Differences in Nanoparticle Uptake in Transplanted and

Mar 13, 2018 - Human pancreatic ductal adenocarcinoma (PDAC) contains a distinctively dense stroma that limits the accessibility of anticancer drugs, ...
0 downloads 5 Views 4MB Size
Subscriber access provided by - Access paid by the | UCSB Libraries

Differences in Nanoparticle Uptake in Transplanted and Autochthonous Models of Pancreatic Cancer Zhimin Tao, Mandar Deepak Muzumdar, Alexandre Detappe, Xing Huang, Eric Xu, Yingjie Yu, Tarek H Mouhieddine, Haiqin Song, Tyler Jacks, and P. Peter Ghoroghchian Nano Lett., Just Accepted Manuscript • DOI: 10.1021/acs.nanolett.7b04043 • Publication Date (Web): 13 Mar 2018 Downloaded from http://pubs.acs.org on March 14, 2018

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

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

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

Nano Letters

Differences in Nanoparticle Uptake in Transplanted and Autochthonous Models of Pancreatic Cancer Zhimin Tao,1,* Mandar Deepak Muzumdar,1,2,3,4* Alexandre Detappe,1,2,3 Xing Huang,1 Eric S. Xu,1 Yingjie Yu,1 Tarek H. Mouhieddine,2,3 Haiqin Song,1 Tyler Jacks,1 and P. Peter Ghoroghchian1,2,3,#

1. Koch Institute for Integrative Cancer Research at MIT, 500 Main Street, Cambridge, MA 02139, USA 2. Dana Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA 3. Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA 4. Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA * Denotes equal author contribution

# To whom correspondence should be addressed:

P. Peter Ghoroghchian, M.D., Ph.D. Charles W. and Jennifer C. Johnson Clinical Investigator Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology 77 Massachusetts Avenue, 76-261F Cambridge, MA 02139, USA (617) 252-1163 [email protected]

1 ACS Paragon Plus Environment

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

1

ABSTRACT

2

Human pancreatic ductal adenocarcinoma (PDAC) contains a distinctively dense stroma that

3

limits the accessibility of anticancer drugs, contributing to its poor overall prognosis. Nanoparticles

4

can enhance drug delivery and retention in pancreatic tumors and have been utilized clinically for

5

their treatment. In preclinical studies, various mouse models differentially recapitulate the

6

microenvironmental features of human PDAC. Here, we demonstrate that through utilization of

7

different organic co-solvents and by doping of a homopolymer of poly(-caprolactone), a diblock

8

copolymer composition of poly(ethylene oxide)-block-poly(-caprolactone) may be utilized to

9

generate biodegradable and nanoscale micelles with different physical properties. Noninvasive

10

optical imaging was employed to examine the pharmacology and biodistribution of these various

11

nanoparticle formulations in both allografted and autochthonous mouse models of PDAC. In

12

contrast to the results reported with transplanted tumors, spherical micelles as large as 300 nm

13

in diameter were found to extravasate in the autochthonous model, reaching a distance of

14

approximately 20 m from the nearest tumor cell clusters. A lipophilic platinum(IV) prodrug of

15

oxaliplatin was further able to achieve a ~7-fold higher peak accumulation and a ~50-fold increase

16

in its retention half-life in pancreatic tumors when delivered with 100 nm-long worm-like micelles

17

as when compared to the free drug formulation of oxaliplatin. Through further engineering of

18

nanoparticle properties, as well as by widespread adoption of the autochthonous tumor model for

19

preclinical testing, future therapeutic formulations may further enhance the targeting and

20

penetration of anticancer agents to improve survival outcomes in PDAC.

21 22

KEYWORDS

23

Drug delivery; Nanomedicine; Pancreatic ductal adenocarcinoma; Platinum(IV); Optical Imaging

2 ACS Paragon Plus Environment

Page 2 of 37

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

Nano Letters

24

Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer death in the United

25

States and a significant contributor to morbidity and mortality worldwide1, 2. In spite of advances

26

in combination chemotherapy (e.g., FOLFIRINOX3 and gemcitabine/nab-paclitaxel4), which offer

27

greater response rates and prolong median survival, long-term survival remains poor at 8%1, 2.

