+ All Categories
Home > Documents > University of Surrey - Designing a bio-inspired bio...

University of Surrey - Designing a bio-inspired bio...

Date post: 23-Jan-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
66
Designing a bio-inspired bio-mimetic in vitro system for the optimisation of ex vivo studies of pancreatic cancer Stella Totti 1 , Spyros I. Vernardis 2 , Lisiane Meira 3 , Pedro A. Pérez-Mancera 4 , Eithne Costello 4,5 , William Greenhalf 5 , Daniel Palmer 4 , John Neoptolemos 4,5 , Athanasios Mantalaris 2 , Eirini. G. Velliou 1,* 1 Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford, GU2 7XH, UK 2 Biological Systems Engineering Laboratory (BSEL), Department of Chemical Engineering, Imperial College London, SW7 2AZ, London, UK 3 Department of Clinical and Experimental Medicine, University of Surrey, Guildford, GU2 7XH, UK 4 Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Daulby Street, Liverpool L693GA, UK 5 NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Daulby Street, Liverpool L69 3GA, UK 1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23
Transcript
Page 1: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

Designing a bio-inspired bio-mimetic in vitro system

for the optimisation of ex vivo studies of pancreatic cancer

Stella Totti1, Spyros I. Vernardis2, Lisiane Meira3, Pedro A. Pérez-Mancera4, Eithne

Costello4,5, William Greenhalf5, Daniel Palmer4, John Neoptolemos4,5, Athanasios Mantalaris2,

Eirini. G. Velliou1,*

1 Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and

Process Engineering, University of Surrey, Guildford, GU2 7XH, UK

2Biological Systems Engineering Laboratory (BSEL), Department of Chemical Engineering, Imperial

College London, SW7 2AZ, London, UK

3Department of Clinical and Experimental Medicine, University of Surrey, Guildford, GU2 7XH, UK

4Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Daulby Street,

Liverpool L693GA, UK

5NIHR Liverpool Pancreas Biomedical Research Unit, University of Liverpool, Daulby Street,

Liverpool L69 3GA, UK

*Corresponding author. Fax: 0044-(0)-1483686577

E-mail address: [email protected]

1

1

2

3

4

5

6

7

8

9

10

11

12

13

14

15

16

17

18

19

20

21

22

23

24

25

26

27

Page 2: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

Abstract

Pancreatic cancer is one of the most aggressive and lethal human malignancies. Drug

therapies and radiotherapy are used for treatment as adjuvants to surgery, but

outcomes remain disappointing. Advances in tissue engineering point that three-

dimensional cultures can reflect the in vivo tumour micro-environment and can

guarantee a physiological distribution of oxygen, nutrients and drugs, therefore, being

promising low cost tools for therapy development. In this work we review crucial

elements, i.e., structural and environmental, that should be considered for an accurate

design of an ex vivo platform for studies of pancreatic cancer. Furthermore, we

propose environmental stress response biomarkers as platform readouts for the

efficient control and further prediction of the pancreatic cancer response to the

environmental and treatment input.

Keywords

Pancreatic cancer, tissue engineering, 3D culture systems, stress biomarkers,

environmental stress, hypoxia, metabolic stress, metabolomics

2

28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

45

46

47

48

49

50

51

52

Page 3: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

1. Introduction

Pancreatic cancer is the fourth leading cause of all cancer-related deaths in the United

States [1] fifth in the UK (see: http://www.cancerresearchuk.org) and eighth

worldwide [2]. Moreover, despite the fact that for most cancers the survival rate has been

increasing, the 5-year survival rate of pancreatic cancer remains persistently low at 3-6% [1]

(see: http://www.nhs.uk). This dismal outcome can be attributed to many factors such

as i) late stage diagnosis due to lack of early diagnostic biomarkers [3,4], ii) high

metastasis likelihood [5,6], iii) resistance to treatment [7]. Understanding the

pancreatic tumour growth/evolution as well as response to treatment is crucial in order

to improve treatment efficacy both for the benefit of society and individual patients.

Animal studies can be very informative, however they are expensive and complex to

reproduce. In vitro studies of cancer so far, are mainly conducted in traditional 2D

monolayer systems. Those systems can provide useful information, but usually they

lack predictability as they differ significantly from the in vivo tumour environment

[8,9]. Thus, in order to accurately understand cellular and tumour behaviour and

response to treatment, it is essential to simulate ex vivo, the in vivo tumour

environment. The latter, is the main aim of 3D tissue engineering constructs. These

3D systems better reflect the in vivo scenario in terms of structure, porosity, and

microenvironmental niches than the conventional two dimensional (2D) systems [9-

12]. Their spatial arrangement provides better cell-cell interactions while coating with

different extracellular matrix proteins (ECM) ensures better cell-matrix interactions

[13,14]. Co-cultivation with stromal cells, additionally to the above characteristics can

lead us one step closer to the accurate recapitulation of the in vivo tumour

3

53

54

55

56

57

58

59

60

61

62

63

64

65

66

67

68

69

70

71

72

73

74

75

76

77

Page 4: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

microenvironment and realistic stromal interactions [15]. Therefore, growing patient

derived samples outside of the patient body in appropriate 3D tissue engineered

platforms could lead to better understanding the tumour behaviour and response to

drug and irradiation therapies [16].

Next to the actual architectural and micro-environmental characteristics of a tumour in

terms of structure, environmental conditions such as oxygen, glucose, nutrients,

temperature are of significant importance for the cancer kinetics. Fluctuations of those

environmental parameters naturally occur in patients with cancer and could alter

tumour proliferation and/or cause adaptation of the pancreatic tumour cells to

treatment [17-21].

The aim of this work is to review crucial elements, i.e., structural and environmental,

that should be considered for an accurate design of an ex vivo platform for studies of

pancreatic cancer. Furthermore, we propose environmental stress response

biomarkers as platform readouts for the efficient control and further prediction of the

pancreatic cancer response to the environmental and treatment input.

2. 3D tissue engineering constructs for in vitro studies of pancreatic cancer

A first challenging and promising step towards understanding the pancreatic cancer

evolution and further response to treatment is to recapitulate ex vivo the in vivo

environment. For decades, conventional two dimensional (2D) technology has been

used in cancer research. Generally, these 2D systems are simple to handle,

reproducible and cheap. Furthermore, they are responsive to drug therapies and

radiotherapy thus they are systematically used for treatment screening [9,22-25]. The

architecture of these systems enables homogenous distribution of nutrients and

oxygen, with no presence of gradients [9,23]. Thus, a significant amount of research

4

78

79

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

Page 5: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

on pancreatic cancer has been conducted in 2D [26-33]. However, experimental 2D

platforms cannot accurately recapitulate the 3D in vivo tumour microenvironment as

they do not capture aspects such as structure, porosity and three dimensional

extracellular matrix distribution [11,13,23,34-40]. Consequently, 2D-grown cells can

differ significantly in their morphology, cell-cell and cell-matrix interactions, as well

as in their response to fluctuations of environmental factors [15,41-44]. Furthermore,

several studies have revealed that cells change their original phenotype in 2D culture

conditions, therefore hindering predictability in vivo [45-47].

In contrast to 2D cultures, three dimensional (3D) tissue engineering constructs are

closer to the in vivo environment in terms of structure, architecture, porosity and

microenvironmental niches [48,49]. The features of a 3D system allow topologically

realistic cell-cell interactions and migration due to the spatial arrangement of these

constructs, as well as 3D cell-matrix interactions through coating with extracellular

matrix proteins [38,40,50,51] and co-cultivation with stromal cells [34]. The latter is

particularly important as it is known that stromal cells have a crucial role in cancer

progression and aggressiveness [52-54]. Especially in pancreatic cancer, presence of

intense extracellular matrix, i.e., dense desmoplasia, can contribute to the

development and progression of this malignancy [39,40,55,56] as well as to the

inhibition of apoptotic pathways, directly affecting the diffusion of the

chemotherapeutic drug reagents [57].

Additional to their architectural/structural characteristics, 3D tissue engineering

constructs provide a unique perspective for the realistic distribution of environmental

input (oxygen, nutrients & temperature gradients) and treatment input (irradiation,

drugs), in terms of mass transfer/diffusion as well as heat transfer and energy

deposition [58-61].

5

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

Page 6: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

All the above design parameters that differ between 2D and 3D culture systems, could

possibly lead to a different response of the cells to treatment, suggesting the 3D as

more accurate and predictive (Figure 1). There are several pancreatic cancer studies

that have been conducted in 3D systems. More specifically, the following 3D systems

have been reported for pancreatic cancer i) spheroids, ii) hydrogels, iii) synthetic

matrices.

