Identification and Preliminary SAR Analysis of Novel Type-I Inhibitors

Nov 30, 2015 - Several large-molecule inhibitors targeting the ANG/TIE-2 axis have recently entered clinical phase for the therapy of various solid tu...
0 downloads 15 Views 2MB Size
Subscriber access provided by UNIV OF NEBRASKA - LINCOLN

Article

Identification and Preliminary SAR Analysis of Novel Type-I Inhibitors of TIE-2 via Structure-Based Virtual Screening and Biological Evaluation in in vitro Models Peichen Pan, Sheng Tian, Huiyong Sun, Xiaotian Kong, Wenfang Zhou, Dan Li, Youyong Li, and Tingjun Hou J. Chem. Inf. Model., Just Accepted Manuscript • DOI: 10.1021/acs.jcim.5b00576 • Publication Date (Web): 30 Nov 2015 Downloaded from http://pubs.acs.org on December 8, 2015

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Journal of Chemical Information and Modeling is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

1 2

Identification and Preliminary SAR Analysis of Novel Type-I

3

Inhibitors of TIE-2 via Structure-Based Virtual Screening and

4

Biological Evaluation in in vitro Models

5 6

Peichen Pana, Sheng Tianb, Huiyong Suna, Xiaotian Kongb, Wenfang Zhoua, Dan Lia,

7

Youyong Lic, Tingjun Houa,b

8 9 a

10

College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China

11 b

12

State Key Lab of CAD&CG, Zhejiang University, Hangzhou, Zhejiang 310058, China

13 14

c

Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.

15 16 17 18

Corresponding authors:

19

Tingjun Hou

20

E-mail: [email protected]

or

[email protected]

21 22 23 24 25 26 27

Keywords: :Angiogenesis, TIE-2, Type-I inhibitor, Structure-based virtual screening,

28

Substructure search

29

1

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

30

For Table of Contents Use Only

31

32 33 34

2

ACS Paragon Plus Environment

Page 2 of 32

Page 3 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

35

Abstract

36

Angiopoietin (ANG) ligands and their downstream TIE receptors have been validated

37

as the second vascular signaling system involving vessel remodeling and maturation.

38

Among them, ANG/TIE-2 signaling pathway is involved in numerous life-threatening

39

diseases and has become an attractive potential therapeutic target. Several

40

large-molecule inhibitors targeting ANG/TIE-2 axis have recently entered clinical

41

phase for the therapy of various solid tumors, but selective small-molecule inhibitors

42

of TIE-2 is still quite limited. In the present work, structure-based virtual screening

43

was performed to search for type-I inhibitors of TIE-2. Of the only 41 compounds

44

selected by our strategy, 8 molecules with the concentration of 25 µg/mL exhibit over

45

50% inhibitory rate against TIE-2 in in vitro enzymatic activity assay, and the IC50

46

values of 2 hits are lower than 1 µM. Further optimization and SAR analysis based on

47

compound TP-S1-30 and 31 were carried out by using substructure searching strategy,

48

leading to the discovery of several sub-100nM inhibitors. Among them, the most

49

potent compound, TP-S1-68, showed an inhibitory IC50 of 0.149 µM. These novel

50

inhibitors of TIE-2 discovered in this study and the analogs of the active core

51

scaffolds can serve as the starting points for further drug development.

52 53

3

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

54

Introduction

55

Blood supply plays a critical role in the progress of tumor growth. Blocking the

56

formation of vascular system is believed to be an effective way to suppress tumor

57

growth and metastasis.1 Vessels can grow in a number of different ways. Early

58

formation of blood vessels occurs by a process known as vasculogenesis, where

59

endothelial cells differentiate, proliferate and then assemble to a primitive vascular

60

network. This primary vascular plexus is modified and extended by another process

61

called angiogenesis, which includes both sprouting and non-sprouting angiogenesis.

62

This emerging capillary plexus is soon remodeled to resemble a mature vasculature

63

through both pruning and vessel enlargement.2 Further maturation of blood vessels is

64

then accomplished where endothelial cells integrate tightly with smooth muscle cells,

65

pericytes and surrounding matrix.1, 3

66

Complex signaling pathways and a high degree of co-ordination among various cell

67

types are involved in the formation of blood vessels. In the past decade, great progress

68

has been made in identifying and characterizing physiological regulators of blood

69

vessel growth. The members of the vascular endothelial cell growth factor (VEGF)

70

receptor family, which includes Flt-1 (VEGF-R1), Flk-1/KDR (VEGF-R2), and Flt-4

71

(VEGF-R3), are potent mediators for angiogenesis.4, 5 A number of kinase inhibitors

72

targeting VEGF signaling and anti-VEGF antibodies have entered clinical trials,

73

among which sunitinib (Sutent), sorafenib (Nexavar), etc. have been approved by the

74

FDA for the treatment of various solid tumors.4, 6, 7 Although clinical outcomes were

75

often encouraging, patients always failed to respond or became resistant to the

76

currently available anti-angiogenic drugs.8-12

77

Of the other factors involved in angiogenesis, TIE-2/TEK (tyrosine kinase with

78

immunoglobulin and epidermal growth factor homology domains-2), a tyrosine kinase

79

that is expressed principally on vascular endothelium, was found to play a critical role

80

in the stabilization, maturation, remodeling and hierarchical organization of

81

preliminary vessels.13-15 Interference with the ANG/TIE-2 signaling pathways has

82

been shown to block or attenuate tumor-induced angiogenesis and suppress primary 4

ACS Paragon Plus Environment

Page 4 of 32

Page 5 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

83

and metastatic tumor growth in a variety of xenograft models.16-19 Recently, it has

84

become clear that combination of both first (VEGF signaling) and second generation

85

(ANG/TIE-2 signaling) agents can lead to better therapeutic outcomes, which is

86

superior to treatments with either agent alone.16, 20 Therefore, ANG/TIE-2 axis has

87

become a very promising anti-angiogenesis target for the therapy of various tumors.

88

Pharmaceutical companies and some academic institutions have been involved in

89

developing ANG/TIE-2 inhibitors in recent years.21-26 Several large molecular

90

antibodies

91

REGN-910 and AMG-780)27-29 targeting ANG ligands (ANG-1 and/or ANG-2) and at

92

least two small molecule candidates of TIE-2, namely CEP-1198121 (Phase I,

93

Cephalon, Inc.) and ARRY-61424 (Phase I, Array Biopharma. Inc.), have been pushed

94

into the clinical phase.

(e.g.

AMG-386/Trebananib,

MEDI-3617,

CVX-060/PF04856884,

95

With the explosive increasing number of synthesized compounds from various

96

chemical libraries, the demand for efficient strategies to identify novel and potent

97

small inhibitors of a certain drug target is also growing. Structure-based virtual

98

screening (VS) method is being widely utilized in hit identification and plays an

99

important role in the preliminary stage of drug discovery.30-36 In this work, a

100

docking-based VS model for discovering type-I inhibitors of TIE-2 was developed

101

and evaluated by in vitro biological inhibitory activity experiments. Several active

102

compounds were successfully identified from the first-round virtual screening

103

campaign, where the inhibitory activities (IC50) of 2 hit compounds were lower than 1

104

µM. Subsequently, preliminary optimization based on the core structures of these

105

compounds were performed using substructure search methodology, and several

106

sub-100nM inhibitors were then discovered. According to the results of both

107

experimental tests and molecular modeling simulations, the structure-activity

108

relationships (SAR) of the inhibitors were analyzed. The active compounds identified

109

in this work and the detailed binding mechanism of these inhibitors to TIE-2 can

110

further guide the design and discovery of more potent inhibitors of TIE-2, which

111

would finally benefit to the targeted therapy of associated fatal diseases.

112 5

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

113

Materials and Methods

114

Initial Structures and Preparations for Molecular Docking. The DFG-in crystal

115

structures of TIE-2/inhibitor complexes used in this study were obtained from RCSB

116

Protein Data Bank,37 including 3L8P21 and 2OO838. The preparations of the protein

117

structures were accomplished with the Protein Preparation wizard in Schrödinger

118

9.0.39 The prediction accuracy of docking may be influenced when water molecules

119

(especially the active site ones) are included in docking simulations, and the positions

120

of water molecules in the binding of different inhibitors may also significantly vary.

