaginous solution or 1545 suspension. The compound can generally be administered intravenously or as an oral dose of 1546 0.1 ug to 20 mg/kg given, for example, every 3 - 12 hours. 1547 [0171] Formulations for oral use can be in the form of hard gelatin capsules n the 1548 active ingredient is mixed with an inert solid diluent, for e, calcium carbonate, 1549 m phosphate or kaolin. They can also be in the form of soft gelatin capsules wherein 1550 the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin 1551 or olive oil. 1552 [0172] Aqueous suspensions normally contain the active materials in admixture with 1553 excipients suitable for the manufacture of aqueous suspension. Such excipients can be (1) 1554 suspending agent such as sodium carboxymethyl cellulose, methyl cellulose, 1555 hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and 1556 gum ; (2) dispersing or wetting agents which can be (a) naturally occurring phosphatide 1557 such as lecithin; (b) a condensation product of an alkylene oxide with a fatty acid, for 1558 example, polyoxyethylene stearate ; (c) a condensation product of ethylene oxide with a long 1559 chain aliphatic alcohol, for example, heptadecaethylenoxycetanol; (d) a condensation product 1560 of ethylene oxide with a partial ester derived from a fatty acid and hexitol such as 1561 polyoxyethylene sorbitol monooleate, or (e) a condensation product of ethylene oxide with a 1562 partial ester d from fatty acids and hexitol anhydrides, for example polyoxyethylene 1563 an monooleate. 1564 [0173] The pharmaceutical compositions can be in the form of a sterile injectable aqueous 1565 or oleagenous suspension. This suspension can be formulated according to known methods 1566 using those le dispersing or wetting agents and suspending agents that have been 1567 ned above. The sterile injectable preparation can also a sterile injectable solution or 1568 suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a 1569 solution in l,3-butanediol. Among the able vehicles and solvents that can be employed 1570 are water, ’s solution, and isotonic sodium chloride solution. In addition, sterile, fixed 1571 oils are conventionally employed as a solvent or suspending medium. For this purpose, any 1572 bland fixed oil can be employed including synthetic mono-or diglycerides. In addition, fatty 1573 acids such as oleic acid flnd use in the preparation of injectables. 1574 [0174] A compound disclosed herein can also be administered in the form of ophthalmic 1575 compositions applied topically to the eye, ably in the form of eye drops. A compound 1576 disclosed herein can also be administered in the form of intravitreal injection. 1577 [0175] A nd disclosed herein can also be administered in the form of suppositories 1578 for rectal administration of the drug. These compositions can be prepared by mixing the drug 1579 with a suitable non-irritating excipient that is solid at ordinary temperature but liquid at the 1580 rectal temperature and will therefore melt in the rectum to release the drug. Such materials 1581 include cocoa butter and polyethylene glycols. 1582 [0176] The compounds disclosed herein as used in the methods disclosed herein can also be 1583 administered in the form of me delivery systems, such as small ellar vesicles, 1584 large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a 1585 variety of phospholipids, such as cholesterol, stearylamine, or phosphatidylcholines. 1586 [0177] For topical use, creams, ointments, s, solutions or suspensions, etc., containing 1587 the compounds disclosed herein, are ed. 1588 [0178] In addition, some of the nds sed herein can form solvates with water 1589 or common organic solvents. Such solvates are encompassed within the scope of the methods 1590 contemplated herein. 1591 B. Effective Dosages 1592 [0179] Pharmaceutical compositions provided herein include compositions wherein the 1593 active ingredient is contained in a therapeutically effective amount, i.e., in an amount 1594 effective to achieve its intended purpose. The actual amount effective for a particular 1595 application will depend, inter alia, on the condition being treated. 1596 [0180] The dosage and frequency (single or multiple doses) of compound administered can 1597 vary depending upon a variety of factors, including route of administration; size, age, sex, 1598 health, body , body mass index, and diet of the recipient; nature and extent of 1599 symptoms of the disease being treated (e.g., the e responsive to inhibition of thrombin, 1600 KLKl and/or KLKBl); presence of other diseases or other health-related problems; kind of 1601 concurrent treatment; and complications from any disease or treatment regimen. Other 1602 therapeutic regimens or agents can be used in conjunction with the methods and compounds 1603 disclosed . 1604 [0181] For any compound described herein, the therapeutically effective amount can be 1605 initially ined from a y of techniques known in the art, e.g., biochemical 1606 characterization of inhibition of enzyme (thrombin, KLKl, or KLKBl), cell culture assays, 1607 and the like. Target concentrations will be those concentrations of active compound(s) that 1608 are capable of decreasing enzymatic activity as ed, for example, using the methods 1609 described. 1610 [0182] Therapeutically effective amounts for use in humans can be determined from animal 1611 models. For example, a dose for humans can be ated to achieve a concentration that 1612 has been found to be effective in s. The dosage in humans can be adjusted by 1613 monitoring enzymatic inhibition and adjusting the dosage upwards or downwards, as 1614 described above. 1615 [0183] Dosages can be varied depending upon the requirements of the patient and the 1616 compound being employed. The dose administered to a patient, in the context of the methods 1617 disclosed herein, should be sufficient to affect a beneficial therapeutic response in the patient 1618 over time. The size of the dose also will be determined by the existence, , and extent of 1619 any adverse side effects. Generally, ent is initiated with smaller dosages, which are less 1620 than the optimum dose of the compound. Thereafter, the dosage is increased by small 1621 increments until the optimum effect under circumstances is reached. In some ments of 1622 a method sed , the dosage range is 0.001% to 10% w/v. In some embodiments, 1623 the dosage range is 0.1% to 5% w/v. 1624 [0184] Dosage amounts and intervals can be adjusted individually to provide levels of the 1625 administered compound effective for the particular clinical tion being treated. This will 1626 provide a therapeutic regimen that is commensurate with the ty of the individual’s 1627 disease state. 1628 [0185] Utilizing the teachings provided herein, an effective prophylactic or therapeutic 1629 treatment regimen can be planned that does not cause substantial toxicity and yet is entirely 1630 effective to treat the clinical ms demonstrated by the particular patient. This planning 1631 should involve the careful choice of active compound by considering factors such as 1632 compound potency, relative bioavailability, patient body weight, ce and severity of 1633 adverse side effects, preferred mode of administration, and the toxicity profile of the selected 1634 agent. 1635 [0186] Accordingly, in some embodiments, dosage levels of the compounds disclosed 1636 herein as used in the present methods are of the order of e. g., about 0.1 mg to about 1 mg, 1637 about 1 mg to about 10 mg, about 0.5 mg to about 20 mg per am body weight, an 1638 average adult weighing 70 ams, with a red dosage range between about 0.1 mg to 1639 about 20 mg per kilogram body weight per day (from about 7.0 mg to about 1.4 gm per 1640 patient per day). The amount of the compound disclosed herein that can be combined with the 1641 r materials to produce a single dosage will vary depending upon the host treated and the 1642 particular mode of administration. For example, a formulation intended for oral 1643 administration to humans can contain about 5 [Lg to l g of a nd disclosed herein with 1644 an appropriate and ient amount of carrier material that can vary from about 5 to 95 1645 percent of the total composition. Dosage unit forms will lly contain between from 1646 about 0.1 mg to 500 mg of a compound disclosed herein. 1647 [0187] It will be understood, however, that the specific dose level for any particular patient 1648 will depend upon a variety of factors including the activity of the specific nd 1649 employed, the age, body weight, general health, sex, diet, time of administration, route of 1650 administration, rate of excretion, drug combination and the severity of the particular disease 1651 undergoing therapy. 