Using in Vitro and in Vivo Models To Evaluate the Oral Bioavailability

Jan 23, 2015 - Once transported into the portal system, the absorbed component is then ... Variations in body weight, organ size, hepatic and renal bl...
0 downloads 0 Views 712KB Size
Subscriber access provided by SELCUK UNIV

Perspective

Using in vitro and in vivo models to evaluate the oral bioavailability of nutraceuticals Yuwen Ting, Qin Zhao, Chunxin Xia, and Qingrong Huang J. Agric. Food Chem., Just Accepted Manuscript • Publication Date (Web): 23 Jan 2015 Downloaded from http://pubs.acs.org on January 28, 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 Agricultural and Food Chemistry 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 27

1 2

Journal of Agricultural and Food Chemistry

Title Header Models to Evaluate Nutraceutical Bioavailability

3 4 5

Using in vitro and in vivo models to evaluate the oral bioavailability of nutraceuticals

6

Yuwen Ting, Qin Zhao, Chunxin Xia and Qingrong Huang*

7

Department of Food Science, Rutgers University, 65 Dudley Road, New Brunswick, New Jersey

8

08901, USA

9 10 11 12 13 14 15 16 17 18 19 20

* To whom correspondence should be addressed. Tel: (848)-932-5514. Fax: (732)-932-6776.

21

Email: [email protected].

22 23

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 2 of 27

24

Abstract. Nutraceuticals are the bioactive compounds found in many dietary sources. Numerous

25

publications reported their ability to prevent the development of degenerative diseases through

26

modulation of the physiological and physiochemical processes in the living system. Having

27

sufficient concentration at the target site of action is the most critical factor for nutraceuticals to

28

deliver the health benefits. For consumers, it is commonly accepted to ingest these bioactive

29

components through oral delivery route since it is convenient, cost-efficient, and allows flexible

30

dosing schedule. Thus, it is important to understand the oral bioavailability of nutraceuticals in

31

order to evaluate their qualifications as disease preventive agents and to calculate the required

32

ingestion dosages. To predict the oral bioavailability of nutraceuticals, many in vitro and in vivo

33

models have been developed to reduce the need of frequent human pharmacokinetic study, which

34

is costly, time-consuming, and involves ethical complications. These models evaluate one or

35

more of the influential factors that contribute to the oral bioavailability, and are efficient

36

screening techniques with the potential of predicting the pharmacokinetic process in human. In

37

order to accurately predict the human oral bioavailability, further research is not only required to

38

develop a better correlation between the in vitro and in vivo models, but also an accurate scaling

39

factor that takes into account of interspecies variations.

40

Key

41

bioaccessibility/ Caco-2/ metabolism/ bioavailability

words:

Nutraceutical/

pharmacokinetics/

TIM-1/

in

42 43 44 45

2

ACS Paragon Plus Environment

vitro

digestion/

lipolysis/

Page 3 of 27

46

Journal of Agricultural and Food Chemistry

Introduction

47

Bioactive compounds, widely found in variety of dietary sources, provide an excellent

48

alternative to medicinal drugs for improving public health. The roles of these compounds to

49

modulate the physiochemical reactions in the biological system were extensively studied. The

50

term, nutraceutical, was then emerged to categorize the dietary compounds that could offer

51

health functionalities in addition to simple satisfaction of the basic nutritional needs for survival

52

of living organisms. Apart from drugs that support immediate relief of diagnosed pathological

53

symptoms, nutraceuticals, on the other hand, are found more effective to prevent the

54

development of degenerative illness, such as cardiovascular diseases and cancer (1, 2). Thus,

55

chronic consumption of nutraceuticals becomes necessary since it is commonly used as disease

56

preventive agent. In this case, oral administration, compare to other delivery routes, is the

57

consumer-preferred method to utilize these bioactive ingredients because it is cheaper, more

58

user-friendly, painless, and allowing flexible dosing schedule.

59

Upon oral administration, the ingested nutraceuticals enter the gastrointestinal (GI) tact.

60

Depending on the chemical structure of ingested compounds, complex physiological and

61

physiochemical environment within the GI tract may variably affect the absorption of bioactives

62

and could eventually prevent it from reaching the system circulation. As most of the

63

nutraceutical merely subject to clinical trials, the biological activities of these bioactive

64

ingredients are frequently characterized using in vitro evidences. According to the in vitro

65

evaluations, the concentration required for nutraceuticals to produce meaningful bioactivities is

66

usually in the micromolar (µM) range (3), which is at least an order of magnitude higher than the

67

plasma concentration (< 1 µM) obtained from normal dietary intake (4). In order to obtain the

68

desirable functionalities, appropriate concentration of the bioactive compound must reach the

3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

69

target site of action. To achieve sufficient system bioavailability, oral dosing concentration of

70

nutraceutical must be selected to accommodate the fraction that will be eliminated by the

71

physiological barriers.

72

The determination of oral bioavailability is then as important as to identify the potential

73

therapeutic uses of neutraceuticals. Even though it is more precise to evaluate oral bioavailability

74

directly using human subjects and clinical testing, it is frequently challenged by the costly

75

consequences involving safety and ethical considerations. Thus, many investigators choose to

76

alternatively study the pharmacokinetic profile of bioactives using animal models. The in vivo

77

animal models provide valuable information to determine the oral bioavailability in a conclusive

78

manner. Unlike the man-made machinery that can be examined step-by-step, in vivo models

79

provide only unilateral end result determining the fraction of ingested dosages that are available

80

to the systemic circulation without providing enough explanation to the events that lead to it.

81

Compared to in vivo pharmacokinetic model, in vitro methods developed to mimic the

82

events occurred after oral administration offer opportunities to study the effect of different

83

physical and chemical parameters that could play critical roles in the systemic availability of

84

bioactive components. In addition, the easy-to-perform in vitro tools are commonly used for

85

rapid prediction on the oral bioavailability of bioactive compounds before performing an in vivo

86

study that are more costly, time-consuming, and may subject to ethical complications. In this

87

perspective, in vitro and in vivo methods for predicting the oral bioavailability of bioactive

88

components will be summarized and compared in relation to their relevance to the living system.

