• 沒有找到結果。

Traditional Chinese Medicine as Dual Guardians against Hypertension and Cancer?

N/A
N/A
Protected

Academic year: 2021

Share "Traditional Chinese Medicine as Dual Guardians against Hypertension and Cancer?"

Copied!
24
0
0

加載中.... (立即查看全文)

全文

(1)

1

Traditional Chinese Medicine as Dual

Guardians against Hypertension and Cancer?

Weng Ieong Tou1, 2, Calvin Yu-Chian Chen2, 3, 4, 5, 6*

1School of Medicine, China Medical University, Taichung, 40402, Taiwan

2Laboratory of Computational and Systems Biology, China Medical University,

Taichung, 40402, Taiwan.

3Department of Medical Research, China Medical University Hospital, Taichung,

40402, Taiwan.

4Department of Biotechnology, Asia University, Taichung, 41354, Taiwan.

5Department of Biomedical Informatics, Asia University, Taichung, 41354, Taiwan.

6China Medical University Beigang Hospital, Yunlin,65152, Taiwan.

*Corresponding author. Tel.: +886-4-2205-3366 ext. 4124

E-mail address: ycc929@MIT.EDU (C.Y.-C. Chen)

(2)

2

Abstract

This study utilizes the comprehensive traditional Chinese medicine database TCM Database@Taiwan (http://tcm.cmu.edu.tw/) in conjunction with structure-based and ligand-based drug design to identify multi-function Src inhibitors. The three potential TCM candidates identified as having suitable docking conformations and

bioactivity profiles were angeliferulate,

(3R)-2'-hydroxy-3',4'-dimethoxyisoflavan-7-O-beta-D-glucoside(HMID) and 3-[2',6-dihydroxy-5'-(2-propenyl)[1,1'-biphenyl]3-yl]-(E)-2-propenoic acid(3PA).

Molecular dynamics (MD) simulation demonstrated that the TCM candidates have more stable interactions with the cleft and in complex with Src kinase compared to Saracatinib. Angeliferulate and HMID, both originated from Angelica sinensis, not only interact with Lys298 and amino acids from different loops in the cleft, but with Asp407 located on the activation loop. These interactions are important to reduce the opening of the activation loop due to phosphorylation, hence stabilize the Src kinase cleft structure and inhibit activation. The TCM candidates also exhibited high affinity to other cancer-related target proteins (EGFR, HER2, HSP90). Our observations suggest that the TCM candidates might have multi-targeting effects in hypertension and cancer.

Keywords: cancer; Src; traditional Chinese medicine (TCM); docking; QSAR;

molecular dynamics (MD)

(3)

3

Introduction

1

Src-family protein kinases are proto-oncogenes that participate in important 2

physiological functions such as cellular differentiation, mobility, and proliferation.

3

Src, a member of the Src-family protein kinases, functions as a signal protein and is 4

implicated in various diseases. Src is ubiquitously expressed within the body, but 5

distribution levels vary depending on individual tissue and organs (1,2). The role of 6

Src in cancer was initially established by its overexpression in colon (3-7) and 7

breast cancer (8-20) and is now well established. Figure 1 illustrates the major 8

cancer types in which Src plays a significant role, and important substrates and 9

factors involved in each pathway (21-39). The critical role of Src in cancer makes it 10

an attractive target for designing novel cancer therapy.

11

The carboxyl terminal of the Src kinase (CSK) is important in regulating 12

conformation and activity of Src. Under normal inactive conditions, the Src protein 13

is locked as an inward-folding conformation through binding between the 14

phosphorylated Tyr527 and the SH2 domain. However, in mitotic cells, Src 15

becomes activated due to thedephosphorylation of Tyr527 and phosphorylation of 16

Tyr419.

17

From N-terminal to C-terminal, Src is composed of a smaller amino-terminal 18

lobe (residues 270-340) which binds ATP, and a larger carboxyl-terminal lobe 19

(residues 345-523) which binds with substrates. The ATP binding site is also 20

partially located in the larger lobe. By regulating the structure of the alpha-helix, the 21

large lobe can move toward or away from the small lobe, resulting in opening or 22

closing of the cleft between the two lobes. The Src catalytic site is located within 23

the cleft. An open conformation allows the entrance of ATP into the cleft and exit of 24

ADP from the cleft. Drugs that can either interact with the residues (404-432) on the 25

(4)

4

activation loop or inhibit the activation loop from moving away and opening the 26

cleft as a result of Tyr419 phosphorylation can effectively inhibit Src activity.

27

The importance of Src is established in cancer, but increasing studies are also 28

suggesting its possible involvement in hypertension (40).Based on the 29

pathophysiology of hypertension, endogenous ouabain (EO) and mutant α-adducin 30

have been shown to increase binding of Na-K-ATPase with the SH2 domain and 31

phosphorylation of Tyr416, thus activating subsequent 32

Na,K-ATPase-Src-EGFR-ERK1/2 signaling cascade (41-43). As Src kinase is a key 33

protein regulating the activation of downstream pathways, inhibiting Src kinase 34

activation and Tyr416 phosphorylation may be a novel approach to controlling 35

hypertension. Observations showing that lung cancer patients also have the 36

tendency to develop hypertension and other cardiovascular diseases (44) also 37

suggest possible linkage between the two diseases. In view of this, we speculate that 38

Src may be a common factor for the development of hypertension and cancer, and 39

inhibiting Src may have multi-targeting effects for both diseases. Though many Src 40

inhibitors have been developed, limited pharmaceutical effectiveness has been 41

observed. Recently, computational techniques are commonly used to screen or 42

predict potent drugs targeting specific diseases (45-58).In view of this shift in drug 43

design, and considering the vast pharmaceutical potential of traditional Chinese 44

medicine (TCM), we constructed the world’s most comprehensive TCM database 45

TCM Database@Taiwan (http://tcm.cmu.edu.tw/) (59,60) and its accompanying 46

cloud-computing webserver iScreen (61) and integrative computational design 47

portal “integrated SysteMs Biology Associated Research with TCM” (iSMART) 48

(62) to facilitate drug development from TCM. TCM Database@Taiwan has been 49

successfully used to identify lead compounds for a variety of important diseases 50

(5)

5

(63-82). In our previous study, we utilized TCM Database@Taiwan to identify 51

potential Src inhibitors (83). Based on the essential role of Src in cancer, we deemed 52

it important to evaluate the effect of TCM compounds with Src inhibitory potential 53

against other cancer target proteins. This study targets not only Src, but also 54

validates the docking potential of these TCM candidates against established cancer 55

targets EGFR (84,85), HER2 (86,87), and HSP90 (88-91). Since Src is also 56

involved in the pathological mechanisms of EGFR and HER2 (92,93), ourTCM 57

candidates may have potential as multi-targeting inhibitors for different cancers.