1652 C. Toxicity 1653 [0188] The ratio between ty and therapeutic effect for a particular compound is its 1654 eutic index and can be expressed as the ratio between LD50 (the amount of compound 1655 lethal in 50% of the population) and ED50 (the amount of compound effective in 50% of the 1656 population). Compounds that exhibit high therapeutic indices are preferred. Therapeutic WO 45986 1657 index data obtained from in vitro assays, cell culture assays and/or animal studies can be used 1658 in formulating a range of dosages for use in humans. The dosage of such compounds 1659 preferably lies within a range of plasma concentrations that include the ED50 with little or no 1660 toxicity. The dosage can vary within this range depending upon the dosage form ed 1661 and the route of administration utilized. See, 6.g. Fingl et al., In: THE PHARMACOLOGICAL 1662 BASE(MFTHERAPEUTKB,C1Ll,pl,1975.Theexactfinnnflafion,unneofadnnnmuafion,and 1663 dosage can be chosen by the individual practitioner in View of the patient’s condition and the 1664 ular method in which the compound is used. For in vitro formulations, the exact 1665 formulation and dosage can be chosen by the indiVidual practitioner in View of the patient’s 1666 condition and the particular method in which the compound is used. 1667 VII. Examples 1668 [0189] The examples below are meant to illustrate certain embodiments of the invention and 1669 not to limit the scope of the invention. iations used herein have their conventional 1670 meaning in the art, unless indicated otherwise. Specific abbreviations include the following: 1671 A = om; AczO = acetic anhydride; AcOH = acetic acid; aq = aqueous; Bt = 1672 benzotriazole; BOC = N—tert—butoxycarbonyl; br = broad; t—BuOH = tert—butanol; 0C = degree 1673 Celsius; d = doublet; DABCO = 1,4-diazabicyclo[2.2.2]octane; DCE = 1,2-dichloroethane; 1674 DCM = romethane; dd = doublet of doublets; DIEA = diethylisopropylamine; DMAP = 1675 4-dimethylaminopyridine; DMF = N,N—dimethylformamide; DMSO = dimethylsulfoxide; 5 = 1676 chemical shift (given in ppm, unless otherwise indicated); EDCI = 1-ethyl(3- 1677 dimethylaminopropyl)carbodiimide; eq = lent; EtzO = diethyl ether; Eth = 1678 triethylamine; EtOAc = ethyl acetate; EtOH = ethanol; g = gram; h (or hr) = hour; HOBt = 1679 hydroxybenzotriazole; HPLC = high performance liquid chromatography; Hz = Hertz; IC50 = 1680 inhibitory concentration at 50% inhibition; J = ng constant (given in Hz, unless 1681 otherwise indicated); LC = liquid chromatography; LHMDS = lithium hexamethyldisilazide; 1682 m = multiplet; M = molar; [M+H]+ = parent mass spectrum peak plus H+; MS = mass 1683 spectrum; ms = molecular sieves; MP = melting point; MeZNH = dimethylamine; MeOH = 1684 methanol; mg = milligram; mL = milliliter; mM = millimolar; mmol = millimole; min = 1685 minute; uL = microliter; uM = micromolar; ng = nanogram; nM = nanomolar; NMR = 1686 nuclear magnetic resonance; ppm = parts per million; q = quartet; Rf = ion ; RT = 1687 room temperature; s = singlet; t = triplet; TFA = trifluoroacetic acid; THF = tetrahydrofuran; 1688 TLC = thin layer chromatography. 1689 [0190] General Scheme 1. A tic scheme useful for synthesis of compounds 1690 described herein is disclosed in General Scheme 1 following, wherein the terms “ ”, 1691 and “R” are independently hydrogen, tuted or tituted alkyl, substituted or 1692 unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted 1693 heterocycloalkyl, tuted or unsubstituted heterocycloalkenyl, substituted or unsubstituted 1694 aryl, or substituted or unsubstituted heteroaryl, or other groups obvious to those skilled in the 1695 art. 1696 General Scheme I o o 1. SOCIZ, DCM, RT HN CH3CN, n-BuLi HN CN —> # \ / C02” 2.ROH \ / COZR THE-78°C \ / Step-1 Step-2 N2H4H20 WAN—12 (i); RXCHO DMF RT HN NHCHZRX \ / N’NH \ / EtOH 85°C (ii); NaCNBH3, AcOH, RT N’NH Step-3 Step-4 O 0 RVCOCI( ) NHCHZRx RCHZCI,2 R2“ , HN \ N NHCHZRX —> \ \ / Et3N,DMF N’NYO \ / K2C03,DMF N’NYO Ry Ry 1697 Step-5 Step-6 1698 Example I - Preparation ofIntermediate 1 1699 [0191] The sis of Intermediate 1 followed General Procedure 1 following. 1700 General Procedure 1 O 0 w 1. SOCI2, DCM, RT HN \ / COZH —’ \ / COZMe 2. MeOH, RT 1701 1702 Intermediate 1 1703 [0192] To a solution of 2-hydroxynicotinic acid (50.0 g, 0.359 moles, 1.0 eq.) in 1704 dichloromethane (500 mL) at 0°C was added thionyl chloride (133.6 mL, 1.798 moles, 5.0 1705 eq.) dropwise. After 30 min tetrahydrofuran (500 mL) was added and the reaction d for 1706 14—15 hours at ambient temperature. The reaction mixture was cooled to 0°C, to it was added 1707 methanol (150 mL) dropwise, and the mixture was d for a r 30 min at room 1708 temperature. The reaction mixture was concentrated under reduced pressure to obtain a solid, 1709 which was then neutralized with aqueous sodium bicarbonate (pH 7—8), and again 1710 trated to obtain solid product. The solid was dissolved in methanol, filtered, and the 1711 filtrate concentrated to give desired product 45.0 g, (yield; 81.8 %) m/z 153.99 [M+H]+ 1H 1712 NMR (DMSO-d6, 400 MHz) 8 8051-074 (1H, q), 7.661-7.682 (1H, q), 6259-6292 (1H, m), 1713 3.734 (3H, s) ppm. 1714 Example 2 - Preparation ofIntermediate 2 1715 [0193] The synthesis of Intermediate 2 followed the ure of General ure 2 1716 following. 1717 General Procedure 2 O O HN CH3CN, n-BULI HN CN \ / COZMe THE-78°C \ / 1718 0 1719 Intermediate 1 ediate 2 1720 [0194] To a cold (-78°C) solution of acetonitrile (8.18 mL, 0.156 moles, 1.2 eq.) in 1721 tetrahydrofuran (300 mL) was added n-BuLi (2.5M in Hexane; 62.68 mL, 0.156 moles, 1.2 1722 eq) dropwise over a period of 60 min. After addition, the reaction was stirred for another 1723 60 min, then to it added methyl 2-oxo-1,2-dihydropyridinecarboxylate (Intermediate 1, 1724 20.0 g, 130 mmol, 1.0 eq) portionwise to reaction mixture and maintained -78°C for 3 hrs. 1725 The reaction was quenched with water and washed with ethyl acetate. The aqueous layer was 1726 ated to obtain crude product, which was suspended in methanol and stirred for 30 min 1727 at room ature. The solid was filtered h suction and dried over high vacuum to 1728 afford Intermediate 2 (1 1 .5 g, 54%). 1729 Example 3 - Preparation ofCompound I 1730 [0195] The synthesis of Compound 1 followed the procedure of General Procedure 3 1731 following. 1732 General Procedure 3 o o WON N2H4.H20 HN NH2 \ / —> \ / \ o ’PrOH, 85°C N’NH 1733 1734 Intermediate 2 nd 1 1735 [0196] To a solution of Intermediate 2 (20.0 g, 0.123 moles, 1.0 eq) in isopropanol (600 1736 mL) and acetic acid (22.2 mL) was added hydrazine monohydrate (7.40 mL, 0.148 moles, 1.2 1737 eq) dropwise and the reaction was heated at 85°C for 4-5 Hrs. After cooling, the reaction 1738 mixture was concentrated to give crude product, which was purified by column 1739 chromatography using neutral silica gel (60-120 mesh), eluting with 10-25% methanol in 1740 dichloromethane as gradient to give the desired product Compound 1 13.25 g (yield-61%) 1741 m/Z 177.06 [M+H]+ 1H NMR (DMSO-d6, 400 MHz) 5 11.831 (1H, s), 7857-7879 (1H, q), 1742 7383-7403 (1H, q), 6303—6336 (1H,n1), 6.048 (1H, s) 4.633 (2H, s) ppm. 1743 1744 Example 4 - Preparation ofCompound 2 1745 [0197] The synthesis of Compound 2 followed the procedure of General ure 4 1746 following. 1747 l Procedure 4 o I \ o I (i); OHCQ\CI ,DMF, RT HN NHfl’CI WW2 S \ / \N/NH —’ \ / \ N’NH 1748 (ii); NaCNBHg, AcOH, RT 1749 Compound 1 Compound 2 1750 [0198] To a solution of Compound 1 in dimethylformamide (100 mL) at 10-15°C was 1751 added acetic acid (11.2 mL) dropwise, followed by 5-chlorothiophenecarbaldehyde 1752 (9.15 g, 0.0624 moles, 1.1 eq) added portionwise. The reaction was stirred for 30-45 min at 1753 room ature. Sodium cyanoborohydride (5.35 g, 0.0851 moles, 1.5 eq.) was added 1754 portionwise over a period of 45 min and reaction was stirred for 2 hours. After completion of 1755 reaction, the mixture was poured into ice cold water under stirring and the t was 1756 extracted with ethyl acetate. The organic layer was dried over sodium sulfate and 1757 concentrated under reduced pressure to obtain crude product, which was purified by column 1758 tography using neutral silica gel and product was eluted with 10-12% Methanol in 1759 dichloromethane as mobile phase to yield pure desired product compound 2 (7.3 g, yield: 1760 42.7%) m/Z[M+H]+ 307.10 1H NMR (DMSO-d6, 400 MHz) 8 12.034 (1H, s), 11.815 (1H, 1761 S), 7869-7882 (1H, q), 7.404-7.415 (1H, d), 6.922-6.931 (1H, d), 6.862-6.871 (1H, d), 1762 6.314-6.331 (1H, d), 6.117 (1H, S), 5867-5898 (1H, t), 4.348-4.363 (2H, d) ppm. 1763 1764 Example 5 - Preparation ofCompound 3 1765 [0199] The synthesis of Compound 3 followed the procedure of l Procedure 5 1766 following. 1767 General ure 5 o H |\ CI 0 HN N s tBuC(O)CI, Et3N, HN HfimS ’ \ \ / \ \ / N’NH ,N o CHZCIZ, 0°C 1768 1769 Compound 2 Compound 3 1770 1771 [0200] To a cooled (0°C) solution of compound 2 in triethylamine (2.98 mL, 0.0215 moles, 1772 3.0 eq.) and dichloromethane (40 mL) was added pivaloyl chloride (0.776 g, 0.00647 moles, 1773 0.9 eq) dropwise over a period of 30 s. The reaction was stirred for 2-3 hours by 1774 maintaining the temperature below 10°C. After completion, the reaction was d with ice 1775 cold water under stirring and the product was extracted with dichloromethane. The organic 1776 phase was dried over sodium sulfate and concentrated under reduced pressure. The resultant 1777 crude product was d by column chromatography using neutral silica gel, eluting with 1778 5-8% methanol in dichloromethane to fiarnish pure desired t (compound 3, 0.76 g, 1779 yield: 43.6%) m/z[M+H]+ 391.24 1H NMR (DMSO-d6, 400 MHz) 5 11.250 (1H, s), 8.086- 1780 8.109 (1H, q), 7.731-7.761 (1H, t), 7.484 (1H, S), 6974-6984 (1H, d), 6.934-6.944 (1H, d), 1781 6.317-6.350 (1H, t), 6.213 (1H, S), 4471-4486 (2H, d), 1.47 (9H, s) ppm. 1782 1783 Example 6 - Preparation ofCompound 4 1784 [0201] The synthesis of Compound 4 followed the procedure of l Procedure 6 1785 following. 1786 General Procedure 6 o / 2 HN N HN N s s \ \ \ / \N,NH —’ \ / \N,N o C|, DIEA, HOBt, DMF 1787 \0 1788 Compound 2 Compound 4 1789 [0202] To a solution of furancarboxylic acid (0.338 g, 0.00301 moles, 1.2 eq) in 1790 dimethylformamide (5.0 mL) was added EDCI.HCl (0.724 g, 0.00337 moles, 1.5 eq), DIEA 1791 (0.811g, 0.00629 moles, 2.5 eq) and finally HOBt (0.074g, 0.00048 moles, 0.5 eq). The 1792 reaction mixture was stirred at room ature for 30 min, followed by the on of 1793 compound 2 (0.770 g, 0.00251 moles, 1.0 eq). The mixture was stirred at 14 hours at room 1794 temperature. After checking that the reaction had reached completion by LC-MS, the mixture 1795 was poured into ice cold water under stirring. The product was extracted with ethyl acetate. 1796 The organic phase was dried over sodium sulfate, concentrated under reduced pressure and 1797 purified by column chromatography using neutral silica gel (60-120 mesh), eluting with 15- 1798 25% ethyl acetate in n-hexane as gradient to give pure desired compound 4 (0.45 g, yield: 1799 45%) m/z[M+H]+ 401.84 1H NMR (DMSO-d6, 400 MHz) 8 11.923 (1H, S), 9024-9029 1800 (1H, q), 8.274-8.297 (1H, q), 7.893 (1H, d), 7.833-7.884 (1H, q), 7.500-7.512 (1H, d), 1801 7.085-7.091 (1H, q), 6.990 (2H, q), 6.313-6.347 (2H, t), 5.771 (1H, S), 560 1802 (1H, d) ppm. 1803 1804 Example 7 - ation ound 5 1805 [0203] The synthesis of Compound 5 followed the ure of General Procedure 7 1806 following. 1807 General Procedure 7 06'1“]:ng cho3 DMF /O\$\oN \ \ 1808 o 0 1809 Compound 4 Compound 5 1810 [0204] To a solution of compound 4 (0.150 g, 0.375 mmoles, 1.0 eq) in DMF (5.0 mL) was 1811 added anhydrous potassium carbonate (0.129 g, 0.937 mmoles, 2.5 eq) and then stirred for 30 1812 minutes at room temperature. 