development and intratumoral distribution of block copolymer micelles … · 2014-07-01 · iii...

159
Development and Intratumoral Distribution of Block Copolymer Micelles as Nanomedicines for the Targeted Delivery of Chemotherapy to Solid Tumors by Andrew S. Mikhail A thesis submitted in conformity with the requirements for the degree of doctor of philosophy Department of Pharmaceutical Sciences and the Institute of Biomaterials and Biomedical Engineering University of Toronto © Copyright by Andrew S. Mikhail 2013

Upload: others

Post on 11-Mar-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

Development and Intratumoral Distribution of Block Copolymer Micelles as Nanomedicines for the Targeted

Delivery of Chemotherapy to Solid Tumors

by

Andrew S. Mikhail

A thesis submitted in conformity with the requirements for the degree of doctor of philosophy

Department of Pharmaceutical Sciences and the Institute of Biomaterials and Biomedical Engineering

University of Toronto

© Copyright by Andrew S. Mikhail 2013

Page 2: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

ii

Development and Intratumoral Distribution of Block Copolymer

Micelles as Nanomedicines for the Targeted Delivery of

Chemotherapy to Solid Tumors

Andrew S. Mikhail

Doctor of philosophy

Department of Pharmaceutical Sciences and the Institute of Biomaterials and Biomedical Engineering

University of Toronto

2013

Abstract

Recent advancements in pharmaceutical technology based on principles of

nanotechnology, polymer chemistry, and biomedical engineering have resulted in the

creation of novel drug delivery systems with the potential to revolutionize current

strategies in cancer chemotherapy. In oncology, realization of significant improvements

in therapeutic efficacy requires minimization of drug exposure to healthy tissues and

concentration of the drug within the tumor. As such, encapsulation of chemotherapeutic

agents inside nanoparticles capable of enhancing tumor-targeted drug delivery is a

particularly promising innovation. Yet, initial investigations into the intratumoral fate of

nanomedicines have suggested that they may be heterogeneously distributed and

achieve limited access to cancer cells located distant from the tumor vasculature. As

such, uncovering the determinants of nanoparticle transport at the intratumoral level is

Page 3: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

iii

critical to the development of optimized delivery vehicles capable of fully exploiting the

therapeutic potential of nanomedicines.

In this work, the chemotherapeutic agent, docetaxel (DTX), was incorporated into nano-

sized, biocompatible PEG-b-PCL block copolymer micelles (BCMs). Encapsulation of

DTX in micelles via chemical conjugation or physical entrapment resulted in a dramatic

increase in drug solubility and customizable drug release rate. The use of multicellular

tumor spheroids (MCTS) was established as a viable platform for assessing the efficacy

and tumor tissue penetration of nanomedicines in vitro. A series of complementary

assays was validated for analysis of DTX-loaded micelle (BCM+DTX) toxicity in

monolayer and spheroid cultures relative to Taxotere®. Cells cultured as spheroids were

less responsive to treatment relative to monolayer cultures due to mechanisms of drug

resistance associated with structural and microenvironmental properties of the 3-D

tissue. Computational, image-based methodologies were used to assess the spatial and

temporal penetration of BCMs in spheroids and corresponding human tumor xenografts.

Using this approach, the tumor penetration of micelles was found to be nanoparticle-

size-, tumor tissue type- and time- dependent. Furthermore, spheroids were found to be

a valuable platform for the prediction of trends in nanoparticle transport in vivo. Overall,

the results reported herein serve to demonstrate important determinants of nanoparticle

intratumoral transport and to establish computational in vitro and in vivo methodologies

for the rational design and optimization of nanomedicines.

Page 4: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

iv

Acknowledgments

Firstly I would like to thank my supervisor Dr. Christine Allen for the opportunity to

undertake a doctoral research project in her lab. It has been a privilege and a pleasure

to work under her guidance and I am truly grateful for her continued support and belief

in my abilities. I am very lucky to have worked under the leadership of such a caring

individual and inspiring researcher. She has made this experience both enjoyable and

deeply rewarding and I would like to sincerely thank her for all that she has done for me.

I am also deeply appreciative of my advisory committee members: Dr. Yu-Ling

Cheng, Dr. Robert Macgregor and Dr. Ian Tannock. Receiving constructive ideas and

critical input from such esteemed investigators was truly one of the greatest privileges of

being a doctoral student. I extend to them my deepest gratitude for finding time in their

busy schedules to support my research. I valued their input tremendously. In particular, I

am very grateful for the scientific guidance provided by Dr. Tannock and the members

of his laboratory and the advice and support of Dr. Macgregor.

I would also like to thank all of my colleagues who have helped me along the way

and who have made this experience enjoyable. A special thanks to Sina Eetezadi for his

efforts in what turned out to be a very productive, and fun, collaboration. Thank you also

to Dr. David Dubins for making my experience as a teaching assistant so enjoyable. I’d

also like to acknowledge Dr. Kazunori Kataoka and Dr. Horacio Cabral for providing me

the opportunity to work at the University of Tokyo.

I am also blessed with great friends in my life who have supported me and with

whom I share so many great memories from this period of my life. There are too many

to list here, but I would be remiss to not acknowledge my great friend Derek Watkins,

who is like a brother to me. Words are not enough to thank you for your support. As this

journey comes to a close and a new one begins, I humbly remember friends of mine

and of my family who have been affected by or lost their lives to cancer.

Finally, with all of my heart, I would like to thank my family for everything they

have done for me. Their unconditional support through thick and thin is what made this

accomplishment possible. To my parents I am forever indebted for all that they have

sacrificed and put second to my well-being and happiness. None of this was possible

without them. I dedicate this accomplishment to my father, Shaheer Mikhail, who is my

greatest mentor and who inspired me to pursue higher learning.

Page 5: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

v

Table of Contents

Acknowledgments ........................................................................................................ iv

Table of Contents ........................................................................................................... v

List of Tables .................................................................................................................. x

List of Figures ............................................................................................................... xi

List of Abbreviations .................................................................................................. xiv

CHAPTER 1. Introduction .............................................................................................. 1

1.1 Overview ............................................................................................................... 3

1.2 Whole body transport and macrodistribution ......................................................... 5

1.3 Thermodynamic and kinetic stability of block copolymer micelles in vivo .............. 6

1.4 Drug retention in vivo ............................................................................................ 7

1.5 Pharmacokinetics and whole body distribution of block copolymer micelles ....... 10

1.6 Influence of blood proteins and MPS clearance .................................................. 11

1.7 Passive tumor accumulation via the EPR effect .................................................. 12

1.8 Tumor physiology and its role in nanoparticle delivery ........................................ 13

1.8.1 Vascular density (density of functional vessels) ....................................... 13

1.8.2 Vascular permeability ............................................................................... 13

1.8.3 Interstitial fluid pressure and transport in the interstitium .......................... 14

1.9 Active targeting ................................................................................................... 16

1.10 Microdistribution: transport within an organ or tissue…………………………….17

1.11 3-D tissue culture and the multicellular tumor spheroid model .......................... 22

1.12 Transport at the cellular level……………………………………………………….23

1.13 Docetaxel .......................................................................................................... 25

1.14 . Summary……………………………………………………………………………...26

1.15 Rationale………………………………………………………………………………27

Page 6: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

vi

1.16 Hypotheses…………………………………………………………………………...28

1.17 Objectives……………………………………………………………………………..28

1.18 Overview of Thesis Chapters……………………………………………………….29

CHAPTER 2. Poly(ethylene glycol)-b-poly(ε-caprolactone) Micelles Containing Chemically Conjugated and Physically Entrapped Docetaxel: Synthesis, Characterization, and the Influence of the Drug on Micelle Morphology ........... 30

2.1 Abstract ............................................................................................................... 32

2.2 Introduction ......................................................................................................... 33

2.3 Experimental Section .......................................................................................... 34

2.3.1 Materials ................................................................................................... 34

2.3.2 Synthesis of CH3O-PEG-b-PCL (PEG-b-PCL) Copolymers ..................... 34

2.3.3 Addition of Carboxylic Acid Group to PCL Block of PEG-b-PCL Copolymers .............................................................................................. 35

2.3.4 Conjugation of DTX to PEG-b-PCL-COOH Copolymers .......................... 35

2.3.5 Characterization of Copolymers and Copolymer-Drug Conjugates .......... 36

2.3.6 Micellization and Measurement of Critical Micelle Concentration (CMC) . 36

2.3.7 Transmission Electron Microscopy (TEM) Analysis .................................. 37

2.3.8 Determination of Micelle Size ................................................................... 37

2.3.9 Evaluation of Drug Loading and Release ................................................. 38

2.3.10 Statistical Analysis .................................................................................... 38

2.4 Results and Discussion ....................................................................................... 38

2.4.1 Synthesis of PEG-b-PCL Copolymer and PEG-b-PCL-DTX Copolymer-Drug Conjugate ........................................................................................ 38

2.4.2 Physico-Chemical Properties of PEG-b-PCL and PEG-b-PCL-DTX Copolymer and Copolymer-Drug Conjugates ........................................... 41

2.4.3 Characterization of the Copolymer-Drug Conjugate and Drug-Loaded Micelles. ................................................................................................... 43

2.4.4 Micelle Morphology ................................................................................... 43

2.4.5 Drug Loading ............................................................................................ 46

Page 7: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

vii

2.4.6 Drug Release............................................................................................ 47

2.5 Conclusions ......................................................................................................... 49

2.6 Acknowledgements ............................................................................................. 50

CHAPTER 3. Cytotoxicity and Growth Inhibitory Effect of Docetaxel-loaded Block Copolymer Micelles and Taxotere® in Monolayer and Spheroid Cultures .......... 51

3.1 Abstract ............................................................................................................... 53

3.2 Introduction ......................................................................................................... 54

3.3 Materials and Methods ........................................................................................ 56

3.3.1 Materials ................................................................................................... 56

3.3.2 Synthesis of CH3O-PEG-b-PCL (PEG-b-PCL) and Alexa Fluor 488-PEG-b-PCL (AF488-PEG-b-PCL) copolymers .................................................. 56

3.3.3 Preparation and characterization of BCMs ............................................... 57

3.3.4 Sizing of BCM+DTX ................................................................................. 57

3.3.5 Transmission Electron Microscopy (TEM) ................................................ 57

3.3.6 Drug Release............................................................................................ 57

3.3.7 Tissue culture and growth of MCTS ......................................................... 58

3.3.8 Immunohistochemical analysis of MCTS .................................................. 58

3.3.9 Measurement of MCTS Growth ................................................................ 59

3.3.10 Cytotoxicity in Monolayer and Spheroid Tissue Culture ........................... 59

3.3.11 Growth Inhibition of MCTS ....................................................................... 60

3.3.12 Clonogenic Survival Assay ....................................................................... 60

3.4 Results ................................................................................................................ 61

3.4.1 Characterization of BCM+DTX ................................................................. 61

3.4.2 Growth of MCTS ....................................................................................... 62

3.4.3 Cytotoxicity in monolayer and MCTS culture ............................................ 63

3.4.4 Inhibition of MCTS growth ........................................................................ 64

3.4.5 Immunohistochemistry .............................................................................. 66

Page 8: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

viii

3.4.6 Clonogenic Survival .................................................................................. 68

3.5 Discussion ........................................................................................................... 69

3.6 Conclusions ......................................................................................................... 73

CHAPTER 4. Image-based Analysis of the Time-dependent Penetration of Polymeric Micelles in Multicellular Tumor Spheroids and Tumor Xenografts ... 74

4.1 Abstract ............................................................................................................... 76

4.2 Introduction ......................................................................................................... 77

4.3 Methods .............................................................................................................. 79

4.3.1 Materials ................................................................................................... 79

4.3.2 Synthesis of CH3O-PEG-b-PCL (PEG-b-PCL) and Alexa Fluor® 488-PEG-b-PCL (AF488-PEG-b-PCL) copolymers .................................................. 79

4.3.3 Preparation and characterization of BCMs ............................................... 80

4.3.4 Transmission electron microscopy (TEM) ................................................ 80

4.3.5 Tissue culture and growth of MCTS ......................................................... 81

4.3.6 Penetration of BCMs in MCTS ................................................................. 81

4.3.7 Animals and growth of tumor xenografts .................................................. 81

4.3.8 Penetration of BCMs in tumor xenografts ................................................. 82

4.3.9 Computational analysis of BCM distribution in tumor sections ................. 83

4.4 Results ................................................................................................................ 83

4.4.1 Synthesis and characterization of PEG-b-PCL copolymers and BCMs .... 83

4.4.2 Growth of MCTS ....................................................................................... 84

4.4.3 Penetration of BCMs in MCTS ................................................................. 85

4.4.4 Penetration of BCMs in tumor xenografts ................................................. 86

4.5 Discussion ........................................................................................................... 92

4.6 Conclusions ......................................................................................................... 96

4.7 Acknowledgements ............................................................................................. 97

Page 9: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

ix

CHAPTER 5. Summary and Future Directions .......................................................... 98

5.1 Conclusions and Summary of Findings ............................................................... 98

5.2 Future Directions ............................................................................................... 100

5.2.1 Improving drug retention and tumor-specific drug delivery ..................... 100

5.2.2 Therapeutic efficacy and the intratumoral fate of DTX ............................ 101

5.2.3 Overcoming transport barriers by modulating the tumor microenvironment…………………………………………………………….102

References.................................................................................................................. 104

Appendix 1: Chapter 3 Supplemental Data .............................................................. 140

Appendix 2: Chapter 4 Supplemental Data .............................................................. 143

Page 10: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

x

List of Tables

Table 2.1 Melting Temperature (Tm), Critical Micelle Concentration (CMC), Morphology,

Diameter, Molecular Weight (Mn), and Polydispersity (Mw/Mn) of Copolymers

and Copolymer-Drug Conjugate. ........................................................................... 40

Table 2.2. GPC Retention Times (Rt) of the Copolymers and Copolymer-Drug

Conjugate .................................................................................................................. 41

Table 4.1. Composition of block copolymers and properties of BCMs. ............................. 84

Page 11: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

xi

List of Figures

Figure 1.1. Micelle transport at the whole body, tissue and cellular levels. ........................ 5

Figure 1.2. Molecular simulation of DTX surrounded by PEG-b-PCL ................................. 9

Figure 1.3. Biodistribution and intratumoral distribution of 111In-PEG-b-PCL micelles. .. 21

Figure 1.4. Cellular uptake of EGF-conjugated PEG-b-PVL micelles . ............................. 19

Figure 2.1. Synthesis of PEG-b-PCL, PEG-b-PCL-COOH, and PEG-b-PCL-DTX ......... 39

Figure 2.2. 1H NMR of PEG-b-PCL copolymers and copolymer-DTX conjugates. ......... 40

Figure 2.3. GPC chromatograms of copolymers and copolymer-DTX conjugates.. ....... 41

Figure 2.4. Diameter and stability of PEG-b-PCL-DTX micelles . ...................................... 42

Figure 2.5 TEM images showing the morphology of micelles containing physically

and/or chemically conjugated docetaxel in different molar ratios. ................. 45

Figure 2.6. Drug loading of BCMs by physical entrapment of DTX at various copolymer

and copolymer-drug conjugate concentrations………………………….....…47

Figure 2.7. Release of chemically conjugated DTX from PEG-b-PCL-DTX micelles. .... 48

Figure 2.8. Release of physically entrapped DTX from PEG2000-b-PCL1000 BCMs and

BCM-DTX................................................................................................................ 49

Figure 3.1. 3-D cultures as an intermediary platform between monolayer cultures and

animal models ........................................................................................................ 55

Figure 3.2. Schematic representation of assays used for analysis of formulation efficacy

in spheroids. ........................................................................................................... 56

Figure 3.3. TEM and size distribution as determined by DLS of BCM+DTX... ................. 61

Figure 3.4. Release of DTX from BCMs (PEG5000-b-PCL5000) and Taxotere® .................. 62

Page 12: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

xii

Figure 3.5. Cell packing density and growth of HeLa and HT29 MCTS ............................ 62

Figure 3.6. Cytotoxicity of Taxotere® and BCM+DTX and BCMs in monolayer and

MCTS cultures as measured using the APH assay. ........................................ 64

Figure 3.7. Growth inhibition of HeLa and HT29 MCTS by BCM+DTX and Taxotere® .. 65

Figure 3.8. Immunohistochemical staining of HeLa and HT29 MCTS cross-sections with

H&E, Ki67 (proliferation) and EF5 (hypoxia). .................................................... 66

Figure 3.9. Ki67 positive signal distribution in HeLa and HT29 MCTS .............................. 67

Figure 3.10. Spatial distribution of features of HeLa and HT29 MCTS

microenvironments. ............................................................................................... 67

Figure 3.11. Clonogenic survival of HeLa and HT29 cells following treatment with

BCM+DTX or Taxotere® as monolayers, disaggregated MCTS and intact

MCTS. ..................................................................................................................... 68

Figure 4.1. Schematic representation of tumor and MCTS cross-sections ...................... 78

Figure 4.2. TEM images of PEG5000-b-PCL5000 and PEG2000-b-PCL1000 BCMs. .............. 84

Figure 4.3. Schematic representation of MCTS growth using the liquid overlay

method. ................................................................................................................... 85

Figure 4.4. H&E immunohistochemical staining of HT29 and HeLa xenografts and

MCTS ...................................................................................................................... 86

Figure 4.5. BCM penetration in HeLa and HT29 MCTS following incubation with BCM-

15 and BCM-55 for 1 h and 24 h ......................................................................... 87

Figure 4.6. Area-normalized distribution of BCM-15 and 55 HeLa and HT29 MCTS

following 1 h and 24 h exposures. ...................................................................... 88

Figure 4.7. Representative computational image-based analysis of BCM penetration in

tumor xenografts .................................................................................................... 89

Page 13: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

xiii

Figure 4.8. Interstitial penetration of BCMs relative to tumor blood vesselsin HeLa and

HT29 tumor xenografts. ........................................................................................ 90

Figure 4.9. Mean fluorescence pixel intensities of BCM-15 and BCM-55 at 100 µm from

the nearest blood vessel in tumor xenografts at 1, 6, and 24 h p.i. ............... 91

Figure 4.10. Potential influence of micelles on the intratumoral distribution of the drug. 96

Page 14: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

xiv

List of Abbreviations

APH Acid phosphatase assay

AUC Area under the curve

BCM(s) Block copolymer micelle(s)

BCM-15 15 nm block copolymer micelles

BCM-55 55 nm block copolymer micelles

CL Caprolactone

CMC Critical micelle concentration

Da Daltons

DLS Dynamic light scattering

DOX Doxorubicin (adriamycin)

DTX Docetaxel

Glu Glutamic acid

GPC Gel permeation chromatography

HLB Hydrophilic-lipophilic balance

HPLC High-performance liquid chromatography

IC50 Half maximal inhibitory concentration

i.v. Intravenous

MCTS Multicellular tumor spheroids

MDR Multi-drug resistant

Mn Number average molecular weight

mPEG Methoxy-poly(ethylene oxide)

MPS Mononuclear phagocytic system

Mw Weight average molecular weight

NMR Nuclear magnetic resonance

PBS Phosphate buffered saline

PCL Poly(ε-caprolactone)

pKb Base dissociation constant

PPO Poly(propylene oxide)

PTX Paclitaxel

PAsp Poly(aspartic acid)

Page 15: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

xv

PEG Poly(ethylene glycol)

P85 Pluronic® P85

PK Pharmacokinetics

SF Surviving fraction

t1/2,α Distribution half-life

t1/2,β Elimination half-life

TAX Taxotere®

TEM Transmission electron microscopy

Tyr Tyrosine

Page 16: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

1

1 Chapter 1. Introduction

Chapter 1. Introduction

Page 17: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

2

This chapter is a reprint with modification of: “Andrew S. Mikhail and Christine Allen.