121

Thus, all crystallographic water molecules were deleted, and hydrogen atoms and

122

partial charges were added to the proteins, and a restrained partial minimization

123

terminated when the root-mean-square deviation (RMSD) reached a maximum value

124

of 0.3 Å.

125

The 3D structures of known TIE-2 inhibitors were obtained from BindingDB

126

database,40 and then the type-II inhibitors and those with poor inhibitory activity

127

(IC50 > 100 µM) were removed.41 The final inhibitor decoy set was generated by

128

extracting 100 structurally diverse type-I inhibitors from the cleared database above

129

using Find Diverse Molecule module in Discovery Studio 2.5.42 A library of

130

approximately 240,000 screening compounds was downloaded from the Specs

131

chemical library, and the non-inhibitor decoy set of 10,000 compounds for

132

establishment and validation of screening models was randomly extracted from the

133

Specs library using Discovery Studio 2.5 package.42 All the compounds used for

134

docking were minimized with Macromodel in Schrödinger using the OPLS-2005

135

force field. Ligand preparations were carried out with LigPrep module in Schrödinger,

136

and correct protonated states were generated at pH = 7.0 ± 2.0. Default settings were

137

used for the other parameters.

138

Docking Study. All molecular docking calculations were carried out using the Glide

139

module in Schrödinger. The grid boxes of proteins for docking were generated and

140

centered on the inhibitors in the binding pockets, and the scaling factors for van der

141

Waals radii was set to 1.0. The maximum partial atomic charge was set to 0.25. 6

ACS Paragon Plus Environment

Page 6 of 32

Page 7 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

142

Subsequently, the studied compounds were docked into the binding site of TIE-2 with

143

different precision modes, which includes HTVS (high throughput virtual screening),

144

SP (standard precision) and/or XP (extra precision). The Epik state penalties to

145

docking score were added.

146

Virtual Screening Workflow. Before screening the Specs chemical library, validation

147

of the performance of molecular docking based on two reported DFG-in crystal

148

structures of TIE-2 in complex with type-I inhibitors (PDB entry: 2WQB and 3L8P)43,

149

44

150

non-inhibitor decoy set randomly extracted from Specs were docked into the two

151

TIE-2 crystal structures using three different scoring modes: HTVS, SP and XP. By

152

comparing the accuracy of Glide docking in distinguishing inhibitors from

153

non-inhibitors based on two different TIE-2 structures, the crystal structure of 3L8P

154

with better prediction accuracy was finally chosen for the following screening

155

campaign. A hierarchical scheme was applied in the virtual screening of TIE-2

156

inhibitors (Figure 1). In the first step, all the screened compounds were docked into

157

the crystal structure of 3L8P, and the binding energies were scored and ranked by

158

Glide SP mode. Subsequently, the top ranked 50,000 molecules were saved and

159

submitted to the following Glide XP docking assessment. According to the ADMET

160

properties of the compounds predicted by ACD/ADME package,45 the 1,000 top

161

ranked molecules from Glide XP docking were then filtered by several criteria, and

162

the compounds in accordance with either criteria were removed: (1) logP/logD

163

(pH=7.0) > 6.0; (2) violation of Lipinski's rule of five ≥ 2;46 (3) violation of Opera's

164

rules of drug-likeness ≥ 4;47 (4) functional groups with toxic, reactive, or otherwise

165

undesirable moieties defined by the REOS rules48. In order to maximize the chemical

166

diversity of the selected compounds for experimental testing, the remaining filtered

167

compounds were clustered into 50 clusters on the basis of the Tanimoto distance

168

predicted from the FCFP_4 fingerprints using Find Diverse Molecule module in

169

Discovery Studio 2.5.42 50 compounds were obtained by selecting the molecule with

170

the lowest docking scores in each cluster. However nine compounds were not

was first carried out. The molecules of both the inhibitor decoy set of TIE-2 and the

7

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

171

available for purchasing from chemical vendor, Specs. So 41 compounds were

172

eventually purchased and tested experimentally for their ability to inhibit the

173

enzymatic activity of TIE-2. Furthermore, hit optimization and initial SAR analysis

174

based on substructure searching of the active core structures was also carried out by

175

querying the entire Specs database for structures with the same core scaffold. Then a

176

second round experimental screening was also accomplished.

177

MD Simulations and MM/GBSA Calculations. Molecular dynamics (MD)

178

simulations and molecular mechanics/generalized born surface area (MM/GBSA)

179

calculations were performed in AMBER14.49 Atomic partial charges of inhibitors

180

were generated by fitting the electrostatic potentials calculated at the HF/6-31G* level

181

using the RESP fitting technique after the small molecules were optimized by

182

semi-empirical AM1 method in Gaussian09.50, 51 The general AMBER force field

183

(gaff)52 was used for the inhibitors, and the ff99SB force field for the proteins.53

184

TIP3P water molecules with a distance of 10 Å extended from any solute atoms were

185

applied into the system. Particle Mesh Ewald (PME) method54 was employed to

186

handle the long-range electrostatics.

187

In the first stage, 500 cycles of steepest descent and 500 cycles of conjugate

188

gradient minimizations were carried out with 50 kcal/mol/Å2 restraint on the backbone

189

carbons. Then, 1000 cycles of steepest descent and 4000 cycles of conjugate gradient

190

minimizations without any restraint were carried out. Subsequently, each system was

191

heated up from 0 to 300 K in 50 ps, and 5 ns NPT (P = 1 atm and T = 300 K) MD

192

simulations were then performed. The SHAKE algorithm was applied to constrain all

193

bonds involving hydrogen atoms.55 The time step was set to 2.0 fs, and the

194

coordinates were saved every 10 ps.

195

The binding free energy (∆Gbind) of each inhibitor/TIE-2 complex was calculated

196

using the Molecular Mechanics/Generalized Born Solvent Area (MM/GBSA)

197

methodology.56,

198

dielectric constant (εin) was set to 1. The polar part of desolvation (∆GGB) was

199

computed with the modified GB model developed by Onufriev et al. (referred as

57

The exterior dielectric constant was set to 80, and the solute

8

ACS Paragon Plus Environment

Page 8 of 32

Page 9 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

200

igb=2 in Amber),58 and the non-polar part of desolvation (∆GSA) was determined

201

based on the solvent accessible surface area (SASA) computed by the LCPO

202

algorithm:59 ∆GSA= 0.0072 × ∆SASA. The change of the conformational entropy

203

(-T∆S) was not considered due to high computational cost and low prediction

204

accuracy.56, 60 In total, 100 snapshots evenly extracted from 2 to 5 ns were used to

205

calculate the energy terms. For each complex, the interaction spectrum between

206

inhibitor and TIE-2 on a per-residue basis was calculated by MM/GBSA

207

decomposition analysis supported by the mm_pbsa module in AMBER.61-63

208

Reagents and Materials for Bioassays. All reagents and anhydrous solvents were

209

purchased from commercial sources. The positive control TIE-2 kinase inhibitor, also

210

referenced under CAS 948557-43-5, was purchased from Merck Calbiochem

211

(Darmstadt, Germany, purity ≥ 97% by HPLC).64 Each compound was dissolved in

212

100% dimethyl sulfoxide (DMSO) as a 10 mM stock solution. The final DMSO

213

concentration in each reaction was less than 1%. Purified recombinant human TIE-2

214

protein (Catalog number: PV3628) and Z′-Lyte® kinase assay kit - ser/thr 5 peptide

215

(Catalog number: PV3178) were purchased from Thermo Fisher Scientific Inc. All the

216

96 test compounds were purchased from Specs, and the purity of these compounds is

217

≥ 95%, which is confirmed by Specs (compound data are summarized in Table S1).

218

In vitro Inhibitory Activity Assay of TIE-2. All the assays were carried out in

219

384-well plate format. The 4×test compounds and 4×ATP solution were firstly

220

prepared before the enzyme reaction starts. The 2×recombinant human TIE-2 protein /

221

Tyr 05 peptide mixture was prepared in 50 mM HEPES (pH 7.5), 0.01% BRIJ-35, 10

222

mM MgCl2, 4 mM MnCl2, 1 mM EGTA and 2 mM DTT. The final 10 µL kinase

223

reaction consists of 5 µL 1×TIE-2 / Tyr 05 peptide mixture (10 ng TIE-2 and 2 µM

224

Tyr 05 peptide), 2.5 µL ATP (100 µM) and 2.5 µL test compounds with desired

225

concentration. The assay plate was shaked on a plate shaker for 30 seconds to mix the

226

reactions thoroughly. After 1 hour kinase reaction incubation at room temperature

227

(20−25°C), 5 µL of a 1:1024 dilution of development reagent A is added, and the

228

assay plate was incubated for another 1 hour at room temperature. Then, the reactions 9

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

229

were terminated by adding 5 µL of Stop Reagent to each well, and the assay plate was

230

placed into a fluorescence plate reader (BioTek Synergy™ 4) to measure both the

231

coumarin and fluorescein emission signals (excitation: 400 nm; emission: 445 and

232

520 nm, respectively).