2-(Chloromethyl)thiophene (0.059 g, 0.45 , 1.2 eq) was 1813 added to the reaction mixture and the reaction stirred for a further 2-3 hours at room 1814 temperature. The mixture was monitored by TLC and LCMS. After completion of reaction, 1815 the reaction mixture was poured into ice cold water under stirring and extracted with ethyl 1816 acetate. The organic phase was dried over sodium sulfate, concentrated under reduced 1817 pressure and purified by column chromatography using neutral silica gel. The product was 1818 eluted with 1-5% ethyl acetate as gradient in n-hexane to fiamish compound 5 (0.036 g, yield- 1819 19.3%) m/z[M+H]+ . 1H NMR (DMSO-d6, 400 MHz) 8 9.020 (1H, S), 8274-8297 2014/030853 1820 (1H, dd), 7.960-7.981 (1H, dd), 7885-7893 (1H, t), 7833-7864 (1H, t), 7519—7539 (1H, 1821 dd), 7430—7434 (1H, d), 7117—7133 (1H, dd), 7.087-7.091 (1H, d), 6975-6987 (1H, t), 1822 6380-6427 (1H, t), 6.435 (1H, s), 5.189(2H, s), 4.550-4.565(2H, d) ppm. 1823 1824 e 8 - Preparation ofCompound 6 1825 [0205] The synthesis of Compound 6 followed the procedure of General Procedure 8 1826 following. 1827 General Procedure 8 CONNEQO | s\ CI Hfim H\N / \ / N,N 032003 DMF 70°C o 060\ 1828 0 1829 Compound 4 Compound 6 1830 [0206] To a solution of compound 4 (0.150 g, 0.375 mmoles, 1.0 eq.) in DMF (5.0 mL) 1831 was added cesium carbonate (0.304 g, 0.937 mmoles, 2.5 eq.). The reaction e was 1832 stirred for 30 min at room temperature, followed by the addition of 4-(chloromethyl)pyridine 1833 hydrochloride (0.073 g, 0.45 mmoles, 1.2 eq). The reaction was stirred for 3-4 hours at 70°C. 1834 The reaction was monitored by TLC and LCMS. After completion of the reaction, the 1835 mixture was poured into ice cold water under ng and extracted into ethyl acetate. The 1836 organic phase was dried over sodium sulfate, filtered and concentrated under reduced 1837 pressure. The crude product was d by column chromatography using neutral silica gel, 1838 eluting with 40-55% ethyl acetate as gradient in n-hexane to furnish compound 6 (0.032 g, 1839 yield: 17.4%) m/z [M+H]+ 491.95. 1H NMR (DMSO-d6, 400 MHz) 8 9.030 (1H, s), 8.541- 1840 8.526 (2H, d), 8.379-8.356 (1H, dd), 8.020-7.999(1H, dd), 7.893-7.836 (2H,m), 7.210-7.195 1841 (2H, d), 7.089(1H, d), 6.968-6.948(2H, t), 6.498-6.463 (1H, t), 6.294 (1H, s), 5.255 1842 (2H, s), 4.542-4.526 (2H,d) ppm. 1843 e 9 - Preparation ofIntermediate 3 1844 [0207] l Scheme 11. A synthetic scheme useful for synthesis of compounds 1845 described herein is disclosed in General Scheme 11 following, wherein the terms “ X”,“Ry”, 1846 and “R” are independently hydrogen, tuted or unsubstituted alkyl, tuted or 1847 unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted 1848 heterocycloalkyl, substituted or unsubstituted heterocycloalkenyl, substituted or unsubstituted 1849 aryl, or substituted or unsubstituted heteroaryl, or other groups obvious to those skilled in the 1850 art. 1851 1852 General Scheme 11 O H H2804 N NH2 RXJJ\OH 2 + H N’ X 2 Ill/H R f 140 °C i. RVCHO, EtOH, 78 °C ii. NaCNBH3, ACOH N Ry RX_<N§( V H RZCOCI N\ NVRV N’ Y0 R —<\N’NHX \r Et3N 1853 RZ 1854 [0208] The synthesis of Intermediate 3 followed General Procedure 9 following. 1855 l Procedure 9 H H so NH ,N NH 2 4 N 2 2 N\ N \ OH + H2N \H/ I 140 0C Q—QY — / NH N’NH 1856 1857 Intermediate 3 1858 [0209] A solution of nicotinic acid (9.9 g, 80.9 mmol) in water (30 mL) was added slowly 1859 portion-wise to a previously stirred mixture of aminoguanidine sulfate (10 g, 73.5 mmol) in 1860 concentrated H2S04 (8.8 mL, 162 mmol), and the reaction mixture was stirred at 140 CC for 1861 72 h. The reaction mixture was diluted with water (50 mL) and lized with saturated 1862 s K2CO3 (30 mL), and the resultant solid was filtered. The residue was washed with 1863 water (2 X 30 mL), EtzO (2 X 30 mL) and dried under vacuum to afford Intermediate 3 (4.6 g, 1864 39%) as an off-white solid. 1H NMR: (DMSO-d6) 8 12.23 (s, 1H), 9.05 (s, 1H), 8.54 (d, J: 1865 2.8 Hz, 1H), 8.17 (d, J: 7.4 Hz, 1H), 7.42-7.52 (m, 1H), 6.19 (s, 2H); MS: 162 [M + H]+; 1866 MP: 234-236 C’C; TLC: 20% H3 in CHClgC Rf: 0.40. 1867 Example 10 - Preparation ofIntermediate 4 H H so N NH2 N\ ,N NH 2 4 \H/ 2 N\ OH + H2N | 140°C </:>_<\Y — / NH N/NH 1868 1869 Intermediate 4 WO 45986 1870 [0210] General Procedure 9 was followed to obtain Intermediate 4 (8.5 g, 46%). 1H NMR: 1871 (DMSO-d6) 8 8.60 (d, J: 4.4 Hz, 1H), 7.86-7.91 (m, 2H), 7.37 (br s, 1H), 5.79 (br s, 2H); 1872 MS: 162 [M + H]+; MP: 218-220 c’C; TLC: 20% MeOH/NH3 in CHClg: Rf: 0.40. 1873 Example I I - Preparation rmediate 5 H NH N H2804 |\ OH + H2N , \H/ O NC\>—<\ 1*N 1874 1875 Intermediate 5 1876 [0211] General Procedure 9 was followed to obtain Intermediate 5 (12 g, 67%). 1H NMR: 1877 (DMSO-d6) 5 12.35 (br s, 1H), 8.59 (d, J: 5.5 Hz, 2H), 7.76-7.78 (m, 2H), 6.23 (s, 2H); MS: 1878 162 [M + H]+; TLC: 20% MeOH/NH3 in CHC13: Rf: 0.40. 1879 Example 12 - Preparation ofIntermediate 6 1880 [0212] The synthesis of Intermediate 6 followed the procedure of l Procedure 10 1881 following. 1882 General Procedure 10 CHO F N NH2 i. EtOH, ms, 78°C H / \ \YN\ . —.N N — NINH ii. NaCNBH3, AcOH / \ 1883 F N,NH 1884 Intermediate 3 Intermediate 6 1885 [0213] 4-Fluorobenzaldehyde (3.1 g, 24.8 mmol, 2 eq) and molecular sieves (4A powder) 1886 were added to a solution of Intermediate 3 (2 g, 12.4 mmol) in EtOH (20 mL) at RT and 1887 refluxed for 8 h. Then was added a catalytic quantity ofAcOH, NaCNBH3 (1.6 g, 24.8 mmol, 1888 2 eq) at 0 CC and with stirring for 15 h at RT. The solvent was distilled off, and the residue 1889 was dissolved in EtOAc (200 mL) and filtered through a Celite® pad to remove inorganic 1890 als. The filtrate was washed with saturated s NaHC03 (2 X 20 mL), water 1891 (20 mL), brine (20 mL), dried over Na2S04, filtered and concentrated in vacuo. The resultant 1892 compound was purified by column chromatography over silica gel (100-200 mesh) by using a 1893 solvent gradient of 0-10% HC13 as the eluent to afford Intermediate 6 (1.7 g, 51%). 1894 1H NMR: (DMSO-d6) 5 12.50 (s, 1H), 9.06 (d, J: 1.4 Hz, 1H), 8.53-8.55 (m, 1H), 8.17-8.20 1895 (m, 1H), 7.33-7.45 (m, 4H), .19 (m, 2H), 4.40 (d, J: 6.4 Hz, 2H); MS: 270 [M + H]+; 1896 MP: 185-186 0C; TLC: 10% MeOH in CHClgC Rf: 0.25. 1897 1898 Example 13 - Preparation ofIntermediate 7 N N NH2 i. EtOH, ms, 78 °C H / \ Y + —> N N \ N .. / \ _ N,NH u. NaCNBHS, AcOH \ Y 1899 _ N’NH 1900 Intermediate 4 Intermediate 7 1901 [0214] General Procedure 10 was ed to obtain Intermediate 7 (2.8 g, 60%). MS: 252 1902 [M + H]+; MP: 226-228 0C; TLC: 10% MeOH in CHC13: Rf: 0.30. 1903 Example 14 - Preparation ofIntermediate 8 CH0 F N N NH2 i.EtOH,ms,78°C / 3% H \ . _. N N _ N,NH ii. NaCNBH3, AcOH / \ \YN 1904 F _ N H 1905 ediate 4 Intermediate 8 1906 [0215] General Procedure 10 was followed to obtain Intermediate 8 (1.6 g, 48%). 1H NMR: 1907 (DMSO-d6) 8 13.15 (br s, 1H), 8.60 (d, J: 4.0 Hz, 1H), 7.86-7.93 (m, 2H), 7.30-7.42 (m, 1908 3H), 7.02-7.15 (m, 2H), 6.84 (br s, 1H), 4.37 (d, J: 6.2 Hz, 2H); MS: 270 [M + H]+; MP: 1909 219-220 c’C; TLC: 10% MeOH in CHClg: Rf: 0.25. 1910 Example 15 - Preparation ofIntermediate 9 CH0 F NH ' o N\ 2 |. EtOH, ms, 78 C N/ \ + —> N \ Y N H\/©/ _ N,NH ii. NaCNBH3,AcOH N/ \ \fi/ 1911 F _ N’ 1912 Intermediate 5 Intermediate 9 1913 [0216] General Procedure 10 was followed to obtain ediate 9 (1.4 g, 42%). MS: 270 1914 [M + H]+; TLC: 10% MeOH in CHClg: Rf: 0.25. 1915 1916 Example I 6 - Preparation ound 7 1917 [0217] The synthesis of Compound 7 followed General Procedure 11 following. 1918 General Procedure 11 0 EN H 3 N N\ H\/©/ N N + / \ \r / \ NPr CIJK/CH —>3 — N’N O _ NH N, f 1919 CH3 1920 ediate 6 Compound 7 1921 [0218] Propionyl chloride (39 uL, 0.44 mmol, 1.2 eq) was added to a solution of 1922 Intermediate 6 (100 mg, 0.37 mmol) in triethylamine (3 mL) at RT and stirred for 5 h. The 1923 reaction mixture was diluted with water (5 mL) and extracted with EtOAc (20 mL). The 1924 organic layer washed with water (2 X 5 mL), saturated aqueous NaHC03 (5 mL), brine (5 1925 mL), dried over Na2S04, d and concentrated in vacuo. The crude compound was 1926 purified by column chromatography over silica gel (100-200 mesh) by using a gradient 1927 mixture of 0-30% EtOAc-hexane as the eluent to afford Compound 7 (40 mg, 33%). 1H 1928 NMR: d6) 5 9.14 (d, J: 1.8 Hz, 1H), 8.66-8.67 (m, 1H), 8.28-8.34 (m, 2H), 7.47- 1929 7.53 (m, 3H), 7.13-7.17 (m, 2H), 4.63 (d, J: 6.2 Hz, 2H), 3.05 (q, J: 7.5 Hz, 2H), 1.16 (t, J 1930 = 7.5 Hz, 3H); MS: 326 [M + H]+; TLC: 50% EtOAc in hexane: Rf: 0.60. 1931 Example I 7 - ation ofCompound 8 0% *9 —* - 5 _ N,NH 1932 1933 Intermediate 7 Compound 8 1934 [0219] General Procedure 11 was ed to obtain Compound 8 (48 mg, 35%). 1H NMR: 1935 (DMSO-d6) 5 8.71 (d, J: 4.0 Hz, 1H), 8.46 (br s, 1H), 8.13-8.23 (m, 3H), 7.92-7.96 (m, 1H), 1936 7.24-7.52 (m, 6H), 6.88-6.89 (m, 1H), 4.74 (d, J: 6.2 Hz, 2H); MS: 346 [M + H]+; MP: 143- 1937 145 oC; TLC: 50% EtOAc in hexane: Rf: 0.60. 1938 Example 18 - Preparation ofCompound 9 Nx/J:::) O ZIV: H EtN3 <:;>F—<N:IrN N / \ HN N + 0| _ " O 1939 1940 Intermediate 7 nd 9 1941 [0220] General Procedure 11 was ed to obtain Compound 9 (25 mg, 16%). 1H NMR: 1942 (DMSO-d6) 5 8.65 (d, J: 4.0 Hz, 1H), 8.26 (br s, 1H), 8.03 (d, J: 8.1 Hz, 1H), 7.90 (t, J: 1943 7.7 Hz, 1H), 7.19-7.48 (m, 11H), 4.67 (d, J: 6.0 Hz, 2H), 3.30-3.41 (m, 2H), 2.99-3.03 (m, 1944 2H); MS: 384 [M + H]+; MP: 0 0C; TLC: 50% EtOAc in hexane: Rf: 0.40. 1945 Example 19 - Preparation ofCompound 10 F H i [ O N\/© H —3 ’ C>_<\,\NrN N + 8 {weN N CI ‘/ “ 0 1946 1947 Intermediate 8 Compound 10 1948 [0221] General Procedure 11 was followed to obtain Compound 10 (40 mg, 28%). 1H 1949 NMR: (DMSO-d6) 8 8.72 (d, J: 4.6 Hz, 1H), 8.47-8.54 (m, 2H), 8.12-8.23 (m, 2H), 7.94- 1950 7.98 (m, 1H), 7.48-7.52 (m, 3H), 7.34-7.36 (m, 1H), 7.16 (t, J: 9.0 Hz, 2H), 4.71 (d, J: 6.1 1951 Hz, 2H); MS: 380 [M + H]+; MP: 159-160 0C; TLC: 50% EtOAc in hexane: Rf: 0.60. 1952 Example 20 - Preparation ofCompound I I O Hog H Et3N / N \ NerN N + \ N Cl —’ / N / — \ S N’ 0 N \ \ Y \ _ NH 1953 \8 1954 Intermediate 9 Compound 11 1955 [0222] General Procedure 11 was followed to obtain Compound 11 (20 mg, 14%). 