Block copolymer micelles for delivery of cancer therapy: Transport at the whole body,

tissue and cellular levels.” Journal of Controlled Release 138 (2009) 214–223; with

permission from Elsevier.

This chapter was written by AS. Mikhail with editing by Dr. C. Allen.

Page 18: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

3

1.1 Overview

The use of block copolymer micelles (BCMs) for the targeted delivery of

chemotherapeutics has proven to be a promising approach for improving the

therapeutic efficacy of drug therapy. Acceleration of the translation of nanomedicines

from the fundamental stages of pre-clinical development to clinical use requires a

greater understanding of the transport mechanisms that influence the fate of nano-

carriers at the whole body, tissue, and cellular levels (Figure 1.1). New imaging-based

techniques to evaluate the intratumoral distribution and tumor penetration of BCMs and

other nanosystems have the potential to revolutionize our understanding and current

approach to drug delivery in this field.

The therapeutic efficacy of conventional chemotherapeutic agents is often limited

by the drug's poor aqueous solubility and systemic toxicity. Side effects resulting from

toxicity to healthy tissues commonly constrain the dose and frequency of chemotherapy

and thus compromise the effectiveness of treatment [1]. Although the use of small

molecule surfactants as excipients in commercial formulations has helped to improve

the solubility of hydrophobic drugs, these chemicals are also intrinsically toxic [2].

Incorporation of drugs within biocompatible and/or biodegradable block copolymer

micelles (BCMs) has been shown to reduce systemic toxicity while increasing drug

solubility and site-specific tumor accumulation [3–7].

BCMs are self-assembled nano-sized aggregates of amphiphilic copolymers. The

hydrophobic micelle core, which acts as a drug reservoir, is surrounded by a hydrophilic

corona that provides a protective interface between the core and the external

environment [8, 9]. A number of other polymer and lipid-based colloidal carriers have

also been explored for improving the solubility and efficacy of various drugs [10–13].

However, an important advantage of micellar drug delivery vehicles is the unique ability

to customize both their core and coronal properties. Alterations to the composition of the

constituent copolymers can influence important performance related parameters

including micelle size, core-drug compatibility, drug loading capacity, drug release

kinetics and stability thus permitting the manipulation of the encapsulated drug's

pharmacokinetic profile and tissue distribution [14–16]. As a result of growing interest in

this promising drug delivery platform over the past two decades, several BCM-based

Page 19: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

4

drug formulations are now in various stages of clinical evaluation with many others in

pre-clinical development [17–21]. The current status of micellar drug delivery vehicles in

clinical trial development has recently been reviewed by Matsumura [22] and Yokoyama

[23]. These achievements along with a survey of the literature using the Scopus™

database (Elsevier B.V.) that reveals more than 10000 publications on BCMs for drug

delivery necessitates reflection on our current state of knowledge and the next steps

required to truly achieve revolutionary advances in this field.

The concept of the “magic bullet”, proposed by Paul Ehrlich in 1906, envisioned a

delivery system that could “bring therapeutically active groups to the organ in question”

[24]. Due to the toxic and non-specific nature of chemotherapeutic agents, this concept

is particularly relevant to the field of cancer therapy. Certainly in oncology,

achievements of the most significant improvements in therapeutic efficacy require

minimization of drug exposure to healthy tissues and concentration of the drug at the

tumor site. In essence, realization of Ehrlich's vision would require the design of delivery

vehicles capable of carrying large quantities of poorly soluble chemotherapeutic drugs

with drug release only occurring once the vehicle has reached the target tumor.

Although the emergence of nanomedicines has served to reduce treatment-

related side effects by improving the tumor-specific delivery of chemotherapy,

corresponding improvements in therapeutic efficacy have often been modest. One

important reason for this is that they may be poorly distributed in tumors resulting in

limited exposure of cancer cells to therapeutically relevant concentrations of drug. Many

systemically administered therapeutic agents remain highly concentrated in the

periphery of the tumor and are confined to perivascular tissue due to their

heterogeneous intratumoral distribution and limited penetration through the tumor

interstitial space [25]. Enlarged intervascular distances become host to gradients in drug

concentration that may promote drug resistance and spare distant cells from treatment

that can subsequently repopulate the tumor [26]. Therapeutic efficacy is further impeded

by resistance associated with properties of the tumor microenvironment including the

presence of gradients in cell proliferation in which quiescent cells are less sensitive to

therapy, and regions of hypoxia in which cellular changes can result in drug resistance

and a more aggressive phenotype [27]. As such, there is an increasing demand for the

establishment of in vitro tissue models that more accurately replicate the complex

Page 20: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

5

transport barriers and resistance mechanisms associated with the microenvironment of

native tumors. Three dimensional (3-D) tissue cultures, such as multicellular tumor

spheroids (MCTS), have the potential to bridge the gap between traditional in vitro and

in vivo models while providing a high-throughput platform for the rapid assessment of

therapeutic candidates [28–30]. Moreover, this approach is highly relevant in the field of

nanomedicine where the rational design and optimization of nanocarrier properties is

critical in expediting the translation of advanced drug delivery systems from conception

to clinical application.

Figure 1.1. Block copolymer micelle transport at the whole body, tissue and cellular

levels. Micelles circulate throughout the body and accumulate in tumor tissues where

they may be taken up by cancer cells.

1.2 Whole body transport and macrodistribution

Encapsulation of a drug in BCMs can increase the drug's circulation lifetime following

systemic administration, allow for site-specific drug delivery, and reduce undesirable

drug distribution to healthy tissues and organs. A thorough understanding of how the

physico-chemical properties of micelles influence the pharmacokinetics (PK) and

biodistribution of encapsulated drugs in vivo is critical to the design of new formulations

with enhanced therapeutic efficacy. It is important to note that the ability of micelle

Endosomal escape

Micelle disassembly

Endocytosis/receptor

mediated cell uptake

Extracellular drug release

Page 21: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

6

delivery vehicles to alter the in vivo disposition and distribution of a drug is dependent

on the extent to which the drug remains encapsulated in the micelle following

administration. Micelles that function primarily as solubilizers increase the aqueous

solubility of the drug but have poor stability and/or limited drug retention following i.v.

injection. As a result, the pharmacokinetic profile and biodistribution of the drug remain

largely unchanged when compared to conventional formulations [31, 32]. In contrast,

the longer the drug remains encapsulated within the micelle, the greater the influence of

the micelle carrier on the drug's fate in vivo [33–35]. In general, the ability of a micelle

delivery system to function as a solubilizer or a true carrier depends on the stability of

the micelles in vivo and drug retention in the presence of blood components.

1.3 Thermodynamic and kinetic stability of block copolymer micelles in vivo

Micelle stability is a function of both thermodynamic and kinetic parameters and has

been discussed extensively elsewhere [15]. Briefly, the concentration at which

amphiphilic copolymer chains are thermodynamically driven to self-assemble in solution

to form micelles is known as the critical micelle concentration (CMC). Above the CMC,

the copolymer exists as micelles in equilibrium with a small population of single chains

while below the CMC, the copolymer exists in solution as single copolymer chains

referred to as unimers. Therefore, the dilution that occurs following intravenous (i.v.)

administration of micelles can present a significant challenge in terms of achieving

adequate thermodynamic stability of the delivery system in vivo. However, it has been

demonstrated that micelles can remain kinetically stable for extended periods of time

below the CMC depending on several properties of the core-forming block including its

glass transition temperature and melting temperature (if any) [36, 37].

To date, radiolabeling of micelles has been the most common method used to

investigate the influence of micelle stability on copolymer fate in vivo, though the

number of studies remains limited and merits further investigation [32, 38–40].

Batrakova et al. recently examined the pharmacokinetics and biodistribution of

radioactively labeled Pluronic® P85 (P85) administered intravenously to mice at

copolymer concentrations that, following dilution in vivo, were above and below the

Page 22: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

7

CMC [41]. The results revealed that the circulation half-life of P85 (t1/2=60–90 h) was

dependent on the concentration of copolymer administered and thus the aggregation

state of the copolymers in vivo. The concentration of copolymer also influenced

copolymer uptake in the liver with the lowest AUC/dose ratio in the liver occurring

following administration of an intermediate copolymer concentration. Therefore, the

presence of Pluronic® copolymers as either micelles or unimers in vivo plays an

important role in determining their biodistribution. In another study, Liu et al. explored

the influence of micelle stability on the pharmacokinetics of radiolabeled PEG-b-poly(ε-

caprolactone) (PEG-b-PCL) block copolymers above and below the CMC [37]. The

pharmacokinetic profile obtained for copolymer administered i.v. below the CMC

revealed rapid elimination of the copolymer from the circulation (t1/2β=10.2 h) and a high

volume of distribution (Vd=7.6 mL). The plasma clearance profile for copolymer

administered above the CMC followed a two compartment model with a rapid

distribution phase (t1/2α=1.5 h) followed by a relatively slow elimination phase (t1/2β=30.8

h). In addition, administration of copolymer as intact micelles, yet at a concentration that

following dilution in the plasma would reach levels below the CMC (i.e.

thermodynamically unstable micelles), resulted in a more rapid distribution phase

(t1/2α=0.8 h) and elimination phase (t1/2β=16.7 h) when compared to micelles

administered above the CMC. The presence of intact micelles was observed up to 24 h

postadministration of both thermodynamically stable and unstable micelles confirming a

high degree of kinetic stability for this system likely imparted by the hydrophobic and

semi-crystalline PCL core. This study demonstrates that the selection of a core-forming

block that provides a high degree of kinetic stability is an important component in the

preparation of micelles that act as true drug carriers and remain intact for prolonged

periods in vivo. However, at high drug to material ratios the drug itself may become an

influential component of the micelle core and alter the nature and/or state of the core as

well as formulation stability.

1.4 Drug retention in vivo

Retention of hydrophobic drugs within the core of stable BCMs is critical to the

development of true site-specific delivery vehicles. The capacity of a micellar system to

solubilize and retain a drug within its core is determined by the compatibility between

Page 23: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

8

the drug and the core-forming polymer block. The miscibility and/or degree of interaction

between the polymer and the drug affects many of the performance related

characteristics of the delivery system including stability, drug loading capacity, drug

loading efficiency, and drug release kinetics [42].

Due to the vast array of drugs each with its own unique chemical structure, no

single core forming block will be suitable for maximizing the loading and retention of all

drugs. Therefore, it is unlikely that a single micelle system can serve as a universal

delivery vehicle for all drugs. Instead, a drug should be matched with compatible

copolymer micelles containing a core–forming block tailored to the specific chemical

properties of the drug [43]. For example, Carstens et al. found that the presence of an

aromatic end group on the core-forming block improved the loading and stability of

taxanes [44]. Paclitaxel (PTX) and docetaxel (DTX) were loaded into mPEG750-b-

oligo(ε-caprolactone)5 micelles containing a hydroxyl (OH), benzoyl (Bz) or naphthoyl

(Np) group at the terminus of the polycaprolactone block. Micelles formed from

copolymers with a hydroxyl terminated core-forming block and encapsulating PTX or

DTX were the least stable and demonstrated rapid drug release compared to micelles

containing aromatic end groups. The improved performance of the copolymer micelles

containing the aromatic end groups was attributed to π–π interactions between the

taxanes and the micelle core resulting in improved core-drug compatibility. Similarly, a

micelle formulation of PTX, known as NK105, that has now reached Phase II clinical

development is composed of copolymers with PEG as the hydrophilic block and

modified poly(aspartic acid) (PAsp) as the core-forming block [45]. The PAsp block is

conjugated with 4-phenyl-1-butanol to increase core-drug compatibility and as a result,

the micelles have a drug loading capacity of 23% (w/w). Alternatively core-drug

compatibility may be improved by covalent attachment of drug molecules to the core-

forming block of the copolymer. This principle was demonstrated by Yokoyama et al.

who found that the conjugation of doxorubicin to the core forming block of (PEG-b-

P(Asp)) micelles led to greater physical encapsulation of doxorubicin in comparison to

micelles formed from the PEG-b-P(Asp) copolymer alone [46].

In order to expedite the selection of highly compatible polymer-drug pairs,

Dwan'Isa et al. demonstrated the ability to use predictive computational approaches for

estimating drug solubility and compatibility in hydrated copolymer micelles [47].

Page 24: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

9

Copolymers of PEG and random copolyesters of ε-caprolactone (CL) and trimethylene

carbonate (TMC) were considered for the assessment of polymer-drug compatibility

using the Flory-Huggins interaction parameter (χsp). A comparison of the experimental

solubilities for 8 model drugs in 10% w/v PEG-b-P(CL-co-TMC) micelle solutions with

the predicted polymer-drug compatibilities demonstrated good agreement between the

experimental and theoretical results. This technique may offer a promising approach for

the rapid screening of drugs suitable for delivery by a specific micelle system or,

conversely, of polymers suitable for formulation of a specific drug. As illustrated in

Figure 1.2, the application of such theoretical or computational models to formulation

design may eliminate the trial and error approach that is commonly used to select

suitable material-drug pairs [42, 48].

Figure 1.2. Docetaxel molecule (yellow) surrounded by PCL2-b-PEG19 copolymer (PCL

in white, PEG in blue). Oxygen atoms are in red. Potential hydrogen bond interaction is

indicated by a dashed green line.

Page 25: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

10

1.5 Pharmacokinetics and whole body distribution of block copolymer micelles

The PK and biodistribution of BCMs can be influenced by many factors including the

chemical nature of the copolymer materials (i.e. nature of hydrophobic and hydrophilic

blocks), characteristics of the copolymer (e.g. polydispersity, CMC), their physico-

chemical characteristics (e.g. size, size distribution, morphology, surface charge) and

stability [14–16, 49]. To some extent, variations in copolymer properties and preparation

as well as differences between animal models make it challenging to identify the

underlying criteria that may be used to predict the biodistribution of all micelle systems

[50]. Changes to any number of these properties can significantly alter the in vivo

performance of BCMs. Yamamoto et al. demonstrated the potential for long circulating

PEG-b-poly(D,L-lactide) (PEG-b-PDLLA) copolymer micelles by introducing a slight

anionic charge on the micelle surface through the conjugation of charged peptides and

selection of an appropriate micelle size with a narrow size distribution (diameter ≈30

nm) [51]. The pharmacokinetic profiles of 125I labeled Tyr and Tyr-Glu-PEG-b-PDLLA

micelles in mice were biphasic with elimination half-lives (T1/2) of 18.8 h and 17.7 h,

respectively. The biodistribution of micelles containing an anionic charge (Tyr-Glu)

displayed a significantly lower uptake into the liver and spleen when compared to

neutral Tyr-conjugated micelles suggesting the importance of the slight anionic surface

charge on non-specific organ uptake. Most notably, 25% of the total injected copolymer

dose, for both the 125I labeled Tyr and Tyr-Glu-PEG-b-PDLLA micelle formulations

remained in circulation after 24 h and only 9% in the liver and spleen compared to ~20–

30% for similar systems [52, 53]. The copolymers used in this study were synthesized

by anionic ring opening polymerization resulting in materials with a narrow

polydispersity (Mw/Mn=1.10) consequently yielding micelles having a narrow and

unimodal size distribution. This study highlights the need to prepare or select the

copolymers to be used as building blocks of micelle systems with care in order to

ensure the micelles have a narrow, monomodal size distribution and are resistant to

self-aggregation.

