26
UNCORRECTED PROOF 1 Review 2 Oral delivery of anticancer drugs: Challenges and opportunities 3 Kaushik Q1 Thanki a , Rahul Gangwal b , Abhay T. Sangamwar b , Sanyog Jain a, 4 a Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar (Mohali), Phase X, Sector-67, 5 Punjab 160062, India 6 b Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar (Mohali), Phase X, Sector-67, Punjab 160062, India 7 8 abstract article info 9 Article history: 10 Received 3 December 2012 11 Accepted 26 April 2013 12 Available online xxxx 13 14 15 16 Keywords Q2 : 17 Oral delivery 18 Anticancer 19 Solubility 20 Permeability 21 GastroPlus 22 Nanoparticles 23 Lipid 24 Polymer 25 The present report focuses on the various aspects of oral delivery of anticancer drugs. The signicance of oral 26 delivery in cancer therapeutics has been highlighted which principally includes improvement in quality of 27 life of patients and reduced health care costs. Subsequently, the challenges incurred in the oral delivery of an- 28 ticancer agents have been especially emphasized. Sincere efforts have been made to compile the various 29 physicochemical properties of anticancer drugs from either literature or predicted in silico via GastroPlus. 30 The later section of the paper reviews various emerging trends to tackle the challenges associated with 31 oral delivery of anticancer drugs. These invariably include efux transporter based-, functional excipient- 32 and nanocarrier based-approaches. The role of drug nanocrystals and various others such as polymer 33 based- and lipid based-nanocarriers in the bioavailability enhancement along with their clinical outcomes 34 has also been discussed exhaustively. Furthermore, an insight on the various absorption mechanisms of 35 these nanocarriers across the gastrointestinal tract has also been highlighted. 36 © 2013 Published by Elsevier B.V. 37 38 39 40 41 42 Contents 43 1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 44 2. Challenges to the oral delivery of anticancer agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 45 2.1. Physicochemical properties of the drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 46 2.2. Biological barriers to drug delivery of anticancer drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 47 2.2.1. Transmembrane efux of drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 48 2.2.2. Pre systemic metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 49 3. Emerging trends in addressing the challenges to oral delivery of anticancer drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 50 3.1. Absorption enhancers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 51 3.1.1. P-Gp inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 52 3.1.2. Functional excipients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 53 3.2. Nanocarrier based approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 54 3.2.1. Drug nanocrystals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 55 3.2.2. Polymeric nanocarriers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 56 3.2.3. Lipid based nanocarriers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 57 3.2.4. Dendrimers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 58 4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 59 5. Future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 60 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0 61 62 1. Introduction 63 Cancer is dened as a complex series of disease condition caused 64 by persistent tissue injury and hostenvironment interactions. The 65 repeated exposure of carcinogens such as tobacco, ultraviolet light 66 and infections leads to various genetic (mutations), epigenetic (loss 67 of heterozygosity) and global transcriptome changes (via inamma- 68 tion pathways) and is associated with increased cancer risk [1]. 69 Owing to increased occurrence of cancer and worldwide prevalence 70 during the last decade, it has posed a great challenge to the health 71 care professionals. The latest WHO statistics suggests about 45% Journal of Controlled Release xxx (2013) xxxxxx Corresponding author. E-mail addresses: [email protected], [email protected] (S. Jain). COREL-06705; No of Pages 26 0168-3659/$ see front matter © 2013 Published by Elsevier B.V. http://dx.doi.org/10.1016/j.jconrel.2013.04.020 Contents lists available at SciVerse ScienceDirect Journal of Controlled Release journal homepage: www.elsevier.com/locate/jconrel Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drugs: Challenges and opportunities, J. Control. Release (2013), http:// dx.doi.org/10.1016/j.jconrel.2013.04.020

Oral delivery of anticancer drugs: Challenges and opportunities

  • Upload
    sanyog

  • View
    238

  • Download
    11

Embed Size (px)

Citation preview

Page 1: Oral delivery of anticancer drugs: Challenges and opportunities

1

2

3Q1

456

7

8910111213141516Q21718192021222324

39

40

4142

43

44

45

46

47

48

49

50

51

52

53

54

55

56

57

58

59

60

61

62

63

64

Journal of Controlled Release xxx (2013) xxx–xxx

COREL-06705; No of Pages 26

Contents lists available at SciVerse ScienceDirect

Journal of Controlled Release

j ourna l homepage: www.e lsev ie r .com/ locate / jconre l

Review

Oral delivery of anticancer drugs: Challenges and opportunities

F

Kaushik Thanki a, Rahul Gangwal b, Abhay T. Sangamwar b, Sanyog Jain a,⁎a Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar (Mohali), Phase X, Sector-67,Punjab 160062, Indiab Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), SAS Nagar (Mohali), Phase X, Sector-67, Punjab 160062, India

O

⁎ Corresponding author.E-mail addresses: [email protected], sanyogjain

0168-3659/$ – see front matter © 2013 Published by Elhttp://dx.doi.org/10.1016/j.jconrel.2013.04.020

Please cite this article as: K. Thanki, et al., Odx.doi.org/10.1016/j.jconrel.2013.04.020

Oa b s t r a c t

a r t i c l e i n f o

25

26

27

28

29

30

31

32

33

34

35

36

Article history:Received 3 December 2012Accepted 26 April 2013Available online xxxx

Keywords:Oral deliveryAnticancerSolubilityPermeabilityGastroPlusNanoparticlesLipidPolymer

ED P

RThe present report focuses on the various aspects of oral delivery of anticancer drugs. The significance of oraldelivery in cancer therapeutics has been highlighted which principally includes improvement in quality oflife of patients and reduced health care costs. Subsequently, the challenges incurred in the oral delivery of an-ticancer agents have been especially emphasized. Sincere efforts have been made to compile the variousphysicochemical properties of anticancer drugs from either literature or predicted in silico via GastroPlus™.The later section of the paper reviews various emerging trends to tackle the challenges associated withoral delivery of anticancer drugs. These invariably include efflux transporter based-, functional excipient-and nanocarrier based-approaches. The role of drug nanocrystals and various others such as polymerbased- and lipid based-nanocarriers in the bioavailability enhancement along with their clinical outcomeshas also been discussed exhaustively. Furthermore, an insight on the various absorption mechanisms ofthese nanocarriers across the gastrointestinal tract has also been highlighted.

© 2013 Published by Elsevier B.V.

3738

T CContents

UNCO

RRE1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

2. Challenges to the oral delivery of anticancer agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 02.1. Physicochemical properties of the drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 02.2. Biological barriers to drug delivery of anticancer drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

2.2.1. Transmembrane efflux of drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 02.2.2. Pre systemic metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

3. Emerging trends in addressing the challenges to oral delivery of anticancer drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 03.1. Absorption enhancers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

3.1.1. P-Gp inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 03.1.2. Functional excipients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

3.2. Nanocarrier based approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 03.2.1. Drug nanocrystals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 03.2.2. Polymeric nanocarriers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 03.2.3. Lipid based nanocarriers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 03.2.4. Dendrimers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 05. Future prospects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

65

66

67

68

1. Introduction

Cancer is defined as a complex series of disease condition causedby persistent tissue injury and host–environment interactions. The

69

70

[email protected] (S. Jain).

sevier B.V.

ral delivery of anticancer dr

repeated exposure of carcinogens such as tobacco, ultraviolet lightand infections leads to various genetic (mutations), epigenetic (lossof heterozygosity) and global transcriptome changes (via inflamma-tion pathways) and is associated with increased cancer risk [1].Owing to increased occurrence of cancer and worldwide prevalenceduring the last decade, it has posed a great challenge to the healthcare professionals. The latest WHO statistics suggests about 45%

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 2: Oral delivery of anticancer drugs: Challenges and opportunities

T

72

73

74

75

76

77

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

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

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

153

154

155

156

157

158

159

160

161

162

Q4

2 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

RREC

increase in the global cancer deaths by 2030, of which 70% would becontributed from developing countries like India [2]. With continuousupgradation in the field of science and technology, the need for ad-dressing the practical problems associated with the drug therapiesincreased proportionately. The major portion of cancer therapy tillthe last couple of decades was based on parenteral route of adminis-tration [3,4]. However, looking at the quality of life and need offollow-up therapy after the diagnosis of the disease, oral route hasgained major focus as compared to the parenteral route [4–6]. Oralroute is considered as one of the most abundant and traditionalways of drug delivery; main advantage being greatest safety, conve-nience and patient compliance. The possibility of tailor-made designas per physicochemical properties of the drug substances further in-creases the attraction of the scientific community. However, diverseproperties of drug substances, limitations in the choice of excipientsand principally, physiological barriers pose great challenge for designand development of oral drug delivery system.

The use of oral anticancer therapy affects the many clinically rele-vant aspects such as the following [7]:

1. An appropriate plasma drug concentration can be maintained toachieve a prolonged exposure of drugs to cancerous cells. Thiswill increase the efficacy and decrease the side effects of the anti-cancer drugs.

2. Modulation of drug release from the dosage forms also provides anadded advantage compared to that in other routes of administration.

3. It further facilitates the use of more chronic treatment regimens.This is especially important for cell cycle specific agents, especiallythose of predominantly cytostatic effect such as angiogenesis inhibi-tors and signal transduction inhibitors. For these agents, prolongedexposure to the drug may lead to pharmacodynamic advantagesover intermittent intravenous administration.

4. Oral chemotherapy avoids the discomfort of injection and can beconducted at home. This approach may enhance the patient coop-eration and their quality of life, which is an important issue andthus deserves high attention for any medical treatment.

5. The risks of infection and extravasations associated with intrave-nous infusion lines is avoided.

6. The treatment cost for the patient can be highly reduced due toavoidance of hospitalization, sterile manufacturing and trainedpersonnel assistance.

7. Apart from the therapeutic applications, oral therapy can also beexplored in the segment of prophylactics due to high level ofease in administration.

An interesting study has been carried out to evaluate the patient'spreference for route of administration and it was found that almost

UNCO

Fig. 1. Chemotherapeutic agents implemented to combat cancer. £P-gp modulator; #novelmetalloprotease inhibitors; ¥tetracycline analogue; ‡selective nonpeptide potent MMPI; TKI

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

78.7%wanted themselves to be treated by oral route for recurring breastcancer disease, whereas nearly 2.7% preferred parenteral route while18.6% landed with no preference [8]. Synchronizing with these results,the current scenario for development of new drug molecules has alsorapidly shifted towards oral delivery. Approximately 20 molecules arealready present in market for oral therapy of cancer, whereas a numberof them are pipeline. This clearly indicates the developer's insight andintentions for oral delivery. Fig. 1 shows the list of drugs presentlyutilized for cancer therapy [4,9–11].

However, oral delivery of anticancer drugs is a great challengeowing to their peculiar physicochemical properties, and physiologicalbarriers such as pre-systemic metabolism and gastrointestinal insta-bility. Upon oral administration of such drugs, only a fraction ofdose is available to systemic circulation for execution of therapeuticresponse e.g. oral bioavailability of paclitaxel, docetaxel, doxorubicin,tamoxifen, etc. is in the range of 5–20% [12–15]. Broadly, this could beattributed to low aqueous solubility, poor intestinal permeability,high level of P-glycoprotein (P-gp) efflux and pre-systemic metabo-lism. The P-gp efflux also has a key role in the execution of multidrugresistance in the tumor cells and thereby needs special considerationwhile designing the formulation of poor biopharmaceutical proper-ties, as the amount which is required to achieve the therapeutic re-sponse might be very high ultimately leading to multidrug resistance.

Furthermore, cost of manufacturing novel formulations of theexisting parenteral drugs and limited therapeutic window of the anti-cancer drugs leading to sub-therapeutic or toxic dose, also restrictsthe developability for oral route of administration [16]. However, recentadvances in nanotechnology based drug delivery system posed poten-tial advantages in overcoming these limitations. This includes polymer-ic nanoparticles, polymeric micelles, microemulsion, self-emulsifyingdrug delivery systems (SEDDS), carbon nanotubes, layersomes, lipo-somes, lipid–drug conjugates, nanocrystals, etc.

The therapeutic efficacy of the formulation depends upon its capa-bility to deliver the drug at the right place and at the right time inamount adequate enough to yield a therapeutic response. Compara-tive therapeutic equivalence of oral and intravenous routes has beenstudied for wide variety of drugs and promising results were ob-served in most of the cases. Cyclophosphamide yields no statisticalsignificant difference in the area under the plasma disappearancecurve (AUC) and generated similar cytotoxic metabolic products uponadministration through oral and parenteral routes thereby suggestingthe therapeutic equivalence, irrespective of the route of delivery [17].Paclitaxel in nanoparticulate dosage form administered by oral routehad shown promising tumor reduction in animals compared tocommercially available intravenous formulation at 50% reduced dose[18]. Co-administration of cyclosporin A further potentiated its oral

oral taxane; ΔDeoxycytidine-type antimetabolite; €Oral fluoropyrimidine; MMPImatrixtyrosine kinase inhibitor; FTIfarnesyl transferase inhibitor.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 3: Oral delivery of anticancer drugs: Challenges and opportunities

T

163

164

165

166

167

168

169

170

171

172

173

174

175

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199

200

201

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

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

252

253

254

255

256

257

258

259

260

261

262

3K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

NCO

RREC

bioavailability, due to inhibition of the P-gp efflux pump and CYP 3A4,both being limitations for oral bioavailability [19]. Similarly, topotecanhas also been found to be equally effective irrespective of the route ofadministration with upper hand in reduced toxicity via oral route[20,21]. The other drugs which have been evaluated include docetaxel,paclitaxel, doxorubicin, cisplatin [22], ifosfamide/mesna combination[23] and melphalan [24] to name a few. The studies suggest that byvirtue of appropriate pharmaceutical/pharmacological interventions,the inherent problems associated with oral route (owing to variousphysiological barriers) can be overcome in comparison to intravenousroute of administration.

The present review highlights comprehensive coverage of variouschallenges encountered for efficient oral delivery of anticancer drugs.In contrast to other literature reports published recently, a correlationbetween physicochemical properties of these anticancer drugs withits in vivo performance has been included specifically. In absence ofthe availability of these data, appropriate in silico predictions havebeen made. Furthermore, various conventional and novel approachesincluding emerging trends to overcome these challenges have alsobeen discussed in detail with special emphasis on polymer and lipidbased drug delivery systems.

2. Challenges to the oral delivery of anticancer agents

Bioavailability is staging of in vivo performance of drug and re-flects the rate and extent to which the drug is absorbed into the sys-temic circulation and thereby available for therapeutic response. Thekey factors affecting the oral bioavailability of drug include its stabil-ity in the gastrointestinal tract, aqueous solubility, dissolution ratefrom the dosage form, intestinal epithelium permeability, stabilityagainst intestinal and liver cytochrome P450 metabolic enzymes,and P-gp efflux pump. Therefore, on these grounds the principal chal-lenges to the oral delivery can be categorized broadly into physico-chemical properties of the drugs and physiological barriers posed bythe body (Fig. 2).

2.1. Physicochemical properties of the drugs

The critical physicochemical properties of the drug affecting itsoral deliverability include solubility and permeability which are fur-ther dependent on the fundamental properties such as log P andpKa. The classical relationship between the solubility, permeabilityand potency has been reported by Lipinski according to which thequantity sufficient solubility of the drug can be determined on thebasis of the potency and permeability of the drug [25]. The in vivo per-formance (biopharmaceutics) of a drug is dependent on pharmacoki-netics (ADME profile) and pharmacodynamics (extent of clinicalresponse). The absorption segment of the pharmacokinetics can bedetermined by the Fick's First law of diffusion, which states that flux(J) of drug for systemic exposure is directly proportional to the per-meability coefficient (inclusive of drug efflux) and drug concentrationin the gastrointestinal lumen (inclusive of solubility, dissolution andstability of drug within the GIT) [26].

U 263

264

265

266

267

268

269

270

271

272

273

274

275

• Intrinsic solubility• Permeability• Stability

Physicochemical properties of

drugs

• Gastrointestinal transit time• Absorption window• Transmembrane efflux of drugs• Pre-systemic metabolism

Biological barriers

Limited oral bioavailability

of drugs

Fig. 2. Schematic representation of the various challenges to the oral delivery of drugs.

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

The limited oral bioavailability of the drugs can therefore be desig-nated as solubility/dissolution rate limited, permeability limited, andboth solubility and permeability limited [27]. The quantity sufficientsolubility of a drug candidate could be predicted on the basis of dosenumber and dose number ≫ 1 is considered as poorly soluble [28].According to BCS classification, these candidates are categorized as IIand need attempt for solubility enhancement which could otherwiseresult in either solubility or dissolution limited absorption leading topoor bioavailability. Classical examples include tamoxifen, rubitecan,sorafenib, gefitinib, etc. On the other hand, candidates with sufficientsolubility but poor permeability are categorized as III and includecyclophosphamide, anastrozole, letrozole, doxorubicin, methotrexate,etc. The permeability values b 10 × 10−6 cm/s are considered poorand need appropriate efforts for improvement [29]. However, veryfew (~25% of literature of drug compounds) pose permeability relatedissues (Table 1). But at the same time, >60% drugs are substrate forone or other types of efflux, suggesting dominating role of drug effluxin the oral bioavailability. Further, >65% of anticancer drugs are avail-able in oral dosage form for clinical use but very few of themare actuallyused which could be attributed to the limited oral bioavailability owingto poor physicochemical properties and efflux mechanisms. However,the exact usage needs to be identified so that rationalized drug deliverysystems can be designed to propagate research in the field of oral anti-cancer drug delivery.

2.2. Biological barriers to drug delivery of anticancer drugs

2.2.1. Transmembrane efflux of drugsTransmembrane efflux of drugs is defined as the expulsion of the

drugmolecules across the cellularmembrane from the cells via a clinical-ly significant systematic transportation system such as P-glycoprotein(P-gp), breast cancer resistant protein (BCRP), cytoplasmic transport,multidrug resistant associated protein (MRP), flurochrome efflux,methotrexate efflux (folates), etc. [91–93]. Table 2 represents an ex-haustive list of anticancer drugs that are potential substrate for variousmembrane efflux proteins present in liver and intestine thereby limit-ing their oral delivery [94].

P-Glycoprotein, encoded by the multidrug resistance-1 (MDR1)gene, localized in enterocytes, is one of the significant efflux trans-porters leading to the excretion of drug back in to the intestinallumen. It is extensively distributed in intestinal epithelia, hepatocytes,kidneys, various glands and capillary endothelial cells comprisingblood–brain and blood–testis barriers. This is a membrane associatedprotein belonging to the superfamily of ATP binding cassette (ABC)transporters with the molecular weight of 170 kDa and N terminal gly-cosylation. Its structure constitutes two homologous chains of equallength, each comprising six units of transmembrane domains and twoATP binding sites separated by a flexible linker polypeptide regionbetween the two homologous chains [95]. It mainly operates at threemajor locations, luminal (apical) membrane enterocytes: the drug islimited by entering in the body; canalicular membrane of hepatocytes:increased elimination in to bile and urine; and, sensitive tissues such asbrain, lymphocytes, testis, and fetal circulation: limiting the drug pene-tration [96]. Most of the anticancer drugs are the substrates for P-gpincluding paclitaxel, docetaxel, etoposide, vinblastine, vincristine anddoxorubicin to name a few (Table 1).

P-Gp expresses two types of ATPase activity: basal stimulated byendogenous lipids and other hydrophobic peptides and drug stimu-lated ATPase activity. Different drugs owing to their unique bindingimpart different types of ATPase activities on the P-gp. Dependingon this property of the drug substances, they can be classified inthree different categories [97]. Class I agents (e.g. vinblastine, verapa-mil, and paclitaxel) stimulate ATPase activity at low concentrationsbut inhibit the activity at high concentrations. Class II compounds(e.g. bisantrene, valinomycin, and tetraphenylphosphonium) enhanceATPase activity in a dose dependent manner without any inhibition.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 4: Oral delivery of anticancer drugs: Challenges and opportunities

UNCO

RRECTED P

RO

OF

276

277

278

279

280

281

Table 1t1:1

t1:2 Physicochemical properties of various anticancer drugs.

t1:3 Drug pKa Log P Solubility(mg/ml)

Permeability(cm/s × 10−6)

Efflux Clinically used orally? Reference

t1:4 5-Fluro uracil 11.5, 7.26 −0.9a 1.070a 65 Substrate Yes [30]t1:5 Altretamine 5.65, 0.09 1.25a 0.091a 877 Non-substrate Yes [31]t1:6 Anagrelide 3.08 1.13a 0.0322 35 Substrate Yes [32,33]t1:7 Anastrazole 3.17 2.1a 0.5a 324 Non-substrate Yes [34]t1:8 Bexarotene 4.4 6.9a 0.00385 266 Non-substrate Yes [35]t1:9 Bicalutamide 12 2.92a 0.005 170 Noncompetitive inhibitora Yes [36,37]t1:10 Bleomycin 13.91, 7.68 −3.62a 8.39 b1 Substrate No [38]t1:11 Capecitabine 8.86 0.4a 26a 33 Non-substrate Yes [39]t1:12 Carboplatin None −2.30a 4.50 11 Substrate No [40]t1:13 Cisplatin 3.16, 0.77 −2.53a 2.530a 18 Non-substratea No [41,42]t1:14 Cladribine 1.91 0.498a 3.42 25 Substrate No [43]t1:15 Cyclophosphamide 8.91 0.73a 12.5 446 Substrate Yes [44]t1:16 Cytarabine 4.30 −2.8a 25.8 7 Substrate No [39]t1:17 Dasatinib 10.29,7.07 1.8a 0.0501 30 Substratea Yes [45,46]t1:18 Docetaxel 10.97 4.1a 0.000025a 1 Substratea No [30,47]t1:19 Doxorubicin 9.99, 6.74 1.3a 0.429 22 Substratea No [30,42]t1:20 Erlotinib 10, 6.15 2.7a 0.4a 194 Substratea Yes [48,49]t1:21 Etoposide 9.21 0.698a 0.0587a 5 Substratea Yes [47,50]t1:22 Exemestane None 3.7a 0.00789a 48 Substrate Yes [51]t1:23 Fadrozole 11 2.18 0.693 280 Substrate No –

t1:24 Finasteride 11.88 3.03a 0.0117a 2 Substrate Yes [52]t1:25 Fludarabine 6.33 −2.83 2.76 17 Substrate Yes –

t1:26 Gefitinib 7.08, 5.32 4.85a 0.0367 206 Inhibitora Yes [53,54]t1:27 Hydroxycarbamide 12.52, 10.82, 9.99 −1.4a 1,000,000a 10 Substrate Yes [55]t1:28 Ibandronic acid 10.09, 8.18 2.33a 31.1 18 Substrate Yes [56]t1:29 Idarubicin 10.06, 8.41 3.0a 0.0772a 38 Substratea Yes [38,57]t1:30 Ifosphamide 14.83, 11.27 0.8a 3.780a 230 Substrate No [58,59]t1:31 Imatinib 13.45 1.99a 0.0412 78 Substratea Yes [45,60]t1:32 Irinotecan 8.1 3.2a 0.106 22 Substratea No [61,62]t1:33 Lapatinib 6.55, 4.27 5.1a 0.000223 121 Substratea Yes [63,64]t1:34 Lenalidomide 9.55 −1.09a 0.4a 10 Substrate Yes [56]t1:35 Letrozole 2.19 2.49a 0.0329 384 Substrate Yes [65]t1:36 Lonafarnib 13.22 5.59 0.0167 656 Inhibitora No [66]t1:37 Melphalan 1.93 −1.70a 0.1a 456 Non-substratea Yes [67,68]t1:38 Mercaptopurine 11.25, 7.54 −0.4a 6.85a 16 Substrate Yes [69]t1:39 Mesna None 0.78 8.55 61 Non-substrate No –

t1:40 Methotrexate 4.7a −1.4a 2.60a 58 Non-substratea Yes [70,71]t1:41 Mitomycin 10.9a −1.6a 8.43a b1 Inhibitora No [38,72]t1:42 Nilutamide 9.68 1.8a 0.029 148 Non-substrate Yes [73]t1:43 Oxaliplatin 3.03, 0.1 −0.47/1.73a 2.75a b1 Non-substratea No [74,75]t1:44 Paclitaxel 11.51 3.5a 0.00612 5.2a Substratea No [28,30,42]t1:45 Procarbazine 11.97 0.06a 1.420a 144 Non-substrate Yes [76]t1:46 Raloxifene 9.30, 8.49 5.7a 0.00025a 157 Substratea Yes [77,78]t1:47 Rubitecan 0.87 1.52 0.00875 286 Non-substrate Yes –

t1:48 Sorafenib 11.50, 8.53 3.8a 0.00199 243 Substratea No [39,45]t1:49 Sobuzoxane 5.12 1.49 0.465 1 Substrate Yes –

t1:50 Sunitinib 12.60, 10.33 2.5a 0.00308a 51 Substratea Yes [39,46]t1:51 Tamibarotene 11.45 5.87 0.0101 142 Non-substrate Yes –

t1:52 Tamoxifen 8.40 7.87a 0.000167a 445 Substratea Yes [79,80]t1:53 Temozolomide 10.33 −0.75 0.951 78 Non-substrate Yes –

t1:54 Thalidomide 9.87 0.04a 0.545a 10 Non-substrate Yes [31]t1:55 Thioguanine 11.51, 8.67 −0.46 0.0658 17 Non-substrate Yes –

t1:56 Tipifarnib 5.95, 3.61 4.94 0.00142 389 Inhibitora No [81]t1:57 Topotecan 11.7 0.8a 1a 159 Substratea Yes [82,83]t1:58 Toremifene 8.33 6.35a 0.000801 440 Substrate Yes [84]t1:59 Treosulfan None −2.11 922.0 10 Non-substrate Yes –

t1:60 Trilostane 5.51 1.76 0.117 9 Substrate No –

t1:61 Trimetrexate 7.67, 4.08 1.99 0.0318 108 Substrate No –

t1:62 Ubenimex 8.62 −1.79 7.36 110 Non-substrate No –

t1:63 Vinblastine 14.55, 6.10 1.68a 0.0138 8 Substratea No [47,85]t1:64 Vincristine 5a 1.16a 0.003a 3 Substratea No [47,86]t1:65 Vinorelbine 14.31, 6.73 1.32a 0.00206 11 Substratea No [86,87]t1:66 Vorinostat 9.2a 12.84, 10.70 0.512 255 Substrate Yes [88]

a Experimental/reported values; rest of values have been predicted in silico. Briefly, for predicting solubility and permeability, 3D structures of molecules were sketch usingSYBYL7.1 [89]. The molecules were later minimized by applying Tripos molecular mechanics force field with conjugate gradient method. The minimization was terminatedwhen the energy gradient convergence criterion of 0.05 kcal/mol was reached or when the 10,000 steps minimization cycle was exceeded. Minimized molecules were then sub-mitted to ADMET Predictor 5.5, GastroPlus, Simulations Plus, USA for prediction of physicochemical properties. All the calculations were performed at pH 6.8. The substrate spec-ificity for efflux was predicted using web server developed based on Support Vector Machine and molecular docking methods [90].t1:67

4 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

In contrast, Class III compounds (e.g. cyclosporin A, rapamycin, andgramicidin D) inhibit both basal- and verapamil-stimulated ATPaseactivities.

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

The mechanism of drug efflux by P-gp can be assessed on the basisof various models such as pore model, flippase model, and hydropho-bic vacuum cleaner (HVC) model, among which HVC model has

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 5: Oral delivery of anticancer drugs: Challenges and opportunities

T

OF

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

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

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

357

358

359

360

361

362

363

364

365

366

367

368

Table 2t2:1

t2:2 List of drugs that are potential substrates for various membrane efflux proteins.

t2:3 Membrane effluxproteins

Drug

t2:4 ABCB1(P-Glycoprotein)

Actinomycin D, Daunorubicin, Docetaxel, Doxorubicin, Etoposide, Mitoxantrone, Paclitaxel, Teniposide, Vinblastine, Vincristine, Mitomycin C

t2:5 ABCB4 (MDR2/3) Daunorubicin, Paclitaxel, Vinblastinet2:6 ABCB11 (BSEP) Vinblastinet2:7 ABCC1 (MRP1) Daunorubicin, Doxorubicin, Etoposide, Methotrexate, Vinblastine, Vincristinet2:8 ABCC2 (MRP2) Camptothecin, Cisplatin, Docetaxel, Doxorubicin, Epirubicin, Etoposide, Methotrexate, Paclitaxel, SN-38 (active metabolite of irinotecan), Vincristinet2:9 ABCC3 (MRP3) Etoposide, Etoposide glucuronide, Methotrexate, Teniposide,t2:10 ABCC4 (MRP4) 6-Mercaptopurine, 6-Thioguanine, Methotrexatet2:11 ABCC5 (MRP5) 5-Flurouracil, 6-Mercaptopurine, Cisplatin, Doxorubicin, Methotrexate, Oxaliplatin, Thioguanine, Pemetrexedt2:12 ABCC6 (MRP6) Cisplatin, Doxorubicin, Etoposide, Teniposidet2:13 ABCG2 (BCRP) Daunorubicin, Doxorubicin, Epirubicin, Epirubicin, Gefitinib, Genistein, Imatinib, Irinotecan, Methotrexate, Methotrexate diglutamate, Methotrexate

triglutamate, Mitoxantrone, Quercetin, SN-38 (active metabolite of irinotecan), SN-38 glucuronide, Teniposide, Topotecan.t2:14 OAT 1 Methotrexate, Azathioprine, Doxorubicin, 5-fluouracilt2:15 MCT Ifosfamide and its metabolites, S-carboxymethylcysteine, thiodiglycolic acid

t2:16 BSEP: Bile salt efflux pump; MDR: Multidrug resistance; MRP: Multiple resistance protein; BCRP: Breast cancer resistance protein; OAT: Organic anionic transporter; MCT:t2:17 Monocarboxylate transporter.