89

Later, the discussion on the potential of using in vitro and in vivo methods for the rapid

90

bioavailability screening of future bioactive ingredient and their related oral formulations will

91

also be included.

4

ACS Paragon Plus Environment

Page 4 of 27

Page 5 of 27

92

Journal of Agricultural and Food Chemistry

Oral bioavailability

93

A sufficient dosage at the target site of action is critical for nutraceutical to generate the

94

desirable biological activities. That is, until compound concentration becomes greater than the

95

threshold dose, there could be no desirable health promoting effect found. To obtain the

96

appropriate concentration at target site, it is important to determine the bioavailability of the

97

bioactive compounds. Bioavailability is a principal pharmacokinetic term describing the fraction

98

of the administered dose reaches the system circulation without being altered. As system

99

concentration obtained from intravenous injection was defined as 100% bioavailability,

100

nutraceuticals administered through other delivery routes generally present lower bioavailability

101

with incomplete absorption due to the physiological or physiochemical barriers.

102

In specific for the system concentration obtain after oral administration, oral

103

bioavailability is greatly depend on the ability of nutraceuticals to survive the structural

104

alteration from pre-absorption degradation and post-absorption metabolism. The pre-absorption

105

events such as pH instability, ionic interaction, enzymatic degradation, and rapid GI elimination

106

rate could significantly decrease the amount of ingested compound available for the gut wall

107

absorption. Once taken up by the intestinal enterocytes, the bioactive compound is then subjected

108

to extensive structural transformation first by intestinal and then by hepatic metabolism. Taking

109

into the account of all events occurred after oral administration (Figure 1), oral bioavailability

110

(Foral) can be summarized and defined by following equation(5, 6).

111

 =  ×  × 

Eq. 1

112

In this mathematical expression, FB is defined as the fraction of ingested dose that

113

become accessible for intestinal absorption. The second component of the equation, FT, describes

114

the fraction of unchanged compound being transported into the portal system. The third

5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

115

component to determine the oral bioavailability, FM, outlined the fraction of ingested dose that

116

survive the metabolic activities as it travels to various systemic organs, such as liver, heart, and

117

lung. Hence, unlike intravenous injection that all applied dose reaches the system circulation, the

118

system bioavailability of orally ingested nutraceutical may be reduced to various degree

119

depending on their specific chemical structure that influences the abovementioned

120

physiochemical parameters.

121

In vitro models for predicting oral bioavailability

122

Oral bioavailability is positively correlated with the bioaccessbility for GI absorption (FB),

123

the gut wall transport coefficient (FT), and the resistibility to the systemic metabolism (FM). Base

124

on the aforementioned theory, in vitro and in vivo models developed for predicting the oral

125

bioavailability of nutraceuticals generally focus on evaluating one or more of the factors.

126

Bioaccessibility. Once ingested from mouth, dietary components enter the GI tract, which is

127

essentially a compartment separating from other interior organs and circulating system with a

128

semipermeable barrier. To get to the inner system, nutraceutical must be first absorbed by the

129

enterocytes. Thus, it is most critical that these bioactives are available at the absorption contact

130

area. The bioaccessibility of a nutraceuitcal is then determined by quantifying the amount of

131

ingested dose reaches the absorption site with its intact structure. GI tract, specialized for the

132

digestion, consists of several sections that each has a distinct physical and physiochemical

133

environment responsible for the breakdown of diverse dietary components.

134

Nutraceutical, just as all other dietary ingredients, go through the entire GI transition

135

process, during which it may be chemically degraded and/or enzymatically metabolized to the

136

product structure with totally different chemical property. In other words, nutraceutical must

137

survive all the degradation activities within the GI tract while being solubilized, or at least

6

ACS Paragon Plus Environment

Page 6 of 27

Page 7 of 27

Journal of Agricultural and Food Chemistry

138

dispersed, in the aqueous lumen in order to become bioaccessible for absorption. To determine

139

the bioaccessibility of a bioactive component, numbers of in vitro models, from simple to

140

complex, are designed based on the separable nature of GI tract. Namely, effect of pH, ionic

141

interaction, enzyme activities, temperature, and luminal solubility on the compound

142

bioaccessibility can be studied individually or systematically using suitable tests.

143

Chemical tests evaluating the stability of nutraceutical at physiological relevant ranges of

144

temperature (37°C), pH (2 - 7.5), and ionic composition are commonly performed methodologies

145

to initially assess the oral dosage efficiency(7-14). However, the events occurs in the actual

146

human GI tract is a rather complex biochemical process than just few simple chemical reactions.

147

Enzyme activities not only directly cause the structural change, such as bond cleavage and sugar

148

moiety substitution, but also interfere with the absorption kinetic when other dietary components,

149

such as lipid, protein, and starch are present (10, 15-18).

150

As an example, the absorption of hydrophobic compound is significantly enhanced when

151

lipid is present due to the improved aqueous solubility after being incorporated into the mixed

152

micelle, which is the vehicle-like structure formed after lipid digestion (5, 19-21). In vitro pH-

153

stat model simulating the digestion environment in the small intestine, the major site for lipid

154

digestion, is an especially suitable analytical tool to evaluate the change in bioaccessibility for

155

hydrophobic compounds when lipid is ingested either as part of the food matrix or oral

156

formulations(22, 23). The single-factored in vitro chemical models is preferred by many

157

investigators for initial screening of dosing efficiency since it is relatively simple to perform,

158

cost-effective, rapid, and allow large sample through-put.

159

In addition to methodologies that contain only single factor, multi-factored models that

160

combine two or more bioaccessibility-influential factors are also available (Figure 2). For

7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

161

example, the in vitro digestion model may simulate the physiochemical condition first in the

162

stomach and then the small intestine(24). However, the in vitro models discussed above,

163

regardless of number of factor addressed, are considered as static model due to their inability to

164

reproduce the dynamic in vivo physical conditions including the peristalsis motion of GI tract,

165

integration of all the influential physiological factors, transitional change in physiological

166

environment, and elimination from the absorption site. Consequently, the bioaccessibility

167

predicted by static models usually overestimate the oral efficiency (25).