Page 26: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

11

1.6 Influence of blood proteins and MPS clearance

Immediately following intravenous injection, various plasma proteins may adhere to the

delivery vehicle in a process known as opsonization. Not only may opsonization lead to

the extraction of the drug from the micelle [54], but it may also result in the elimination of

the micelles by the mononuclear phagocytic system (MPS). The MPS is comprised

primarily of the Kupffer cells and hepatocytes of the liver and the macrophages of the

spleen. These phagocytotic cells have the ability to rapidly remove micelles from the

bloodstream by recognizing specific proteins (opsonins) bound to their surface. The

susceptibility of circulating polymeric micelles to clearance via the MPS is determined

primarily by the size and surface properties of the micelle that influence the extent of

opsonization. In general, the smaller the circulating colloidal carrier, the greater is its

accumulation and uptake in the liver by hepatocytes likely due to the diameter of the

sinusoidal endothelial fenestrations which range from 100 to 150 nm [55–57]. Larger

particles (>200 nm) or particle aggregates may be removed from the circulation by the

spleen [12].

The properties of the corona-forming block also strongly influence the biodistribution

and clearance of BCMs. Various properties of the corona including the thickness,

density, charge, presence of functional groups, and surface bound targeting moieties

may impact the extent of opsonization and clearance. Presence of a non-ionic, steric

stabilizing hydrophilic polymer, such as polyethylene glycol (PEG) [58], as the corona-

forming block has been shown to prolong the circulation time of BCMs by avoiding

opsonization and subsequent clearance by the MPS [52, 59, 60]. PEG has also been

shown to reduce self-aggregation of lipid-based colloidal carriers, extending their

circulation longevity [61]. Colloidal carriers that avoid clearance by the MPS are

eventually excreted in the urine by the kidneys [32, 41, 62, 63] and/or undergo

hepatobiliary excretion in the feces [51]. For materials to be eliminated from circulation

by glomerular filtration in the kidneys, they must generally have a molecular weight of

less than 50,000 g/mol [64, 65] and have a diameter of less than approximately 5.5 nm

[66]. Since the total molecular weight and size of micelles is typically higher than this, it

may be assumed that only individual copolymer chains and free drug are eliminated via

renal excretion.

Page 27: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

12

1.7 Passive tumor accumulation via the EPR effect

BCMs have been shown to increase drug accumulation at solid tumor sites via passive

targeting. Passive targeting relies on the enhanced permeation and retention (EPR)

effect defined by the leaky vasculature and poor lymphatic drainage commonly

associated with solid tumors that enables the extravasation and accumulation of nano-

sized delivery systems at these sites [67, 68]. Importantly an extended circulation

lifetime is required for exploitation of the EPR effect. Therefore, passive targeting relies

primarily on the size of the delivery system but is also indirectly influenced by other

physico-chemical characteristics (e.g. surface properties) that may influence the

circulation lifetime of the BCMs. Indeed, passive targeting of drugs to tumors using

colloidal systems has been reported to result in as much as a 10–50 fold increase in

drug accumulation in tumors relative to normal tissues [69]. For example, Kwon and

Kataoka reported that the PEG-b-PAsp doxorubicin conjugated micelle system

increased the tumor accumulation of doxorubicin from 0.9% of the total injected dose

per gram of tumor for free DOX to 10% for the polymer-drug conjugated micelles 24 h

following i.v. administration in tumor bearing mice [70]. Recently, Kataoka and

coworkers also reported that polymeric micelles incorporating (1,2-

diaminocyclohexane)platinum(II) (DACHPt) resulted in accumulation of 10% of the total

injected dose per gram of tumor at 24 h p.i. in an orthotopic gastric cancer model (6-fold

higher than that achieved following administration of oxaliplatin) [71]. In addition,

elevated accumulation was observed in metastatic lymph nodes (15% total injected

dose per gram of tissue) resulting in a reduction in metastatic tumor growth.

There are several physiological factors that are responsible for determining the

extent of extravasation of colloidal carriers via the EPR effect including the density,

permeability and perfusion of the tumor vasculature as well as the interstitial fluid

pressure [72, 73]. Therefore, the delivery of drugs encapsulated in colloidal carriers

depends not only on the properties of the carrier itself, but also on the physiological

features of the tumor. It should be noted, however, that tumor physiology is highly

heterogeneous on an intra- and inter-tumor basis as well as within an animal or patient

population [74, 75]. These differences can have a profound effect on the efficacy of

EPR-mediated therapies and other forms of tumor-targeted therapies [74, 76–78].

Page 28: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

13

1.8 Tumor physiology and its role in nanoparticle delivery

1.8.1 Vascular density (density of functional vessels)

Since micelle delivery vehicles arrive at the tumor via the circulatory system, tumor

vascularization is an important factor in determining their initial distribution and ability to

exploit the EPR effect. The vascular density within a tumor is heterogeneous, with

vascular growth, perfusion or function often limited to specific regions [79]. As a result,

colloidal delivery vehicles may only be distributed to well-perfused compartments of the

tumor. In accordance with this phenomenon, Yuan et al. showed that following systemic

administration of fluorescently labeled liposomes in tumor bearing mice, the liposomes

preferentially accumulated in certain regions of the tumor and were entirely absent from

others [80]. The impact of heterogeneous delivery vehicle distribution in a tumor on

therapeutic efficacy is likely to depend on the rate of drug release and the ability of the

drug to penetrate within the extravascular space in order to reach cells located in

avascular regions of the tumor.

The extent and nature of tumor vascularization depends on several factors

including the type of tumor, its location, and its growth rate [79]. For example, Fukumura

et al. compared the microcirculation of a colon adenocarcinoma metastasized to the

liver with that of the same tumor grown subcutaneously [81]. They found that tumors

that had metastasized to the liver had significantly lower vessel density particularly in

the necrotic center in comparison to the subcutaneous tumors. The aforementioned

variations in tumor physiology may have important implications when comparing the

efficacy of formulations studied in different animal models and in the translation of

therapies from pre-clinical animal models to humans.

1.8.2 Vascular permeability

The permeability of tumor vasculature is an important determinant of BCM transport into

the tumor interstitium [82]. Extravasation of colloidal carriers from the blood into the

interstitial space occurs primarily via diffusion and convection through endothelial

fenestrae which may range in size from 200 nm to 1.2 um depending on the tumor type

[83]. A study by Dreher et al. showed that the tumor vascular permeability of

macromolecular carriers (i.e. dextrans) with molecular weights of 3.3, 10, 40, 70 kDa

Page 29: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

14

and 2 MDa was inversely correlated with their molecular size [84]. The potential

modulation of vascular permeability as a means of increasing the extravasation of

colloidal carriers was demonstrated by Monsky et al. using vascular endothelial growth

factor (VEGF) [85]. Injection of various nanoparticles combined with the topical

administration of VEGF (10 ng/ml) to the tumor via a dorsal skin chamber was found to

double the maximum pore size (from 400 to 800 nm) in human colon carcinoma

xenografts resulting in the extravasation of larger nanoparticles when compared to

controls. Similarly, Ogawara et al. demonstrated that the degree of in vivo accumulation

of radiolabeled PEG liposomes containing DOX was greater in tumors secreting higher

quantities of vascular endothelial growth factor (VEGF), independent of the extent of

vascularization [86]. Therefore the extent of BCM tumor accumulation will be influenced

by the unique vascular permeability associated with a specific tumor model or cancer.

As a result, BCMs of appropriate size should be selected for drug delivery based in part

on the vascular permeability of the tumor in order to maximize BCM transvascular

transport via the EPR effect [83, 84, 87]. Alternatively, modulation of tumor vascular

permeability may be an effective strategy to enhance tumor-specific drug delivery and to

overcome heterogeneity associated with EPR-mediated transvascular transport [68].

1.8.3 Interstitial fluid pressure and transport in the interstitium

Interstitial fluid pressure is elevated in tumors due to an increase in fluid and plasma

protein transport into the interstitial space resulting from the highly permeable nature of

tumor vessels and the absence of adequate lymphatic drainage [88]. The resulting

reduction in hydrostatic and oncotic pressure differences across the vessel wall

presents a barrier to the transport of colloidal carriers into the interstitium [89].

Furthermore, interstitial fluid pressure may be elevated in the center of the tumor

relative to the periphery resulting in reduced extravasation of macromolecules in the

center and a convective flow which forces macromolecules towards the tumor periphery

[90, 91]. The high cell density, presence of certain extracellular matrix components and

interactions with cells also decreases the diffusion of delivery systems in the tumor

interstitial compartment [92–94]. Indeed, colloids must be below a certain size in order

to achieve significant penetration from the vascular surface into the tumor interstitium

[84]. The study by Dreher et al., introduced above, also evaluated the three-dimensional

Page 30: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

15

penetration of differently sized (i.e. 3.3, 10, 40 and 70 kDa and 2 MDa) and

fluorescently labeled dextrans in mice bearing solid tumors using the dorsal skin fold

window chamber model and intravital laser scanning microscopy. The 10 kDa dextrans

were found to reach the same level of deep penetration as the 3 kDa dextrans but in a

longer period of time while the penetration of the 40 and 70 kDa dextrans were reported

to be similar in their penetration heterogeneity throughout the 30 minute study. Although

the permeability of larger macromolecules was decreased, the associated reduction in

the rate of clearance combined with a greater vascular AUC resulted in optimal

macromolecule molecular weights of 40 and 70 kDa with respect to total tumor

accumulation. The 2 MDa dextran was only detected in appreciable amounts up to 5 μm

from the vascular surface. The molecular weights of the dextrans (3.3, 10, 40, 70 kDa

and 2 MDa) used in this study were reported to correspond to hydrodynamic diameters

of about 4, 6, 11, 15 and 49 nm, respectively. Recently, Kataoka and coworkers

demonstrated that sub-100 nm micelles had no size dependent limitations on

penetration in tumors with a highly permeable structure, while only micelles smaller than

50 nm were capable of penetrating the interstitium of hypovascular tumors with low

structural permeability [95]. The limited transport of delivery systems away from tumor

entry sites may indeed preclude further entry of particles into the tumor [75]. Also, since

intervascular distances and avascular regions within tumors can be significant, the

limited penetration of a delivery system may prevent the eradication of cells distant from

the tumor vasculature; however this will also be dependent on the release and

distribution properties of the drug.

Importantly, in the selection of the size of a delivery system, a balance must be

achieved between optimizing blood residence time and tumor penetration. An extended

circulation lifetime is required for the delivery system to fully exploit the EPR effect: in

mice and rats it has been reported that the “area under the time-concentration curve

(AUC) must remain high for >6 h” [69]. In general, an increase in the molecular size or

hydrodynamic diameter of macromolecules or colloidal delivery systems is known to

increase their residence time in the bloodstream [66, 84, 96]. Yet, beyond a certain size,

significant uptake and accumulation in MPS organs such as the liver and spleen tends

to occur [97, 98]. Also, larger sized particles (>30–45 kDa) are retained within the tumor

tissue longer than small molecules which rapidly clear by diffusion [99, 100]. Therefore

Page 31: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

16

the size of the nano-carrier should be adjusted accordingly in order to balance the

advantage of passive uptake at the tumor via the EPR effect with undesirable uptake in

other tissues such as the liver and spleen and tumor penetration. Achieving this balance

and selecting an optimal size may be confounded by the differences in physiology

between animals and humans that may significantly alter the observed performance of a

colloidal carrier system, such as BCMs, in humans. Some of the most promising

preclinical results for a BCM formulation were reported by Hamaguchi et al. using

NK105, a PEO-b-poly(4-phenyl-1-butanoate-L-aspartamide) [PEO-b-PPBA] block

copolymer micelle [34]. Micelles containing physically entrapped PTX were

administered to C26 tumor bearing CDF1 mice resulting in a 90-fold increase in PTX

plasma AUC and a 25-fold increase in its tumor AUC relative to free PTX. The treatment

resulted in complete tumor regression following a single injection of a PTX-equivalent

dose of 100 mg/kg. However, in phase I clinical trials, the increase in plasma AUC

following administration of NK105 was significantly less in humans (~15 fold increase)

when compared to the results of the preclinical animal studies. Translation of

formulations from pre-clinical animal models to humans is challenging owing to

differences in their MPS activity, vascular characteristics at the tumor site and tumor

physiology. Discrepancies between the physiology of animal tumor xenografts and the

physiology of native human tumors are likely to profoundly influence the performance of

colloidal delivery systems. These same differences may exist between individual

patients and highlight the need to work towards the implementation of personalized

therapies.

1.9 Active targeting

BCMs may be actively targeted by incorporating surface grafted recognition moieties

that impart an affinity for cellular receptors or components that are present on and/or

upregulated by tumor cells [101]. Ligands that have been pursued for active targeting of

BCMs to different cancers include sugar moieties such as galactose [102], epidermal

growth factor [103], folate [104], antibodies [105], and peptides [106]. In vitro cell culture

studies have demonstrated that coupling of targeting moieties to the micelle surface

results in their selective accumulation in target cells via receptor-mediated endocytosis

[104, 105, 107]. Furthermore, in vitro studies have shown that active targeting can also

Page 32: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

17

overcome multidrug resistance and direct delivery systems to specific subcellular

compartments [108–110]. Drug formulations relying on actively targeted BCMs and

other colloidal systems have also been shown to result in significant improvements in

efficacy in pre-clinical animal models, in comparison to the non-targeted drug

formulations [111–115]. However, few studies have examined the influence of active

targeting on the cellular uptake of delivery systems within tumors in vivo [99, 116–118].

In one example, Mamot et al. demonstrated that anti-EGFR immunoliposomes

achieved a 6-fold increase in cellular uptake at the tumor site compared to nontargeted

liposomes 24 h following i.v. administration [113]. The influence of the presence of a

targeting moiety on the pharmacokinetics, biodistribution and tumor accumulation of

nanosystems is controversial [99]. In some cases active targeting of a delivery system

has been shown to increase tumor accumulation while in others the degree of tumor

accumulation remains constant [99]. For example, Kirpotin et al. showed that active

targeting of liposomes using the anti-HER2 monoclonal antibody did not result in

increased tumor localization in mice bearing HER2-overexpressing breast cancer tumor

xenografts but did lead to a greater degree of cellular internalization at the tumor site

and an increase in efficacy [117]. Similarly, Bae et al. found no significant difference in

tumor accumulation between folate-targeted and non-targeted micelles suggesting that

the EPR effect is the primary factor in tumor localization [119]. In contrast, Hussain et al.

found that the tumor accumulation of liposomes was 2 fold greater for epithelial cell

adhesion molecule targeted liposomes compared to non-targeted liposomes [114].

Overall, the influence of a targeting moiety on the in vivo pathway and fate of delivery

systems is dependent on the nature of the targeting moiety as well as its density at the

surface of the vehicle and biological factors such as tumor physiology [119]. However,

further studies are needed to fully elucidate the influence of active targeting on BCMs in

vivo. It is likely that although passive targeting is the primary mechanism for tumor

accumulation via the EPR effect, active targeting may promote retention of nano-

carriers in the tumor interstitium and increase cellular internalization.

1.10 Microdistribution: transport within an organ or tissue

Traditionally, the primary goal of drug delivery is to increase the concentration of a

therapeutic agent at the tumor site while limiting systemic exposure and toxicity to non-

Page 33: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

18

target tissues and organs. However, in order for chemotherapeutic drugs to be effective,

they must be capable not only of reaching the tumor but also of penetrating it sufficiently

in order to expose all constituent tumor cells to a therapeutically relevant dose. There is

increasing evidence that tumor repopulation following treatment is not only mediated by

cellular mechanisms of drug resistance but also by a drug's limited penetration and

distribution within the tumor which results in insufficient elimination of malignant cells

[25, 26, 120, 121]. For chemotherapy to be curative, the drug should have access to all

cells in the tumor in lethal quantities, as certain cells that remain following treatment

may be capable of initiating tumor recurrence [25]. Due to limited tumor vascular

density, a drug may be required to penetrate > 100 µm from the nearest blood vessel to

reach distant cells [122]. This is in contrast to normal tissues in which cells usually

remain within a few cell diameters from a vessel [122]. Therefore, the extent of

penetration within the interstitial space of tumors is critical in determining the dose of

drug received by cells distant from the tumor vasculature.

The transport of drugs within the extravascular space of solid tumors is

influenced by both tumor physiology and the physicochemical properties of the drug.

Several excellent reviews on the penetration of drugs and nanomedicines in tumors can

be found elsewhere [73, 122, 123]. In general, the supply of drug to a tumor depends on

the injected dose, the drug's pharmacokinetic profile and biodistribution, tumor

vascularization, and, in the case of drugs encapsulated within colloidal delivery vehicles,

the extent of extravasation via the EPR effect. Once in the interstitium, drugs may be

transported by convection and/or diffusion [73, 124]. Convection in the interstitial space

is determined by the pressure gradient established according to intravascular hydraulic

pressure, transvascular fluid flow and the rate of resorption into the lymphatic vessels.

However, due to the lack of functional lymphatic drainage in tumors, fluid accumulates

in the interstitial space resulting in increased interstitial pressure and limited convective

flow and interstitial transport [123]. The rate and extent of diffusion through the tumor

interstitium is determined by properties of the drug including its size, charge and

structure, the extent of its cellular uptake and binding, as well as the structure and

composition of the interstitial compartment (refer to section 1.8.3 for further details) [92,

124–126].

Page 34: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

19

Transport of the free drug through the tumor tissue occurs through extracellular

or trans-cellular pathways depending on the drug's relative aqueous and lipid solubility.

Consumption of the drug due to cellular metabolism or intra-cellular binding at the site of

action (e.g. microtubules, DNA) will reduce drug penetration [122]. In many cases, these

factors act to minimize the transport of drugs away from the vasculature, minimizing

access of the drug to distant cells.