1H 1956 NMR: (DMSO-d6) 5 9.19 (d, J: 1.3 Hz, 1H), 8.63-8.73 (m, 3H), 8.00 (d, J: 5.7 Hz, 2H), 1957 7.72-7.88 (m, 2H), 7.50-7.54 (m, 2H), 7.17 (t, J: 8.8 Hz, 2H), 4.70 (d, J: 6.2 Hz, 2H); MS: 1958 380 [M + H]+; MP: 187-188 °C; TLC: 50% EtOAc in : Rf: 0.60. 1959 Example 21 - Preparation ofCompound 12 1960 [0223] The synthesis of Compound 12 ed General Procedure 12 following. 1961 General Procedure 12 H«0+ CIJLN/fi —>DABCO / \ N N,N o — @IN N \ \ be DMF EIj 1962 0 1963 Intermediate 6 Compound 12 1964 [0224] A solution of Intermediate 6 (100 mg, 0.37 mmol) in dry DMF (2 mL) was added to 1965 a solution of morpholinecarbonyl chloride (86 uL, 0.74 mmol, 2 eq), DABCO (124 mg, 1.11 1966 mmol, 3 eq) in DMF (3 mL) at RT and stirred for 2 h. The reaction mixture was diluted with 1967 water (10 mL) and extracted with EtOAc (30 mL). The organic layer washed with water (2 X 1968 5 mL), ted aqueous NaHC03 (2 X 5 mL), brine (10 mL), dried over Na2S04, d and 1969 concentrated in vacao to get a crude e. The crude compound was purified by column 1970 chromatography over silica gel (100-200 mesh) by using a nt mixture of 0-50% 1971 EtOAc-hexane as the eluent to afford Compound 12 (33 mg, 23%). 1H NMR: (DMSO-d6) 5 1972 9.11 (s, 1H), 8.64 (d, J: 4.8 Hz, 1H), 8.25 (d, J: 7.9 Hz, 1H), 7.90 (s, 1H), 7.46-7.52 (m, 1973 3H), 7.16 (t, J: 8.8, 2H), 4.59 (d, J: 6.2 Hz, 2H), 3.70-3.99 (m, 8H); MS: 383 [M + H]+; 1974 TLC: 50% EtOAc in hexane: Rf: 0.40. 1975 Example 22 - Preparation ofCompound 13 1976 [0225] A useful scheme for the preparation of compounds of the type of Compound 13 is 1977 provided in Scheme 1 following. 1978 Schemel N NH NB 10° N H M NH N EtOH THF I H CH3 Intermediate 10 Intermediate 11 \ CI Et3N,CHCI3 H3C O ©_<\N\(NCH3 CIJK/CHS H C 3N H4N2°H20 N \ N N N ,N _ \CHs | N # / )I\ ,CH3 1979 Compound 13 Intermediate 13 Intermediate 12 1980 [0226] A detailed description of the preparation of Intermediates 10-13 and Cmpd 24 1981 s. 1982 Preparation of ediate 10 [::1: NN. CNBr1d%Na0H ’ iii N N acetone, EtOH <:;>\[<1 ‘ Intermediate 10 1983 1984 [0227] A solution of cyanogen bromide (1.3 g, 12.6 mmol) in acetone (5 mL) was added 1985 portion-wise slowly to a mixture of benzotriazole (3 g, 25.2 mmol, 2 eq) in EtOH (50 mL) 1986 followed by 10% aqueous NaOH (6 mL, 12.6 mmol, 1 eq) at 0 OC. The on mixture was 1987 then stirred at RT for 30 min. Solid formation was observed. The solid was filtered and 1988 washed with cold EtOH. The resulting material was recrystallized from benzene to afford 1989 Intermediate 10 (2.2 g, 33%) as a white solid. 1H NMR: (DMSO-d6) 5 11.76 (s, 1H), 8.29- 1990 8.39 (m, 2H), 7.86-8.09 (m, 2H), 7.44-7.72 (m, 4H), MS: 264 [M + H]+; TLC: 30% EtOAc in 1991 hexane: Rf: 0.50. 1992 Preparation of Intermediate 11 NH NH N’N M62NH Btii‘ 03, H =N N=N CH3 Intermediate 10 Intermediate 11 1993 1994 [0228] Dimethylamine (1.59 mL, 7.60 mmol, 1 eq) was added to Intermediate 10 (2 g, 7.60 1995 mmol) in THF (30 mL) at RT and the resulting mixture was allowed to stir for 24 h. The 1996 solvent was evaporated and the residue was dissolved in DCM (100 mL). The organic layer 1997 was washed with 10% N32CO3 (3 X 5 mL), brine (10 mL), dried over Na2S04, filtered and 1998 concentrated in vacuo to afford ediate 11 (1.2 g, 71%) as a light yellow liquid which 1999 was used without additional purification. 1H NMR: (DMSO-d6) 5 8.17 (d, J = 8.4 Hz, 1H), 2000 7.65-7.80 (m, 3H), 7.49-7.53 (m, 1H), 2.87 (s, 6H); MS: 190 [M + H]+; TLC: 30% EtOAc in 2001 hexane: Rf: 0.30. 2002 Preparation of Intermediate 12 NH O I. (COCI)21 DMF DCM 1 N \ N I I ii. Et3N,CHCI3 / / Bt N/CH3 CH3 | Intermediate 11 Intermediate 12 2003 2004 [0229] Oxalyl chloride (2 mL, 23.3 mmol, 1.4 eq) was added to a solution of nicotinic acid 2005 (2 g, 16.3 mmol) in DCM followed by catalytic amount of DMF (0.5 mL) at 0 CC and stirred 2006 for 5 h at RT. The solvent was then evaporated to afford nic acid chloride as a yellow 2007 solid. Nicotinic acid chloride (1.1 g, 7.93 mmol, 1.5 eq) was then added to a solution of 2008 Intermediate 11 (1 g, 5.29 mmol) in CHC13 (30 mL) ed by Et3N (0.7 mL, 5.29 mmol, 1 2009 eq) at 0 CC. The reaction mixture was d to warm to RT for stir for 18 h. The mixture 2010 was then diluted with CHC13 (20 mL). The organic layer was washed with water (10 mL), 2011 brine (10 mL), dried over Na2S04, filtered and concentrated in vacuo. The resulting 2012 nd was ed by column chromatography over silica gel (100-200 mesh) by using a 2013 gradient mixture of 0-50% EtOAc-hexane as the eluent to afford Intermediate 12 (900 mg, 2014 60%) as a white solid. MS: 295 [M + H]+; TLC: 50% EtOAc in DCM: Rf: 0.40. 2015 Preparation of Intermediate 13 N \ N NH2NH2‘H20 N I I Q43,N_ N \ \CH3 / 8&1,CH3 cm.
Intermediate 12 Intermediate 13 2016 2014/030853 2017 [0230] Hydrazine hydrate (5 mL) was added solution of Intermediate 12 (900 mg, 25.2 2018 mmol) in chloroform (20 mL) at RT and the resulting e was allowed to stir for 24 h. 2019 The mixture was diluted with excess CHC13 (20 mL). The organic layer was then washed 2020 with water (15 mL), brine (10 mL), dried over NazSO4, filtered and concentrated in vacuo. 2021 The crude residue was partially purified by column chromatography over silica gel 00 2022 mesh) by using a gradient mixture of 0-50% EtOAc-hexane as the eluent to afford 2023 Intermediate 13 (150 mg) as a thick brown mass. MS: 190 [M + H]+; TLC: 10% MeOH in 2024 CHC13: Rf: 0.30. 2025 Preparation of Compound 13 Et3N \—/ ©_<N\rNCH3 N,NH ' f0 2026 CH3 2027 Intermediate 13 Compound 13 2028 [0231] General Procedure 11 was followed to obtain Compound 13 (13 mg, 6%). 1H NMR: 2029 (DMSO-d6) 5 9.15 (s, 1H), 8.68 (d, J: 3.5 Hz, 1H), 8.31 (d, J: 7.9 Hz, 1H), 7.53 (dd, J: 2030 7.9, 4.8 Hz, 1H), 3.04-3.14 (m, 8H), 1.15 (t, J: 7.3 Hz, 3H); MS: 246 [M + H]+; TLC: 50% 2031 EtOAc in DCM: Rf: 0.50. 2032 Example 23 - Preparation ofCompound 14 2033 [0232] A general chemical scheme for the formation of compounds of the type of 2034 Compound 14 is provided in General Scheme 111 following, wherein the terms “ , X”,“Ry”, 2035 and “R” are independently hydrogen, substituted or unsubstituted alkyl, substituted or 2036 unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or tituted 2037 cycloalkyl, substituted or unsubstituted heterocycloalkenyl, substituted or unsubstituted 2038 aryl, or substituted or tituted aryl, or other groups obVious to those skilled in the 2039 art. General Scheme III H 0 0 pyrIdIne. . H KOH SH RX CI 8 H H20 NzNH 100°C N S‘Ry RZCOCI Rx_<\ T N\ Sway N’ \f0 Et3N RX—<\ 2040 RZ 2041 [0233] A detailed description of the preparation of Intermediates 14, 15 and Compound 14 2042 follows. 2043 Preparation of Intermediate 14 O O _I-(COC|)2,DMF H KOH N SH ,N NH2 N \ OH —>.. N \ N \n/ —> / \ >( . . . \ | ||. thlosemlcarbaZIde | H H20 _ / pyridine / S N,NH 100 °c 2044 2045 Intermediate 14 2046 [0234] Oxlayl chloride (5.4 mL, 61.0 mmol, 1.5 eq) and DMF (3 mL) was added 2047 sequentially to a solution of nicotinic acid (5 g, 40.7 mmol) in dry DCM (300 mL) at RT. The 2048 reaction mixture was d to stir at RT for 2 h. The solvent was removed and co-distilled 2049 with dry toluene (2 X 50 mL) and to afford 5 g of crude nicotinic acid de (5 g, 2050 35.5 mmol). This material was added slowly portion-wise to a solution of thiosemicarbazide 2051 (5 g, 54.9 mmol, 1.5 eq) in pyridine (50 mL) at 0 0C over a period of 1 h and then allowed to 2052 stir at RT for 14 h. The reaction mixture was neutralized with saturated aqueous NaHC03 2053 (30 mL) and extracted with t—BuOH (3 X 100 mL) and dried over NaZSO4, filtered and 2054 concentrated in vacuo. The crude residue was dissolved in water (20 mL) along with 10% 2055 aqueous KOH (50 mL) and the ing mixture was allowed to stir at 100 CC for 3 h. The 2056 on e was then cooled to 0 OC and lized with 10% aqueous AcOH (60 mL), 2057 extracted with EtOAc (2 X 150 mL), dried over NaZSO4, filtered and concentrated in vacuo to 2058 afford crude Intermediate 14 (1.2 g) as an off-white solid. MS: 179 [M + H]+; TLC: 20% 2059 MeOH/NH3 in CHCl3C Rf: 0.30. 2014/030853 2060 Preparation of Intermediate 15 N SH WYN\ Br H20,THF + —»N s — N’NH F / \ \\ 2061 _ N,NH 2062 Intermediate 14 Intermediate 15 2063 [0235] 4-Fluorobenzyl bromide (0.12 mL, 1.01 mmol, 0.6 eq) was added to a solution of 2064 Intermediate 14 (300 mg, 1.68 mmol) in water (5 mL) and THF (15 mL) at -10 CC and the 2065 reaction mixture was d to stir at -10 0C for 8 h. The solvent was removed and the 2066 residue was diluted with water (10 mL) and extracted with EtOAc (50 mL). The c layer 2067 was washed with water (15 mL), saturated aqueous NaHC03 (10 mL), brine, dried over 2068 Na2S04, filtered and concentrated in vacuo. The crude compound was purified by column 2069 chromatography over silica gel 00 mesh) by using a solvent gradient mixture of 0-10% 2070 MeOH-CHC13 as the eluent to afford Intermediate 15 (110 mg, 23%) as an off-white solid. 2071 MS: 287 [M + H]+; TLC: EtOAc: Rf: 0.40. 2072 Preparation of Compound 14 o OCH3 N ,\ orN N s —> Mr_ N Cl _ N,N 0 N,NH OCH3 2073 2074 Intermediate 15 Compound 14 2075 [0236] General Procedure 11 was followed to obtain Compound 14 (20 mg, 30%). 1H 2076 NMR: (DMSO-d6) 8 9.13 (s, 1H), 8.71 (d, J: 4.0 Hz, 1H), 8.26 (d, J: 7.9 Hz, 1H), 7.53- 2077 7.67 (m, 5H), 7.09-7.25 (m, 4H), 4.64 (s, 2H), 3.75 (s, 3H); MS: 421 [M + H]+; MP: 108-112 2078 0C; TLC: 30% EtOAc in hexane: Rf: 0.40. 2079 Example 24 - Preparation ofIntermediate 1 6 2080 O H O NH pyridine H KOH N + H N’ —> ,N NH2 —> I \ §r \ \ H H20 ,NH S 3 N 3 3 3 100°C 2081 2082 Intermediate 16 2083 [0237] 2-Thiophene carboxylic acid chloride (6.5 mL, 60.4 mmol) was added slowly 2084 portionwise to a on of thiosemicarbazide (5 g, 54.9 mmol, 1.1 eq) in pyridine (50 mL) at 2085 0 °C over a period of 1 h and then allowed to stir at RT for 14 h. The reaction mixture was 2086 neutralized with saturated s NaHC03 (50 mL) and extracted with t—BuOH (3 X 100 2087 mL) and dried over Na2S04, filtered and concentrated in vacuo. The crude residue was 2088 dissolved in water (30 mL) along with 10% aqueous KOH (60 mL) and the resulting mixture 2089 was allowed to stir at 100 0C for 3 h. The reaction mixture was then cooled to 0 oC and 2090 neutralized with 10% aqueous AcOH, extracted with EtOAc (2 X 150 mL), dried over 2091 Na2S04, filtered and concentrated in vacao to afford crude Intermediate 16 (1.2 g) as an off- 2092 white solid. MS: 184 [M + H]+; TLC: 10% MeOH/NH3 in CHCl3C Rf: 0.60. 