233

Results and Discussion

234

Evaluation of Virtual Screening Strategy. At present, five crystal structures of

235

TIE-2 are available in RCSB Protein Data Bank, namely 2OO8, 2OSC, 2P4I, 2WQB

236

and 3L8P. However, the structures of 2OO8, 2OSC and 2P4I are all DFG-out

237

conformations of TIE-2, which bind to type-II inhibitors. Thus, only 2WQB and 3L8P

238

with DFG-in conformations can be utilized to screen type-I inhibitors of TIE-2. In

239

order to determine the best screening strategy, the docking accuracy for both 2WQB

240

and 3L8P was evaluated to distinguish inhibitors from non-inhibitors using different

241

Glide docking modes. Each molecule in the validation set containing inhibitor and

242

non-inhibitor decoy sets was docked into the active pockets of both 2WQB and 3L8P

243

using three different Glide modes (HTVS, SP and XP).

244

The validation results were summarized in Figure 2. The student’s t-test was

245

utilized to assess the significance of the difference between two data sets, which can

246

be indicated by P-values. As shown in Figure 2, the P-values of 3L8P are obviously

247

lower than those of 2WQB in all three Glide modes, indicating that the discrimination

248

power of 3L8P in distinguishing inhibitors from non-inhibitors is better than that of

249

2WQB. So the structure of 3L8P was employed in the virtual screening workflow. It

250

was also observed that the P-values of HTVS Glide mode for both 2WQB and 3L8P

251

are quite high implying a poor discrimination power. Though HTVS mode is

252

extremely efficient in docking procedure, it is not used in our screening campaign due

253

to its low prediction accuracy. The XP Glide mode exhibited the best discrimination

254

power among all three models. However, XP mode is highly time-consuming when

255

entire Specs database is docked, thus SP mode with moderate prediction accuracy was

256

firstly carried out. Then, the top ranked 50,000 molecules were saved and re-docked 10

ACS Paragon Plus Environment

Page 10 of 32

Page 11 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

257

using Glide XP mode. In this way, both prediction accuracy and computing efficiency

258

were balanced. Subsequently, the top 1,000 compounds ranked by Glide XP docking

259

were filtered and clustered according to their predicted ADMET properties, and 41

260

compounds were finally purchased for bioassays (Figure 1).

261

In vitro Biological Activity of Virtual Screening Compounds. To evaluate the in

262

vitro TIE-2 enzymatic activity of the 41 candidate compounds, we developed a

263

quantitative FRET-based Z’-Lyte assay as described above. Commercially available

264

TIE-2 inhibitor (CAS: 948557-43-5) was included as a positive control to validate the

265

screening assay. Under the same assay conditions, TIE-2 inhibitor inhibited the kinase

266

activity of TIE-2 with an IC50 value of 0.31 µM, which is consistent with the reported

267

data (0.25 µM). In the first step, all the obtained 41 compounds were tested for

268

inhibition of TIE-2 activity at 25 µg/mL. The results were summarized and plotted in

269

Figure 3. Eight compounds (TP-S1-11, 18, 22, 30, 31, 37, 40 and 41) inhibited at least

270

50% of TIE-2 enzymatic activity at the concentration of 25 µg/mL, with four of them

271

(TP-S1-18, 30, 31 and 41) inhibiting at least 80 % of TIE-2 activity. We can find that

272

several compounds exhibited negative inhibitory activity, especially compounds

273

TP-S1-5, 9 and 20 (-101.76%, -240.97% and -94.16%, respectively). This

274

phenomenon may be caused by the auto-fluorescence property of the tested

275

compounds, which finally affects the detection of FRET signal. The inhibitory

276

activities of these compounds were actually not correctly characterized, so these

277

compounds were treated as inactive. The four compounds with over 80% inhibitory

278

activity were further tested to check their ability to inhibit TIE-2 activity at a lower

279

concentration, and the dose-dependent effects of these compounds (the inhibitory IC50

280

values) were then determined as well (Figure 4). The IC50 values for compounds

281

TP-S1-18, 30, 31 and 41 are 11.71 µg/mL (27.39 µM), 0.32 µg/mL (0.89 µM), 0.16

282

µg/mL (0.48 µM), 5.79 µg/mL (14.82 µM), respectively. Among them, compounds

283

TP-S1-30 and TP-S1-31 are in low micro molar range.

284

The novelty of these 4 active compounds was also evaluated by comparing the

285

structural similarity of each inhibitor to known TIE-2 inhibitors. Based on the 11

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

286

FCFP_4 fingerprints, the pairwise Tanimoto similarity, which is demonstrated to be a

287

preferential method of choice for computing molecular similarities,65 were calculated

288

using the Find Similar Molecules by Fingerprints protocol in Discovery Studio 2.5.

289

The values of Tanimoto coefficients range from zero (no bits in common) to unity (all

290

bits the same). According to our results, all the 4 inhibitors exhibit low Tanimoto

291

similarities to the known TIE-2 inhibitors obtained from BindingDB (TP-S1-18:

292

0.138, TP-S1-30: 0.148, TP-S1-31: 0.130 and TP-S1-41: 0.159). Therefore, the 4

293

inhibitors discovered from VS are structurally novel compared with the reported

294

TIE-2 inhibitors.

295

The active site residues Ile830, Glu832, Val838, Lys855, Leu876, Ala905, Leu971,

296

Ala981 and Asp982 are involved in the binding of the best four compounds to TIE-2.

297

However, the dominating residues for the binding of each inhibitor-protein complex

298

are different. According to the results of the Glide XP docking simulations shown in

299

Figure 5A, the key binding residues of the TP-S1-18/TIE-2 complex are Asp982,

300

Lue971, Val838 and Ile830. A hydrogen bond is formed between Asp982 and a

301

carbonyl group close to the benzene ring. The arene ring of isoindole in TP-S1-18 can

302

form hydrophobic interactions contacts (arene-H interaction) with both Lue971 and

303

Val838. Another hydrophobic contact can be observed between Ile830 and the

304

benzene ring of phenylethanone, which is an obvious difference compared with the

305

other inhibitors due to the extended phenylethanone structure in this region.

306

Compounds TP-S1-30 and 31 share the same [1,2,4]triazolo[3,4-b][1,3,4]thiadiazole

307

core structure linked with two conjugate ring group in 3- and 6-position, respectively.

308

Thus the key binding patterns of compounds TP-S1-30 and 31 are quite similar.

309

Residue Lys855 in both TP-S1-30 and 31 complexes can form cation-pi interactions

310

with the thiadiazole group of the core structure. Though the substitutions at 3-position

311

of the core structure are different, the same benzene ring can form similar

312

hydrophobic interaction with Val838 in both systems. However, different substitutions

313

at 3- and 6-positions of the core structure still have significant influence on the

314

binding modes of TP-S1-30 and 31. A hydrogen bond interaction was formed between

315

the residue Asp982 in TP-S1-31/TIE-2 complex and the substituted pyridine group at 12

ACS Paragon Plus Environment

Page 12 of 32

Page 13 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

316

6-position, but this phenomenon was not observed in the TP-S1-30/TIE-2 complex,

317

where the 3-chlorobenzene group is substituted at 6-position. So this suggests that

318

incorporating a hydrogen bond acceptor in the region close to Asp982 would help to

319

form similar hydrogen bonds and therefore benefit to inhibitor binding. In addition,

320

the binding patterns of the substituents at 3-position are also different. For the

321

TP-S1-30/TIE-2 complex, the oxygen of the 1,3-benzodioxole group can form

322

hydrogen bond interaction with the backbone of Ala905. But the dioxole group is

323

replaced by a benzene group in compound TP-S1-31, which can interact with residue

324

Leu971 via hydrophobic contact. In the binding of TP-S1-41 to TIE-2, the

325

hydrophobic interactions of the residues Lys855, Val838, and Ala981 are observed.