5K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

gained wider acceptance [98]. ATP binding and hydrolysis are the im-portant processes for the drug efflux. At the cost of two molecules ofthe ATPs, one molecule of the drug is effluxed. The drug efflux occursin two cycles, each cycle consuming one ATP molecule. The first cyclestarts with the binding of the drug and ATP at their respective sitesfollowed by the conformational change which is regained in the sec-ond cycle at the cost of hydrolysis of yet another ATP molecule [99]. Itis generally found to be overexpressed in the multidrug resistant cellsand is capable to expelling large variety of non-host compounds fromthe cells. It acts both at intracellular compartments and cell surface.P-Gp has major contributions in ADME of the drug molecules dueits wider spectrum of presence throughout the body. This has alsobeen supported experimentally where the oral bioavailability of anti-cancer drugs such as paclitaxel has been increased significantly inP-gp knockout mice [100].

2.2.2. Pre systemic metabolismOral bioavailability is a cumulative function of fraction of dose

absorbed through the gastrointestinal tract, fraction of drug absorbedfrom the entero-hepatic circulation and drug available post first passhepatic metabolism [101]. Gastro-intestinal and hepatic availability isdefined as the drug escaping the metabolizing effects of the gastro-intestinal tract and the liver. The gastro-intestinal metabolism, alsoreferred as luminal metabolism is performed by digestive enzymessecreted by pancreas such as amylase, lipase, and peptidases andfrom the bacterial flora present especially in the lower part of the gas-trointestinal tract. The first pass intestinal metabolism includes thebrush border metabolism and the intracellular metabolism [102].The brush border activity is generally to its highest in the proximalsmall intestine and is destined by enzymes such as alkaline phospha-tase, sucrase and isomaltase and various peptidases [103]. The intra-cellular metabolism in the gut is principally carried out by extra-hepatic microsomal enzymes present within the cytoplasm on theendoplasmic reticulum. Cytochrome P4503A family, especially CYP3A4, phase I metabolizing enzymes, are present in the enterocyteswhich leads to the metabolism of the drug substances at the gastroin-testinal wall. Phase II metabolizing enzymes such as glutathione-S-transferases, esterases, etc. are also reported to present in the intestine[104]. Though intestinal epithelium acts as a site for pre-absorptivemetabolism, contributing to low bioavailability of therapeutic peptidesand ester type drugs like capecitabine [105], it can also serve as a keytarget for delivery of ester or amide prodrugs.

Once the drug gets absorbed from the gastrointestinal tract, it en-ters in to the entero-hepatic portal vein and reaches to liver, where afraction of absorbed dose is metabolized, being referred as First PassHepatic Metabolism (FPHM). Liver is the hub of various enzymes

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

ROand is referred as “metabolic clearing house” for both endogenous

chemicals (e.g., cholesterol, steroid hormones, fatty acids, and pro-teins) and xenobiotics [106]. This first pass metabolism is consideredas major contributor for low oral bioavailability of many drugs e.g. ta-moxifen [15]. Worsening the case, the drugs which are substrates forcytochromes are also substrates for P-gp, thereby leading to furtherdecrease in the bioavailability. Both these work in tandem and needscareful consideration while designing of oral drug delivery systemfor their substrates, e.g. level of CYP 3A4 decreases from proximal tosmall intestine whereas P-gp expression increases in same flow [107].

3. Emerging trends in addressing the challenges to oral delivery ofanticancer drugs

Nevertheless, in spite of abovementioned challenges, oral delivery ofvarious anticancer drugs has been evaluated for their efficacy and toxic-ity profiles. Themost conventional approach includes co-administrationof a therapeutic agent or functional excipient that either circumvents thebiological barriers or facilitates the absorption of the drug across gastro-intestinal tract by altering the physicochemical properties of drug sub-stances. Recently, carrier based approaches have been implementedwhich can bypass majority of the challenges and is able to achievedesired delivery in most efficient form. However, the selection, designand development of drug specific carrier system are a state-of-art andrequire thorough understanding of the physicochemical properties ofdrug substances and its behavior in physiological conditions. The subse-quent sections report various such approaches implemented to achieveoral delivery of various difficult-to-deliver anticancer drugs. Fig. 3 dem-onstrates various strategies to improve the oral bioavailability of drugsubstances.

3.1. Absorption enhancers

3.1.1. P-Gp inhibitorsTransmembrane efflux of drugs can be tackled by co-administration

of various P-gp modulators or inhibitors along with the drugs. The po-tential mechanisms by which the inhibition of efflux pump occurs in-clude altered membrane fluidity, inhibited ATPase activity, blocking ofdrug binding site, decreasing P-gp expression (e.g. Peceole), depletionof ATP (e.g. Pluronics), interaction with membrane (e.g. Triton X 100)and interference with the ATP binding sites [108]. Fig. 4 reflects variousP-gp based approaches for improving the oral bioavailability of drugs.

P-gp inhibitors/modulators have been broadly classified to threeclasses viz. first generation, second generation and third generationinhibitors [93]. First-generation comprised of pharmacological agentsthat act as competitive inhibitors and hence block the drug efflux

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 6: Oral delivery of anticancer drugs: Challenges and opportunities

T

PRO

OF

369

370

371

372

373

374

375

376

377

378

379

380

381

382

383

384

385

386

387

388

389

390

391

392

393

394

395

396

397

398

399

400

401

402

Fig. 3. Strategies to improve the oral bioavailability of drug substances.

6 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

RREC

e.g. verapamil and cyclosporin A. But considering their poor bindingaffinities (needing higher doses to achieve desired results) and pharma-cokinetic interactions (due to induction/inhibition of CYP 3A enzymes)these are not clinically advisable. Cyclosporin A is widely used as P-gpinhibitor for improving the oral bioavailability of various drug sub-stances. About 10-fold increase in the oral bioavailability of paclitaxel[109] and docetaxel [110] was observed upon co-administration withcyclosporin A. Second generation included those lacking pharmacolog-ical activity andwere developedwith the intention of high P-gp bindingand inhibiting effect alongwith lower toxicities. However, they fail to re-main inert with CYPs thereby limiting their use e.g. the cyclosporin A an-alogue valspodar (PSC833). Therefore the third generation came in toplay and highly P-gp specific inhibitors such as elacridar (GF120918),zosuquidar (LY335979) and tariquidar (XR9576) were developed.

This approach is scarcely used clinically owing to associated clinicalcomplications such as suppression of immune system thus causing longterm medical complications. Hence, the novel approach of exploitingsimilar properties of excipients can be sought. Some commonly used

UNCO

P-gp inhibition

Decrease in P-gp

expression

Depletion of ATP

Inhibition of ATPase activity

Interference with ATP

binding sites

Blockade of drug

binding site

Interaction with

membrane

Altered membrane

fluidity

First• The

efflu• 10-f

doc

S•

Third• High• E.g.

Tari

Fig. 4. Various P-gp based approaches for im

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

EDfunctional excipients, such as surfactants, polymers, lipids, etc. can be

employed as bioavailability enhancers [98].

3.1.2. Functional excipientsA large variety of the functional excipients have been studied to in-

crease the oral bioavailability of drugs. Mechanistically, they are foundto modulate the activity of the P-gp efflux pump, facilitate wetting, in-crease solubilization, increase permeability across the gastrointestinaltract, etc. These include polysaccharides, polyethylene glycols and de-rivatives, surfactants, lipids, thiolated polymers and amphiphilic blockcopolymers to name a few.

3.1.2.1. Natural polymers. The natural polymers such as dextrans, an-ionic gums and sodium alginate are reported to inhibit the P-gp effluxpumps [111]. Anionic gums such as xanthan gum, gellan gum, algi-nates, flavicam and ascophyllum with carboxyl functional groups orsalts thereof increase serosal transport of drugs such as vinblastineand doxorubicin by inhibiting their efflux [112]. In addition, some

generationse act as competitive inhibitors and hence block the drug x. E.g. Verapamil and cyclosporin Aold increase in the oral bioavailability of paclitaxel and etaxel upon co-admintration with Cyclosporin A

econd generationLacks pharmacologiocal activity, high p-gp binding and inhibiting effect and lower toxicities. Fail to remain inert with CYPs thereby limiting their use. E.g. Valspodar (PSC833).

generationly specific P-gp inhibitors Elacridar (GF120918), Zosuquidar(LY335979) and quidar (XR9576).

proving the oral bioavailability of drugs.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 7: Oral delivery of anticancer drugs: Challenges and opportunities

T

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

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

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

527

528

529

530

531

7K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

polymers such as acacia also possess MDR reversal capability e.g. in-tracellular accumulation of epirubicin was significantly increasedwhen tested against MDR Caco-2 cell lines in the presence of acacia[113].

Recently, amember of flavanoid class, quercetin, has been evaluatedas a potential P-gp modulator and CYP modulator [114]. Quercetin isfound to interact either with the ATP binding site or with the substratebinding site of P-gp. Co-administration of quercetin with tamoxifen cit-rate has led to an increase in relative bioavailability by about 1.5 fold, in-dicative of its inhibitory effect on transmembrane efflux and CYPs [15].Similar appreciation in the oral bioavailability in the order of 2-fold wasalso observed with paclitaxel [115], doxorubicin [116] and etoposide[117]. Furthermore, co-administration of other functional agents suchas narigrin, geniestein, morin, biochanin A, schisandrol B, kaempferol,myricetin, silymarin, baicalein, recombinant interleukin-2 (pretreat-ment) and curcumin has also been reported to substantially increasethe oral bioavailability of anticancer agents such as paclitaxel, docetaxel,doxorubicin, etoposide, etc. Similarly, quinidine is also found to inhibitthe carrier mediated efflux of etoposide when tested against Caco-2cell monolayers, rat and human intestine. The inhibitory effect wasfound to be concentration- and temperature-dependent suggestingthe involvement of active process [118].

3.1.2.2. Polyethylene glycol (PEG) and its derivatives. These are wellreported to inhibit the P-gp efflux of drug compounds, although themechanistic understanding is still elusive [119–122]. PEGs were foundto inhibit the secretory transport of various compounds such as rhoda-mine 123, paclitaxel, doxorubicin, etc. irrespective of the molecularweights, when tested against Caco-2 cell monolayers [120,121]. How-ever, PEGs withmolecular weight b 200 lacked the P-gp interaction ca-pabilitywhile satisfactory P-gp inhibitionwas observed in case of >300molecular weight [123]. The P-gp inhibition capability is also retainedwhen PEGylation of drug is practiced, e.g. PEGylated paclitaxel poseshigher oral bioavailability as compared to plain paclitaxel [124]. Apartof efflux inhibition, solubilization potential of PEGs also contributes sig-nificantly in increasing the oral bioavailability of drugs. Similar inhibito-ry response on the P-gp efflux of doxorubicin was also observed inMCF-7/ADR cells when administered in the micellar form preparedwith PEGylated phosphatidylethanolamine [125]. The inhibitory activi-ty of PEG could be further enhanced by thiol modification and about3.3-fold increase in the transport of rhodamine with PEG-g-PEIco-polymer was observed with significant decrease in the secretorytransport at tested concentrations as compared to free rhodamine. Sub-strate competition or ATP depletion was proposed to be the probablemechanism of P-gp inhibition by such novel grafted polymer systems[126].

3.1.2.3. Thiolated polymers. Thiolated polymers have recently beenstudied for their P-gp and CYP enzyme inhibitory activity [126–129].The rationale for such studies was based on the feasibility of the cova-lent binding of sulfhydryl substituted purines with P-gp [130]. Thethiolated polymer chitosan-4-thio-butylamidine (chitosan-TBA) signif-icantly increased the absorptive transport (+118%) and markedlyreduced the secretory transport (−37%) of rhodamine 123 [131]. Thisinhibitory effect was attributed to the capability of the sulfhydrylgroup to react with the cysteine residues located within the Walker Aconsensus sequences of each of the two ATP binding domains of P-gp[132].While these cysteine residues are not important for P-gp functionthere lays a probability of steric hindrance at the catalytic activities ofthe P-gp [133,134]. Additionally, considering the non-penetrating char-acteristics of the modified chitosan, due to high molecular weight, theinhibitory activity ismainly concentrated at the cell surface [131]. In ad-dition, a significant decrease in the transepithelial electrical resistancein the Caco-2 cell monolayers was also observed in the presence ofchitosan [135]. It was further revealed that the modified chitosans arecapable of altering the microviscosity of the surrounding lipid bilayer

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

and thus imparts molecular conformational changes in the structureof P-gp, thereby inhibiting the efflux of drugs. Additionally, themolecu-lar weight of these modified chitosans also plays a crucial role inexhibiting the polymeric characteristics. At lower molecular weight,lower mucoadhesive and cohesive properties are exhibited, which arebeneficial for immobilization of thiol groups on polymeric backbone,but at the same time the higher interpenetration is exhibited, hence-forth an optimum molecular weight of chitosan should be chosen tohave desired results [136]. The added advantage of these modified chi-tosan resides in the fact that they are not significantly absorbed fromthe GIT and remain concentrated on the gastrointestinal membrane[137]. However, the gastrointestinal irritation of the said polymersshould be carefully monitored. Thiolated polycarbophil has also beenfound to significantly improve the plasma levels of paclitaxel and reduc-tion in tumor growth when administered orally in mammary cancerbearing rats. The effects were attributed to the P-gp inhibition propertyof these novel polymers [138].

Mechanistically, the alteration in the concentration of a tyrosinephosphatase inhibitor, glutathione, initiation of the tight junction open-ing cascade is also reported in the literature [139,140]. Glutathione iscapable of raising the absorptive transport to 200% and lowers thesecretory transport to−42%, viamodulation of tight junctions. Concur-rently, the thiolated polymer also prevents the oxidation of the glutathi-one at the mucosal surface [141]. The cysteine groups were found tohave very critical role in the inhibition of the tyrosine phosphatases,enzyme responsible for the dephosphorylation to render the tight junc-tions in closed state [142]. The hypothesis could be corroborated by theexperimental finding that chitosan–N-acetyl cysteine conjugates in-creased the absorptive transport of rhodamine-123 by 1.5 fold anddecreased the secretory transport by 1.9 fold [143].

3.1.2.4. Surfactants. Surfactants have been widely used in formulationswith the intention of enhanced drug absorption by means of wettingand solubilization. However, their P-gp inhibition capability has re-cently gained widespread acceptance [144]. Mainly polysorbates,pluronic block co-polymers, castor oil derivatives, fatty acid ester sur-factants such as hydroxyl stearates e.g. Solutol HS 15, Cremophor EL,etc. and vitamin E derivatives have been evaluated for their P-gp ef-flux inhibition [145]. Fig. 5 reflects the inhibitory activity of varioussurfactants on the uptake of [3H] mitoxantrone via different mem-brane transporter proteins such as P-gp and BCRP. Among the testedsurfactants, Cremophor EL, Tween 20, Span 20, Pluronic P85 and Brij30 exhibited significant increase in the uptake of [3H]mitoxantronein BCRP-expressing cells whereas Cremophor EL, Cremophor RH40,Tween 20, Tween 80, Span 20, Pluronic P85, vitamin E TPGS, Brij 30,Myrj 52 and Gelucire 44/14 posed significant increase in the uptakeof [3H]mitoxantrone in P-gp-expressing cells [146]. These surfactantsact even at very low concentration, e.g. ~0.01% v/v Tween 80 signifi-cantly increased the accumulation of the daunorubicin in the resistantEhrlich Ascites Tumor cells [147]. Similarly, the basolateral to apicaltransport of the epirubicin on the Caco-2 cell lines was significantlyreduced in presence of Myrj [148]. The oral bioavailability of the rho-damine 123 increased by ~2.8 fold upon co-administration with Myrj[149]. P-gp inhibition is often found to be concentration dependentwith most of the surfactants such as Cremophor EL, Tween 80, etc.Mechanistically, these alter the membrane fluidity and bind with hy-drophobic domain of P-gp thereby changing its conformation leadingto reduced functionality [150]. On the other hand some surfactantssuch as Labrasol open the tight junctions of intestinal epithelium viainteraction with F-actin and ZO-1 [151]. A dose dependent increasein the intestinal permeability of mannitol was observed when co ad-ministered with Labrasol.

Pluronic block co-polymers are known to sensitize the P-gp ofoverexpressing MDR cancer cells and are reported to deplete theATP pool of the cells; adhere to the cell membranes and alter thelipid bilayer leading to significant decrease in the ATPase activity

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 8: Oral delivery of anticancer drugs: Challenges and opportunities

ECT

532

533

534

535

536

537

538

539

540

541

542

543

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

576

577

578

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

604

605

606

607

608

609

610

611

612

613

614

615

616

617

618

619

620

Fig. 5. Comparative inhibitory activity of various surfactants on the uptake of [3H]mitoxantrone in BCRP-(A) and P-gp-expressing MDCK-II cells (B); BCRP: breast cancerresistance protein, GFP: green fluorescent protein, P-gp: P-glycoprotein, MDCK:Mardin Darby canine kidney.Adapted with permission from [146].

8 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RR[152]. Microgel formulations containing hydrophobic (L-68) and hy-

drophilic (F-127) pluronic co-polymer significantly increased the oralbioavailability of megestrol acetate [153]. However, vitamin E TPGShad shown a dose dependent inhibition only on the substrate inducedATPase activity of the P-gp owing to its PEG content [154]. The oral bio-availability of the paclitaxel increased by about 6-fold as compared tothe control group upon co-administration with vitamin E TPGS. Addi-tionally, the absorption permeability increased by about 4.7-fold andsecretory permeability decreased by about 0.66-fold as measured onthe bidirectional-transport in excised rat ileum. These results wereattributed to micellar solubilization and enhanced permeability of pac-litaxel due to inhibition of P-gp efflux pump by vitamin E TPGS [155].

3.1.2.5. Cyclodextrins. Yet another class of functional excipients, cyclo-dextrins, has been evaluated for their P-gp interaction capability.Methylated cyclodextrins have been reported to interact with thelipid components of the biological membranes, especially cholesterol,modifying their fluidity and permeability [156]. Mechanistically,these are reported to reduce the activation energy required to incor-porate cholesterol in the hydrophobic cavity of the membranes[157–159]. Furthermore, the solubility advantage also contributes tooverall bioavailability enhancement by these cyclodextrins. Signifi-cant increase in the susceptibility of MCF-7 and MDA-MB-231 celllines to carboplatin and 5-fluorouracil was observed when pretreatedwith methylated cyclodextrins [160]. Intracellular accumulation ofdoxorubicin was found to about 2–4-fold higher in HL-60 S and

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

HL-60 R cell lines pretreated with methyl cyclodextrins as comparedto those treated with doxorubicin alone [161]. Similar results werealso obtained with docetaxel when tested against K562 S, MCF7 S andA2780 S cell lines [162]. Furthermore, significant increase in the invitro cytotoxicity against cancer cell lines was observed when paclitaxelwas complexed with cyclodextrins; probable reason identified was in-creased solubilization and permeation, owing to inhibition of effluxpumps [163]. Extrapolating these findings, the permeation studiesacross excised intestinal epithelium of the rats were performed andabout 12-fold increase in the apparent permeability of paclitaxel wasfound from cyclodextrin-polyanhydride nanoparticles as comparedto Taxol® [164]. Furthermore, significant increase in the oral bio-availability of the tacrolimus was also found when complexed withdimethyl-β-cyclodextrin (DM-β-CD). Permeability studies revealed de-creased basolateral to apical transport thereby contributing to the over-all bioavailability enhancement along with the solubility advantage[165]. Recently, nanosponges made up of β-cyclodextrins have beenimplemented to improve the oral bioavailability of paclitaxel andabout 3-fold increase in the area under the plasma concentrationcurve (AUC) was observed for paclitaxel loaded nanosponges as com-pared to Taxol® [166]. Similarly, about 3-fold increase in the apparentpermeability of exemestane across Caco-2 cell monolayers was ob-served upon complexation with hydroxypropyl β-cyclodextrin [167].

3.2. Nanocarrier based approaches

Nano-engineered drug delivery systems have shown their poten-tial to increase the oral delivery of various anticancer drugs. Substan-tial efforts have been made to improve the oral bioavailabilityvis-à-vis therapeutic efficacy and safety profile. It has now becomequite evident that a variety of nanocarriers have gained a substantialattention for enhancing the oral deliverability of anticancer drugs.The maintenance of an optimal drug concentration in plasma and inthe vicinity of tumors is the prime requirement of the effective cancertherapy. Carrying forward the discussion of utilizing functional excip-ients in improving the oral delivery of anticancer drugs, a more logicaland scientific approach could be formulating the nanoparticles ofthese functional excipients capable of altered absorption pathways.These altered absorption pathways further appreciate the incrementin oral bioavailability of difficult to deliver drugs.

The principal advantages of nanocarriers include their increasedsolubilization potential, superior encapsulation, altered absorptionpathways, prevention of metabolic degradation within gastrointesti-nal tract, chemical versatility of materials eligible for nanomedicines,flexibility in surface functionalization, drug and disease specific tailormade design capability, targeting potential and ability to incorporatewide variety of drug substances. Recently, the prevalence of drug–drug interactions in cancer patients treated with oral anticancerdrugs is reported which is alarming in the sense that conventionaldrug delivery system (both oral and intravenous) is dangerous to pa-tients. About 46% of patients receiving oral anticancer therapy devel-oped potential drug–drug interactions among which ~16% wereconsidered major [168]. These drug interactions could be fruitfullyavoided by utilization of carrier based approach where drug is encap-sulated within carrier matrices. In addition, it also prevents the cyto-toxic effects to the gastrointestinal tract which is very critical forpatients on chronic cancer therapy via oral route. A variety of bio-pharmaceutical parameters have been reviewed to manipulate theirin vivo fate upon oral administration. Briefly, particle size, shape andsurface properties of the nanoparticles play a crucial role in the up-take across the gastrointestinal membrane and were found to signifi-cantly affect the absorption profile. The nanocarriers with particlesize of 50–300 nm, positive zeta potential and hydrophobic surfacewere found to have preferential uptake from gastrointestinal tractas compared to their counterparts [169]. However, the retention ofthese properties in the gastrointestinal lumen is equally important

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 9: Oral delivery of anticancer drugs: Challenges and opportunities

T

621

622

623

624

625

626

627

628

629

630

631

632

633

634

635

636

637

638

639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

661

662

663

664

665

666

667

668

669

670

671

672

673

674

675

676

677

678

679

680

681

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

707

708

709

710

711

712

713

714

715

716

717

718

719

720

721

722

723

724

725

726

727

728

729

730

731

732

733

734

735

736

737

738

739

740

741

742

743

744

745

746

747

748

749

9K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

and efforts should be made in this direction to maximize the deliveryefficiency of the carrier system.

Numerous research groups, including ours, are actively involved inidentifying the different absorption mechanisms of these nanocarriersacross the intestinal epithelium and a significant success has beenachieved in this area. Nanocarriers by the virtue of their ability areable to bypass thedifferent hurdles that are responsible for poor oral ab-sorption of majority of anticancer drugs. Various identified absorptionmechanisms through which nanocarriers increase the oral bioavailabil-ity of drug molecules include increased absorption from enterocytes(due to increased solubilization and dissolution), mucoadhesion (inter-action between the positively charged nanocarrier with negativelycharged mucin) [170], tight junction modulation (capability of nano-carriers to interact with the tight junction proteins) [171], receptorme-diated endocytosis and transcytosis (clathrin- and calveolae-dependentand -independent endocytosis) [172], phagocytosis via specializedmicrofold cells (M cells) of the Peyer's patches and other mucosa asso-ciated lymphoid tissues (MALT) [173] and lymphatic absorption viachylomicron uptake mechanism from the enterocytes (mediated by li-pase for various lipid based drug delivery systems) [174]. The readersare redirected to the specific literature for further details on variousmechanisms. Hitherto, substantial efforts have been dedicated in thedevelopment of wide variety of nanoformulations with the aim to aug-ment the oral bioavailability vis-à-vis the efficacy of the anticancerdrugs. Various formulation strategies include nanocrystals, drug–poly-mer conjugates, polymeric nanocapsules, polymericmicelles, polymericnanoparticles, lipid based nanocarriers and surfacemodified liposomes.The subsequent sections convey the information on recent develop-ments in the formulation of a variety of nanocarriers which wereemployed for the oral bioavailability enhancement of anticancer drugs.

3.2.1. Drug nanocrystalsRecently, nanocrystal approach has gained a great deal of impor-

tance considering its capability to impart higher saturation solubility,enhanced dissolution and reproducibility for oral absorption of drugmolecules, encompass high dose drugs, thereby increasing the overallbioavailability (Fig. 6) [175]. The residence time of nanocrystals in theGIT could be increased by improving the adhesiveness of nanocrystalsto lumen by mucoadhesive polymers. Various approaches have beenreported to impart mucoadhesion to the nanocrystals which includesuspension layering, spray drying, etc. However, there is always a limi-tation in choice of excipients with dual functionality of mucoadhesionand nanocrystal stabilization. Hence, a novel approach of incorporatingthe nanocrystal in mucoadhesive gels was implemented [176]. In addi-tion, increased permeability is also reported to contribute to the overallbioavailability enhancement to some extent [177]. The other advan-tages with nanocrystal approach include high drug pay load, drug sta-bility, improved drug efficacy, high level of scalability and widespreadindustrial adaptability. Currently, two approaches i.e. bottom-up andtop-down methods have been reported for the formulation of drugnanocrystals and on that basis various technologies have been adaptedby the industry such as pearl milling (NANOCRYSTALS™, Elan), homog-enization inwater (NANOEDGE™, Baxter), homogenization in alternatedispersion media (NANOPURE™, Pharamasol), homogenization withmicrofluidizers (IDD-P™, SkyePharma), piston gap homogenizationin surfactant based aqueous phase (DISSOCUBES™, SkyePharma), pre-cipitation to yield amorphous material (NANOMORPH™, Soliqs) andcombination of approaches such as precipitation, spray drying andlyophilization (SMARTCRYSTAL™, Abbott).

Incorporation of the functional excipients, such as P-gp inhibitors,solubilizers, in the nanocrystals may further potentiate the efficacy ofthe final formulation [178].