Figure 1.3. (a) EGF-conjugated PEG-b-PVL micelles labeled with a hydrophobic

fluorescent probe, CM-DiI, via physical encapsulation. (b) Fluorescent confocal

microscopy images of EGFR-overexpressing breast cancer cells (MDA-MB-468)

incubated with non-targeted and targeted micelles for two hours at 37°C. (c) MDA-MB-

468 cells were incubated with EGFR-targeted micelles, and were subsequently counter-

stained with Hoechst 33258 for visualization of the cell nuclei. The white arrows

indicate localization of the EGFR-targeted micelles in the nucleus. (b) and (c) are taken

from Zeng et al. [108].

Page 35: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

20

A significant limitation associated with the current methodology for assessing the

accumulation of drugs and/or delivery systems in solid tumors in vivo is the assumption

that their tumor distribution is homogeneous. For example, biodistribution studies are

commonly performed by excising and processing the entire tumor for quantitative

analysis of tumor accumulation using radioactively labeled drugs or drug delivery

vehicles. Although this kind of analysis may provide important information regarding

total tumor accumulation, it provides no insight into the spatial distribution of a drug or

carrier within the tumor. As a result, image-based methodologies have recently been

employed in order to improve our understanding of drug and drug delivery vehicle

distribution within tumors in vivo. For example, intravital laser-scanning confocal

microscopy has been used in murine tumor xenograft models via a dorsal skin fold

window chamber to assess penetration of fluorescently labeled macromolecules from

the vascular surface into the tumor interstitium [127]. Using this technique, Yuan et al.

showed that fluorescently labeled liposomes between 86 and 90 nm in size remained

near the vasculature with minimal penetration through the interstitium [80]. Following

extravasation, the liposomes formed clusters that were not significantly transported into

the interstitial space. Recently, Kataoka and coworkers have implemented an intravital

confocal microscopy technique that permits real-time analysis of the tissue penetration

and distribution of micelles in tumors and other organs [95, 129, 130]

In addition, studies by Zheng et al. have used computed tomography (CT) to

track the biodistribution and tumor accumulation of liposomes with an average diameter

of 80 nm in rabbit models [131, 132]. The CT-based assessment enabled noninvasive

and real-time visualization of the heterogeneous intratumoral distribution of the

liposomes at submillimeter resolution in VX-2 sarcoma bearing New Zealand White

rabbits [131]. As well Hoang et al. employed both the traditional method and

microSPECT/CT imaging to quantify the biodistribution of an 111Indium-labeled (111In)

PEG-b-PCL copolymer micelle formulation [132]. The 111In-copolymer was administered

intravenously to healthy and MDA-MB-231 tumor-bearing mice as micelles with an

average hydrodynamic diameter of 58 nm. Data obtained by traditional evaluation of

biodistribution was found to correlate well with data acquired using non-invasive image-

based analysis. Moreover, the intratumoral distribution of the micelles in vivo was

revealed by microSPECT/CT imaging. As shown in Figure 1.4, the intratumoral

Page 36: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

21

Figure 1.4. a) MIP and saggital image of tissue accumulation of 111In-PEG-b-PCL

micelles 48 h p.i in an athymic BALB/c mouse bearing an MDA-MB-231 tumor xenograft

after i.v. administration of 111In-micelles. Clear visualization of the liver, spleen, bladder

and tumor was observed. b) Tissue distribution of 111In-micelles acquired via

conventional methodology and MicroSPECT/CT ROI analyses in MDA-MB-231 tumor-

bearing mice at 48 h p.i. c) Transversal slices of tumor accumulation illustrating non-

homogeneous distribution of 111In-micelles. Taken from Hoang et al. [132].

a) b)

c)

Page 37: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

22

distribution of the micelles was found to be heterogeneous and limited primarily to the

tumor periphery. This information is critical for the development of drug delivery vehicles

with improved distribution and penetration in tumors and is otherwise unavailable

through traditional evaluation of biodistribution.

In general, there remains a limited understanding of the independent fate of drugs

and colloidal carriers in tumors. It is likely that following extravasation, rapid release of

the drug from the colloidal carrier would result in a similar distribution of drug within the

tumor as would be observed for freely administered drug. Carriers that demonstrate a

sustained release effect but remain confined to certain tumor compartments may act as

drug reservoirs that deliver the drug within the tumor over extended periods of time

even following a reduction in plasma concentration. In this scenario, the efficacy of

treatment may be highly dependent on the ability of the drug itself to penetrate within

the tumor tissue. Drug delivery vehicles that strongly retain the drug and do not

distribute within the tumor may inhibit drug penetration and treatment efficacy. For drugs

that display poor penetration in tissues, encapsulation within delivery vehicles that

enhance transport would be ideal, though this possibility has yet to be examined.

1.11 3-D tissue culture and the multicellular tumor spheroid model

Evaluating penetration in relevant 3-D in vitro models can be useful for the systematic

analysis of the influence of drug or delivery vehicle properties on tissue transport. To

date, 3-D in vitro tumor tissue models have served to enhance our understanding of

biological processes associated with tumor growth and resistance to treatment. Several

models with varying degrees of biological complexity have been implemented including

multicellular layers [133–135], natural and synthetic tissue scaffolds [136–138], and

multicellular tumor spheroids [29, 139, 140]. Of these models, spheroids, which are

growing spherical aggregates of tumor cells, are particularly appealing for their potential

use in high-throughput screening protocols and the ability to measure their growth in

response to treatment. The use of spheroids for evaluating anti-cancer therapies was

largely pioneered by Sutherland et al. via a series of systematic studies investigating

their response to radiotherapy in the early 1970’s [141–143]. The spheroid model has

also been applied in evaluating the influence of the tumor microenvironment on

fundamental cellular processes such as proliferation, metabolism, differentiation,

Page 38: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

23

invasion and metastasis [144–146]. Furthermore, mechanisms of resistance to

chemotherapy and limitations in drug penetration have also been revealed in spheroid

cultures [147–149]. Yet, to date, the application of spheroids in the design and

evaluation of nanomedicines has been limited [150–152].

In general, spheroids contain a gradient in cellular proliferation from the surface to

the core with a greater proportion of proliferating cells located closer to the spheroid

periphery and quiescent cells located centrally. This structural organization reflects the

radial distribution of tissues surrounding tumor blood vessels in which proliferative cells

reside proximally and less metabolically active cells reside distally. In addition,

spheroids possess an extensive extracellular matrix as well gradients in pH and pO2

reflective of the acidic and hypoxic microenvironment of solid tumors and avascular

tumor nodules [153]. Together, these characteristics serve to approximate conditions of

the tumor microenvironment enabling better prediction of therapeutic response in vivo

relative to conventional 2-D cultures. In addition, the symmetrical, spherical morphology

of spheroids makes them a useful platform for evaluating the transport of drugs and

nano-carriers by assessing their penetration from the spheroid surface towards the core

[154].

1.12 Transport at the cellular level

Following extravasation of drug loaded micelles into the tumor interstitium, hydrophobic

drugs may be transported into cancer cells by diffusion following their release from

intact or disassembled micelles or by cellular internalization of the drug-loaded micelle

[155–158]. The transport of hydrophobic molecules across the cell membrane is

governed by the difference in drug concentration between the interstitial fluid and the

intracellular compartment. Therefore, the rate of release of the drug from the micelle

can be expected to influence the rate of diffusion and intracellular drug accumulation.

Alternatively, drug loaded micelles may undergo cellular internalization prior to drug

release. However, unlike small hydrophobic drug molecules, BCMs cannot diffuse

through the cell membrane but rather are internalized by endocytosis. Following cell

uptake, micelles are contained within acidic endosomes and may follow various

transport pathways including endosomal fusion with lysosomes or exocytosis

Page 39: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

24

(recycling). Detailed reviews of the endocytotic pathways and endocytosis of

nanosystems have been recently published [159–162].

BCMs have been shown to alter the subcellular distribution of drugs. To date,

studies examining the subcellular distribution of the components of block copolymer-

based formulations (i.e. drug and copolymer) have primarily relied upon confocal

microscopy in combination with fluorescently labeled drug and/or copolymer and various

organelle specific stains [116, 163–166]. Rapoport et al. demonstrated that fluorescently

labeled Pluronic® micelles internalized by fluid phase endocytosis can alter the

distribution of a drug between acidic vesicles and the cytosol [109]. When administered

in PBS, the drug ruboxyl was not found in the nuclei of multi-drug resistant (MDR) cells,

whereas following administration of ruboxyl in Pluronic P-105 micelles, significant

accumulation was observed in the nuclei due to drug release from cytosolic vesicles.

They suggested that the permeabilization of acidic vesicle membranes by the Pluronic®

copolymer surfactant resulted in the release of the drug into the cytoplasm. The

subcellular distribution of micelles and their cargo has also been shown to be altered by

the addition of a targeting moiety to the surface of the delivery system. As shown in

Figure 1.3, Zeng et al. demonstrated that conjugation of epidermal growth factor (EGF)

to the surface of PEG-b-PVL micelles resulted in preferential perinuclear and nuclear

localization of the micelles in the epidermal growth factor receptor overexpressing cell

line MDA-MB-468 [108].

Stimuli-responsive BCMs containing surface-bound active targeting moieties have

been developed to specifically target the acidic environment of endosomes. For

example, BCMs containing drugs conjugated to the hydrophobic core by pH-labile

chemical linkers have been developed to release the drug when exposed to the acidic

environment of endosomes [167–170]. In another approach, BCMs composed of

polymers containing ionizable groups have been designed such that the micelle is

destabilized following polymer charge conversion in response to acidic conditions [107,

[171, 172]. Furthermore, some cationic polymers have demonstrated the ability to

disrupt the endosomal membrane based on the “proton sponge effect” whereby

ionizable groups become protonated and therefore act to resist the natural acidification

of endosomes leading to further influx of cytosolic protons, accompanying Cl- ions and

osmotically-driven swelling and membrane rupture [173–178]. Kataoka's group has

Page 40: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

25

developed a polyplex triblock copolymer micelle system for the delivery of DNA to solid

tumors containing an endosomal disrupting segment composed of a poly(aspartamide)

derivative, (PAsp(DET)). The highly efficient transfection achieved by this system was

attributed to the change in protonation of (PAsp (DET)) in the acidic environment of

endosomes and subsequent disruption of the endosomal membrane and release of

DNA into the cytoplasm [173]. Notably, BCM systems have also demonstrated particular

effectiveness against MDR cell lines [171, 179]. Bae's group has developed a mixed

micelle system composed of poly(L-histidine)-block-poly(ethylene glycol) (PolyHis-b-

PEG) and poly(L-lactic acid) (PLLA)-b-PEG-folate that undergoes protonation of polyHis

below its pKb and deprotonation above its pKb. As a result, micelles are formed at high

pH, and are destabilized in acidic environments due to the increase in hydrophilicity of

the micelle core. PolyHis-b-PEG unimers induce disruption of the endosomal membrane

resulting in enhanced drug release into the cytoplasm and greater cytotoxicity towards

ovarian MDR carcinoma cells compared to pH insensitive micelles [110]. Intracellular

accumulation of drugs in resistant cancer cell lines was also increased in the presence

of Pluronic® polymers. Specifically, Kabanov's research has shown that Pluronic®

copolymers, with HLB values b19 and intermediate lengths for the PPO blocks, are

effective drug efflux modulators [180]. Burt's group also showed that PEG-b-PCL

copolymers were capable of increasing the accumulation of a P-gp substrate in Caco-2

cells [181].

1.13 Docetaxel

DTX is an anti-mitotic agent, currently administered as Taxotere® (Sanofi-Aventis) or as

a generic (Hospira), belonging to the taxane family of drugs and is approved for

treatment of cancers of the breast, prostate, lung, head and neck, and stomach [182].

DTX, like its sibling paclitaxel, has been shown to exert an anti-cancer effect primarily

by impeding mitosis via stabilization of microtubules [183]. Microtubules are an

important cellular component involved in several essential cell processes including

intracellular transport and cell division. DTX binds to the microtubule subunit β-tubulin,

inducing hyperstabilization that leads to cell-cycle arrest and apoptosis [184]. DTX has

demonstrated greater efficacy than paclitaxel in preclinical and clinical studies due to its

improved cellular uptake and increased binding and stabilization of microtubules [185].

Page 41: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

26

In a randomized phase III study comparing docetaxel to paclitaxel in patients with

advanced breast cancer, the median overall survival, median time to progression and

overall response rate were greater for patients receiving docetaxel [186]. DTX has also

demonstrated superior response rates and median time to progression in anthracycline

and alkylating-agent resistant metastatic breast cancer relative to doxorubicin and high

activity as first-line chemotherapy [187–189].

Following i.v. administration, it has been reported that 92% of docetaxel binds with

plasma proteins [190]. Elimination occurs primarily by metabolism in the liver and

excretion into the feces via the bile [185]. Clearance from systemic circulation is

biphasic (for doses up to 70 mg/m2) with associated plasma half-lives of 5.4 minutes

and 1.4 hours following a 1 hour infusion at a dose of 70 mg/m2 in humans [185, 191].

Docetaxel is also associated with serious toxicities, most commonly neutropenia, that in

certain cases require dose reduction [192]. Taxotere® is comprised of docetaxel

dissolved in polysorbate 80 (Tween® 80), a non-ionic surfactant which has been linked

to hypersensitivity reactions [1]. In light of its poor solubility, toxicity and rapid plasma

clearance, docetaxel is an ideal candidate for encapsulation in BCMs.

1.14 Summary

BCMs have proven to be a viable and versatile platform for the development of novel

chemotherapeutic formulations with the potential to significantly improve cancer

treatment outcomes. However, effective development of BCM-based drug formulations

from pre-clinical evaluation to clinical application requires the use of improved in vitro

models which mimic mechanisms of drug resistance associated with the tumor

microenvironment. The use of 3-D tissue cultures such as multicellular tumor spheroids

for the rational design and optimization of nanosystems will serve to expedite the

translational process. Establishment of advanced techniques for evaluating the

intratumoral transport of nanosystems is required in order to better understand the

complex relationships between nano-carrier properties, intratumoral penetration and

therapeutic efficacy in the context of the tumor microenvironment. Integration of

computational, imaging-based methodologies within the field of drug delivery research

promises to provide powerful and unique insight into the transport and fate of

nanosystems in vivo with the ultimate goal of realizing their full therapeutic potential.

Page 42: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

27

1.15 Rationale

According to the World Health Organization (WHO), cancer accounts for more than 7.6

million deaths worldwide every year [193]. In Canada, it is estimated that approximately

40% of women and 45% of men will develop cancer during their lifetimes and 1 in every

4 Canadians will die from this disease [194]. Chemotherapy is a critical component of

most cancer treatment strategies. However, its efficacy hinges on the ability of the

therapeutic agent to selectively harm cancer cells while inflicting as little damage as

possible to healthy tissues and organs. Since the primary selective mechanism of many

current cytostatic drugs relies upon their inherent capacity to exert toxicity to rapidly

proliferating cells, collateral damage to dividing cells throughout the body which are

non-cancerous is inevitable. This limits the maximum tolerable dose or may result in

side effects that necessitate dose reductions that hinder treatment efficacy or in some

cases may require discontinuation of therapy [193, 194].

Encapsulation of chemotherapeutic agents in nanoparticles such as block

copolymer micelles (BCMs) has proven to be a promising approach for reducing toxicity

to healthy tissues and organs and improving therapeutic efficacy. As such, there are

now seven BCM-based formulations undergoing clinical trial evaluation [23]. While

some of these formulations serve primarily to enhance the solubility of the drug,

advances in nanoengineering have led to the development of carrier vehicles capable of

tumor-targeted drug delivery. Yet, despite achieving increased tumor-specific drug

accumulation, realization of commensurate improvements in therapeutic efficacy has

proven elusive. Indeed, recent evidence suggests that nanoparticles may be

heterogeneously distributed in tumors, potentially limiting the exposure of cancer cells to

encapsulated drugs. For chemotherapy to be most effective the drug must be

distributed throughout a tumor in lethal quantities so as to minimize the survival of

clonogenic cells capable of initiating tumor repopulation. Therefore, the focus of this

research was to elucidate critical determinants of nanoparticle intratumoral transport

using novel in vitro and in vivo computational methodologies with the ultimate goal of

establishing rational design criteria for the development of nanomedicines with

enhanced intratumoral distribution and efficacy.

Page 43: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

28

1.16 Hypotheses

1) The 3-D tumor microenvironment imposes substantial obstacles which limit the

effective transport and therapeutic efficacy of nanomedicines. The penetration of

nanoparticles in tumor tissues is a dynamic process wherein the rate and extent

depend on nanoparticle physicochemical properties, such as particle size, and

physiological properties of the tumor tissues.

2) Multicellular tumor spheroids can be used to evaluate the transport and

therapeutic efficacy of nanomedicines in vitro and to facilitate prediction of their

interstitial penetration and performance in vivo.

1.17 Objectives

In accordance with the hypotheses described above, the specific research objectives

were as follows:

Objective 1: To synthesize a polymeric nanomedicine containing docetaxel and to

characterize its physicochemical properties including drug loading capacity, release

kinetics and morphology.

Objective 2: To establish a 3-D tissue model and corresponding in vitro methodologies

for evaluating the performance of the nanomedicine and Taxotere®.

Objective 3: To implement computational, image-based methodologies for evaluating

the spatio-temporal penetration of nanoparticles in 3-D tissue cultures (spheroids) and

human tumor xenografts.