2.1. Spheroids

Spheroids are the simplest and most widely used tissue engineering constructs, where

a small aggregate of cells grows in three dimensions without adhering to a solid

surface [62,63]. In vitro spheroid formation can be achieved by the forced floating

method [64], the hanging drop method [64,65] and agitation approaches [64] (Figure

1). Spheroids allow cells to interact with each other, with the (produced) extracellular

matrix and the microenvironment [23,66]. Matsuda et al. created 3D spheroid culture

systems for different pancreatic cell lines and captured differences in the expression

levels of cytoskeletal proteins between the developed spheroids and 2D conventional

monolayer cultures. [67]. Longati et al. captured differences in proliferation,

metabolism and chemoresistance between 2D cultures and 3D methylcellulose based

spheroid cell cultures, using various human pancreatic ductal adenocarcinoma

(PDAC) cell lines. Proliferation was lower in 3D compared to 2D, and lactate

accumulation increased in the spheroids. Additionally, higher production of collagen I

and fibronectin I and higher resistance to gemcitabine (GEM), i.e., commonly used

chemotherapeutic drug for pancreatic cancer treatment, was observed in 3D [68]. Wen

et al. developed a 3D spheroid based system using the forced floating method for

human pancreatic cell lines. Higher resistance of PANC-1 to the drugs gemcitabine

6

128

129

130

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

Page 7: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

and 5-fluorouracil was observed in the spheroids as compared to classical 2D

monolayers [69]. As can be seen from the above findings, pancreatic cell lines behave

differently in spheroids as compared to 2D cultures and generally they display an

increased resistance to treatment in the spheroids, making the latter a very attractive

system for in vitro studies. However, spheroids have some disadvantages as their

spatial characteristics result in very high gradients of nutrients/oxygen [9,23,70].

Moreover, it is difficult to control the shape and the morphology of the formed

spheroids [9,11] . A summary of the advantages and disadvantages of spheroids for

pancreatic cancer screening are shown on Table 1.

2.2. Hydrogels

Hydrogels are water swollen and cross-linked polymeric networks produced by a

polymeric reaction of one or more monomers [71,72]. Due to their ability to simulate

the native tissues in terms of architectural and spatial characteristics, their

biocompatibility and their hydrate structure that allows nutrient and oxygen diffusion,

hydrogels are attractive materials for 3D ex vivo cancer models [13,73-75] (Figure 1).

Hydrogels can be formed from purely non-natural molecules such as poly(ethylene

glycol) (PEG) [76] and poly(vinyl alcohol) (PVA) [77]. Sempere et al. developed a

hydrogel, composed of 3% collagen IV/laminin-rich gelatinous medium (Matrigel®)

and soft agar, to study the effect of transforming growth factor (TGFβ) on the growth

of human pancreatic cancer cell lines. Although TGFβ had an inhibitory effect in soft

agar, TGFβ had a growth stimulatory effect in the 3D culture [78]. Raza et al.

developed a hydrogel system for PANC-1 using 4-arm poly(ethylene glycol)-tetra-

norbornene (PEG4NB) and four different cross-linkers. Morphological differences

were observed in the hydrogel as compared to 2D. Specifically, the pancreatic cells

7

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

Page 8: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

formed clusters within the hydrogels within 4 days of cultivation. Moreover, matrix

characteristics such as stiffness and the type of cross-linkers used, influenced the cell

growth as well as the structure of the hydrogels. In particular, softer matrices lead to

enhanced metabolic activity of cell populations and cyst like formations. Furthermore,

matrices cross linked with the MMP (Matrix Metalloproteinase) supported cell growth

& enhanced metabolic activity compared to a dithiothreitol linker to form the

hydrogel and resulted in formation of cyst-like cell structures [79]. Ki et al. developed

a hydrogel system for pancreatic cell lines and evaluated the effect of hydrogel matrix

stiffness and epidermal growth factor receptor (EGFR) on cell proliferation within the

different hydrogel matrices. Higher metabolic activity was observed in softer matrices

as compared to the medium and stiff ones. Moreover, treatment with EGFR inhibitor

reduced the cell viability on stiff hydrogels but had no effect on soft hydrogels nor in

2D cultures [80]. Ki et al. developed a semi-synthetic in vitro microenvironment

mimicking pancreatic desmoplasia. More specifically, a hydrogel was developed and

coated with collagen type I. Increased cell proliferation and drug resistance to

gemcitabine was observed in the hydrogel matrix compared to the 2D system [81].

Boj et al. developed a 3D organoid matrigel system for pancreatic cancer from

primary pancreatic ductal adenocarcinoma cells that can accurately recapitulate the

histology and the disease stage –specific characteristics of the tissue from which they

were derived (normal and neoplastic human tissue). This organoid system managed to

retain both the tumour and stromal production [82,83].

However, despite their great advantages in comparison to classical 2D cultures,

hydrogels cannot provide a consistent in vitro tumour model, due to the lack of

uniform spatial distribution of cells within them [9,84]. Additionally, hydrogels

8

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

Page 9: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

present weak mechanical strength, provoking difficulties in handling them [13,85]

(Table 1).

2.3. Synthetic polymeric scaffolds

Polymeric scaffolds are a promising approach for ex vivo modelling of pancreatic

cancer. Their architectural characteristics provide a realistic ex vivo 3D spatial

arrangement of the cells in the scaffold (Figure 1). Moreover, the possibility of

controlling accurately the porosity of such structures allows mass and energy transport

phenomena such as diffusion of oxygen, nutrients, drugs as well as heat and

irradiation (radiotherapy) to occur to a more similar manner to the actual in vivo

behaviour [35,86-89]. Additionally, polymeric scaffolds, depending on their

fabrication, can have strong mechanical properties [9,12,35,70]. Furthermore, features

of the tumour microenvironment such as the presence of extracellular matrix proteins

and co-culture of stromal cells can take place in order to imitate the in vivo

progression of cancer [43,49]. Therefore, cellular proliferation, signalling, migration,

differentiation and additional bio-physical-chemical and mechanical phenomena that

occur in the tumour microenvironment in vivo can take place in those polymeric

systems, rendering them strong candidates for 3D culture platforms. A synthetic

scaffolding system consisting of poly(vinyl alcohol) – gelatine (PVA/G) sponges was

constructed by Funel et al. Cell growth of primary pancreatic cells was further studied

in the polymeric scaffolds. The results indicated viable cells that adhered within the

sponge, with enhanced metabolism in the 3D model compared to the traditional 2D

culture, resulting in a growth ratio (3D/2D) of 1.38 [90]. Wang et al. indicated that

polyglyconate/gelatine electrospun scaffolds provided a favourable microenvironment

for pancreatic cancer stem cells derived from patients, ensuring increased proliferation

9

202

203

204

205

206

207

208

209

210

211

212

213

214

215

216

217

218

219

220

221

222

223

224

225

226

Page 10: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

capacity in comparison to the 2D system for a time-spam of 7 days [91]. Additionally,

He et al. created a disk-like polymeric scaffolding system based on poly(glycolide-co-

trimethylene carbonate) and gelatine (PGA-TMC)/G. Pancreatic cancer stem cells

were seeded in the scaffold and proliferated for up to 7 days. This polymeric scaffold

model demonstrated better neoplastic formation and accelerated tumour evolution as

compared to the 2D system. [92]. Ricci et al. developed three different biocompatible

scaffold types based on two polymers (poly(ethylene oxide

terephthalate)/poly(butylene terephthalate) and poly(vinyl alcohol)/gelatine) and two

polymeric formulations (fibre mesh, sponge like). The type of polymer and the

formulation technique alter the internal architecture, therefore, affecting the cell

growth and morphology as well as the tumour-specific MMPs synthesis of PDAC

[48]. Totti et al. developed a highly porous (90% porosity) polyurethane (PU) based

scaffold which successfully supported the long term (up to 5 weeks) cultivation of

pancreatic cancer cell lines [93].

Polymeric scaffolds are sometimes complex to produce as compared to other 3D

systems [9] and present difficulties in retrieving cells after the culture formation [64]

(Table 1). However, their overall properties as compared to the other 3D systems

make them very strong tissue engineering candidates. Namely, the possibility of an

accurate monitoring of porosity (size, type, distribution), and the ability to recapitulate

and actual 3D structure with controlled size makes them ideal for ex vivo drug

screening. Furthermore, their strong mechanical strength enable the construction of

robust perfusion systems that could allow vascularisation mimicry.

Overall, from all the above studies, it is clear that there are significant differences in

the biological behaviour as well as the response/resistance to treatment of pancreatic

cancer cells in 3D tissue engineering constructs compared to 2D culture systems, with

10

227

228

229

230

231

232

233

234

235

236

237

238

239

240

241

242

243

244

245

246

247

248

249

250

251

Page 11: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

3D tissue engineering constructs featuring more accurately elements of the tumour

microenvironment (Table 2).