2093 Example 25 - Preparation ofIntermediate 1 7 2094 SH 8 04*N CH ‘°’ I, 1M NaOH \ EtOH [HiN 2095 S N,NH S N,NH 2096 Intermediate 16 Intermediate 17 2097 [0238] A solution of methyl iodide (65 uL, 1.04 mmol, 1.6 eq) in EtOH (2 mL) was added 2098 to a solution of Intermediate 16 (120 mg, 0.66 mmol) in 1M aqueous NaOH (3 mL) at RT 2099 and the resulting mixture was allowed to stir for 3 h. The reaction mixture was then 2100 lized with 10% s AcOH (5 mL) and extracted with EtOAc (30 mL). The organic 2101 layer was washed with water (10 mL), saturated aqueous NaHC03 (5 mL), brine, dried over 2102 Na2S04, filtered and concentrated in vacuo. The crude compound was d by column 2103 chromatography over silica gel (100-200 mesh) by using a solvent gradient mixture of 0-10% 2104 MeOH-CHC13 as the eluent to afford Intermediate 17 (90 mg, 70%) as an off-white solid. 1H 2105 NMR: d6) 8 14.19 (br s, 1H), .67 (m, 2H), 7.16-7.18 (m, 1H), 2.60 (s, 3H); 2106 MS: 198 [M + H]+; TLC: 50% EtOAc in hexane: Rf: 0.50. 2107 Example 26 - Preparation ofCompound 15 \ N\ S\CH3 o OCH3 / T N \§(N S\CH Et3N — N/N o / \ 3 + Cl —> _ N,NH OCH3 2108 2109 Intermediate 18 Compound 15 2110 2111 [0239] General Procedure 11 was followed to obtain Cmpd 29 (30 mg, 29%). 1H NMR: 2112 (DMSO-d6) 8 7.72 (d, J: 4.8 Hz, 1H), 7.56-7.65 (m, 3H), 7.25 (d, J: 8.8 Hz, 1H), 7.09-7.24 2113 (m, 2H), 3.77 (s, 3H), 2.73 (s, 3H); MS: 332 [M + H]+; MP: 165-167 c’C; TLC: 30% EtOAc in 2114 hexane: Rf: 0.40. 2115 Example 2 7 - Preparation ofCompound I 6 2116 [0240] General Scheme IV. A synthetic scheme useful for synthesis of compounds 2117 described herein including Compound 16 is disclosed in General Scheme IV following 2118 wherein the terms “ X”,“Ry”, and “R” are independently en, substituted or 2119 unsubstituted alkyl, substituted or unsubstituted alkyl, substituted or unsubstituted 2120 cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or tituted 2121 heterocycloalkenyl, substituted or tituted aryl, or substituted or unsubstituted 2122 heteroaryl, or other groups obVious to those skilled in the art. 2123 General Scheme IV HZN,N\n/NH2 0 SOCI2 o NaOMe NYNHz Rx OH EtOH,78°C t MeOH, 65°C NHX i. RVCHO, EtOH, 78 °c ii. NaCNBH3, AcOH N Ry RX{N§( V RZCOCI N HVRV \N’N O \F Et3N RX—QY N’NH 2124 RZ 2125 [0241] A description of the sis of Intermediates l9-24 and Compound 16 follows. 2126 Synthesis of Intermediate 19 followed General Procedure 13 following. 2127 Preparation of Intermediate 19 [General Procedure 13] 2128 [0242] General Procedure 13 was followed in the preparation of Intermediate 19. 2129 General Procedure 13 O O SOCI N 2 N If \ OH —°> If \ oACHs N / EtOH, 78 c N / 2130 2131 Intermediate 19 2132 [0243] Thionyl chloride (3.55mL, 48.4 mol, 3 eq) was added dropwise to a solution of 2133 pyrimidinecarboxylic acid (2 g, 16.1 mmol) in EtOH (15 mL) and the ing e 2134 was heated to reflux for 14 h. The mixture was then cooled to RT and made alkaline with 2135 saturated aqueous NaHC03 to pH 8. The basic solution was then extracted with EtOAc (4 X 2136 50 mL). The combined organic layers were washed with brine (30 mL), dried over , 2137 filtered and concentrated in vacuo to afford Intermediate 19 (1.7g, 77%). 1H NMR: (DMSO- 2138 d6) 5 9.40 (d, J: 1.0 Hz, 1H), 9.10 (d, J: 5.1 Hz, 1H), 8.05 (dd, J: 5.1, 1.3 Hz, 1H), 4.39 2139 (q, J: 7.1 Hz, 2H), 1.35 (t, J: 7.1 Hz, 3H); MS: 153 [M + H]+; TLC: 40% hexane in EtOAc: 2140 Rf: 0.40. 2141 Preparation of Intermediate 20 O O SOCI2 N \ OH —> N \ o/\CH3 i / EtOH, 78°C l / 2142 N N 2143 ediate 20 2144 [0244] General Procedure 13 was followed to obtain crude Intermediate 20 (950 mg, 86%). 2145 1H NMR: (DMSO-d6) 8 9.43 (s, 1H), 9.26 (s, 2H), 4.39 (q, J: 7.1 Hz, 2H), 1.35 (t, J: 7.1 2146 Hz, 3H); TLC: 40% EtOAc in hexane: Rf: 0.50. 2147 Preparation of Intermediate 21 [General Procedure 14] 2148 [0245] General Procedure 14 was followed in the preparation of ediate 21. 2149 General Procedure 14 N N OMea NH2 /\ H2N’N\n/NH2 N\/_>_<\\(/_N N\ f \ 0 CH3 + N / — N’NH 2150 NH MeOH,0—65°C 2151 Intermediate 19 Intermediate 21 2152 [0246] Intermediate 19 (1.6 g, 10.5 mmol) was added dropwise to a vigorously stirring 2153 mixture of aminoguanidine sulfate (10.3 g, 42.1 mmol, 4 eq) in freshly prepared NaOMe 2154 (using 968 mg, 42.1 mmol ofNa in 28 mL of dry MeOH) at 0 CC. The resulting mixture was 2155 heated to reflux for 20 h. The mixture was then cooled to RT, carefully poured over ice cold 2156 water (20 mL) and concentrated in vacuo. The crude residue was purified over neutral 2157 alumina using 4-10% MeOH-CHC13 as the eluent to give Intermediate 21 (500mg, 26%). 2158 MS: 163 [M + H]+; TLC: 20% MeOH in CHC13: Rf: 0.20. 2159 Preparation of Intermediate 22 H NaOMe N N NH2 N \ o/\CH3 + HN/N NH2 \ \7/ m 2 \H/ </ \ N/ NH MeOH, 0—65 °c N— N’ 2160 2161 Intermediate 20 Intermediate 22 2162 [0247] General Procedure 14 was followed to obtain Intermediate 22 (500 mg, 45%). 1H 2163 NMR: (DMSO-d6) 8 12.44 (br s, 1H), 9.17-9.18 (m, 3H), 6.32 (s, 2H); TLC: 20% MeOH in 2164 CHC13: Rf: 0.20. 2165 Preparation of Intermediate 23 N NH ' EtOH N 2 I.
° H : , 78 c NCH# N or VN + —. Nfl _ N’NH ii. NaCNBHg, ACOH — N’NH 2166 2167 Intermediate 21 Intermediate 23 2168 [0248] l Procedure 10 was followed to obtain Intermediate 23 (210 mg, 34%). 1H 2169 NMR: d6) 8 12.80 (s, 1H), 9.18 (s, 1H), 8.83 (s, 1H), 7.92 (d, J: 4.4 Hz, 1H), 7.25- 2170 7.40 (m, 5H), 4.44 (d, J: 5.7 Hz, 2H); TLC: EtOAc: Rf: 0.30. 2171 Preparation of Intermediate 24 «“546NH2 i. EtOH, 78 °c N + —» @m ii. NaCNBHg, ACOH N— NrNH N_ N’NH 2172 2173 ediate 22 Intermediate 24 2174 [0249] General Procedure 10 was followed to obtain Intermediate 24 (160 mg, 20%). MS: 2175 253 [M + H]+; TLC: EtOAc: Rf: 0.30. 2176 Preparation of Compound 16 al Procedure 11] 2177 [0250] General Procedure 11 was followed in the preparation of Compound 16. 2178 General Procedure 11 2179 2180 Intermediate 23 Compound 16 2181 [0251] 2-Methoxybenzoyl chloride (72 uL, 0.54 mmol, 2 eq) was added to a solution of 2182 Intermediate 23 (70 mg, 0.27 mmol) in Eth (0.18 mL, 1.35 mmol) and DCM (3 mL) at 0 CC. 2183 The resulting mixture was allowed to stir at RT for 2 h. The reaction mixture was then diluted 2184 with water (5 mL) and extracted with DCM (3 X 15 mL). The combined organic layers were 2185 washed with saturated aqueous NaHC03 (10 mL), water (2 X 5 mL), brine (15 mL), dried 2186 over Na2S04, filtered and concentrated in vacuo. The crude material was purified by column 2187 chromatography over silica gel (100-200 mesh) using a gradient mixture of 0-70% EtOAc- 2188 hexane as the eluent to afford Compound 16 (45 mg, 29%). 1H NMR: (DMSO-d6) 5 9.21 (s, 2189 1H), 8.90 (d, J: 5.1 Hz, 1H), 8.59 (t, J: 6.0 Hz, 1H), 7.93 (d, J: 5.1 Hz, 1H), 7.08-7.60 (m, 2190 10H), 4.72 (d, J: 5.7 Hz, 2H), 3.77 (s, 3H); MS: 387 [M + H]+; MP: 5 C’C; TLC: 40% 2191 hexane in EtOAc: Rf: 0.30. 2192 Example 28 - Preparation ofCompound I 7 H\/© O OCH3 N </ :\>—<\NT IHN N \ C©DCEt3N N“ N’ 2193 2194 Intermediate 24 Compound 17 2195 [0252] General ure 11 was ed by preparative HPLC purification to obtain 2196 Compound 17 (30 mg, 16%). 1H NMR: d6) 8 9.26 (s, 1H), 9.11 (s, 2H), 8.64 (t, J: 2197 6.3 Hz, 1H), 7.07-7.60 (m, 9H), 4.71 (d, J: 6.3 Hz, 2H), 3.78 (s, 3H); MS: 387 [M + H]+; 2198 MP: 154-157 0C; TLC: 40% EtOAc in hexane: Rf: 0.20. 2199 Example 29 - Preparation ofCompound 18 2200 [0253] General Scheme V. A tic scheme useful for synthesis of compounds 2201 described herein including Compound 18 is disclosed in General Scheme V following, 1 00 2202 wherein the terms “ X”,“Ry”, and “R” are independently hydrogen, substituted or 2203 unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted 2204 lkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted 2205 heterocycloalkenyl, substituted or unsubstituted aryl, or substituted or unsubstituted 2206 heteroaryl, or other groups obvious to those skilled in the art.General Scheme V N NH 0 (00002 O i. pyridine NYNHZ OH M RxJkCi ii. H20,100°C RX_<\N,NH i. RYCHO, EtOH, 78 °c ii. NaCNBHg, AcOH Ry H NerNV y RZCOCI N ‘— MN§|/"‘\/R N’ O Y [NH Et3N N 2207 RZ 2208 [0254] A description of the synthesis of Intermediates 25, 26 and Compound 18 follows. 2209 Preparation of ediate 25 HZN,N\n/NH2 “1ng C(OC|)2 MCI i. pyridine OH —> CN/ N\ DMF, DCM ii. H20, 100 °C \N’NH >_<NYNHZ 2210 {/N 2211 Intermediate 25 2212 [0255] Oxalyl chloride (2.36 mL, 24.2 mmol, 1.5 eq) and a catalytic quantity ofDMF were 2213 added to a solution of pyrimidinecarboxylic acid (2 g, 16.1 mmol) in dry DCM (30 mL) at 2214 0 CC. The resulting mixture was allowed to warm to RT and stir for 3 h. The volatiles were 2215 removed in vacuo and the e was ghly dried to afford pyrimidinecarboxylic 2216 acid de (2.1 g, 14.8 mmol) as a black solid. The crude material was added portion-wise 2217 to a solution of aminogaunidine sulfate (5.5 g, 22.2 mmol, 1.5 eq) in pyridine (20 mL) at 2218 0 CC. The resulting mixture was allowed to warm to RT and stir for 14 h. The mixture was 2219 then neutralized with saturated s NaHCOg, ted with t—BuOH (5 X 50 mL), dried 2220 over NaZSO4, filtered and concentrated in vacuo. The crude material was dissolved in water 2221 (45 mL) and the resulting solution was heated to 100 CC for 24 h. The reaction mixture was 2222 then cooled to RT, extracted with t—BuOH (5 X 30 mL), dried over Na2S04, filtered and 2223 concentrated in vacao to afford Intermediate 25 (650 mg, 25 %) as off-white solid. TLC: 30% 2224 MeOH in CHC13: Rf: 0.20. 2225 Preparation of Intermediate 26 {w +6N N NW LBOH78m N {m N\ HQ 2226 -—N NgNH iLNaCNBH3Ad3H -—N N/NH 2227 Intermediate 25 Intermediate 26 2228 [0256] General Procedure 10 was followed to obtain Intermediate 26 (120 mg, 17%). MS: 2229 253[NI+Ifl+;TLC:Efl)Ac:Rg(l30 2230 Preparation of nd 18 H\/© N N o OCH3 N\ Et3N / H \r N N N\ + CI —> — ’N <::§_2g‘7/ N N O 2231 2232 Intermediate 26 Compound 18 2233 2234 [0257] General Procedure 11 was followed to obtain Compound 18 (32 mg, 21%). 1H 2235 NMR: (DMSO-d6) 8 8.86 (d, J: 5.1 Hz, 2H), 8.44 (t, J: 6.0 Hz, 1H), 7.08-7.59 (m, 10H), 2236 4.73 (d, J: 6.3 Hz, 2H), 3.77 (s, 3H); MS: 387 [M + H]+; MP: 203-205 c’C; TLC: 40% 2237 hexane in EtOAc: Rf: 0.40. 