58

Materials and Methods

59

Docking and Candidate Screening 60

The protein structure used in this study was downloaded from Protein Data 61

Bank (PDB: 2H8H) (94). The binding site was defined as the space occupied by 62

Saracatinib within the 2H8H crystal structure, and TCM compounds from TCM 63

Database@Taiwan docked and screened. Using Discovery Studio 2.5, the ligands 64

were first passed through Lipinski’s Rule of Five, and then screened for contour of 65

TCM ligands with the Src kinase binding site using LigandFit. LigandFit is a 66

receptor-rigid docking algorithm that uses Monte Carlo simulation to match ligands 67

with designate binding sites on a given protein. Results from docking are ranked by 68

binding energy and ligand similarity to the Saracatinib within the 2H8H crystal 69

structure, and three candidates are selected for further analysis. Each candidate 70

ligand was minimized with smart minimizer setting in Minimization Algorithm 71

under the force field of CHARMm (95), and a maximum of five docking poses were 72

generated. Each generated docking pose was redocked into the protein for a second 73

time, and five poses from each re-dock were generated. The 25 poses for each 74

ligand are then visually compared with that of Saracatinib within the crystal 75

(6)

6

structure, and docking poses that are most similar to Saracatinib were selected.

76

Hydrophobic contacts between the ligands and Src kinase are calculated with 77

LigPlot v.2.2.25(96). The absorption, distribution, metabolism, excretion and 78

toxicity (ADMET) analysis in D.S. 2.5 was used to calculate pharmacology and 79

toxicity of the derivatives in human bodies. To assess the general applicability of the 80

candidates on other cancer target proteins, the top three candidates from screening 81

were further docked into EGFR, HER2, and HSP. Protein structures used for this 82

application spectrum verification were EGFR (PDB: 2ITY) (97) and HSP90 (PDB:

83

3K97) (98). The HER2 model used was adopted from our previous study (72,99) 84

and was built from structures 2ITY and 2J5E (97,100).

85

Bioactivity Prediction by Multiple Linear Regression (MLR) and Support 86

Vector Machine (SVM) Models 87

Linear MLR (101) and nonlinear SVM (102) were used to construct 88

quantitative structure-activity relationship (QSAR) models for predicting the 89

bioactivity of the TCM compounds. A total of 20 Src ligands (103) were randomly 90

separated into a training set of 15 compounds and an external validation test set of 91

five compounds.

92

Prior to constructing QSAR models, genetic function approximation (GFA) 93

(104) was applied to identify representative descriptor sets from the large pool of 94

descriptors generated from the training set. The MLR model was built based on the 95

representative descriptors using MATLAB in the form of equation [1]:

96

n n n

x

pIC = +

1 0

50 α α [1]

97

where α0 is a constant value and αn is the coefficient value of descriptor Xn.

98

The generated MLR model was validated with cross-validation and independent 99

(7)

7

tests and verified by calculating the square correlation coefficients (R2) between 100

predicted and actual pIC50 of the training set.

101

SVM are groups of supervised methods that allow categorization of hard-to 102

separate patterns through the use nonlinear of generalized portrait algorithms (105).

103

The SVM algorithm was extended for regression (SVMR) from its original use for 104

classification through the use of a ε-insensitive loss function (106) with the goal of 105

identifying a function f(x) in which all training points has a maximum deviation ε 106

from experimental values and has a maximum margin (107). A final nonlinear SVM 107

regression giving the modeled property for a pattern x was obtained by introducing 108

Lagrange multipliers and kernels to map input patterns into a higher dimension 109

space, the formula being equation [2]:

110

( )

K

(

x x

)

b

x f

m

i

k i i i

k =

− +

=

+

1

, )

( λ λ

[2]

111

whereλi+i are Lagrange multipliers and K(xi, xk) is the kernel function.

112

Our SVM model was constructed using the LibSVM program(108). Key 113

parameters determining the SVM model fit are C cost, ε, γ, the kernel type, and the 114

corresponding kernel parameters. The kernel selected for training the SVM model 115

was the Gaussian radial basis function kernel equation [3]:

116

( )



 −

= 2

2

exp 2

, σ

y x x

x

K i k [3]

117

Optimum C, ε, γ were determined using the gridregression.py command within 118

LibSVM. Cross-validation was conducted according to default settings in LibSVM.

119

The validated MLR and SVM models to the TCM candidates to predict individual 120

pIC50 values of the compounds.

121

3D-QSAR Modeling and Analysis 122

(8)

8

3D-QSAR methods such as comparative molecular field analysis (CoMFA) 123

(109) and comparative molecular similarity analysis (CoMSIA) (110) are widely 124

used as activity prediction tools in drug design. Since traditional QSAR (MLR and 125

SVM) do not take into account the 3D structure of the compounds, CoMFA and 126

CoMSIA models were constructed to further test the robustness of TCM candidates 127

as ligands with biological activity against Src. The Src ligands from (103) were 128

randomly divided into a training set of 15 compounds and an external validation test 129

set of five compounds.

130

Partial least square (PLS) analysis is a statistical tool used for establishing a 131

linear model describing the correlation between dependent and independent 132

variables and has the advantage of being directly applicable for prediction(111). In 133

this section, CoMFA and CoMSIA descriptors were used as independent variables 134

and pIC50 values were the dependent variables.

135

The cross-validated coefficient, q2, which is calculated by equation [4], was used to 136

evaluate the prediction accuracy:

137

( )

( )

− −

= 2

_ 50 _

50

2 _ 50 _

2 1 50

mean actual

actual predicted

pIC pIC

pIC

q pIC [4]

138

Conventional correlation coefficient r2 and the standard error, SEE, were also 139

computed for each PLS model. Models with the highest q2, r2, and lowest SEE were 140

selected as the optimum CoMFA and CoMSIA model. Once the optimum model is 141

established, relevant compound descriptors are projected into the PLS model to 142

make external predictions on the test set.

143

CoMFA and CoMSIA structure building was performed using the SYBYL 144

program. The CoMFA descriptors steric and electrostatic field energies were 145

(9)

9

calculated by Lennard Jones function (112) and Coulombic function (113), 146

respectively, using the SYBYL default parameters: avan der Waals (vdW) radius of 147

1.52 Å, a C1+ probe atom, grid point spacing of 2Å, and energy cut-off value of 30 148

kcal/mol. The field contributions in CoMSIA, namely steric, electrostatic, 149

hydrophobic, H-bond donor and H-bond acceptor descriptors, were calculated with 150

identicalC1+ probe atom and grid spacing parameters. The probe radius was set at 151

1.0 Å. The default attenuation factor (R) value of 0.3 was used. Column filtering 152

was set at2.0 kcal/mol. The TCM ligands were overlaid against the generated 153

models to evaluate biological activities based on the CoMFA and CoMSIA models.