Nanocrystals for the oral bioavailability enhancement of antican-cer drugs have been scarcely reported. A few research groups likeLiu et al. have reported the development of paclitaxel nanocrystalsby surface stabilization with Pluronic F127 and studied the effect of

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

stabilizer and stability of paclitaxel nanocrystals in the mechanisticmanner [179]. The same group further employed D-α-tocopheryl poly-ethylene glycol 1000 succinate (TPGS) as a surface stabilizer for thedevelopment which posed superior and significant efficacy in themulti drug resistant cancer cell lines as compared to marketed controlgroup and free drug [178]. On the similar line, novel nanocrystal formu-lation of paclitaxel and camptothecin has been developed usingthree-phase nanoparticle engineering technology (3PNET technology),which includes a 3 step process: amorphous precipitate; hydratedamorphous aggregate and finally stabilized nanocrystal utilizing F127as polymer stabilizer [180]. The developed nanocrystal formulationupon oral administration showed comparable antitumor efficacy asthat of intravenous administration of taxol at lower doses (20 mg/kg)and significantly higher efficacy at higher doses of 60 mg/kg. Further-more, the efficacy was 3-fold greater as compared to that of free drugsuspension, clearly indicative of advantage of nanocrystal in the drugabsorption. However, the authors still feel need to improve the oral bio-availability of nanocrystal to achieve comparable to that attained by itsintravenous counterpart. Henceforth folate receptor targeted nanocrys-tal design is under evaluation. However, principal problems associatedwith nanocrystal strategy includes higher toxicity potential to the gas-trointestinal tract, poor intellectual property perks and classical to BCSclass II drugs. Therefore, efforts should bemade in the direction of func-tional coating to nanocrystals, stabilization of nanocrystals with hydro-phobic amino acids, absorption enhancers, etc. Further efforts canalso be made to stabilize the nanocrystals in gastrointestinal tract andthus make them eligible for M cell uptake classical to polymericnanoparticles (Fig. 6). β-Casein stabilized paclitaxel nanocrystals wereformulated and no significant depreciation in the in vitro cytotoxicitywas found as compared to the free drug when tested against humangastric cancer N-87 cell lines in the presence of simulated gastrointesti-nal fluids. The authors proposed the protective role of β-casein in theformulation to avoid the toxicity to the oral and esophagus liningswhen administered orally without compromising the cytotoxicity[181]. However, the systemic bioavailability of drug from such systemsis questionable and hence a rationalized system, paclitaxel nanocrystalsloaded porous quaternized chitosan nanoparticleswas developed [182].These modified nanocrystals exhibited significantly higher intracellularaccumulation and in vitro cytotoxicity as compared to free drug whentested inCaco-2 cell lines and Lewis lung carcinoma (LLC) cell lines, re-spectively. Synchronizing with the in vitro results, in vivo studies re-vealed about 5-fold increase in the accumulation of drug at the tumorsite alongwith higher antitumor efficacy and safety profile as comparedto standard formulation. Recently, an investigational new chemical en-tity, 2-methoxyestradiol (2ME2), has been developed as Nanocrystal®dispersion (NCD) and is presently under phase II clinical trial in patientswith taxanes refractory, metastatic castrate-resistant prostate cancer(CRPC) [183].

3.2.2. Polymeric nanocarriers

3.2.2.1. Polymeric nanoparticles. Polymeric nanoparticles are nano-colloidal cargos, preferably in the size range of 10–1000 nm, made upof wide variety of polymers. They have been widely studied and evalu-ated for the oral delivery of chemotherapeutic agents. The principal ad-vantage of this system includes their robust structural characteristicsimparting very high stability in the gastrointestinal tract. Furthermore,the hydrophobicity and hydrophilicity within the polymeric systemcan be manipulated to accommodate wide variety of drug molecules[184]. The properties of biocompatibility and biodegradation furtherenhance their delivery potential. A large number of polymers includingthe co-polymers have been employed for the preparation of polymericnanoparticles (nanocapsules and matrix based nanoparticles). Theseinclude natural polymers such as gelatin, dextran, albumin, chitosanand alginate to name a few, among which chitosan and its derivativeshave been widely explored [185]. Recent trends include the utilization

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 10: Oral delivery of anticancer drugs: Challenges and opportunities

T

PRO

OF

750

751

752

753

754

755

756

757

758

759

760

761

762

763

764

765

766

767

768

769

770

771

772

773

774

775

776

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

802

803

804

805

806

807

808

809

810

811

812

813

814

815

816

817

818

819

820

821

822

823

824

825

826

827

1 mm

Size Reduction

10 µm

100 nm

BCS Class II drugs

•••

DissolutionSolubility Dose

BCS Class IV drugs

•• Solubility• Permeation •

Dissolution

Stability in GIT• Uptake by M cells in Peyer’s patches

Particle size

Surface

area

Size Reduction

Conventional drugTSA: 6 mm2

*TSA: Total surface area

Micronized drugTSA: 600 mm2

Nanonized drugTSA: 60000 mm2

Fig. 6. Mechanistic representation of absorption via nanocrystals.

10 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

of the synthetic biodegradable polymers such as polylactic acid (PLA),polyglycolic acid (PGA), copolymers of lactic and glycolic acid (PLGA),poly(ε-caprolactone) (PCL), poly-alkyl cyanoacrylate (PACA), polyeth-ylene imine (PEI), poly(L-lysine), poly(aspartic acid), etc. [186].

The polymeric nanoparticles tend to show very high degree ofsustained release of drug molecules, which could be of special signifi-cance for oral delivery in terms of ensuring that no drug is releasedfrom the formulation till it reaches systemic circulation therebybypassing various physiological barriers to oral delivery of difficult-to-deliver drugs [187]. Numerous reports on rapid absorption of polymericnanoparticles from gastrointestinal tract upon oral administration andsubsequent drug release from longer period of time (exceeding gas-trointestinal transit time) have been reported. The same could be at-tributed to the preferential uptake of polymeric nanoparticles byspecialized Peyer's patches (M cells) and the isolated follicles of thegut-associated lymphoid tissue present in the gastrointestinal tract(Fig. 7). Our group has exhaustively studied PLGA nanoparticles fororal delivery of various anticancer agents including tamoxifen anddoxorubicin. Significant improvement in the cellular uptake of doxo-rubicin loaded PLGA nanoparticles (Dox-NPs) was observed inmouse breast cancer, C127I cell lines as compared to free drug [188].The internalized Dox-NPs were found to preferentially localize in thevicinity of nucleus, site of action of doxorubicin (Fig. 8). Furthermore,the developed formulation posed both time- and concentration-dependent increases in the Caco-2 cell uptake as compared to freedrug solution. Considering increased permeation across the gastroin-testinal tract, the developed formulation showed ~49.06% reductionin the tumor burden upon oral administration of Dox-NPs in30 days; whereas in contrast 158% increase in tumor burden wasobserved in untreated group. Although, intravenous administrationof free doxorubicin could reduce the tumor burden to greater extent(~69.28%) as compared to developed formulation, significantly highercardiotoxicity was observed. The developed formulation in contrastposed significantly lower cardiotoxicity. Therefore, it could be con-cluded that efforts should be made in improving the therapeutic effi-cacy of a formulation without compromising the safety profile andthe same could be achieved using nanocarrier based approaches. Onthe similar line of action, tamoxifen loaded PLGA nanoparticles(Tmx-NPs) were also developed and evaluated for their in vitro and

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED

in vivo performances. Although, similar cellular cytotoxicity profile offree Tmx and Tmx-NPswas observed against C127I cell lines at all test-ed time points till 72 h, significantly higher cytotoxicity was observedin case of recovery experiments which could be attributed to rapid in-ternalization and retention of nanoparticles within cells as comparedto free drug.

About 3.84-fold and 11.19-fold increase in the oral bioavailability oftamoxifenwas observedupon incorporation in to PLGAnanoparticles ascompared to commercial analogue, tamoxifen citrate and free base, re-spectively. Furthermore, significantly higher antitumor efficacy of ta-moxifen and reduced hepatotoxicity were also observed from PLGAnanoparticles as compared to free base (Fig. 9) [189]. Similar resultswere also observed in case of CoQ10 loaded PLGA nanoparticles [190].

Very often the surfacemodification of the polymeric nanoparticles iscarried out to achieve desired properties meeting intended application.The hydrophobicity and surface charge of the prepared nanoparticlescan be manipulated to modulate the absorption kinetics from thegastrointestinal tract. This includes surface functionalization such asPEGylation, co-polymerization with functional polymers, polyelectro-lyte coatings and ligand anchoring [191]. An exhaustive study onengineered polymeric nanoparticles for cancer therapeutics has beenrecently reviewed by our group [192]. PEGylation is one of the mostcommon approaches implemented to circumvent classical problemsassociated with nanoparticles such as particle aggregation, stability inthe biological milieu and rapid clearance from body. Various aspectsof nanoparticle PEGylation and their pros and cons in the drug deliveryhave been reviewed recently [193,194]. PEG chains are reported toimpart stealth characteristics to the nanoparticles [195], improvebioadhesion in the gastrointestinal tract [196], provide surface hydro-philicity and facilitate passive targeting via EPR effect [197].

Ligand anchoring on the polymeric nanoparticles is also one of theapproaches to improve their oral deliverability. However, this ap-proach is less studied owing to existence of very few receptor medi-ated transport systems in the gastrointestinal tract. The principalligands that are exploited for the said purpose include folic acid[198], bioadhesins such as lectins, pectins [199]; peptidic ligandssuch as RGD (arginine–glycine–aspartate) [200]; and bile acids suchas deoxycholic acid [201]. Bioadhesins are the wide variety of pro-teins and glycoproteins capable of interacting preferentially with

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 11: Oral delivery of anticancer drugs: Challenges and opportunities

TD P

RO

OF

828

829

830

831

832

833

834

835

836

837

838

839

840

841

842

843

844

845

846

847

848

849

850

851

852

853

854

855

856

857

858

859

860

861

862

863

864

Receptor mediated endocytosis

Fluid Phase Macropinocytosis

Lymphatic absorption

Macrophage

Systemic circulation

Polymeric nanoparticles

M-cell

Polymeric micelles

Inhibition of P-gpUptake by

M cells

Enterocytes

Polymer drug conjugates

Lysosomes

Inhibition of P-gp

Chitosan nanoparticles

Interaction with tight junction

proteins

P-gp↑ Bioadhesiveness

Fig. 7. Potential absorption mechanisms implemented by polymeric nanocarriers for increasing oral bioavailability of drug substances.

11K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

CO

RREC

carbohydrate residues. Among the wide variety of known bioadhesins,lectins are themostwidely studied for targeted drug delivery to specificlocations along the gastrointestinal tract for variety of purposes such astumor targeting, mucosal immunization, etc. [202]. The lectin anchoredpolymeric nanocarriers are specifically taken up by antigen samplingcells of gastrointestinal tract, M-cells of the Peyer's patches; whichfurther process it to transepithelial antigen transport system. Once in-ternalized, these carriers pave their way to systemic circulation via lym-phatic system; thereby bypassing various absorption barriers [203].In addition, lysosomal targeting mediated by lectin has also been pro-posed in Caco-2 cell lines [204]. On the other hand, peptidic ligandssuch as RGDpeptide are found to interact with the β1 integrin receptorslocalized at apical pole on the M cells of the Peyer's patches alongthe gastrointestinal tract [205]. Similarly, various bile acids have alsobeen evaluated for improving the oral deliverability of polymericnanoparticles. These bile acids such as deoxycholic acid are activelytransported via bile acid transporters from the gastrointestinal tract toliver. Therefore, bile acid as a targeting ligand can be used specificallyfor liver targeting via oral route of administration [201]. Table 3 reveals

UN

Fig. 8. Critical quality attributes of doxorubiciAdapted with permission from [188].

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

Evarious types of polymeric nanoparticles that have been implementedto improve the oral delivery of anticancer drugs.

The principle disadvantages associated with polymeric nanopar-ticles especially PLGA based drug delivery system are excessive cost in-curred. However, as far as oral anticancer therapy is concerned, everyeffort should be made to have cost effective formulations. Hence, rela-tively inexpensive polymeric materials should be sought such as chito-san. But the concern associated with crosslinking of chitosan forpreparation of nanoparticles should be carefully addressed.

3.2.2.2. Polymeric micelles. Polymeric micelles are the systems con-taining hydrophobic cores surrounded by hydrophilic corona that isexposed to aqueous environment. The hydrophobic cores act as a res-ervoir for lipophilic drug molecules and corona acts as the steric stabi-lizer of the overall system thereby assuring the integrity of the systemin aqueous environment holding the adequate amount of guest drugmolecules [226]. Polymeric micelles have recently gained wide accep-tance as the carrier systems considering their various advantagessuch as superior stability compared to surfactant micelles, enhanced

n loaded PLGA nanoparticles (Dox-NPs).

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 12: Oral delivery of anticancer drugs: Challenges and opportunities

T

F

865

866

867

868

869

870

871

872

873

874

875

876

877

878

879

880

881

882

883

884

885

886

887

888

889

890

891

892

893

894

895

896

Fig. 9. (A) Plasma concentration time profiles of tamoxifen (Tmx) after oral administration to SD rats at 10 mg/kg dose formulated in the PLGA NPs, compared with the oral ad-ministration of Tmx free base and Tmx salt; (B) tumor progression after repetitive oral administration of Tmx citrate and Tmx-NPs (3 mg/kg). Tumor volume was taken as 100%at the start of drug treatment and tumor progression monitored till the end of the study; each data point is represented as mean ± SEM (n = 6).Adapted with permission from [189].

t3:1

t3:2

t3:3

t3:4

t3:5

t3:6

t3:7

t3:8

t3:9

t3:10

t3:11

t3:12

t3:13

t3:14

t3:15

t3:16

t3:17

t3:18

t3:19

t3:20

t3:21

t3:22

t3:23

t3:24

t3:25

t3:26

t3:27

t3:28

12 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

solubilizing power, longer circulating time due to outer hydrophilicshell, small size and targeting capability [227]. An exhaustive reviewon the various mechanisms of drug absorption via polymeric micellesfrom gastrointestinal tract is already available in literature and hencebeen kept out of the scope of this report [226]. Briefly, these include al-teration in the membrane permeability, absorption of micelles via fluidphase pinocytosis, receptor mediated endocytosis (upon attachment ofspecific ligands to themicellar structures), inhibition of the efflux trans-porter proteins (majority of the polymers formingmicelles are reportedto interact with efflux transporter proteins) and mucoadhesion alongthe gastrointestinal tract (Fig. 7). These systems generally have A–Bdiblock structure; A, hydrophilic polymers constituting coronawhereasB, hydrophobic polymers, forms cores. However, tri block copolymerA–B–A systems have also been studied to some extent and under consid-eration for its use as potential drug delivery system. The cores of the poly-meric micelles are generally made of biodegradable polymer such as

UNCO

RRECTable 3

List of polymers implemented to improve the oral deliverability of various anticancer agen

Polymeric system Functionalization Active Outcome

Chitosan TPP cross linking (−)-Epigallocatechin gallate 1.5-FoldChitosan Caseinophosphopeptides (−)-Epigallocatechin gallate SuperiorPAA-cysteine GSH Paclitaxel About 7-

bioavailaPCCL-PEG-PCCL Chitosan 10-Hydroxycamptothecin SignificanPolyanhydrides Cyclodextrins Paclitaxel Relative BPolyanhydrides PEG 2000 Paclitaxel Relative bPolymethylmethacrylate

Thiolated chitosan Docetaxel Significanfor 72 h

PLA Vitamin E TPGS Docetaxel BioavailaPLA TPGS Paclitaxel 1.8-Fold

nanopartPLGA DMAB-TPGS Docetaxel About 16

lines as cPLGA Montmorillonite Paclitaxel Significan

and 11–5PLGA Dextran sulfate Vincristine sulfate 12.4-FoldPLGA Vitamin E TPGS – 4–6-FoldPLGA Pectin Thymopentin IncreasedPLGA Deoxycholic acid Rhodamine BioavailaPLGA WGA Paclitaxel About 2-

comparePLGA WGA Paclitaxel SignificanNMA622 – Rapamycin SignificanPLGA – 5-Fluorouracil BioavailaPLGA – Curcumin BioavailaPLGA – Doxorubicin BioavailaPLGA – Paclitaxel 7-Fold hi

2780 ADPLGA – Tamoxifen Bioavaila

PLGA: Polylactide-co-glycolic acid; PCL: Polycaprolactones; PCCL: poly(caprolactone-co-lacacid; NMA622: N-isopropylacrylamide, methylmethacrylate, and acrylic acid in the molar rat

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

Opoly(β-benzyl-L-aspartate), poly(DL-lactic acid), poly(ε-caprolcatones),etc. whereas the shell is made up of biocompatible polymers such aspolyethylene oxide. Themicellesmay either be functionalizedwith cer-tain polymers such as poly(N-isopropylacrylamide) [228] or poly(alkyl-acrylic acid) [229] to impart temperature or pH sensitivity, or canfurther be conjugated with ligands to impart targeting characteristics[230,231].

Recently, functional micelles for intracellular targeting via oralroute of administration have been reported [232]. These sophisticatedsystems utilize dequalinium, as a targeting ligand for preferentialco-localization in the mitochondria, guided by transmembrane elec-tric potential within the cell. The cell culture experiments revealedstrong inhibitory action of these functional nanomicelles againstMCF-7 and MCF-7/Adr cell lines with selective accumulation in themitochondria and endoplasmic reticulum within the cells. The per-meability studies revealed about 36.4-fold higher transport of these

ts.

Ref

increase in oral bioavailability [206]biocompatibility, increased Caco-2 permeability and oral bioavailability [207]fold increase in apparent permeability and about 5-fold increase in oralbility as compared to free paclitaxel

[208]

t improvement in the absorptive transport across Caco-2 cell monolayers [209]ioavailability up to 80% [210]ioavailability up to 70% [196]t increase in the in vitro cytotoxicity against MCF-7 cell lines maintainedas compared to free drug

[211]

bility increased by 22-fold [212]increase in the cellular uptake by HT29 cell lines as compared to plain PLGAicles and 40% reduction in IC50 values as compared to taxol

[213]

-fold decrease in the IC50 value for in vitro cytotoxicity against MCF-7 cellompared to marketed formulation at 72 h

[214]

t improvement in the entrapment efficiency by 57–177% for Caco-2 cells5% for HT-29 cells was observed

[215]

higher uptake by MCF-7/Adr cells as compared to free drug [216]increase in the cellular uptake by Caco-2 cells [217]in vivo efficacy [218]

bility increased by 1.8-fold [201]fold increase in the cellular uptake by Caco-2 and HT-29 cell lines asd to plain nanoparticles

[219]

tly higher cytotoxicity against A549 and H1299 cell lines [220]t reduction in tumor inhibition [221]bility increased by 2.27-fold [222]bility increased by 22-fold [223]bility increased by 3.63-fold [224]gher toxicity of formulation in A2780 cell lines and significant activity inresistant cell lines

[225]

bility increased by 11-fold [189]

tide); PLA: Polylactic acid; PEG: Polyethylene glycol; RGD: Arginine–Glycine–Asparticios of 60:20:20; WGA:Wheat germ agglutinin; PAA: polyacrylic acid; GSH: glutathione.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 13: Oral delivery of anticancer drugs: Challenges and opportunities

T

897

898

899

900

901

902

903

904

905

906

907

908

909

910

911

912

913

914

915

916

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

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

971

972

973

974

975

976

977

978

979

980

981

982

983

t4:1

t4:2

t4:3

t4:4

t4:5

t4:6

t4:7

t4:8

t4:9

t4:10

t4:11

t4:12

t4:13

t4:14

t4:15

t4:16

t4:17

t4:18

t4:19

t4:20

13K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

RREC

functional micelles as compared to free drug when evaluated againstCaco-2 cell monolayers. Furthermore, everted gut sac method revealedabout 52.9-fold higher permeability of these functionalmicelles as com-pared to free drug.Moreover, these functionalmicelles revealed compa-rable in vivo antitumor efficacy against xenografted resistant MCF-7/Adr cancer bearing mice by oral and intravenous routes of administra-tion; both being higher than free drug by ~5-fold and 3-fold, respective-ly. This remarkable antitumor efficacy could also be attributed to thepharmacological activity of dequalinium which shows significantlyhigher antitumor efficacy as compared to most of the established anti-cancer agents [233]. However, preferential localization of dequaliniumat the tumor site via oral route is a state-of-art which can be achievedby nanocarriers, per se. Table 4 reflects the list of anticancer drugsbenefitted with the polymeric micelle approach for improving theiroral deliverability.

3.2.2.3. Polymer–drug conjugates. Polymer–drug conjugates are thenovel drug delivery systems comprised of covalently linked drugmole-cules to the polymer backbone. Along with the drug molecules, addi-tional functional excipients such as diagnostic agent, targeting ligand,PEG chains to improve hydrophilicity can also be accommodated inthe polymer backbone thereby resulting in to formation of nanocargosthat are highly specific to disease condition. The resulting conjugate isthen eligible for active transport from gastrointestinal tract to facilitateimproved absorption (Fig. 7). In addition, functional excipients havingcapability to inhibit drug efflux can also be employed. Further, targetingpotential offered by polymer–drug conjugate is especially important incase of cancer therapy owing to highly toxic effects of anticancer agentsto the normal tissues of the body. This also facilitates the stabilizationand subsequent delivery of various enzymes and immunocomponents[246]. Furthermore, upon exploitation of the structure–activity relation-ship of the drug molecules, various benefits such as increased solubili-zation, enhanced plasma half-life, bioavailability enhancement due toincrease in the hydrodynamic volume and reduced excretion by kid-neys, protection towards degrading enzymes, prevention or reductionof aggregation or immune responses, reduction in toxic side effectsand targeting capability can be achieved via polymer–drug conjugates[246,247].

The most critical aspect in case of polymer–drug conjugate ap-proach is identification of functional group for covalent modification.The said functional group should preferably be not involved in phar-macological activity. In absence of the same, it should be ensured thatthe drug in concern is released in free form upon exposure to physio-logical conditions or at target site. Secondly, type of linkage could also

UNCO

Table 4List of anticancer drugs benefitted by polymeric micelles approach for improving their oral

Drug System Outcome

Paclitaxel Dequalinium-Vit TPGS1000-PEG2000-DSPE Oral antitumcomparable

Paclitaxel PEG-DSPE-TPGS Solubility inPaclitaxel PEG-b-P(VBODENA) Solubility inPaclitaxel PEG-b-VBODENA-Acrylic acid Complete dTamoxifen PHEA-PEG-C(16) SignificantlyTamoxifen PAHy-PEG(2000)-C(16) 3000-fold inMeso-tetraphenyl porphine Pluronic F127/PEG–DSPE 90% drug loα-Mangostin PVP About 13,00Doxorubicin Stearic acid-g-chitosan Increased oDoxorubicin Pluronic-L92-PAA-EGDMA- ~2-Fold incDoxorubicin PLGA-dextran SignificantlyPaclitaxel N-deoxycholic acid-N,O-hydroxyethyl chitosan BioavailabilPaclitaxel N-octyl-O-sulfate chitosan BioavailabilQuercetin PEG-PE 110-Fold in

MOG: Monoethylene glycol; P(VBODENA): poly(2-(4-vinylbenzyloxy)-N,N-diethylnicotinaPAHy-PEG(2000)-C(16): PEG 2000 and palmitic acid grafted on polyaspartylhydrazide; PHEacid); EGDMA: ethylene glycol dimethacrylate; PE: phosphatidylethanolamine.

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

be carefully employed for preparation of polymer–drug conjugate, toachieve target site specificity. The principal types of linkages includeamide linkages, ester linkages, sulfhydryl linkages, hydrazone link-ages, enzymatically degradable linkages, etc. [248]. It should benoted that ester and hydrazone linkages are acid labile and shouldbe less favored while designing oral drug delivery system. Thirdly,the release of drug from conjugate system in timely manner and ther-apeutically active state of released compound is one of the critical as-pects and should be ensured. This obviously necessitates highlysensitive analytical techniques while performing various in vitro andin vivo studies, which is a principal concern in case of polymer–drugconjugates. Further, owing to covalent attachment of the drug mole-cule with the polymer backbone, obvious concern for differentialpharmacokinetics in physiological conditions raises and should becarefully addressed.

The classical approach for formulation of polymer–drug conjugatescan be broadly categorized as PEGylation, involving covalent bindingof PEG chains to drug molecules. PEGylated paclitaxel showed about4-fold increase in the oral bioavailability as compared to free drug,which could be attributed to increased solubility, permeability andreduced presystemic metabolism [124]. Recently, nanoconjugates ofcamptothecin with polylactide have been successfully synthesized[249] and were found to preferentially circulate in body via lymphaticsystem [250]. A variety of carrier linked prodrugs for anticancer agentswith antibodies, proteins and polymers such as glutamic acid, PEG andpolylactic acid have been developed and are presently under clinicalstudies [251] but the aspects related to oral delivery are yet at infantstage and need serious efforts tomaterialize the concept. Recently, oral-ly administrable doxorubicin conjugated monomethoxy-polyethyleneglycol-polylactic acid was developed. The prepared conjugate wasabsorbed from intestine via the transepithelial pathway, accumulatedpreferentially in the liver and thus was exploited in the managementof liver metastasis in mice [252]. On the similar line of action, an oralcolon-specific drug delivery was developed for 9-aminocamptothecin(9-AC) by copolymerizationwithN-(2-Hydroxypropyl)methacrylamide(HPMA) and was found to have potential in management of colon can-cer [253]. Furthermore, lectin functionalization to these HPMA drugconjugates has been shown to improve the oral deliverability of difficultto deliver drugs such as insulin [254]. Henceforth attempts could bemade to apply the same principle for oral delivery of anticancer agents;however the careful selection of spacers for conjugation is one of thecritical steps.

Although, the conjugated systems are difficult to be delivered by oralroute considering their limited absorption, some functional excipients

bioavailability.

Ref

or efficacy in the xenografted resistant MCF-7/Adr cancers in miceto that of intravenous administration

[232]

creased to 5 mg/ml [234]creased to 39 mg/ml [235]rug release in 12 h [236]higher anticancer activity against MCF-7 cell lines as compared to free drug [237]crease in the solubility of Tamoxifenading efficiency could be achieved [238]0-fold increase in solubility of α-mangostin [239]ral bioavailability [240]rease in intracellular accumulation by Caco-2 cell monolayers [241]higher in vitro cytotoxicity against doxorubicin resistant HuCC-T1 cell lines [242]

ity increased by 3-fold [243]ity increased by 6-fold [244]crease in solubility and significant increase in the antitumor efficacy [245]

mide); SA: Succinic anhydride; POE: polyoxyethylene; HPC: Hydroxypropylcellulose;A-PEG-C(16): PEG and palmitic acid grafted on polyhexadecylamine; PAA: poly(acrylic

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 14: Oral delivery of anticancer drugs: Challenges and opportunities

T

984

985

986

987

988

989

990

991

992

993

994

995

996

997

998

999

1000

1001

1002

1003

1004

1005

1006

1007

1008

1009

1010

1011

1012

1013

1014

1015

1016

1017

1018

1019

1020

1021

1022

1023

1024

1025

1026

1027

1028

1029

1030

1031

1032

1033

1034

1035

1036

1037

1038

1039

1040

1041

1042

1043

1044

1045

1046

1047

1048

1049

1050

1051

1052

1053

14 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

along with formulation intervention have to be implemented. The clas-sical approaches include nanocarrier based approach alongwith surfacemodification by functional excipients. Recently, chitosan, owing to itsbiodegradable, biocompatible, mucoadhesive and non toxic nature,has been studied as a novel platform for oral delivery of drugs [255].Unlike high molecular weight chitosan, the low molecular weightchitosan (≤1 lac Dalton) possesses higher solubility, relative lower tox-icity profile and narrower molecular weight distribution [256]. Addi-tionally, these are also reported to reversibly open the tight junctionsbetween the epithelial cells of the Caco-2 cell lines more stronglyas compared to high molecular weight chitosans (≥20 lacs Dalton)[257]. Chitosans are proposed to preferentially interact with theF-actin present in the intestinal lamina in a charge dependent fashionleading to its depolarization and disbandment of ZO-1 protein, an inte-gral part of tight junction. Additionally, this effect leads to improvementin only apical transport of drugs confirming themechanismas increasedpermeability by paracellular pathway [258]. Furthermore, the effect ofdegree of acetylation and molecular weight was also found to signifi-cantly affect the absorption across the Caco-2 cell monolayers [259].Chitosan conjugated paclitaxel has been studied and offered several ad-vantages such as improved water solubility due to increased hydrophi-licity, prolonged retention of the conjugate in the gastrointestinalsystem due to mucoadhesive properties of chitosan resulting in overallincreased uptake, opening of tight junctions and enabling the para-cellular route for absorption and ability to bypass the CYP-450 depen-dent metabolism as the conjugated paclitaxel would no longer be thesubstrate of these enzymes. The absolute oral bioavailability of paclitax-el in rats was found to be ~42% upon conjugation with chitosan and asignificant increase in the in vivo antitumor efficacy and comparabletoxicity profile as compared to free drugwere observed. The hypothesiswas further confirmed by evaluating the effect of cyclosporin A on bio-availability of paclitaxel in the conjugate form. Significant increase incase of free drug and no effect on bioavailability were observed incase of conjugated paclitaxel, suggesting P-gp independent absorptionmechanism for chitosan conjugated systems [260].

UNCO

RREC

Fig. 10. Key advantages associated with

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

3.2.3. Lipid based nanocarriersSince many decades, lipids have been widely used for improving

the oral bioavailability of various difficult-to-deliver drugs. The lipidicexcipients principally comprises of monoglycerides, diglycerides, tri-glycerides, oils constituting various combinations of glycerides, phos-pholipids, sphingolipids and even high fat meal [261]. This could beattributed to some unique properties of lipids such as high solubiliza-tion potential, biocompatibility, manufacturing scalability, industrialadaptability, and distinct route of absorption thereby eliminating var-ious physiological barriers such as pre-systemic metabolism, gastro-intestinal degradation, P-gp efflux and permeability related issues,etc. [262]. The usual problem associated with most of the drugs in-cludes either low aqueous solubility or poor intestinal permeability,both of which can adequately be taken care by lipid based drug deliv-ery systems.