28

These therapies also induce considerable adverse effects, limiting their use to patients with good

29

performance status. Given that most patients are diagnosed with unresectable disease at the time

30

of initial presentation2, novel approaches for the treatment of advanced pancreatic cancers are

31

desperately needed.

32 33

A defining feature of PDAC is the extraordinary heterogeneity of the tumor microenvironment5-7.

34

Pancreatic tumor cells are surrounded by a dense stroma that possesses diverse cellular (e.g.,

35

fibroblasts, macrophages, immune cells) and acellular components (e.g., extracellular matrix

36

proteins, hyaluronic acid), which together can comprise > 90% of the tumor by volume6. These

37

stromal features contribute to increased interstitial pressures, impairing intratumoral blood flow

38

and therapeutic delivery to PDAC cells. Efforts to modify the cellular (e.g., through hedgehog

39

inhibition8) and acellular microenvironment (e.g., with the use of hyaluronidase9, 10) have improved

40

vascularization and tumor responses in preclinical PDAC models; they have also shown early

41

promise in clinical trials11, 12. Overcoming the stromal barrier can, thus, enhance the efficacy of

42

existing cytotoxic chemotherapies directed against PDAC, potentially affording greater tumor

43

responses at lower doses and with fewer systemic side effects.

44 45

Nanomedicine offers a means by which to improve anticancer drug delivery through both passive

46

and active targeting mechanisms that promote nanoparticle accumulation in tumors13-16.

47

Nanoparticles have also been successfully utilized to improve clinical outcomes in pancreatic

48

cancers4, 17. Conjugation of cancer cell-specific targeting moieties to nanoparticles can potentially

49

promote greater drug accumulation at tumor sites, minimizing side effects and maximizing 3 ACS Paragon Plus Environment

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

50

therapeutic activity18-20. While lipid-based nanoparticles have been widely-adopted in translational

51

research due to their outstanding biocompatibility, their labile nature may affect stability during

52

both product storage and in vivo administration; polymeric nanoparticles can be better engineered

53

for controlled biodegradability but must overcome concerns related to potential toxicity and

54

immunogenicity.21-26 As an example of the successful preclinical application of the latter, platinum-

55

based anticancer agents have been incorporated into polymeric micelles to prolong their blood

56

circulation times and to enhance tumor accumulation in transgenic as well as in transplanted

57

models of human PDAC27-30. These studies have revealed the potential for nanoparticles of

58

smaller sizes to exhibit preferential accrual in pancreatic tumors28, 31. Little is known, however,

59

about the effects of other nanoparticle characteristics, such as shape, on affecting drug delivery.

60

Moreover, the inconsistent findings reported from previous nanoparticle studies may be

61

attributable to differences in vascular density, in the sizes of endothelial gap junctions, and in the

62

extent of the desmoplastic stroma in the specific transgenic or transplanted tumor model that was

63

utilized; these key features play critical roles in disease pathogenesis, in limiting nanoparticle

64

extravasation and parenchymal drug diffusion, and in stymying the efficacy of antitumor therapies.

65

It is important to underscore that all transgenic and transplanted tumor models fail to faithfully

66

recapitulate the genetic and biological characteristics of human PDAC.8

67 68

Genetically-engineered mouse models of pancreatic cancer have been recently developed in

69

which tumors arise spontaneously in the mouse’s own pancreas (i.e., autochthonous models)32-

70

34.

71

wise progression of human PDAC, including the development of its desmoplastic stroma32-34. We,

72

therefore, utilized both subcutaneous transplant and autochthonous murine models of PDAC to

73

systematically evaluate the interplay between different nanoparticle characteristics (e.g., size and

74

shape) and their impact on overcoming the unique features of each tumor model (i.e., differences

75

in vascular and stromal density) to augment in vivo delivery.