It is therefore essential to move towards the development of efficient 3D systems as

in vitro models for studying, understanding and eventually predicting the pancreatic

tumour evolution and response to different types of treatment.

11

252

253

254

255

256

Page 12: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

3. Towards a biomimetic distribution and control of the environmental (stress)

input in pancreatic cancer tissue engineering platforms

The formation of desmoplasia around the pancreatic tumour coupled with an

avascular tumour microenvironment leads to hypoperfusion of nutrients as well as

hypoxia [26,94,95]. Therefore, environmental parameters should be monitored in 3D

experiments, especially since within a tissue engineering construct those parameters

could fluctuate as a result of diffusional limitations and/or increased cell density at

different locations of the 3D [43]. Furthermore, additionally to cell survival, easily

readable quantitative measurements, i.e., in situ as well as in the cell culture medium

surrounding the 3D systems should be considered for the detection of

stressed/unstressed regions within the 3D culture systems that could be linked to

resistance to treatment (Table 3). Such environmental stress monitoring would lead to

a more efficient control of the input of the environmental fluctuations on the cell

evolution (Figure 1). In particular, a correlation of metabolic (biomarker) information

and the overall cell survival (cell growth/inactivation kinetics) under different levels

of environmental (stress) stimuli in 3D could be highly beneficial as it would enable a

more accurate control and further prediction of the disease progression (in vitro). The

following section summarizes important environmental parameters and suggests

biomarker candidates that could enable efficient control of such parameters in 3D

systems.

3.1.The Impact of Oxygen Stress on Pancreatic Cancer Evolution

Low oxygen levels (hypoxia) as well as fluctuations on the levels of oxygen, naturally

occur within a solid tumour [96]. These hypoxic regions are correlated with altered

metabolism and resistance of a tumour to drug therapies and irradiation [96,97]. For

12

257

258

259

260

261

262

263

264

265

266

267

268

269

270

271

272

273

274

275

276

277

278

279

280

281

Page 13: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

example, Shibamoto et al. showed that four different pancreatic cell lines displayed

resistance to radiotherapy under hypoxic conditions as compared to normoxic

conditions [98]. On the contrary, Mizumoto et al. showed enhanced cytotoxicity of

the drug PR-350 under hypoxia. More specifically, under hypoxia the cytotoxicity of

PR-350 was significantly enhanced almost 5% [99].Yokoi and Fidler observed that

pancreatic cells are more resistant to chemotherapy under hypoxic conditions. In

particular, they showed that under hypoxic conditions, the highly metastatic

pancreatic cell line L3.6pl was resistant to apoptosis triggered by the drug

gemcitabine [17]. Cheng et al. observed that pancreatic cells grown in hypoxic

conditions were resistant to chemotherapy in comparison with cells grown in

normoxic conditions [100]. Furthermore, Maftouh et al. showed that pancreatic

tumour spheroids under hypoxia exhibited increased chemoresistance to gemcitabine

than the corresponding spheroids in a normoxic environment [101].

Oxidative stress biomarkers

Oxygen homeostasis and hypoxia signaling is mediated by the hypoxia-inducible

factors (HIFs) [102]. HIF-1α, the master regulator of O2 homeostasis, is

overexpressed in 88 % of pancreatic cancer tissues, versus in only 16% of healthy

pancreas [94,103]. Hypoxia mediated HIF-1 induction in cancer correlates with

resistance to apoptosis, tumour size and cell proliferation, invasion and metastasis

[102,104,105]. For example, Kizaka-Kindoh et al. indicated that killing/deactivation

of HIF-active pancreatic cells, impaired the tumour evolution [106]. Additionally,

Schwartz et al. showed that the in vitro and in vivo use of HIF-1α inhibitors led to a

decrease in pancreatic cancer cell survival after fractionated radiation [103]. Another

study by Kang et al. showed that HIF-1 degradation triggers pancreatic tumour cell

death [107].

13

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

Page 14: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

Collectively, the above studies indicate hypoxia is a direct cause of therapeutic

resistance. Both radiotherapy and chemotherapy are significantly influenced by

hypoxia, therefore, it is very important to include oxygen input as a parameter on the

design as well as the readout (by HIF status monitoring) of the pancreatic cancer in

vitro platform (Figure 1 & Table 4).

3.2. Metabolic stress- Nutrient deprivation

As previously stated, hypoperfusion is a common characteristic of pancreatic cancer

due to the desmoplasia which surrounds the tumour. Therefore, it may lead to not only

oxygen but also nutrient limitations in the tumour area [108]. Environmental stress

can affect normal tissue cells and turn them into other types, after accumulation of

genomic and epigenomic alterations (Figure 2). The metabolism of proliferative cells

(cancer cells) tends to be more glycolytic and dependent less on oxidative

phosphorylation than a normal/healthy cell (Figure 2) even when oxygen is sufficient

(Warburg effect). This kind of metabolism covers the needs of proliferative cells in

macromolecules’ building blocks (monomers), energy and establishes a redox

potential balance [109]. Nevertheless, pancreatic tumours show an inherited ability to

survive under starvation conditions. A study by Izuishi et al. showed that different

pancreatic cell lines displayed remarkable survival under nutrient starvation

conditions. More specifically, the cells maintained 50 % of their viability up to 48 h in

nutrient deprived cultured medium [21]. Similarly, Kim et al. observed that among

different cancer cell lines (lung carcinoma, colorectal carcinoma), pancreatic cells

displayed remarkable tolerance to extreme nutrient deprivation. [20]. However,

several studies showed a beneficial effect of metabolic stress on treatment of

pancreatic cancer. For example, Lu et al. reported that Kigamicin D, a potential

cytotoxic compound, resulted in enhanced cytotoxic effect in glucose deprived culture

14

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

Page 15: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

medium, compared to nutrient rich conditions [110]. Similarly, Momose et al.

investigated the effect of various potential cytotoxic compounds on the survival of

PANC-1. The presence of those agents led to enhanced cytotoxicity under nutrient

deprived conditions compared with nutrient abundant conditions [111]. Ueda et al.

observed increased cytotoxicity of the drug Grandifloracin in PANC-1 under nutrient

deprivation [112].

From all the above studies it is clear that the cocktail of nutrients plays a key role on

the pancreatic cancer response to drugs, therefore, the nutrient level and content is a

necessary input when screening pancreatic cancer ex vivo (Figure 1).

Metabolic stress biomarkers

The relevance of metabolic stress on the response of pancreatic cancer to treatment

underscores the potential clinical value of candidate biomarkers of metabolic stress.

Indeed, several studies identify biomarkers that are expressed under metabolic stress

conditions. Guo et al. reported that PANC-1cells under starvation have higher

expression of LC3-II protein levels. LC3-II is the autophagy –associated form of the

microtubule-associated protein 1 light chain 3 [113-115]. Treatment with the

autophagy inhibitor chloroquine under nutrient deprived conditions significantly

increased LC3-II expression and inhibited cell growth [116]. Moreover Ueda et al.

presented that combination of nutrient deprivation and treatment with the natural

compound Grandifloracin increased LC3-II expression up to 22 fold in comparison to

nutrient abundance and resulted to reduction of cell viability [112]. Similarly,

Hashimoto et al. showed that nutrient deprivation increased LC3-II levels by almost

70% in PANC-1 cells compared to nutrient rich conditions. Additionally, combination

treatment of PANC-1 and BxPC-3 cells with common chemotherapeutic drugs and

15

332

333

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

Page 16: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

chloroquine led to an increase in LC3-II protein expression which was coupled with

enhanced cytotoxicity for both cell lines [27].

Overall, as stated above, LC3-II is one of the key proteins in the molecular

mechanism of autophagy, which is a general environmental cell stress response

mechanism to metabolic stress (nutrient deprivation) [20,112,116], but also to

oxidative stress [117-119] and treatment (chemotherapy and/or radiotherapy)

[28,31,120-122]. This molecular mechanism (autophagy) is highly elevated in

pancreatic cancer [123,124]. Therefore, the LC3-II protein could be an interesting

biomarker candidate to be quantified and monitored in 3D tissue engineering systems,

especially due to the occurrence of oxygen/nutrient/drug gradients in such systems

(Figure 1, Table 4).