The higher lipophilicity of the drug molecules can be fruitfullyexploited tomake them soluble in lipid based systemswhich can subse-quently be delivered via suitable route of administration, preferablyoral. Two critical quality attributes for development of lipid baseddrug delivery systems have been identified which include drug solubil-ity within lipidic system and subsequent physiological processing ofdrug loaded lipidic system to achieve adequate drug absorption(Fig. 10). The intraluminal processing of the lipidic excipients hasbeen exhaustively reviewed elsewhere [263]. Briefly, gastric and linguallipases digest triglycerides in diglycerides and fatty acids in stomachand are responsible for generation of crude emulsion of lipids which isstabilized bydietary proteins, polysaccharides, etc. However, the dietaryphospholipids and cholesterol esters are not attacked by these lipases.Subsequent to gastric emptying, the partially digested and crude emul-sified lipidic system gets exposed to bile salts, cholesterol and phospho-lipids preferentially phosphatidylcholine secreted by gall bladder andpancreatic lipase/co-lipase secreted by pancreas. The secreted compo-nents then get adsorbed on the surface of the crude emulsion droplets.The adsorption leads to further digestion and formation of stable smallsized emulsion droplets. These small sized lipid emulsion droplets are

lipid based drug delivery systems.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 15: Oral delivery of anticancer drugs: Challenges and opportunities

T

1054

1055

1056

1057

1058

1059

1060

1061

1062

1063

1064

1065

1066

1067

1068

1069

1070

1071

1072

1073

1074

1075

1076

1077

1078

1079

1080

1081

1082

1083

1084

1085

1086

1087

1088

1089

1090

1091

1092

1093

1094

1095

1096

1097

1098

1099

1100

1101

1102

1103

1104

1105

1106

1107

1108

1109

1110

1111

1112

1113

1114

1115

1116

1117

1118

1119

1120

1121

15K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

further acted by lipid digestion products such as 2-monoglycerides andfatty acids to form mixed micelles of lipidic system, thereby makingprocess of lipolysis self-promoting [264]. The formed micelles willthen be absorbed by enterocytes, where it gets converted to the chylo-microns upon re-esterification via monoacyl glycerol or phosphatidicacid pathway and subsequent stabilization by phospholipids. However,the penetration of unstirred water layer and mucin in gastrointestinaltract is rate limiting factor. The formed chylomicrons are then subjectedto lymphatic transport system via mesenteric lymph and ultimatelyenter the systemic circulation by lymphatic drainage at thoracic duct[265]. The other transcellular routes by which lipid based drug deliverysystems get transported across enterocytes include macropinocytosis,clathrin-mediated, caveolae-mediated, and clathrin- and caveolae-independent endocytosis (principally lipid rafts comprising ofsphingolipids- and cholesterol-rich microdomains) [172]. Fig. 11 de-picts various absorption mechanisms by which lipid nanocarriersimprove the oral bioavailability of drug substances. Various types oflipid based nanocarriers implemented for improving the oral deliveryof drugs include microemulsions, nanoemulsions, lipid nanocapsules,self-emulsifying systems, lipid nanoparticles, hybrid lipid nanoparticles,liposomes and surface engineered liposomes to name a few.

3.2.3.1. Emulsions. Emulsion based approach has been one of the mostancient methods for delivery of lipophilic drugs. Presently two ver-sions of emulsions viz. microemulsions and nanoemulsions havebeen employed for improving the oral deliverability of BCS class IIor IV drugs. The unique characteristics include thermodynamic(microemulsions) or kinetic (nanoemulsions) stability, supersolvency,small droplet size, high industrial scalability (as low energy require-ments formanufacturing) and use of lipidic excipients as absorption en-hancers [266]. Traditionally, these systems comprises of appropriateblend of oil, surfactant and co-surfactant dispersed in aqueous phasetailor made as per the physicochemical properties of drug substances.The most common oils used for preparation of nanoemulsions includeomega 3- and 6- containing polyunsaturated fatty acids (PUFA) such

UNCO

RREC

Lymphatic absor

Macrophage

Systemic c

Solid lipid Nanoparticles (SLNs)

M-cell

Uptake by M cells

Enterocy

LayersomeLiposome

Nanostructured lipid carriers (NLCs)

P-gpParacellular

absorption Fluid Phase Macropinocytosis

Pab

Fig. 11. Absorption mechanisms implemented by lipidic nanocarr

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

as pine seed oil, fish oil, flax seed oil, safflower oil, hemp and wheatgerm oil, etc. The principal reasons for their widespread usages in prep-aration of nanoemulsions could be attributed to their preferential ab-sorption across the gastrointestinal barrier due to paucity of theseagents in physiological conditions [267]. Furthermore, reports suggesttheir probable interaction with cell membrane via lipid exchangeor endocytosis [268]. In addition, PUFA also contributes to reductionin the protein production and efflux transporters MDR1/P-gp. Re-cently this mechanismwas proposed to promote the in vitro cytotox-icity of paclitaxel against Caco-2 cell lines [269]. These interactionscan further be strengthened by incorporating surface charge to thenanoemulsions. The negative charge imparting agents include lecithin,cholesterol and PEGylated phospholipids while positive charge can beimparted by cationic lipids such as stearylamine and 1,2-dioleoyl-3-trimethylammonium-propane (DOTAP), etc. Negatively charged nano-emulsions were found to have higher reticulo-endothelial system(RES) uptake while positively charged preferentially accumulated inlungs followed by redistribution in the liver and spleen [270].

Recently, nanoemulsions have also been reported to improve theoral deliverability of various protein based vaccines such as MAGE1-HSP70/SEA complex [271]. The antitumor immune responses fromnanoemulsion loaded vaccine via oral and subcutaneous routeswere studied and comparable efficacy in terms of delay in tumorgrowth and tumor occurrence in mice challenged with B16-MAGE-1tumor cells was observed. Sincere efforts in this direction are present-ly under pipeline and soon some deeper insights on various aspects ofsaid area are anticipated. Table 5 reveals various anticancer drugsbenefited from emulsion based approach.

3.2.3.2. Self-emulsifying drug delivery systems (SEDDS). SEDDS are themost advanced approach of emulsion based drug delivery systemsand relies on the physiological fluids for the in-situ formation ofmicro/nanoemulsion. This formulation strategy comprises of drugdissolved in oils and stabilized by surfactants and co-surfactants,which upon exposure to the aqueous environment under gentle

Fluid Phase Macropinocytosis

ption

irculation

Self emulsifying formulations

tes

ss

Lipid nanocapsules

Emulsified lipids

Mixed micelles

PhospholipidsCholesterol

Chylomicrons

Inhibition of P-gp

aracellularsorption

iers for improving the oral bioavailability of drug substances.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 16: Oral delivery of anticancer drugs: Challenges and opportunities

T

OO

F

1122

1123

1124

1125

1126

1127

1128

1129

1130

1131

1132

1133

1134

1135

1136

1137

1138

1139

1140

1141

1142

1143

1144

1145

1146

1147

1148

1149

1150

1151

1152

1153

1154

1155

1156

1157

1158

1159

1160

1161

1162

1163

1164

1165

1166

1167

1168

1169

1170

1171

1172

1173

1174

1175

1176

1177

1178

1179

1180

1181

1182

1183

1184

1185

1186

1187

1188

1189

1190

1191

1192

1193

1194

1195

1196

1197

1198

1199

1200

1201

1202

1203

1204

1205

1206

1207

Table 5t5:1

t5:2 List of anticancer agents benefited by emulsion based carrier systems for improving their oral bioavailability.

t5:3 Drug Formulation Outcome Ref

t5:4 Benzylisothiocyanate

Flax seeds, egg phosphatidyl choline and glycerol based nanoemulsion About 3-fold increase in the apparent permeability across Caco-2 cellmonolayers and significantly higher in vitro cytotoxicity againstA549 and SKOV-3 cell lines as compared to free drug

[272]

t5:5 Curcumin Lecithin, soyabean oil and Tween 80 based microemulsion incombination with ultrasound energy

In vitro cytotoxicity established against two oral squamous cellcarcinoma cell lines, OSCC-4 and OSCC-25.

[273]

t5:6 Curcumin Curcumin oraganogel and Tween 20 based nanoemulsion 9-fold increase in the oral bioavailability in mice as compared tounformulated curcumin

[274]

t5:7 Docetaxel Capryol 90, Cremophor EL and Transcutol based microemulsion About 25-fold increase in the permeability across Caco-2 cell mono-layers and about 5.19-fold increase in the oral bioavailability wereobserved as compared to Taxotere®

[150]

t5:8 Melphalan Capmul MCM, Tween 80 and Transcutol P based nanoemulsion About 4.83-fold increase in oral bioavailability and 2-fold higherdistribution in ovaries as compared to free drug

[275]

t5:9 Methotrexate Soyabean oil, Cremophor EL, Span 80 and isopropyl alcohol basedmicroemulsion

Significantly higher in vitro cytotoxicity against MCF-7 cell lines ascompared to free drug

[276]

t5:10 Paclitaxel Pine nut oil and lecithin based nanoemulsion Significantly higher oral bioavailability as compared to aqueoussuspension with preferential distribution in liver, kidneys, and lungs

[270]

t5:11 Paclitaxel Capmul, polysorbate 80 and myvacet oil based microemulsion 11-Fold in the permeability across rat intestine and 3-fold increase inthe oral bioavailability as compared to taxol

[277]

t5:12 Paclitaxel Labrasol, vitamin E-TPGS and Labrafil M1944CS based nanoemulsion 70% absolute bioavailability upon oral administration; [278]t5:13 Paclitaxel Drug loaded nanoemulsion co-administered with curcumin 5.2-Fold increase in oral bioavailability and 3.2-fold increase in

tumor uptake was observed as compared to free drug[279]

16 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

agitation leads to spontaneous formation of emulsion. Although theexact mechanism of self emulsification is yet to be understood andis unclear however it has been postulated that it happens when en-tropy change for dispersion exceeds the energy required to increasethe surface area of dispersion [280].

The self-emulsification process is also reported to be affected by theconstituent oils, surfactants, co-surfactants, co-solvents and physico-chemical properties of drug to be incorporated. The details of variousfactors affecting the SEDDS are already available in literature [281]. Innutshell, oils play a very critical role in the solubilization of drug andare responsible for facilitating the drug transport across the gastrointes-tinal barriers via specialized transport mechanisms such as intestinallymphatic system as discussed previously. Different types of oils thatare known to form SEDDS include long- and medium-chain triglycer-ides with different degrees of saturation. The selection of oils solelydepends on its capability to solubilize the drug in concern. The currenttrends in design of SEDDS include utilization of modified or hydrolyzedvegetable oils and novel semi synthetic medium chain derivatives;later having the upper hand in terms of amphiphilic naturewith surfac-tant properties. On the other hand, surfactants, co-surfactants andco-solvents play a major role in the formation and stabilization of thein-situ emulsion. Generally, nonionic surfactantswith higher hydrophil-ic lipophilic balance (HLB) values are preferred; however, based on ourexperience, it should be noted that precipitation of drug upon dilutionneeds careful monitoring in this case. Furthermore, the toxicity associ-ated with higher usage of surfactants is a major limiting factor.

A newer generation of SEDDS based on supersaturation principlehas been designed to exclude the side effects of surfactants and enhancethe rate of absorption of drugs from the gastrointestinal tract [282].These formulations include low levels of surfactants and polymeric pre-cipitation inhibitors to yield and stabilize a temporary supersaturatedstate of drug. HPMC and other cellulosic polymers are well recognizedfor their capability to inhibit the crystallization of drugs and therebyleading to formation of supersaturated state of drug for prolong dura-tion. Recently, solid supersaturable SEDDS of docetaxel via spray dryingtechnique have been prepared and it was found to have about 8.77-foldand 1.45-fold higher oral bioavailability as compared to free drug andconventional SEDDS, respectively; clearly indicating the superior bene-fits at reduced surfactant level by this novel approach [283]. Further-more, ligand anchored SEDDS have also been developed to exploit thetargeting potential of nanocarriers. Recently, folate modified SEDDSfor delivery of curcumin have been designed and developed for colontargeting [284]. The developed formulation was found to significantly

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

Reffective against HELA and HT29 cell lines as compared to the plainSEDDS and free curcumin. Interestingly, the in vitro cytotoxicity was re-duced in the presence of external folate supplementation clearly reveal-ing the role of folate modification on the activity of formulation.

The principal advantage of the SEDDS lies in their superior stabil-ity profile that surpasses the stability related issues associated withthe conventional emulsions. However, the inherent stability of theoils should be critically assessed to predict the overall shelf life ofthe formulation. The stability of these formulations can further be en-hanced by adsorbing the liquid or semisolid SEDDS on to suitableinert adsorbents such as silicates, dextran, magnesium hydroxide,crosspovidone, carbon nanotubes, fullerenes, charcoal, etc. via variousprocess such as spray drying, lyophilization, melt granulation andmelt extrusion to name a few [285]. However, the process of adsorp-tion may also be associated with problems such as alterations in re-lease profile, process loss and increased manufacturing steps andexcess cost. Therefore, newer advancement includes utilization of ex-cipients that serve dual purpose of solidifying and emulsifying agentssuch as poloxamer 188 [286].

Our group is also actively involved in the formulation develop-ment of SEDDS for various difficult-to-deliver drugs. Recently, tamox-ifen loaded SEDDS have been developed and an Indian patentapplication has been filed [287]. A PCT application has also beenfiled and recently got favorable ISR/WO [288]. The developed formula-tion showed about 3.8-fold and 9-fold enhancement in oral bioavailabil-ity as compared to clinically used tamoxifen citrate and tamoxifen freebase, respectively. Furthermore, significantly higher antitumor efficacyin 7,12-dimethylbenz[α]anthracene (DMBA) induced breast cancermodel in rats was observed as compared to tamoxifen citrate. The tox-icity studies revealed no observable hepatotoxicity markers when test-ed in mice. The formulation was further found to be stable in simulatedgastrointestinal fluids for 8 h. In addition, accelerated stability was alsoestablished for developed formulation as per ICH guidelines. Usually thechronic therapy of anticancer agents is associated with the reduced an-tioxidant levels due to higher reactive oxygen species (ROS) generationmediated by these agents for execution of their cytotoxic effect. Hence-forth, co-administration of antioxidants is generally prescribed. Howev-er, the physicochemical properties of antioxidants such as CoenzymeQ10, quercetin and curcumin generally make them difficult-to-deliver,which can be specifically tackled by nanocarrier based approach [189].We have successfully developed and patented the SEDDS of quercetinfor improving its oral bioavailability [289]. About 4.98-fold increase inthe oral bioavailability of quercetin was observed as compared to free

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 17: Oral delivery of anticancer drugs: Challenges and opportunities

T

1208

1209

1210

1211

1212

1213

1214

1215

1216

1217

1218

1219

1220

1221

1222

1223

1224

1225

1226

1227

1228

1229

1230

1231Q31232

1233

1234

1235

1236

1237

1238

1239

1240

1241

1242

1243

1244

1245

1246

1247

1248

1249

1250

1251

1252

1253

1254

1255

1256

1257

1258

1259

1260

1261

1262

1263

1264

1265

1266

1267

1268

1269

1270

1271

1272

1273

1274

1275

1276

1277

1278

1279

1280

1281

1282

1283

1284

1285

1286

1287

t6:1

t6:2

t6:3

t6:4

t6:5

t6:6

t6:7

t6:8

t6:9

t6:10

t6:11

t6:12

t6:13

t6:14

t6:15

t6:16

t6:17

17K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

REC

drug. Further, the developed formulation also posed significantly higherprophylactic and therapeutic antitumor efficacy as compared to freequercetin in DMBA induced breast cancer model in rats (unpublisheddata). In addition, combination of antioxidant loaded SEDDS with anti-cancer agents is also reported to have higher therapeutic efficacy ascompared to individual anticancer agents. Recently, co-administrationof docetaxel with curcumin loaded SEDDS led to about 3.2-fold increasein the oral bioavailability of docetaxel as compared to the free drug. Theinhibitory effect of curcumin on P-gp and cytochrome P450 was attrib-uted to this increment in oral bioavailability of docetaxel [290]. Table 6reflects the exhaustive list of self emulsifying formulation approach forimproving the oral bioavailability of anticancer agents.

3.2.3.3. Solid lipid nanoparticles (SLNs). SLNs are the alternative versionof emulsions in which the liquid oil is replaced by solid lipids. Specificadvantages include modulation of drug release, increased drug stabili-ty, exclusion of organic solvents from the manufacturing process,manufacturing scalability and industrial adaptability [303]. These aregenerally prepared by various commonly used techniques such as hothomogenization, cold homogenization, microemulsion technique,solvent emulsification/evaporation, spray drying and so on. The lipidsthat are used to prepare SLNs include fatty acids (e.g. stearic acid),fatty acid esters (e.g. glyceryl monostearate, glyceryl behenate), triglyc-erides (e.g. tristearin, trilaurin), steroids (e.g. cholesterol) and waxes(e.g. cetyl palmitate). Depending on the charge, molecular weight andtheir capability to stabilize the dispersion, single or combination ofemulsifiers are chosen and incorporated into the system. Commonlyused emulsifiers and co-emulsifiers include lecithin, poloxamers, cho-lates, and so on. Our group has exhaustively reviewed various aspectsof solid lipid nanoparticles for improving the oral bioavailability ofdifficult-to-deliver drugs [174]. Recently, paclitaxel has been formulat-ed as SLNs using stearic acid as lipid and lecithin and poloxamer as sur-factants [304]. The paclitaxel loaded SLNs showed superior in vitrocytotoxicity as compared to free drug. A similar study for camptothecinhas been carried with an intention of sustaining the drug release [305].In order to further modulate the drug release; SLNs were further conju-gated with dextran methacrylate hydrogels [306]. Reviewing the solidlipid nanoparticles, it has been generally found that the drug releaseis often retarded due to high lipophilic nature of the overall system,but considering the urge to increase the drug release rate; polymerlipid hybrid nanoparticles were developed [173]. These are based on

UNCO

RTable 6List of anticancer agents benefited by self-emulsifying drug delivery systems for improving

Drug Oils Surfactants

9-Nitrocamptothecin Ethyl oleate Tween 80 and PEG 400Curcumin Castor oil Tween 80 and ethanolCurcumin Lauroglycol Labrasol and Transcutol HP

Curcumin Gelucire 44/14 Labrasol, Vitamin E TPGS and PEG 400

Curcumin Isopropyl myristate Cremophor RH 40 and ethanol

Docetaxel Glyceryl tricaprylate Cremophor RH 40, Diethylene Glycol MEther; HPMC as super-saturation prom

Etoposide PC-complexed; Octyl anddecyl mono-glyceride

Cremophor EL and PEG 400

Exemestane Capryol 90 Transcutol P and Cremophor ELP

Mitotane Capryol 90 Tween 80 and Cremophor EL

Raloxifene Capmul MCM C8 Tween-20 and Akrysol K140 and PEG-

Paclitaxel Vitamin E DOC-Na, TPGS, Propylene glycol, CremPaclitaxel Glyceryl dioleate Cremophor EL, PEG 400; HPMC as supe

promoterPaclitaxel DL-alpha tocopherol TPGS, tyloxapol, DOC-Na

PC: Phosphatidylcholine; DOC-Na: Sodiumdeoxy cholate; TPGS: D-alpha-tocopheryl polyethylen

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

same principle of SLNs, and incorporation of the ionic polymers helpsin improving the encapsulation of water soluble ionic drugs such asdoxorubicin hydrochloride [307]. This system showed 8-fold highertumor cell inhibition in multi drug resistant cell lines compared tofree drug along with greater drug uptake and retention by cell lines[308]. Table 7 represents the applicability of SLNs in improving thedeliverability of various anticancer drugs.

3.2.3.4. Nanostructured lipid carriers (NLCs). NLCs are the advancedgeneration of SLNs overcoming the problems associated with thelater such as limited drug loading capacity, restructuring during stor-age and subsequently expulsion of drug during storage. The saidadvantages could be attributed to higher solubility of drugs in oilsas compared to solid lipids. However, inclusion of drug beyond oil/lipid solubility is practically difficult and often needs alternative strat-egy. Hence high dose compounds are poor candidates for these sys-tems. NLCs are prepared by similar technique as reported for SLNswith only difference that both solid lipid and liquid oil are mixedfor formation of NLCs in contrast to only solid lipid for SLNs. Thisleads to formation of porous matrix structure resulting in higherdrug pay load that is entrapped throughout the shelf life of the prod-uct [318]. The stability of NLCs can further be enhanced by addition ofpreservatives such as propylene glycol, pentylene glycol, etc. Variousstabilization aspects of NLCs have been recently studied. Briefly, theparameters which need consideration include affinity of the preserva-tive to the particle surface, surface hydrophobicity of the particles,anchoring of stabilizer onto/into surface, ability of preservative to re-duce zeta potential, nature of the particle stabilizer and interaction ofpreservative with stabilizer layer [319]. Recently, tocotrienol loadedNLCs were developed and evaluated for their antiproliferative effectagainst +SA epithelium cell lines and were found to be about 2-foldmore effective as compared to free tocotrienol [320]. Furthermore,NLCs have also been implemented for oral delivery of etoposide andabout 3.5-fold increase in the oral bioavailability was observed ascompared to free drug [321]. Increased permeation across, decreasedclearance and specialized uptake of nanoparticles were attributed forthe probable reasons for increased oral bioavailability. Furthermore,the developed formulation posed about 5-fold increase in the in vitrocytotoxicity against A549 cell lines as compared to free drug, suggestiveof potential of NLCs based drug delivery system for improving the oraldeliverability of various anticancer drugs.

their oral bioavailability.

Outcome Ref

2.23-Fold increase in bioavailability [291]About 2000-fold increase in the solubility of curcumin [292]7.6-Fold increase in the oral bioavailability as compared tofree drug.

[293]

35.8-Fold increase in the oral bioavailability as compared tocontrol

[294]

12-Fold increase in the oral bioavailability as compared tofree drug.

[295]

onoethyloter

8.77-Fold increase in the total AUC [283]

60-Fold increase in bioavailability [296]

About 2.8-fold increase in the oral bioavailability as comparedto free drug

[297]

About 3.4-fold increase in oral bioavailability as compared toclinical formulation

[298]

200 About 2-fold increase in the intestinal permeability across therat intestine as compared to plain drug

[299]

ophor RH 40 1.2–1.5-Fold increase in bioavailability [300]rsaturation 5-Fold higher bioavailability [301]

3-Fold increase in the solubility of Paclitaxel [302]

e glycol 1000 succinate; PEG: Polyethylene glycol;HPMC: Hydroxypropylmethyl cellulose.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 18: Oral delivery of anticancer drugs: Challenges and opportunities

T

F

1288

1289

1290

1291

1292

1293

1294

1295

1296

1297

1298

1299

1300

1301

1302

1303

1304

1305

1306

1307

1308

1309

1310

1311

1312

1313

1314

1315

1316

1317

1318

1319

1320

1321

1322

1323

1324

1325

1326

1327

1328

1329

1330

1331

1332

1333

1334

1335

1336

1337

1338

1339

1340

1341

1342

1343

1344

1345

1346

1347

1348

1349

1350

1351

1352

1353

1354

1355

1356

1357

1358

1359

1360

1361

1362

1363

1364

1365

1366

1367

1368

1369

1370

1371

1372

1373

1374

1375

1376

1377

1378

1379

Table 7t7:1

t7:2 List of anticancer drugs benefited from solid lipid nanoparticles for improvement in oral bioavailability.

t7:3 Drug Ingredients Outcomes Ref

t7:4 5-Flurouracil Dynasan, soya lecithin, polyvinyl alcohol Sustained release of drug till 48 h [309]t7:5 Camptothecin Stearic acid, Soya lecithin, poloxamer 188 About 1.4-fold increase in the oral bioavailability as compared to free drug was observed [305]t7:6 Doxorubicin Emulsifyingwax, Brij 78 and Vitamin E TPGS About 9-fold increase in the in vitro cytotoxicity against P388/ADR cancer cell lines as compared to free drug [310]t7:7 Emodin Stearic acid, poloxamer 188 and Tween 80 Significantly higher in vitro cytotoxicity against MCF-7 and MDA-MB-231 cancer cell lines as compared to

free drug.[311]

t7:8 Methotrexate Stearic acid, Soya lecithin and sodiumtaurodeoxy-cholate

Significant improvement in the half life and mean residence time of formulation as compared to free drug.The life span of Ehrlich Ascites Carcinoma bearing mice was increased upon treatment with formulation ascompared to free drug

[312]

t7:9 Tamoxifencitrate

Glycerol behenate, sodiumtauroglycocholate

About 3.5-fold increase in the mean residence time of formulation as compared to free drug [313]

t7:10 Paclitaxel Stearic acid, lecithin and poloxamer 188 Significant improvement in the in vitro cytotoxicity of paclitaxel as compared to free drug [304]t7:11 Paclitaxel Folate-(PEG)-phosphatidyl-ethanolamine Significant improvement in in vivo antitumor efficacy of formulation as compared to free drug [314]t7:12 Paclitaxel Glyceryl palmitostearate nanoparticles Retention of the in vitro cytotoxicity of paclitaxel against B16F10 cell lines [315]t7:13 Paclitaxel Folate modified monostearin About 11-fold increase in the in vitro cytotoxicity against A549 cell lines as compared to free drug [316]t7:14 Vinorelbine PEG2000-stearic acid About 27-fold and 25-fold increase in the in vitro cytotoxicity of vinorelbine in developed formulation

against MCF-7 and A549 cell lines, respectively was observed as compared to free drug.[317]

18 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

3.2.3.5. Lipid nanocapsules. Lipid nanocapsules are the novel colloidalnanocarriers having typical core shell arrangement of the constituentswith liquid reservoir (usually oils) as core coated by protective mem-brane. The protectivemembrane generally comprises of polymer chainspresent in the surfactant which is aligned as exterior coat during thepreparation of lipid nanocapsules [322]. Similar to nanoemulsions,various methods have been proposed for formulation of lipid nano-capsules, out of which phase inversion temperature is most commonlyadapted [323]. Briefly, the method relies on the properties of thenon-ionic surfactants such as PEG hydroxystearates, having tendencyto pose temperature dependent solubility. The lipid nanocapsules areprincipally known for their biocompatibility, biodegradability, superiorencapsulation of drug molecules, very high drug payloads, specific ab-sorption mechanism across the gastrointestinal tract, sustained release,membrane efflux transporter inhibition, targeting potential (active andpassive) and most importantly oral deliverability [322]. Furthermore,the aqueous-core lipid nanocapsules have also been proposed that arecapable of incorporating both hydrophilic and lipophilic drugmolecules[324].

Recently, the stability of the paclitaxel loaded lipid nanocapsuleswas established in the gastrointestinal tract. The stability of these sys-tems was attributed to the presence of the surface PEG coating, whichspecifically protected the aggregation of internal lipid constituentsdue to acidic pH in the stomach and against pancreatin activity in in-testine. However, weaker protection was exhibited in the presence ofbile salts and hence pre-prandial administration of lipid nanocapsulesfor better stability profile along the gastrointestinal tract could besought [325]. With these promising results, the uptake of the lipidnanoparticles across the in vitro Caco-2 cell monolayer model wasevaluated. About 3.5-fold increase in the apparent permeability ofpaclitaxel upon its loading in lipid nanocapsules was observed ascompared to free drug; vesicle mediated transcytosis being the pro-posed mechanism for improved permeability [172]. These resultsare in correlation with pharmacokinetic studies conducted by anothergroup which posed about 3-fold increase in the oral bioavailability ofpaclitaxel when loaded in lipid nanocapsules as compared to freedrug [12]. Similarly, about 18-fold increase in the apparent perme-ability of SN-38 in Caco-2 cell monolayer model was observed whenloaded in lipid nanocapsules as compared to free drug [326].