These models accurately mimic the genetic and molecular changes that occur during the step-

4 ACS Paragon Plus Environment

Page 4 of 37

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

Nano Letters

76 77

Generation of polymeric nanoparticles of varying size and shape for in vivo delivery

78

Poly(ethylene oxide)-block-poly(-caprolactone) (PEO-b-PCL) is an amphiphilic diblock

79

copolymer composition comprised of two FDA-approved building blocks that impart favorable

80

physicochemical and biological characteristics, including prolonged in vivo stability, enhanced

81

aqueous solubility (imparted by the PEO block), full biodegradability (of the PCL block), and low

82

in vitro or in vivo toxicity; moreover, while repeated administration of any PEO-coated nanoparticle

83

may elicit a humoral immune response, PEO-b-PCL-based vehicles have been shown to induce

84

minimal immunogenicity.35 We have previously explored the phase behavior of different PEO-b-

85

PCL molecular weight compositions, identifying those that readily generate nanoscale vesicles

86

and spherical micelles in aqueous suspension36, 37. We have demonstrated that PEO-b-PCL-

87

based vesicles can incorporate both chemotherapeutic species36 and near-infrared (NIR)

88

fluorophores38. Additionally, we have conducted in vivo optical imaging studies that have

89

determined preferential accumulation of PEO-b-PCL-based spherical nanoparticles within

90

peritoneal tumor implants after intraperitoneal as opposed to intravenous (IV) administration 39.

91

Previous investigations have also reported that PEO-b-PCL-based worm-like micelles (as

92

opposed to nanospheres) may achieve enhanced penetration through dense tumor tissues40, 41.

93 94

Here, we sought to explore the size and shape dependence of PEO-b-PCL-based nanoparticles

95

to accumulate in pancreatic tumors in hopes of identifying key physical characteristics that may

96

enhance drug delivery. While other studies have explored the size dependencies of nanoparticles

97

to accumulate in pancreatic tumor xenografts28, the Kras- and p53-mutant genetically-engineered

98

mouse model (KPC; see Materials and Methods) more readily recapitulates the key genetic,

99

histologic, and microenviromental features of PDAC33, 34, 42, 43 (Figure 1A, 1B and Supplementary

100

Figure S1). Previous work revealed that transplant and autochthonous PDAC models display

101

marked differences in drug delivery. Specifically, transplant tumors show greater responses to 5 ACS Paragon Plus Environment

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

102

chemotherapy and increased perfusion compared to the autochthonous PDAC model. Further,

103

both human and mouse transplanted tumors exhibit greater vascularity (i.e., greater blood vessel

104

numbers and smaller vessel-to-tumor cell distances) than their naturally arising counterparts.8 As

105

such, we sought to conduct a comparative study of nanoparticle delivery between the

106

autochthonous KPC and traditional transplant models.

107 108

We first focused on identifying fabrication conditions that could give rise to nanoparticles that

109

differed with respect to size and shape, utilizing two PEO-b-PCL compositions (PEO5k-PCL10k and

110

PEO5k-PCL16k), a homopolymer of PCL30k, and two lipophilic encapsulants that could be loaded

111

within the resultant nanoparticles (Figure 1C). The near-infrared (NIR-) emissive carbocyanine

112

dye DiR (1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide) was selected as a

113

model imaging agent and a novel lipophilic prodrug of oxaliplatin (C16-oxaliplatin(IV)-C16) was

114

synthesized for nanoparticle-based encapsulation and delivery to pancreatic tumors.

115

Encapsulation of lipophilic DiR in PEO-b-PCL-based nanoparticles has previously been used to

116

enable their accurate in vivo imaging in subcutaneous tumor xenograft models.44, 45 We have also

117

utilized DiR that was encapsulated in PEO-b-PCL-wrapped lanthanide nanoparticles to

118

successfully image and compare the extent of tumor localization after either intraperitoneal or

119

intravenous administration into a disseminated cell-line xenograft model of ovarian cancer,

120

validating tumor cell (delineated by emission of red fluorescent protein, RFP) and nanoparticle

121

biodistribution (demarcated by the non-overlapping NIR emission of DiR) in vivo as well as ex

122

vivo both in whole-excised organs and in tissue sections via microscopy.39

123 124

To generate nanoparticles of varying size and shape for comparative analyses, we dissolved the

125

PEO-b-PCL copolymers with and without the PCL homopolymer and/or the lipophilic

126

encapsulants (i.e., DiR and/or C16-oxaliplatin(IV)-C16) into two different organic solvents (Figure

6 ACS Paragon Plus Environment

Page 6 of 37

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

Nano Letters

127

1D, Supplementary Scheme S1 and Materials and Methods). When dissolved in THF (and

128

subsequently diluted into larger aqueous volumes), PEO-b-PCL diblock copolymers produced

129

suspensions of spherical nanoparticles; polymers of higher molecular weight formed