3.3. Temperature stress

Increased temperature could affect cell proliferation and consequently influence the

response/resistance to treatment. For that reason, there are clinical studies that

investigate the synergistic effect of hyperthermia and chemotherapy and/or

radiotherapy [125,126]. Ohguri et al. notified that chemoradiotherapy combined with

regional hyperthermia applied to 29 patients with advanced pancreatic carcinoma led

to better survival than the chemoradiotherapy alone [125]. Similarly, Assogna et al.

reported the beneficial effect of applied hyperthermia on the adverse effects of

chemotherapy for 25 pancreatic cancer patients. Apart from the clinical studies, in

vitro studies also examined the role of temperature stress on pancreatic cancers’

resistance/ sensitivity to treatment [126]. For instance, Liao et al. reported that when

different pancreatic cell lines were exposed to heat stress at 45oC they overexpressed

the BAG-3 (Bcl-2 Associated Athanogene 3) protein, a modulator of cellular anti-

16

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

Page 17: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

apoptotic activity [127]. Furthermore, Mocan et al. studied the growth of pancreatic

cancer cells, as influenced by hyperthermia via laser treatment, in a nano-biosystem.

The cell viability was significantly reduced after thermal treatment [128]. Similarly,

Guo et al. indicated that hyperthermia increased the gemcitabine sensitivity in

pancreatic cell lines [129]{Guo, 2015 #273;Guo, 2015 #273}.

Temperature stress biomarkers

Cells respond to heat by inducing the synthesis of a group of proteins called the heat

shock proteins or hsps [130]. HSP27 belongs to the family of heat shock proteins that

basically act as molecular chaperones in cells exposed to different types of stress,

such as heat shock and/ or irradiation, oxidative stress, chemotherapeutic drugs

[130 ,131-133]. Schäfer et al. indicated that the application of hyperthermia (at

41.8oC), increased the over-expression of heat shock protein 27 (HSP27) and the

sensitivity of pancreatic cell lines to the chemotherapeutic agent gemcitabine [134].

Similar findings on HSP27 were reported by Guo et al. [129]. In contrast, heat shock

protein HSP 27 overexpression has also been reported to increase gemcitabine

resistance in pancreatic cancer cell lines [18,19,135]. Overall, these studies strongly

indicate that temperature stress affects the response of pancreatic cells to treatment

and it is very important to monitor the quantitative evolution of the HSP27 as readout

of the platform (Figure 1 &Table 4).

4. Monitoring environmental stress using metabolomics: a great potential for

novel pancreatic cancer biomarker detection in 3D cultures

Further to the previously listed biomarker candidates for monitoring the

environmental stress response in 3D systems new biomarker molecules can be

detected through metabolomics. Metabolomics is the high-throughput analysis of all

17

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

403

404

405

406

Page 18: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

the measurable free metabolite pools [136]. It offers a quantitative and holistic insight

of the metabolome of a particular biological system. There is already a significant

interest of the scientific community in the application of metabolomics analysis in

deciphering aspects of cancer metabolism, as it offers a metabolic snapshot of the

cancerous state. Metabolites are a very big group of compounds (41993 now

described at the Human Metabolome Database [137,138]) and it is highly likely that

some of them are correlated with specific diseases. Additionally, metabolic profiles,

which consist of the relative concentrations of many metabolites simultaneously, can

be proven very sensitive and accurate as biomarkers as they reflect the total metabolic

condition of a sample and can include subtle differences which cannot be easily

detected by classic single metabolite quantitative methods. Moreover, they do not

require the full knowledge of the metabolic network and metabolomics samples’

collection is not necessarily invasive, as samples can be collected from body fluids

such as plasma, saliva, tears, sweat and urine [139].

Several metabolomics analyses have already been applied for the study of pancreatic

cancer. Most of them concern of human samples and not pancreatic cells, but plasma

[140,141], saliva [142], urine [143],serum [144-147], tumour tissues [148] and all the

common metabolomics platforms, such as GC-MS, LC-MS, CE-MS and NMR have

been applied. Specifically, Berger et al. applied lipidomics profiling and managed to

discriminate normal plasma profiles and profiles collected from pancreatic cancer

patients [140]. Based on the same idea, Urayama et al. found differences on the

metabolic profiles of pancreatic cancer plasma samples vs. healthy plasma samples

with lactate characteristically increased in pancreatic cancer plasma samples [141].

Sugimoto et al. used saliva samples to compare oral, breast and pancreatic cancer and

managed to identify 57 metabolites that could be used for early detection purposes

18

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

Page 19: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

[142]. Napoli et al. used urine samples to discriminate the metabolic profiles of males

with PDAC and healthy ones [143]. Tesiram et al. applied NMR to serum samples

and detected significant differences in lactate, taurine, cholines and fatty acids

between normal samples and samples collected from patients with pancreatic cancer

[146], while Kobayashi et al. claim to have reached a diagnostic-level of sensitivity

with the same kind of samples [147]. In a study with a total number of 99 samples,

Bathe et al. compared the serum metabolic profiles of benign hepatobiliary diseased

and pancreatic cancer individuals [145]. They were able to distinguish the two types

of profiles while there are important differences in glutamate, glucose, creatine and

glutamine concentrations.

Pancreatic cancer cell lines were also compared with pancreatic ductal epithelial cells

revealing that normal ductal epithelial cell line (H6C7) displayed metabolic profiles

that were significantly distinct from three pancreatic cancer cell lines (MIA PaCa-2,

PANC-1, AsPC-1).[149].

However, all the above studies have been conducted with cell lines in 2D systems.

Metabolomic analysis in 3D pancreatic cancer culture systems, simulating in vivo

conditions, could be translated into sensitive, dynamic and accurate monitoring of the

cancerous system ex vivo. 3D systems monitoring with the use of metabolomics

analysis offers a quick and robust snapshot of the metabolic physiology which reflects

even to subtle changes of the cell physiology, something that cannot be achieved with

other omics analyses. More interestingly, the ability to combine intracellular

(fingerprint) and extracellular (footprint) metabolomics data could give the

opportunity to achieve a good level of knowledge on the biological system and watch

the dynamic transitions of the cells towards different physiological conditions, as the

population of cancer cells within a 3D matrix is not homogenous.

19

432

433

434

435

436

437

438

439

440

441

442

443

444

445

446

447

448

449

450

451

452

453

454

455

456

Page 20: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

Overall, quantitative knowledge on the relation of the production/evolution as well as

the potential distribution of one or more biomarkers within a tissue engineering

construct as influenced by (micro-) environmental conditions as well as treatment

would be a significant step towards enhancement of predictability of treatment

response of pancreatic cancer [43,150-152].

6. Conclusions

For decades, pancreatic cancer remains a highly lethal disease. Despite the useful

information retrieved from animal studies they still lack translatability and they are

expensive and not always reproducible. Furthermore, the classically used 2D cultures

for in vitro drug and/or irradiation screening have been unable to efficiently predict

the in vivo response to treatment. Clinical trials have shown that combinatory drug

therapy may be beneficial for pancreatic cancer, as summarised in Table 5 [153,154].

The development of in vitro tumour models which would capture elements of the in

vivo pancreatic tissue are a very promising, low cost, accurate approach for

accelerating drug development and application of novel therapies from bench to bed.

Advances in tissue engineering have enabled the construction of in vitro platforms

that recapitulate mammalian tissue aspects such as structure, porosity, extracellular

matrix mimicry, and cell-cell interactions. Next to the ex vivo architectural tissue

mimicry, monitoring the impact of the environmental stress adaptation through the

quantitative control of the production and/or distribution of stress biomarkers within

the in vitro tissue (in situ analysis) could allow a more accurate correlation of the

pancreatic cancer cell survival with the cellular environmental (stress) metabolic

response, therefore, enhancing treatment predictability.

20

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 21: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

Acknowledgements

This work was supported by the Department of Chemical and Process Engineering of

the University of Surrey as well as an Impact Acceleration Grant (IAA-KN9149C) of

the University of Surrey, an IAA-EPSRC Grant (RN0281J) and the Royal Society.

21

482

483

484

485

486

Page 22: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

References

1 American Cancer Society. (2016) Cancer Facts and Figures 2016. (Vol.

2016)

2 Siegel, R.L. et al. (2016) Cancer statistics, 2016. CA Cancer J Clin 66, 7-30

3 Cascinu, S. et al. (2010) Pancreatic cancer: ESMO Clinical Practice

Guidelines for diagnosis, treatment and follow-up. Annals of Oncology 21

(Supplement 5), 55-58

4 Herreros-Villanueva M., and Bujanda, L. (2016) Non-invasive biomarkers in

pancreatic cancer diagnosis: what we need versus what we have. Ann Transl

Med 4, 134

5 Kamisawa, T. et al. (1995) Hematogenous metastases of pancreatic ductal

carcinoma. Pancreas 11, 345-349

6 Yachida, S. and Iacobuzio-Donahue, C.A. (2009) The pathology and genetics

of metastatic pancreatic cancer. Arch Pathol Lab Med 133, 413-422

7 Chand, S., et al. (2016) The Landscape of Pancreatic Cancer Therapeutic

Resistance Mechanisms. Int J Biol Sci 12,273-282.