Objective 4: To assess the role of nanoparticle size and tumor tissue properties on the

spatio-temporal distribution of nanoparticles in spheroids and corresponding tumor

xenografts and to evaluate the “in vivo” predictive capacity of the spheroid model.

Page 44: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

29

1.18 Overview of Thesis Chapters

The studies addressing the specific aims of this research, described above, are

described in Chapters 2 - 4 of the thesis. Chapter 2 describes the preparation and

physico-chemical characterization of PEG-b-PCL micelles as well as two methods of

incorporating DTX into the micelles, namely, physical entrapment and chemical

conjugation. This chapter also demonstrates the release kinetics of DTX from the

micelles and examines micelle morphology. Specifically, the influence of chemical

conjugation of DTX to the micelle core on drug release and micelle morphology was

reported and potential implications for drug delivery were discussed. Chapter 3

describes the performance of DTX-loaded PEG-b-PCL BCMs relative to Taxotere®

using a 3-D in vitro tumor model. Multicellular tumor spheroids were grown using colon

and cervical cancer cell lines and characterized in terms of their growth rate, cell

packing density, and tissue architecture. The cytotoxicity of BCM+DTX was compared in

monolayer and spheroid cultures in order to assess the influence of the 3-D

microenvironment on resistance to therapy. The ability of BCM+DTX and Taxotere® to

inhibit the growth of MCTS over a 30 day period was also evaluated. Resistance of

MCTS to treatment was assessed with respect to cell line-specific differences in MCTS

microenvironment and tissue structure. Chapter 4 describes the evaluation of BCM

penetration in tumor tissues using both in vitro and in vivo tumor models. A

computational methodology was developed for quantifying the relative distribution of

fluorescently labeled micelles following incubation with MCTS. In addition, a

computational procedure was established for the evaluation of BCM interstitial tissue

penetration in tumor xenografts following systemic administration. The influence of

micelle size and structural properties of the tumor tissues on micelle penetration was

evaluated. Chapter 5 summarizes the main findings of this research, presents

conclusions, and outlines potential future research.

Page 45: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

30

2 Chapter 2

Poly(ethylene glycol)-b-poly(ε-caprolactone) Micelles Containing

Chemically Conjugated and Physically Entrapped Docetaxel:

Synthesis, Characterization, and the Influence of the Drug on

Micelle Morphology

Chapter 2

Page 46: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

31

This chapter is a reprint of: “Andrew S. Mikhail and Christine Allen. Poly(ethylene

glycol)-b-poly(ε-caprolactone) Micelles Containing Chemically Conjugated and

Physically Entrapped Docetaxel: Synthesis, Characterization, and the Influence of the

Drug on Micelle Morphology.” Biomacromolecules 2010, 11, 1273–1280; with

permission from the American Chemical Society.

All experiments were performed by AS. Mikhail.

This chapter was written by AS. Mikhail and edited by Dr. C. Allen.

Page 47: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

32

2.1 Abstract

Docetaxel was coupled to the hydrophobic block of poly(ethylene glycol)-b-poly(ε-

caprolactone) (PEG-b-PCL) copolymers synthesized by metal free ring-opening

polymerization. Synthesis of the copolymers and the copolymer-drug conjugate (PEG-b-

PCL-DTX) were confirmed by 1H NMR and GPC analyses. The PEG-b-PCL-DTX

conjugates had a ∼1:3 drug/copolymer ratio (w/w) and a low critical micelle

concentration in aqueous solution (14 mg/L). Encapsulation of DTX in PEG-b-PCL-DTX

micelles resulted in an 1840-fold increase in the aqueous solubility of the drug. Release

of physically encapsulated DTX from PEG-b-PCL-DTX micelles was slower than drug

release from PEG-b-PCL micelles due to the improved compatibility between DTX and

the micelle core. Core-conjugated DTX was released over the course of one week

indicating that PEG-b-PCL-DTX micelles have the capacity for sustained drug release in

the absence of physically encapsulated drug. Importantly, conjugation of DTX to the

core-forming block had a profound effect on the morphology of the copolymer

aggregates.

Page 48: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

33

2.2 Introduction

The use of block copolymer micelles (BCMs) as nano-sized drug delivery vehicles is a

viable and versatile strategy for improving the toxicity profile and efficacy of

chemotherapeutic agents [197]. However, effective design of advanced delivery

systems of this nature demands a thorough understanding of how the physicochemical

properties of BCMs influence their performance as drug delivery vehicles [198].

Alterations to the composition of the constituent copolymers allows for the manipulation

of important properties such as the compatibility between the drug and the carrier. Good

compatibility between the drug and the core-forming block of the micelle affords high

drug-loading levels [199], improved drug solubility [47], and sustained release of the

encapsulated drug [200]. Several strategies have been employed to improve core-drug

compatibility, including optimization of the structure [34, 201, 202] and end group [44] of

the hydrophobic block and chemical conjugation of drug molecules to the core [203].

Recently, BCM morphology has been identified as an important determinant of

their performance as drug delivery vehicles [204, 205]. Specifically, the morphology of

BCMs has been shown to influence their drug-loading capacity and drug release profile

[206]. In addition, Christian et al. showed that administration of the anticancer drug

paclitaxel in filamentous block copolymer aggregates, called filomicelles, resulted in a

higher maximum tolerated dose for the drug and more sustained tumor shrinkage in

comparison to administration of the drug in spherical micelles [207]. Discher’s group has

also demonstrated that the shape of the micelle can influence delivery vehicle transport

and their interactions with phagocytic cells [208]. Therefore BCM morphology must be

carefully considered in the design and evaluation of copolymer-based drug formulations.

In many of the published studies performed to date, the morphology of BCM-

based drug formulations is largely ignored or simply assumed to be spherical. As such,

there remains a limited understanding of the factors that control the morphology of

copolymer aggregates formed from biocompatible and biodegradable copolymer

materials [209, 210]. If greater morphological control is achieved, efforts may be made

to more clearly elucidate the influence of BCM morphology on drug delivery vehicle

biodistribution and transport in vivo. In the work presented herein, the physicochemical

characteristics of biocompatible BCMs comprised of poly(ethylene glycol)-b-poly(ε-

Page 49: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

34

caprolactone) (PEG-b-PCL) and a novel micelle-forming copolymer-drug conjugate

(PEG-b-PCL-DTX) containing docetaxel (DTX) are reported. The copolymer-drug

conjugate contains DTX that is chemically conjugated to the hydrophobic PCL core of

the micelle in order to improve the compatibility between the core and physically

entrapped DTX. The influence of chemical conjugation of DTX on drug loading, drug

release, and morphology of the micelles is evaluated. Importantly, the conjugation of the

drug to the core-forming block is not only found to have a profound effect on drug

loading but also on the morphology of the copolymer aggregates. As well, the chemical

entrapment of a drug into BCMs is shown to alter their morphology.

2.3 Experimental Section

2.3.1 Materials

α-Methoxy-ω-hydroxypoly(ethylene glycol) (CH3O-PEGOH, Mn = 2000, Mw/Mn = 1.06,

as determined by SEC) was obtained from Sigma-Aldrich (Oakville, ON, Canada) and

dried by azeodistillation of toluene. ε-Caprolactone and dichloromethane were

purchased from Sigma-Aldrich and were dried using calcium hydride. Hydrogen chloride

(1.0 M in diethyl ether), 1,6-diphenyl-1,3,5-hexatriene (DPH), succinic anhydride, N,N-

dicyclohexylcarbodiimide (DCC), 4-dimethylaminopyridine (DMAP), N,N-

dimethylformamide (DMF), diethyl ether, hexane, and acetonitrile were also purchased

from Sigma-Aldrich and used without further purification. Anhydrous docetaxel was

purchased from Jari Pharmaceutical Co. (Jiangsu, China) and 3H-labeled docetaxel was

purchased from American Radiolabeled Chemicals (St. Louis, MO).

2.3.2 Synthesis of CH3O-PEG-b-PCL (PEG-b-PCL) Copolymers

PEG-b-PCL copolymers were prepared by metal-free cationic ring-opening

polymerization of ε-CL using CH3O-PEG-OH as macroinitiator in the presence of HCl,

as previously reported [37]. Typically, 2.0 g of CH3O-PEG-OH (1 mmol, Mn) 2000,

Mw/Mn ) 1.06) was added to a flame dried flask, dried by toluene azeodistillation, and

dissolved in 12 mL of dichloromethane. ε-Caprolactone (1.0 or 2.0 g; 8.76 or 17.52

Page 50: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

35

mmol, distilled over calcium hydride) was added, followed by 3 mL of hydrogen chloride

(0.003 mmol, 1.0 M in diethyl ether). The reaction solution was stirred for 24 h at room

temperature prior to termination upon the addition of triethylamine (0.5 mL). Finally, the

triethylamine hydrochloride salt was removed by filtration, and the copolymer was

collected via precipitation in diethyl ether and hexane (50:50, v/v%) and dried under

vacuum at room temperature.

2.3.3 Addition of Carboxylic Acid Group to PCL Block of PEG-b-PCL Copolymers

PEG-b-PCL block copolymers were modified by reaction with succinic anhydride to

generate carboxyl-terminated PEG-b-PCL (PEG-b-PCL-COOH) copolymers [211].

Specifically, 0.5 g of PEG-b-PCL was placed in a flame-dried flask containing 50x molar

excess of succinic anhydride. The reactants were dissolved in 10 mL of pyridine and

heated to 60 °C for 4 h before being cooled to room temperature and allowed to stir for

20 h. Pyridine was removed in vacuo and the polymer was dissolved in acetonitrile,

dialyzed against water for 3 days, and lyophilized. The reaction yield was 72%.

2.3.4 Conjugation of DTX to PEG-b-PCL-COOH Copolymers

Conjugation of DTX to PEG-b-PCL-COOH copolymers was conducted by reaction with

equimolar equivalents of 4-dimethylaminopyridine (DMAP) and N,N′-

dicyclohexylcarbodiimide (DCC) in dry dichloromethane. The copolymer was first placed

in a flame-dried flask and dried by azeodistillation with toluene. DCC and DMAP were

then added and the reaction flask was purged with N2 prior to the addition of

dichloromethane. The flask and its contents were then cooled to 0 °C in an ice bath for

2 h and subsequently stirred for 22 h at room temperature. The reaction solution was

then filtered to remove the N,N′-dicyclohexylurea precipitate and dried under vacuum to

recover the product. Free DTX was removed by washing thoroughly in diethyl ether.

Successful conjugation of DTX to the copolymer was confirmed using gel permeation

chromatography (GPC), high performance liquid chromatography (HPLC), and nuclear

magnetic resonance (1H NMR) analyses.

Page 51: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

36

2.3.5 Characterization of Copolymers and Copolymer-Drug Conjugates

1H NMR spectra were obtained on an Oxford 400 spectrometer (400 MHz) using CDCl3

as the solvent. Chemical shifts were reported in ppm. GPC measurements were

performed using a Waters 590 liquid chromatography system equipped with three

Waters Styragel HR 4E columns and a 410 differential refractometer detector. THF with

1% triethylamine was used as the mobile phase at a flow rate of 1.0 mL/min through the

columns at 40 °C. Narrow polystyrene standards (Polysciences Inc., Warrington, PA)

were used to generate a GPC calibration curve. The data obtained were recorded and

assessed using the Windows-based Millenium 2.0 software package (Waters Inc.,

Milford, MA). The amount of DTX conjugated to the copolymer was determined using

HPLC (Agilent series 1200) connected to a UV detector (Waters 2487).

Chromatographic separation of PEG-b-PCLDTX and DTX was achieved using an

XTerra C18 reverse phase column and acetonitrile/water (51/49, v/v%) as the mobile

phase with UV detection at 227 nm. To determine the efficiency of DTX conjugation to

the copolymer, a sample of PEG-b-PCL-DTX before and after the removal of free DTX

was dissolved in acetonitrile and the elution peak of DTX in the sample was quantified

using a calibration curve generated from a series of DTX samples of known

concentration. The extent of DTX conjugation was also determined by scintillation

counting (Beckman Coulter, LS 6500) following the conjugation of 3H-DTX to the

copolymer.

2.3.6 Micellization and Measurement of Critical Micelle Concentration (CMC)

Micelles were prepared from PEG-b-PCL copolymers of various molecular weights and

the PEG-b-PCL-DTX copolymer-drug conjugate using a dry-down method. Copolymer

or copolymer-drug conjugate was dissolved in DMF and allowed to stir overnight.

Following evaporation of DMF under N2 and vacuum, polymer films were heated to 60

°C in a water bath. Hot filtered distilled water or buffer (PBS, pH 7.4) was subsequently

added to vials containing the polymer films and thoroughly vortexed. Resulting micelle

solutions were stirred for 3 days. The CMCs of the copolymers and copolymer-drug

conjugate were determined by an established fluorescence-based method [212]. Briefly,

an aliquot of a 1,6-diphenyl-1,3,5-hexatriene (DPH) stock solution prepared in

Page 52: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

37

chloroform was added to a series of glass vials such that the final concentration of DPH

in each solution was 1.175 mg/L. Copolymer or copolymer-drug conjugate stock

solutions were generated in chloroform and added to the vials resulting in copolymer or

copolymer-drug concentrations ranging from 0.125 to 500 mg/L. The samples were

vortexed and dried thoroughly under nitrogen. Once the films had dried, the vials were

heated to 60 °C, and 1 mL of double-distilled water, also at 60 °C, was added to each

vial. The hydrated samples were protected from light and stirred at 60 °C for 2 h and

then at room temperature overnight. The fluorescence emission of the samples was

measured at 350 nm (λex) and 430 nm (λem) with a dual-scanning microplate

spectrofluorometer (Spectra GeminiXS, Molecular Devices, Sunnyvale, CA).

2.3.7 Transmission Electron Microscopy (TEM) Analysis

Micelle morphology was observed using TEM with a Hitachi 7000 microscope operating

at an acceleration voltage of 75 kV (Schaumburg, IL). The micelle solutions (10 mg/mL)

were diluted in double-distilled water immediately prior to analysis and negatively

stained with a 1% uranyl acetate (UA) solution. The negative stain provided an electron-

dense layer resulting in reverse-contrast, negative-electron images. The samples mixed

with UA were deposited on copper grids that had been pre-coated with carbon and

negatively charged (Ted Pella Inc., Redding, CA). The final copolymer concentration on

the grid was 0.5 mg/mL. The copper grids were briefly left to stand to allow the solvent

to evaporate.

2.3.8 Determination of Micelle Size

The average hydrodynamic diameter of spherical micelles was determined by DLS

using a 90Plus Particle Size Analyzer (Brookhaven Instruments Corp., Holtsville, NY) at

an angle of 90° and temperature of 25 °C. The samples were diluted to 0.5 mg/mL prior

to DLS measurement. The effective mean diameter was obtained using the 90Plus

Particle Sizing Software. For aggregates with nonspherical morphologies the mean

micelle diameters were obtained by analysis of TEM images. The average diameters of

the copolymer aggregates in the TEM images were obtained using SigmaScan Pro

software (Jandel Scientific). Representative images were selected and the diameters of

a minimum of 20 spherical aggregates were recorded.

Page 53: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

38

2.3.9 Evaluation of Drug Loading and Release

The level of physically entrapped and chemically conjugated DTX was quantified using

3H-DTX (0.001% (w/w) of total DTX) with subsequent analysis of samples by scintillation

counting. For drug-loading studies, DTX was stirred with the copolymer or copolymer-

drug conjugate (1:10 free DTX to copolymer or copolymer-drug conjugate containing

24% conjugated DTX (w/w)) in DMF prior to formation of the copolymer film as

described above. The micelle solutions were centrifuged (Eppendorf 5804R) at 2000

rpm for 5 min to remove drug crystals prior to determination of drug content. For the

drug release studies and evaluation of copolymer-drug conjugate hydrolysis a dialysis

method was employed. Briefly, 1 mL aliquots of micelle samples were placed in dialysis

bags (molecular weight cutoff = 1 kDa) at a copolymer concentration of 10 mg/mL and

dialyzed against 500 mL of buffer (PBS, 0.01 M, pH 7.4) that was replaced at 1, 3, 8,

and 24 h and every 24 h thereafter. At selected time intervals, 10 μL samples were

withdrawn from the dialysis bag for scintillation counting. The volume in the dialysis bag

was also measured at each time point by withdrawing and replacing the micelle solution

using a micropipette to account for changes in total sample volume.

2.3.10 Statistical Analysis

All results were obtained from data groups of n ≥ 4 and are expressed as mean ±

standard deviation (SD). A two sample t-test was used to measure statistical

significance between pairs of results, and p < 0.05 was considered to be significant.

2.4 Results and Discussion

2.4.1 Synthesis of PEG-b-PCL Copolymer and PEG-b-PCL-DTX Copolymer-Drug Conjugate

Synthesis of the diblock copolymer, PEG-b-PCL, was achieved via metal-free cationic

ring-opening polymerization. PEG-b-PCL copolymers containing two different PCL block

molecular weights of 1000 g/mol (PEG-b-PCL(2k1k)) and 2000 g/mol (PEG-b-

PCL(2k2k)) were generated by altering the ratio of ε-CL to CH3O-PEG-OH in the

reaction solution. Following polymerization of PEG-b-PCL(2k1k), the PCL block was

functionalized by attachment of a carboxylic acid terminal group to facilitate conjugation

Page 54: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

39

of DTX. Carboxyl-terminated PEG-b-PCL (PEG-b-PCL-COOH) copolymers were then

conjugated with DTX using a well-established coupling mechanism described above

and previously reported [213]. The synthetic procedures employed for the preparation of

PEG-b-PCL copolymers and the PEG-b-PCL-DTX conjugate are outlined in Figure 2.1.