130

nanoparticles of larger size as assessed by cryogenic transmission electron microscopy (cryo-

131

TEM; Figure 2A). Interestingly, by first dissolving in water-miscible DMF (followed by aqueous

132

dilution), the same PEO-b-PCL compositions led to the formation of small worm-like micelles. To

133

our knowledge, this is the first example by which different shaped nanoparticles are produced

134

from the same diblock copolymer compositions simply by changing the miscible organic solvent

135

used for aqueous emulsion. Dynamic Light Scattering (DLS) revealed that the spherical

136

nanoparticles made from PEO5k-PCL10k, PEO5k-PCL16k, and PEO5k-PCL16k:PCL30k (1:1 molar

137

ratio) possessed hydrodynamic diameters of ~70 nm, ~100 nm, and ~300 nm, respectively. In

138

contrast, worm-like micelles made from PEO5k-PCL10k had a mean length of 100 nm. We

139

confirmed the stability of each PEO-b-PCL-based formulation; sizes (Figure 2B), polydispersity

140

indexes (PDIs) (Figure 2C), and zeta potential measurements (Figure 2D) from all nanoparticle

141

suspensions were relatively constant over the course of one week. Their surface charges were

142

mildly negative and were likely attributable to their neutrally-charged PEO shells. Through the

143

selection of different initial molecular weight compositions and the simple exchange of organic

144

solvents used for aqueous emulsion, we were, thus, able to reproducibly generate stable

145

polymeric nanoparticles of varying size and shape for further in vitro and in vivo experimentation.

146 147

Prior to their use for in vivo delivery of anticancer agents, we first examined the in vitro cytotoxicity

148

of the various PEO-b-PCL-based nanoparticle formulations. We treated Kras-mutant B22 murine

149

PDAC cells (see Materials and Methods) with different concentrations of PEO-b-PCL-based

150

nanoparticles of varying size and morphology and measured in vitro cell viability at 72 h

151

(Supplementary Figure S2). The IC50 values were determined to be 10, 17.8 and 177.8 mg/mL

152

(corresponding to 0.5, 0.7, and 11.9 mM of polymer) for PEO5k-PCL16k (THF), PEO5k7 ACS Paragon Plus Environment

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

153

PCL16k/PCL30k (THF), and PEO5k-PCL10k (DMF) nanoparticles, respectively. The cytotoxicity of

154

each nanoparticle formulation correlated with its decreasing size and spherical shape: the toxicity

155

imparted by PEO5k-PCL16k (THF; 100 nm diameter spherical micelles) > PEO5k-PCL16k/PCL30k

156

(THF; 300 nm-diameter spherical micelles) > PEO5k-PCL10k (DMF; 100 nm-length worm-like

157

micelles). The worm-like micelles displayed the lowest toxicity on B22 cells as compared to each

158

spherical nanocarrier. While smaller particles may have been expected to introduce slightly more

159

cytotoxicity (at a given polymer concentration) due to improvements in intracellular uptake, the

160

dramatic differences in cytotoxicity between nanoparticles of different shape but of the same

161

overall dimensions was not expected and warrants further study. Nevertheless, these data

162

confirmed that the PEO-b-PCL-based nanoparticles were suitably non-toxic for in vivo use.

163 164

To facilitate in vivo optical imaging of PEO-b-PCL-based nanoparticles, we loaded them with the

165

NIR fluorophore DiR (see Materials and Methods). We observed linear concentration-dependent

166

changes in the fluorescence emission (Figure 3A) and the solution absorption of DiR (Figure 3B

167

and Supplementary Figure S3) when encapsulated in PEO5k-PCL16k–based nanospheres in

168

water. With increasing DiR concentrations, emission and absorbance initially rose but then

169

declined (Figure 3A and 3B), likely due to the self-quenching effect and solubility limit,

170

respectively, of DiR confined in the polymer shell. Consistent with the lowered energy states

171

imparted by the self-quenching and stacking of dye molecules, the peak emission wavelength of

172

DiR displayed a bathochromic shift with increasing concentration in nanoparticle suspensions. To

173

minimize self-quenching, we adopted an initial solution concentration of DiR equal to 5 µM for

174

generation of all nanoparticles used for subsequent in vivo studies. To ensure that this loading

175

concentration enabled linear concentration-dependent detection of particles by in vivo imaging,