168

To take the dynamic factor into account, a sophisticated instrument, TIM-1

169

gastrointestinal simulation system, featuring a continuous in vitro digestion process was

170

developed by TNO quality of life, The Netherlands. Physiological and physiochemical factors,

171

such as body temperature, peristalsis movement, gastric and intestinal residence time, pH,

172

luminal composition, and elimination, are closely approximated by an automated computer

173

system(26). The bioaccessible fraction is determined by continuous sampling from jejunum and

174

ileum sections over a pre-determined time period(25, 27). Another advantage to use an open-

175

compartment system, in which the unabsorbed portion is eliminated and collected as efflux,

176

allows estimation of loss due to degradation since it can be quantified by subtracting the total

177

recovered portion from the original input dosage. TIM system, by far, is the most sophisticated

178

and precise instrument for bioaccessiblity estimation of dietary components.

179

Transport coefficient. To travel across the intestinal lining, nutraceuticals may undertake either

180

the transcellular or paracellur route. When transcellular route is assumed, nutraceutical from the

181

gastric lumen is first absorbed into the enterocytes through either passive diffusion that is driven

182

by concentration gradient or active transportation through the transcellular protein channels.

183

During the in-and-out passage through the enterocytes, cellular metabolism significantly reduces

8

ACS Paragon Plus Environment

Page 8 of 27

Page 9 of 27

Journal of Agricultural and Food Chemistry

184

the fraction of intact compound being release into the portal circulation (28). Apart from the

185

transcellular route, small molecules (Mw < 200) permeate through the membrane paracellularly

186

through tight junction and do not go through the enterocytes. Nutraceuticals utilized paracellular

187

route escaped from the intestinal metabolism and retain the aglycone structure when streaming

188

into the portal blood (28, 29).

189

Hence, the rate of compound transported from GI tract into the portal system is depended

190

on its permeability to the cellular membrane and resistibility to intestinal metabolism. Unlike

191

bioaccessiblity is determined majorly by the physiochemical events in the GI lumen and can be

192

easily simulated using in vitro chemical studies, transport coefficient cannot be characterized

193

using only the chemical experiment. Gastrointestinal model that include a dialysis step after the

194

in vitro digestion is the most simplified in vitro experiment for measuring the membrane

195

transport rate of dissolved compound(30). Since no biological system was involved, dialysis

196

method that allows permeation of small molecules over concentration gradient can only be used

197

for estimating the fraction of compound that is transported through the paracellualr route.

198

The intestinal walls composed of single layer of epithelial cells forms the rate-limiting

199

barrier to the transport of bioactive compounds. Cell culture model (Figure 3) of properly

200

differentiated Caco-2 human colon carcinoma cell into phenotype with the characteristic

201

physiochemical functions of the enterocytes is the commonly used in vitro method for

202

elucidating the transport mechanism of nutraceutical (31-35). Caco-2 monolayer allows effective

203

in vitro prediction of compound permeability and absorption while taking into account all the

204

profound physical, chemical, and biological events during the intestinal transportation(36).

205

Moreover, Caco-2 monolayer model is also a well-developed model to effectively evaluate the

206

lymphatic transport (alterative transport route into the system circulation) of lipid-soluble

9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

207

compounds when included in lipid-based oral formulations(37-39). However, the transport

208

studies by Caco-2 monolayer still have numbers of disadvantages since the consistency of this

209

study is highly depended on the quality of the monolayer. Factors such as cell condition, passage

210

number, length of cultivation, circumstance under which the study is performed, and the

211

transport unit varies among laboratories(40). Thus, the absolute transport coefficient value

212

generated by different laboratories deviate accordingly and could not be compared directly.

213

Moreover, the fact that Caco-2 is human colon carcinoma cell with different physiological

214

characteristics than the normal intestinal cell, such as lacking mucin, biofilm, and other epithelial

215

cell types, can also cause unrealistic prediction of transport coefficient value in human(24).

Page 10 of 27

216 217

In addition to the cell culture model, single-pass intestinal perfusion (SPIP) is an in situ animal

218

model that provides enhanced approximation of intestinal physiology and allows the study of

219

difference in the absorption behavior at various intestinal regions (41). The experimental

220

parameters, such as the compound concentration and the flow rate through the perfused section,

221

are closely controlled to ensure the intra- and inter- experimental comparability. In this model,

222

the compound permeation efficiency is quantified as the concentration difference between the

223

inlet and outlet of perfusate normalized by volume decrease due to water absorption(42).

224

However, permeation rate quantified by the lost of compound in the perfusate retain a

225

shortcoming of being unable to differentiate between the metabolized compound and the

226

unchanged fraction, which is the main parameter for calculating the transport coefficient.

227

Resistibility to systemic metabolism. Once being transported into the portal system, absorbed

228

component is then conveyed to liver where they will be facing the rigorous first-pass metabolic

229

activities. Due to the high density of the metabolic related cytochromes and enzymes, liver is the

10

ACS Paragon Plus Environment

Page 11 of 27

Journal of Agricultural and Food Chemistry

230

principal facility for biotransformation of xenobiotic and is thought to be the one of major rate-

231

limiting factors that reduce the system bioavailability of orally ingested nutraceuticals (43).

232

Cytochrome P-450 (CYPs) represent a family of membrane associated proteins that is variably

233

expressed in different organ tissues (44, 45) and is responsible for the metabolism of more than

234

75% of endogenous compounds (46). The major biotransformation of nutraceuticals by CYP450

235

monooxygenase includes oxidation, hydroxylation and demethylation (47, 48). The isozymes

236

mainly involve in the metabolism of bioactive compounds are CYP1A1, CYP2B, and CYP3A

237

(43, 49, 50).