154

Molecular Dynamics (MD) Simulation 155

To verify the stability of TCM candidates under dynamic conditions, molecular 156

dynamics (MD) simulations were conducted on the Src-candidate complexes using 157

DS2.5. The energy of each complex was minimized with 500 steps each of Steepest 158

Descent and Conjugate Gradient. The system was heated for 50 ps to increase 159

temperature from 50K to 310K and allowed to equilibrate for 200 ps once the target 160

temperature was reached. Canonical ensemble (NVT; constant temperature) was 161

selected for the 40 ns production process and snapshots were taken at 20 ps intervals.

162

Time steps were set at 2fs. Electrostatic interactions (114) were calculated using 163

Particle Mesh Ewald (PME) method. The MD results were used to analyze energy 164

trajectories, H-bond formation and distances, and torsion angles which provide 165

insights to the interaction between TCM candidates and Src kinase.

166

Results and Discussion

167

Docking and Candidate Screening 168

. Table I lists the top ten TCM candidates with the lowest Binding Energy and 169

(10)

10

Ligand Internal Energy. The complete list of top 100 TCM ligands based on Binding 170

Energyand Ligand Internal Energy can be viewed in Supplementary Table I.

171

Considering the ability to form multiple bonds with Src and their chemical structure, 172

Angeliferulate,(3R)-2'-hydroxy-3',4'-dimethoxyisoflavan-7-O-beta-D-glucoside 173

(HMID), and 3-[2',6-dihydroxy-5'-(2-propenyl)[1,1'-biphenyl]3-yl]-(E)-2-propenoic 174

acid (3PA) were selected as our top candidates. The structural scaffolds of the TCM 175

candidates and the control Saracatinib are shown in Figure 2.Significantly lower 176

binding energy and LIE values were estimated for the TCM candidates (Table I).A 177

higher binding energy, such as the case with Saracatinib, implies that the ligand 178

binding with the protein is more unstable. Results for ADMET are shown in 179

Supplementary Figure 1.

180

Figures 3-6 illustrates interactions and Src protein residues that may be of 181

importance for each ligand. Figure 7 highlights amino acids that form hydrophobic 182

contacts with each test ligand. Saractatinib formed two types of interactions with 183

Src. Pi-interaction was formed with the positively charged alkyl group in Lys298 184

(Figure 3A), and hydrophobic interactions were formed with amino acids located on 185

different loops (Figure 7A). The low calculated binding energy and LIE for 186

Angeliferulate (Top 1) could be the result of H-bonds formed with Lys298, Asp351, 187

Asp407 (Figure 4A), and hydrophobic interactions with Val284, Gly347, and 188

Leu396 (Figure 7B). HMID is docked within Src through H-bonds with Leu276, 189

Lys298, and Asp407 (Figure 5A) and hydrophobic interactions with seven amino 190

acids (Figure 7C). These interactions are formed with amino acids either on the 191

(11)

11

activation loop (Ala407, Asp407) or on different loops (Leu276, Ser348), helping to 192

maintain the stability of the cleft. Similarly, 3PA also docked in Src through the 193

formation of H-bonds (Lys298, Ser348, and Asp351) (Figure 6A) and hydrophobic 194

interactions formed with amino acids on different loops (Figure 7D).

195

Lys298 is an amino acid of critical importance in Src activation. Under open 196

conformations, ATP will enter the cleft, bind atLys298, undergo hydrolysis to 197

release a phosphate and exit as ADP. The released phosphate is used to 198

phosphorylate downstream reactions. A ligand that can effectively interact with 199

Lys298and form stable and permanent interactions with neighboring amino acid 200

residues has potential to block ATP from the binding site and inhibit activation of 201

downstream reactions. Whilst Saracatinib formed a pi-interaction with Lys298, lack 202

of H-bonds with other residues renders Saracatinib unstable. Higher stability of 203

TCM candidates over Saracatinib may be likely due to their ability to form H-bonds 204

with multiple amino acids including Lys298. Hydrophobic contacts, through 205

significantly weaker than pi-interactions and H-bonds, may also contribute to 206

stability. In particular, hydrophobic interactions formed by Saracatinib, HMGF, and 207

3PA with Leu276 and Ser348 (located on different amino acid chains) (Figure 7) 208

can increase stability of the cleft. HMID also forms hydrophobic interactions with 209

activation loop amino acids Ala406 and Asp407 which can resist removal of the 210

activation loop from the cleft during Tyr419 activation.

211

Bioactivity Prediction by Multiple Linear Regression (MLR)and Support 212

Vector Machine (SVM) Models 213

The MLR and SVM models were developed with ten structural descriptors 214

identified by GFA, namely ALogP, ES_Sum_dsCH, ES_Sum_aaCH, ES_Sum_sCl, 215

Molecular_Weight, Molecular_SurfaceArea, Molecular_PolarSurfaceArea, 216

(12)

12

Jurs_RPCG, Jurs_WNSA_1, and PMI_X.ALogPis a measurement of molecular 217

hydrophobicity based on Ghose and Crippen’s method (115); ES_Sum_dsCHis 218

theelectrotopological count for carbons with one single bond and one double 219

bond,ES_Sum_aaCH stand for the electrotopological count for carbons with two 220

aromatic bonds; ES_Sum_sCl is related to the electrotopological count of chlorides 221

with a single bond; Molecular_Weight is the sum of atomic weight;

222

Molecular_SurfaceArea and Molecular_PolarSurfaceAreacalculate total surface 223

area and polar surface area, respectively; Jurs_RPCGdescribes relative positive 224

charge; Jurs_WNSA_1describes the total molecular solvent-accessible surface, and 225

PMI_X is a spatial descriptor related to the orientation and conformational rigidity 226

of the ligand.

227

The generated MLR model is expressed as follows and has good prediction ability 228

(R2= 0.8043; Figure 8A):

229

X PMI WNSA

Jurs

RPCG Jurs

ceArea PolarSurfa

Molecular

a SurfaceAre Molecular

Weight Molecular

sCl Sum ES aaCH

Sum ES

dsCH Sum

ES ALogP

pIC

_ 0060

. 0 1 _ _

0050 . 0

_ 3521

. 7 _

0567 . 0

_ 0107

. 0 _

0433 . 0

_ _ 9470 . 33 _

_ 879 . 0

_ _ 2497 . 9 2414

. 0 7 .

50 387

× +

× +

×

×

× +

×

+

×

×

× +

×

=

230

The SVM model constructed using the aforementioned descriptors also 231

generated a prediction model where predicted values were highly correlated to 232

actual observed values (R2=0.937; Figure 8B). The robustness of our models were 233

validated through external validation tests using the test set. Good correlation 234

between observed and predicted pIC50 values were observed for both models.