176

we next administered DiR-encapsulated PEO5k-PCL16k–based nanospheres at different

177

concentrations to nude mice by IV tail-vein injection. We subsequently measured the fluorescence

178

intensities values of the particles in blood. We observed an increasing linear correlation between 8 ACS Paragon Plus Environment

Page 8 of 37

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

Nano Letters

179

fluorescence efficiency and the DiR concentration at the administered dose level (Figure 3C). We

180

also measured the effects of DiR loading on the chemical properties of all the various nanoparticle

181

formulations. While their final concentrations of encapsulated DiR varied by as much as 2-fold in

182

different solutions (Table 1), the DiR-loaded nanoparticles were highly photostable in water, in

183

phosphate-buffered saline (PBS), and in fetal-bovine serum (FBS) (Figure 3D).

184 185

Delivery of PEO-b-PCL-based nanoparticles to allografted PDAC tumors in mice

186

We evaluated whether the efficiency of tumor accumulation of our PEO-b-PCL-based

187

nanoparticles depended on their size and/or shape, using an allografted murine model of PDAC.

188

We transduced B22 Kras; p53 mutant murine PDAC cells derived from a mouse model that closely

189

recapitulates the human disease46 with a retroviral construct, permitting expression of both

190

luciferase

191

immunofluorescence analysis of tumor sections). We transplanted these cells into the flanks of

192

immunocompromised nude mice, allowing for subcutaneous tumor growth, and then administered

193

various DiR-encapsulated, PEO-b-PCL-based nanoparticle formulations via IV tail-vein injection.

194

The nanoparticles were monitored by their DiR fluorescence intensities (ex= 710 nm; em= 800

195

nm) in tumors/organs (for biodistribution studies) and in blood (for pharmacokinetic analysis) by

196

IVIS imaging at various time points. For the spherical nanoparticles, circulation half-lives

197

decreased as a function of increasing particle size (Table 1 and Supplementary Figures S4-

198

S6), which was consistent with previously reported studies47. The worm-like micelles (100 nm in

199

length) had a t1/2 = (6.6 + 1.9) h (Figure 4A and Supplementary Figure S7), which was

200

significantly shorter than the t1/2 of the 100 nm-diameter spherical nanoparticles (10.9 + 1.0 h;

201

Table 1). Fluorescence imaging (of nanoparticles) was spatially mapped to luminescence signals

202

(from the tumors; e.g., as depicted for the 100 nm-length worm-like micelles in Figure 4A) to

203

determine the extent of nanoparticle accumulation within tumors as compared to other organs.

(LUC;

for

in

vivo

imaging)

and

green

fluorescence

9 ACS Paragon Plus Environment

protein

(GFP;

for

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

204

Note that 80-90% of the maximal tumor accumulation for each nanoparticle formulation was

205

achieved by 24 h after intravenous administration (Supplementary Figure S4-S7).

206 207

The 300-nm diameter spherical nanoparticles displayed minimal accumulation in subcutaneous

208

(SQ) tumors as compared to their 100 nm- or 70 nm-diameter spherical counterparts or to the

209

100 nm-length worm-like micelles (Supplementary Figure S4-S7); in particular, the later

210

construct showed improved tumor accumulation (Table 1), supporting its further development as

211

a pancreatic drug delivery platform. At 72 h post-injection, mice were sacrificed and their major

212

organs were dissected to measure DiR fluorescence distribution ex vivo (e.g., Figure 4A, right

213

insert). The fluorescence radiant efficiency of every organ was normalized to that of the liver in

214

the same mouse (Figure 4B). For all injected suspensions (except for the 300 nm-diameter

215

nanospheres), the tumors showed the highest accumulation of PEO-b-PCL-based nanoparticles

216

as compared to all other organs (n=5 mice per formulation).