2238 Example 30 - Preparation ofCompound 19 2239 [0258] General Scheme VI. A synthetic scheme useful for synthesis of nds 2240 described herein including Compound 19 is disclosed in General Scheme VI following, 2241 wherein the terms “ ”, and “R” are independently hydrogen, substituted or 2242 unsubstituted alkyl, tuted or unsubstituted heteroalkyl, substituted or unsubstituted 2243 cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted 2244 heterocycloalkenyl, substituted or unsubstituted aryl, or substituted or unsubstituted 2245 aryl, or other groups s to those skilled in the art. 2246 General Scheme VI R H R H i. RVCHO, EtOH, 78 °c NVRY RZCOCI N\/Ry , _> RX RX ii. NaCNBH3, AcOH \N’NH EtaN \N’NYO 2247 R = H, CH3 Rz 2248 Preparation of Intermediate 27 2249 [0259] General Procedure 10 was followed in the ation of Intermediate 27. 2250 General Procedure 10 i EtOH MS 78 °C N.g N,NH \ ii. 3 AcOH \N’NH 2251 2252 Intermediate 27 2253 [0260] 4-Fluorobenzaldehyde (0.54 mL, 5.03 mmol, 2 eq) and molecular sieves (4 A 2254 powder) were added to a solution of ophenylpyrazole (400 mg, 2.51 mmol) in 2255 EtOH (20 mL) at RT and the resulting mixture was heated to reflux. After 8 h, the reaction 2256 mixture was cooled to 0 oC and AcOH (0.4 mL) and NaCNBH3 (316 mg, 5.03 mmol, 2 eq) 2257 were added. The mixture was then allowed to warm to RT and stir for 15 h. The solvent was 2258 evaporated and the residue was dissolved in EtOAc (100 mL) and filtered through a Celite 2259 pad to remove nic materials. The filtrate was then washed with saturated aqueous 2260 NaHC03 (2 X 20 mL), water (20 mL), brine (20 mL), dried over Na2S04, filtered and 2261 concentrated in vacuo. The crude material was purified by column chromatography over 2262 silica gel (100-200 mesh) by using a solvent gradient of 0-50% EtOAc-petroleum ether as the 2263 eluent to afford Intermediate 27 (240 mg, 36%) as an off white solid. MS: 268 [M + H]+; 2264 TLC: EtOAc: Rf: 0.60. 2265 ation of Compound 19 2266 [0261] General Procedure 15 was followed in the preparation of Compound 19. 2267 General Procedure 15 H\/©/ + O EtSN \ zNH H30 CH3 H3C CH3 2268 H30 2269 ediate 27 nd 19 2270 [0262] Pivaloyl chloride (32 uL, 0.26 mmol, 1.2 eq) was added to a solution of 2271 Intermediate 27 (60 mg, 0.22 mmol) in triethylamine (3 mL) at RT and stirred for 3 h. The 2272 reaction mixture was diluted with water (5 mL) and extracted with EtOAc (20 mL). The 2273 organic layer was washed with water (2 X 5 mL), saturated aqueous NaHC03 (5 mL), brine 2274 (5 mL), dried over Na2S04, filtered and concentrated in vacuo. The crude nd was 2275 purified by column chromatography over silica gel (100-200 mesh) by using a gradient 2276 mixture of 0-10% EtOAc-hexane as the eluent to afford Compound 19 (23 mg, 29%). 1H 2277 NMR: (DMSO-d6) 5 7.79-7.84 (m, 3H), 7.37-7.49 (m, 5H), 7.17 (t, J: 8.8 Hz, 2H), 5.89 (s, 2278 1H), 4.38 (d, J: 6.2 Hz, 2H), 1.49 (s, 9H); MS: 352 [M + H]+; TLC: 20% EtOAc in hexane: 2279 Rf: 0.60. 2280 Example 31 - Preparation ofCompound 20 2281 [0263] General Scheme VII. A synthetic scheme useful for synthesis of compounds 2282 described herein including Compound 20 is disclosed in General Scheme VII following, 2283 wherein the terms “ X”,“Ry”, and “R” are independently hydrogen, substituted or 2284 unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted 2285 cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or tituted 2286 heterocycloalkenyl, substituted or unsubstituted aryl, or substituted or tituted 2287 heteroaryl, or other groups obvious to those d in the art. 2288 l Scheme VII 0 SOC'z 0 0 NaH, CH3CN H2NNH2'H20 xJ\ —> o XJ\ R OH EtOH, 78 c R OEt touene,I 65 °c RXJk/CN EtOH, 78 co H Ry . NVRy NH2 I. RYCHO, EtOH,78°C RCOCIZ v N,NH ||.NaCNBH3,AcOH \N’NH Et3N N’ Y0 2289 R2 2290 [0264] A description of the syntheses of Intermediates 28-31 and Compound 20 follows. 2291 Preparation of Intermediate 28 [General Procedure 16] 2292 [0265] General Procedure 16 was followed in the preparation of Intermediate 28. 2293 l ure 16 o o SOCI2 N N \ OH —>o \ OEt | Et H,78 °c | 2294 / / 2295 Intermediate 28 2296 [0266] Thionyl chloride (5.4 mL, 73.2 mmol, 3 eq) was added to a solution of picolinic 2297 acid (3 g, 24.4 mmol) in EtOH (50 mL) at 0 CC. The resulting mixture was heated to reflux 2298 and allowed to stir for 2 h. The mixture was then cooled and the solvent was evaporated. The 2299 resulting residue was poured into saturated aqueous NaHC03 and extracted with EtOAc (2 X 2300 50 mL). The combined organic layers were dried over NaZSO4, d and concentrated in 2301 vacuo. The crude material was purified by column chromatography over silica gel (100-200 2302 mesh) by using DCM as the eluent to afford Intermediate 28 (3 g, 81%) as a colorless liquid. 2303 MS: 152 [M + H]+; TLC: 10% H3 in CHClg: Rf: 0.70. 2304 Preparation of Intermediate 29 al Procedure 17] 2305 [0267] General Procedure 17 was followed in the preparation of Intermediate 29. 2306 General Procedure 17 O O N NaH, CH ON N\ CN \ CE —3, | | 2307 toluene, 65 °C / 2308 Intermediate 28 Intermediate 29 2309 [0268] A solution of ediate 28 (3 g, 19.6 mmol) and CH3CN (0.8 mL, 19.6 mmol, 1 2310 eq) in dry toluene (10 mL) was slowly added to a mixture ofNaH (784 mg, 19.6 mmol, 1 eq, 2311 60% in mineral oil) in toluene (50 mL) at 65 CC. The resulting e was allowed to stir at 2312 65 0C for 16 h. The reaction mixture was then cooled to RT and quenched with ice cold water 2313 (20 mL). The resulting solid was filtered to afford Intermediate 29 (1.5 g, 53%) as a brown 2314 solid. 1H NMR: (CDC13)5 8.70 (d, J: 4.8 Hz, 1H), 8.12 (d, J: 7.5 Hz, 1H), 7.90-7.94 (m, 2315 1H), 7.56-7.60 (m, 1H), 4.41 (s, 2H); MS: 147 [M + H]+; TLC: EtOAc: Rf: 0.40. 2316 Preparation of Intermediate 30 [General Procedure 18] 2317 [0269] General Procedure 18 was followed in the preparation of Intermediate 30. 1 05 2318 l Procedure 15 N CN 'H20 \ \ ’ / N\ | o \ EtOH, 78 c _ N,NH 2319 / 2320 Intermediate 29 Intermediate 30 2321 [0270] Hydrazine hydrate (0.34 mL, 6.8 mmol, 1 eq) was added to a solution of 2322 Intermediate 29 (1 g, 6.8 mmol) in EtOH (30 mL) at RT. The mixture was then heated to 2323 reflux and allowed to stir for 20 h. The solvent was then evaporated. The resulting crude 2324 material was triturated with EtzO (2 X 20 mL) and dried under vacuum to afford Intermediate 2325 30 (700 mg, 64%) as a brown liquid. 1H NMR: (DMSO-d6) 8 8.53 (d, J: 4.4 Hz, 1H), 7.78 2326 (d, J: 4.4 Hz, 2H), 7.23-7.26 (m, 1H), 5.95 (s, 1H), 4.84 (br s, 2H); MS: 161 [M + H]+; TLC: 2327 EtOAc: Rf: 0.20. 2328 ation of Intermediate 31 N i. EOH, MS,t 78 °C N H\/©/ MT”I/_ / \ \\ ii. NaCNBH3 AcOH — N’NH 2329 2330 Intermediate 30 Intermediate 31 2331 [0271] General Procedure 10 was followed to afford Intermediate 31 (450 mg). MS: 269 2332 [M + Hr; TLC: EtOAc: Rf: 0.40. 2333 Preparation of Compound 20 /\\ /\\ H30 CH \ _ \N,NH MIN 0 HacICH3 2334 H30 2335 Intermediate 31 Compound 20 2336 [0272] General Procedure 11 was followed to afford Compound 20 (40 mg, 30%). 1H 2337 NMR: (DMSO-d6) 8 8.58 (d, J: 4.4 Hz, 1H), 7.86-7.98 (m, 3H), 7.38-7.46 (m, 3H), 7.18 (t, 2338 J: 8.8 Hz, 2H), 5.84 (s, 1H), 4.40 (d, J: 6.2 Hz, 2H), 1.50 (s, 9H); MS: 353 [M + H]+; MP: 2339 3 0C; TLC: 20% EtOAc in hexane: Rf: 0.60. 2340 Example 33 - General Scheme VIII 2341 [0273] A synthetic scheme useful for synthesis of compounds described herein is disclosed 2342 in General Scheme VIII following, wherein the terms “Ar,” “R1” and “R2” are as defined in 2343 Example 1. 2344 General Scheme VIII O N\ NH2 NVLOE N (EtO)3CH \\ ArNHNH2, NaOAc Ar—N’ —> OEt —> / A020 140 °c | 0 A020, H20, 110 °C EtO OEt RlX, NaH N\ N~R1 Ar—N' H LTiAIFH4 / a. KOH MeOH THF ,N\ Ar_N o Ar_N b. HN(OMe)Me-HCI O ““9, \(.3 EDC|,HOBt,DMF , DMAP Et3N, THF BOC eoc H N\ N\ 1 R2MgX R1 R a MnO2, THF Ar—NIN\ Ar—N H—> Ar—N’N\ —> / OH b. TFA, DCM O 2345 R2 2346 Example 34 - Preparation ofIntermediate 32 (EtO)3CH N\\ —> OEt OEt I A020 140°C 2347 EtO 2348 Intermediate 32 2349 2350 [0274] A on of ethyl cyanoacetate (20 g, 176.8 mmol) and triethyl orthoformate 2351 (29.4 mL, 176.8 mmol) in acetic anhydride (100 mL) was heated to 140 °C and allowed to 2352 stir for 5 h. The solvent was then evaporated to afford crude Intermediate 32 (23 g, 76%) as a 2353 low g solid. MS: 170 [M + H]+; TLC: 30% EtOAc in hexane: Rf: 0.40. 2354 Example 35 - Preparation ofIntermediate 33 EtIO#03 N aOAc A020 , \ + 0"“N / _ 0 H20 110 °c 2355 OEt 2356 Intermediate 32 Intermediate 33 2357 2358 2359 [0275] Sodium acetate (8.2 g, 100 mmol, 2 eq) was added to a solution of Intermediate 32 2360 (8.45 g, 50.0 mmol) and 2-hydrazinopyridine (5 g, 45.5 mmol, 0.9 eq) in AcOH (100 mL) 2361 and water (20 mL). The resulting mixture was heated at 110 CC and allowed to stir for 16 h. 2362 The mixture was then allowed to cool and ice-cold water was added. The precipitate was 2363 collected by filtration and washed with EtzO and dried under vacuum to afford Intermediate 2364 33 (4 g, 38%) as a pale yellow solid. 1H NMR: (DMSO-d6) 8 8.48-8.49 (m, 1H), 8.00-8.04 2365 (m, 1H), 7.87 (d, J: 8.3 Hz, 1H), 7.79 (s, 1H), 7.65 (br s, 2H), 7.33-7.36 (m, 1H), 4.22 (q, .1 2366 = 7.0 Hz, 2H), 1.28 (t, J: 7.0 Hz, 3H); MS: 233 [M + H]+; TLC: 15% EtOAc in hexane: Rf: 2367 0.50. 2368 Example 36 - Preparation ound 2] CHE: NaH DMF N N N\ H33 °c to RT — 0 2369 CE 2370 Intermediate 33 Compound 21 2371 2372 [0276] Sodium hydride (603 mg, 15.1 mmol, 1 eq, 60% in mineral oil) was added to a 2373 solution of ediate 33 (3.5 g, 15.1 mmol) in DMF (300 mL) at 0°C. After 30 minutes, a 2374 solution of 4-fiuorobenzyl bromide (2.85 g, 15.1 mmol, 1 eq) in DMF (50 mL) was added 2375 and the resulting mixture was allowed to warm to RT. After 5 h, the reaction mixture was 2376 d with water (100 mL) and extracted with EtOAc (3 X 100 mL). The combined organic 2377 layers were washed with water (5 X 50 mL), brine (50 mL), dried over , filtered and 2378 concentrated in vacuo. The crude material was purified by column chromatography over 2379 silica gel (100-200 mesh) by using a nt e of 0-5% EtOAc-hexane as the eluent to 2380 afford a partially pure product. The material was then recrystallized from EtzO and pentane to 2381 afford Compound 21 (2.8 g, 55%) as a pale yellow solid. 