238

As CYPs is most abundantly expressed in liver, isolated liver microsomes are usually

239

employed in the in vitro metabolic model that characterizes the biotransformation of

240

nutraceutical. During the metabolic test, compounds are incubated with liver microsome and the

241

resulting product composition is usually identified by selected analytical instrument. In vitro

242

metabolism model is effective method to evaluate the metabolic stability of nutraceuticals as it

243

allow rapid, low-cost, and large through-put sampling. However, the use of animal liver

244

microsome to predict human metabolite composition is somewhat unreliable since the expression

245

of CYPs is variable among animal species and is also highly dependent on the incubation and

246

sampling interval.

247

In vivo pharmacokinetic model and correlation to human oral bioavailability

248

The bioavailability of orally ingested compound is the product of a complex process that

249

integrates series of physiological or physiochemical reactions. In vitro methods designed to

250

simulate an aspect of physiochemical process are useful tools for characterization and prediction

251

of the in vivo pharmacokinetics in early screening of bioactive components. Since the

252

bioavailability is affected by multiple factors, in vitro tests conducted in isolation usually present

11

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 12 of 27

253

inadequate in vitro-in vivo correlations (IVIVC). Using the combinations of two or more in vitro

254

models, the precision of estimating the oral bioavailability will increase since more influential

255

factors are addressed. However, even the TIM-1 system being the most complex and closest

256

simulation of pre-absorption events in the human GI tract still cannot perfectly predict the oral

257

bioavailability due the lacking of biological factors, such as enterocytes absorption, systemic

258

metabolism, and volumes of body distribution. Thus, even though in vivo pharmacokinetic study

259

is time consuming, costly, and sometimes inhumane, it is still considered as the best described

260

technique to predict human oral bioavailability. Since completely reproducing the

261

pharmacokinetic process using in vitro model is overly complicated and unrealistic, in vivo

262

animal model offers a living system imitating the full dynamic physiological and physiochemical

263

events during the absorption, distribution, metabolism, and elimination of orally ingested

264

nutraceuticals. Even though the mechanism that underlines the pharmacokinetic process cannot

265

be fully explained, analyzing the timed serum profile from the in vivo studies provide useful

266

information on the oral bioavailability of ingested compounds as well as the metabolite profile.

267

Although animal data, especially pharmacokinetic profile of rodents, have been widely

268

used to predict oral bioavailability in human, the correlation in the absorption of bioactive

269

compounds between animals and human has not been extensively studied. The variations in the

270

body weight, organ size, hepatic and renal blood flow, metabolism, distribution, and elimination

271

rate result in significantly different pharmacokinetic profiles among different animal species. As

272

an example, the pharmacokinetic parameters, such as peak concentration time (Tmax), Peak serum

273

concentration (Cmax), total area under curve (AUC), and urinary excretion, of quercetin deviate

274

accordingly when different in vivo models were used (table 1). Developed according to the body

275

weight and physiological parameters, allometric scaling was applied to in vivo animal data to

12

ACS Paragon Plus Environment

Page 13 of 27

Journal of Agricultural and Food Chemistry

276

better reflect the actual pharmacokinetic process in human(51, 52). However, due to the

277

complexity of physiological events in the living system, methods or scaling to 100% correlate

278

the in vivo data to human oral bioavailability is still not available (53).

279

Besides physiological factors of modeling systems, the pharmacokinetic profile is also

280

significantly affected when different experimental designs were applied. For oral ingestion, the

281

most influential parameter to the bioavailability is the physical and chemical characteristic of

282

dosing formulation. By using different dosing media, the absorption of nutraceuticals can

283

deviate appreciably due to change in the bioaccessibility, a positively correlated parameter to the

284

system bioavailability. In particular for hydrophobic compound, oral absorption is improved

285

when its solubility, the important factor for compound to be available for gut wall absorption,

286

become higher in the dosing medium (54-56). In support of this idea, Piskula et al. found the

287

oral bioavailability of quercetin was highest when solubilized in 100% propylene glycol,

288

followed by 25% propylene glycol solution and then water (55). Inspired by the finding that

289

better bioaccessibility leads to higher absorption rate, various oral delivery systems was designed

290

to improve the pharmacokinetic profile of nutraceuticals with diverse chemical properties(5, 6).

291

As an example, the oral bioavailability of hydrophobic compound, such as curcumin (table 2), is

292

improved by the oral delivery system that enhanced its aqueous solubility. On the other hand,

293

delivery systems that could enhance the luminal stability and membrane permeability will lead to

294

better oral bioavailability of EGCG, a hydrophilic compound which could be rapidly degraded in

295

the GI tract (Table 3).

296

Due to the lack of consensus in the selection of animal models, dosing formulation, and

297

delivery system, the in vivo pharmacokinetic data published by different investigators can rarely

298

be comparable. Thus, the prediction of human oral bioavailability is difficult without systematic

13

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 14 of 27

299

collation of available pharmacokinetic data. Despite of being the best pre-clinical model to

300

evaluate the oral bioavailability of bioactive compounds, pharmacokinetic study itself does not

301

provide enough mechanistic information to allow efficient data organization. In this sense, in

302

vitro test should be collectively conducted to study the pharmacokinetic behavior of

303

nutraceuticals.

304

Prediction of human oral bioavailability

305

To prevent the development of degenerative diseases, nutraceuticals are desirable to have

306

good oral dosage efficiency since it is usually consumed either as part of (functional food) or

307

supplementary (dietary supplement) to the diet. Oral bioavailability of bioactive compound, as

308

discussed in earlier sections, is majorly influenced by its bioaccessibility in the GI lumen,

309

transport rate across the gut wall, and resistibility to the systemic metabolism. In order to better

310

understand their contribution to the oral bioavailability individually and conjointly, these

311

parameters could be evaluated by using proper in vitro and in vivo modeling systems since

312

evaluating the pharmacokinetic profile using human subject is expensive and may involve ethical

313

issues. Thus, it is further desirable that these modeling systems could eventually allow accurate

314

prediction of the pharmacokinetic events in human body. Even though in vitro studies

315

conveniently examine the factors that contribute to oral bioavailability, it cannot accurately

316

predict human pharmacokinetic profile without proper correlation to the in vivo system and the

317

application of allometric scaling, which is calculated with regard to the interspecies difference.