217 218

There were no significant differences in the relative tissue distributions of 70 nm- and 100 nm-

219

diameter spherical nanoparticles as compared to the 100 nm-length worm-like micelles, revealing

220

similar ex vivo tumor distribution profiles for the injected doses. In contrast, the relative

221

accumulation of the 300 nm spherical nanoparticles was lower in the tumors than in the liver and

222

was highest in the spleen, consistent with its clearance by the mononuclear phagocytic system

223

(MPS)47. Together, these data suggest that 100 nm-diameter nanoparticles may be delivered

224

efficiently to allograft PDAC tumors in vivo. While exhibiting similar relative ex vivo biodistribution

225

profiles (at 24 h after injection), the worm-like micelles demonstrated a shorter overall circulation

226

half-life and an enhanced in vivo tumor accumulation as compared to the spherical nanoparticles

227

(Table 1), which possessed comparable overall sizes and which were constructed from the same

228

PEO-b-PCL composition, highlighting the potential for shape alterations to improve nanoparticle

229

delivery. 10 ACS Paragon Plus Environment

Page 10 of 37

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

Nano Letters

230 231

Delivery of PEO-b-PCL-based nanoparticles to autochthonous PDAC tumors in mice

232

Previous work has suggested significant differences in vascularity, blood flow, and

233

chemotherapeutic drug delivery between transplanted and autochthonous models of PDAC8. In

234

particular, autochthonous mouse models that harbor mutations in the proto-oncogene Kras and

235

the tumor suppressor gene p53 generate pancreatic tumors with a dense desmoplastic stroma33,

236

34, 43;

237

and, they exhibit hypoperfusion and reduced drug delivery as compared to allograft models8. As

238

discussed previously (vide supra), the potential for vascularity to affect nanoparticle delivery to

239

pancreatic tumor cells has been explored using xenograft models of human cell lines that induce

240

extensive stroma, suggesting a strong dependence on nanoparticle size. No prior work has

241

examined the effects of size on nanoparticle accumulation in autochthonous Kras;p53 mutant

242

tumors or those of nanoparticle shape on ultimate drug delivery to hypoperfused pancreatic

243

tumors.

they demonstrate the hallmark genetic and histologic features observed in human tumors42;

244 245

We generated Kras;p53 mutant tumors, using a genetically-engineered immunocompetent mouse

246

model of PDAC,8, 32, 33 and administered our PEO-b-PCL-based nanoparticles (of varying size and

247

shape) via IV tail-vein injection. Whole-animal fluorescence imaging by IVIS was used to

248

independently monitor tumor growth (by tomato red fluorescent protein - RFP; see Materials and

249

Methods) and nanoparticle location (by DiR; Figure 4C). 24 h after nanoparticle injection, mice

250

were sacrificed and their organs were harvested to measure the relative ex vivo fluorescence

251

distribution of the tumor cells and the nanoparticles. The harvested pancreas revealed strong

252

RFP (Figure 4D) and DiR signals (Figure 4E), verifying successful delivery of the polymeric

253

nanoparticles to the primary pancreatic tumor(s) in the autochthonous model. Note that

254

normalization of emission intensities where conducted with respect to the organ that displayed

255

the largest value in each data set, which was the pancreas in the case of RFP emission (i.e., the 11 ACS Paragon Plus Environment

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

256

organ of highest PDAC cell localization) and the liver in the case of DiR emission (i.e., the healthy

257

organ of greatest NP accumulation). A high RFP signal was also detected in the dissected

258

intestines and stomach, likely due to the local spread of primary tumor cells. While the 100-nm

259

diameter nanospheres appeared to demonstrate higher tumor accumulation than the more

260

modest accrual levels displayed by the 300-nm diameter nanospheres, the results obtained with

261

either formulation were not significantly different when compared to the relative tumor

262

accumulation levels for the 70 nm-diameter nanospheres and the 100 nm-length worm-like

263

micelles. These data, thus, reveal that nanoparticles as large as 100 nm are capable of readily

264

accumulating in the poorly-permeable pancreatic tumor(s) of the autochthonous model.

265

Moreover, we did not observe a significant advantage of the worm-like micelle shape in improving

266

delivery to these primary pancreatic tumors, differing from reported studies claiming their

267

improved capabilities for solid tumor penetration41.