8 Voskoglou-Nomikos, T., et al. (2003) Clinical Predictive Value of the in Vitro

Cell Line, Human Xenograft, and Mouse Allograft Preclinical Cancer Models.

Clin Cancer Res 9,4227-4239.

9 Burdett, E., et al. (2010) Engineering Tumors: A Tissue Engineering

Perspective in Cancer Biology. Tissue Eng Part B Rev 16,351-359.

10 Matsusaki, M., et al. (2014)Three-dimensional cell culture technique and

pathophysiology. Adv Drug Deliv Rev,74,95-103

11 Asghar, W. et al. (2015) Engineering cancer microenvironments for in vitro 3-

D tumor models. Mater Today 18, 539-553

22

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

Page 23: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

12 Lee, J. et al. (2008) Three-dimensional cell culture matrices: state of the art.

Tissue Eng Part B Rev 14, 61-86

13 Nyga, A. et al. (2011) 3D tumour models: novel in vitro approaches to cancer

studies. J Cell Commun Signal 5, 239-248

14 Gaspar, D. and Zeugolis, D.I. (2016) Engineering in vitro complex

pathophysiologies for drug discovery purposes. Drug Discov Today 21,1341-

1344

15 Birgersdotter, A. et al. (2005) Gene expression perturbation in vitro-A

growing case for three-dimensional (3D) culture systems. Semin Cancer Biol

15, 405-412

16 Shamir, E.R. and Ewald, A.J. (2014) Three-dimensional organotypic culture:

experimental models of mammalian biology and disease. Nat Rev Mol Cell

Biol 15, 647-664

17 Yokoi, K. and Fidler, I.J. (2004) Hypoxia Increases Resistance of Human

Pancreatic Cancer Cells to Apoptosis Induced by Gemcitabine. Clin Cancer

Res 10, 2299-2306

18 Mori-Iwamoto, S. et al. (2008) A proteomic profiling of gemcitabine

resistance in pancreatic cancer cell lines. Mol Med Rep 1, 429-434

19 Mori-Iwamoto, S. et al. (2007) Proteomics finding heat shock protein 27 as a

biomarker for resistance of pancreatic cancer cells to gemcitabine. Int J Oncol

31, 1345-1350

20 Kim, S.E. et al. (2015) Autophagy sustains the survival of human pancreatic

cancer PANC-1 cells under extreme nutrient deprivation conditions. Biochem

Biophys Res Commun 463, 205-210

23

512

513

514

515

516

517

518

519

520

521

522

523

524

525

526

527

528

529

530

531

532

533

534

535

Page 24: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

21 Izuishi, K. et al. (2000) Remarkable Tolerance of Tumor Cells to Nutrient

Deprivation: Possible New Biochemical Target for Cancer Therapy. Cancer

Res 60, 6201-6207

22 Johnson, J. et al. (2001) Relationships between drug activity in NCI

preclinical in vitro and in vivo models and early clinical trials. Br J Cancer 84,

1424-1431

23 Edmondson, R. et al. (2014) Three-Dimensional Cell Culture Systems and

Their Applications in Drug Discovery and Cell-Based Biosensors. Assay Drug

Devel Technol 12, 207-218

24 Holbeck, S.L. (2004) Update on NCI in vitro drug screen utilities. Eur J

Cancer 40, 785-793

25 Sittampalam, S. et al. (2015) Three-Dimensional Cell Culture Assays: Are

They More Predictive of In Vivo Efficacy than 2D Monolayer Cell-Based

Assays? Assay Drug Dev Technol 13, 254-261

26 Huang, Z.Q. et al. (2011) Molecular targeted approaches for treatment of

pancreatic cancer. Curr Pharm Des 17, 2221-2238

27 Hashimoto, D. et al. (2014) Autophagy is needed for the growth of pancreatic

adenocarcinoma and has a cytoprotective effect against anticancer drugs. Eur

J Cancer 50, 1382-1390

28 Mukubou, H. et al. (2010) The role of autophagy in the treatment of pancreatic

cancer with gemcitabine and ionizing radiation. Int J Oncol 37, 821-828

29 Oonishi, K. et al. (2012) Different effects of carbon ion beams and X-rays on

clonogenic survival and DNA repair in human pancreatic cancer stem-like

cells. Radiother Oncol 105, 258-265

24

536

537

538

539

540

541

542

543

544

545

546

547

548

549

550

551

552

553

554

555

556

557

558

559

Page 25: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

30 Porcelli, L. et al. (2013) Optimize radiochemotherapy in pancreatic cancer:

PARP inhibitors a new therapeutic opportunity. Mol Oncol 7, 308-322

31 Koshkina, N.V. et al. (2014) Autophagy and Enhanced Chemosensitivity in

Experimental Pancreatic Cancers Induced by Noninvasive Radiofrequency

Field Treatment. Cancer Lett 120, 480-491

32 Wang, Y. et al. (2014) Gemcitabine Induces Poly (ADP-Ribose) Polymerase-1

(PARP-1) Degradation through Autophagy in Pancreatic Cancer. Plos One,

DOI: 10.1371/journal.pone.0109076 (https://www.ncbi.nlm.nih.gov)

33 Dai, Z.-J. et al. (2013) Antitumor Effects of Rapamycin in Pancreatic Cancer

Cells by Inducing Apoptosis and Autophagy. Int J Mol Sci, 14,273-285

34 Kim, J.B. (2005) Three-dimensional tissue culture models in cancer biology.

Cancer Biol 15, 365-377

35 O’Brien, F.J. (2011) Biomaterials & scaffolds for tissue engineering. Mater

Today 14, 88-95

36 Antoni, D. et al. (2015) Three-dimensional cell culture: a breakthrough in

vivo. Int J Mol Sci 16 (3), 5517-5527

37 Lu, P. et al. (2012) The extracellular matrix: a dynamic niche in cancer

progression. J Cell Biol 196 (4), 395-406

38 Kim, S.H. et al. (2011) Extracellular matrix and cell signalling: the dynamic

cooperation of integrin, proteoglycan and growth factor receptor. J Endocrinol

209, 139-151

39 Xie, D. and Xie, K. (2015) Pancreatic cancer stromal biology and therapy.

Genes Dis 2, 133-143

40 Pickup, M.W. et al. (2014) The extracellular matrix modulates the hallmarks

of cancer. EMBO Rep 15, 1243-1253

25

560

561

562

563

564

565

566

567

568

569

570

571

572

573

574

575

576

577

578

579

580

581

582

583

584

Page 26: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

41 Nelson, C.M. and Bissell, M.J. (2006) Of Extracellular Matrix, Scaffolds, and

Signaling: Tissue Architecture Regulates Development, Homeostasis, and

Cancer. Annu Rev Cell Dev Biol. 22, 287-309

42 Cukierman, E. et al. (2002) Cell interactions with three-dimensional matrices.

Curr Opin Cell Biol 14, 633-639

43 Velliou, E.G. et al. (2015) Towards unravelling the kinetics of an acute

myeloid leukaemia model system under oxidative and starvation stress: a

comparison between two- and three-dimensional cultures. Bioprocess Biosyst

Eng 38, 1589-1600

44 Villasante, A. and Vunjak-Novakovic, G. (2015) Tissue-engineered models of

human tumors for cancer research. Expert Opin Drug Discov 10, 257-268

45 Yamada, K.M. and Cukierman, E. (2007) Modeling Tissue Morphogenesis

and Cancer in 3D. Cell 130, 601-608

46 Weaver, V.M. et al. (1997) Reversion of the Malignant Phenotype of Human

Breast Cells in Three-Dimensional Culture and In Vivo by Integrin Blocking

Antibodies. JCB 137, 231-245

47 Baker, L.A. et al. (2016) Modeling pancreatic cancer with organoids. Trends

Cancer 2, 176-190

48 Ricci, C. et al. (2014) Interfacing polymeric scaffolds with primary pancreatic

ductal adenocarcinoma cells to develop 3D cancer models. Biomatter DOI:

10.4161/21592527.2014.955386 (https://www.ncbi.nlm.nih.gov).