235

Predicted pIC50values of the TCM candidates and Saracatinib using the MLR 236

and SVM models are listed in Table I. Results suggest that the TCM candidates 237

have good bioactivity towards Src.

238

3D-QSAR Modeling and Analysis 239

(13)

13

Table II shows the results of the CoMFA and CoMSIA models generated 240

through PLS algorithm. PLS statistics led to a CoMFA model in which steric 241

features were the dominant contributing factor. Several CoMSIA model 242

combinations were generated and the model considering electrostatic (E), 243

hydrophobic (H), and H-bond donor (D) properties was selected as the optimum 244

model based on high cross-validation and non-cross validation correlation 245

coefficients (q2=0.482, r2=0.877).

246

To test the predictive capabilities of the models, they were used to predict the 247

pIC50 values of an external test set excluded from the original training set.

248

Bioactivity predictions of the training and test set are listed in Table III.The 249

correlation curves show high square correlation coefficients of R2=0.9721 for 250

CoMFA (Figure 9A) and R2=0.9414 for CoMSIA (Figure 9B), implying models of 251

good prediction power.

252

In Figure 10, the heteroaromatic ring of Saracatinib, the benzene of 253

Angeliferulate, and the hydroxyl group of HMID, and partial regions of the hexane 254

ring in 3PA fall within the steric favor region, but only Angeliferulate forms 255

interaction bonds. All test ligands fall within the regions between Leu276/Asp351 256

and Lys298/Asp407, satisfying the CoMFA steric favoring region located at 257

Leu276/Asp351. However, only TCM candidates have interactions with the amino 258

acids located within the Lys298/Asp407 region. When the CoMSIA contour was 259

superimposed, Lys298 was located within the region where hydrophilic interactions 260

were desirable for bioactivity (white)(Figure 11). The ability of Angeliferulate, 261

HMID and 3PA to form H-bonds at Lys298 contour to this hydrophilic region and 262

most likely contribute to higher bioactivities. The hydroxyl group of Angeliferulate 263

falls adjacent to the electrostatic favoring (orange) region of Leu276/Asp351 264

(14)

14

(Figure 11B). The hydrophobic benzene ring moiety of 3PA is located close to the 265

hydrophobic disfavoring region (white) of Leu276/Asp351 (Figure 11D). Violation 266

of the hydrophobic disfavor region contour matches the lower bioactivity predicted 267

by our SVM/MLR models (Table I).

268

Molecular Dynamics (MD) Simulation 269

RMSDs and total energy trajectories 270

Protein-ligand complex RMSDs, ligand RMSDs, and total energy level 271

changes during MD are shown in Figure 12. Saracatinib has the highest complex, 272

ligand RMSDs, and the highest total energy of the four test compounds. Most 273

notably, the significant increase in ligand RMSD (Figure 12B) indicates high 274

instability. Total energy of the TCM candidates by increasing order 275

were3PA<Angeliferulate<HMID, all of which were lower than that of Saracatinib 276

(Figure 12C). The lower total energy trajectories indicate a more stable state of the 277

ligand-protein complex during dynamic situations.

278

Saracatinib-Src interactions during MD 279

Saracatinib formed the least amount of interactions with Src during docking, a 280

phenomenon also observed during MD (Figure 3A).As indicated by the H-bond 281

distance trajectories (Figure 3B) and H-bond occupancy analysis (Table IV), the 282

only stable H-bonds formed during MD were with Lys298 and Ser348. Saracatinib 283

could not form H-bonds with neighboring residues Leu276, Asp351, and Asp407 284

were the underlying reasons for the inability to form H-bonds.

285

Angeliferulate-Src interactions during MD 286

The primary stabilizing interactions formed between Angeliferulate and Src 287

were at Ser348, Asp351, and Asp407 (Figure 4B). H-bonds with Lys298 were also 288

initially observed, but rotations on Angeliferulate increased the distance and 289

(15)

15

discouraged interactions with Lys298 after 9.86 ns. Distance from Asp351 reduced 290

from 4.47Å to 2.45Å at 7.64ns, enabling the formation of multiple H-bonds. Small 291

fluctuations on the H-bond distanceswithSer348 and Asp407 were observed, but 292

since the distance was greater than the 2.50Å cut-off distance, the H-bonds were 293

presumed to be weaker.

294

HMID-Src interactions during MD 295

A significant directional shift of HMID was observed during MD due to the 296

formation of H-bonds with new amino acid residues (Table IV). Initially, the 297

H-bond with Leu276 during directed HMID towards the small lobe (Figure 5A).

298

During MD, the loss of the Leu276 H-bond and the formation of H-bond with 299

Ser348 (Figure 5B) shifted HMID towards the large lobe (Figure 5A). HMID 300

formed seven H-bonds with Lys298, but as the HZ1, HZ2, and HZ3 on Lys298 301

continuously rotated, only a 50.65% occupancy was recorded. The H-bond with 302

Asp407 observed during docking was stable throughout MD.

303

3PA-Src interactions during MD 304

3PA was primarily stabilized by Lys298 and Asp351 during MD (Table IV).

305

Multiple H-bonds were formed with Lys298. Distance fluctuations recorded in 306

Figure 6B were normal circumstances caused by the constant rotation of the Lys298 307

H atoms. A stable H-bond with occupancies of greater than 99% was maintained 308

with Asp351. Contrary to the docking pose, no high occupancy H-bonds were 309

observed for Ser348.

310

Torsion angle changes during MD 311

More information on bond formation during MD can be explained through 312

torsion angle changes. As shown in Figure 3C, torsion of a and b contribute to the 313

relative spatial angle of the chloride-containing moiety of Saracatinib. The inability 314

(16)

16

of Saracatinib to maintain interactions with Lys298 and Asp407 could be attributed 315

to conformational shifts brought on by these torsions which increase the distance of 316

these residues from Saracatinib. Oxane could not form interactions due to large 317

torsion fluctuations in c and d and its orientation away from the pocket. Piperazine 318

groups could not form H-bonds due to the large torsion changes observed in e, f, g, 319

and h.

320

Angeliferulate was a largely flexible ligand as indicated by the torsion angles 321

(Figure 4C). Large torsion changes at a-d disrupted the ability of the terminal 322

aromatic ring to form stable H-bonds. Torsion angles at e-h also showed that 323

constant fluctuations. Though m and n were more stable, the dynamic outer regions 324

of the cleft limited the methoxy group and hydroxyl groups of m and n from 325

forming H-bonds.

326

Fluctuations of the torsion angles were also observed in HMID, but H-bond 327

formations were rarely affected (Figure 5C). The stability of b, e, and f enabled the 328

formation of stable H-bonds with Lys298 and Asp407 through O17 and O28, 329

respectively. Other recorded fluctuations at a-d, and g did not affect stability of 330

these two H-bonds. Two large torsion changes were observed at h (8.98 ns and 15 331

ns), both of which directly caused H-bond distance changes between H55 and 332

Leu276, Ser348, and Asp351 (Figure 5B).