318

To take full advantages of the pre-clinical modeling system to realistically predicting human oral

319

bioavailability of nutraceutical, the mathematical expression of oral bioavailability could be re-

320

write as follow:

321

 =  ×  ×  × × 

14

ACS Paragon Plus Environment

Eq. 2

Page 15 of 27

Journal of Agricultural and Food Chemistry

322

The mathematical equation reveals a future perspective, in which the evaluation of

323

human oral bioavailability can be projected using only the in vitro modeling systems. In this

324

equation, the bioaccessibility (FB), transport rate (FT), and resistibility to systemic metabolism

325

(FM) can all be determined with appropriate in vitro studies. The other two components represent

326

the correlation factors of in vitro and in vivo modeling system (CA) and interspecies scaling (CS).

327

To determine CA and CS, a consistent experimental procedure is required when carrying out

328

pharmacokinetic study using both animal and human subjects. The in vivo animal models serve

329

as an important bridging factor that links the in vitro data to the actual event occurring in human

330

body.

331

In order to accurately calculating CA and CS, there are definitely much more work need to

332

done and problems to be solve. However, the benefit from successfully developing such

333

mathematical relationship between in vitro measurement and human pharmacokinetic process is

334

numerous. First of all, the cost and disadvantages of using models involve living creatures could

335

be minimized. Secondly, factors that change the pharmacokinetic process can be easily taken

336

into account by conducting in vitro testing of affected parameters. Third, rapid estimation of

337

required dosage for disease prevention is much more reliable and realistic to the actual value in

338

human subject. Fourth, the effectiveness of oral formulation to enhance bioavailability can be

339

efficiently determined without going through complicated evaluations. Overall, integration of

340

modeling systems covering all aspect of physiological and physiochemical reactions that

341

contribute to the oral bioavailability of nutraceticals offers the possibility to further elucidate

342

human pharmacokinetic process with much more simplified and efficient procedure.

343

15

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389

Page 16 of 27

References 1. Tewfik, S.; Tewfik, I., Nutraceuticals, functional foods and botanical dietary supplements; promote wellbeing and underpin public health. World Rev. Sci. Technol. Sustain. Dev. 2008, 5, (2), 104-123. 2. Go, V. L. W.; Harris, D. M.; Srihari, P., Global Overview of the Role of Nutraceuticals in Cancer. In Sarkar, F. H., Ed. Springer Netherlands: 2012; pp 1-10. 3. Scheepens, A.; Tan, K.; Paxton, J. W., Improving the oral bioavailability of beneficial polyphenols through designed synergies. Genes Nutr. 2010, 5, (1), 75-87. 4. Scalbert, A.; Williamson, G., Dietary intake and bioavailability of polyphenols. The Journal of nutrition 2000, 130, (8), 2073S-2085S. 5. Ting, Y. W.; Li, S.; Ho, C.-T.; Huang, Q., Emulsion in Oral Delivery of Bioactive Lipophilic Phytochemicals. In Nutrition, Functional and Sensory Properties of Foods, Ho, C.T.; Mussinan, C.; Shahidi, F.; Colitis, E. T., Eds. Royal Society of Chemistry: 2013; Vol. 344, pp 231-242. 6. Ting, Y.; Jiang, Y.; Ho, C.-T.; Huang, Q., Common delivery systems for enhancing in vivo bioavailability and biological efficacy of nutraceuticals. J. Funct. Food 2014, 7, 112128. 7. Zhu, Q. Y.; Zhang, A.; Tsang, D.; Huang, Y.; Chen, Z.-Y., Stability of green tea catechins. J. Agr. Food Chem. 1997, 45, (12), 4624-4628. 8. Zhu, Q. Y.; Holt, R. R.; Lazarus, S. A.; Ensunsa, J. L.; Hammerstone, J. F.; Schmitz, H. H.; Keen, C. L., Stability of the flavan-3-ols epicatechin and catechin and related dimeric procyanidins derived from cocoa. J. Agr. Food Chem. 2002, 50, (6), 1700-1705. 9. Wen, X.; Walle, T., Methylated Flavonoids Have Greatly Improved Intestinal Absorption and Metabolic Stability. Drug Metab. Dispos. 2006, 34, 1786-1792. 10. Wang, X. Y.; Wang, Y.-W.; Huang, Q. R., Enhancing Stability and Bioavailability of Polyphenols using Nanoemulsions. In Micro/Nano Encapsulation of Active Food Ingredients, Huang, Q. R.; Given, P.; Qian, M., Eds. Oxford University Press: 2009; Vol. 1007. 11. Su, Y. L.; Leung, L. K.; Huang, Y.; Chen, Z. Y., Stability of tea theaflavins and catechins. Food Chem. 2003, 83, (2), 189-195. 12. McDougall, G. J.; Fyffe, S.; Dobson, P.; Stewart, D., Anthocyanins from red cabbage– stability to simulated gastrointestinal digestion. Phytochemistry 2007, 68, (9), 1285-1294. 13. Bermudez-Soto, M. J.; Tomas-Barberan, F. A.; Garcia-Conesa, M. T., Stability of polyphenols in chokeberry (Aronia melanocarpa) subjected to in vitro gastric and pancreatic digestion. Food Chem. 2007, 102, (3), 865-874. 14. Bell, L. N., Stability testing of utraceuticals and functional foods. In Handbook of Nutraceuticals and functional foods, Wildman, R. E. C., Ed. CRC Press.: New York, 2001; pp 501-516. 15. Yang, X.; Tian, H.; Ho, C.-T.; Huang, Q., Stability of Citral in Emulsions Coated with Cationic Biopolymer Layers. J. Agr. Food Chem. 2011, 60, (1), 402-409. 16. McClements, D. J.; Decker, E. A.; Park, Y.; Weiss, J., Designing food structure to control stability, digestion, release and absorption of lipophilic food components. Food Biophys. 2008, 3, (2), 219-228. 17. Kim, M. K.; Park, K. S.; Yeo, W. S.; Choo, H.; Chong, Y., In vitro solubility, stability and permeability of novel quercetin-amino acid conjugates. Bioorg. Med. Chem. 2009, 17, (3), 1164-71. 16