3.2.3.6. Liposomes. Liposomes are the most sophisticated class of lipidbased drug delivery systems known for improving the deliverabilityof various difficult-to-deliver drugs. These are vesicular structuresprepared from phospholipids and are capable of accommodatingboth hydrophilic and lipophilic drug substances within them. Theseare reported to have huge potential in delivery of various anticanceragents especially in targeted drug delivery [327–329]. However, they

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

O

are not stable in harsh gastrointestinal environment. The principal rea-sons for their instability comprises of acidic environment in the stom-ach (that leads to aggregation of constituent phospholipids) andpresence of degrading enzymes in intestine (such as pancreatic lipase).Henceforth various attempts have been made in this direction to im-prove their oral stability. The classical approach includes surface modi-fication such as PEGylation [330], transphosphatidylation [331], mucincoating (mucin liposomes) [332], polymer coatings (polymerized lipo-somes) [333], polyelectrolyte coatings (nanocapsules) [334] and chito-san coating [335]. Recently, exemestane loaded proliposomes wereprepared using distearoyl phosphatidylcholine (DSPC), cholesteroland dimyristoyl-phosphatidylglycerol (DMPG). These proliposomesare in the free flowing powder form which upon exposure to aqueousenvironment results in to formation of multilamellar liposomes thatmay be optionally sonicated for generation of desired sized liposomes.The developed formulation was found to exhibit about 3-fold increasein the permeability across the rat intestine as compared to the plaindrug [336]. The probable mechanisms for increased oral bioavailabilityinclude higher accumulation at the brush bordermembrane thereby in-creasing the concentration gradient across the intestinal epithelium, in-creased paracellular absorption, receptor mediated endocytosis andpredominantly byM-cells present over Peyer's patches along the intes-tine [337]. Although both hydrophilic and lipophilic drugs could beloaded in to liposomes, the concerns on drug loading, entrapment effi-ciency, cost incurred, and long term stability should be considered be-fore developing a particular drug compound in liposome based drugdelivery system.

3.2.3.7. Layersomes. Layersomes, by definition, are composed of thelayer by layer (LbL) coating of the polyelectrolytes over liposomesto increase stability of the formulation in the biological milieu andlong term storage. Our group has pioneered the multiple polyelectro-lyte stabilized polymeric nanocapsules for oral delivery of therapeuticagents. Recently, this patented approach has been successfullyimplemented for design and development of oral delivery of variousdifficult-to-deliver anticancer agents [338]. About 5.89-fold increasein the oral bioavailability of doxorubicin as compared to the freedrug and comparable therapeutic efficacy and toxicity profile wereobserved as compared to i.v. LipoDox® [339]. Similar results werealso observed in case of paclitaxel with about 4.07 fold increase inthe oral bioavailability, superior therapeutic efficacy and safety pro-file as compared to i.v. Taxol [340]. Recently, doxorubicin loadedpoly(L-glutamic acid) microcapsules have also been prepared usingLbL approach and was evaluated on multi-drug-resistant (MDR) celllines [341]. A significant level of accumulation was found in the celllines and internalization of the prepared polyelectrolyte polymer cap-sules was attributed to macropinocytosis [342].

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 19: Oral delivery of anticancer drugs: Challenges and opportunities

T

1380

1381

1382

1383

1384

1385

1386

1387

1388

1389

1390

1391

1392

1393

1394

1395

1396

1397

1398

1399

1400

1401

1402

1403

1404

1405

1406

1407

1408

1409

1410

1411

1412

1413

1414

1415

1416

1417

1418

1419

1420

1421

1422

1423

1424

1425

1426

1427

1428

1429

1430

1431

1432

1433

1434

1435

1436

1437

1438

1439

1440

1441

1442

1443

1444

1445

1446

1447

1448

1449

1450

1451

1452

1453

1454

1455

1456

1457

1458

1459

1460

1461

1462

1463

1464

1465

1466

1467

1468

1469

1470

1471

1472

1473

1474

1475

1476

1477

1478

1479

1480

1481

1482

1483

1484

1485

1486

1487

1488

1489

1490

1491

1492

1493

1494

1495

1496

1497

1498

1499

1500

1501

1502

1503

1504

1505

1506

1507

1508

1509

19K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

3.2.3.8. Liquid crystalline nanoparticles (LCNPs). LCNPs are consideredas sophisticated lipid based nanocarriers prepared from polar lipids,having capability to encapsulate both hydrophilic and lipophilicdrugs [343]. The different types of lipids/surfactants which could beemployed for preparation of LCNPs include Glyceryl monooleate,selachyl alcohol, glyceryl dioleate, phytantriol, oleyl glycerate,phytanyl glycerate, Brij 30/Brij 35 combination, monopalmitolein,ethoxylated fatty alcohols, monoolein/phosphatidyl choline combina-tion, etc. [344]. Our group has recently employed LCNPs for improvingthe antitumor efficacy of various anticancer agents such as doxorubi-cin, tamoxifen, docetaxel, CoQ10, curcumin etc. About 5.34-foldincrease Cmax and 17.74-fold increase in area under curve was ob-served upon incorporation of doxorubicin in to LCNPs as comparedto free drug (unpublished data). Further an insignificant differencein tumor inhibition of LCNPs (oral) vs. LipoDox® (i.v.) was observedin DMBA induced breast cancer model in rats till 20 days. Additional-ly, the developed formulation posed negligible cardiotoxicity incontrast to clinical formulation. The prophylactic and therapeuticantitumor efficacy of CoQ10 was significantly increased upon incor-poration in to LCNPs as compared to free drug (unpublished data).Mechanistically, LCNPs are known to increase the oral bioavailabilityby virtue of bioadhesiveness, membrane fusing properties, clathrin/caveolae/lipid raft-mediated endocytosis, superior encapsulation, sol-ubilization, etc. [345].

3.2.3.9. Lipid–drug conjugates. Lipid–drug conjugate approach is one ofthe most advanced versions of the lipid based drug delivery systemsin which the concerned drug is covalently linked to lipids thereby im-proving the drug payload into the system up to as high as 40–50% w/w [318]. The lipids that can be used for formulation of lipid–drug con-jugates include phospholipids, fatty acids such as docosahexaenoicacid, stearic acid, oleic acid, etc. and lipoamino acids. Methotrexateconjugated dihexanoylphosphatidylethanolamine was synthesizedfor improving its impaired transport across cell membranes, fre-quently leading to resistance. The developed system was found~120-fold more effective against methotrexate resistant humanT-lymphoblastic leukemia cell line as compared to free methotrexate[346]. Furthermore, methotrexate conjugated with lipoamino acidswas found ~150-fold more effective than free methotrexate undersimilar set of conditions [347]. In another set of conditions, paclitaxelconjugated docosahexaenoic acid showed about 61-fold higher accu-mulation of conjugated form at tumor site as compared to free drug atequitoxic doses whereas about 8-fold increase was observed at equi-molar doses [348]. Furthermore, stearic acid conjugated methotrex-ate has been designed and developed which showed significantimprovement in the oral bioavailability of methotrexate as comparedto free drug [349]. Recently, phytanyl pro-drug analogue of capecitabinehas been synthesized and evaluatedwhich revealed significantly higherantitumor efficacy as compared to parent compound [350]. Important-ly, the resulting lipid–drug conjugate is amphiphilic in nature and iscapable of self assembling with definite structure which could furtherassist in the stabilization and superior absorption of drug in the gastro-intestinal tract. Again here also the principal concerns as applicableto polymer–drug conjugates equally stands well and should beconsidered.

3.2.4. DendrimersDendrimers are the hyper branched and uniformly distributed

macromolecules that possess definite molecular weight, shape, sizeand specific chemical and physical properties including host–guestentrapment properties [351]. The tailor made design makes these so-phisticated systems eligible for incorporation of wide variety of drugsthat otherwise pose potential delivery challenges. The drug moleculescan be either physically entrapped or chemically conjugated to thedendritic structures during or after synthesis of macromolecularsystems. Furthermore, encapsulation of drug molecules within the

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

dendritic structures selectively eliminates solubility and permeabilityrelated issues. Henceforth, these systems can be exploited to accom-plish the oral delivery of anticancer agents. The applicability ofdendrimers in cancer therapeutics has been exhaustively reviewedrecently [352]. However, very few reports are available on utilizationof these systems for oral delivery.

The principal reason for intuitions on dendrimers having potentialfor oral delivery of drugmolecules includes their high solubilization ca-pacity [353], increased permeation capability across the gastrointestinaltract via paracellular or transcellular (preferentially via clathrin-dependent endocytosis) pathway [354], encapsulation of the drugmol-ecules, thereby protection fromharsh gastrointestinal environment andhigh drug payload (in contrast to other nanocarriers in race) [355]. Theproperties of dendrimers such as size, conformation and surface chargehave been found to significantly affect the absorption of these systemsacross the GIT [351]. Dendrimers with particle size ≤ 3 nm werefound to permeate via transcellular or paracellular pathway whereashigher sized permeated through nonspecific adsorptive endocytosis[356]. Concomitantly, anionic dendrimers showed relatively higherserosal transfer as compared to anionic dendrimers, which could be at-tributed to the higher interaction of these systems with negativelycharged intestinal epithelium. Furthermore, cationic dendrimers alsoshowed some evidence of toxicity in glucose-active transport assay athigher concentrations. Cumulatively, anionic dendrimers posed greaterpotential for oral drug delivery in compassion to its cationic counterpart[357].

The permeability studies of various dendrimer generations againstCaco-2 cell monolayers revealed significant reduction in thetransepithelial electrical resistance values and increase in the [14C]mannitol permeability as a function of donor concentration, incuba-tion time and generation number, thereby confirming the potentialof these systems for oral delivery of drug molecules. In addition, thelower generations in the order of G2 were found nontoxic to theCaco-2 cells, which further paved the way for subsequent studies[358]. The hypothesis was confirmed when doxorubicin complexedpoly(amido amine) (PAMAM) dendrimers were developed and eval-uated. The developed complex showed about 200-fold higher oralbioavailability as compared to free doxorubicin [359]. Similar resultshave also been observed for an active metabolite of irinotecan(SN-38) loaded PAMAM dendrimer, which showed pH dependent re-lease profile (no release at physiological pH 7.4 and significantlyhigher drug release at tumor pH 5.5) with about 10-fold increase inthe permeability across and about 100-fold increase in cellular uptakeby Caco-2 cells [360]. The permeation enhancer effect of PAMAMdendrimers has also been proven recently, which revealed reversalof permeation irrespective of the time, even within 10 min of pre-treatment. This was further confirmed by absence of any membranetoxicity in intestinal epithelia of rats measured as a function of re-leased lactate dehydrogenase (LDH) and protein post treatment[361].

The properties of these dendrimer systems can be further enhancedupon surface functionalization such as PEGylation, polyamine func-tionalization, acetylation or lipid anchoring. The PEGylation of theanionic dendrimers altered their transport mechanism across the gas-trointestinal tract and in fact decreased transport was observed in alltested cases of G3 and G4 dendrimers. However, the decrease in thetransport of these dendrimers was marginal and comparable to othermacromolecules. Therefore, authors concluded that this decrease canbe afforded to avail the advantages offered by PEGylation such as facil-itated conjugation and improved biodistribution post absorption [362].On the contrary, the surface modification of PAMAM dendrimers withpolyamines such as arginine and ornithine significantly improvedthe apparent permeability of modified dendrimers as compared tounmodified dendrimers when tested in Caco-2 cell model. The authorsproposed polyamine transported system for improved permeability.However, the toxicity of argininemodified dendrimerswas significantly

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 20: Oral delivery of anticancer drugs: Challenges and opportunities

T

1510

1511

1512

1513

1514

1515

1516

1517

1518

1519

1520

1521

1522

1523

1524

1525

1526

1527

1528

1529

1530

1531

1532

1533

1534

1535

1536

1537

1538

1539

1540

1541

1542

1543

1544

1545

1546

1547

1548

1549

1550

1551

1552

1553

1554

1555

1556

1557

1558

1559

1560

1561

1562

1563

1564

1565

1566

1567

1568

1569

1570

1571

1572

1573

1574

1575

1576

1577157815791580158115821583158415851586158715881589159015911592159315941595159615971598159916001601160216031604160516061607160816091610161116121613161416151616161716181619162016211622162316241625162616271628162916301631163216331634163516361637163816391640164116421643164416451646164716481649165016511652

20 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

higher at all tested concentrations as compared to unmodified and orni-thine conjugated dendrimers [363]. In yet another set of studies the tox-icity of the cationic PAMAM dendrimers was reduced by about 10-foldby acetylation of their terminal amine groups. No compromise on thepermeability was observed rather permeability was increased due toavoidance of any non specific binding to highly reactive amine groups[364]. Furthermore, about 8–10-fold increase in the apparent perme-ability of [14C]mannitol and significant reduction in toxicity wereobserved upon surface engineering of cationic PAMAM dendrimerswith lauroyl chloride as compared to unmodified cationic PAMAMdendrimers [365]. The probable reason for this increment in perme-ation by surface lipid functionalization could be attributed to the exploi-tation of phospholipase-C (PLC)-dependent pathway and transcellularpathway (via perturbation and change of fluidity of the cellmembrane).Similar results were also observed upon conjugating lipid (C12 chains)with polylysine dendrimers revealing the potential of these lipidfunctionalizations on dendrimers for oral delivery [366].

4. Conclusion

Although the newer generations of anticancer agents which can bedelivered orally are at priority in developmental pipeline, the classicaldrug substances can also bedelivered efficiently via specific formulationdesign approach. The poor physicochemical and biopharmaceuticalproperties associated with the various anticancer drugs hinderingtheir oral deliverability can be effectively circumvented by utilizationof absorption enhancers (P-gp inhibitors and functional excipients)and pharmaceutical approaches such as nanocrystals and nanocarriers.These novel drug delivery systems owing to their special properties areable to bypass various barriers of drug delivery across the gastrointesti-nal tract. Furthermore, the targeting potential of these systems is of spe-cial interest in the cancer therapy. Passive targeting via enhancedpermeation and retention is one of the common and important advan-tages offered by almost all types of nanocarriers. The oral delivery ofanticancer agents via such drug delivery systems is of great interestfor improving the quality of life of patients suffering cancer. In addition,the pharmacoeconomic advantage with oral delivery of ‘injection only’drugs will fetch significant attraction of health care agencies by reduc-ing the overall cost incurred in health care management.

5. Future prospects

With the advent of nanotechnology in the drug delivery applica-tions for improving the deliverability of various difficult-to-deliverdrugs, there arises obvious concerns that need to be addressed. Theprincipal concern is the safety profile of these nanocarriers for chronictreatment which is already under consideration and dedicated effortsare being made by scientific community and health care agencies. Theresults so far seems to be promising which is evident from the factthat there are numerous nanotechnology based products approvedby regulatory agencies and are in clinical use. The increasing numberof such products under clinical trials clearly reflects the thrust in thescientific community in this area. Sincere efforts are made in thefield of understanding the mechanism of cellular trafficking of thesenanocarriers and its correlation with the efficacy and toxicity profileis one of the priority research areas. The insight of these will lead todevelopment in the newer technologies that would be able to exploitthe physiological principles for drug delivery purposes withoutcompromising the safety profile. Furthermore, more attempts canbe made for translation of the laboratory developments in to productdevelopment. This could be achieved by efforts in the area of improv-ing the drug payload within the nanocarriers, simple manufacturingsteps, usage of inexpensive excipients and robust formulation design.Furthermore, attempts can also be made to further exploit thetargeting potential of nanocarriers so as to increase their uptakefrom gastrointestinal tract to an extent comparable to that of

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

parenteral administration. In addition, the utilization of the functionalexcipients within the carrier systems can also be exploited to greaterextent so as to design and develop rationalized drug delivery systemsfor various difficult to treat diseases such as cancer.

ED P

RO

OF

References

[1] C.H. Lieu, W.N. William, S.M. Lippman, Cancer chemoprevention, in: M. Hidalgo,S.G. Eckhardt, E. Garrett-Mayer, N.J. Clendeninn (Eds.), Principles of AnticancerDrug Development, Springer Science, New York, 2010, pp. 463–482.

[2] WHO, Cancer-fact sheets in, http://www.who.int/mediacentre/factsheets/fs297/en/index.html, (WHO, Accessed on January 23, 2012).

[3] K. Ruddy, E. Mayer, A. Partridge, Patient adherence and persistence with oral an-ticancer treatment, CA Cancer J. Clin. 59 (2009) 56–66.

[4] V.J. O'Neill, C.J. Twelves, Oral cancer treatment: developments in chemotherapyand beyond, Br. J. Cancer 87 (2002) 933–937.

[5] L.A. Jeanneret, M.P. Schneider, S. Troxler, O. Bugnon, F. Luthi, Therapeutic adher-ence to oral cancer therapy and interdisciplinary management, Rev. Med. Suisse7 (2011) 1154–1158.

[6] A.H. Partridge, J. Avorn, P.S. Wang, E.P. Winer, Adherence to therapy with oralantineoplastic agents, J. Natl. Cancer Inst. 94 (2002) 652–661.

[7] G.L. Banna, E. Collovą, V. Gebbia, H. Lipari, P. Giuffrida, S. Cavallaro, R. Condorelli,Anticancer oral therapy: emerging related issues, Cancer Treat. Rev. 36 (2010)595–605.

[8] J. Wojtacki, G. Rolka-Stempniewicz, M. Grzegorczyk, Breast cancer patients pref-erences for oral versus intravenous second-line anticancer therapy, Eur. J. Can-cer (Suppl.) 4 (2006) 159–160.

[9] N. Ramnath, P.J. Creaven, Matrix metalloproteinase inhibitors, Curr. Oncol. Rep.6 (2004) 96–102.

[10] N.M.G.M. Appels, J.H. Beijnen, J.H.M. Schellens, Development of farnesyl trans-ferase inhibitors: a review, Oncologist 10 (2005) 565–578.

[11] M.S. Gordon, D.S. Mendelson, G. Kato, Tumor angiogenesis and novelantiangiogenic strategies, Int. J. Cancer 126 (2010) 1777–1787.

[12] S. Peltier, J.M. Oger, F. Lagarce, W. Couet, J.P. Benoît, Enhanced oral paclitaxelbioavailability after administration of paclitaxel-loaded lipid nanocapsules,Pharm. Res. 23 (2006) 1243–1250.

[13] I. Kuppens, T.M. Bosch, M.J. van Maanen, H. Rosing, A. Fitzpatrick, J.H. Beijnen,J.H.M. Schellens, Oral bioavailability of docetaxel in combination withOC144-093 (ONT-093), Cancer Chemother. Pharmacol. 55 (2005) 72–78.

[14] M.D. Troutman, D.R. Thakker, Novel experimental parameters to quantify themodulation of absorptive and secretory transport of compounds byP-glycoprotein in cell culture models of intestinal epithelium, Pharm. Res. 20(2003) 1210–1224.

[15] S.C. Shin, J.S. Choi, X. Li, Enhanced bioavailability of tamoxifen after oral admin-istration of tamoxifen with quercetin in rats, Int. J. Pharm. 313 (2006) 144–149.

[16] A.S. Narang, D.S. Desai, Anticancer drug development, in: Y. Lu, R.I. Mahato(Eds.), Pharmaceutical Perspectives of Cancer Therapeutics, Springer, NewYork, 2009, pp. 49–92.

[17] R.F. Struck, D.S. Alberts, K. Horne, J.G. Phillips, Y.M. Peng, D.J. Roe, Plasma phar-macokinetics of cyclophosphamide and its cytotoxic metabolites after intrave-nous versus oral administration in a randomized, crossover trial, Cancer Res.47 (1987) 2723–2726.

[18] V. Bhardwaj, D. Ankola, S. Gupta, M. Schneider, C.M. Lehr, M.N.V.R. Kumar, PLGAnanoparticles stabilized with cationic surfactant: safety studies and applicationin oral delivery of paclitaxel to treat chemical-induced breast cancer in rat,Pharm. Res. 26 (2009) 2495–2503.

[19] J. van Asperen, O. van Tellingen, M.A. van der Valk, M. Rozenhart, J.H. Beijnen,Enhanced oral absorption and decreased elimination of paclitaxel in micecotreated with cyclosporin A, Clin. Cancer Res. 4 (1998) 2293–2297.

[20] M. Gore, A. Oza, G. Rustin, J. Malfetano, H. Calvert, D. Clarke-Pearson, J.Carmichael, G. Ross, R. Beckman, S. Fields, A randomised trial of oral versusintravenous topotecan in patients with relapsed epithelial ovarian cancer, Eur.J. Cancer 38 (2002) 57–63.

[21] J. von Pawel, U. Gatzemeier, J.L. Pujol, L. Moreau, S. Bildat, M. Ranson, G.Richardson, C. Steppert, A. Rivière, I. Camlett, Phase II comparator study of oralversus intravenous topotecan in patients with chemosensitive small-cell lungcancer, J. Clin. Oncol. 19 (2001) 1743–1749.

[22] S. Urien, E. Brain, R. Bugat, X. Pivot, I. Lochon, M.L.V. van, F. Vauzelle, F. Lokiec,Pharmacokinetics of platinum after oral or intravenous cisplatin: a phase 1study in 32 adult patients, Cancer Chemother. Pharmacol. 55 (2005) 55–60.

[23] C. Manegold, P. Drings, A. Pawinski, M.A. Lentz, M. van Glabbeke, N. vanZandwijk, P. Bachmann, Y. Schnaars, P. Zatloukal, J. Dolensky, Oral ifosfamide/mesna versus intravenous ifosfamide/mesna in non-small-cell lung cancer: arandomized phase II trial of the EORTC lung cancer cooperative group, Ann.Oncol. 7 (1996) 637–639.

[24] A.G. Bosanquet, E.D. Gilby, Pharmacokinetics of oral and intravenous melphalanduring routine treatment of multiple myeloma, Eur. J. Cancer Clin. Oncol. 18(1982) 355–362.

[25] C.A. Lipinski, Drug-like properties and the causes of poor solubility and poorpermeability, J. Pharmacol. Toxicol. 44 (2000) 235–249.

[26] J. Brouwers, M.E. Brewster, P. Augustijns, Supersaturating drug delivery systems:the answer to solubility-limited oral bioavailability? J. Pharm. Sci. 98 (2009)2549–2572.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 21: Oral delivery of anticancer drugs: Challenges and opportunities

T

16531654165516561657165816591660166116621663166416651666166716681669167016711672167316741675167616771678167916801681168216831684168516861687168816891690169116921693169416951696169716981699170017011702170317041705170617071708170917101711171217131714171517161717171817191720172117221723172417251726172717281729173017311732173317341735173617371738

17391740174117421743174417451746174717481749175017511752175317541755175617571758175917601761176217631764176517661767176817691770177117721773177417751776177717781779178017811782178317841785178617871788178917901791179217931794179517961797179817991800180118021803180418051806180718081809181018111812181318141815181618171818181918201821182218231824

21K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

[27] S. Kiyohiko, Fraction of a dose absorbed estimation for structurally diverse lowsolubility compounds, Int. J. Pharm. 405 (2011) 79–89.

[28] M.V. Varma, R. Panchagnula, Prediction of in vivo intestinal absorption enhance-ment on P-glycoprotein inhibition, from rat in situ permeability, J. Pharm. Sci. 94(2005) 1694–1704.

[29] S. Yee, In vitro permeability across Caco-2 cells (colonic) can predict in vivo (smallintestinal) absorption in man—fact or myth, Pharm. Res. 14 (1997) 763–766.

[30] W. Fayad, L. Rickardson, C. Haglund, M.H. Olofsson, P. D'Arcy, R. Larsson, S.Linder, M. Fryknas, Identification of agents that induce apoptosis of multicellulartumour spheroids: enrichment for mitotic inhibitors with hydrophobic proper-ties, Chem. Biol. Drug Des. 78 (2011) 547–557.

[31] J.A. Platts, M.H. Abraham, D. Butina, A. Hersey, Estimation of molecular linearfree energy relationship descriptors by a group contribution approach. 2. Predic-tion of partition coefficients, J. Chem. Inf. Comput. Sci. 40 (2000) 71–80.

[32] S.N.H. Oo, N.H. Ho, Guanidines, acylguanidines, and biguanides, in: F.Z. Dorwald(Ed.), Lead Optimization for Medicinal Chemists: Pharmacokinetic Properties ofFunctional Groups and Organic Compounds, Wiley VCH Verlag & Co. KGaA,Weinheim, Germany, 2012, pp. 122–132.

[33] O. Ottmann, H. Dombret, G. Martinelli, B. Simonsson, F. Guilhot, R.A. Larson, G.Rege-Cambrin, J. Radich, A. Hochhaus, A.M. Apanovitch, A. Gollerkeri, S.Coutre, Dasatinib induces rapid hematologic and cytogenetic responses inadult patients with Philadelphia chromosome positive acute lymphoblastic leu-kemia with resistance or intolerance to imatinib: interim results of a phase 2study, Blood 110 (2007) 2309–2315.

[34] N. Suvannang, C. Nantasenamat, C. Isarankura-Na-Ayudhya, V. Prachayasittikul,Molecular docking of aromatase inhibitors, Molecules 16 (2011) 3597–3617.

[35] A. Schulz, Y. Han, Z. He, T.K. Bronich, A.V. Kabanov, R. Luxenhofer, R. Jordan,Poly(2-oxazoline)s: an all-around drug delivery system? Polym. Prepr. 53(2012) 354–355.

[36] A. Patil, Y. Pore, B. Kuchekar, Effect of L-arginine on bicalutamide complexationwith hydroxypropyl-beta-cyclodextrin, Digest J. Nanomat. Biostruct. 3 (2008)89–98.

[37] A. Palmeira, F. Rodrigues, E. Sousa, M. Pinto, M.H. Vasconcelos, M.X. Fernandes,Pharmacophore-based screening as a clue for the discovery of newP-glycoprotein inhibitors, in: M.P. Rocha, F.F. Riverola, H. Shatkay, J.M.Corchado (Eds.), Advances in Bioinformatics, Springer, Heidelberg, 2010,pp. 175–180.

[38] M. Sasaki, M. Okamura, A. Ideo, J. Shimada, F. Suzuki, M. Ishihara, H. Kikuchi, Y.Kanda, S. Kunii, H. Sakagami, Re-evaluation of tumor-specific cytotoxicity of mi-tomycin C, bleomycin and peplomycin, Anticancer. Res. 26 (2006) 3373–3380.

[39] L. Jain, E.R. Gardner, W.D. Figg, M.S. Chernick, H.H. Kong, Lack of association be-tween excretion of sorafenib in sweat and hand–foot skin reaction, Pharmaco-therapy 30 (2010) 52–56.

[40] V.B. Jadhav, Y.J. Jun, J.H. Song, M.K. Park, J.H. Oh, S.W. Chae, I.S. Kim, S.J. Choi, H.J.Lee, Y.S. Sohn, A novel micelle-encapsulated platinum(II) anticancer agent, J.Control. Release 147 (2010) 144–150.

[41] A. Robertazzi, J.A. Platts, Hydrogen bonding, solvation, and hydrolysis of cisplat-in: a theoretical study, J. Comput. Chem. 25 (2004) 1060–1067.

[42] W.C. Yen, W.W. Lamph, The selective retinoid X receptor agonist bexarotene(LGD1069, Targretin) prevents and overcomes multidrug resistance in advancedbreast carcinoma, Mol. Cancer Ther. 4 (2005) 824–834.

[43] V. Reichelova, J. Liliemark, F. Albertioni, Structure–activity relationships of2-Chloro-2-arabino-fluoro-2-deoxyadenosine and related analogues: proteinbinding, lipophilicity, and retention in reversed-phase LC, J. Liq. Chromatogr.Relat. Technol. 18 (1995) 1123–1135.

[44] H.S. Friedman, O.M. Colvin, S.M. Ludeman, S.C. Schold Jr., V.L. Boyd, L.H.Mulhbaier, D.D. Bigner, Experimental chemotherapy of human medulloblasto-ma with classical alkylators, Cancer Res. 46 (1986) 2827–2833.

[45] J.S. Lagas, R.A. van Waterschoot, R.W. Sparidans, E. Wagenaar, J.H. Beijnen, A.H.Schinkel, Breast cancer resistance protein and P-glycoprotein limit sorafenibbrain accumulation, Mol. Cancer Ther. 9 (2010) 319–326.

[46] A.Haouala, N.Widmer,M.Montemurro, T. Buclin, L. Decosterd, Cardiovascular druginteractions with tyrosine kinase inhibitors, Cardiovasc. Med. 13 (2010) 147–154.

[47] M.M. Gottesman, T. Fojo, S.E. Bates, Multidrug resistance in cancer: role ofATP-dependent transporters, Nat. Rev. Cancer 2 (2002) 48–58.

[48] P. Dong, Z. Fang, Y. Zhang, G. Ge, Y. Mao, L. Zhu, Y. Qu, W. Li, L. Wang, C. Liu,Substrate-dependent modulation of the catalytic activity of CYP3A by erlotinib,Acta Pharmacol. Sin. 32 (2011) 399–407.

[49] M. Jamal-Hanjani, J. Spicer, Epidermal growth factor receptor tyrosine kinase in-hibitors in the treatment of epidermal growth factor receptor-mutant non-smallcell lung cancer metastatic to the brain, Clin. Cancer Res. 18 (2012) 938–944.