49 Blanco, T.M. et al. (2010) The development of a three-dimensional scaffold

for ex vivo biomimicry of human acute myeloid leukaemia. Biomaterials 31,

2243-2251

26

585

586

587

588

589

590

591

592

593

594

595

596

597

598

599

600

601

602

603

604

605

606

607

608

Page 27: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

50 Debnath, J. and Brugge, J.S. (2005) Modelling Grandular Epithelial Cancers

In Three - Dimensional Cultures. Nature 5, 675-689

51 Schlie-Wolter, S. et al. (2013) The selective role of ECM components on cell

adhesion, morphology, proliferation and communication in vitro. Exp Cell Res

319, 1553-1561

52 Nielsen, M.F.B. et al. (2016) Key players in pancreatic cancer-stroma

interaction: cancer-associated fibroblasts, endothelial and inflammatory cells.

WJG 22, 2678-2700

53 Waghray, M. et al. (2013) Deciphering the Role of Stroma in Pancreatic

Cancer. Curr Opin Gastroenterol 29 (5), 537-543

54 Kleeff, J. et al. (2016) Pancreatic cancer. Nat Rev Dis Primers 2, 16022

55 Gress, T.M. et al. (1998) Role of extracellular matrix in pancreatic diseases.

Digestion 59, 625-637

56 Pandol, S. et al. (2009) Desmoplasia of pancreatic ductal adenocarcinoma.

Clin Gastroenterol Hepatol 7 (11 Suppl), S44-47

57 Whatcott, C.J. et al. (2012) Desmoplasia and chemoresistance in pancreatic

cancer. In Pancreatic Cancer and Tumor Microenvironment (Grippo, P.J. and

Munshi, H.G., eds.),Transworld Research Network

58 Zanoni, M. et al. (2016) 3D tumor spheroid models for in vitro therapeutic

screening: a systematic approach to enhance the biological relevance of data

obtained. Sci Rep, DOI: 10.1038/srep19103 (http://www.nature.com/).

59 Anastasov, N., et al. (2015).A 3D-microtissue-based phenotypic screening of

radiation resistant tumor cells with synchronized chemotherapeutic treatment.

BMC Cancer, DOI: 10.1186/s12885-015-1481-9

(https://www.ncbi.nlm.nih.gov).

27

609

610

611

612

613

614

615

616

617

618

619

620

621

622

623

624

625

626

627

628

629

630

631

632

633

Page 28: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

60 Simon, K.A. et al. (2016) Metabolic response of lung cancer cells to radiation

in a paper-based 3D cell culture system. Biomaterials 95, 47-59

61 Pan, D. et al. (2014) Ionizing radiation induced biological effects in three-

dimensional cell cultures. Rend. Fis. Acc. Lincei 25 (Suppl 1), S81-S86

62 Fennema, E. et al. (2013) Spheroid culture as a tool for creating 3D complex

tissues. Trends Biotechnol 31, 108-115

63 Cesarz, Z. and Tamama, K. (2016) Spheroid Culture of Mesenchymal Stem

Cells. Stem Cells Int, DOI:10.1155/2016/9176357

(https://www.ncbi.nlm.nih.gov).

64 Breslin, S. and O’Driscoll, L. (2013) Three-dimensional cell culture: the

missing link in drug discovery. Drug Discov Today 18, 241-248

65 Foty, R. (2011) A Simple Hanging Drop Cell Culture Protocol for Generation

of 3D Spheroids. JoVE, DOI: 10.3791/2720 (http://www.jove.com).

66 Nath, S. and Devi, G.R. (2016) Three-dimensional culture systems in cancer

research: Focus on tumor spheroid model. Pharmacol Ther 163, 94-108

67 Matsuda, Y. et al. (2010) Morphological and cytoskeletal changes of

pancreatic cancer cells in three-dimensional spheroidal culture. Med Mol

Morphol 43, 211-217

68 Longati, P. et al. (2013) 3D pancreatic carcinoma spheroids induce a matrix-

rich, chemoresistant phenotype offering a better model for drug testing. BMC

Cancer DOI:10.1186/1471-2407-13-95 (http://www.biomedcentral.com).

69 Wen, Z. et al. (2013) A spheroid-based 3-D culture model for pancreatic

cancer drug testing, using the acid phosphatase assay. Braz J of Med Biolog

Res 46, 634-642

28

634

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

Page 29: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

70 Knight, E. and Przyborski, S. (2015) Advances in 3D cell culture technologies

enabling tissue-like structures to be created in vitro. J Anat 227, 746-756

71 Ahmed, E.M. (2015) Hydrogel: Preparation, characterization, and

applications: A review. J Adv Res 6, 105-121

72 Peppas, N.A. et al. (2000) Hydrogels in pharmaceutical formulations. Eur J

Pharm Biopharm 50, 27-46

73 Tibbitt, M.W. and Anseth, K.S. (2009) Hydrogels as Extracellular Matrix

Mimics for 3D Cell Culture. Biotechnol Bioeng. 103, 655-663

74 Saha, K. et al. (2007) Designing synthetic materials to control stem cell

phenotype. Curr Opin ChemBiol, 11, 381-387

75 El-Sherbiny, I.M. and Yacoub, M.H. (2013) Hydrogel scaffolds for tissue

engineering: Progress and challenges. Glob Cardiol Sci Pract 2013, 316-342

76 Sawhney, A.S. et al. (1993) Bioerodible Hydrogels Based on

Photopolymerized Poly(ethyleneglycol)-co-poly(a-hydroxy acid) Diacrylate

Macromers. Macromolecules 26, 581-587

77 Martens, P. and Anseth, K.S. (2000) Characterization of hydrogels formed

from acrylate modified poly(vinyl alcohol) macromers. Polymer 41, 7715-

7722

78 Sempere, L.F. et al. (2011) A novel 3-dimensional culture system uncovers

growth stimulatory actions by TGFβ in pancreatic cancer. Cancer Biol Ther

12, 198-207

79 Raza, A. et al. (2013) The influence of matrix properties on growth and

morphogenesis of human pancreatic ductal epithelial cells in 3D. Biomaterials

24, 5117-5127

29

658

659

660

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

681

Page 30: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

80 Ki, C.S. et al. (2013) Effect of 3D Matrix Compositions on the Efficacy of

EGFR Inhibition in Pancreatic Ductal Adenocarcinoma Cells.

Biomacromolecules 14, 3017-3026

81 Ki, C.S. et al. (2014) Thiol-ene hydrogels as desmoplasia-mimetic matrices

for modeling pancreatic cancer cell growth, invasion, and drug resistance.

Biomaterials 35, 9688-9677

82 Boj, S.F. et al. (2016) Model organoids provide new research opportunities for

ductal pancreatic cancer. Mol Cell Oncol, DOI:

10.1080/23723556.2015.1014757 (http://www.tandfonline.com).

83 Boj, S.F. et al. (2015) Organoid models of human and mouse ductal pancreatic

cancer. Cell 160, 324-338

84 Ricci, C. et al. (2013) Cancer tissue engineering—new perspectives in

understanding the biology of solid tumours—a critical review. OA Tissue

Engineering, https://doi.org/10.13172/2052-9643--1-607

( http://www.oapublishinglondon.com)

85 Hutmacher, D.W. (2001) Scaffold design and fabrication technologies for

engineering tissues — state of the art and future perspectives. J. Biomater. Sci.

Polymer Edn 12, 107-124

86 Dhandayuthapani, B. et al. (2011) Polymeric Scaffolds in Tissue Engineering

Application: A Review. Int J Polym Sci, DOI:10.1155/2011/290602

(https://www.ncbi.nlm.nih.gov).

87 Xu, F. and Burg, K.J. (2007) Three-dimensional polymeric systems for cancer

cell studies. Cytotechnology 54, 135-143

88 Mooney, D.J. et al. (1997) Long-term engraftment of hepatocytes transplanted

on biodegradable polymer sponges. Biomed Mater 37, 413-420

30

682

683

684

685

686

687

688

689

690

691

692

693

694

695

696

697

698

699

700

701

702

703

704

705

706

Page 31: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

89 Ouriemchi, E.M. and Vergnaud, J.M. (2000) Processes of drug transfer with

three different polymeric systems with transdermal drug delivery. Comput

Theor Polym Sci 10, 391-401

90 Funel, N. et al. (2008) In Vitro Model of Pancreatic Ductal Adenocarcinoma:

New Strategy to Study Pancreatic Ductal Carcinoma. JOP 9 (6 Suppl),810-

811

91 Wang, X. et al. (2013) A bioengineered metastatic pancreatic tumor model for

mechanistic investigation of chemotherapeutic drugs. J Biotechnol 166, 166-

179

92 He, Q. et al. (2013) A tissue-engineered subcutaneous pancreatic cancer model

for antitumor drug evaluation. Int J Nanomedicine 8, 1167-1176

93 Totti, S. et al. (2016) On the evaluation of long term ex vivo cultivation of

pancreatic cancer in a 3D scaffolding system. Eur Cell Mater, 31 (Supp 1),

441

94 Erickson, L.A. et al. (2015) Targeting the hypoxia pathway to treat pancreatic

cancer. Drug Des Devel and Ther 9, 2029-2031

95 Guillaumond, F. et al. (2013) Strengthened glycolysis under hypoxia supports

tumor symbiosis and hexosamine biosynthesis in pancreatic adenocarcinoma.