ACS Paragon Plus Environment

Page 17 of 27

390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435

Journal of Agricultural and Food Chemistry

18. Cohen, R.; Schwartz, B.; Peri, I.; Shimoni, E., Improving Bioavailability and Stability of Genistein by Complexation with High-Amylose Corn Starch. J. Agr. Food Chem. 2011, 59, (14), 7932-7938. 19. Trevaskis, N. L.; Charman, W. N.; Porter, C. J. H., Lipid-based delivery systems and intestinal lymphatic drug transport: A mechanistic update. Adv. Drug Deliver. Rev. 2008, 60, (6), 702-716. 20. Porter, C. J. H.; Trevaskis, N. L.; Charman, W. N., Lipids and lipid-based formulations: optimizing the oral delivery of lipophilic drugs. Nat. Rev. Drug Discov. 2007, 6, (3), 231-248. 21. Fricker, G.; Kromp, T.; Wendel, A.; Blume, A.; Zirkel, J.; Rebmann, H.; Setzer, C.; Quinkert, R. O.; Martin, F.; Muller-Goymann, C., Phospholipids and Lipid-Based Formulations in Oral Drug Delivery. Pharm. Res. 2010, 27, (8), 1469-1486. 22. McDougall, G. J.; Dobson, P.; Smith, P.; Blake, A.; Stewart, D., Assessing potential bioavailability of raspberry anthocyanins using an in vitro digestion system. J. Agr. Food Chem. 2005, 53, (15), 5896-5904. 23. McClements, D. J.; Li, Y., Review of in vitro digestion models for rapid screening of emulsion-based systems. Food Funct. 2010, 1, (1), 32-59. 24. Failla, M. L.; Huo, T.; Thakkar, S. K., In vitro screening of relative bioaccessibility of carotenoids from foods. Asia Pac. J. Clin. Nutr. 2008, 17, (S1), 200-203. 25. Ting, Y.; Jiang, Y.; Lan, Y.; Xia, C.; Lin, Z.; Roger, M. A.; Huang, Q., Use of Viscoelastic Emulsions to Increase the Oral Bioavailability of Tangeretin. Mol. Nutr. Food Res. Submited. 26. Blanquet, S.; Zeijdner, E.; Beyssac, E.; Meunier, J.-P.; Denis, S.; Havenaar, R.; Alric, M., A dynamic artificial gastrointestinal system for studying the behavior of orally administered drug dosage forms under various physiological conditions. Pharm. Res. 2004, 21, (4), 585-591. 27. Zhao, Q.; Lan, Y.; Xia, C.; Rogers, M. A.; Huang, Q., Comparison of in vitro models in determining bioaccessibility of nutraceuticals: the case of coenzyme Q10. J. Agr. Food Chem. Submited. 28. Lin, J. H.; Chiba, M.; Baillie, T. A., Is the role of the small intestine in first-pass metabolism overemphasized? Pharm. Rev. 1999, 51, (2), 135-158. 29. Pelkonen, O.; Boobis, A. R.; Gundert-Remy, U., In vitro prediction of gastrointestinal absorption and bioavailability: an experts' meeting report. Eur. J. Clin. Pharmacol. 2001, 57, (9), 621-629. 30. Bouayed, J.; Hoffmann, L.; Bohn, T., Total phenolics, flavonoids, anthocyanins and antioxidant activity following simulated gastro-intestinal digestion and dialysis of apple varieties: Bioaccessibility and potential uptake. Food Chem. 2011, 128, (1), 14-21. 31. Zhang, L.; Zheng, Y.; Chow, M. S. S.; Zuo, Z., Investigation of intestinal absorption and disposition of green tea catechins by Caco-2 monolayer model. Int. J. Pharm. 2004, 287, (12), 1-12. 32. Walle, U. K.; Galijatovic, A.; Walle, T., Transport of the flavonoid chrysin and its conjugated metabolites by the human intestinal cell line Caco-2. Biochem. Pharmacol. 1999, 58, (3), 431-438. 33. Konishi, Y.; Kobayashi, S.; Shimizu, M., Transepithelial transport of p-coumaric acid and gallic acid in caco-2 cell monolayers. Biosci. Biotech. Bioch. 2003, 67, (11), 2317-2324. 34. Chabane, M. N.; Al Ahmad, A.; Peluso, J.; Muller, C. D.; Ubeaud, G., Quercetin and naringenin transport across human intestinal Caco-2 cells. J. Pharm. Pharmacol. 2009, 61, (11), 1473-1483. 17