326

Different from compounds TP-S1-18, 30 and 31, the phenylethanone group extends

327

into a different region close to Glu832, where a hydrogen bond is formed between the

328

carbonyl oxygen and the backbone of Glu832.

329

Preliminary Optimization and SAR Analysis Based on the Core Structures of

330

TP-S1-30 and 31. To obtain a preliminary understanding of the structure−activity

331

relationships, substructure searches were carried based on the Specs database.

332

According to the results from our virtual screening campaign, two most potent

333

candidate inhibitors, TP-S1-30 and TP-S1-31 (IC50 = 0.89 µM and 0.48 µM,

334

respectively), were selected and submitted to initial optimization and SAR analysis

335

using substructure search. Unfortunately, few derivatives were available in the

336

database when the frame structure of either TP-S1-30 or TP-S1-31 was taken as the

337

template

338

([1,2,4]triazolo[3,4-b][1,3,4]thiadiazole) of both compounds TP-S1-30 and 31 linked

339

with two phenyl groups at 3- and 6-position was taken as one of the starting

340

structures.

scaffold.

Thus,

the

identical

core

structure

341

Hundreds of compounds were retrieved through substructure search, and most

342

substituents located in the two phenyl groups at 3- and 6-positions of the core

343

structure. To give an explicit SAR analysis, only the compounds with substituents at

344

one side (3- or 6-position phenyl) were selected and purchased for the following 13

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

345

biological evaluation. 26 compounds listed in Table 1 mainly focus on R1

346

modifications at 6-position of the phenyl group. A preliminary enzymatic inhibitory

347

test of these compounds was first carried out. We can observe that almost half of the

348

tested compounds inhibited over 95% TIE-2 activity at the concentration of 5 µg/mL.

349

However, there were still eight compounds exhibiting low percentage inhibitory

350

activity, so the dose-dependent effects of these compounds were not determined. The

351

inhibitory IC50 values varied significantly among the determined compounds, ranging

352

from 0.169 µM to 60.95 µM, which implies that substitutions at different positions of

353

R1 have a great influence on the activity of the derivatives.

354

Compound TP-S1-44 is the most potent inhibitor among the 26 compounds in

355

Table 1 with an IC50 value of 0.169 µM, which is almost 10-fold more active than

356

compound TP-S1-42 (IC50 = 1.267 µM) without any substitution at R1 and/or R2.

357

MM/GBSA has been validated as a successful method to compare the binding

358

affinities of related inhibitors and to gain rational insights into their differences.66, 67

359

Therefore, in order to understand the difference of the binding mechanism among

360

these derivatives, the MM/GBSA binding free energy calculations and decomposition

361

analysis based on MD simulations were carried out. According to the residue-inhibitor

362

interaction spectrum of the dominating amino acids depicted in Figure 6, the

363

contributions of residues Val838 and Ile902 determine the higher binding affinity of

364

compound TP-S1-44 over TP-S1-42 and 46. Though hydrophobic contacts are

365

observed in the binding of TP-S1-42 to residues Val838 and Lue971, the interactions

366

are relatively weak. Introducing the methoxy group at 2-R1 position leads to the

367

interaction between the R1 phenyl ring and Lue876, which is not found in the binding

368

of TP-S1-42 and 46. Incorporation of strong electron withdrawing groups (2-F or

369

2-CF3) at R1 phenyl dramatically decreases compound activity. However, the effect of

370

the 3-F or 3-Cl modification at R1 phenyl is not significant. In contrast to TP-S1-44,

371

substitution of the methoxy group at 3-R1 position opposes binding. For the

372

modifications at 4-R1 position, the impact of introducing the methoxy, Cl, F or ethoxy

373

group is quite limited, but modifications with the Br, ethyl, propyl or isopropyl group

374

significantly decrease activity. The replacement of two or more positions at R1 phenyl 14

ACS Paragon Plus Environment

Page 14 of 32

Page 15 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

375

is not favorable to inhibitor binding with at least 40 fold decline in activity compared

376

to TP-S1-42, which can be roughly reflected by their poor performance in docking

377

simulations (not shown).

378

Chemical modifications of moieties at R2 phenyl are listed in Table 2. We can

379

find that the compounds containing the methoxy substitution at 3-R2 position show

380

the best activity, e.g. TP-S1-73 and 85. Compound activity is significantly influenced

381

when the trifluoromethyl group is introduced to either 3-R2 or 4-R2 position

382

(compounds TP-S1-75 and 84). Interestingly, the activity of TP-S1-79 with the F

383

substituted at 4-R2 position, which is also a strong electron withdrawing group, is

384

much higher compared with TP-S1-84. In order to explore the structural mechanism

385

underlying the difference in compound activity, the binding modes of compounds

386

TP-S1-73, 79 and 84 were analyzed and the contributions of the important residues

387

for binding were also calculated and compared. As plotted in Figure 7, the R2 phenyl

388

ring of compound TP-S1-73 can form hydrophobic interactions with residues Val838

389

and Lue971. Similar interactions can also be observed in the binding of compounds

390

TP-S1-79 and 84, indicating that the decreased activity of TP-S1-84 may not be

391

caused by the fluctuations of residues Val838 and Lue971. This phenomenon is also

392

demonstrated by the energy decomposition analysis depicted in Figure 7D, where the

393

contributions of residues Val838 and Lue971 vary slightly upon binding to the three

394

compounds. By comparing the difference of the contribution of each amino acid, we

395

can observe that the impaired interactions with residues Gly831, Lys855 and Ile902

396

determine the inferior binding affinity of compound TP-S1-84. Though the inhibitory

397

activities of compounds TP-S1-73 and 79 are similar, several key interactions for

398

binding are strikingly different. The interactions of compound TP-S1-73 with residues

399

Lys855, Ile886 and Ile902 are much stronger than those of TP-S1-79. For the binding

400

of TP-S1-79, the contributions of residues Gly831, Val838 are greater. Of the other

401

molecules in Table 2, compounds TP-S1-81 and 86 also exhibit superior inhibitory

402

activities with IC50 values of 0.301 nM and 0.283 nM, respectively.

403

In addition, several derivatives of the core structure of compound TP-S1-31 with

404

the pyridine group replaced by the phenyl ring were also retrieved from the Specs 15

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

405

database. As we can see in Table 3, the IC50 values of most compounds are higher

406

than 2 µM, except for compound TP-S1-90 with Cl at the 3 position of R3 phenyl.

407

This phenomenon, on the contrary, implies the significance of the pyridine group to

408

the binding of TP-S1-31 to TIE-2, where a hydrogen bond is formed between the

409

residue Asp982 and pyridine.

410

Because of the limitation of substructure search methodology, the diversity of the

411

modified compounds is far from sufficient and needs to be further optimized.

412

According to the discussions above, some crucial interaction patterns between these

413

inhibitors and TIE-2 are determined. Most binding pocket residues of TIE-2 are

414

nonpolar, especially the regions around R1 and R2 phenyl, so the hydrophobic

415

interactions with the surrounding residues (e.g. Val838, Leu876 and Leu971) should

416

be maintained. Not only the phenyl groups but also various aromatic rings can be

417

incorporated into both 3- and 6-position of the [1,2,4]triazolo[3,4-b][1,3,4]thiadiazole

418

core structure. In spite of the hydrophobic property of the binding site, the regions

419

close to the flank of the core structure (e.g. residues Lys855 and Asp982) are

420

hydrophilic. Thus introducing moieties containing hydrogen donors or acceptors into

421

the core structure, which is not realized by substructure search, might benefit to form

422

H-bond interactions. The inhibitors identified and the information presented in this

423

work will help to guide further design and optimization of more potent inhibitors.