[50] P.B. Reddy, D.V. Paul, S.K. Agrawal, A.K. Saxena, H.M. Kumar, G.N. Qazi, Design,synthesis, and biological testing of 4beta-[(4-substituted)-1,2,3-triazol-1-yl]podophyllotoxin analogues as antitumor agents, Arch. Pharm. (Weinheim)341 (2008) 126–131.

[51] R.C. Coombes, L.S. Kilburn, C.F. Snowdon, R. Paridaens, R.E. Coleman, S.E. Jones, J.Jassem, C.J.H. van de Velde, T. Delozier, I. Alvarez, L. Del Mastro, O. Ortmann, K.Diedrich, A.S. Coates, E. Bajetta, S.B. Holmberg, D. Dodwell, E. Mickiewicz, J.Andersen, P.E. Lønning, G. Cocconi, J. Forbes, M. Castiglione, N. Stuart, A.Stewart, L.J. Fallowfield, G. Bertelli, E. Hall, R.G. Bogle, M. Carpentieri, E.Colajori, M. Subar, E. Ireland, J.M. Bliss, Survival and safety of exemestane versustamoxifen after 2–3 years' tamoxifen treatment (Intergroup ExemestaneStudy): a randomised controlled trial, Lancet 369 (2007) 559–570.

[52] Y. Javadzadeh, J. Shokri, S. Hallaj-Nezhadi, H. Hamishehkar, A. Nokhodchi, Enhance-ment of percutaneous absorption of finasteride by cosolvents, cosurfactant andsurfactants, Pharm. Dev. Technol. 15 (2010) 619–625.

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

[53] S. Hofer, K. Frei, H.P. Rutz, Gefitinib accumulation in glioblastoma tissue, CancerBiol. Ther. 5 (2006) 483–484.

[54] T. Kitazaki, M. Oka, Y. Nakamura, J. Tsurutani, S. Doi, M. Yasunaga, M. Takemura,H. Yabuuchi, H. Soda, S. Kohno, Gefitinib, an EGFR tyrosine kinase inhibitor,directly inhibits the function of P-glycoprotein in multidrug resistant cancercells, Lung Cancer 49 (2005) 337–343.

[55] S. Syvanen, J. Barletta, G. Blomquist, B. Langstrom, M. Bergstrom, PET-evaluatedtransport of [11C] hydroxyurea across the rat blood–brain barrier-lack of influ-ence of cyclosporin and probenecid, Drug Metab. Lett. 1 (2007) 189–194.

[56] S. So3H, O. Oh, O.H.N. Nn, O. Nos, Sulfonic acids, in: F.Z. Dorwald (Ed.), Lead Op-timization for Medicinal Chemists: Pharmacokinetic Properties of FunctionalGroups and Organic Compounds, Wiley VCH Verlag & Co. KGaA, Weinheim,Germany, 2012, pp. 203–205.

[57] P.R. Wielinga, H.V. Westerhoff, J. Lankelma, The relative importance of passiveand P-glycoprotein mediated anthracycline efflux from multidrug-resistantcells, Eur. J. Biochem. 267 (2000) 649–657.

[58] T. Wagner, Ifosfamide clinical pharmacokinetics, Clin. Pharmacokinet. 26 (1994)439–456.

[59] L.M. Allen, P.J. Creaven, R.L. Nelson, Studies on the human pharmacokinetics ofisophosphamide (NSC-109724), Cancer Treat. Rep. 60 (1976) 451–458.

[60] B. Peng, P. Lloyd, H. Schran, Clinical pharmacokinetics of imatinib, Clin.Pharmacokinet. 44 (2005) 879–894.

[61] G.G. Chabot, Clinical pharmacokinetics of irinotecan, Clin. Pharmacokinet. 33(1997) 245–259.

[62] T. Bansal, G. Mishra, M. Jaggi, R.K. Khar, S. Talegaonkar, Effect of P-glycoproteininhibitor, verapamil, on oral bioavailability and pharmacokinetics of irinotecanin rats, Eur. J. Pharm. Sci. 36 (2009) 580–590.

[63] P. Ballard, B.C. Barlaam, R.H. Bradbury, A. Dishington, L.F.A. Hennequin, D.M.Hickinson, I.M. Hollingsworth, J.G. Kettle, T. Klinowska, D.J. Ogilvie, Neutral5-substituted 4-anilinoquinazolines as potent, orally active inhibitors of erbB2receptor tyrosine kinase, Bioorg. Med. Chem. Lett. 17 (2007) 6326–6329.

[64] D.A. Smith, K.M. Koch, N. Arya, C.J. Bowen, J.M. Herendeen, A. Beelen, Effects ofketoconazole and carbamazepine on lapatinib pharmacokinetics in healthy sub-jects, Br. J. Clin. Pharmacol. 67 (2009) 421–426.

[65] P. Paixão, L.F. Gouveia, J.A.G. Morais, Prediction of the human oral bioavailabilityby using in vitro and in silico drug related parameters in a physiologically basedabsorption model, Int. J. Pharm. 429 (2012) 84–98.

[66] E.J. Wang, W.W. Johnson, The farnesyl protein transferase inhibitor lonafarnib(SCH66336) is an inhibitor of multidrug resistance proteins 1 and 2, Chemother-apy 49 (2003) 303–308.

[67] H.S. Friedman, S.C. Schold Jr., D.D. Bigner, Chemotherapy of subcutaneous andintracranial human medulloblastoma xenografts in athymic nude mice, CancerRes. 46 (1986) 224–228.

[68] J.D. Allen, R.F. Brinkhuis, L. van Deemter, J. Wijnholds, A.H. Schinkel, Extensivecontribution of the multidrug transporters P-glycoprotein and Mrp1 to basaldrug resistance, Cancer Res. 60 (2000) 5761–5766.

[69] M.C. Dubinsky, Azathioprine, 6-mercaptopurine in inflammatory bowel disease:pharmacology, efficacy, and safety, Clin. Gastroenterol. Hepatol. 2 (2004) 731–743.

[70] G. Yousefi, S.M. Foroutan, A. Zarghi, A. Shafaati, Synthesis and characterization ofmethotrexate polyethylene glycol esters as a drug delivery system, Chem.Pharm. Bull. (Tokyo) 58 (2010) 147–153.

[71] Y. Shibayama, Y. Takeda, K. Yamada, Effect of methotrexate treatment on ex-pression levels of organic anion transporter polypeptide 2, P-glycoprotein andbile salt export pump in rats, Biol. Pharm. Bull. 32 (2009) 493–496.

[72] M.A. Ihnat, A.M. Nervi, S.P. Anthony, R.C. Kaltreider, A.J. Warren, C.A. Pesce, S.A.Davis, J.P. Lariviere, J.W. Hamilton, Effects of mitomycin C and carboplatin pre-treatment on multidrug resistance-associated P-glycoprotein expression andon subsequent suppression of tumor growth by doxorubicin and paclitaxel inhuman metastatic breast cancer xenografted nude mice, Oncol. Res. 11 (1999)303–310.

[73] A. Decensi, R. Torrisi, V. Fontana, Stimulation of erythropoiesis by the non-steroidalanti-androgen nilutamide in men with prostate cancer: evidence for an agonisticeffect? Br. J. Cancer 69 (1994) 617–619.

[74] J. Graham, M. Muhsin, P. Kirkpatrick, Oxaliplatin, Nat. Rev. Drug Discov. 3 (2004)11–12.

[75] T.-B. Lee, J.-H. Park, Y.-D.Min, K.-J. Kim, C.-H. Choi, Epigeneticmechanisms involvedin differential MDR1mRNA expression between gastric and colon cancer cell linesand rationales for clinical chemotherapy, BMC Gastroenterol. 8 (2008) 33.

[76] J.A. Beisler, G.W. Peng, J.S. Driscoll, Potential antitumor agents: procarbazine an-alogs and other methylhydrazine derivatives, J. Pharm. Sci. 66 (1977) 849–852.

[77] R.W. Lee, D.B. Shenoy, R. Sheel, Micellar nanoparticles: applications for topicaland passive transdermal drug delivery, in: V.S. Kulkarni (Ed.), Handbook ofNon-invasive Drug Delivery System, Elsevier Inc., Burlington, 2010, pp. 37–58.

[78] J.H. Chang, C.J. Kochansky, M. Shou, The role of P-glycoprotein in thebioactivation of raloxifene, Drug Metab. Dispos. 34 (2006) 2073–2078.

[79] A.F. El-Kattan, C.S. Asbill, N. Kim, B.B. Michniak, The effects of terpene enhancerson the percutaneous permeation of drugs with different lipophilicities, Int. J.Pharm. 215 (2001) 229–240.

[80] D. Iusuf, S.F. Teunissen, E. Wagenaar, H. Rosing, J.H. Beijnen, A.H. Schinkel, P-Glycoprotein (ABCB1) transports the primary active tamoxifenmetabolites endoxifenand 4-hydroxytamoxifen and restricts their brain penetration, J. Pharmacol. Exp. Ther.337 (2011) 710–717.

[81] B.F. Goemans, C.M. Zwaan, A. Harlow, A.H. Loonen, B.E. Gibson, K. Hahlen, D.Reinhardt, U. Creutzig, M.C. Heinrich, G.J. Kaspers, In vitro profiling of the sensi-tivity of pediatric leukemia cells to tipifarnib: identification of T-cell ALL and FABM5 AML as the most sensitive subsets, Blood 106 (2005) 3532–3537.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 22: Oral delivery of anticancer drugs: Challenges and opportunities

T

18251826182718281829183018311832183318341835183618371838183918401841184218431844184518461847184818491850185118521853185418551856185718581859186018611862186318641865186618671868186918701871187218731874187518761877187818791880188118821883188418851886188718881889189018911892189318941895189618971898189919001901190219031904190519061907190819091910

19111912191319141915191619171918191919201921192219231924192519261927192819291930193119321933193419351936193719381939194019411942194319441945194619471948194919501951195219531954195519561957195819591960196119621963196419651966196719681969197019711972197319741975197619771978197919801981198219831984198519861987198819891990199119921993199419951996

22 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

[82] V.M. Herben, H.W.W. ten Bokkel, J.H. Beijnen, Clinical pharmacokinetics oftopotecan, Clin. Pharmacokinet. 31 (1996) 85–102.

[83] J. Shen, A.M. Carcaboso, K.E. Hubbard, M. Tagen, H.G. Wynn, J.C. Panetta, C.M.Waters, M.A. Elmeliegy, C.F. Stewart, Compartment-specific roles of ATP-binding cassette transporters define differential topotecan distribution in brainparenchyma and cerebrospinal fluid, Cancer Res. 69 (2009) 5885–5892.

[84] A. Martinsen, J. Gynther, Retention behaviour of triphenylethylene derivativesin reverse phase liquid chromatography, J. Liq. Chromatogr. Relat. Technol. 19(1996) 2117–2128.

[85] B. El Hafny, N. Cano, M. Piciotti, A. Regina, J.M. Scherrmann, F. Roux, Role ofP-glycoprotein in colchicine and vinblastine cellular kinetics in an immortalizedrat brain microvessel endothelial cell line, Biochem. Pharmacol. 53 (1997)1735–1742.

[86] I.V. Zhigaltsev, N. Maurer, Q.F. Akhong, R. Leone, E. Leng, J. Wang, S.C. Semple,P.R. Cullis, Liposome-encapsulated vincristine, vinblastine and vinorelbine: acomparative study of drug loading and retention, J. Control. Release 104(2005) 103–111.

[87] D.J. Adams, V.C. Knick, P-glycoprotein mediated resistance to 5′-nor-anhydro-vinblastine (Navelbine), Invest. New Drugs 13 (1995) 13–21.

[88] E. Kantharaj, R. Jayaraman, Histone deacetylase inhibitors as therapeutic agentsfor cancer therapy: drug metabolism and pharmacokinetic properties, in: C.Rundfeldt (Ed.), Drug Development — A Case Study Based Insight into ModernStrategies, InTech, Rijeka, Croatia, 2011.

[89] ADMET Predictor, Simulations Plus Inc., 10th Street West Lancaster, CA, USA,2012.

[90] Z. Bikadi, I. Hazai, D. Malik, K. Jemnitz, Z. Veres, P. Hari, Z. Ni, T.W. Loo, D.M.Clarke, E. Hazai, Q. Mao, Predicting P-glycoprotein-mediated drug transportbased on support vector machine and three-dimensional crystal structure ofP-glycoprotein, PLoS One 6 (2011) e25815.

[91] I.L.O. Buxton, Pharmacokinetics and pharmacodynamics: the dynamics of drugabsorption, distribution, action and elimination, in: L.L. Brunton, J.S. Lazo, K.L.Parker (Eds.), Goodman & Gilman's The Pharmacological Basis of Therapeutics,McGraw-Hill Publishers, New York, 2006, pp. 1–40.

[92] J. Hunter, B.H. Hirst, Intestinal secretion of drugs: the role of p-glycoprotein andrelated drug efflux systems in limiting oral drug absorption, Adv. Drug Deliv.Rev. 25 (1997) 129–157.

[93] P. Breedveld, J.H. Beijnen, J.H.M. Schellens, Use of P-glycoprotein and BCRP in-hibitors to improve oral bioavailability and CNS penetration of anticancerdrugs, Trends Pharmacol. Sci. 27 (2006) 17–24.

[94] M.E. Morris, Y.A. Gandhi, ABC transporters in intestinal and liver efflux, in: M.Hu, X. Li (Eds.), Oral Bioavailability: Basic Principles, Advanced Concepts, andApplications, John Wiley & Sons, New Jersey, 2011, pp. 381–400.

[95] S.V. Ambudkar, S. Dey, C.A. Hrycyna, M. Ramachandra, I. Pastan, M.M.Gottesman, Biochemical, cellular, and pharmacological aspects of the multidrugtransporter, Annu. Rev. Pharmacol. Toxicol. 39 (1999) 361–398.

[96] M.F. Fromm, Importance of P-glycoprotein at blood–tissue barriers, TrendsPharmacol. Sci. 25 (2004) 423–429.

[97] S.V. Ambudkar, M. Ramachandra, C.O. Cardarelli, I. Pastan, M.M. Gottesman,Modulation of human P-glycoprotein ATPase activity by interaction betweenoverlapping substrate-binding sites, Proc. Annu. Meet., Am. Assoc. Cancer Res.,1996, p. 325.

[98] M.V.S. Varma, Y. Ashokraj, C.S. Dey, R. Panchagnula, P-Glycoprotein inhibitors andtheir screening: a perspective from bioavailability enhancement, Pharmacol. Res.48 (2003) 347–359.

[99] Z.E. Sauna, M.M. Smith, M. Müller, K.M. Kerr, S.V. Ambudkar, The mechanism ofaction of multidrug-resistance-linked P-glycoprotein, J. Bioenerg. Biomembr. 33(2001) 481–491.

[100] A. Sparreboom, J. van Asperen, U. Mayer, A.H. Schinkel, J.W. Smit, D.K. Meijer, P.Borst, W.J. Nooijen, J.H. Beijnen, O. van Tellingen, Limited oral bioavailability andactive epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein in theintestine, Proc. Natl. Acad. Sci. U. S. A. 94 (1997) 2031–2035.

[101] C.Y. Wu, L.Z. Benet, M.F. Hebert, S.K. Gupta, M. Rowland, D.Y. Gomez, V.J.Wacher, Differentiation of absorption and first-pass gut and hepatic metabolismin humans: studies with cyclosporine, Clin. Pharmacol. Ther. 58 (1995) 492–497.

[102] B.N. Singh, K.H. Kim, Drug delivery: oral route, in: J. Swarbrick (Ed.), Encyclope-dia of Pharmaceutical Technology, Informa Healthcare, New York, 2007,pp. 1242–1265.

[103] L. Barthe, J. Woodley, G. Houin, Gastrointestinal absorption of drugs: methodsand studies, Fundam. Clin. Pharm. 13 (1999) 154–168.

[104] K.E. Thummel, K.L. Kunze, D.D. Shen, Enzyme-catalyzed processes of first-passhepatic and intestinal drug extraction, Adv. Drug Deliv. Rev. 27 (1997) 99–127.

[105] T. Tabata, M. Katoh, S. Tokudome, M. Nakajima, T. Yokoi, Identification of the cy-tosolic carboxylesterase catalyzing the 5′-deoxy-5-fluorocytidine formationfrom capecitabine in human liver, Drug Metab. Dispos. 32 (2004) 1103–1110.

[106] F.J. Gonzalez, R.H. Tukey, Drug metabolism, in: L.L. Brunton, J.S. Lazo, K.L. Parker(Eds.), Goodman & Gilman's The Pharmacological Basis of Therapeutics,McGraw-Hill Publishers, New York, 2006, pp. 71–92.

[107] S. Mouly, M.F. Paine, P-Glycoprotein increases from proximal to distal regions ofhuman small intestine, Pharm. Res. 20 (2003) 1595–1599.

[108] T. Bansal, N. Akhtar, M. Jaggi, R.K. Khar, S. Talegaonkar, Novel formulation ap-proaches for optimising delivery of anticancer drugs based on P-glycoproteinmodulation, Drug Discov. Today 14 (2009) 1067–1074.

[109] T.J.M. Meerum, M.M. Malingré, J.H. Beijnen, H.W.W. ten Bokkel, H. Rosing, F.J.Koopman, O. van Tellingen, M. Swart, J.H. Schellens, Coadministration of oral cy-closporin A enables oral therapy with paclitaxel, Clin. Cancer Res. 5 (1999)3379–3384.

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

[110] M.M. Malingré, D.J. Richel, J.H. Beijnen, H. Rosing, F.J. Koopman, W.W.T.B.Huinink, M.E. Schot, J.H.M. Schellens, Coadministration of cyclosporine stronglyenhances the oral bioavailability of docetaxel, J. Clin. Oncol. 19 (2001)1160–1166.

[111] M. Werle, Natural and synthetic polymers as inhibitors of drug efflux pumps,Pharm. Res. 25 (2008) 500–511.

[112] B. Carreno-Gomez, R. Duncan, Compositions with enhanced oral bioavailability,US Patent App. 20030211072 A1 filed on January 16, 2001.

[113] Y.L. Lo, C.Y. Hsu, J.D. Huang, Comparison of effects of surfactants with other MDRreversing agents on intracellular uptake of epirubicin in Caco-2 cell line, Anti-cancer. Res. 18 (1998) 3005–3009.

[114] C. Chen, J. Zhou, C. Ji, Quercetin: a potential drug to reverse multidrug resistance,Life Sci. 87 (2010) 333–338.

[115] J.S. Choi, B.W. Jo, Y.C. Kim, Enhanced paclitaxel bioavailability after oral admin-istration of paclitaxel or prodrug to rats pretreated with quercetin, Eur. J. Pharm.Biopharm. 57 (2004) 313–318.

[116] J.S. Choi, Y.J. Piao, K.W. Kang, Effects of quercetin on the bioavailability of doxo-rubicin in rats: role of CYP3A4 and P-gp inhibition by quercetin, Arch. Pharm.Res. 34 (2011) 607–613.

[117] X. Li, J.S. Choi, Effects of quercetin on the pharmacokinetics of etoposide afteroral or intravenous administration of etoposide in rats, Anticancer. Res. 29(2009) 1411–1415.

[118] V.D. Makhey, A. Guo, D.A. Norris, P. Hu, J. Yan, P.J. Sinko, Characterization of theregional intestinal kinetics of drug efflux in rat and human intestine and inCaco-2 cells, Pharm. Res. 15 (1998) 1160–1167.

[119] B.M. Johnson, W.N. Charman, C.J.H. Porter, An in vitro examination of the impactof polyethylene glycol 400, pluronic P85, and vitamin E da-tocopheryl polyeth-ylene glycol 1000 succinate on p-glycoprotein efflux and enterocyte-basedmetabolism in excised rat intestine, AAPS J. 4 (2002) 193–205.

[120] E.D. Hugger, K.L. Audus, R.T. Borchardt, Effects of poly(ethylene glycol) on effluxtransporter activity in Caco-2 cell monolayers, J. Pharm. Sci. 91 (2002) 1980–1990.

[121] Q. Shen, Y. Lin, T. Handa, M. Doi, M. Sugie, K. Wakayama, N. Okada, T. Fujita, A.Yamamoto, Modulation of intestinal P-glycoprotein function by polyethyleneglycols and their derivatives by in vitro transport and in situ absorption studies,Int. J. Pharm. 313 (2006) 49–56.

[122] J.D.R. Schulze, W.A. Waddington, P.J. Ell, G.E. Parsons, M.D. Coffin, A.W. Basit,Concentration-dependent effects of polyethylene glycol 400 on gastrointestinaltransit and drug absorption, Pharm. Res. 20 (2003) 1984–1988.

[123] D.A.I. Ashiru-Oredope, N. Patel, B. Forbes, R. Patel, A.W. Basit, The effect ofpolyoxyethylene polymers on the transport of ranitidine in Caco-2 cell mono-layers, Int. J. Pharm. 409 (2011) 164–168.

[124] J.S. Choi, B.W. Jo, Enhanced paclitaxel bioavailability after oral administration ofpegylated paclitaxel prodrug for oral delivery in rats, Int. J. Pharm. 280 (2004)221–227.

[125] J. Wang, H. Qu, L. Jin, W. Zeng, L. Qin, F. Zhang, X. Wei, W. Lu, C. Zhang, W. Liang,Pegylated phosphotidylethanolamine inhibiting P-glycoprotein expression andenhancing retention of doxorubicin in MCF7/ADR cells, J. Pharm. Sci. 100(2011) 2267–2277.

[126] J. Iqbal, J. Hombach, B. Matuszczak, A. Bernkop-Schnürch, Design and in vitroevaluation of a novel polymeric P-glycoprotein (P-gp) inhibitor, J. Control. Re-lease 147 (2010) 62–69.

[127] M. Greindl, F. Föger, J. Hombach, A. Bernkop-Schnürch, In vivo evaluation ofthiolated poly(acrylic acid) as a drug absorption modulator for MRP2 effluxpump substrates, Eur. J. Pharm. Biopharm. 72 (2009) 561–566.

[128] J. Iqbal, D. Sakloetsakun, A. Bernkop-Schnürch, Thiomers: inhibition of cyto-chrome P450 activity, Eur. J. Pharm. Biopharm. 78 (2011) 361–365.

[129] N. Langoth, H. Kahlbacher, G. Schöffmann, I. Schmerold, M. Schuh, S. Franz, P.Kurka, A. Bernkop-Schnürch, Thiolated chitosans: design and in vivo evaluationof a mucoadhesive buccal peptide drug delivery system, Pharm. Res. 23 (2006)573–579.

[130] M.K. Al-Shawi, I.L. Urbatsch, A.E. Senior, Covalent inhibitors of P-glycoproteinATPase activity, J. Biol. Chem. 269 (1994) 8986–8992.

[131] M. Werle, M. Hoffer, Glutathione and thiolated chitosan inhibit multidrug resis-tance P-glycoprotein activity in excised small intestine, J. Control. Release 111(2006) 41–46.

[132] A.E. Senior, P. Gros, I.L. Urbatsch, Residues in p-glycoprotein catalytic sites thatreact with the inhibitor 7-chloro-4-nitrobenzo-2-oxa-1,3-diazole, Arch. Biochem.Biophys. 357 (1998) 121–125.

[133] R. Liu, F.J. Sharom, Site-directed fluorescence labeling of P-glycoprotein on cys-teine residues in the nucleotide binding domains, Biochemistry 35 (1996)11865–11873.

[134] T.W. Loo, D.M. Clarke, Membrane topology of a cysteine-less mutant of humanP-glycoprotein, J. Biol. Chem. 270 (1995) 843–848.

[135] G. Borchard, H.L. Lueβen, A.G. de Boer, J. Verhoef, C.M. Lehr, H.E. Junginger, Thepotential of mucoadhesive polymers in enhancing intestinal peptide drug ab-sorption. III: effects of chitosan–glutamate and carbomer on epithelial tightjunctions in vitro, J. Control. Release 39 (1996) 131–138.

[136] M.E. Imam, M. Hornof, C. Valenta, G. Reznicek, A. Bernkop-Schnürch, Evidencefor the interpenetration of mucoadhesive polymers into the mucous gel layer,STP Pharma Sci. 13 (2003) 171–176.

[137] J.M. Staddon, K. Herrenknecht, C. Smales, L.L. Rubin, Evidence that tyrosinephosphorylation may increase tight junction permeability, J. Cell Sci. 108(1995) 609–619.

[138] F. Foger, S. Malaivijitnond, T. Wannaprasert, C. Huck, A. Bernkop-Schnurch, M.Werle, Effect of a thiolated polymer on oral paclitaxel absorption and tumorgrowth in rats, J. Drug Target. 16 (2008) 149–155.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 23: Oral delivery of anticancer drugs: Challenges and opportunities

T

19971998199920002001200220032004200520062007200820092010201120122013201420152016201720182019202020212022202320242025202620272028202920302031203220332034203520362037203820392040204120422043204420452046204720482049205020512052205320542055205620572058205920602061206220632064206520662067206820692070207120722073207420752076207720782079208020812082

20832084208520862087208820892090209120922093209420952096209720982099210021012102210321042105210621072108210921102111211221132114211521162117211821192120212121222123212421252126212721282129213021312132213321342135213621372138213921402141214221432144214521462147214821492150215121522153215421552156215721582159216021612162216321642165216621672168

23K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

[139] A. Bernkop-Schnurch, C.E. Kast, D. Guggi, Permeation enhancing polymers inoral delivery of hydrophilic macromolecules: thiomer/GSH systems, J. Control.Release 93 (2003) 95–103.

[140] A. Bernkop-Schnürch, C.E. Kast, Chemically modified chitosans as enzyme inhibi-tors, Adv. Drug Deliv. Rev. 52 (2001) 127–137.

[141] A.E. Clausen, C.E. Kast, A. Bernkop-Schnürch, The role of glutathione in the perme-ation enhancing effect of thiolated polymers, Pharm. Res. 19 (2002) 602–608.

[142] W.C. Barrett, J.P. DeGnore, S. König, H.M. Fales, Y.F. Keng, Z.Y. Zhang, M.B. Yim,P.B. Chock, Regulation of PTP1B via glutathionylation of the active site cysteine215, Biochemistry 38 (1999) 6699–6705.

[143] T. Schmitz, J. Hombach, A. Bernkop-Schnürch, Chitosan-N-acetyl cysteine conjugates:in vitro evaluation of permeation enhancing and P-glycoprotein inhibiting properties,Drug Deliv. 15 (2008) 245–252.

[144] B.D. Rege, J.P.Y. Kao, J.E. Polli, Effects of nonionic surfactants on membrane trans-porters in Caco-2 cell monolayers, Eur. J. Pharm. Sci. 16 (2002) 237–246.

[145] E.M. Collnot, C. Baldes, M.F. Wempe, J. Hyatt, L. Navarro, K.J. Edgar, U.F. Schaefer,C.M. Lehr, Influence of vitamin E TPGS poly(ethylene glycol) chain lengthon apical efflux transporters in Caco-2 cell monolayers, J. Control. Release 111(2006) 35–40.

[146] T. Yamagata, H. Kusuhara, M. Morishita, K. Takayama, H. Benameur, Y. Sugiyama,Effect of excipients on breast cancer resistance protein substrate uptake activity,J. Control. Release 124 (2007) 1–5.

[147] E. Friche, P.B. Jensen, M. Sehested, E.J. Demant, N.N. Nissen, The solventscremophor EL and tween 80 modulate daunorubicin resistance in the multidrugresistant Ehrlich ascites tumor, Cancer Commun. 2 (1990) 297–303.

[148] Y. Lo, Relationships between the hydrophilic–lipophilic balance values of phar-maceutical excipients and their multidrug resistance modulating effect inCaco-2 cells and rat intestines, J. Control. Release 90 (2003) 37–48.

[149] F. Foger, H. Hoyer, K. Kafedjiiski, M. Thaurer, A. Bernkop-Schnurch, In vivo com-parison of various polymeric and low molecular mass inhibitors of intestinalp-glycoprotein, Biomaterials 27 (2006) 5855–5860.

[150] Y.M. Yin, F.D. Cui, C.F. Mu, M.K. Choi, J.S. Kim, S.J. Chung, C.K. Shim, D.D. Kim,Docetaxel microemulsion for enhanced oral bioavailability: preparation and invitro and in vivo evaluation, J. Control. Release 140 (2009) 86–94.

[151] X. Sha, G. Yan, Y. Wu, J. Li, X. Fang, Effect of self-microemulsifying drug deliverysystems containing Labrasol on tight junctions in Caco-2 cells, Eur. J. Pharm. Sci.24 (2005) 477–486.

[152] E.V. Batrakova, S. Li, Y. Li, V.Y. Alakhov, A.V. Kabanov, Effect of pluronic P85 onATPase activity of drug efflux transporters, Pharm. Res. 21 (2004) 2226–2233.