333

Figure 6C shows that c, which connects the two benzene moieties in 3PA were 334

very stable. Torsions at e, f, g did not fluctuate greatly, resulting in the stable 335

H-bond with Lys298. 3PA remained in a relatively linear conformation until changes 336

observed at d. Nonetheless, aromatic rings remained in a planar state after 22 ns.

337

Discussion 338

Based on the results of docking and MD, Leu276, Lys298, Ser348, Asp351, 339

(17)

17

and Asp407 are important amino acid residues for stability within Src (Table V).

340

MD results generally supported the findings of docking but showed additional 341

H-bond formation. This implies that under dynamic physiological conditions, all 342

ligands tested could form and remain in complex with Src. Comparison between 343

ligands show that the TCM candidates were more stable and had lower total energy 344

than Saracatinib. The higher stability of the TCM candidates can be attributed to the 345

formation of more H-bonds that remain stable throughout MD. Angeliferulate and 346

HMID not only formed H-bond with the ATP binding site Lys298, but also with 347

Asp407. This can effectively limit the phosphorylated Tyr419 from moving away 348

from the cleft, thus exhibiting important characteristics for being potential Src 349

inhibitors.

350

Conclusion

351

This study utilizes computational methods to virtually screen small molecules 352

found in TCM for potential Src inhibitors. Potential of each candidate was validated 353

using structure-based and ligand-based drug design. As shown in Supplementary 354

Video 1, Angeliferulate and HMID have multiple stable interactions with the two 355

Src cleft loops while simultaneously interacting with Asp407, hindering the 356

activation loop from activation. 3PA also exhibits drug-like potential by primarily 357

interacting with Src through via cleft loop amino acids, but may be less potent than 358

its TCM counterparts due to the lack of direct interaction with the activation loop.

359

Considering the aforementioned interactions with Src and high affinity with EGFR, 360

HER2, and HSP90, we suggest that Angeliferulate and HMID which both originate 361

from the TCM Angelica sinensis may have potential as multi-targeting drug leads.

362

Supplementary Materials

363

Supplementary material include the top 100 candidates from screening, ADMET 364

(18)

18

results of the top three TCM candidates, and a video depicting the mode of 365

inhibition of the TCM candidates on Src.

366

Acknowledgements

367

The research was supported by grants from the National Science Council of 368

Taiwan (NSC 100-2325-B-039-001), Committee on Chinese Medicine and 369

Pharmacy (CCMP100-RD-030), China Medical University and Asia University 370

(DMR-101-094). This study is also supported in part by Taiwan Department of 371

Health Clinical Trial and Research Center of Excellence (DOH101-TD-B-111-004) 372

and Taiwan Department of Health Cancer Research Center of Excellence 373

(DOH101-TD-C-111-005). We are grateful to the National Center of 374

High-performance Computing for computer time and facilities and Dr. Su-sen 375

Chang for discussions and technical assistance with the manuscript preparations. We 376

also wish to express thanks to cloud-computing facilities at Asia University.

377

(19)

19

References

378

1. G. Manning, D. B. Whyte, R. Martinez, T. Hunter, and S. Sudarsanam.

379

Science 298, 1912-1934 (2002).

380

2. S. M. Thomas and J. S. Brugge. Annu Rev Cell Dev Biol 13, 513-609 (1997).

381

3. C. Oneyama, E. Morii, D. Okuzaki, Y. Takahashi, J. Ikeda, N. Wakabayashi, 382

H. Akamatsu, M. Tsujimoto, T. Nishida, K. Aozasa, and M. Okada.

383

Oncogene, (Advanced Online Publication) doi: 10.1038/onc.2011.1367 384

(2011).

385

4. J. Huang, L. Yao, R. Xu, H. Wu, M. Wang, B. S. White, D. Shalloway, and 386

X. Zheng. Embo J 30, 3200-3211 (2011).

387

5. G. Swaminathan and C. A. Cartwright. Oncogene, (Advanced Online 388

Publication) doi:10.1038/onc.2011.1242 (2011).

389

6. R. R. Brady, C. J. Loveridge, M. G. Dunlop, and L. A. Stark.

390

Carcinogenesis 32, 1069-1077 (2011).

391

7. D. Gianni, N. Taulet, C. DerMardirossian, and G. M. Bokoch. Mol Biol Cell 392

21, 4287-4298 (2010).

393

8. B. Pohorelic, R. Singh, S. Parkin, K. Koro, A. D. Yang, C. Egan, and A.

394

Magliocco. Breast Cancer Res Tr, (Advanced Online Publication) doi:

395

10.1007/s10549-10011-11753-10542 (2011).

396

9. J. Jian, Q. Yang, and X. Huang. J Biol Chem 286, 35708-35715 (2011).

397

10. A. J. Bernier, J. Zhang, E. Lillehoj, A. R. Shaw, N. Gunasekara, and J. C.

398

Hugh. Mol Cancer 10, 93 (2011).

399

11. V. Ratushny, H. B. Pathak, N. Beeharry, N. Tikhmyanova, F. Xiao, T. Li, S.

400

Litwin, D. C. Connolly, T. J. Yen, L. M. Weiner, A. K. Godwin, and E. A.

401

Golemis. Oncogene, doi: 10.1038/onc.2011.1314 (2011).

402

12. N. Tikhmyanova and E. A. Golemis. PLoS One 6, e22102 (2011).

403

13. T. Miyake and S. J. Parsons. Oncogene, doi: 10.1038/onc.2011.1332 (2011).

404

14. M. E. Irwin, N. Bohin, and J. L. Boerner. Cancer Biol Ther 12, 718-726 405

(2011).

406

15. T. J. Shackleford, Q. Zhang, L. Tian, T. T. Vu, A. L. Korapati, A. M.

407

Baumgartner, X. F. Le, W. S. Liao, and F. X. Claret. Breast Cancer Res 13, 408

R65 (2011).

409

16. J. D. Bjorge, A. S. Pang, M. Funnell, K. Y. Chen, R. Diaz, A. M. Magliocco, 410

and D. J. Fujita. PLoS One 6, e19309 (2011).

411

17. Z. Hou, D. J. Falcone, K. Subbaramaiah, and A. J. Dannenberg.

412

Carcinogenesis 32, 695-702 (2011).

413

18. C. C. Mader, M. Oser, M. A. Magalhaes, J. J. Bravo-Cordero, J. Condeelis, 414

A. J. Koleske, and H. Gil-Henn. Cancer Res 71, 1730-1741 (2011).

415

(20)

20

19. K. K. Haenssen, S. A. Caldwell, K. S. Shahriari, S. R. Jackson, K. A.

416

Whelan, A. J. Klein-Szanto, and M. J. Reginato. J Cell Sci 123, 1373-1382 417

(2010).