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481

Page 18 of 27

35. Boyer, J.; Brown, D.; Liu, R. H., In vitro digestion and lactase treatment influence uptake of quercetin and quercetin glucoside by the Caco-2 cell monolayer. Nutr. J. 2005, 4. 36. Hubatsch, I.; Ragnarsson, E. G. E.; Artursson, P., Determination of drug permeability and prediction of drug absorption in Caco-2 monolayers. Nat. Protoc. 2007, 2, (9), 21112119. 37. Levy, E.; Mehran, M.; Seidman, E., Caco-2 cells as a model for intestinal lipoprotein synthesis and secretion. FASEB J. 1995, 9, (8), 626-635. 38. Seeballuck, F.; Lawless, E.; Ashford, M. B.; O’Driscoll, C. M., Stimulation of triglyceride-rich lipoprotein secretion by polysorbate 80: in vitro and in vivo correlation using Caco-2 cells and a cannulated rat intestinal lymphatic model. Pharm. Res. 2004, 21, (12), 2320-2326. 39. Simmons, A. L.; Chitchumroonchokchai, C.; Vodovotz, Y.; Failla, M. L., Isoflavone retention during processing, bioaccessibility, and transport by caco-2 cells: effects of source and amount of fat in a soy soft pretzel. J. Agr. Food Chem. 2012, 60, (49), 1219612203. 40. Bailey, C. A.; Bryla, P.; Malick, A. W., The use of the intestinal epithelial cell culture model, Caco-2, in pharmaceutical development. Adv. Drug Deliver. Rev. 1996, 22, (1-2), 85103. 41. Zakeri-Milani, P.; Valizadeh, H.; Tajerzadeh, H.; Azarmi, Y.; Islambolchilar, Z.; Barzegar, S.; Barzegar-Jalali, M., Predicting human intestinal permeability using single-pass intestinal perfusion in rat. J. Pharm. Pharm. Sci. 2007, 10, (3), 368-379. 42. Fagerholm, U.; Johansson, M.; Lennernas, H., Comparison between permeability coefficients in rat and human jejunum. Pharm. Res. 1996, 13, (9), 1336-1342. 43. Wilkinson, G. R., Drug metabolism and variability among patients in drug response. New Engl. J. Med. 2005, 352, (21), 2211-2221. 44. Ding, X. X.; Kaminsky, L. S., Human extrahepatic cytochromes P450: Function in xenobiotic metabolism and tissue-selective chemical toxicity in the respiratory and gastrointestinal tracts. Annu. Rev. Pharmacol. 2003, 43, 149-173. 45. Botto, F.; Seree, E.; Elkhyari, S.; Desousa, G.; Massacrier, A.; Placidi, M.; Cau, P.; Pellet, W.; Rahmani, R.; Barra, Y., Tissue-Specific Expression and Methylation of the Human Cyp2e1 Gene. Biochem. Pharmacol. 1994, 48, (6), 1095-1103. 46. Guengerich, F. P., Cytochrome p450 and chemical toxicology. Chem. Res. Toxicol. 2007, 21, (1), 70-83. 47. Siess, M. H., Leclerc, J., Canivenc-Lavier, M. C., Rat, P., & Suschetet, M. Thanou, J. C. Verhoef and H. E. Junginger, Heterogenous effects of natural flavonoids onmonooxygenase activities in human and rat liver microsomes. . Toxicol. Appl. Pharm. 1995, 130, 73-78. 48. Nielsen, S. E.; Breinholt, V.; Justesen, U.; Cornett, C.; Dragsted, L. O., In vitro biotransformation of flavonoids by rat liver microsomes. Xenobiotica 1998, 28, (4), 389401. 49. Canivenclavier, M. C.; Brunold, C.; Siess, M. H.; Suschetet, M., Evidence for Tangeretin O-Demethylation by Rat and Human Liver-Microsomes. Xenobiotica 1993, 23, (3), 259-266. 50. Bursztyka, J.; Perdu, E.; Tulliez, J.; Debrauwer, L.; Delous, G.; Canlet, C.; De, S. G.; Rahmani, R.; Benfenati, E.; Cravedi, J. P., Comparison of genistein metabolism in rats and humans using liver microsomes and hepatocytes. Food Chem.Toxicol. 2008, 46, (3), 939948. 18

ACS Paragon Plus Environment

Page 19 of 27

482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526

Journal of Agricultural and Food Chemistry

51. Boxenbaum, H., Interspecies Scaling, Allometry, Physiological Time, and the Ground Plan of Pharmacokinetics. J. Pharmacokinet Biop. 1982, 10, (2), 201-225. 52. Dedrick, R.; Bischoff, K. B.; Zaharko, D. S., Interspecies correlation of plasma concentration history of methotrexate (NSC-740). Cancer Chemother. Rep. 1970, 54, (2), 95-101. 53. Obach, R. S.; Baxter, J. G.; Liston, T. E.; Silber, B. M.; Jones, B. C.; MacIntyre, F.; Rance, D. J.; Wastall, P., The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. J. Pharmacol. Exp. Ther. 1997, 283, (1), 46-58. 54. Vallejo, F.; Larrosa, M.; Escudero, E.; Zafrilla, M. P.; Cerda, B.; Boza, J.; Garcia-Conesa, M. T.; Espin, J. C.; Tomas-Barberan, F. A., Concentration and Solubility of Flavanones in Orange Beverages Affect Their Bioavailability in Humans. J. Agr. Food Chem. 2010, 58, (10), 6516-6524. 55. Piskula, M. K.; Terao, J., Quercetin's solubility affects its accumulation in rat plasma after oral administration. J. Agr. Food Chem. 1998, 46, (10), 4313-4317. 56. Das, S.; Lin, H. S.; Ho, P. C.; Ng, K. Y., The impact of aqueous solubility and dose on the pharmacokinetic profiles of resveratrol. Pharm. Res. 2008, 25, (11), 2593-2600. 57. Chen, X.; Yin, O. Q.; Zuo, Z.; Chow, M. S., Pharmacokinetics and modeling of quercetin and metabolites. Pharm. Res. 2005, 22, (6), 892-901. 58. Hou, Y.; Chao, P.; Ho, H.; Wen, C.; Hsiu, S., Profound difference in pharmacokinetics between morin and its isomer quercetin in rats. J. Pharm. Pharmacol. 2003, 55, (2), 199203. 59. Reinboth, M.; Wolffram, S.; Abraham, G.; Ungemach, F. R.; Cermak, R., Oral bioavailability of quercetin from different quercetin glycosides in dogs. Brit. J. Nutr. 2010, 104, (2), 198-203. 60. Cermak, R.; Landgraf, S.; Wolffram, S., The bioavailability of quercetin in pigs depends on the glycoside moiety and on dietary factors. J. Nutr. 2003, 133, (9), 2802-2807. 61. Gugler, R.; Leschik, M.; Dengler, H., Disposition of quercetin in man after single oral and intravenous doses. Eur. J. Clin. Pharmacol. 1975, 9, (2-3), 229-234. 62. Goldberg, D. M.; Yan, J.; Soleas, G. J., Absorption of three wine-related polyphenols in three different matrices by healthy subjects. Clin. Biochem. 2003, 36, (1), 79-87. 63. Moon, Y. J.; Wang, L.; DiCenzo, R.; Morris, M. E., Quercetin pharmacokinetics in humans. Biopharm. Drug Dispos. 2008, 29, (4), 205-217. 64. Li, J.; Jiang, Y. Y.; Wen, J.; Fan, G. R.; Wu, Y. T.; Zhang, C., A rapid and simple HPLC method for the determination of curcumin in rat plasma: assay development, validation and application to a pharmacokinetic study of curcumin liposome. Biomed. Chromatogr. 2009, 23, (11), 1201-1207. 65. Liu, A. C.; Lou, H. X.; Zhao, L. X.; Fan, P. H., Validated LC/MS/MS assay for curcumin and tetrahydrocurcumin in rat plasma and application to pharmacokinetic study of phospholipid complex of curcumin. J. Pharmaceut. Biomed. 2006, 40, (3), 720-727. 66. Hu, L. D.; Jia, Y. H.; Niu, F.; Jia, Z.; Yang, X.; Jiao, K. L., Preparation and Enhancement of Oral Bioavailability of Curcumin Using Microemulsions Vehicle. J. Agr. Food Chem. 2012, 60, (29), 7137-7141. 67. Liu, Z. F.; Chiu, M.; Wang, J.; Chen, W.; Yen, W.; Fan-Havard, P.; Yee, L. D.; Chan, K. K., Enhancement of curcumin oral absorption and pharmacokinetics of curcuminoids and curcumin metabolites in mice. Cancer Chemoth. Pharm. 2012, 69, (3), 679-689.