268 269

To more carefully assess nanoparticle delivery at the cellular level, we analyzed the spatial

270

distribution of DiR fluorescence (which identified nanoparticle locations; white) relative to the

271

tumor cells (seen by RFP imaging; red) in frozen sections of tumors that were excised from treated

272

mice and fixed at 24 h after nanoparticle administration (Figure 4F and Supplementary Figure

273

S8). There were no significant differences between the four nanoparticle formulations with respect

274

to the mean distances between nanoparticles and their nearest tumor cells (Figure 4G). While

275

the overall percentage of the delivered dose was lower, even the 300 nm-diameter nanospheres

276

displayed comparable nanoparticle-to-tumor distances as compared to the smaller-sized

277

particles. While the possibility exists that differences in tumor accumulation might be observed at

278

longer or shorter time points after the administration of different nanoparticle formulations, the

279

results would support that once nanoparticles extravasate within pancreatic tumors, their ultimate

280

penetration distance is independent of their size or shape.

281 12 ACS Paragon Plus Environment

Page 12 of 37

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

Nano Letters

282

Generation of oxaliplatin-loaded nanoparticles

283

We next evaluated the capacity of our nanoparticles to deliver a chemotherapeutic agent to

284

pancreatic tumors. As a proof-of-principle example, we chose oxaliplatin, which is a platinum-

285

containing anticancer agent used in first-line therapy of human PDAC3. To generate an oxaliplatin-

286

loaded nanoparticle, we constructed a novel lipophilic oxaliplatin(IV) prodrug (C 16-oxaliplatin(IV)-

287

C16), where oxaliplatin was oxidized and conjugated to palmitic acid (Figure 1C). The lipophilic

288

prodrug was subsequently encapsulated in the hydrophobic core of PEO-b-PCL-based

289

nanoparticles during their fabrication (Figure 1D and Supplementary Scheme S1). By

290

measuring the relative fluorescence yield and platinum (Pt) content of the final nanoparticle

291

suspensions, we determined that the optimal polymer-to-oxaliplatin (P:O) weight ratio for

292

formation was 100:10, suggesting that 10 wt% loading of nanoparticles with the lipophilic

293

oxaliplatin(IV) prodrug was the most efficient level for incorporation (Figure 5A). DLS

294

measurements revealed that nanoparticles formed at a P:O=100:10 remained ~100 nm in

295

dimension (Figure 5B), which was similar to the results obtained with their unloaded PEO5k-

296

PCL16k-based counterparts. Cryo-TEM images showed that the DiR-containing PEO-b-PCL-

297

based nanoparticles retained a worm-like architecture after oxaliplatin(IV) loading (Figure 5C).

298

Thus, it was determined that the oxaliplatin species could be readily incorporated within the

299

micelles without altering nanoparticle size or shape.

300 301

We next investigated the drug release profiles of the oxaliplatin(IV)-loaded worm-like micelles.

302

PEO-b-PCL-based nanoparticles are known to be stable at neutral pH, while acidic conditions,

303

such as those found within endosomes, facilitate release of their contents36, 38. Under neutral or

304

acidic pH conditions and in the presence of low concentrations of exogenously added glutathione

305

(GSH), platinum (Pt) release remained minimal; at an acidic pH and with higher GSH

306

concentrations, which mimicked conditions known to exist within tumor cells

307

concentrations of free Pt species were detected in nanoparticle suspensions over time (Figure 13 ACS Paragon Plus Environment

48,

increasing

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

308

5D). Pt release from worm-like micelles was, thus, found to be triggered under acidic and

309

reductive in situ conditions, supporting that similar release features could be expected after the

310

rapid uptake of the worm-like micelles within in vivo tumor locations. It should be noted that the

311

shortened circulatory half-life of oxaliplatin(IV)-loaded worm-like micelles would be expected to

312

reduce premature drug release prior to systemic nanoparticle clearance. Given these desirable

313

features, we next examined the in vitro potency of the drug-laden micelles as compared to free

314

oxaliplatin in both murine (B22) and human (8988T) PDAC cell lines (Supplementary Figure

315

S9). From their IC50 values, it was evident that free oxaliplatin and oxaliplatin(IV)-loaded worm-

316

like micelles exhibited similar in vitro antitumor activity; note that cellular incubation with unloaded

317

PEO-b-PCL-based nanoparticles of varying size and shape did not result in comparable cellular

318

toxicity, supporting the biocompatibility of the nanoparticle carrier. Empty nanoparticles that were

319

incubated with B22 and 8999T cells at concentrations that were equivalent to the in vivo dose

320

used for IV tail-vein injection (i.e., NP1: 5.2 mM or 78 ug/uL of PEO-b-PCL copolymer) did not

321

result in significant in vitro cytotoxicity (Supplementary Figures S9D and S9H, respectively).