418

20. K. E. Reeder-Hayes, L. A. Carey, and W. M. Sikov. Breast Dis 32, 123-136 419

(2010).

420

21. S. Thomas, J. B. Overdevest, M. D. Nitz, P. D. Williams, C. R. Owens, M.

421

Sanchez-Carbayo, H. F. Frierson, M. A. Schwartz, and D. Theodorescu.

422

Cancer Res 71, 832-841 (2011).

423

22. N. Said and D. Theodorescu. Cancer Metastasis Rev 28, 327-333 (2009).

424

23. A. Prinetti, T. Cao, G. Illuzzi, S. Prioni, M. Aureli, N. Gagliano, G. Tredici, 425

V. Rodriguez-Menendez, V. Chigorno, and S. Sonnino. J Biol Chem 47, 426

40900-40910 (2011).

427

24. S. Charoenfuprasert, Y. Y. Yang, Y. C. Lee, K. C. Chao, P. Y. Chu, C. R.

428

Lai, K. F. Hsu, K. C. Chang, Y. C. Chen, L. T. Chen, J. Y. Chang, S. J. Leu, 429

and N. Y. Shih. Oncogene 30, 3570-3584 (2011).

430

25. H. S. Kim, H. D. Han, G. N. Armaiz-Pena, R. L. Stone, E. J. Nam, J. W. Lee, 431

M. M. Shahzad, A. M. Nick, S. J. Lee, J. W. Roh, M. Nishimura, L. S.

432

Mangala, J. Bottsford-Miller, G. E. Gallick, G. Lopez-Berestein, and A. K.

433

Sood. Clin Cancer Res 17, 1713-1721 (2011).

434

26. E. L. Leung, J. C. Wong, M. G. Johlfs, B. K. Tsang, and R. R. Fiscus. Mol 435

Cancer Res 8, 578-591 (2010).

436

27. X. G. Liu, Y. Guo, Z. Q. Yan, M. Y. Guo, Z. G. Zhang, and C. A. Guo.

437

Zhonghua Zhong Liu Za Zhi 33, 340-344 (2011).

438

28. Y. Ding, X. Wang, A. Xu, X. Xu, K. Tian, C. Y. Young, and H. Yuan. J Cell 439

Biochem 112, 818-828 (2011).

440

29. T. Kobayashi, T. Inoue, Y. Shimizu, N. Terada, A. Maeno, Y. Kajita, T.

441

Yamasaki, T. Kamba, Y. Toda, Y. Mikami, T. Yamada, T. Kamoto, O.

442

Ogawa, and E. Nakamura. Mol Endocrinol 24, 722-734 (2010).

443

30. S. Zhang, H. E. Zhau, A. O. Osunkoya, S. Iqbal, X. Yang, S. Fan, Z. Chen, 444

R. Wang, F. F. Marshall, L. W. Chung, and D. Wu. Mol Cancer 9, 9 (2010).

445

31. J. DaSilva, D. Gioeli, M. J. Weber, and S. J. Parsons. Cancer Res 69, 446

7402-7411 (2009).

447

32. G. Pandini, M. Genua, F. Frasca, R. Vigneri, and A. Belfiore. Ann N Y Acad 448

Sci 1155, 263-267 (2009).

449

33. F. Leve, T. G. Marcondes, L. G. Bastos, S. V. Rabello, M. N. Tanaka, and J.

450

A. Morgado-Diaz. Eur J Pharmacol 671, 7-17 (2011).

451

34. L. M. Sturla, P. O. Zinn, K. Ng, M. Nitta, D. Kozono, C. C. Chen, and E. M.

452

Kasper. Brit J Cancer 105, 1235-1243 (2011).

453

(21)

21

35. A. E. Al Moustafa, A. Yasmeen, and A. Achkhar. Med Hypotheses 77, 454

812-814 (2011).

455

36. S. Saini, S. Arora, S. Majid, V. Shahryari, Y. Chen, G. Deng, S. Yamamura, 456

K. Ueno, and R. Dahiya. Cancer Prev Res 4, 1698-1709 (2011).

457

37. X. F. Le, W. Mao, G. He, F. X. Claret, W. Xia, A. A. Ahmed, M. C. Hung, 458

Z. H. Siddik, and R. C. Bast, Jr. J Natl Cancer I 103, 1403-1422 (2011).

459

38. A. Migliaccio, G. Castoria, and F. Auricchio. Methods Mol Biol 776, 460

361-370 (2011).

461

39. J. C. Lee, M. C. Maa, H. S. Yu, J. H. Wang, C. K. Yen, S. T. Wang, Y. J.

462

Chen, Y. Liu, Y. T. Jin, and T. H. Leu. Mol Carcinog 43, 207-214 (2005).

463

40. M. Ferrandi, I. Molinari, L. Torielli, G. Padoani, S. Salardi, M. P. Rastaldi, P.

464

Ferrari, and G. Bianchi. Sci Transl Med 2, 59ra86 (2010).

465

41. P. M. Kearney, M. Whelton, K. Reynolds, P. Muntner, P. K. Whelton, and J.

466

He. Lancet 365, 217-223 (2005).

467

42. P. Ferrari. Biochim Biophys Acta 1802, 1254-1258 (2010).

468

43. P. Ferrari, M. Ferrandi, G. Valentini, and G. Bianchi. Am J Physiol-Reg I 469

290, R529-R535 (2006).

470

44. P. C. Chen, C. H. Muo, Y. T. Lee, Y. H. Yu, and F. C. Sung. Stroke 42, 471

3034-3039 (2011).

472

45. A. P. Guimaraes, A. A. Oliveira, E. F. da Cunha, T. C. Ramalho, and T. C.

473

Franca. J Biomol Struct Dyn 28, 455-469 (2011).

474

46. E. P. Semighini, J. A. Resende, P. de Andrade, P. A. B. Morais, I. Carvalho, 475

C. A. Taft, and C. H. T. P. Silva. J Biomol Struct Dyn 28, 787-796 (2011).

476

47. Y. D. Cai, J. F. He, and L. Lu. J Biomol Struct Dyn 28, 797-804 (2011).

477

48. Z. H. Mei, J. Liu, and H. W. Yu. J Biomol Struct Dyn 28, 871-879 (2011).

478

49. K. C. Chen and C. Y. C. Chen. Soft Matter 7, 4001-4008 (2011).

479

50. H. J. Huang, K. J. Lee, H. W. Yu, H. Y. Chen, F. J. Tsai, and C. Y. C. Chen.

480

J Biomol Struct Dyn 28, 187-200 (2010).