1H NMR: (DMSO-d6) 8 9.50 (t, J = 2382 6.6 Hz, 1H), 8.45-8.46 (m, 1H), 8.00-8.05 (m, 1H), .89 (m, 2H), 7.24-7.38 (m, 3H), 2383 7.11 (t, J: 8.8 Hz, 2H), 4.88 (d, J: 6.6 Hz, 2H), 4.17 (q, J: 7.0 Hz, 2H), 1.24 (t, J: 7.0 Hz, 2384 3H); MS: 341 [M + H]+; MP: 99-100 oC; TLC: 15% EtOAc in hexane: Rf: 0.40. 2385 e 3 7 — cornealpermeability ofcompounds 4 and 22 2386 [0277] In this example, trans-comeal permeability of compounds are measured in-vitro 2387 across excised rabbit corneas. Excised rabbit cornea are acquired from Pel-Freez and 2388 delivered in DMEM culture media on ice overnight. The test apparatus is a curved 9 mm 2389 Franz chamber (Permegear) suitable for rabbit eyes. Test compound is ed in PBS 2390 buffer. The compound solution is added to the donor chamber of the Franz chamber and the 2391 entire apparatus placed in an incubator at 37°C for four hours. During incubation and at every 2392 subsequent hour, a sample is removed from the receiver chamber and analyzed by HPLC 2393 (Shimadzu Prominence) using a C18 column (Phenomenex 00FE0) in reverse phase 2394 with acetonitrile in water. The apparent permeability coefficient Papp (cm/s) is calculated as 3;. mil“.. x‘ 2395 .~§ ii: - 1535.;1.3% 2396 where dM/dt is the flux (nmol/s) across the cell layers or cornea, A is the exposed surface 2397 area (cm2) of the insert membrane of rabbit cornea, and C0 is the initial drug concentration 2398 (uM) in the donor compartment. 2399 [0278] In Table E following, measured trans-comeal permeability Papp in units of cm/s is 2400 presented for example compounds. 2401 Table E 2402 2403 Example 38 — Pharmacokinetz'cs in mice 2404 [0279] In this example, pharmacokinetics in mice is ted for series of example 2405 compounds #4, #23, #24, #25, #26. Each nd is administered intravenously (i.v.) as a 2406 single dose via tail vein or orally (p.o.) as a single dose via c gavage to male CD-1 mice 2407 of nominal weights n 20 g and 26 g. Nominal doses are 1 mg/kg and 5 mg/kg for iv. 2408 and p.o., respectively. In some examples (dose type A), both p.o. and iv. doses are prepared 2409 by dissolving the test compound in 5% dimethyl acetamide and diluted in tetraethylene glycol 1 09 2410 for a final concentration of 0.25 mg/mL. In other es (dose type B), i.v. doses are 2411 prepared by dissolving test compounds in 20% dimethyl acetamide, 40% polyethylene glycol 2412 300 and 40% phosphate buffered saline, and p.o. doses are prepared by dissolving test 2413 compounds in carboxymethyl cellulose suspension (1% by weight) in water and 2.5 % 2414 dimethyl acetamide. 2415 [0280] Animals are housed in standard holding cages with food and water available ad 2416 libitum except for animals used for p.o. dosing which are fasted overnight prior to dosing. 2417 Samples are taken in triplicate via cardiac puncture at times prior to dosing and at 0.083 (i.v. 2418 only), 0.25, 0.5, l, 2, 4, 8, and 24 hours after administration. Plasma is obtained by filge 2419 and stored frozen until analyzed by LC-MS/MS using a Shimadzu VP System HPLC coupled 2420 to a Applied tems MDS SCIEX API 3000 triple quadrapole MS. Assay results are 2421 calibrated using reference samples prepared in a range between 1.5 and 5000 ng/mL. 2422 [0281] cokinetic parameters are calculated from mean concentration values using a 2423 non-compartmental analysis as described in the following and as nt to those of 2424 ordinary skill in the art. Half-lives (tl/g) and elimination rate constants (k) are ined by 2425 log linear regression using equal weighting on the last three finite sample time points. 2426 Concentration at time zero (Co) for the i.v. data is ished by the extrapolation of log 2427 linear regression using equal weighting on the first three sample time points. Area under the 2428 curve (AUC) values are ated using linear trapezoidal integration. Systemic clearance 2429 (CL) is calculated as the ratio of dosage and AUC. The apparent volume of distribution (Vd) 2430 is calculated as the ratio of CL and 9». Percent oral bioavailability (%F) is determined from the 2431 ratio of iv. and p.o. AUC values weighted by dosage. 2432 [0282] In Table F following resulting pharmacokinetic ters are listed for five 2433 example compounds, rounded to the nearest significant digit. 2434 Table F Cmpd No 4 23 Dose type A A 1.V. 121/2 (11) 1 0.5 i.v. C0 (ng/ml) 300 400 i.v. AUC (h-ng/ml) 100 100 i.V. Vd (ml/kg) 10000 5000 iv. CL(n11/kg/h) 10000 8000 p.0. t1/2 (h) 6 0.6 p.o. AUC (h-ng/ml) 300 80 %F 60 10 2435 2436 [0283] The contents of all references, patents, and published ations cited herein are 2437 hereby incorporated by reference in their entirety and for all es. 2438 [0284] While the invention has been described in detail with reference to certain preferred 2439 embodiments thereof, it will be understood that modifications and variations are Within the 2440 spirit and scope of that which is described and claimed. 2441

Claims (38)

WHAT IS CLAIMED IS:
1. Use of a compound having a structure according to Formula (V): R5 L5 O N L1 R2 (V) or pharmaceutically acceptable salt, ester, or solvate thereof, or a pharmaceutical composition comprising said compound, for the manufacture of a medicament for the ent of a thrombotic disorder, a kallikrein-related disorder, or a disease or disorder responsive to inhibition of thrombin and/or kallikrein in a subject, n the kallikrein-related disorder comprises at least one of an ophthalmic disease, a thrombotic disease, a fibrinolytic disease, a type of cancer, an inflammatory condition, or a dermatological condition; L1 is -NR7-; L2 is –C(=O)-; L5 is a bond or substituted or unsubstituted alkylene, provided that if L5 is a bond, R5 is hydrogen; R1 is tuted alkyl; R2 is substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkenyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heterocycloalkenyl, substituted or tituted aryl, substituted or unsubstituted fused ring aryl, or tuted or unsubstituted aryl; R5 is hydrogen, halogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkenyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heterocycloalkenyl, substituted or tituted aryl, substituted or unsubstituted fused ring aryl, or substituted or unsubstituted heteroaryl; and R7 is hydrogen, or substituted or unsubstituted alkyl; and wherein the term “alkyl” means a straight or branched chain, which can be fully saturated, or mono- or polyunsaturated; and wherein the term oalkyl” means a ht or branched chain, consisting of at least one carbon atom and at least one heteroatom selected from the group consisting of O, N, P, Si, and S; and wherein any substituted group may be substituted with one or more tuent group selected from the following moieties: (A) -OH, -NH2, -SH, -CN, -CF3, -NO2, oxo, n, -COOH, unsubstituted C1-C24 alkyl, unsubstituted 2- to 20 ed heteroalkyl, unsubstituted C3-C8 cycloalkyl, unsubstituted C3-C8 cycloalkenyl, unsubstituted 4- to ered heterocycloalkyl, unsubstituted 4- to 8-membered heterocycloalkenyl, unsubstituted aryl, unsubstituted heteroaryl, and (B) C1-C24 alkyl, 2- to 20 ed heteroalkyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, 4- to 8-membered heterocycloalkyl, 4- to 8-membered heterocycloalkenyl, aryl, and heteroaryl, substituted with at least one tuent selected from: (i) oxo, -OH, -NH2, -SH, -CN, -CF3, -NO2, halogen, -COOH, unsubstituted C1- C24 alkyl, unsubstituted 2- to 20 membered heteroalkyl, unsubstituted C3-C8 cycloalkyl, unsubstituted C3-C8 cycloalkenyl, unsubstituted 4- to 8-membered heterocycloalkyl, unsubstituted 4- to 8-membered heterocycloalkenyl, unsubstituted aryl, unsubstituted heteroaryl, and (ii) C1-C24 alkyl, heteroalkyl, C3-C8 cycloalkyl, C3-C8 cycloalkenyl, 4- to 8- membered heterocycloalkyl, 4- to 8-membered heterocycloalkenyl, aryl, and heteroaryl, substituted with at least one substituent selected from: (a) oxo, -OH, -NH2, -SH, -CN, -CF3, -NO2, halogen, -COOH, unsubstituted C1-C24 alkyl, unsubstituted 2- to 20 membered heteroalkyl, unsubstituted C3-C8 cycloalkyl, unsubstituted C3-C8 cycloalkenyl, unsubstituted 4- to 8-membered heterocycloalkyl, tituted 4- to 8-membered heterocycloalkenyl, unsubstituted aryl, unsubstituted heteroaryl, and (b) C1-C24 alkyl, 2- to 20- membered heteroalkyl, C3-C8 cycloalkyl, C3- C8 cycloalkenyl, 4- to 8-membered heterocycloalkyl, 4- to 8-membered cycloalkenyl, aryl, or heteroaryl, substituted with at least one substituent selected from: oxo, -OH, -NH2, -SH, -CN, -CF3, -NO2, halogen, -COOH, unsubstituted C1-C24 alkyl, unsubstituted 2- to 20 membered heteroalkyl, unsubstituted C3-C8 cycloalkyl, tituted C3-C8 cycloalkenyl, unsubstituted 4- to 8-membered heterocycloalkyl, unsubstituted 4- to 8-membered heterocycloalkenyl, unsubstituted aryl, and unsubstituted heteroaryl; and provided the compound is not selected from: Compound 4, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(furanyl)carbonyl]- azolyl)-1,2-dihydropyridinone; Compound 23, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(furan yl)carbonyl]-1H-pyrazolyl)methyl-1,2-dihydropyridinone; Compound 25, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(furan yl)carbonyl]-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridinone; Compound 26, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(furan yl)carbonyl]-1H-pyrazolyl)(furanylmethyl)-1,2-dihydropyridinone; Compound 73, 3-(1-benzoyl[(5-chlorothiophenyl)methyl]amino-1H-pyrazol yl)methyl-1,2-dihydropyridinone; and Compound 132, [(5-chlorothiophenyl)methyl]amino}[(2- methoxyphenyl)carbonyl]-1H-pyrazolyl)-1,2-dihydropyridinone.
2. The use according to claim 1, wherein L2 is -C(=O)-, and R2 is substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted cycloalkenyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkenyl, tuted or unsubstituted aryl, substituted or unsubstituted fused ring aryl, or substituted or tituted heteroaryl.
3. The use according to claim 1, wherein said disease or disorder is at least one of a thrombotic disorder, a disease or disorder involving a blood clot us, and a e or disorder which can lead to formation of a blood clot thrombus.
4. The use according to claim 3, wherein said thrombotic disorder ses at least one of acute coronary syndrome, thromboembolism, and thrombosis.