Figure 2.2 includes the 1H NMR spectra for the copolymer, DTX, and the PEG-b-PCL-

DTX copolymer-drug conjugate with corresponding peak assignments. As has been

reported for PTX, a taxane similar in structure to DTX, hydroxyl groups at the 2′, 7, and

10 positions are suitable sites for conjugation [214, 215]. However, reaction occurs

preferentially at the 2′ position due to the limited reactivity of the 7 and 10 positions

[216, 217]. The disappearance of the resonance at δ = 4.62 ppm in the DTX spectrum

corresponds to a change in resonance of the proton at the carbon 2′ position, confirming

successful conjugation of the drug to the copolymer.

Figure 2.1. Synthesis of (a) PEG-b-PCL, (b) PEG-b-PCL-COOH, and (c) PEG-b-PCL-

DTX

Page 55: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

40

Figure 2.2. 1H NMR spectra and corresponding peak assignments for PEG-b-PCL (a)

and PEG-b-PCL-COOH (b) copolymers. 1H NMR spectra of DTX (c) and PEG-b-PCL-

DTX (d).

Table 2.1 Melting temperature (Tm), critical micelle concentration (CMC), morphology,

diameter, molecular weight (Mn), and polydispersity (Mw/Mn) of copolymers and

copolymer-drug Conjugate.

aDiameter of spherical micelles as determined by TEM measurements. bDiameter

determined by DLS as an average of five measurements over a 7 day period (n = 3).

Page 56: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

41

Table 2.2. GPC retention times (Rt) of the copolymers and copolymer-drug conjugate

The resonance of this proton is likely shifted to 5.3 ppm. The chemical shift and peak

assignments are as follows: peaks at 1.38 ppm (2H, CO-CH2-CH2-CH2-CH2-CH2), 1.6

ppm (4H, CO-CH2-CH2-CH2-CH2-CH2), 2.3 ppm (2H, CO-CH2-CH2-CH2-CH2-CH2), and

4.1 ppm (2H, CO-CH2-CH2-CH2-CH2-CH2) were assigned to the PCL block, whereas the

peaks at 3.38 ppm (3H, CH3-O), and 3.65 ppm (4H, O-CH2-CH2-O) were assigned to

the PEG block. The appearance of a peak at 2.65 ppm (4H, CO-CH2-CH2-COOH)

indicates the successful addition of the carboxylic acid functional group to the PCL

block. Ratios of the hydrophobic block to the hydrophilic block were determined from the

relative intensities of the PCL proton signal at 2.3 ppm and the PEG proton signal at

3.65 ppm.

Figure 2.3. GPC chromatograms of (a) PEG-b-PCL(2k1k); (b) PEG-b-PCL(2k1k)-DTX;

(c) PEG-b-PCL(2k2k).

2.4.2 Physico-Chemical Properties of PEG-b-PCL and PEG-b-PCL-DTX Copolymer and Copolymer-Drug Conjugates

The CMC represents the concentration at which micellization occurs in aqueous

solution and provides an indication of the thermodynamic stability of the micelles. The

CMC of the PEG-b-PCL copolymer was reduced from 20.6 to 14.0 mg/L following the

Retention time (Rt), min

Page 57: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

42

conjugation of DTX. Because the micelles are subjected to significant dilution in the

blood, a low CMC is important to promote stability following i.v. administration.

However, several other factors contribute to maintaining micelle stability at

concentrations below the CMC [15]. Table 2.1 includes a summary of the physical and

thermal properties of the copolymers and copolymer-drug conjugate.

Dynamic light scattering at a fixed scattering angle (90°) cannot be used to

determine the diameter of non-spherical micelles since the correlation function analysis

is based on the assumption that the particles are spherical. Therefore, the diameter of

spherical micelles in samples containing both spheres and rods was determined by

analysis of TEM images. DLS measurements are reported for micelle solutions that

contain only spherical aggregates.

Figure 2.4. Mean diameter of PEG-b-PCL(2k1k)-DTX micelles at a concentration of 10

mg/mL (equivalent to 2.5 mg/mL DTX), as measured by DLS over a period of one week.

Successful conjugation of DTX to the copolymer was also confirmed by GPC. The

reduction in retention time (Rt) and shift in the GPC chromatogram of the PEG-b-

PCL(2k1k)-DTX conjugate relative to the chromatogram of the PEG-b-PCL(2k1k)

copolymer indicates an increase in the copolymer molecular weight (Mn) following the

attachment of DTX (Table 2.2. GPC retention times (Rt) of the copolymers and

copolymer-drug conjugate, Figure 2.3). Both PEG-b-PCL copolymers and PEG-b-

PCL(2k1k)-DTX copolymer-drug conjugate possess a monomodal molecular weight

distribution and low polydispersity (Mw/Mn; Table 1). The stability of PEG-b-PCL(2k1k)-

DTX micelles was assessed by DLS over a period of one week (Figure 2.4). No

Page 58: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

43

significant change in the mean diameter of the micelles was observed indicating good

stability and an absence of secondary particle aggregation.

2.4.3 Characterization of the Copolymer-Drug Conjugate and Drug-Loaded Micelles.

Docetaxel was conjugated to the PCL block of PEG-b-PCL(2k1k). HPLC analysis

revealed that 89% of PEG-b-PCL(2k1k) was coupled to DTX (mol %). Unreacted PEG-

b-PCL(2k1k) was not removed. The final product contained 24% DTX (wt%). It was

determined that <1% (w/w) unreacted DTX remained following purification of the

copolymer-drug conjugate. Although copolymer micelle concentrations of up to 50

mg/mL (equivalent to ∼12 mg/mL docetaxel) were generated, a 10 mg/mL PEG-b-

PCL(2k1k)-DTX concentration (equivalent to ∼2.4 mg/mL docetaxel) was selected for

further investigation based on results demonstrating the stability of the micelles formed

from the copolymer-drug conjugate at this concentration over one week (i.e., clear

solution, constant particle size, Figure 2.4)). This concentration represents an ∼480-fold

increase in the water solubility of DTX (DTX aqueous solubility: 5.0 μg/mL [218]).

2.4.4 Micelle Morphology

The morphology of BCMs in aqueous solution is controlled by a number of properties

including the nature of the copolymer blocks, ratio of hydrophobic to hydrophilic block

length, and total copolymer molecular weight [219]. A general model describing the free

energy of a copolymer molecule in a micelle, as put forth by Halperin and Alexander

[220], includes the following three components: interfacial tension (Fint), steric repulsions

between adjacent coronal polymer chains (Fcorona), and core chain deformation (Fcore).

These terms may be combined to approximate the free energy per chain in a single

micelle (Fmicelle = Fint + Fcorona + Fcore). This model has been used to explain the

morphologies produced in aqueous solution by block copolymers of varied compositions

[209, 221, 222].

In the present study, the morphology of aggregates formed from the PEG-b-PCL

copolymers and the PEG-b-PCL(2k1k)-DTX copolymer-drug conjugate were observed

by TEM (Figure 2.5). The PEG-b-PCL(2k1k) copolymer formed spherical aggregates

∼10 nm in diameter and rod-like aggregates ranging in length from 0.3 to 1.8 μm. In

Page 59: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

44

contrast, the PEG-b-PCL(2k1k)-DTX copolymer-drug conjugate formed only spherical

aggregates. The rod to sphere transition was also apparent when aggregates formed

from varying ratios of PEG-b-PCL(2k1k) and PEG-b-PCL(2k1k)-DTX were found to

contain a larger population of spheres when the PEG-b-PCL(2k1k)-DTX content was

increased. Interestingly, similar to PEG-b-PCL(2k1k) but in contrast to PEG-b-

PCL(2k1k)-DTX micelles, aggregates of PEG-b-PCL(2k2k) contained rod-like

morphologies, suggesting that the molecular weight of the hydrophobic block alone

(both PEG-b-PCL(2k2k) and PEG-b-PCL(2k1k)-DTX have similar Mn values) was not

responsible for generating the pure spherical morphology of the PEG-b-PCL(2k1k)-DTX

micelles. Furthermore, the morphology of PEG-b-PCL(2k1k) and PEG-b-PCL(2k2k)

copolymer micelles (a mixed population of rods and spheres) was not significantly

altered following physical entrapment of DTX in the micelle core. Therefore, only when

DTX was chemically conjugated to the micelle core was an alteration in micelle

morphology observed.

The morphology of copolymer aggregates containing a crystalline core is

determined by the balance of entropic forces that arise from the stretching of corona

chains and an enthalpic component associated with core chain folding [223, 224]. In the

case of brush micelles where the corona forming block is longer than the core forming

block, spherical micelles are generally favored due to the larger area available per

corona chain relative to rod or lamellar aggregate morphologies. However, an increase

in the number of crystalline folds in the core leads to a reduction in corona chain

crowding thus favoring rod-like and lamellar aggregate morphologies. For example, Du

et al. examined the influence of systematically increasing the PCL block length and,

presumably, the amount of core chain folding in PEG-b-PCL micelles while maintaining

a constant PEG chain length and observed the formation of spherical, cylindrical, worm-

like, and lamellar aggregates, respectively [225]. In a study by Zhang et al., the

crystallization tendency of the core forming block in copolymer micelles containing PEG

as the hydrophilic block and either crystalline poly(caprolactone-b-L-lactide) (P(CL-LLA))

or amorphous poly(caprolactone-b-D,L-lactide) (P(CL-DLLA)) as the hydrophobic core

forming block was demonstrated to be an important determinant of micelle morphology

[226].

Page 60: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

45

Figure 2.5 TEM images of micelles formed from PEG-b-PCL(2k1k)-DTX and PEG-b-

PCL(2k1k) in molar ratios of 9:1 (a), 7:3 (b), 3:7 (c), 1:9 (d), and PEG-b-PCL (2k2k) (e).

Micelles containing physically entrapped DTX (2% w/w): PEG-b-PCL(2k1k) + DTX (f),

PEG-b-PCL(2k2k) + DTX (g), and PEG-b-PCL(2k1k)-DTX + DTX (h). The concentration

of all copolymers and the copolymer-drug conjugate was 10 mg/mL.

The authors suggested that the high enthalpy of crystallization of the P(CL-LLA) core-

forming block was the driving force for the observed sphere to rod transition. In the

present study, PEG-b-PCL(2k1k) micelles were found to undergo a rod to sphere

transition upon chemical conjugation of DTX to the core forming block of the micelle. It

is postulated that the conjugation of the drug to the micelle core disrupts the

semicrystalline structure of PCL, thereby inhibiting core chain folding and leading to the

formation of spherical aggregates as opposed to the mixed sphere and rod

morphologies of the PEG-b-PCL(2k1k) micelles. However, further examination of the

state of the micelle core is required to support this mechanism. In agreement with the

current study, Elsabahy et al. did not observe a DTX-induced transition in the

Page 61: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

46

morphology of the PEG-b-PCL aggregates when DTX was loaded into PEG-b-

poly(butylene oxide) (PEG-b-PBO) and PEG-b-poly(styrene oxide) (PEG-b-PSO)

micelles [227]. A spherical morphology was observed for the PEG-b-PBO micelles and

a rod morphology for the PEG-b-PSO micelles before and after drug loading.

2.4.5 Drug Loading

DTX loading capacity of PEG-b-PCL(2k1k), PEG-b-PCL(2k2k), and PEG-b-PCL(2k1k)-

DTX micelles was compared. The highest drug-loading capacity was observed for PEG-

b-PCL(2k1k)-DTX micelles at all copolymer and copolymer-drug conjugate

concentrations (Figure 2.6).

The superior drug-loading capacity of PEG-b-PCL(2k1k)-DTX micelles reflects an

improvement in core-drug compatibility following conjugation of DTX to the PCL block.

This phenomenon may be attributed to the significant increase in core hydrophobicity

and the presence of π-π interactions between the core-conjugated drug and the free

drug [201]. This is in agreement with a study by Carstens et al. that found the presence

of an aromatic end group on the core-forming block of mPEG750-b-oligo(ε-

caprolactone)5 improved the loading capacity of DTX in the micelles [44]. Similarly,

Yokoyama et al. found that the conjugation of doxorubicin to the core forming block of

(PEG-b-P(Asp)) micelles led to greater physical encapsulation of doxorubicin in

comparison to micelles formed from the PEG-b-P(Asp) copolymer alone [46].

The length of the core forming block also influenced drug loading at a high

copolymer concentration (30 mg/mL) as the PEG-b-PCL(2k2k) copolymer displayed a

higher loading capacity than PEG-b-PCL(2k1k). Chemical and physical entrapment of

DTX in PEG-b-PCL(2k1k)-DTX copolymer-drug conjugate micelles resulted in a total

drug-loading capacity of 3.15 mg/mL DTX equiv. (0.75 mg/mL physically entrapped DTX

+ 2.4 mg/mL chemically conjugated DTX) at a PEG-b-PCL(2k1k)-DTX concentration of

10 mg/mL and up to 9.2 mg/mL DTX equiv. (2 mg/mL physically entrapped DTX + 7.2

mg/mL chemically conjugated DTX) at a PEG-b-PCL(2k1k)-DTX concentration of 30

mg/mL. Therefore, this formulation strategy affords a 630- to 1840-fold increase in the

aqueous solubility of DTX.

Page 62: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

47

Figure 2.6. Physical entrapment of DTX in BCMs formed from PEG-b-PCL(2k1k), PEG-

b-PCL(2k2k), and PEG-b-PCL(2k1k)-DTX at various copolymer and copolymer-drug

conjugate concentrations (n = 5). *Denotes statistically significant difference (p < 0.05).

2.4.6 Drug Release

Drug release from PEG-b-PCL(2k1k)-DTX micelles containing only chemically

conjugated DTX is expected to occur via hydrolysis of the ester bond joining DTX to the

PCL block. An initial rapid release of DTX was observed from the micelles followed by

sustained drug release over the course of one week (Figure 2.7). The apparent burst

release observed during the first 3 h represents the release of free DTX present as a

result of hydrolysis that occurred during the 3 days of stirring that is part of micelle

preparation. HPLC analysis revealed that 48.7% of the total conjugated DTX initially

present in the PEG-b-PCL(2k1k)-DTX micelles is cleaved from the copolymer during

this time (data not shown). Due to the large quantity of DTX solubilized by this system

(2.4 mg/mL), the copolymer-drug conjugate micelles may be capable of delivering

therapeutically relevant doses without the need for supplementary physical entrapment

of DTX. Because drug release occurs via hydrolysis, it is reasonable to suggest that the

rate of drug release will depend on the ability of water to penetrate the micelle core (i.e.,

the length and nature of the hydrophobic block) and the stability of the micelles in vivo.

Co

nce

ntr

atio

n o

f D

TX

, m

g/m

L

Polymer or polymer-drug conjugate concentration, mg/mL

Page 63: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

48

Therefore, it may be possible to modulate the rate of drug release by altering the length

and chemical structure of the core-forming block.

The release of physically entrapped DTX from the micelles was assessed under

sink conditions (Figure 2.8). The drug release profile of all micelle solutions displayed

rapid initial release of DTX followed by extended release over a 24 h period. PEG-b-

PCL(2k1k)-DTX micelles displayed the slowest release of physically entrapped DTX

compared to PEG-b-PCL(2k2k) and PEG-b-PCL(2k1k) micelles likely due to superior

compatibility between free DTX and the micelle core containing chemically bound DTX.

Figure 2.7. Release of chemically conjugated DTX from PEG-b-PCL-DTX micelles at

10 mg/mL (2.4 mg/mL DTX equivalent) in PBS buffer (pH = 7.4), n = 4.

Because the PEG-b-PCL(2k1k)-DTX micelles were found to be stable (i.e., they did not

aggregate or degrade significantly based on size measurements by DLS) in PBS buffer

at pH 7.4 for at least one week, the release of DTX from the micelles is likely to occur

primarily by diffusion. Furthermore, a longer hydrophobic block may provide greater

protection against both hydrolysis of the ester bond between PCL and DTX and a

greater barrier against DTX diffusion, resulting in a slower rate of drug release.

Rele

ase

of

che

mic

ally

co

nju

ga

ted

DT

X,

%

Time, days

Page 64: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

49

Figure 2.8. Drug release profile of physically entrapped DTX from copolymer micelles

formed from PEG-b-PCL(2k1k) ( ), PEG-b-PCL(2k2k) ( ), and PEG-b-PCL(2k1k)-

DTX ( ) at a concentration of 10 mg/mL (2% free DTX w/w of physically entrapped

DTX to copolymer or copolymer-drug conjugate). * and ** denote a statistically

significant difference (p < 0.05) between PEG-b-PCL(2k1k) and PEG-b-PCL(2k2k) and

PEG-b-PCL(2k1k)-DTX and both PEG-b-PCL(2k1k) and PEG-b-PCL(2k2k),

respectively. “Release of physically entrapped DTX” refers to the amount of DTX

released relative to the total physically entrapped DTX initially present and does not

include DTX released due to the hydrolysis of PEG-b-PCL(2k1k)-DTX during the

experiment.