481

51. C. Y. C. Chen. J Biomol Struct Dyn 27, 627-640 (2010).

482

52. K. Bhargavi, P. Kalyan Chaitanya, D. Ramasree, M. Vasavi, D. K. Murthy, 483

and V. Uma. J Biomol Struct Dyn 28, 379-391 (2010).

484

53. M. T. Cambria, D. Di Marino, M. Falconi, S. Garavaglia, and A. Cambria. J 485

Biomol Struct Dyn 27, 501-509 (2010).

486

54. G. Ompraba, D. Velmurugan, P. A. Louis, and Z. A. Rafi. J Biomol Struct 487

Dyn 27, 489-499 (2010).

488

55. E. F. F. da Cunha, E. F. Barbosa, A. A. Oliveira, and T. C. Ramalho. J 489

Biomol Struct Dyn 27, 619-625 (2010).

490

56. C. Y. C. Chen. J Taiwan Inst Chem E 41, 143-149 (2010).

491

(22)

22

57. C. Y. C. Chen. J Taiwan Inst Chem E 40, 55-69 (2009).

492

58. C. Y. C. Chen. J Chin Chem Soc-Taip 54, 653-658 (2007).

493

59. C. Y. C. Chen. PLoS One 6, e15939 (2011).

494

60. K. Sanderson. Nat Med 17, 1531 (2011).

495

61. T. Y. Tsai, K. W. Chang, and C. Y. C. Chen. J Comput Aid Mol Des 25, 496

525-531 (2011).

497

62. K. W. Chang, T. Y. Tsai, K. C. Chen, S. C. Yang, H. J. Huang, T. T. Chang, 498

M. F. Sun, H. Y. Chen, F. J. Tsai, and C. Y. C. Chen. J Biomol Struct Dyn 499

29, 243-250 (2011).

500

63. S. S. Chang, H. J. Huang, and C. Y. C. Chen. PLoS Comput Biol 7, 501

e1002315 (2011).

502

64. S. S. Chang, H. J. Huang, and C. Y. C. Chen. Mol Biosyst 7, 3366-3374 503

(2011).

504

65. K. C. Chen, M. F. Sun, S. C. Yang, S. S. Chang, H. Y. Chen, F. J. Tsai, and 505

C. Y. C. Chen. Chem Biol Drug Des 78, 679-688 (2011).

506

66. T. T. Chang, K. C. Chen, K. W. Chang, H. Y. Chen, F. J. Tsai, M. F. Sun, 507

and C. Y. C. Chen. Mol Biosyst 7, 2702-2710 (2011).

508

67. M. F. Sun, H. Y. Chen, F. J. Tsai, S. H. Lui, and C. Y. C. Chen. J Biomol 509

Struct Dyn 29, 325-337 (2011).

510

68. M. F. Sun, T. T. Chang, K. W. Chang, H. J. Huang, H. Y. Chen, F. J. Tsai, J.

511

G. Lin, and C. Y. C. Chen. J Biomol Struct Dyn 28, 895-906 (2011).

512

69. T. T. Chang, M. F. Sun, H. Y. Chen, F. J. Tsai, M. Fisher, J. G. Lin, and C.

513

Y. C. Chen. J Biomol Struct Dyn 28, 773-786 (2011).

514

70. C. H. Lin, T. T. Chang, M. F. Sun, H. Y. Chen, F. J. Tsai, K. L. Chang, M.

515

Fisher, and C. Y. C. Chen. J Biomol Struct Dyn 28, 471-482 (2011).

516

71. P. C. Chang, J. D. Wang, M. M. Lee, S. S. Chang, T. Y. Tsai, K. W. Chang, 517

F. J. Tsai, and C. Y. C. Chen. J Biomol Struct Dyn 29, 471-483 (2011).

518

72. S. C. Yang, S. S. Chang, and C. Y. C. Chen. PLoS One 6, e28793 (2011).

519

73. S. C. Yang, S. S. Chang, H. Y. Chen, and C. Y. C. Chen. PLoS Comput Biol 520

7, e1002189 (2011).

521

74. T. T. Chang, H. J. Huang, K. J. Lee, H. W. Yu, H. Y. Chen, F. J. Tsai, M. F.

522

Sun, and C. Y. C. Chen. J Biomol Struct Dyn 28, 309-321 (2010).

523

75. C. Y. Chen and C. Y. C. Chen. J Mol Graph Model 29, 21-31 (2010).

524

76. H. J. Huang, K. J. Lee, H. W. Yu, C. H. Hsu, H. Y. Chen , F. J. Tsai, and C.

525

Y. C. Chen. J Biomol Struct Dyn 28, 23-37 (2010).

526

77. C. Y. C. Chen. J Taiwan Inst Chem E 40, 155-161 (2009).

527

78. C. Y. Chen, Y. H. Chang, D. T. Bau, H. J. Huang, F. J. Tsai, C. H. Tsai, and 528

C. Y. C. Chen. Acta Pharmacol Sin 30, 1186-1194 (2009).

529

(23)

23

79. C. Y. C. Chen. J Chin Inst Chem Eng 39, 291-299 (2008).

530

80. C. Y. C. Chen. J Chin Inst Chem Eng 39, 663-671 (2008).

531

81. C. Y. C. Chen, G. W. Chen, and W. Y. C. Chen. J Chin Chem Soc-Taip 55, 532

297-302 (2008).

533

82. C. Y. Chen, H. J. Huang, F. J. Tsai, and C. Y. C. Chen. J Taiwan Inst Chem 534

E 41, 8-15 (2010).

535

83. W. I. Tou and C. Y. C. Chen. PLoS One In press. (2012).

536

84. H. W. Lo, S. C. Hsu, W. Xia, X. Cao, J. Y. Shih, Y. Wei, J. L. Abbruzzese, 537

G. N. Hortobagyi, and M. C. Hung. Cancer Res 67, 9066-9076 (2007).

538

85. H. W. Lo, S. C. Hsu, M. Ali-Seyed, M. Gunduz, W. Xia, Y. Wei, G.

539

Bartholomeusz, J. Y. Shih, and M. C. Hung. Cancer Cell 7, 575-589 (2005).

540

86. M. Casimiro, O. Rodriguez, L. Pootrakul, M. Aventian, N. Lushina, C.

541

Cromelin, G. Ferzli, K. Johnson, S. Fricke, F. Diba, B. Kallakury, C.

542

Ohanyerenwa, M. Chen, M. Ostrowski, M. C. Hung, S. A. Rabbani, R. Datar, 543

R. Cote, R. Pestell, and C. Albanese. Cancer Res 67, 4364-4372 (2007).

544

87. C. Bartholomeusz, H. Itamochi, L. X. Yuan, F. J. Esteva, C. G. Wood, N.

545

Terakawa, M. C. Hung, and N. T. Ueno. Cancer Res 65, 8406-8413 (2005).