[153] V. Alakhov, G. Pietrzynski, K. Patel, A. Kabanov, L. Bromberg, T.A. Hatton,Pluronic block copolymers and pluronic poly(acrylic acid) microgels in oral de-livery of megestrol acetate, J. Pharm. Pharmacol. 56 (2004) 1233–1241.

[154] E.M. Collnot, C. Baldes, M.F. Wempe, R. Kappl, J. Hüttermann, J.A. Hyatt, K.J.Edgar, U.F. Schaefer, C.M. Lehr, Mechanism of inhibition of P-glycoprotein medi-ated efflux by vitamin E TPGS: influence on ATPase activity and membrane flu-idity, Mol. Pharm. 4 (2007) 465–474.

[155] M.V.S. Varma, R. Panchagnula, Enhanced oral paclitaxel absorption with vitaminE-TPGS: effect on solubility and permeability in vitro, in situ and in vivo, Eur. J.Pharm. Sci. 25 (2005) 445–453.

[156] A. Garrigues, A.E. Escargueil, S. Orlowski, Themultidrug transporter, P-glycoprotein,actively mediates cholesterol redistribution in the cell membrane, Proc. Natl. Acad.Sci. U. S. A. 99 (2002) 10347–10352.

[157] M.E. Davis, M.E. Brewster, Cyclodextrin-based pharmaceutics: past, present andfuture, Nat. Rev. Drug Discov. 3 (2004) 1023–1035.

[158] E.P. Kilsdonk, P.G. Yancey, G.W. Stoudt, F.W. Bangerter, W.J. Johnson, M.C.Phillips, G.H. Rothblat, Cellular cholesterol efflux mediated by cyclodextrins,J. Biol. Chem. 270 (1995) 17250–17256.

[159] K. Yunomae, H. Arima, F. Hirayama, K. Uekama, Involvement of cholesterol inthe inhibitory effect of dimethyl-[beta]-cyclodextrin on P-glycoprotein andMRP2 function in Caco-2 cells, FEBS Lett. 536 (2003) 225–231.

[160] A.K. Upadhyay, S. Singh, R.R. Chhipa, M.V. Vijayakumar, A.K. Ajay, M.K. Bhat,Methyl-beta-cyclodextrin enhances the susceptibility of human breast cancercells to carboplatin and 5-fluorouracil: involvement of Akt, NF-kappaB andBcl-2, Toxicol. Appl. Pharmacol. 216 (2006) 177–185.

[161] P.Y. Grosse, F. Bressolle, F. Pinguet, Methyl-β-cyclodextrin in HL-60 parental andmultidrug-resistant cancer cell lines: effect on the cytotoxic activity and intra-cellular accumulation of doxorubicin, Cancer Chemother. Pharmacol. 40(1997) 489–494.

[162] P.Y. Grosse, F. Bressolle, F. Pinguet, Antiproliferative effect of methyl-beta-cyclodextrin in vitro and in human tumour xenografted athymic nude mice,Br. J. Cancer 78 (1998) 1165–1169.

[163] S. Alcaro, C.A. Ventura, D. Paolino, D. Battaglia, F. Ortuso, L. Cattel, G. Puglisi, M.Fresta, Preparation, characterization, molecular modeling and in vitro activity ofpaclitaxel–cyclodextrin complexes, Bioorg.Med. Chem. Lett. 12 (2002) 1637–1641.

[164] M. Agueros, L. Ruiz-Gaton, C. Vauthier, K. Bouchemal, S. Espuelas, G. Ponchel,J.M. Irache, Combined hydroxypropyl-beta-cyclodextrin and poly(anhydride)nanoparticles improve the oral permeability of paclitaxel, Eur. J. Pharm. Sci. 38(2009) 405–413.

[165] H. Arima, K. Yunomae, F. Hirayama, K. Uekama, Contribution of P-glycoproteinto the enhancing effects of dimethyl-β-cyclodextrin on oral bioavailability oftacrolimus, J. Pharmacol. Exp. Ther. 297 (2001) 547–555.

[166] S.J. Torne, K.A. Ansari, P.R. Vavia, F. Trotta, R. Cavalli, Enhanced oral paclitaxelbioavailability after administration of paclitaxel-loaded nanosponges, DrugDeliv. 17 (2010) 419–425.

[167] B. Yavuz, E. Bilensoy, I. Vural, M. Sumnu, Alternative oral exemestane formulation:improved dissolution and permeation, Int. J. Pharm. 398 (2010) 137–145.

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

[168] R.W. van Leeuwen, D.H. Brundel, C. Neef, T. van Gelder, R.H. Mathijssen, D.M.Burger, F.G. Jansman, Prevalence of potential drug–drug interactions in cancerpatients treated with oral anticancer drugs, Br. J. Cancer 108 (2013) 1071–1078.

[169] E. Roger, F. Lagarce, E. Garcion, J.P. Benoit, Biopharmaceutical parameters to con-sider in order to alter the fate of nanocarriers after oral delivery, Nanomedicine5 (2010) 287–306.

[170] S.K. Lai, Y.Y. Wang, J. Hanes, Mucus-penetrating nanoparticles for drug and genedelivery to mucosal tissues, Adv. Drug Deliv. Rev. 61 (2009) 158–171.

[171] M.A. Deli, Potential use of tight junction modulators to reversibly open membra-nous barriers and improve drug delivery, Biochim. Biophys. Acta 1788 (2009)892–910.

[172] E. Roger, F. Lagarce, E. Garcion, J.P. Benoit, Lipid nanocarriers improve paclitaxeltransport throughout human intestinal epithelial cells by using vesicle-mediatedtranscytosis, J. Control. Release 140 (2009) 174–181.

[173] S. Jain, P.U. Valvi, N.K. Swarnakar, K. Thanki, Gelatin coated hybrid lipid nano-particles for oral delivery of amphotericin B, Mol. Pharm. 9 (2012) 2542–2553.

[174] H. Harde, M. Das, S. Jain, Solid lipid nanoparticles: an oral bioavailability enhanc-er vehicle, Expert Opin. Drug Deliv. 8 (2011) 1407–1424.

[175] R. Shegokar, R.H. Müller, Nanocrystals: industrially feasible multifunctional for-mulation technology for poorly soluble actives, Int. J. Pharm. 399 (2010)129–139.

[176] C. Jacobs, O. Kayser, R. Muller, Production and characterisation of mucoadhesivenanosuspensions for the formulation of bupravaquone, Int. J. Pharm. 214 (2001)3–7.

[177] P. Fasinu, V. Pillay, V.M.K. Ndesendo, L.C. du Toit, Y.E. Choonara, Diverse ap-proaches for the enhancement of oral drug bioavailability, Biopharm. DrugDispos. 32 (2011) 185–209.

[178] Y. Liu, L. Huang, F. Liu, Paclitaxel nanocrystals for overcoming multidrug resis-tance in cancer, Mol. Pharm. 7 (2010) 863–869.

[179] J. Deng, L. Huang, F. Liu, Understanding the structure and stability of paclitaxelnanocrystals, Int. J. Pharm. 390 (2010) 242–249.

[180] F. Liu, J.Y. Park, Y. Zhang, C. Conwell, Y. Liu, S.R. Bathula, L. Huang, Targeted can-cer therapy with novel high drug-loading nanocrystals, J. Pharm. Sci. 99 (2010)3542–3551.

[181] A. Shapira, I. Davidson, N. Oron, Y.G. Assaraf, Y.D. Livney, β-Casein nanoparticle-based oral drug delivery system for potential treatment of gastric carcinoma:stability, target-activated release and cytotoxicity, Eur. J. Pharm. Biopharm. 80(2012) 298–305.

[182] P.P. Lv, W. Wei, H. Yue, T.Y. Yang, L.Y. Wang, G.H. Ma, Porous quaternized chito-san nanoparticles containing paclitaxel nanocrystals improved therapeutic effi-cacy in non-small-cell lung cancer after oral administration, Biomacromolecules12 (2011) 4230–4239.

[183] M.R. Harrison, N.M. Hahn, R. Pili, W.K. Oh, H. Hammers, C. Sweeney, K. Kim, S.Perlman, J. Arnott, C. Sidor, G. Wilding, G. Liu, A phase II study of2-methoxyestradiol (2ME2) nano crystal dispersion (NCD) in patients withtaxane-refractory, metastatic castrate-resistant prostate cancer (CRPC), Invest.New Drugs 29 (2011) 1465–1474.

[184] L. Plapied, N. Duhem, A. des Rieux, V. Préat, Fate of polymeric nanocarriers fororal drug delivery, Curr. Opin. Colloid. 16 (2011) 228–237.

[185] J.H. Park, G. Saravanakumar, K. Kim, I.C. Kwon, Targeted delivery of low molec-ular drugs using chitosan and its derivatives, Adv. Drug Deliv. Rev. 62 (2010)28–41.

[186] A. Kumari, S.K. Yadav, S.C. Yadav, Biodegradable polymeric nanoparticles baseddrug delivery systems, Colloids Surf. B 75 (2010) 1–18.

[187] L. Brannon-Peppas, Recent advances on the use of biodegradable microparticlesand nanoparticles in controlled drug delivery, Int. J. Pharm. 116 (1995) 1–9.

[188] A.K. Jain, N.K. Swarnakar, M. Das, C. Godugu, R.P. Singh, P.R. Rao, S. Jain, Aug-mented anticancer efficacy of doxorubicin-loaded polymeric nanoparticlesafter oral administration in a breast cancer induced animal model, Mol. Pharm.8 (2011) 1140–1151.

[189] A.K. Jain, N.K. Swarnakar, C. Godugu, R.P. Singh, S. Jain, The effect of the oral ad-ministration of polymeric nanoparticles on the efficacy and toxicity of tamoxi-fen, Biomaterials 32 (2011) 503–515.

[190] N.K. Swarnakar, A.K. Jain, R.P. Singh, C. Godugu, M. Das, S. Jain, Oral bioavailability,therapeutic efficacy and reactive oxygen species scavenging properties of coen-zyme Q10-loaded polymeric nanoparticles, Biomaterials 32 (2011) 6860–6874.

[191] A. des Rieux, V. Fievez, M. Garinot, Y.J. Schneider, V. Preat, Nanoparticles aspotential oral delivery systems of proteins and vaccines: a mechanistic approach,J. Control. Release 116 (2006) 1–27.

[192] A.K. Jain, M. Das, N.K. Swarnakar, S. Jain, Engineered PLGA nanoparticles: anemerging delivery tool in cancer therapeutics, Crit. Rev. Ther. Drug CarrierSyst. 28 (2011) 1–45.

[193] J.V. Jokerst, T. Lobovkina, R.N. Zare, S.S. Gambhir, Nanoparticle PEGylation forimaging and therapy, Nanomedicine (Lond) 6 (2011) 715–728.

[194] K. Knop, R. Hoogenboom, D. Fischer, U.S. Schubert, Poly(ethylene glycol) in drugdelivery: pros and cons as well as potential alternatives, Angew. Chem. Int. Ed.Engl. 49 (2010) 6288–6308.

[195] J. Park, P.M. Fong, J. Lu, K.S. Russell, C.J. Booth, W.M. Saltzman, T.M. Fahmy,PEGylated PLGA nanoparticles for the improved delivery of doxorubicin,Nanomedicine 5 (2009) 410–418.

[196] V. Zabaleta, G. Ponchel, H. Salman, M. Agueros, C. Vauthier, J.M. Irache, Oral admin-istration of paclitaxel with pegylated poly(anhydride) nanoparticles: permeabilityand pharmacokinetic study, Eur. J. Pharm. Biopharm. 81 (2012) 514–523.

[197] L.E. van Vlerken, T.K. Vyas, M.M. Amiji, Poly(ethylene glycol)-modified nanocarriers for tumor-targeted and intracellular delivery, Pharm. Res. 24 (2007)1405–1414.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 24: Oral delivery of anticancer drugs: Challenges and opportunities

T

21692170217121722173217421752176217721782179218021812182218321842185218621872188218921902191219221932194219521962197219821992200220122022203220422052206220722082209221022112212221322142215221622172218221922202221222222232224222522262227222822292230223122322233223422352236223722382239224022412242224322442245224622472248224922502251225222532254

22552256225722582259226022612262226322642265226622672268226922702271227222732274227522762277227822792280228122822283228422852286228722882289229022912292229322942295229622972298229923002301230223032304230523062307230823092310231123122313231423152316231723182319232023212322232323242325232623272328232923302331233223332334233523362337233823392340

24 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

[198] S. Jain, V.V. Rathi, A.K. Jain, M. Das, C. Godugu, Folate-decorated PLGA nano-particles as a rationally designed vehicle for the oral delivery of insulin,Nanomedicine (Lond) 7 (2012) 1311–1337.

[199] M.A. Jepson, M.A. Clark, B.H. Hirst, M cell targeting by lectins: a strategy for mu-cosal vaccination and drug delivery, Adv. Drug Deliv. Rev. 56 (2004) 511–525.

[200] M. Garinot, V. Fiévez, V. Pourcelle, F. Stoffelbach, A. des Rieux, L. Plapied, I.Theate, H. Freichels, C. Jérôme, J. Marchand-Brynaert, PEGylated PLGA-basednanoparticles targeting M cells for oral vaccination, J. Control. Release 120(2007) 195–204.

[201] R.M. Samstein, K. Perica, F. Balderrama, M. Look, T.M. Fahmy, The use ofdeoxycholic acid to enhance the oral bioavailability of biodegradable nano-particles, Biomaterials 29 (2008) 703–708.

[202] N. Mishra, S. Tiwari, B. Vaidya, G.P. Agrawal, S.P. Vyas, Lectin anchored PLGAnanoparticles for oral mucosal immunization against hepatitis B, J. Drug Target.19 (2011) 67–78.

[203] H. Chen, V. Torchilin, R. Langer, Lectin-bearing polymerized liposomes as potentialoral vaccine carriers, Pharm. Res. 13 (1996) 1378–1383.

[204] M. Wirth, C. Kneuer, C.M. Lehr, F. Gabor, Lectin-mediated drug delivery: discrim-ination between cytoadhesion and cytoinvasion and evidence for lysosomalaccumulation of wheat germ agglutinin in the Caco-2 model, J. Drug Target. 10(2002) 439–448.

[205] W.J. Kim, J.W. Yockman, M. Lee, J.H. Jeong, Y.H. Kim, S.W. Kim, Soluble Flt-1 genedelivery using PEI-g-PEG-RGD conjugate for anti-angiogenesis, J. Control. Release106 (2005) 224–234.

[206] A. Dube, J.A. Nicolazzo, I. Larson, Chitosan nanoparticles enhance the plasmaexposure of (−)-epigallocatechin gallate in mice through an enhancement inintestinal stability, Eur. J. Pharm. Sci. 44 (2011) 422–426.

[207] B. Hu, Y. Ting, X. Yang, W. Tang, X. Zeng, Q. Huang, Nanochemoprevention by en-capsulation of (−)-epigallocatechin-3-gallate with bioactive peptides/chitosannanoparticles for enhancement of its bioavailability, Chem. Commun. 48(2012) 2421–2423.

[208] J. Iqbal, F. Sarti, G. Perera, A. Bernkop-Schnurch, Development and in vivo evalu-ation of an oral drug delivery system for paclitaxel, Biomaterials 32 (2011)170–175.

[209] L. Zhang, M. Sun, R. Guo, Z. Jiang, Y. Liu, X. Jiang, C. Yang, Chitosan surface-modified hydroxycamptothecin loaded nanoparticles with enhanced transportacross Caco-2 cell monolayer, J. Nanosci. Nanotechnol. 6 (2006) 2912–2920.

[210] M. Agüeros, V. Zabaleta, S. Espuelas, M. Campanero, J. Irache, Increased oral bio-availability of paclitaxel by its encapsulation through complex formation withcyclodextrins in poly(anhydride) nanoparticles, J. Control. Release 145 (2010)2–8.

[211] S. Saremi, F. Atyabi, S.P. Akhlaghi, S.N. Ostad, R. Dinarvand, Thiolated chitosannanoparticles for enhancing oral absorption of docetaxel: preparation, in vitroand ex vivo evaluation, Int. J. Nanomedicine 6 (2011) 119–128.

[212] S.S. Feng, L. Mei, P. Anitha, C.W. Gan, W. Zhou, Poly(lactide)–vitamin Ederivative/montmorillonite nanoparticle formulations for the oral delivery ofdocetaxel, Biomaterials 30 (2009) 3297–3306.

[213] Z. Zhang, S.S. Feng, Self-assembled nanoparticles of poly(lactide)–vitamin ETPGS copolymers for oral chemotherapy, Int. J. Pharm. 324 (2006) 191–198.

[214] H. Chen, Y. Zheng, G. Tian, Y. Tian, X. Zeng, G. Liu, K. Liu, L. Li, Z. Li, L. Mei, L.Huang, Oral delivery of DMAB-modified docetaxel loaded PLGA-TPGS nano-particles for cancer chemotherapy, Nanoscale Res. Lett. 6 (2011) 4–13.

[215] Y. Dong, S.S. Feng, Poly(D, L-lactide-co-glycolide)/montmorillonite nanoparticlesfor oral delivery of anticancer drugs, Biomaterials 26 (2005) 6068–6076.

[216] G. Ling, P. Zhang, W. Zhang, J. Sun, X. Meng, Y. Qin, Y. Deng, Z. He, Development ofnovel self-assembled DS-PLGAhybrid nanoparticles for improving oral bioavailabil-ity of vincristine sulfate by P-gp inhibition, J. Control. Release 148 (2010) 241–248.

[217] K.Y. Win, S.S. Feng, Effects of particle size and surface coating on cellular uptakeof polymeric nanoparticles for oral delivery of anticancer drugs, Biomaterials 26(2005) 2713–2722.

[218] Y.S. Yin, D.W. Chen, M.X. Qiao, Z. Lu, H.Y. Hu, Preparation and evaluation oflectin-conjugated PLGA nanoparticles for oral delivery of thymopentin, J. Control.Release 116 (2006) 337–345.

[219] C. Wang, P.C. Ho, L.Y. Lim, Wheat germ agglutinin-conjugated PLGA nanoparticlesfor enhanced intracellular delivery of paclitaxel to colon cancer cells, Int.J. Pharm. 400 (2010) 201–210.

[220] Y. Mo, L.Y. Lim, Preparation and in vitro anticancer activity of wheat germ agglu-tinin (WGA)-conjugated PLGA nanoparticles loaded with paclitaxel and isopro-pyl myristate, J. Control. Release 107 (2005) 30–42.

[221] S. Bisht, G. Feldmann, J.B. Koorstra, M. Mullendore, H. Alvarez, C. Karikari, M.A.Rudek, C.K. Lee, A. Maitra, In vivo characterization of a polymeric nanoparticleplatform with potential oral drug delivery capabilities, Mol. Cancer Ther. 7(2008) 3878–3888.

[222] X.M. Li, Y.L. Xu, G.G. Chen, P. Wei, Q.N. Ping, PLGA nanoparticles for the oral de-livery of 5-fluorouracil using high pressure homogenization–emulsification asthe preparation method and in vitro/in vivo studies, Drug Dev. Ind. Pharm. 34(2008) 107–115.

[223] Y.M. Tsai, W.C. Jan, C.F. Chien, W.C. Lee, L.C. Lin, T.H. Tsai, Optimized nano-formulation on the bioavailability of hydrophobic polyphenol, curcumin, infreely-moving rats, Food Chem. 127 (2011) 918–925.

[224] D.R. Kalaria, G. Sharma, V. Beniwal, M.N.V.R. Kumar, Design of biodegradablenanoparticles for oral delivery of doxorubicin: in vivo pharmacokinetics and tox-icity studies in rats, Pharm. Res. 26 (2009) 492–501.

[225] V. Bhardwaj, J.A. Plumb, J. Cassidy, M.N.V.R. Kumar, Evaluating the potential ofpolymer nanoparticles for oral delivery of paclitaxel in drug-resistant cancer,Cancer Nanotechnol. 1 (2010) 29–34.

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

[226] G. Gaucher, P. Satturwar, M.C. Jones, A. Furtos, J.C. Leroux, Polymeric micelles fororal drug delivery, Eur. J. Pharm. Biopharm. 76 (2010) 147–158.

[227] M.C. Jones, J.C. Leroux, Polymeric micelles—a new generation of colloidal drugcarriers, Eur. J. Pharm. Biopharm. 48 (1999) 101–111.

[228] J.E. Chung, M. Yokoyama, K. Suzuki, T. Aoyagi, Y. Sakurai, T. Okano, Reversiblythermo-responsive alkyl-terminated poly(N-isopropylacrylamide) core–shellmicellar structures, Colloids Surf. B 9 (1997) 37–48.

[229] W.Y. Chen, P. Alexandridis, C.K. Su, C.S. Patrickios, W.R. Hertler, T.A. Hatton, Ef-fect of block size and sequence on the micellization of ABC triblock methacrylicpolyampholytes, Macromolecules 28 (1995) 8604–8611.

[230] Y. Nagasaki, T. Okada, C. Scholz, M. Iijima, M. Kato, K. Kataoka, The reactive poly-meric micelle based on an aldehyde-ended poly(ethylene glycol)/poly(lactide)block copolymer, Macromolecules 31 (1998) 1473–1479.

[231] C. Scholz, M. Iijima, Y. Nagasaki, K. Kataoka, A novel reactive polymeric micellewith aldehyde groups on its surface, Macromolecules 28 (1995) 7295–7297.

[232] H.J. Yao, R.J. Ju, X.X. Wang, Y. Zhang, R.J. Li, Y. Yu, L. Zhang, W.L. Lu, The antitumorefficacy of functional paclitaxel nanomicelles in treating resistant breast cancersby oral delivery, Biomaterials 32 (2011) 3285–3302.

[233] M.J. Weiss, J.R. Wong, C.S. Ha, R. Bleday, R.R. Salem, G.D. Steele Jr., L.B. Chen,Dequalinium, a topical antimicrobial agent, displays anticarcinoma activitybased on selective mitochondrial accumulation, Proc. Natl. Acad. Sci. U. S. A. 84(1987) 5444–5448.

[234] R.D. Dabholkar, R.M. Sawant, D.A. Mongayt, P.V. Devarajan, V.P. Torchilin, Poly-ethylene glycol-phosphatidylethanolamine conjugate (PEG-PE)-based mixedmicelles: some properties, loading with paclitaxel, and modulation ofP-glycoprotein-mediated efflux, Int. J. Pharm. 315 (2006) 148–157.

[235] S.C. Lee, K.M. Huh, J. Lee, Y.W. Cho, R.E. Galinsky, K. Park, Hydrotropic polymericmicelles for enhanced paclitaxel solubility: in vitro and in vivo characterization,Biomacromolecules 8 (2007) 202–208.

[236] S. Kim, J.Y. Kim, K.M. Huh, G. Acharya, K. Park, Hydrotropic polymer micellescontaining acrylic acid moieties for oral delivery of paclitaxel, J. Control. Release132 (2008) 222–229.

[237] G. Cavallaro, L. Maniscalco, M. Licciardi, G. Giammona, Tamoxifen-loaded poly-meric micelles: preparation, physico-chemical characterization and in vitro eval-uation studies, Macromol. Biosci. 4 (2004) 1028–1038.

[238] Z. Sezgin, N. Yuksel, T. Baykara, Preparation and characterization of polymericmicelles for solubilization of poorly soluble anticancer drugs, Eur. J. Pharm.Biopharm. 64 (2006) 261–268.

[239] A.F. Aisha, Z. Ismail, K.M. Abu-Salah, A.M. Majid, Solid dispersions of alpha-mangostin improve its aqueous solubility through self-assembly of nanomicelles,J. Pharm. Sci. 101 (2012) 815–825.

[240] H. Yuan, L. Lu, Y.Z. Du, F.Q. Hu, Stearic acid-g-chitosan polymeric micelle for oral drugdelivery: in vitro transport and in vivo absorption, Mol. Pharm. 8 (2011) 225–238.

[241] L. Bromberg, V. Alakhov, Effects of polyether-modified poly(acrylic acid)microgels on doxorubicin transport in human intestinal epithelial Caco-2 celllayers, J. Control. Release 88 (2003) 11–22.

[242] Y.I. Jeong, H. Kim do, C.W. Chung, J.J. Yoo, K.H. Choi, C.H. Kim, S.H. Ha, D.H. Kang,Doxorubicin-incorporated polymeric micelles composed of dextran-b-poly(DL-lactide-co-glycolide) copolymer, Int. J. Nanomedicine 6 (2011) 1415–1427.

[243] H. Li, M. Huo, J. Zhou, Y. Dai, Y. Deng, X. Shi, J. Masoud, Enhanced oral absorptionof paclitaxel in N-deoxycholic acid-N,O-hydroxyethyl chitosan micellar system,J. Pharm. Sci. 99 (2010) 4543–4553.

[244] R. Mo, X. Jin, N. Li, C. Ju, M. Sun, C. Zhang, Q. Ping, The mechanism of enhance-ment on oral absorption of paclitaxel by N-octyl-O-sulfate chitosan micelles,Biomaterials 32 (2011) 4609–4620.

[245] B.J. Tan, Y. Liu, K.L. Chang, B.K. Lim, G.N. Chiu, Perorally active nanomicellarformulation of quercetin in the treatment of lung cancer, Int. J. Nanomedicine7 (2012) 651–661.

[246] R. Duncan, Polymer conjugates as anticancer nanomedicines, Nat. Rev. Cancer 6(2006) 688–701.

[247] G. Pasut, F.M. Veronese, Polymer–drug conjugation, recent achievements andgeneral strategies, Prog. Polym. Sci. 32 (2007) 933–961.

[248] M.C. Garnett, Targeted drug conjugates: principles and progress, Adv. DrugDeliv. Rev. 53 (2001) 171–216.

[249] R. Tong, J. Cheng, Controlled synthesis of camptothecin–polylactide conjugatesand nanoconjugates, Bioconjug. Chem. 21 (2010) 111–121.

[250] E.J. Chaney, L. Tang, R. Tong, J. Cheng, S.A. Boppart, Lymphatic biodistribution ofpolylactide nanoparticles, Mol. Imaging 9 (2010) 153–162.

[251] F. Kratz, K. Abu Ajaj, A. Warnecke, Anticancer carrier-linked prodrugs in clinicaltrials, Expert Opin. Investig. Drugs 16 (2007) 1037–1058.

[252] O. Benny, O. Fainaru, A. Adini, F. Cassiola, L. Bazinet, I. Adini, E. Pravda, Y.Nahmias, S. Koirala, G. Corfas, R.J. D'Amato, J. Folkman, An orally deliveredsmall-molecule formulation with antiangiogenic and anticancer activity, Nat.Biotechnol. 26 (2008) 799–807.

[253] S.Q. Gao, Z.R. Lu, B. Petri, P. Kopeckova, J. Kopecek, Colon-specific9-aminocamptothecin–HPMA copolymer conjugates containing a 1,6-eliminationspacer, J. Control. Release 110 (2006) 323–331.

[254] B.Y. Kim, J.H. Jeong, K. Park, J.D. Kim, Bioadhesive interaction and hypoglycemiceffect of insulin-loaded lectin–microparticle conjugates in oral insulin deliverysystem, J. Control. Release 102 (2005) 525–538.

[255] M. Thanou, J. Verhoef, H. Junginger, Oral drug absorption enhancement by chitosanand its derivatives, Adv. Drug Deliv. Rev. 52 (2001) 117–126.

[256] M.N.V.R. Kumar, R.A.A. Muzzarelli, C. Muzzarelli, H. Sashiwa, A.J. Domb, Chitosanchemistry and pharmaceutical perspectives, Chem. Rev. 104 (2004) 6017–6084.

[257] S.Y. Chae, M.K. Jang, J.W. Nah, Influence of molecular weight on oral absorptionof water soluble chitosans, J. Control. Release 102 (2005) 383–394.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 25: Oral delivery of anticancer drugs: Challenges and opportunities

T

23412342234323442345234623472348234923502351235223532354235523562357235823592360236123622363236423652366236723682369237023712372237323742375237623772378237923802381238223832384238523862387238823892390239123922393239423952396239723982399240024012402240324042405240624072408240924102411241224132414241524162417241824192420242124222423242424252426

24272428242924302431243224332434243524362437243824392440244124422443244424452446244724482449245024512452245324542455245624572458245924602461246224632464246524662467246824692470247124722473247424752476247724782479248024812482248324842485248624872488248924902491249224932494249524962497249824992500250125022503250425052506250725082509251025112512

25K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

[258] N.G.M. Schipper, S. Olsson, J.A. Hoogstraate, A.G. deBoer, K.M. Vårum, P.Artursson, Chitosans as absorption enhancers for poorly absorbable drugs 2:Mechanism of absorption enhancement, Pharm. Res. 14 (1997) 923–929.