2.5 Conclusions

DTX was coupled to the hydrophobic block of PEG-b-PCL copolymers synthesized by

metal free ring-opening polymerization. PEG-b-PCL(2k1k)-DTX copolymer-drug

conjugates formed micelles in aqueous solution at a CMC significantly lower than that of

PEG-b-PCL(2k1k). Analysis of drug loading via physical entrapment of DTX within the

copolymer-drug conjugate micelles revealed a maximum increase in DTX solubility of

1840-fold. Release of physically encapsulated DTX from PEG-b-PCL(2k1k)-DTX was

rapid, though slower than drug release from PEG-b-PCL(2k1k) micelles, suggesting that

this formulation may act primarily as a drug solubilizer. However, release of the core-

Rele

ase

of

ph

ysic

ally

en

trapp

ed

DT

X,

%

Time, h 25

Page 65: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

50

conjugated drug was slower (90% released over 1 week), demonstrating the copolymer-

drug conjugate micelle’s capacity for sustained release of DTX. The morphologies of the

BCMs were influenced by the presence of chemically conjugated DTX in the micelle

core. Block copolymer aggregates adopted a spherical morphology following the

attachment of DTX to the PCL core forming block rather than a mixed population of rod-

and sphere-like aggregates as was observed for copolymer micelles in the absence of

core conjugated DTX. Finally, an increasing population of BCMs with a rod-like

morphology was observed as the ratio of copolymer to copolymer-drug conjugate was

increased. In light of recent evidence demonstrating the influence of BCM morphology

on drug loading, circulation longevity and tissue penetration, these findings demonstrate

the importance of characterizing the morphology of similar systems in the absence and

presence of drug.

2.6 Acknowledgements

This research is funded by an NSERC operating grant to C. Allen. A. Mikhail is also

grateful to NSERC for a post graduate scholarship.

Page 66: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

51

3 Chapter 3

Cytotoxicity and Growth Inhibitory Effect of Docetaxel-loaded

Block Copolymer Micelles and Taxotere® in Monolayer and

Spheroid Cultures

Chapter 3

Page 67: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

52

The following procedures were performed by AS Mikhail: Synthesis and characterization

of PEG-b-PCL copolymers, formulation of BCM+DTX, TEM imaging, drug release

assays, histological analyses, copolymer biocompatibility assay, the clonogenic assay,

portions of the APH assay validation, establishment of spheroid growth protocol and all

data analysis and writing.

The following procedures were performed by S. Eetezadi under the guidance of AS.

Mikhail: Measurement of spheroid packing density and growth rate, the APH assay,

growth inhibition assay and illustration of Figure 3.1 and Figure 3.2.

Immunohistochemical staining was performed by the Pathology Research Program

(PRP) at the University Health Network (Toronto, ON).

This chapter was written by AS. Mikhail and edited by Dr. C. Allen.

Page 68: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

53

3.1 Abstract

While 3-D tissue models have received increasing attention over the past several

decades in the development of traditional anti-cancer therapies, their potential

application for the evaluation of advanced drug delivery systems such as

nanomedicines has been largely overlooked. In particular, new insight into drug

resistance associated with the 3-D tumor microenvironment has called into question the

use of monolayer cultures for prediction of in vivo anti-tumor activity. In this work, a

series of complementary assays was established for evaluating the in vitro efficacy of

docetaxel (DTX) -loaded block copolymer micelles (BCM+DTX) and Taxotere® in 3-D

multicellular tumor spheroid (MCTS) cultures. Spheroids were found to be significantly

more resistant to treatment than monolayer cultures in a cell line dependent manner.

Limitations in treatment efficacy were attributed to mechanisms of resistance associated

with properties of the spheroid microenvironment. DTX-loaded micelles demonstrated

greater therapeutic effect in both monolayer and spheroid cultures in comparison to

Taxotere®. Overall, this work demonstrates the use of spheroids as a viable platform for

the evaluation of nanomedicines in conditions which more closely reflect the in vivo

tumor microenvironment relative to traditional monolayer cultures. By adaptation of

traditional cell-based assays, spheroids have the potential to serve as intermediaries

between traditional in vitro and in vivo models for high-throughput assessment of

therapeutic candidates.

Page 69: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

54

3.2 Introduction

Resistance to chemotherapy is not only facilitated by processes at the cellular level, but

also by mechanisms associated with the tumor microenvironment [228, 229]. In growing

tumors, the heterogeneous architecture of the vasculature, irregular blood flow, large

intervascular distances and nature of the extracellular matrix limit the access of cells to

oxygen, nutrients, and systemically administered therapies [122, 123]. Within the tumor

interstitium, gradients in the rate of cell proliferation are established wherein rapidly

dividing cells reside close to the tumor vasculature and quiescent cells are situated

deep within the extravascular space. However, many anti-neoplastic agents exert

limited toxicity against slowly- or non-proliferating cells and are less effective in the

hypoxic and acidic microenvironments of poorly perfused tissues [27, 230]. These

therapeutic limitations are exacerbated by high interstitial fluid pressure which inhibits

the penetration of chemotherapeutic agents through the tumor interstitium by limiting

convective transport [73]. As a result cells located far from blood vessels may be less

sensitive to treatment and also be exposed to sub-therapeutic drug concentrations.

The use of in vitro cell culture is critical for rapid identification of drug candidates

and for investigating mechanisms of drug efficacy at the cellular and molecular levels. In

contrast to in vivo tumor models, in vitro cultures are better suited for systematic studies

of formulation parameters in a highly controlled environment. However, cytotoxic effects

observed in monolayer cultures often fail to translate into similar effects in vivo [231,

232]. This is due to the inherent inability of conventional cultures to account for

mechanisms of drug resistance and transport restrictions associated with the 3-D tumor

microenvironment. As such, there is increasing interest in applying in vitro tissue models

that enable rapid, high throughput screening of drug formulations for selection of lead

candidates to move forward for evaluation in vivo [28, 139, 233].

As depicted in Figure 3.1, 3-D tissue cultures such as multlicellular tumor

spheroids (MCTS) serve as an intermediary between the oversimplified structure of

monolayer cultures and the highly complex nature of in vivo tumors. MCTS cultures

possess a complex network of cell-cell contacts and advanced extracellular matrix

development, as well as pH, oxygen, metabolic and proliferative gradients analogous to

the microenvironmental conditions in poorly vascularized and avascular regions of solid

Page 70: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

55

tumors [234–236]. In general, a spheroid is comprised of an outer region of proliferating

cells which surrounds intermediate layers of quiescent cells and, if the spheroid is large

enough, a necrotic core. This arrangement parallels the radial organization of tissues

surrounding tumor blood vessels. To date, a variety of 3-D in vitro tissue models have

been applied for the study of anticancer therapies including natural and synthetic tissue

scaffolds [237, 238], multicellular layers [25, 125, 134, 239, 240], and multicellular tumor

spheroids [29, 135, 237]. MCTS are particularly relevant in the development of

nanomedicines since their penetration in 3-D tumor tissues may be restricted by

properties of the delivery vehicle. To date, however, there remain limited examples of

the use of MCTS for the evaluation of nanomedicines [150–152, 168, 241].

DTX is a potent chemotherapeutic agent that is currently administered as

Taxotere® (Sanofi-Aventis) and used for treatment of cancers of the breast, prostate,

lung, head and neck, and stomach [182]. However, Taxotere is known to be

associated with side effects that can require reductions to the administered dose [192].

Encapsulation of chemotherapeutic agents within biocompatible nanosystems such as

block copolymer micelles (BCMs) has proven to be a promising approach for mitigating

the burden of toxicity on normal tissues and increasing tumor-specific drug

accumulation [198]. In this study, traditional cell-based assays are adapted and applied

in a systematic and complementary manner for the evaluation of a DTX-loaded

nanomedicine and Taxotere® in both monolayer and MCTS cultures (Figure 3.2).

Figure 3.1. Intermediate in complexity, 3-D cultures permit the systematic, high-

throughput assessment of formulation properties in a controlled environment that

approximates important components of in vivo tumors in the absence of complex

parameters which may confound data interpretation.

Page 71: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

56

Figure 3.2. Schematic representation of assays used for analysis of formulation efficacy

in spheroids.

3.3 Materials and Methods

3.3.1 Materials

Methoxy poly(ethylene glycol) (CH3O-PEG-OH; Mn = 5000, Mw/Mn = 1.06) was

obtained from Sigma-Aldrich (Oakville, ON, Canada). ε-Caprolactone and

dichloromethane (Sigma-Aldrich) were dried using calcium hydride prior to use.

Hydrogen chloride (HCl) (1.0 M in diethyl ether), N,N-dimethylformamide (DMF), diethyl

ether, hexane and acetonitrile were used without further purification. Alexa Fluor 488

(AF488) carboxylic acid succinimidyl ester was purchased from Molecular Probes

(Eugene, OR). The hypoxia marker, EF5, and Cy5-conjugated anti-EF5 antibody were

purchased from the Department of Radiation Oncology, University of Pennsylvania,

(Philadelphia, PA). DTX was purchased from Jari Pharmaceutical Co. (Jiangsu, China).

3.3.2 Synthesis of CH3O-PEG-b-PCL (PEG-b-PCL) copolymers

PEG-b-PCL copolymer was prepared as previously described [242]. Briefly, CH3O-PEG-

OH was used to initiate the ring-opening polymerization of ε-CL in the presence of HCl.

The reaction was carried out for 24 h at room temperature prior to termination by

addition of triethylamine (TEA) and precipitation in diethyl ether and hexane (50:50,

v/v%). The product was dried under vacuum at room temperature.

Page 72: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

57

3.3.3 Preparation and characterization of BCM+DTX

PEG-b-PCL copolymer and DTX were dissolved at a copolymer:drug weight ratio of

20:1 in DMF and stirred for 30 min. DMF was evaporated under N2 at 30oC and residual

solvent was removed under vacuum. Dry copolymer-drug films were then heated to 60

°C in a water bath prior to the addition of PBS buffer (pH 7.4) at the same temperature.

Resultant micelle solutions were vortexed, stirred for 24 h at room temperature and

finally sonicated (Laboratory Supplies Co., NY) for 1 h. Undissolved drug crystals were

removed by centrifugation at 4400 g for 12 min (Eppendorf 5804R). The final copolymer

concentration was 10 mg/mL. The amount of physically entrapped DTX in BCM

samples was determined by HPLC analysis (Agilent series 1200) with UV detection

(Waters 2487) at a wavelength of 227 nm. An XTerra C18 reverse phase column was

employed with ACN/water (60/40, v/v%) as the mobile phase. Drug loading was

quantified using a calibration curve generated from a series of DTX standards.

3.3.4 Sizing of BCM+DTX

The average hydrodynamic diameter of the BCMs was determined by dynamic light

scattering (DLS) using a 90Plus Particle Size Analyzer (Brookhaven Instruments Corp.,

Holtsville, NY) at an angle of 90° and temperature of 25 °C. The samples were diluted to

a copolymer concentration of 0.5 mg/mL prior to measurement. Analysis was performed

using the 90Plus Particle Sizing Software.

3.3.5 Transmission Electron Microscopy (TEM)

BCMs were observed by TEM using a Hitachi 7000 microscope operating at an

acceleration voltage of 75 kV (Schaumburg, IL). Samples were diluted in double distilled

water immediately prior to analysis and negatively stained with a 1 % uranyl acetate

(UA) solution. The final copolymer concentration was 0.5 mg/mL. The samples were

then deposited on copper grids that had been pre-coated with carbon and negatively

charged (Ted Pella Inc., Redding, CA) and briefly air-dried prior to analysis.

3.3.6 Drug Release

The release of DTX from BCMs and Taxotere® was analyzed using a dialysis method.

Aliquots (1 mL) of BCM+DTX, DTX in DMSO and Taxotere® were placed in individual

Page 73: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

58

dialysis bags (MWCO 2 kDa, Spectra/Por, Rancho Dominguez, CA) and dialyzed

against 2 L of PBS at pH 7.4 in an incubator at 37oC ensuring that sink conditions were

maintained. At selected timepoints, 50 µL samples were withdrawn from the dialysis

bags and DTX content was measured by HPLC as described above. The volume in the

dialysis bags was also measured at each time point by withdrawing the samples,

measuring their mass and returning them in order to account for possible dilution or

concentration of the samples over time.

3.3.7 Tissue culture and growth of MCTS

Human cervical (HeLa) and colon (HT29) (ATCC, Manassas, VA) cancer cells were

incubated at 37 °C and 5% CO2 in DMEM containing 1% penicillin-streptomycin and

supplemented with 10% FBS. For growth of MCTS, cells were suspended using trypsin-

EDTA and seeded onto non-adherent 96-well round-bottomed Sumilon PrimeSurface™

plates (MS-9096U; Sumitomo Bakelite, Tokyo, Japan) in 200 µL of media per well.

During growth, 50% of the media was exchanged every other day. MCTS were grown

until they reached ~ 500 µm in diameter before use.

3.3.8 Immunohistochemical analysis of MCTS

MCTS were washed in PBS and transferred onto a vinyl specimen mold (Cryomold®,

Tissue-Tek, Sakura Finetek, CA) prior to addition of Tissue-Tek® O.C.T. compound

(Sakura Finetek, Torrance, CA). MCTS were then submersed in an isopentane bath

cooled by liquid nitrogen, cut into 5 μm thick sections using a microtome and mounted

on glass slides. Histological staining was conducted for the identification of cellular

proliferation (Ki67) and stained with hematoxylin and eosin (H&E). For identification of

hypoxic regions, MCTS were incubated with 0.5 mM EF5 and soaked in PBS prior to

cryosectioning. EF5 in the MCTS sections was identified by binding with cyanine-5-

conjugated mouse anti-EF5 (1/50) antibody. The positive signal distribution for Ki67 was

analyzed using a customized MATLAB® algorithm, as described previously [243].

Briefly, images containing Ki67-stained MCTS sections were thresholded for positive

color intensity. Using a distance map, signal intensities were summed within three

concentric regions of equidistant thickness, each equivalent to 1/3 of the MCTS radius:

Page 74: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

59

periphery, intermediate and core. The distribution of Ki67 positive signal is expressed as

a percentage of total positive signal in the MCTS section.

3.3.9 Measurement of MCTS Growth

Spheroids were imaged using a light microscope with a 10x objective lens (VWR

VistaVision TM) connected to a digital camera (VWR DV-2B). The diameter and volume

of MCTS were determined by measuring their cross-sectional area using an automated

image analysis macro developed for use with the ImageJ software package (NIH,

Bethesda, MD, Version 1.44m). The automated method was validated by comparison to

manual determination of spheroid diameter and volume (Appendix 1, Figure A1). For

the automated method, images were converted into 8-bit greyscale and the perimeter of

an individual MCTS was recognized by an automated threshold function and the image

converted to a 2-D mask. The area of the spheroid mask was recorded, applying an

image of known scale as calibration. Finally, the volume of the MCTS was calculated by

assuming a spherical shape as follows: V=4/3*π*(d/2)3. Data was fit using the Gompertz

equation for tumor growth as follows: V(t)=V(0)exp(α/β(1−exp(−β∗t))) where V(t) is

volume at time t, V(0) the initial volume and α and β are constants [244].

3.3.10 Cytotoxicity in Monolayer and Spheroid Tissue Cultures

The cytotoxicity of BCM+DTX and Taxotere® in monolayer and spheroid cell cultures

was determined using the established acid phosphatase (APH) assay which is based on

quantification of cytosolic acid phosphatase activity [245]. For this assay, p-nitrophenyl

phosphate is added in cell culture and hydrolyzed in viable cells to p-nitrophenol via

intracellular acid phosphatase. Briefly, MCTS and monolayer cultures were treated with

Taxotere® or BCM+DTX for 24 h over a range of drug concentrations. Following

treatment, cells were washed three times with fresh media and cultured for an additional

48 h. Cells were washed with PBS buffer prior to the addition of reaction buffer (2 mg/ml

p-nitrophenyl phosphate (Sigma) and 0.1 % v/v Triton-X-100 in 0.1 M sodium acetate

buffer at pH 5.5). Following incubation for 2 h in the cell incubator, 1 M sodium

hydroxide was added to each well and acid phosphatase activity was determined by

measuring the UV absorbance at 405 nm using an automated 96-well plate reader

(SpectraMax Plus 384, Molecular Devices, Sunnyvale, CA). Results were normalized to

Page 75: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

60

controls as follows: % viability = (Atreatment – Amedia)/(Acontrol – Amedia), where A = mean

absorbance. All experiments were performed in triplicate.

3.3.11 Growth Inhibition of MCTS

BCM+DTX or Taxotere® was administered to spheroids for 24 h at a DTX equivalent

concentration of 2, 20 or 200 ng/mL. The culture media was replaced following the

incubation period. Subsequently, half of the culture media was replaced in each well

every other day. Images of spheroids were captured using a light microscope with a 10x

objective lens (VWR VistaVisionTM) connected to a digital camera (VWR DV-2B).

Spheroid size was determined by measuring their 2-D cross-sectional area using the

automated image analysis method described previously. The data are reported as the

mean volume of six spheroids ± SD.

3.3.12 Clonogenic Survival Assay

The clonogenic assay was used to determine the ability of single cells to replicate and

form colonies (>50 cells) following exposure to BCM+DTX and Taxotere®. Single cell

suspensions derived from monolayers and disaggregated spheroids were diluted in

culture media and cells were plated in 6-well plates in desired numbers. MCTS were

disaggregated by incubation in trypsin-EDTA for 10 min, followed by gentle agitation.

Drug formulations were added immediately at a DTX equivalent concentration of 20

ng/mL. This concentration was selected due to its intermediate cytotoxicity and growth

inhibitory effect. After treatment for 24 h, cells were washed with PBS and 2 mL of fresh

media was added to each well. For treatment of intact spheroids, drug formulations

were added directly into wells containing individual MCTS. After 24 h, MCTS were

collected and rinsed in PBS, suspended as single-cell suspensions in fresh media

following trypsinization, and seeded onto 6-well plates. Cells were incubated for 14-16

days prior to fixation with methanol and staining with 1% crystal violet solution. Colonies

consisting of at least 50 cells were counted. The surviving fraction (SF) was expressed

as the number of colonies divided by the product of the number of cells plated and the

plating efficiency. The plating efficiency was determined by dividing the number of

colonies formed by the number of cells plated for untreated controls.