424

Conclusions

425

Due to the critical role of ANG/TIE-2 signaling pathway in the angiogenesis process

426

of preliminary vessels, TIE-2 inhibitors have gained significant attention and are now

427

believed to be potential preventative and therapeutic agents for a variety of

428

life-threatening diseases. Several candidate inhibitors of TIE-2 have been reported by

429

pharmaceutical companies in recent years, and some agents have entered clinical trials

430

for the treatment of a few cancer types. However, the development of selective

431

small-molecule inhibitors of TIE-2 is also quite limited. In this work, we developed a

432

structure-based virtual screening approach to discover TIE-2 inhibitors, and have

433

successfully identified several active compounds. Although most of the hit 16

ACS Paragon Plus Environment

Page 16 of 32

Page 17 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

434

compounds discovered showed inhibitory activity in moderate to low micromolar

435

range, the scaffold of each inhibitor is novel and thereby represents a potential starting

436

structure for further drug development. Based on substructure search technology,

437

preliminary optimization and SAR analysis of the core structures of both compound

438

TP-S1-30 and 31 were performed, leading to the discovery of several sub-100nM

439

inhibitors. According to the binding properties of the optimized inhibitors from MD

440

simulations and MM/GBSA predictions, the relationship between inhibitor structure

441

and biological activity was analyzed at length. The detailed mechanism of action of

442

these compounds binding to TIE-2 and the information obtained from SAR analysis

443

justify further investigation to improve the inhibitory activity of these candidate

444

compounds.

445

Associated Content

446

Supporting Information

447

Supporting Information Available: ** Chemical information and 2-D structures of the

448

purchased compounds from Specs database.** This material is available free of

449

charge via the Internet at http://pubs.acs.org.

450

Acknowledgments

451

This study was supported by the National Science Foundation of China (21575128).

452

References

453 454 455 456 457 458 459 460 461 462 463 464

1.

Villanueva, M. T., Angiogenesis: A Sudden Rush of Blood to the Tumour. Nat. Rev. Cancer. 2015,

15, 135-135. 2.

Risau, W., Mechanisms of Angiogenesis. Nature. 1997, 386, 671-674.

3.

Lindahl, P.; Johansson, B. R.; Levéen, P.; Betsholtz, C., Pericyte Loss and Microaneurysm

Formation in Pdgf-B-Deficient Mice. Science. 1997, 277, 242-245. 4.

Musumeci, F.; Radi, M.; Brullo, C.; Schenone, S., Vascular Endothelial Growth Factor (Vegf)

Receptors: Drugs and New Inhibitors. J. Med. Chem. 2012, 55, 10797-10822. 5.

Ishak, R. S.; Aad, S. A.; Kyei, A.; Farhat, F. S., Cutaneous Manifestations of Anti-Angiogenic

Therapy in Oncology: Review with Focus on Vegf Inhibitors. Crit. Rev. Oncol. Hemat. 2014, 90, 152-164. 6.

Demetri, G. D.; van Oosterom, A. T.; Garrett, C. R.; Blackstein, M. E.; Shah, M. H.; Verweij, J.;

McArthur, G.; Judson, I. R.; Heinrich, M. C.; Morgan, J. A., Efficacy and Safety of Sunitinib in Patients with Advanced Gastrointestinal Stromal Tumour after Failure of Imatinib: A Randomised Controlled 17

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508

Trial. Lancet 2006, 368, 1329-1338. 7.

Escudier, B.; Eisen, T.; Stadler, W. M.; Szczylik, C.; Oudard, S.; Staehler, M.; Negrier, S.; Chevreau,

C.; Desai, A. A.; Rolland, F., Sorafenib for Treatment of Renal Cell Carcinoma: Final Efficacy and Safety Results of the Phase Iii Treatment Approaches in Renal Cancer Global Evaluation Trial. J. Clin. Oncol. 2009, 27, 3312-3318. 8.

Bergers, G.; Hanahan, D., Modes of Resistance to Anti-Angiogenic Therapy. Nat. Rev. Cancer. 2008,

8, 592-603. 9.

Casanovas, O.; Salazar, R.; Tabernero, J., Multi-Target Angiokinase Inhibitors to Fight Resistance.

Cell Cycle. 2014, 13, 2649-2650. 10. Vasudev, N. S.; Reynolds, A. R., Anti-Angiogenic Therapy for Cancer: Current Progress, Unresolved Questions and Future Directions. Angiogenesis. 2014, 17, 471-494. 11. Tozer, G.; Kanthou, C.; Lewis, G.; Prise, V.; Vojnovic, B.; Hill, S., Tumour Vascular Disrupting Agents: Combating Treatment Resistance. Br. J. Radiol. 2014. 12. Rivera, L. B.; Meyronet, D.; Hervieu, V.; Frederick, M. J.; Bergsland, E.; Bergers, G., Intratumoral Myeloid Cells Regulate Responsiveness and Resistance to Antiangiogenic Therapy. Cell Rep. 2015, 11, 577-591. 13. Augustin, H. G.; Koh, G. Y.; Thurston, G.; Alitalo, K., Control of Vascular Morphogenesis and Homeostasis through the Angiopoietin–Tie System. Nat. Rev. Mol. Cell Biol. 2009, 10, 165-177. 14. Hu, J.; Srivastava, K.; Wieland, M.; Runge, A.; Mogler, C.; Besemfelder, E.; Terhardt, D.; Vogel, M. J.; Cao, L.; Korn, C., Endothelial Cell-Derived Angiopoietin-2 Controls Liver Regeneration as a Spatiotemporal Rheostat. Science. 2014, 343, 416-419. 15. Biel, N. M.; Siemann, D. W., Targeting the Angiopoietin-2/Tie-2 Axis in Conjunction with Vegf Signal Interference. Cancer Lett. 2014. 16. Gerald, D.; Chintharlapalli, S.; Augustin, H. G.; Benjamin, L. E., Angiopoietin-2: An Attractive Target for Improved Antiangiogenic Tumor Therapy. Cancer Res. 2013, 73, 1649-1657. 17. Jendreyko, N.; Popkov, M.; Rader, C.; Barbas, C. F., Phenotypic Knockout of Vegf-R2 and Tie-2 with an Intradiabody Reduces Tumor Growth and Angiogenesis in Vivo. Proc. Natl. Acad. Sci. 2005, 102, 8293-8298. 18. Daly, C.; Eichten, A.; Castanaro, C.; Pasnikowski, E.; Adler, A.; Lalani, A. S.; Papadopoulos, N.; Kyle, A. H.; Minchinton, A. I.; Yancopoulos, G. D., Angiopoietin-2 Functions as a Tie2 Agonist in Tumor Models, Where It Limits the Effects of Vegf Inhibition. Cancer Res. 2013, 73, 108-118. 19. Chen, H.; Tsai, C.; Hung, W., Foretinib Inhibits Angiogenesis, Lymphangiogenesis and Tumor Growth of Pancreatic Cancer in Vivo by Decreasing Vegfr-2/3 and Tie-2 Signaling. Oncotarget. 2015, 17, 14940-14952. 20. Brown, J. L.; Cao, Z. A.; Pinzon-Ortiz, M.; Kendrew, J.; Reimer, C.; Wen, S.; Zhou, J. Q.; Tabrizi, M.; Emery, S.; McDermott, B.; Pablo, L.; McCoon, P.; Bedian, V.; Blakey, D. C., A Human Monoclonal Anti-Ang2 Antibody Leads to Broad Antitumor Activity in Combination with Vegf Inhibitors and Chemotherapy Agents in Preclinical Models. Mol. Cancer Ther. 2010, 9, 145-156. 21. Hudkins, R. L.; Becknell, N. C.; Zulli, A. L.; Underiner, T. L.; Angeles, T. S.; Aimone, L. D.; Albom, M. S.; Chang, H.; Miknyoczki, S. J.; Hunter, K., Synthesis and Biological Profile of the Pan-Vascular Endothelial Growth Factor Receptor/Tyrosine Kinase with Immunoglobulin and Epidermal Growth Factor-Like

Homology

Domains

2

(Vegf-R/Tie-2)

Inhibitor

11-(2-Methylpropyl)-12,

13-Dihydro-2-Methyl-8-(Pyrimidin-2-Ylamino)-4 H-Indazolo [5, 4-a] Pyrrolo [3, 4-C] Carbazol-4-One (Cep-11981): A Novel Oncology Therapeutic Agent. J. Med. Chem. 2012, 55, 903-913. 18