5. The use according to claim 4, wherein the thromboembolism comprises at least one of venous oembolism, arterial thromboembolism, and genic thromboembolism.
6. The use according to claim 3, wherein said thrombotic disorder involves dysfunctional coagulation or disseminated intravascular coagulation.
7. The use according to claim 1, wherein said disease or disorder comprises at least one of fibrosis, Alzheimer’s Disease, multiple sclerosis, pain, cancer, and inflammation.
8. The use according to claim 4, wherein the thrombosis is arterial thrombosis.
9. The use ing to claim 1, wherein said ophthalmic e is diabetic macular edema, age-related macular degeneration, or diabetic pathy.
10. The use according to claim 1, wherein said type of cancer is al-, testicular-, or non-small-cell lung adenocarcinoma.
11. The use according to claim 1, wherein said inflammatory condition is sepsis, inflammatory bowel disease, ic inflammatory response syndrome, hereditary angioedema, or rheumatoid arthritis.
12. The use according to claim 1, wherein said dermatological ion is atopic dermatitis, psoriasis, or Netherton Syndrome.
13. The use according to claim 1, wherein said compound acts by inhibiting kallikrein.
14. The use according to claim 13, n said compound acts by inhibiting tissue kallikrein.
15. The use ing to claim 13, wherein said compound acts by inhibiting plasma kallikrein.
16. The use according to claim 1, wherein said kallikrein-related disorder is an ophthalmic disease, and wherein the medicament is formulated for oral administration, is in the form of an ophthalmic composition for topical application to the eye, or is in the form of an ophthalmic composition for intravitreal injection.
17. The use according to claim 16, wherein the ophthalmic composition is in the form of eye drops.
18. The use according to claim 16, wherein said compound acts by reducing l vascular permeability.
19. The use according to claim 1, wherein said kallikrein-related disorder is an inflammatory condition and the medicament is formulated for oral administration.
20. The use according to claim 1, wherein said compound is selected from the group consisting of: Compound 5, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(furanyl)carbonyl]- 1H-pyrazolyl)(thiophenylmethyl)-1,2-dihydropyridinone; Compound 49, 1-benzyl(5-{[(5-chlorothiophenyl)methyl]amino}[(furan yl)carbonyl]-1H-pyrazolyl)-1,2-dihydropyridinone; Compound 52, 1-benzyl(5-{[(5-chlorothiophenyl)methyl]amino}[(thiophen- 3-yl)carbonyl]-1H-pyrazolyl)-1,2-dihydropyridinone; nd 62, 3-(1-benzoyl{[(5-chlorothiophenyl)methyl]amino}-1H-pyrazol- 1-(2-methoxyethyl)-1,2-dihydropyridinone; Compound 81, 3-(5-{[(5-chlorothiophenyl)methyl]amino}(2,2- dimethylpropanoyl)-1H-pyrazolyl)(pyridazinylmethyl)-1,2-dihydropyridinone; Compound 94, 3-(5-{[(5-chlorothiophenyl)methyl]amino}(3-hydroxy-2,2- dimethylpropanoyl)-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridinone; Compound 100, 3-(5-{[(5-chlorothiophenyl)methyl]amino}(3-methoxy-2,2- dimethylpropanoyl)-1H-pyrazolyl)(pyridazinylmethyl)-1,2-dihydropyridinone; Compound 104, 3-(5-{[(5-chlorothiophenyl)methyl]amino}(3-methoxy-2,2- dimethylpropanoyl)-1H-pyrazolyl)(pyrimidinylmethyl)-1,2-dihydropyridinone; Compound 109, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2,3-dihydro-1,4- benzodioxinyl)carbonyl]-1H-pyrazolyl)(2-methoxyethyl)-1,2-dihydropyridinone; Compound 113, [(5-chlorothiophenyl)methyl]amino}[(2,3-dihydro-1,4- benzodioxinyl)carbonyl]-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridin one; Compound 116, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2,3-dihydro-1,4- benzodioxinyl)carbonyl]-1H-pyrazolyl)(pyrimidinylmethyl)-1,2-dihydropyridin- 2-one; Compound 122, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2,4- dimethoxyphenyl)carbonyl]-1H-pyrazolyl)(2-methoxyethyl)-1,2-dihydropyridinone; Compound 125, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2,4- dimethoxyphenyl)carbonyl]-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridin one; nd 126, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2,4- dimethoxyphenyl)carbonyl]-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridin one; Compound 131, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2,4- dimethoxyphenyl)carbonyl]-1H-pyrazolyl)[2-(morpholinyl)ethyl]-1,2- dihydropyridinone; Compound 138, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2- methoxyphenyl)carbonyl]-1H-pyrazolyl)(pyridazinylmethyl)-1,2-dihydropyridin one; Compound 140, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2- methoxyphenyl)carbonyl]-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridin one; Compound 145, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2- methoxyphenyl)carbonyl]-1H-pyrazolyl)[2-(morpholinyl)oxoethyl]-1,2- dihydropyridinone; Compound 149, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(4-methyloxan yl)carbonyl]-1H-pyrazolyl)(2-methoxyethyl)-1,2-dihydropyridinone; Compound 160, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(furan yl)carbonyl]-1H-pyrazolyl)(2-methoxyethyl)-1,2-dihydropyridinone; Compound 182, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(thiophen yl)carbonyl]-1H-pyrazolyl)(2-methoxyethyl)-1,2-dihydropyridinone; Compound 194, [(5-chlorothiophenyl)methyl]amino}[(thiophen yl)carbonyl]-1H-pyrazolyl)(2-methoxyethyl)-1,2-dihydropyridinone; Compound 196, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(thiophen bonyl]-1H-pyrazolyl)(furanylmethyl)-1,2-dihydropyridinone; Compound 198, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(thiophen yl)carbonyl]-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridinone; Compound 201, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(thiophen yl)carbonyl]-1H-pyrazolyl)(pyrimidinylmethyl)-1,2-dihydropyridinone; Compound 203, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(thiophen yl)carbonyl]-1H-pyrazolyl)(thiophenylmethyl)-1,2-dihydropyridinone; Compound 208, 3-(5-{[(5-chlorothiophenyl)methyl]amino}{[4-(morpholin nyl]carbonyl}-1H-pyrazolyl)(2-methoxyethyl)-1,2-dihydropyridinone; Compound 213, 3-(5-{[(5-chlorothiophenyl)methyl]amino}{[4-(morpholin yl)phenyl]carbonyl}-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridinone; Compound 214, 3-(5-{[(5-chlorothiophenyl)methyl]amino}{[4-(morpholin yl)phenyl]carbonyl}-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridinone; Compound 218, [(5-chlorothiophenyl)methyl]amino}{[4-(morpholin yl)phenyl]carbonyl}-1H-pyrazolyl)[2-(morpholinyl)ethyl]-1,2-dihydropyridinone; Compound 238, 3-[5-(benzylamino)[(furanyl)carbonyl]-1H-pyrazolyl] (pyridinylmethyl)-1,2-dihydropyridinone; Compound 242, 3-{1-[(2-chlorophenyl)carbonyl]{[(5-chlorothiophen yl)methyl]amino}-1H-pyrazolyl}(2-methoxyethyl)-1,2-dihydropyridinone; Compound 243, 3-{1-[(2-chlorophenyl)carbonyl]{[(5-chlorothiophen yl)methyl]amino}-1H-pyrazolyl}(furanylmethyl)-1,2-dihydropyridinone; Compound 244, (2-chlorophenyl)carbonyl]{[(5-chlorothiophen yl)methyl]amino}-1H-pyrazolyl}(furanylmethyl)-1,2-dihydropyridinone; Compound 247, 3-{1-[(2-chlorophenyl)carbonyl]{[(5-chlorothiophen yl)methyl]amino}-1H-pyrazolyl}(pyridinylmethyl)-1,2-dihydropyridinone; Compound 248, 3-{1-[(2-chlorophenyl)carbonyl]{[(5-chlorothiophen yl)methyl]amino}-1H-pyrazolyl}(pyridinylmethyl)-1,2-dihydropyridinone; and nd 265, {1-[(5-{[(5-chlorothiophenyl)methyl]amino}(1-methyloxo- 1,2-dihydropyridinyl)-1H-pyrazolyl)carbonyl]cyclopropyl}methyl (hydroxymethyl)cyclopropanecarboxylate.
21. Use of a compound selected from the group consisting of: Compound 4, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(furanyl)carbonyl]- 1H-pyrazolyl)-1,2-dihydropyridinone; nd 23, [(5-chlorothiophenyl)methyl]amino}[(furan yl)carbonyl]-1H-pyrazolyl)methyl-1,2-dihydropyridinone; Compound 25, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(furan yl)carbonyl]-1H-pyrazolyl)(pyridinylmethyl)-1,2-dihydropyridinone; Compound 26, [(5-chlorothiophenyl)methyl]amino}[(furan yl)carbonyl]-1H-pyrazolyl)(furanylmethyl)-1,2-dihydropyridinone; and Compound 132, 3-(5-{[(5-chlorothiophenyl)methyl]amino}[(2- methoxyphenyl)carbonyl]-1H-pyrazolyl)-1,2-dihydropyridinone, or ceutically acceptable salt, ester, or solvate thereof, or a pharmaceutical ition comprising said compound, for the manufacture of a medicament for the treatment of a thrombotic disorder, a kallikrein-related disorder, or a disease or disorder responsive to inhibition of in and/or kallikrein in a subject, wherein the kallikrein-related disorder comprises at least one of an ophthalmic disease, a otic disease, a fibrinolytic disease, a type of cancer, an inflammatory condition, or a dermatological condition.
22. The use according to claim 21, wherein said disease or disorder is at least one of a thrombotic disorder, a disease or disorder involving a blood clot thrombus, and a disease or disorder which can lead to formation of a blood clot thrombus.
23. The use according to claim 22, wherein said thrombotic disorder comprises at least one of acute coronary syndrome, thromboembolism, and thrombosis.
24. The use according to claim 23, wherein the thromboembolism comprises at least one of venous thromboembolism, arterial oembolism, and cardiogenic thromboembolism.
25. The use according to claim 22, wherein said thrombotic disorder involves dysfunctional coagulation or disseminated intravascular coagulation.
26. The use according to claim 21, wherein said disease or disorder comprises at least one of fibrosis, Alzheimer’s Disease, multiple sis, pain, , and inflammation.
27. The use according to claim 23, n the osis is arterial thrombosis.
28. The use according to claim 21, wherein said lmic disease is diabetic r edema, age-related macular degeneration, or diabetic retinopathy.
29. The use according to claim 21, wherein said type of cancer is cervical-, testicular-, or non-small-cell lung adenocarcinoma.
30. The use according to claim 21, n said inflammatory condition is sepsis, inflammatory bowel disease, systemic inflammatory response syndrome, hereditary angioedema, or rheumatoid arthritis.
31. The use according to claim 21, wherein said dermatological condition is atopic dermatitis, psoriasis, or Netherton Syndrome.
32. The use according to claim 21, wherein said compound acts by inhibiting kallikrein.
33. The use according to claim 32, wherein said compound acts by inhibiting tissue kallikrein.
34. The use according to claim 32, wherein said compound acts by inhibiting plasma kallikrein.
35. The use according to claim 21, wherein said kallikrein-related disorder is an ophthalmic disease, and wherein the medicament is formulated for oral administration, is in the form of an ophthalmic composition for topical application to the eye, or is in the form of an ophthalmic composition for intravitreal injection.
36. The use according to claim 35, wherein the ophthalmic composition is in the form of eye drops.
37. The use according to claim 35, wherein said compound acts by reducing retinal ar permeability.
38. The use according to claim 21, wherein said kallikrein-related disorder is an inflammatory ion and the medicament is formulated for oral administration.
NZ749214A 2013-03-15 2014-03-17 Multisubstituted aromatic compounds as serine protease inhibitors NZ749214B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361789358P 2013-03-15 2013-03-15
US61/789,358 2013-03-15
NZ711064A NZ711064A (en) 2013-03-15 2014-03-17 Pyridone-substituted pyrazolyl compounds as serine protease inhibitors

Publications (2)

Publication Number Publication Date
NZ749214A NZ749214A (en) 2021-04-30
NZ749214B2 true NZ749214B2 (en) 2021-08-03

Family

ID=

Similar Documents

Publication Publication Date Title
AU2017225000B2 (en) Multisubstituted aromatic compounds as serine protease inhibitors
US10653674B2 (en) Multisubstituted aromatic compounds as inhibitors of thrombin
AU2015317522A1 (en) Pyrazolyl-substituted pyridone compounds as serine protease inhibitors
US9951025B2 (en) Halogenopyrazoles as inhibitors of thrombin
NZ749214B2 (en) Multisubstituted aromatic compounds as serine protease inhibitors