546

88. Y. Yufu, J. Nishimura, and H. Nawata. Leukemia Res 16, 597-605 (1992).

547

89. M. Ferrarini, S. Heltai, M. R. Zocchi, and C. Rugarli. Int J Cancer 51, 548

613-619 (1992).

549

90. A. Jameel, M. Law, R. Coombes, and Y. Luqmani. Int J Oncol 2, 1075-1080 550

(1993).

551

91. L. Whitesell, E. G. Mimnaugh, B. De Costa, C. E. Myers, and L. M. Neckers.

552

P Natl Acad Sci U S A 91, 8324-8328 (1994).

553

92. T. H. Leu and M. C. Maa. Front Biosci 8, s28-38 (2003).

554

93. E. M. Poole, K. Curtin, L. Hsu, R. J. Kulmacz, D. J. Duggan, K. W. Makar, 555

L. Xiao, C. S. Carlson, M. L. Slattery, B. J. Caan, J. D. Potter, and C. M.

556

Ulrich. Int J Mol Epidemiol Genet 2, 300-315 (2011).

557

94. L. F. Hennequin, J. Allen, J. Breed, J. Curwen, M. Fennell, T. P. Green, C.

558

Lambert-van der Brempt, R. Morgentin, R. A. Norman, A. Olivier, L.

559

Otterbein, P. A. Ple, N. Warin, and G. Costello. J Med Chem 49, 6465-6488 560

(2006).

561

95. B. R. Brooks, C. L. Brooks, 3rd, A. D. Mackerell, Jr., L. Nilsson, R. J.

562

Petrella, B. Roux, Y. Won, G. Archontis, C. Bartels, S. Boresch, A. Caflisch, 563

L. Caves, Q. Cui, A. R. Dinner, M. Feig, S. Fischer, J. Gao, M. Hodoscek, 564

W. Im, K. Kuczera, T. Lazaridis, J. Ma, V. Ovchinnikov, E. Paci, R. W.

565

Pastor, C. B. Post, J. Z. Pu, M. Schaefer, B. Tidor, R. M. Venable, H. L.

566

Woodcock, X. Wu, W. Yang, D. M. York, and M. Karplus. J Comput Chem 567

(24)

24

30, 1545-1614 (2009).

568

96. R. A. Laskowski and M. B. Swindells. J Chem Inf Model 51, 2778-2786 569

(2011).

570

97. C. H. Yun, T. J. Boggon, Y. Li, M. S. Woo, H. Greulich, M. Meyerson, and 571

M. J. Eck. Cancer Cell 11, 217-227 (2007).

572

98. P. P. Kung, B. Huang, G. Zhang, J. Z. Zhou, J. Wang, J. A. Digits, J.

573

Skaptason, S. Yamazaki, D. Neul, M. Zientek, J. Elleraas, P. Mehta, M. J.

574

Yin, M. J. Hickey, K. S. Gajiwala, C. Rodgers, J. F. Davies, and M. R.

575

Gehring. J Med Chem 53, 499-503 (2010).

576

99. M. F. Sun, S. C. Yang, K. W. Chang, T. Y. Tsai, H. Y. Chen, F. J. Tsai, J. G.

577

Lin, and C. Y. C. Chen. Mol Simulat 37, 884-892 (2011).

578

100. J. A. Blair, D. Rauh, C. Kung, C. H. Yun, Q. W. Fan, H. Rode, C. Zhang, M.

579

J. Eck, W. A. Weiss, and K. M. Shokat. Nat Chem Biol 3, 229-238 (2007).

580

101. B. K. Slinker and S. A. Glantz. Circulation 117, 1732-1737 (2008).

581

102. R. Burbidge, M. Trotter, B. Buxton, and S. Holden. Comput Chem 26, 5-14 582

(2001).

583

103. D. H. Boschelli, D. Wang, Y. Wang, B. Wu, E. E. Honores, A. C. Barrios 584

Sosa, I. Chaudhary, J. Golas, J. Lucas, and F. Boschelli. Bioorg Med Chem 585

Lett 20, 2924-2927 (2010).

586

104. M. Tufail and L. E. Ormsbee. J Hydroinform 8, 193-206 (2006).

587

105. V. Vapnik and A. Lerner. Automat Remote Contr 24, 774-780 (1963).

588

106. V. Vapnik. The Nature of Statistical Learning Theory. (Springer, 1995).

589

107. O. Ivanciuc. Reviews in Computational Chemistry 23, 291-400 (2007).

590

108. C.-C. Chang and C.-J. Lin. ACM TSIT, 2:27:21--27:27 (2011).

591

109. R. D. Cramer, 3rd, D. E. Patterson, and J. D. Bunce. Prog Clin Biol Res 291, 592

161-165 (1989).

593

110. G. Klebe, U. Abraham, and T. Mietzner. J Med Chem 37, 4130-4146 (1994).

594

111. T. Langer and S. D. Bryant. in The Practice of Medicinal Chemistry (ed 595

C. G. Wermuth) Ch. 29, 587-604 (Path Press, 2008).

596

112. R. J. Gillespie, D. Bayles, J. Platts, G. L. Heard, and R. F. W. Bader. J Phys 597

Chem A 102, 3407-3414 (1998).

598

113. G. Tang and B. K. Ma. Chinese Phys 9, 737-741 (2000).

599

114. M. Kawata and M. Mikami. Chem Phys Lett 313, 261-266 (1999).

600

115. A. K. Ghose and G. M. Crippen. J Chem Inf Comput Sci 27, 21-35 (1987).

601 602 603

參考文獻

相關文件

From the aspect of the representational function of poetry, both Kukai’s and Chinese mountain living poems has the tendencies of “ego lyricizing”. and “intercourse dialogue”,

Light rays start from pixels B(s, t) in the background image, interact with the foreground object and finally reach pixel C(x, y) in the recorded image plane. The goal of environment

• Each student might start from a somewhat different point of view and experience the encounters with works of art and ideas in a different way... Postmodern

 Authorized by the State Education Ministry, International College of Traditional Chinese Medicine (ICTCM) was established in 1992 within TUTCM..  It is in TUTCM where

8.2.1 In the 2012 Study, only the enrolment ratio method was used in projecting demand from local students. In the present study, both the enrolment ratio and the grade transition

To help students achieve the curriculum aims and objectives, schools should feel free to vary the organization and teaching sequence of learning elements. In practice, most

Biases in Pricing Continuously Monitored Options with Monte Carlo (continued).. • If all of the sampled prices are below the barrier, this sample path pays max(S(t n ) −

With regards for learner's learning behaviors, traditional adaptive testing research do not focus on learning curve analysis and personality.. For the learning potential and