322 323

In vivo delivery and tumor retention of oxaliplatin(IV)-loaded PEO-b-PCL-based micelles

324

Following administration to nude mice harboring B22 subcutaneous pancreatic tumors, we

325

compared the delivery and retention of our oxaliplatin(IV)-loaded worm-like micelles with

326

equivalent doses of the free drug formulation of unmodified oxaliplatin. Pharmacokinetic analyses

327

of the Pt content of blood samples taken at various time points after IV tail-vein injection of free

328

oxaliplatin revealed its short circulatory half-life (t1/2 = 1.88 h; Supplementary Figure S10A). The

329

circulatory half-life of oxaliplatin was increased by over three-fold (to 6.5 + 0.7 h) when loaded

330

within and delivered by DiR-containing worm-like micelles (as measured by DiR fluorescence;

331

Supplementary Figure S10B). Note that the half-life of the drug-laden worm-like micelles was

332

similar to that of their unloaded counterparts (t1/2 = 6.6 + 1.9 h), suggesting that loading of the Pt

333

species does not slow nor accelerate the circulation time of the nanoparticles in blood. 14 ACS Paragon Plus Environment

Page 14 of 37

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

Nano Letters

334 335

We further examined whether the worm-like micelles could enhance drug delivery to poorly

336

vascularized tumors, using the autochthonous KPC model of PDAC. The Pt content of various

337

organs was determined after IV tail-vein injection of either oxaliplatin(IV)-loaded worm-like

338

micelles (NP-oxaliplatin) or free oxaliplatin. NP-oxaliplatin displayed both a relative (with respect

339

to other organs) and an absolute increase (~7-fold higher amounts) in the levels of Pt

340

accumulation within the pancreas as compared to the free drug formulation at 24 h and as

341

assessed by atomic absorption spectroscopy (AAS) (Figure 5E). Note that the pancreas is the

342

major organ wherein the tumors are located in these mice; hence, it serves as a surrogate to

343

evaluate the extent of intratumoral accumulation. We also measured the amounts of Pt that

344

persisted within the pancreatic tumors (as measured by AAS) of SQ-allografted mice that were

345

administered either NP-oxaliplatin or free oxaliplatin at identical dose equivalents and that were

346

then sacked at similar time intervals. Importantly, the Pt content of the tumors was prolonged after

347

worm-like micelle delivery, exhibiting a retention half-life that was ~50x that of free oxaliplatin,

348

which was otherwise rapidly cleared from the tumor microenvironment (t1/2 = 5.9 h; Figure 5F).

349

Together, these data reveal that the 100 nm-length, PEO-b-PCL-based worm-like micelles are

350

capable of delivering drugs to poorly-permeable pancreatic tumors with improved biodistribution

351

and prolonged tumor retention as compared to free drug formulations.

352 353

As nab-paclitaxel is an established first-line therapy, and given the recent approval of an

354

irinotecan-containing

355

considerable interest in pancreatic cancer. To date, however, there have been few published

356

studies that have sought to examine the effects of nanoparticle size on tumor penetration in

357

murine models of PDAC. Using SQ models, Cabral et al.28 and Wang et al.31 have reported that

358

nanoparticles that are sub-30 nm in diameter augment tumor uptake while those that are larger

359

than 100 nm exhibit little to no accumulation, presumably due to size-dependent limitations to

nano-liposome17,

novel

nanoparticle

15 ACS Paragon Plus Environment

therapeutics

are

garnering

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

360

nanoparticle extravasation. In contrast to these findings and by using a similar SQ model of

361

PDAC, Kobes et al. demonstrated significant tumor uptake for polymeric nanoparticles that were

362

greater than 450 nm in size49. Additional investigations have shown that ultra-small nanoparticles

363

(< 10 nm in size) demonstrate improved tumor uptake in similar models15, 50.

364 365

A few studies have been performed in more advanced model systems, including in patient-derived

366

xenograft (PDX) and in syngeneic orthotopic transplant models of PDAC; they have demonstrated

367

enhanced tumor uptake of inorganic nanoparticles that range in size from 40 to 125 nm51, 52. These

368

findings, however, contradict those of another study that has compared the sizes of gold

369

nanoparticles in a different syngeneic transplant model, determining that nanoparticles that are