ACS Paragon Plus Environment

Page 18 of 32

Page 19 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552

22. Arcari, J. T.; Beebe, J. S.; Berliner, M. A.; Bernardo, V.; Boehm, M.; Borzillo, G. V.; Clark, T.; Cohen, B. D.; Connell, R. D.; Frost, H. N.; Gordon, D. A.; Hungerford, W. M.; Kakar, S. M.; Kanter, A.; Keene, N. F.; Knauth, E. A.; LaGreca, S. D.; Lu, Y.; Martinez-Alsina, L.; Marx, M. A.; Morris, J.; Patel, N. C.; Savage, D.; Soderstrom, C. I.; Thompson, C.; Tkalcevic, G.; Tom, N. J.; Vajdos, F. F.; Valentine, J. J.; Vincent, P. W.; Wessel, M. D.; Chen, J. M., Discovery and Synthesis of Novel 4-Aminopyrrolopyrimidine Tie-2 Kinase Inhibitors for the Treatment of Solid Tumors. Bioorg. Med. Chem. Lett. 2013, 23, 3059-3063. 23. Hodous, B. L.; Geuns-Meyer, S. D.; Hughes, P. E.; Albrecht, B. K.; Bellon, S.; Bready, J.; Caenepeel, S.; Cee, V. J.; Chaffee, S. C.; Coxon, A.; Emery, M.; Fretland, J.; Gallant, P.; Gu, Y.; Hoffman, D.; Johnson, R. E.; Kendall, R.; Kim, J. L.; Long, A. M.; Morrison, M.; Olivieri, P. R.; Patel, V. F.; Polverino, A.; Rose, P.; Tempest, P.; Wang, L.; Whittington, D. A.; Zhao, H., Evolution of a Highly Selective and Potent 2-(Pyridin-2-Yl)-1,3,5-Triazine Tie-2 Kinase Inhibitor. J. Med. Chem. 2007, 50, 611-626. 24. Gross, S.; Winski, S.; Pheneger, J.; Anderson, D.; Munson, M.; Rodriguez, M., Arry-614, a Small Molecule Inhibitor of P38, Abl, Tie2 and Vegfr2 with Multiple Potential Therapeutic Applications. Kinases Proceedings of the Cancer Meeting: Lessons from the Clinic. 2006, 14-19. 25. Hasegawa, M.; Nishigaki, N.; Washio, Y.; Kano, K.; Harris, P. A.; Sato, H.; Mori, I.; West, R. I.; Shibahara, M.; Toyoda, H., Discovery of Novel Benzimidazoles as Potent Inhibitors of Tie-2 and Vegfr-2 Tyrosine Kinase Receptors. J. Med. Chem. 2007, 50, 4453-4470. 26. Cee, V. J.; Albrecht, B. K.; Geuns-Meyer, S.; Hughes, P.; Bellon, S.; Bready, J.; Caenepeel, S.; Chaffee, S. C.; Coxon, A.; Emery, M.; Fretland, J.; Gallant, P.; Gu, Y.; Hodous, B. L.; Hoffman, D.; Johnson, R. E.; Kendall, R.; Kim, J. L.; Long, A. M.; McGowan, D.; Morrison, M.; Olivieri, P. R.; Patel, V. F.; Polverino, A.; Powers, D.; Rose, P.; Wang, L.; Zhao, H., Alkynylpyrimidine Amide Derivatives as Potent, Selective, and Orally Active Inhibitors of Tie-2 Kinase. J. Med. Chem. 2007, 50, 627-640. 27. Leow, C. C.; Coffman, K.; Inigo, I.; Breen, S.; Czapiga, M.; Soukharev, S.; Gingles, N.; Peterson, N.; Fazenbaker, C.; Woods, R., Medi3617, a Human Anti-Angiopoietin 2 Monoclonal Antibody, Inhibits Angiogenesis and Tumor Growth in Human Tumor Xenograft Models. Int. J. Oncol. 2012, 40, 1321-1330. 28. Huang, H.; Lai, J.-Y.; Do, J.; Liu, D.; Li, L.; Del Rosario, J.; Doppalapudi, V. R.; Pirie-Shepherd, S.; Levin, N.; Bradshaw, C., Specifically Targeting Angiopoietin-2 Inhibits Angiogenesis, Tie2-Expressing Monocyte Infiltration, and Tumor Growth. Clin. Cancer Res. 2011, 17, 1001-1011. 29. Papadopoulos, K.; Chau, N.; Patnaik, A.; Adriaens, L.; Lalani, A.; Daly, C.; Siu, L. A Phase I First-in-Human Study of Regn910, a Fully Human and Selective Angiopoietin-2 Monoclonal Antibody, in Patients with Advanced Solid Tumor Malignancies. In ASCO Annual Meeting Proceedings., 2011; 2011; Vol. 29; p TPS159. 30. Saha, I.; Rak, B.; Bhowmick, S. S.; Maulik, U.; Bhattacharjee, D.; Koch, U.; Lazniewski, M.; Plewczynski, D., Binding Activity Prediction of Cyclin-Dependent Inhibitors. J. Chem. Inf. Model. 2015, 55, 1469-1482. 31. Xu, L.; Zhang, Y.; Zheng, L.; Qiao, C.; Li, Y.; Li, D.; Zhen, X.; Hou, T., Discovery of Novel Inhibitors Targeting the Macrophage Migration Inhibitory Factor Via Structure-Based Virtual Screening and Bioassays. J. Med. Chem. 2014, 57, 3737-3745. 32. Shen, M.; Yu, H.; Li, Y.; Li, P.; Pan, P.; Zhou, S.; Zhang, L.; Li, S.; Lee, S. M.-Y.; Hou, T., Discovery of Rho-Kinase Inhibitors by Docking-Based Virtual Screening. Mol. BioSyst. 2013, 9, 1511-1521. 33. Singh, N.; Tiwari, S.; Srivastava, K. K.; Siddiqi, M. I., Identification of Novel Inhibitors of Mycobacterium Tuberculosis Pkng Using Pharmacophore Based Virtual Screening, Docking, Molecular Dynamics Simulation, and Their Biological Evaluation. J. Chem. Inf. Model. 2015, 55, 1120-1129. 19

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596

Page 20 of 32

34. Tian, S.; Sun, H.; Pan, P.; Li, D.; Zhen, X.; Li, Y.; Hou, T., Assessing an Ensemble Docking-Based Virtual Screening Strategy for Kinase Targets by Considering Protein Flexibility. J. Chem. Inf. Model. 2014, 54, 2664-2679. 35. Li, N.; Ainsworth, R. I.; Ding, B.; Hou, T.; Wang, W., Using Hierarchical Virtual Screening to Combat Drug Resistance of the Hiv-1 Protease. J. Chem. Inf. Model. 2015. 36. Lindh, M.; Svensson, F.; Schaal, W.; Zhang, J.; Sköld, C.; Brandt, P.; Karlén, A., Toward a Benchmarking Data Set Able to Evaluate Ligand-and Structure-Based Virtual Screening Using Public Hts Data. J. Chem. Inf. Model. 2015, 55, 343-353. 37. Berman, H. M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.; Weissig, H.; Shindyalov, I. N.; Bourne, P. E., The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235-242. 38. Hodous, B. L.; Geuns-Meyer, S. D.; Hughes, P. E.; Albrecht, B. K.; Bellon, S.; Caenepeel, S.; Cee, V. J.; Chaffee, S. C.; Emery, M.; Fretland, J., Synthesis, Structural Analysis, and Sar Studies of Triazine Derivatives as Potent, Selective Tie-2 Inhibitors. Bioorg. Med. Chem. Lett. 2007, 17, 2886-2889. 39. Schrödinger, M., Version 9.0. Schrödinger LLC, New York, NY 2009. 40. Nicola, G.; Liu, T.; Hwang, L.; Gilson, M., Bindingdb: A Protein-Ligand Database for Drug Discovery. Biophys. J. 2012, 102, 61a. 41. Liu, T.; Lin, Y.; Wen, X.; Jorissen, R. N.; Gilson, M. K., Bindingdb: A Web-Accessible Database of Experimentally Determined Protein–Ligand Binding Affinities. Nucleic Acids Res. 2007, 35, D198-D201. 42. Discovery Studio 2.5 Guide, Accelrys Inc., San Diego, 2009, Http://Www.Accelrys.Com, 2009. 43. Luke, R. W. A.; Ballard, P.; Buttar, D.; Campbell, L.; Curwen, J.; Emery, S. C.; Griffen, A. M.; Hassall, L.; Hayter, B. R.; Jones, C. D.; McCoull, W.; Mellor, M.; Swain, M. L.; Tucker, J. A., Novel Thienopyrimidine and Thiazolopyrimidine Kinase Inhibitors with Activity against Tie-2 in Vitro and in Vivo. Bioorg. Med. Chem. Lett. 2009, 19, 6670-6674. 44. Hudkins, R. L.; Becknell, N. C.; Zulli, A. L.; Underiner, T. L.; Angeles, T. S.; Aimone, L. D.; Albom, M. S.; Chang, H.; Miknyoczki, S. J.; Hunter, K.; Jones-Bolin, S.; Zhao, H.; Bacon, E. R.; Mallamo, J. P.; Ator, M. A.; Ruggeri, B. A., Synthesis and Biological Profile of the Pan-Vascular Endothelial Growth Factor Receptor/Tyrosine Kinase with Immunoglobulin and Epidermal Growth Factor-Like Homology Domains 2