19

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

527 528 529 530 531 532 533 534 535 536 537 538 539 540

Page 20 of 27

68. Kakkar, V.; Singh, S.; Singla, D.; Kaur, I. P., Exploring solid lipid nanoparticles to enhance the oral bioavailability of curcumin. Mol. Nutr. Food Res. 2011, 55, (3), 495-503. 69. Wan, S. X.; Sun, Y. Q.; Qi, X. X.; Tan, F. P., Improved Bioavailability of Poorly WaterSoluble Drug Curcumin in Cellulose Acetate Solid Dispersion. AAPS Pharmscitech 2012, 13, (1), 159-166. 70. Pietta, P.; Simonetti, P.; Gardana, C.; Brusamolino, A.; Morazzoni, P.; Bombardelli, E., Relationship between rate and extent of catechin absorption and plasma antioxidant status. Biochem. Mol. Biol. Int. 1998, 46, (5), 895-903. 71. Dube, A.; Nicolazzo, J. A.; Larson, I., Chitosan nanoparticles enhance the plasma exposure of (-)-epigallocatechin gallate in mice through an enhancement in intestinal stability. Eur. J. Pharm. Sci. 2011, 44, (3), 422-426.

20

ACS Paragon Plus Environment

Page 21 of 27

541 542 543

Journal of Agricultural and Food Chemistry

Table 1: Pharmacokinetic profile different among animal species.

Phenolics

Model

Dose

Quercetin

SD Rats SD Rats

10 mg/kg 50 mg/kg 100 mg/kg 70.6mg/kg 50 mg/kg 50 – 65 mg/kg 10 mg/70 kg 500 mg

Dogs Pigs Human Human Human

Tmax (hr) 0.078 1.2 2 0.5 3

Cmax (µmol/L) 0.69 4.9 9.5 0.23 1.19 < 0.001 0.16 – 0.45 0.05

AUC (µmol/Lhr) 0.2 48.44 80.3 2.59 6.55 0.21

544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575

21

ACS Paragon Plus Environment

Urinary excretion (%) 2.9 – 7.0 1.18

Ref. (57) (58) (59) (60) (61) (62) (63)

Journal of Agricultural and Food Chemistry

576 577 578

Page 22 of 27

Table 2: Effect of oral formulation on the oral bioavailability of curcumin. Results

Ref.

Liposome

Functional Mechanisms solubility

bioavailability

(64)

Phospholipid complex

solubility

bioavailability

(65)

Microemulsion

solubility

bioavailability

(66)

Nanoemulsion

solubility

bioavailability

(67)

Solid lipid nanoparticle

solubility stability

bioavailability

(68)

Solid dispersion (with cellulose acetate)

solubility

bioavailability longer elimination time

(69)

Formulations

579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603

22

ACS Paragon Plus Environment

Page 23 of 27

604 605 606

Journal of Agricultural and Food Chemistry

Table 3: Effect of oral formulation on the oral bioavailability of EGCG. Formulations Phospholipid complex Chitosan-nanoparticle

Functional Mechanisms permeability stability permeability stability paracellular transport

Results

Ref.

bioavailability

(70)

bioavailability

(71)

607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627

23

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 24 of 27

628 629 630 631

Figure 1: Process of orally ingested compound becomes bioavailable and factors that influence

632

the oral bioavailability.

633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654

24

ACS Paragon Plus Environment

Page 25 of 27

Journal of Agricultural and Food Chemistry

655 656 657

Figure 2: Common in vitro methods to evaluate the bioaccessibility of orally ingested

658

nutraceuticals. Single factor model simulate only the digestion activity with small intestine.

659

Multi-factor model simulated the dynamic GI digestion events in both stomach and small

660

intestine.

661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 25

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 26 of 27

685 686 687 688 689

690 691 692 693

Figure 3: Transport coefficient evaluated using Caco-2 monolayer model. Caco-2 monolayer

694

simulated the transport activity of intestinal epithelium utilizing both paracellular and

695

transcellular transport route.

696 697

26

ACS Paragon Plus Environment

Page 27 of 27

698 699 700

Journal of Agricultural and Food Chemistry

TOC

701 702

27

ACS Paragon Plus Environment