[259] N.G.M. Schipper, K.M. Vårum, P. Artursson, Chitosans as absorption enhancersfor poorly absorbable drugs. 1: Influence of molecular weight and degree ofacetylation on drug transport across human intestinal epithelial (Caco-2) cells,Pharm. Res. 13 (1996) 1686–1692.

[260] E. Lee, J. Lee, I.H. Lee, M. Yu, H. Kim, S.Y. Chae, S. Jon, Conjugated chitosan as anovel platform for oral delivery of paclitaxel, J. Med. Chem. 51 (2008)6442–6449.

[261] R.G. Crounse, Human pharmacology of griseofulvin: the effect of fat intake ongastrointestinal absorption, J. Investig. Dermatol. 37 (1961) 529–533.

[262] S. Chakraborty, D. Shukla, B. Mishra, S. Singh, Lipid—an emerging platform fororal delivery of drugs with poor bioavailability, Eur. J. Pharm. Biopharm. 73(2009) 1–15.

[263] C.J. Porter, C.W. Pouton, J.F. Cuine, W.N. Charman, Enhancing intestinal drugsolubilisation using lipid-based delivery systems, Adv. Drug Deliv. Rev. 60(2008) 673–691.

[264] S. Talegaonkar, O. Bhushinge, S. Jain, Self nanoemulsifying system for enhancedlymphatic uptake: an approach to increase oral bioavailability, in: S.P. Vyas,R.S.R. Murthy, R.K. Narang (Eds.), Nano-colloidal Carriers: Site Specific andControlled Drug Delivery, CBS Publishers and Distributors, New Delhi, India,2010, pp. 320–364.

[265] C.J.H. Porter, W.N. Charman, Uptake of drugs into the intestinal lymphatics afteroral administration, Adv. Drug Deliv. Rev. 25 (1997) 71–89.

[266] N. Anton, T.F. Vandamme, Nano-emulsions and micro-emulsions: clarificationsof the critical differences, Pharm. Res. 28 (2011) 978–985.

[267] S. Ganta, D. Deshpande, A. Korde, M. Amiji, A review of multifunctionalnanoemulsion systems to overcome oral and CNS drug delivery barriers, Mol.Membr. Biol. 27 (2010) 260–273.

[268] S.A. Wickline, G.M. Lanza, Nanotechnology for molecular imaging and targetedtherapy, Circulation 107 (2003) 1092–1095.

[269] C.Y. Kuan, T.H. Walker, P.G. Luo, C.F. Chen, Long-chain polyunsaturated fattyacids promote paclitaxel cytotoxicity via inhibition of the MDR1 gene in thehuman colon cancer Caco-2 cell line, J. Am. Coll. Nutr. 30 (2011) 265–273.

[270] S.B. Tiwari, M.M. Amiji, Improved oral delivery of paclitaxel following administra-tion in nanoemulsion formulations, J. Nanosci. Nanotechnol. 6 (2006) 3215–3221.

[271] W. Ge, Y. Li, Z.S. Li, S.H. Zhang, Y.J. Sun, P.Z. Hu, X.M. Wang, Y. Huang, S.Y. Si, X.M.Zhang, Y.F. Sui, The antitumor immune responses induced by nanoemulsion-encapsulated MAGE1-HSP70/SEA complex protein vaccine following peroraladministration route, Cancer Immunol. Immunother. 58 (2009) 201–208.

[272] H.S. Qhattal, S. Wang, T. Salihima, S.K. Srivastava, X. Liu, Nanoemulsions of can-cer chemopreventive agent benzyl isothiocyanate display enhanced solubility,dissolution, and permeability, J. Agric. Food Chem. 59 (2011) 12396–12404.

[273] H.Y. Lin, J.L. Thomas, H.W. Chen, C.M. Shen, W.J. Yang, M.H. Lee, In vitro suppres-sion of oral squamous cell carcinoma growth by ultrasound-mediated deliveryof curcumin microemulsions, Int. J. Nanomedicine 7 (2012) 941–951.

[274] H. Yu, Q. Huang, Improving oral bioavailability of curcumin using novelorganogel-based nanoemulsions, J. Agric. Food Chem. 60 (2012) 5373–5379.

[275] P. Rajpoot, V. Bali, K. Pathak, Anticancer efficacy, tissue distribution and bloodpharmacokinetics of surface modified nanocarrier containing melphalan, Int. J.Pharm. 426 (2012) 219–230.

[276] H.Y. Karasulu, B. Karabulut, E. Goker, T. Guneri, F. Gabor, Controlled release ofmethotrexate from w/o microemulsion and its in vitro antitumor activity, DrugDeliv. 14 (2007) 225–233.

[277] A.O. Nornoo, H.A. Zheng, L.B. Lopes, B. Johnson-Restrepo, K. Kannan, R. Reed,Oral microemulsions of paclitaxel: in situ and pharmacokinetic studies, Eur. J.Pharm. Biopharm. 71 (2009) 310–317.

[278] S. Khandavilli, R. Panchagnula, Nanoemulsions as versatile formulations for pac-litaxel delivery: peroral and dermal delivery studies in rats, J. Investig. Dermatol.127 (2006) 154–162.

[279] S. Ganta, M. Amiji, Coadministration of paclitaxel and curcumin in nanoemulsionformulations to overcome multidrug resistance in tumor cells, Mol. Pharm. 6(2009) 928–939.

[280] H. Reiss, Entropy-induced dispersion of bulk liquids, J. Colloid Interface Sci. 53(1975) 61–70.

[281] R.N. Gursoy, S. Benita, Self-emulsifying drugdelivery systems (SEDDS) for improvedoral delivery of lipophilic drugs, Biomed. Pharmacother. 58 (2004) 173–182.

[282] P. Gao, M.E. Guyton, T. Huang, J.M. Bauer, K.J. Stefanski, Q. Lu, Enhanced oral bio-availability of a poorly water soluble drug PNU-91325 by supersaturatable for-mulations, Drug Dev. Ind. Pharm. 30 (2004) 221–229.

[283] Y. Chen, C. Chen, J. Zheng, Z. Chen, Q. Shi, H. Liu, Development of a solidsupersaturatable self-emulsifying drug delivery system of docetaxel with im-proved dissolution and bioavailability, Biol. Pharm. Bull. 34 (2011) 278–286.

[284] L. Zhang, W. Zhu, C. Yang, H. Guo, A. Yu, J. Ji, Y. Gao, M. Sun, G. Zhai, A novelfolate-modified self-microemulsifying drug delivery system of curcumin forcolon targeting, Int. J. Nanomedicine 7 (2012) 151–162.

[285] B. Tang, G. Cheng, J.C. Gu, C.H. Xu, Development of solid self-emulsifying drugdelivery systems: preparation techniques and dosage forms, Drug Discov.Today 13 (2008) 606–612.

[286] A.V. Shah, A.T. Serajuddin, Development of solid self-emulsifying drug deliverysystem (SEDDS) I: use of poloxamer 188 as both solidifying and emulsifyingagent for lipids, Pharm. Res. 29 (2012) 2817–2832.

[287] S. Jain, O.M. Bhushinge, A.K. Jain, N.K. Swarnakar, H. Harde, Pharmaceutical com-positions for enhancing anticancer efficacy of tamoxifen, Indian patent applica-tion no. 1960/DEL/2011 filed on July 13, 2011

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

[288] S. Jain, O.M. Bhushinge, A.K. Jain, N.K. Swarnakar, H. Harde, Pharmaceutical com-positions for enhancing anticancer efficacy of tamoxifen, World IntellectualProperty Office (WIPO). Patent Collaboration Treaty (PCT) patents, PCT/IB2012/001361 filed on July 11, 2012

[289] S. Jain, M. Pohekar, A.K. Jain, Self emulsifying pharmaceutical composition fororal delivery of quercetin, Indian patent application no. 3526/DEL/2011 filedon December 05, 2011.

[290] Y.D. Yan, N. Marasini, Y.K. Choi, J.O. Kim, J.S. Woo, C.S. Yong, H.G. Choi, Effect ofdose and dosage interval on the oral bioavailability of docetaxel in combinationwith a curcumin self-emulsifying drug delivery system (SEDDS), Eur. J. DrugMetab. Pharmacokinet. 37 (2012) 217–224.

[291] J.L. Lu, J.C. Wang, S.X. Zhao, X.Y. Liu, H. Zhao, X. Zhang, S.F. Zhou, Q. Zhang,Self-microemulsifying drug delivery system (SMEDDS) improves anticancer ef-fect of oral 9-nitrocamptothecin on human cancer xenografts in nude mice, Eur.J. Pharm. Biopharm. 69 (2008) 899–907.

[292] Y.H. Wang, S.H. Wang, R.P. Yang, X.J. Ma, Design, optimization and quality eval-uation of curcumin self-emulsifying drug delivery system (SEDDS), Zhong YaoCai 33 (2010) 1933–1937.

[293] Y.D. Yan, J.A. Kim, M.K. Kwak, B.K. Yoo, C.S. Yong, H.G. Choi, Enhanced oral bio-availability of curcumin via a solid lipid-based self-emulsifying drug deliverysystem using a spray-drying technique, Biol. Pharm. Bull. 34 (2011) 1179–1186.

[294] Y.B. Pawar, H. Purohit, G.R. Valicherla, B. Munjal, S.V. Lale, S.B. Patel, A.K. Bansal,Novel lipid based oral formulation of curcumin: development and optimizationby design of experiments approach, Int. J. Pharm. 436 (2012) 617–623.

[295] X. Wu, J. Xu, X. Huang, C. Wen, Self-microemulsifying drug delivery system im-proves curcumin dissolution and bioavailability, Drug Dev. Ind. Pharm. 37(2011) 15–23.

[296] Z. Wu, D. Guo, L. Deng, Y. Zhang, Q. Yang, J. Chen, Preparation and evaluation of aself-emulsifying drug delivery system of etoposide–phospholipid complex, DrugDev. Ind. Pharm. 37 (2011) 103–112.

[297] A.K. Singh, A. Chaurasiya, A. Awasthi, G. Mishra, D. Asati, R.K. Khar, R. Mukherjee,Oral bioavailability enhancement of exemestane from self-microemulsifyingdrug delivery system (SMEDDS), AAPS PharmSciTech 10 (2009) 906–916.

[298] D. Attivi, I. Ajana, A. Astier, B. Demore, S. Gibaud, Development of microemulsionof mitotane for improvement of oral bioavailability, Drug Dev. Ind. Pharm. 36(2010) 421–427.

[299] H. Thakkar, J. Nangesh, M. Parmar, D. Patel, Formulation and characterization oflipid-based drug delivery system of raloxifene-microemulsion andself-microemulsifying drug delivery system, J. Pharm. Bioallied. Sci. 3 (2011)442–448.

[300] S. Yang, R.N. Gursoy, G. Lambert, S. Benita, Enhanced oral absorption of paclitax-el in a novel self-microemulsifying drug delivery system with or without con-comitant use of P-glycoprotein inhibitors, Pharm. Res. 21 (2004) 261–270.

[301] P. Gao, B.D. Rush, W.P. Pfund, T. Huang, J.M. Bauer, W. Morozowich, M.S. Kuo,M.J. Hageman, Development of a supersaturable SEDDS (S-SEDDS) formulationof paclitaxel with improved oral bioavailability, J. Pharm. Sci. 92 (2003)2386–2398.

[302] N. Gursoy, J.S. Garrigue, A. Razafindratsita, G. Lambert, S. Benita, Excipient effectson in vitro cytotoxicity of a novel paclitaxel self-emulsifying drug delivery sys-tem, J. Pharm. Sci. 92 (2003) 2411–2418.

[303] W. Mehnert, K. Mäder, Solid lipid nanoparticles: production, characterizationand applications, Adv. Drug Deliv. Rev. 47 (2001) 165–196.

[304] D. Pandita, A. Ahuja, T. Velpandian, V. Lather, T. Dutta, R.K. Khar, Characteriza-tion and in vitro assessment of paclitaxel loaded lipid nanoparticles formulatedusing modified solvent injection technique, Pharmazie 64 (2009) 301–310.

[305] S. Yang, J. Zhu, Y. Lu, B. Liang, C. Yang, Body distribution of camptothecin solidlipid nanoparticles after oral administration, Pharm. Res. 16 (1999) 751–757.

[306] M.A. Casadei, F. Cerreto, S. Cesa, M. Giannuzzo, M. Feeney, C. Marianecci, P.Paolicelli, Solid lipid nanoparticles incorporated in dextran hydrogels: a newdrug delivery system for oral formulations, Int. J. Pharm. 325 (2006) 140–146.

[307] H.L. Wong, A.M. Rauth, R. Bendayan, X.Y. Wu, In vivo evaluation of a new poly-mer–lipid hybrid nanoparticle (PLN) formulation of doxorubicin in a murinesolid tumor model, Eur. J. Pharm. Biopharm. 65 (2007) 300–308.

[308] H.L. Wong, R. Bendayan, A.M. Rauth, H.Y. Xue, K. Babakhanian, X.Y. Wu, A mech-anistic study of enhanced doxorubicin uptake and retention in multidrug resis-tant breast cancer cells using a polymer–lipid hybrid nanoparticle system, J.Pharmacol. Exp. Ther. 317 (2006) 1372–1381.

[309] A.E. Yassin, M.K. Anwer, H.A. Mowafy, I.M. El-Bagory, M.A. Bayomi, I.A. Alsarra,Optimization of 5-flurouracil solid–lipid nanoparticles: a preliminary study totreat colon cancer, Int. J. Med. Sci. 7 (2010) 398–408.

[310] P. Ma, X. Dong, C.L. Swadley, A. Gupte, M. Leggas, H.C. Ledebur, R.J. Mumper, De-velopment of idarubicin and doxorubicin solid lipid nanoparticles to overcomePgp-mediated multiple drug resistance in leukemia, J. Biomed. Nanotechnol. 5(2009) 151–161.

[311] S. Wang, T. Chen, R. Chen, Y. Hu, M. Chen, Y. Wang, Emodin loaded solid lipidnanoparticles: preparation, characterization and antitumor activity studies, Int.J. Pharm. 430 (2012) 234–246.

[312] K. Ruckmani, M. Sivakumar, P.A. Ganeshkumar, Methotrexate loaded solid lipidnanoparticles (SLN) for effective treatment of carcinoma, J. Nanosci.Nanotechnol. 6 (2006) 2991–2995.

[313] L.H. Reddy, K. Vivek, N. Bakshi, R.S. Murthy, Tamoxifen citrate loaded solid lipidnanoparticles (SLN): preparation, characterization, in vitro drug release, andpharmacokinetic evaluation, Pharm. Dev. Technol. 11 (2006) 167–177.

[314] L. Wu, C. Tang, C. Yin, Folate-mediated solid–liquid lipid nanoparticles forpaclitaxel-coated poly(ethylene glycol), Drug Dev. Ind. Pharm. 36 (2010)439–448.

ugs: Challenges and opportunities, J. Control. Release (2013), http://

Page 26: Oral delivery of anticancer drugs: Challenges and opportunities

T

2513251425152516251725182519252025212522252325242525252625272528252925302531253225332534253525362537253825392540254125422543254425452546254725482549255025512552255325542555255625572558255925602561256225632564256525662567256825692570257125722573257425752576257725782579258025812582258325842585

258625872588258925902591259225932594259525962597259825992600260126022603260426052606260726082609261026112612261326142615261626172618261926202621262226232624262526262627262826292630263126322633263426352636263726382639264026412642264326442645264626472648264926502651265226532654265526562657

2658

26 K. Thanki et al. / Journal of Controlled Release xxx (2013) xxx–xxx

UNCO

RREC

[315] V.S. Shenoy, R.P. Gude, R.S. Murthy, Paclitaxel-loaded glyceryl palmitostearatenanoparticles: in vitro release and cytotoxic activity, J. Drug Target. 17 (2009)304–310.

[316] H. Yuan, J. Miao, Y.Z. Du, J. You, F.Q. Hu, S. Zeng, Cellular uptake of solid lipidnanoparticles and cytotoxicity of encapsulated paclitaxel in A549 cancer cells,Int. J. Pharm. 348 (2008) 137–145.

[317] F. Wan, J. You, Y. Sun, X.G. Zhang, F.D. Cui, Y.Z. Du, H. Yuan, F.Q. Hu, Studies onPEG-modified SLNs loading vinorelbine bitartrate (I): preparation and evalua-tion in vitro, Int. J. Pharm. 359 (2008) 104–110.

[318] M. Muchow, P. Maincent, R.H. Muller, Lipid nanoparticles with a solid matrix (SLN,NLC, LDC) for oral drug delivery, Drug Dev. Ind. Pharm. 34 (2008) 1394–1405.

[319] W.M. Obeidat, K. Schwabe, R.H. Muller, C.M. Keck, Preservation of nanostruc-tured lipid carriers (NLC), Eur. J. Pharm. Biopharm. 76 (2010) 56–67.

[320] H. Ali, A.B. Shirode, P.W. Sylvester, S. Nazzal, Preparation and in vitroantiproliferative effect of tocotrienol loaded lipid nanoparticles, Colloids Surf.A 353 (2010) 43–51.

[321] T. Zhang, J. Chen, Y. Zhang, Q. Shen, W. Pan, Characterization and evaluation ofnanostructured lipid carrier as a vehicle for oral delivery of etoposide, Eur. J.Pharm. Sci. 43 (2011) 174–179.

[322] N.T. Huynh, C. Passirani, P. Saulnier, J.P. Benoit, Lipid nanocapsules: a new platformfor nanomedicine, Int. J. Pharm. 379 (2009) 201–209.

[323] B. Heurtault, P. Saulnier, B. Pech, J.E. Proust, J.P. Benoit, A novel phase inversion-basedprocess for the preparation of lipid nanocarriers, Pharm. Res. 19 (2002) 875–880.

[324] N. Anton, P. Saulnier, C. Gaillard, E. Porcher, S. Vrignaud, J.P. Benoit, Aqueous-core lipid nanocapsules for encapsulating fragile hydrophilic and/or lipophilicmolecules, Langmuir 25 (2009) 11413–11419.

[325] E. Roger, F. Lagarce, J.P. Benoit, The gastrointestinal stability of lipid nano-capsules, Int. J. Pharm. 379 (2009) 260–265.

[326] E. Roger, F. Lagarce, J.P. Benoit, Development and characterization of a novellipid nanocapsule formulation of Sn38 for oral administration, Eur. J. Pharm.Biopharm. 79 (2011) 181–188.

[327] C.R. Dass, Drug delivery in cancer using liposomes, Methods Mol. Biol. 437(2008) 177–182.

[328] A. Gabizon, Liposomes as a drug delivery system in cancer chemotherapy, Horiz.Biochem. Biophys. 9 (1989) 185–211.

[329] Y. Malam, M. Loizidou, A.M. Seifalian, Liposomes and nanoparticles: nanosizedvehicles for drug delivery in cancer, Trends Pharmacol. Sci. 30 (2009) 592–599.

[330] H. Li, J.H. Song, J.S. Park, K. Han, Polyethylene glycol-coated liposomes for oraldelivery of recombinant human epidermal growth factor, Int. J. Pharm. 258(2003) 11–19.

[331] M.A. Kisel, L.N. Kulik, I.S. Tsybovsky, A.P. Vlasov, M.S. Vorob'Yov, E.A. Kholodova,Z.V. Zabarovskaya, Liposomes with phosphatidylethanol as a carrier for oraldelivery of insulin: studies in the rat, Int. J. Pharm. 216 (2001) 105–114.

[332] K. Iwanaga, S. Ono, K. Narioka, K. Morimoto, M. Kakemi, S. Yamashita, M. Nango,N. Oku, Oral delivery of insulin by using surface coating liposomes: improve-ment of stability of insulin in GI tract, Int. J. Pharm. 157 (1997) 73–80.

[333] J. Okada, S. Cohen, R. Langer, In vitro evaluation of polymerized liposomes as anoral drug delivery system, Pharm. Res. 12 (1995) 576–582.

[334] P. Jain, S. Jain, K.N. Prasad, S.K. Jain, S.P. Vyas, Polyelectrolyte coated multilayeredliposomes (nanocapsules) for the treatment of Helicobacter pylori infection, Mol.Pharm. 6 (2009) 593–603.

[335] H. Takeuchi, Y. Matsui, H. Sugihara, H. Yamamoto, Y. Kawashima, Effectivenessof submicron-sized, chitosan-coated liposomes in oral administration of peptidedrugs, Int. J. Pharm. 303 (2005) 160–170.

[336] P.S. Hiremath, K.S. Soppimath, G.V. Betageri, Proliposomes of exemestane forimproved oral delivery: formulation and in vitro evaluation using PAMPA,Caco-2 and rat intestine, Int. J. Pharm. 380 (2009) 96–104.

[337] S.S. Ling, E. Magosso, N.A. Khan, K.H. Yuen, S.A. Barker, Enhanced oral bioavail-ability and intestinal lymphatic transport of a hydrophilic drug using liposomes,Drug Dev. Ind. Pharm. 32 (2006) 335–345.

[338] S. Jain, D. Kumar, N.K. Swarnakar, K. Thanki, Novel layersome composition for oraldelivery of anti cancer agents, Indianpatent application no. 3246/DEL/2011filed onNovember 15, 2011.

[339] S. Jain, S.R. Patil, N.K. Swarnakar, A.K. Agrawal, Oral delivery of doxorubicinusing novel polyelectrolyte-stabilized liposomes (layersomes), Mol. Pharm. 9(2012) 2626–2635.

[340] S. Jain, D. Kumar, N.K. Swarnakar, K. Thanki, Polyelectrolyte stabilizedmultilayeredliposomes for oral delivery of paclitaxel, Biomaterials 33 (2012) 6758–6768.

[341] Y. Yan, C.J. Ochs, G.K. Such, J.K. Heath, E.C. Nice, F. Caruso, Bypassing multidrugresistance in cancer cells with biodegradable polymer capsules, Adv. Mater. 22(2010) 5398–5403.

[342] Y. Yan, A.P.R. Johnston, S.J. Dodds, M.M.J. Kamphuis, C. Ferguson, R.G. Parton, E.C.Nice, J.K. Heath, F. Caruso, Uptake and intracellular fate of disulfide-bonded

Please cite this article as: K. Thanki, et al., Oral delivery of anticancer drdx.doi.org/10.1016/j.jconrel.2013.04.020

ED P

RO

OF

polymer hydrogel capsules for doxorubicin delivery to colorectal cancer cells,ACS Nano 4 (2010) 2928–2936.

[343] N.K. Swarnakar, V. Jain, V. Dubey, D. Mishra, N.K. Jain, Enhanced oromucosaldelivery of progesterone via hexosomes, Pharm. Res. 24 (2007) 2223–2230.

[344] S. Jain, N. Bhankur, N.K. Swarnakar, K. Thanki, Novel folic acid functionalized liq-uid crystalline nanoparticles for enhanced tumor delivery of docetaxel, Indianpatent application number 680/DEL/2013 filed on March 8, 2013.

[345] N. Zeng, X. Gao, Q. Hu, Q. Song, H. Xia, Z. Liu, G. Gu, M. Jiang, Z. Pang, H. Chen, J.Chen, L. Fang, Lipid-based liquid crystalline nanoparticles as oral drug deliveryvehicles for poorly water-soluble drugs: cellular interaction and in vivo absorp-tion, Int. J. Nanomedicine 7 (2012) 3703–3718.

[346] S.C. Kinsky, J.E. Loader, Circumvention of the methotrexate transport system bymethotrexate-phosphatidylethanolamine derivatives: effect of fatty acid chainlength, Biochim. Biophys. Acta 921 (1987) 96–103.

[347] R. Pignatello, G. Spampinato, V. Sorrenti, C. Di Giacomo, L. Vicari, J.J. McGuire,C.A. Russell, G. Puglisi, I. Toth, Lipophilic methotrexate conjugates withantitumor activity, Eur. J. Pharm. Sci. 10 (2000) 237–245.

[348] M.O. Bradley, C.S. Swindell, F.H. Anthony, P.A. Witman, P. Devanesan, N.L. Webb,S.D. Baker, A.C. Wolff, R.C. Donehower, Tumor targeting by conjugation of DHAto paclitaxel, J. Control. Release 74 (2001) 233–236.

[349] R. Paliwal, S. Rai, S.P. Vyas, Lipid drug conjugate (LDC) nanoparticles asautolymphotrophs for oral delivery of methotrexate, J. Biomed. Nanotechnol. 7(2011) 130–131.

[350] X. Gong, M.J. Moghaddam, S.M. Sagnella, C.E. Conn, S.J. Danon, L.J. Waddington,C.J. Drummond, Lyotropic liquid crystalline self-assembly material behavior andnanoparticulate dispersions of a phytanyl pro-drug analogue of capecitabine—achemotherapy agent, ACS Appl. Mater. Interfaces 3 (2011) 1552–1561.

[351] Y. Cheng, Z. Xu, M. Ma, T. Xu, Dendrimers as drug carriers: applications in differentroutes of drug administration, J. Pharm. Sci. 97 (2008) 123–143.

[352] R.K. Tekade, P.V. Kumar, N.K. Jain, Dendrimers in oncology: an expanding horizon,Chem. Rev. 109 (2009) 49–87.

[353] S. Svenson, A.S. Chauhan, Dendrimers for enhanced drug solubilization,Nanomedicine (Lond) 3 (2008) 679–702.

[354] S. Sadekar, H. Ghandehari, Transepithelial transport and toxicity of PAMAMdendrimers: implications for oral drug delivery, Adv. Drug Deliv. Rev. 64(2012) 571–588.

[355] N.K. Jain, U. Gupta, Application of dendrimer–drug complexation in the en-hancement of drug solubility and bioavailability, Expert Opin. Drug Metab.Toxicol. 4 (2008) 1035–1052.

[356] N. Pantzar, S. Lundin, L. Wester, B.R. Westrom, Bidirectional small-intestinalpermeability in the rat to some common marker molecules in vitro, Scand.J. Gastroenterol. 29 (1994) 703–709.

[357] R. Wiwattanapatapee, B. Carreno-Gomez, N. Malik, R. Duncan, Anionic PAMAMdendrimers rapidly cross adult rat intestine in vitro: a potential oral delivery sys-tem? Pharm. Res. 17 (2000) 991–998.

[358] M. El-Sayed, M. Ginski, C. Rhodes, H. Ghandehari, Transepithelial transport ofpoly(amidoamine) dendrimers across Caco-2 cell monolayers, J. Control. Release81 (2002) 355–365.

[359] W. Ke, Y. Zhao, R. Huang, C. Jiang, Y. Pei, Enhanced oral bioavailability of doxorubi-cin in a dendrimer drug delivery system, J. Pharm. Sci. 97 (2008) 2208–2216.

[360] R.B. Kolhatkar, P. Swaan, H. Ghandehari, Potential oral delivery of 7-ethyl-10-hydroxy-camptothecin (SN-38) using poly(amidoamine) dendrimers, Pharm.Res. 25 (2008) 1723–1729.

[361] Y. Lin, T. Fujimori, N. Kawaguchi, Y. Tsujimoto, M. Nishimi, Z. Dong, H. Katsumi,T. Sakane, A. Yamamoto, Polyamidoamine dendrimers as novel potential ab-sorption enhancers for improving the small intestinal absorption of poorlyabsorbable drugs in rats, J. Control. Release 149 (2011) 21–28.

[362] D.M. Sweet, R.B. Kolhatkar, A. Ray, P. Swaan, H. Ghandehari, Transepithelialtransport of PEGylated anionic poly(amidoamine) dendrimers: implicationsfor oral drug delivery, J. Control. Release 138 (2009) 78–85.

[363] D.S. Pisal, V.K. Yellepeddi, A. Kumar, R.S. Kaushik, M.B. Hildreth, X. Guan, S.Palakurthi, Permeability of surface-modified polyamidoamine (PAMAM)dendrimers across Caco-2 cell monolayers, Int. J. Pharm. 350 (2008) 113–121.

[364] R.B. Kolhatkar, K.M. Kitchens, P.W. Swaan, H. Ghandehari, Surface acetylation ofpolyamidoamine (PAMAM) dendrimers decreases cytotoxicity while maintainingmembrane permeability, Bioconjug. Chem. 18 (2007) 2054–2060.

[365] R. Jevprasesphant, J. Penny, D. Attwood, N.B. McKeown, A. D'Emanuele, Engi-neering of dendrimer surfaces to enhance transepithelial transport and reducecytotoxicity, Pharm. Res. 20 (2003) 1543–1550.

[366] A.T. Florence, T. Sakthivel, I. Toth, Oral uptake and translocation of a polylysinedendrimer with a lipid surface, J. Control. Release 65 (2000) 253–259.

ugs: Challenges and opportunities, J. Control. Release (2013), http://