(Vegf-R/Tie-2)

Inhibitor

11-(2-Methylpropyl)-12,13-Dihydro-2-Methyl-8-(Pyrimidin-2-Ylamino)-4h-Indazolo[5,4-a]Pyrrolo[3,4-C ]Carbazol-4-One (Cep-11981): A Novel Oncology Therapeutic Agent. J. Med. Chem. 2012, 55, 903-913. 45. Acd/Adme Suite 5.0, Advanced Chemistry Development Inc., Toronto, Canada. 2011. 46. Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J., Experimental and Computational Approaches to Estimate Solubility and Permeability in Drug Discovery and Development Settings. Adv. Drug Delivery Rev. 2012, 64, Supplement, 4-17. 47. Oprea, T. I., Property Distribution of Drug-Related Chemical Databases. J. Comput.-Aided Mol. Des. 2000, 14, 251-264. 48. Walters, W. P.; Namchuk, M., Designing Screens: How to Make Your Hits a Hit. Nat. Rev. Drug Discovery 2003, 2, 259-266. 49. Case, D.; Babin, V.; Berryman, J.; Betz, R.; Cai, Q.; Cerutti, D.; Cheatham III, T.; Darden, T.; Duke, R.; Gohlke, H., Amber 14 (University of California). San Francisco 2014. 50. Frisch, M.; Trucks, G.; Schlegel, H.; Scuseria, G.; Robb, M.; Cheeseman, J.; Montgomery Jr, J.; Vreven, T.; Kudin, K.; Burant, J., Gaussian 03, Revision D. 01. Gaussian, Inc.: Wallingford, CT 2004. 51. Bayly, C. I.; Cieplak, P.; Cornell, W. D.; Kollman, P. A., A Well-Behaved Electrostatic Potential Based Method Using Charge Restraints for Deriving Atomic Charges: The Resp Model. J. Phys. Chem. 1993, 97, 20

ACS Paragon Plus Environment

Page 21 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638

10269-10280. 52. Wang, J. M.; Wolf, R. M.; Caldwell, J. W.; Kollman, P. A.; Case, D. A., Development and Testing of a General Amber Force Field. J. Comput. Chem. 2004, 25, 1157-1174. 53. Duan, Y.; Wu, C.; Chowdhury, S.; Lee, M. C.; Xiong, G. M.; Zhang, W.; Yang, R.; Cieplak, P.; Luo, R.; Lee, T.; Caldwell, J.; Wang, J. M.; Kollman, P., A Point-Charge Force Field for Molecular Mechanics Simulations of Proteins Based on Condensed-Phase Quantum Mechanical Calculations. J. Comput. Chem. 2003, 24, 1999-2012. 54. Darden, T.; York, D.; Pedersen, L., Particle Mesh Ewald: An W Log (N) Method for Ewald Sums in Large Systems. J. Chem. Phys. 1993, 98, 10089-10092. 55. Ryckaert, J. P.; Ciccotti, G.; Berendsen, H. J. C., Numerical Integration of the Cartesian Equations of Motion of a System with Constraints: Molecular Dynamics of< I> N-Alkanes. J. Comput. Phys. 1977, 23, 327-341. 56. Wang, J. M.; Hou, T. J.; Xu, X. J., Recent Advances in Free Energy Calculations with a Combination of Molecular Mechanics and Continuum Models. Curr. Comput.-Aided Drug Des. 2006, 2, 287-306. 57. Kollman, P. A.; Massova, I.; Reyes, C.; Kuhn, B.; Huo, S.; Chong, L.; Lee, M.; Lee, T.; Duan, Y.; Wang, W., Calculating Structures and Free Energies of Complex Molecules: Combining Molecular Mechanics and Continuum Models. Accounts Chem. Res. 2000, 33, 889-897. 58. Onufriev, A.; Bashford, D.; Case, D. A., Exploring Protein Native States and Large-Scale Conformational Changes with a Modified Generalized Born Model. Proteins: Struct., Funct., Bioinf. 2004, 55, 383-394. 59. Weiser, J.; Shenkin, P. S.; Still, W. C., Approximate Atomic Surfaces from Linear Combinations of Pairwise Overlaps (Lcpo). J. Comput. Chem. 1999, 20, 217-230. 60. Hou, T. J.; Li, Y. Y.; Wang, W., Prediction of Peptides Binding to the Pka Rii Alpha Subunit Using a Hierarchical Strategy. Bioinformatics 2011, 27, 1814-1821. 61. Hou, T.; Li, N.; Li, Y.; Wang, W., Characterization of Domain-Peptide Interaction Interface: Prediction of Sh3 Domain-Mediated Protein-Protein Interaction Network in Yeast by Generic Structure-Based Models. J. Proteome Res. 2012, 11, 2982. 62. Hou, T.; Zhang, W.; Case, D. A.; Wang, W., Characterization of Domain-Peptide Interaction Interface: A Case Study on the Amphiphysin-1 Sh3 Domain. J. Mol. Biol. 2008, 376, 1201-1214. 63. Hou, T. J.; Xu, Z.; Zhang, W.; McLaughlin, W. A.; Case, D. A.; Xu, Y.; Wang, W., Characterization of Domain-Peptide Interaction Interface. Mol. Cell. Proteomics. 2009, 8, 639-649. 64. Semones, M.; Feng, Y.; Johnson, N.; Adams, J. L.; Winkler, J.; Hansbury, M., Pyridinylimidazole Inhibitors of Tie2 Kinase. Bioorg. Med. Chem. Lett. 2007, 17, 4756-4760. 65. Willett, P., Similarity-Based Virtual Screening Using 2d Fingerprints. Drug Discovery Today 2006, 11, 1046-1053. 66. Shen, M.; Zhou, S.; Li, Y.; Pan, P.; Zhang, L.; Hou, T., Discovery and Optimization of Triazine Derivatives as Rock1 Inhibitors: Molecular Docking, Molecular Dynamics Simulations and Free Energy Calculations. Mol. BioSyst. 2013, 9, 361-374. 67. Sun, H.; Li, Y.; Tian, S.; Xu, L.; Hou, T., Assessing the Performance of Mm/Pbsa and Mm/Gbsa Methods. 4. Accuracies of Mm/Pbsa and Mm/Gbsa Methodologies Evaluated by Various Simulation Protocols Using Pdbbind Data Set. Phys. Chem. Chem. Phys. 2014, 16, 16719-16729.

639 21

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

640

Legend of the Figures

641 642

Figure 1. Workflow of the structure-based virtual screening.

643 644

Figure 2. Distributions of the glide docking scores of the inhibitor decoy and

645

non-inhibitor decoy dataset for the two crystal complexes of TIE-2. (A) PDB entry:

646

2WQB, Glide HTVS docking, (B) PDB entry: 2WQB, Glide SP docking, (C) PDB

647

entry: 2WQB, Glide XP docking, (D) PDB entry: 3L8P, Glide HTVS docking, (E)

648

PDB entry: 3L8P, Glide SP docking, (F) PDB entry: 3L8P, Glide XP docking;

649 650

Figure 3. Inhibitory activity of the 41 hit compounds from VS campaign at the

651

concentration of 25 µg/mL. All values are the average of n ≥ 2 ± standard deviation.

652

The plots of the compounds with < -100% inhibitory activity were not fully presented.

653 654

Figure 4. Dose-dependent analysis of TIE-2 inhibition by compound TP-S1-18 (A),

655

TP-S1-30 (B), TP-S1-31 (C), and TP-S1-41 (D). For determination of IC50 values, the

656

resulting inhibitory activity was plotted against the concentration of inhibitor, and the

657

data was fitted to a dose-response curve with a variable slop.