PNAS 110, 3919-3924

96 Dewhirst, M.W. et al. (2008) Cycling hypoxia and free radicals regulate

angiogenesis and radiotherapy response. Nat Rev Cancer 8, 425-436

97 Bertout, J.A. et al. (2008) The impact of O2 availability on human cancer. Nat

Rev Cancer 8, 967-975

31

707

708

709

710

711

712

713

714

715

716

717

718

719

720

721

722

723

724

725

726

727

728

729

Page 32: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

98 Shibamoto, Y. et al. (2000) Radiosensitivity of human pancreatic cancer cells

in vitro and in vivo, and the effect of a new hypoxic cell sensitizer,

doranidazole. Radiother Oncol 56, 265-270

99 Mizumoto, K. et al. (2002) A Nitroimidazole Derivative, PR-350, Enhances

the Killing of Pancreatic Cancer Cells Exposed to High-dose Irradiation under

Hypoxia. J.Radiat. Res. 43, 43-51

100 Cheng, Z.X. et al. (2014) Effects of the HIF-1alpha and NF-kappaB loop on

epithelial-mesenchymal transition and chemoresistance induced by hypoxia in

pancreatic cancer cells. Oncol Rep 31, 1891-1898

101 Maftouh, M. et al. (2014) Synergistic interaction of novel lactate

dehydrogenase inhibitors with gemcitabine against pancreatic cancer cells in

hypoxia. Br J Cancer 110, 172-182

102 Yuen, A. and Díaz, B. (2014) The impact of hypoxia in pancreatic cancer

invasion and metastasis. Hypoxia 2, 91-106

103 Schwartz, D.L. et al. (2010) Radiosensitization and Stromal Imaging Response

Correlates for the HIF-1 Inhibitor PX-478 Given with or without

Chemotherapy in Pancreatic Cancer. Mol Cancer Ther 9 (7), 2057-2067

104 Akakura, N. et al. (2001) Constitutive Expression of Hypoxia-inducible

Factor-1 Renders Pancreatic Cancer Cells Resistant to Apoptosis Induced by

Hypoxia and Nutrient Deprivation. Cancer Res 61, 6548-6554

105 Kitada, T. et al. (2003) Clinicopathological significance of hypoxia-inducible

factor-1a expression in human pancreatic carcinoma. Histopathology 43, 550-

555

32

730

731

732

733

734

735

736

737

738

739

740

741

742

743

744

745

746

747

748

749

750

751

752

Page 33: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

106 Kizaka-Kondoh, S. et al. (2009) Selective Killing of Hypoxia-Inducible

Factor-1–Active Cells Improves Survival in a Mouse Model of Invasive and

Metastatic Pancreatic Cancer. Clin Cancer Res 15, 3433-3441

107 Kang, R. et al. (2014) RAGE is essential for oncogenic KRAS-mediated

hypoxic signaling in pancreatic cancer. Cell Death Dis 5,

DOI:10.1038/cddis.2014.445 (https://www.ncbi.nlm.nih.gov)

108 Kamphorst, J.J. et al. (2015) Human pancreatic cancer tumors are nutrient

poor and tumor cells actively scavenge extracellular protein. Cancer Res 75,

544-553

109 Cairns, R.A. et al. (2011) Regulation of cancer cell metabolism. Nat Rev

Cancer 11, 85-95

110 Lu, J. et al. (2004) Kigamicin D, a novel anticancer agent based on a new anti-

austerity strategy targeting cancer cells’ tolerance to nutrient starvation.

Cancer Sci 95, 547-552

111 Momose, I. et al. (2010) Mitochondrial inhibitors show preferential

cytotoxicity to human pancreatic cancer PANC-1 cells under glucose-deprived

conditions. Biochem Biophys Res Commun 392, 460-466

112 Ueda, J. et al. (2014) (+)-Grandifloracin, an antiausterity agent, induces

autophagic PANC -1 pancreatic cancer cell death. Drug Des Devel Ther 8, 39-

47

113 He, C. and Klionsky, D.J. (2009) Regulation Mechanisms and Signaling

Pathways of Autophagy. Ann. Rev. Genet. 43, 67-93

114 Tanida, I. (2011) Autophagy basics. Microbiology and Immunology, DOI:

doi:10.1111/j.1348-0421.2010.00271.x ( https://www.ncbi.nlm.nih.gov)

33

753

754

755

756

757

758

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

776

Page 34: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

115 Russell, R.C. et al. (2014) Autophagy regulation by nutrient signaling. Cell

Res, DOI: 10.1038/cr.2013.166 (http://www.nature.com/)

116 Guo, J.Y. et al. (2011) Activated Ras requires autophagy to maintain oxidative

metabolism and tumorigenesis. Genes Dev 25, 460-470

117 Fujii, S. et al. (2008) Autophagy is activated in pancreatic cancer cells and

correlates with poor patient outcome. Cancer Sci 99, 1813-1819

118 Kang, R. et al. (2011) The Receptor for Advanced Glycation End-Products

(RAGE) Protects Pancreatic Tumor Cells Against Oxidative Injury. Antioxod

Redox Signal 15, 2175-2184

119 Zhang, L. et al. (2015) The Relevance of Nrf2 Pathway and Autophagy in

Pancreatic Cancer Cells upon Stimulation of Reactive Oxygen Species. Oxid

Med Cell Longev, DOI : 10.1155/2016/3897250 (https://www.hindawi.com)

120 Pardo, R. et al. (2010) Gemcitabine Induces the VMP1-Mediated Autophagy

Pathway to Promote Apoptotic Death in Human Pancreatic Cancer Cells.

Pancreatology 10, 19-26

121 Donadelli, M. et al. (2011) Gemcitabine/cannabinoid combination triggers

autophagy in pancreatic cancer cells through a ROS-mediated mechanism.

Cell Death Dis, DOI:10.1038/cddis.2011.36 ( https://www.ncbi.nlm.nih.gov)

122 Chiu, H. et al. (2015) Synergistic antitumor effects of radiation and

proteasome inhibitor treatment in pancreatic cancer through the induction of

autophagy and the downregulation of TRAF6. Cancer Lett 365, 229-239

123 Yang, S. et al. (2011) Pancreatic cancers require autophagy for tumor growth.

Genes Dev 25, 717-729

124 Kang, R. and Tang, D. (2012) Autophagy in pancreatic cancer pathogenesis

and treatment. Am J Cancer Res 2, 383-396

34

777

778

779

780

781

782

783

784

785

786

787

788

789

790

791

792

793

794

795

796

797

798

799

800

801

Page 35: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

125 Ohguri, T. et al. (2008) Concurrent chemoradiotherapy with gemcitabine plus

regional hyperthermia for locally advanced pancreatic carcinoma: initial

experience. Radiat Med 26, 587-596

126 Assogna, M. et al. (2013) Chemotherapy combined with regional

hyperthermia in locally advanced unresectable pancreatic cancer: clinical and

anthropological benefits Oncotherm J 7, 116-118

127 Liao, Q. et al. (2001) The anti-apoptotic protein BAG-3 is overexpressed in

pancreatic cancer and induced by heat stress in pancreatic cancer cell lines.

FEBS Lett 503 (151-157)

128 Mocan, T. et al. (2014) Photothermal Treatment of Human Pancreatic Cancer

Using PEGylated Multi-Walled Carbon Nanotubes Induces Apoptosis by

Triggering Mitochondrial Membrane Depolarization Mechanism. J Cancer, 5,

679-688

129 Guo, Y. et al. (2015) Overexpression of heat shock protein 27 (HSP27)

increases gemcitabine sensitivity in pancreatic cancer cells through S-phase

arrest and apoptosis. J. Cell. Mol. Med. 19, 340-350

130 Lindquist, S. (1988) The Heat-Shock Proteins. Annu. Rev. Genet 22, 631-677

131 Georgopoulos, C. (1993) Role of the Major Heat Shock Proteins as Molecular

Chaperons. Annu Rev Cell Biol. 9, 601-634

132 Arrigo, A. et al. (2007) Hsp27 (HspB1) and aB-crystallin (HspB5) as

therapeutic targets. FEBS Lett 581, 3665-3674

133 Garrido, C. et al. (2006) Heat Shock Proteins 27 and 70: Anti-Apoptotic

Proteins with Tumorigenic Properties. Cell Cycle 5, 2592-2601

35

802

803

804

805

806

807

808

809

810

811

812

813

814

815

816

817

818

819

820

821

822

823

824

Page 36: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

134 Schäfer, C. et al. (2011) Heat shock protein 27 as a prognostic and predictive

biomarker in pancreatic ductal adenocarcinoma. J. Cell. Mol. Med. 16, 1776-

1791

135 Liu, Q. et al. (2012) Role of heat shock protein 27 in gemcitabine-resistant

human pancreatic cancer: Comparative proteomic analyses. Mol Med Rep 6,

767-773

136 Kanani, H. et al. (2008) Standardizing GC-MS metabolomics. J Chromatogr

B Analyt Technol Biomed Life Sci. 871, 191-201

137 Wishart, D.S. et al. (2013) HMDB 3.0--The Human Metabolome Database in

2013. Nucleic Acids Res 41 (Database issue), D801-807

138 Wishart, D.S. et al. (2016) Cancer Metabolomics and the Human Metabolome

Database. Metabolites, DOI : doi:10.3390/metabo6010010

(http://www.mdpi.com).

139 Fiehn, O. (2002) Metabolomics--the link between genotypes and phenotypes.

Plant Mol Biol 48, 155-171

140 Beger, R. et al. (2006) Metabonomic models of human pancreatic cancer using

1D proton NMR spectra of lipids in plasma. Metabolomics 2, 125-134

141 Urayama, S. et al. (2010) Comprehensive mass spectrometry based metabolic

profiling of blood plasma reveals potent discriminatory classifiers of

pancreatic cancer. Rapid Commun Mass Spectrom 24, 613-620

142 Sugimoto, M. et al. (2010) Capillary electrophoresis mass spectrometry-based

saliva metabolomics identified oral, breast and pancreatic cancer-specific

profiles. Metabolomics 6, 78-95

36

825

826

827

828

829

830

831

832

833

834

835

836

837

838

839

840

841

842

843

844

845

846

847

Page 37: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

143 Napoli, C. et al. (2012) Urine metabolic signature of pancreatic ductal

adenocarcinoma by (1)h nuclear magnetic resonance: identification, mapping,

and evolution. J Proteome Res 11, 1274-1283

144 OuYang, D. et al. (2011) Metabolomic profiling of serum from human

pancreatic cancer patients using 1H NMR spectroscopy and principal

component analysis. Appl Biochem Biotechnol 165, 148-154

145 Bathe, O.F. et al. (2011) Feasibility of identifying pancreatic cancer based on

serum metabolomics. Cancer Epidemiol Biomarkers Prev 20, 140-147

146 Tesiram, Y.A. et al. (2012) Utility of nuclear magnetic resonance

spectroscopy for pancreatic cancer studies. Pancreas 41, 474-480

147 Kobayashi, T. et al. (2013) A novel serum metabolomics-based diagnostic

approach to pancreatic cancer. Cancer Epidemiol Biomarkers Prev 22, 571-

579

148 Kaur, P. et al. (2012) Metabolomic profiling for biomarker discovery in

pancreatic cancer. Int J Mass Spectrom 310, 44-51

149 Watanabe, M. et al. (2012) Metabolic Profiling Comparison of Human

Pancreatic Ductal Epithelial Cells and Three Pancreatic Cancer Cell Lines

using NMR Based Metabonomics. J Mol Biomark Diagn. DOI :

10.4172/2155-9929.S3-002 (https://www.ncbi.nlm.nih.gov)

150 Velliou, E.G. et al. (2014) Key Environmental Stress Biomarker Candidates

For The Optimization Of Chemotherapy Treatment Of Leukaemia. MJHS 1,

29-34

151 Fuentes-Garía, M. et al. (2015) A systematic framework for the design,

simulation and optimizationof personalized healthcare: Making and healing

bloodMaría. Computers and Chemical Engineering 81, 80-93

37

848

849

850

851

852

853

854

855

856

857

858

859

860

861

862

863

864

865

866

867

868

869

870

871

872

Page 38: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

152 Velliou, E. et al. (2014) A framework for the design, modeling and

optimization of biomedical systems. Comput Chem Eng 34, 225-236

153 Cunningham, D. et al. (2009) Phase III randomized comparison of

gemcitabine versus gemcitabine plus capecitabine in patients with advanced

pancreatic cancer. J Clin Oncol 27, 5513-5518

154 Neoptolemos, J.P. et al. (2016) Comparison of adjuvant gemcitabine and

capecitabine with gemcitabine monotherapy in patients with resected

pancreatic cancer (ESPAC-4): a phase 3 open-label randomised multicentre

trial. Lancet In press

155 Edmondson, R. et al. (2014) Three-dimensional cell culture systems and their

applications in drug discovery and cell-based biosensors. Assay Drug Dev

Technol 12, 207-218

156 Hoffman, A.S. (2012) Hydrogels for biomedical applications. Adv Drug Deliv

Rev 64, 18-23

157 Hoff, D.D.V. et al. (2013) Increased Survival in Pancreatic Cancer with nab-

Paclitaxel plus Gemcitabine. N Engl J Med 369, 1691-1703

158 Heinemann, V. et al. (2013) Gemcitabine plus erlotinib followed by

capecitabine versus capecitabine plus erlotinib followed by gemcitabine in

advanced pancreatic cancer: final results of a randomised phase 3 trial of the

'Arbeitsgemeinschaft Internistische Onkologie' (AIO-PK0104). Gut 62, 751-

759

159 Neoptolemos, J.P. et al. (2010) Adjuvant chemotherapy with fluorouracil plus

folinic acid vs gemcitabine following pancreatic cancer resection: a

randomized controlled trial. JAMA 304, 1073-1081

38

873

874

875

876

877

878

879

880

881

882

883

884

885

886

887

888

889

890

891

892

893

894

895

896

Page 39: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

160 Dahan, L. et al. (2010) Combination 5-fluorouracil, folinic acid and cisplatin

(LV5FU2-CDDP) followed by gemcitabine or the reverse sequence in

metastatic pancreatic cancer: final results of a randomised strategic phase III

trial (FFCD 0301). Gut 59, 1527-1534

161 Conroy T, et al. (2011) FOLFIRINOX versus gemcitabine for metastatic

pancreatic cancer. N Engl J Med 364, 1817-25

162 Van Cutsem, E. et al. (2009) Phase III trial of bevacizumab in combination

with gemcitabine and erlotinib in patients with metastatic pancreatic cancer. J

Clin Oncol 27, 2231-2237

163 Neoptolemos JP, et al.(2009) Adjuvant 5-fluorouracil and folinic acid vs

observation for pancreatic cancer: composite data from the ESPAC-1 and -

3(v1) trials. Br J Cancer 100, 246-250

164 Oettle H, et al. (2013) Adjuvant chemotherapy with gemcitabine and long-

term outcomes among patients with resected pancreatic cancer: the CONKO-

001 randomized trial. JAMA 310, 1473-1481

165 Uesaka et al. (2016) Adjuvant chemotherapy of S-1 versus gemcitabine for

resected pancreatic cancer: a phase 3, open-label, randomised, non-inferiority

trial (JASPAC 01). Lancet 388, 248–257.

39

897

898

899

900

901

902

903

904

905

906

907

908

909

910

911

912

913

914

915

916

Page 40: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

Figures Legends

Figure 1: Designing a bio-mimetic bio-inspired in vitro platform for pancreatic cancer

screening

Figure 2: Metabolism of healthy and cancer cells. Fluctuations of the micro-

environment and time evolution may accumulate mutations in normal tissue cells,

leading them to a cancer stem cell (CSC) phenotype or to a malignant cell. Normal

and cancer cell metabolism are characterized by activities of metabolic pathways.

40

917

918

919

920

921

922

923

924

925

926

927

928

929

930

931

932

933

934

935

936

937

938

939

940

941

Page 41: University of Surrey - Designing a bio-inspired bio …epubs.surrey.ac.uk/813320/1/Totti_et_al_2016_R1.docx · Web view. developed a highly porous (90% porosity) polyurethane (PU)

Tables Legends

Table 1: Advantages and disadvantages of current systems for in vitro studies for

pancreatic cancer

Table 2: 3D culture systems for pancreatic cancer screening

Table 3: Effect of environmental stress factors on the response of pancreatic cancer to

treatment

Table 4: Environmental stress response biomarkers candidates for pancreatic cancer

Table 5: Recent clinical trials of combinatory drug therapies for pancreatic cancer

41

942

943

944

945

946

947

948

949

950

951

952

953

954

955

956

957

958

959

960

961

962

963

964

965

966

967

968

969

970


Recommended