658 659

Figure 5. Schematic representation of the interactions between TIE-2 and four active

660

compounds. (A) TP-S1-18, (B) TP-S1-30, (C) TP-S1-31, and (D) TP-S1-41. All the

661

binding complexes are predicted by Glide XP docking using 3L8P crystal structure as

662

TIE-2 template.

663 664

Figure 6. 2-D schematic diagrams of the binding patterns for compound (A)

665

TP-S1-42, (B) TP-S1-43, and (C) TP-S1-44. (D) Contribution of the important

666

residues for ligand binding. All structures are average conformations generated from

667

the last 3 ns snapshots of each MD system. The averaged energetic contributions of

668

the dominating residues from MM/GBSA decomposition analysis are also plotted. 22

ACS Paragon Plus Environment

Page 22 of 32

Page 23 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

669 670

Figure 7. 2-D schematic diagrams of the binding patterns for compound (A)

671

TP-S1-73, (B) TP-S1-79, and (C) TP-S1-84. (D) Contribution of the important

672

residues for ligand binding. All structures are average conformations generated from

673

the last 3 ns snapshots of each MD system. The averaged energetic contributions of

674

the dominating residues from MM/GBSA decomposition analysis are also plotted.

675 676

23

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 32

Table 1. TIE-2 Kinase Inhibitory Activity @ 5 µg/mL and Half-Maximal Inhibitory Concentrations (IC50) for TP-S1-42 ~ 67 Analogues with R1 Modifications

a

Compd

R1

R2

Kinase Inhibition (%) @ 5 µg/mL (±SD) a

IC50, µM (±SD) a

TP-S1-42 TP-S1-43 TP-S1-44 TP-S1-45 TP-S1-46 TP-S1-47 TP-S1-48 TP-S1-49 TP-S1-50 TP-S1-51 TP-S1-52 TP-S1-53 TP-S1-54 TP-S1-55 TP-S1-56 TP-S1-57 TP-S1-58 TP-S1-59 TP-S1-60 TP-S1-61 TP-S1-62 TP-S1-63 TP-S1-64 TP-S1-65

-H 2-F 2-OCH3 2-I 2-OCH2CH3 2-CF3 2-Cl 3-Cl 3-F 3-OCH3 4-OCH3 4-Cl 4-F 4-OCH2CH3 4-Br 4-CH2CH3 4-CH2CH2CH3 4-CH(CH3)2 3-Cl, 4-F 3, 4-OCH3 3, 4, 5-OCH3 3, 5-CH3 2, 4-CH3 2, 3, 4, 5-F

-H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H

97.33 ± 0.45 17.57 ± 4.49 99.59 ± 0.16 97.74 ± 0.34 97.74 ± 1.07 48.41 ± 2.27 97.02 ± 0.96 99.81 ± 1.04 98.45 ± 0.21 82.27 ± 1.55 98.44 ± 0.07 99.88 ± 3.03 98.93 ± 1.18 94.26 ± 0.14 16.90 ± 3.41 12.08 ± 4.35 20.62 ± 0.77 14.42 ± 0.31 45.14 ± 0.95 26.31 ± 3.90 38.83 ± 2.35 15.77 ± 4.21 42.33 ± 0.32 47.75 ± 2.33

1.267 ± 0.129 NT b 0.169 ± 0.055 2.208 ± 0.059 1.877 ± 0.066 44.36 ± 6.59 8.478 ± 0.401 4.826 ± 0.793 0.97 ± 0.093 22.93 ± 2.56 1.208 ± 0.102 0.822 ± 0.028 4.484 ± 0.38 4.388 ± 0.1 NT b NT b NT b NT b 41.62 ± 3.82 NT b NT b NT b 47.31 ± 25.22 60.95 ± 16.29

TP-S1-66

97.59 ± 1.55

0.768 ± 0.006

TP-S1-67

89.63 ± 2.24

6.739 ± 0.736

All values are the average of n ≥ 2 ± standard deviation. bNT, not tested. 24

ACS Paragon Plus Environment

Page 25 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

677 Table 2. TIE-2 Kinase Inhibitory Activity @ 5 µg/mL and Half-Maximal Inhibitory Concentrations (IC50) for TP-S1-68 ~ 87 Analogues with R2 Modifications

a

Compd

R1

R2

Kinase Inhibition (%) @ 5 µg/mL (±SD) a

IC50, µM (±SD) a

TP-S1-68 TP-S1-69 TP-S1-70 TP-S1-71 TP-S1-72 TP-S1-73 TP-S1-74 TP-S1-75 TP-S1-76 TP-S1-77 TP-S1-78 TP-S1-79 TP-S1-80 TP-S1-81 TP-S1-82 TP-S1-83 TP-S1-84 TP-S1-85 TP-S1-86 TP-S1-87

-H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H -H

2-Cl 2-OCH3 2-Br 3-Cl 3-Br 3-OCH3 3, 3-CH3 3-CF3 3-F 3-CH3 4-Cl 4-F 4-Br 4-CH3 4-N(CH3)2 4-OCH3 4-CF3 3, 4, 5-OCH3 3, 4-OCH3 2, 4-Cl

101.62 ± 1.01 76.15 ± 0.11 6.09 ± 1.91 54.81 ± 0.06 50.12 ± 8.93 97.84 ± 0.89 48.67 ± 3.44 8.26 ± 1.04 37.05 ± 2.77 76.78 ± 3.12 34.24 ± 5.35 96.69 ± 3.22 45.98 ± 0.61 99.74 ± 4.11 91.61 ± 2.14 48.86 ± 1.46 20.03 ± 2.96 97.15 ± 4.78 98.6 ± 1.54 92.9 ± 6.09

3.650 ± 0.35 30.88 ± 1.29 NT b 26.25 ± 1.47 46.50 ± 2.14 0.177 ± 0.049 49.63 ± 10.85 NT b 55.90 ± 41.69 32.75 ± 0.39 NT b 0.436 ± 0.057 9.029 ± 1.786 0.301 ± 0.03 0.464 ± 0.04 35.72 ± 3.48 NT b 0.159 ± 0.006 0.283 ± 0.039 11.70 ± 1.52

All values are the average of n ≥ 2 ± standard deviation. bNT, not tested.

678 679 680 681

25

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Table 3. TIE-2 Kinase Inhibitory Activity @ 5 µg/mL and Half-Maximal Inhibitory Concentrations (IC50) for TP-S1-88 ~ 96 Analogues with R3 Phenyl Modifications

a

Compd

R3

Kinase Inhibition (%) @ 5 µg/mL (±SD) a

IC50, µM (±SD) a

TP-S1-67 TP-S1-88 TP-S1-89 TP-S1-90 TP-S1-91 TP-S1-92 TP-S1-93 TP-S1-94 TP-S1-95 TP-S1-96

-H 2-OCH3 2-Br 3-Cl 3-F 3-CH3 4-N(CH3)2 4-OCH3 4-CH3 4-CF3

89.63 ± 2.24 87.45 ± 4.77 -169.15 ± 10.55 100.2 ± 1.59 52.21 ± 1.55 99.15 ± 0.38 94.09 ± 0.92 52.93 ± 1.9 77.64 ± 9.08 67.74 ± 1.45

6.739 ± 0.736 2.691 ± 0.161 NT b 0.149 ± 0.013 29.60 ± 2.88 2.589 ± 0.204 6.548 ± 0.246 9.393 ± 1.095 2.226 ± 0.112 30.05 ± 1.379

All values are the average of n ≥ 2 ± standard deviation. bNT, not tested.

682 683

26

ACS Paragon Plus Environment

Page 26 of 32

Page 27 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

684 685

Figure 1

686 687

27

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

688 689

Figure 2

690 691

28

ACS Paragon Plus Environment

Page 28 of 32

Page 29 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

692 693

Figure 3

694 695

696 697

Figure 4

698

29

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

699 700

Figure 5

701

30

ACS Paragon Plus Environment

Page 30 of 32

Page 31 of 32

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Chemical Information and Modeling

702 703

Figure 6

704

31

ACS Paragon Plus Environment

Journal of Chemical Information and Modeling

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

705 706

Figure 7

707

32

ACS Paragon Plus Environment

Page 32 of 32