Page 76: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

61

3.4 Results

3.4.1 Characterization of BCM+DTX

PEG-b-PCL copolymer micelles containing physically encapsulated DTX were

generated with a spherical morphology (Figure 3.3a). The size distribution of the

micelles was monomodal with an average hydrodynamic diameter of 54.6 ± 4.9 nm

(Figure 3.3b). Drug loading resulted in a final DTX equivalent concentration of 258.7 ±

35.5 µg/mL at a loading efficiency of 52.7 ± 7.1 %. Release of DTX from BCMs occurred

over the course of 24h with 74% of the drug released by 12 h. In contrast, the transport

of docetaxel from Taxotere® into the surrounding PBS release media was complete by

12h.

0 50 100

0

20

40

60

80

100

Hydrodynamic diameter (nm)

Re

lative

nu

mb

er

Figure 3.3. a) Transmission electron micrograph (Scale bar in represents 100 nm) and

b) size distribution of BCM+DTX as determined by DLS.

a) b)

Page 77: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

62

Figure 3.4. Release of DTX from dialysis bags containing BCM+DTX, Taxotere®, and

DTX in DMSO, n = 3. The data is fit to a two-phase exponential association model.

3.4.2 Growth of MCTS

Spheroids were grown using a modified liquid overlay technique by seeding HT29 or

HeLa cells onto non-adherent U-bottom tissue culture wells without the use of an

agarose surface coating. MCTS were spherical, followed a sigmoidal growth profile, and

were grown until a diameter of ~ 500 µm was reached prior to use (Figure 3.5).

0 5.010 7 1.0108 1.510 80

110 0 4

210 0 4

310 0 4

410 0 4

510 0 4

HeLa

HT29

Volume per spheroid (µm3)

Cells

per

sphero

id

0 5 10 15 20 25 300

210 0 8

410 0 8

610 0 8

810 0 8

HT29

HeLa

Days

MC

TS

vo

lum

e (m

3)

Figure 3.5. a) Cell packing density (number of cells per spheroid) of HeLa and HT29

MCTS, n=12. Dashed line indicates the volume of MCTS used in studies. b) Growth of

HeLa and HT29 MCTS, n=6. Data was fit using the Gompertz equation for tumor

growth. The dashed lines indicate spheroid properties used in the studies.

a) b)

Page 78: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

63

3.4.3 Cytotoxicity in monolayer and MCTS culture

The metabolic activity of cells following exposure to BCM+DTX or Taxotere® was

assessed using the APH assay (Figure 3.6). This assay was validated by assessing the

relationship between UV absorbance and cell number in both monolayer and spheroid

cultures. If the production of p-nitrophenol (i.e. the activity of acid phosphatase) is to be

used as a measure of cellular metabolic activity and, in this capacity reflect cell viability,

the UV absorption per cell must be independent of spheroid size and proliferative

activity (i.e. cell cycle distribution). As shown in Figure A2 (Appendix 1), a linear

relationship was observed between UV absorbance and spheroid diameter (i.e. cell

number). A well-established tetrazolium salt-based assay (WST-8) was also evaluated

and did not yield a similar correlation (Appendix 1, Figure A3). The NADH/NADPH-

dependent production of formazan per cell was reduced (i.e. a non-linear relationship

between UV absorbance and cell number was established) as the cell number per

spheroid increased.

Spheroid cultures were less sensitive to BCM+DTX and Taxotere® relative to

their monolayer counterparts. HeLa cells were less responsive to treatment with either

BCM+DTX or Taxotere® than HT29 cells in monolayer culture, while the opposite was

observed for cells grown as spheroids. The IC50 of HeLa and HT29 monolayer cultures

treated with BCM+DTX were 0.37+/-0.01 and 0.01+/-0.004 ng/mL, respectively. When

treated with Taxotere®, the IC50 of HeLa and HT29 monolayer cultures were 2.2+/-0.5

and 0.09+/-0.01 ng/mL, respectively. Only HeLa spheroids demonstrated a measurable

IC50 (1396 ± 198 ng/mL for BCM+DTX and 1558 ± 103 ng/mL for Taxotere®) whereas

HT29 MCTS maintained a viability above 80% at all drug concentrations.

Page 79: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

64

10 - 6 10 - 4 10 - 2 10 0 10 2 10 4 1060

50

100 ML: BCM+DTX

ML: Taxotere

MCTS: Taxotere

MCTS: BCM+DTX

[DTX] (ng/ml)

Cell

surv

ival,

%

10 - 6 10 - 4 10 - 2 100 102 104 1060

50

100

ML: Taxotere

ML: BCM+DTX

MCTS: Taxotere

MCTS: BCM+DTX

[DTX] (ng/ml)

Cell

surv

ival,

%

1 10 100 1000 100000

50

100

HT29

HeLa

Concentration (mg/ml)

Cell

viabili

ty,

%

Figure 3.6. Cytotoxicity of Taxotere® and BCM+DTX in a) HeLa and b) HT29 monolayer

and MCTS cultures as measured using the APH assay. Data is expressed as the

percent viability relative to untreated controls and fit to the Hill equation. c) Cytotoxicity

of blank PEG-b-PCL micelles as a function of copolymer concentration. Each plot

represents the mean of three independent experiments ± SD (n=3).

3.4.4 Inhibition of MCTS growth

MCTS volume was plotted over a 30 day period following a 24 h incubation with 2, 20,

and 200 ng/mL BCM+DTX or Taxotere® (Figure 3.7). The growth of HeLa MCTS was

completely impeded following incubation with DTX concentrations of 20 and 200 ng/mL.

No significant difference in growth was observed following exposure to 2 ng/mL of DTX

relative to untreated controls. In the case of HT29 MCTS, incubation with 20 ng/mL of

BCM+DTX and Taxotere® only resulted in a partial reduction in MCTS volume.

Following re-treatment 14 days after the initial drug incubation, BCM+DTX

a) b)

c)

Cell

via

bili

ty, %

Cell

via

bili

ty, %

C

ell

via

bili

ty, %

Page 80: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

65

Figure 3.7. a) Representative images of HeLa and HT29 MCTS following treatment with

BCM+DTX at a concentration of 20 ng/mL. Bars represent 100 µm. Growth inhibition of

HeLa and HT29 MCTS by BCM+DTX and Taxotere® at concentrations of 2, 20 and 200

ng/mL. Cells were re-treated after two weeks (arrow). Plots c) and e) are expanded

regions of plots b) and d). Data is expressed as the mean volume of six spheroids (n=6)

± SD. “*” represents a statistically significant difference, p < 0.05.

b) c)

d) e)

a)

Page 81: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

66

demonstrated greater inhibition of HT29 MCTS growth than Taxotere® at a

concentration of 20 ng/mL. Similarly to HeLa MCTS, complete inhibition of growth was

observed following incubation with 200 ng/mL of drug.

3.4.5 Immunohistochemistry

Immunohistochemical analysis of MCTS cross-sections was performed in order to

identify regions of necrosis, cellular proliferation and hypoxia (Figure 3.8). Staining with

the proliferation marker Ki67 revealed a greater proportion of proliferative cells in HeLa

MCTS relative to HT29. Image analysis revealed that 88.6% of proliferating cells were

located within the periphery of HT29 MCTS (Figure 3.9).

Figure 3.8. HeLa (a-c) and HT29 (d-f) MCTS cross-sections stained with H&E (a, d),

Ki67 proliferation marker (b, e) and EF5 (c, f), a marker of hypoxia. Scale bars represent

100 µm.

a)

a

b)

a

d) a

e)

a

f)

a

c)

a

Page 82: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

67

Figure 3.9. Ki67 positive signal distribution relative to radial position in a) HeLa and b)

HT29 MCTS as a percent of total positive stain, n=6.

Figure 3.10. Relative spatial distribution of features of HeLa and HT29 MCTS

microenvironments. “++”, “+” , and “-”, indicate high, intermediate and low levels of the

corresponding feature, respectively.

In contrast, only 51% of the total proliferating cells were located in the periphery of HeLa

MCTS and 25% and 24% were located in the intermediate region and core,

respectively. Signs of necrosis were visible following staining with H&E in HT29 MCTS.

Incubation of MCTS with EF5 allowed for identification of regions of hypoxia following

exposure to Cy5- conjugated anti-EF5 antibody. Hypoxic conditions were observed

a) a

b)

a

Page 83: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

68

primarily in the core and intermediate regions of HT29 MCTS. In contrast, HeLa MCTS

did not demonstrate any regional hypoxia. The relative distributions of cellular

proliferation, hypoxia and necrosis in the MCTS are summarized in Figure 3.10.

3.4.6 Clonogenic Survival

The surviving fractions (SF) of HeLa and HT29 cells were determined following

treatment with BCM+DTX or Taxotere® as monolayer and MCTS cultures (Figure 3.11).

Figure 3.11. Clonogenic survival of HeLa and HT29 cells following 24 h treatment with

20 ng/mL of BCM+DTX or Taxotere® (TAX) as monolayers, disaggregated spheroids

and intact spheroids.

The SF was higher for all MCTS cultures relative to monolayers. HeLa cells were less

sensitive to treatment than HT29 when cultured as monolayers but more sensitive than

HT29 cells when the cells were exposed to treatment as MCTS. In all cases, the SF

was lower when treated with BCM+DTX compared to Taxotere®. Furthermore, cells

exposed to treatment immediately following MCTS disaggregation demonstrated

residual resistance to both BCM+DTX and Taxotere®.

Page 84: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

69

3.5 Discussion

In recent years, the tumor microenvironment has been implicated in the coordination of

tumor growth, metastasis and resistance to anti-cancer therapies [246, 247]. Therefore,

effective evaluation of novel therapeutic agents requires the use of tissue models which

closely mimic native conditions within the intratumoral space. Yet, the vast majority of

chemotherapeutic agents are screened for cytotoxic effects in monolayer cultures which

do not account for critical mechanisms of drug resistance associated with the tumor

microenvironment. As a result, these models poorly predict a drug’s therapeutic efficacy

in vivo [231]. In contrast, 3-D MCTS better approximate the state of cancer cells in their

native environment and thus can be used to more accurately estimate a drug’s

therapeutic potential. A variety of methods have been used to grow MCTS for use in

cancer research including spinning culture flasks [248], hanging drops [249], liquid

overlay on agarose [250], micropatterned plates [251], and recently, using inter-cellular

linkers [252]. However, many of these techniques are impractical, time-consuming, and

involve delicate handling procedures, limiting the use of the MCTS model in drug

screening and development. In addition, practical application of traditional cell-based

assays in MCTS cultures remains poorly established. In the current study, the

performance of BCM+DTX and Taxotere® was evaluated in monolayer and MCTS

cultures using a straightforward and robust MCTS culture technique and by adaptation

of conventional cytotoxicity and survival assays.

MCTS grew according to sigmoidal growth patterns reflective of tumor growth in

vivo (Figure 3.5) and possessed histological features similar to those of the native tumor

microenvironment including gradients in cell proliferation and regions of hypoxia and

necrosis (Figure 3.8, Appendix 1 - Figure A4). Cells grown in spheroid cultures

demonstrated considerably greater resistance to treatment with BCM+DTX or Taxotere®

relative to cells grown in monolayer cultures. This may be a result of the limited

exposure of cells within MCTS to treatment due to poor penetration of DTX or BCMs,

the limited sensitivity of cells within MCTS to DTX due to a reduction in cellular

proliferation and/or resistance associated with 3-D cell adhesion (i.e. contact effect). In

a study by Kyle et al., the penetration half-depth (the depth from the surface at which

the amount of drug falls to half of its maximum concentration) of DTX in multicellular

Page 85: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

70

layers was found to be < 25 µm following a 2h incubation at a concentration of 0.3 µM

[253]. Peak tissue levels did not increase proportionally following a 10-fold increase in

drug concentration although the depth of penetration was improved indicating partial

saturation of tissue binding. Therefore, it is likely that high intracellular binding and

consumption of DTX by peripheral cells in the MCTS limits the toxicity to cells distant

from the surface. For drugs which are rapidly consumed by cells, encapsulation in

BCMs which minimize interactions and uptake by cells may improve drug penetration

[254]. For example, Pun et al. reported ameliorated penetration of doxorubicin into

MCTS when encapsulated in poly(ethylene oxide)-b-poly[(R)-3-hydroxybutyrate]-b-

poly(ethylene oxide) (PEO-b-PHB-b-PEO) triblock copolymer micelles [150]. However,

BCMs which penetrate poorly through tissues may limit the penetration of the

encapsulated drug. Overall, the extent to which the BCMs influence drug penetration

will depend on the relative rates of drug release and BCM penetration in the MCTS. We

have previously demonstrated that 55 nm PEG-b-PCL BCMs can achieve a

homogeneous distribution in MCTS following a 24 h incubation [255].

In addition to potential limitations in MCTS penetration associated with the drug

and BCMs, the discrepancy between MCTS and monolayer cytotoxicity may also be a

result of drug resistance imparted by the MCTS microenvironment. A marked decrease

in the proportion of proliferating cells was observed in MCTS with increasing depth from

the surface (Figure 3.9). Since DTX exerts its therapeutic effect on cycling cells, cells

located near the MCTS surface will respond to treatment similarly to cells cultured as

monolayers. By contrast, quiescent cells that are located in the intermediate and core

regions of the MCTS will be less sensitive to treatment. This notion is supported by the

observation that cells exposed to treatment immediately following disaggregation of

MCTS demonstrated greater clonogenic survival than monolayer cells, but less than

cells treated as intact MCTS. Therefore, there exists a population of cells within the

MCTS that is more resistant to treatment than cells cultured as monolayers even in the

absence of any physical barrier to drug penetration. The limited sensitivity of MCTS to

treatment is likely a result of both restricted transport and mechanisms of drug

resistance associated with the MCTS microenvironment.

The extent to which culturing cells as MCTS influenced the therapeutic effect of

BCM+DTX and Taxotere® relative to monolayers was found to be cell-line specific. In

Page 86: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

71

monolayer cultures, BCM+DTX and Taxotere® demonstrated greater cytotoxicity against

HT29 cells relative to HeLa cells. However, culturing cells as MCTS imparted a greater

enhancement in therapeutic resistance (i.e. greater increase in IC50) to HT29 cells than

to HeLa cells. We have previously shown significantly greater penetration of HeLa

MCTS than HT29 MCTS by BCMs due to the former’s lower cell packing density and

large intercellular channels [255]. In the current study, significant cell line-dependent

differences in MCTS microenvironment were observed. Limited permeability of HT29

MCTS and/or high consumption of nutrients and oxygen by peripheral cells was

reflected by the presence of central hypoxia and necrosis. Furthermore, HT29 MCTS

contained a greater proportion of non-proliferating cells relative to HeLa MCTS. It is

likely that some quiescent cells within the MCTS retained their clonogenic potential

following exposure to inadequate amounts of DTX and were capable of recommencing

proliferative activity when re-plated as monolayers. The greater clonogenic potential of

HeLa cells following disaggregation of MCTS relative to HT29 cells likely reflects the

greater sensitivity of HT29 monolayer cells to DTX rather than greater residual

resistance of MCTS-derived HeLa cells.

One of the important advantages of the MCTS model is that it allows for

treatment efficacy to be observed over an extended period of time. In order to evaluate

the potential of surviving cells to repopulate MCTS, the growth of MCTS following

treatment with BCM+DTX and Taxotere® was evaluated for 28 days with treatment re-

applied after 14 days. The results of this study demonstrate both dose- and time-

dependent changes in MCTS growth following incubation with the drug formulations.

Near complete elimination of HeLa MCTS was observed following treatment at 20

ng/mL or greater with either BCM+DTX or Taxotere®. In contrast, only partial growth

inhibition was observed in HT29 MCTS when exposed to the same concentration. This

observation is consistent with the results obtained from the cytotoxicity and clonogenic

assays in which HT29 MCTS demonstrated greater resistance to treatment relative to

HeLa MCTS. A slight inhibitory effect in HT29 MCTS following administration of DTX

formulations at 2 ng/mL was likely due to the cytotoxicity and shedding of surface cells,

consistent with the response of HT29 cells to treatment in monolayer cultures. In

addition, the apparent discrepancy between the limited cytotoxicity in HT29 spheroids

revealed using the APH assay (measured 2 days post drug incubation) and the marked

Page 87: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

72

growth inhibition at 20 ng/mL is consistent with the observed 4 day delay in growth

inhibitory effect. Interestingly, little difference in spheroid growth inhibition was observed

between BCM+DTX and Taxotere® following initial treatment. It should be noted,

however, that following retreatment after 14 days of culture, BCM+DTX demonstrated a

greater growth inhibitory effect relative to Taxotere®.

Several factors may have contributed to the greater cytotoxicity of BCM+DTX

relative to Taxotere® in monolayer and MCTS cultures. It has been hypothesized that

DTX is taken up more rapidly by cells following release from BCMs in close proximity to

the cell membrane due to an increase in the local transmembrane concentration

gradient [256–258]. Slower efflux of BCM-encapsulated DTX relative to free DTX, by

avoidance of membrane efflux pumps, may also contribute to the greater therapeutic

effect of the DTX-loaded BCMs [111, 259, 260]. For instance, the level of expression of

certain genes and their associated transport proteins, such as P-glycoprotein (P-gp) or

multidrug resistance associated protein 7 (MRP7), for which docetaxel is a substrate,

may influence differences in cytotoxicity between free docetaxel and docetaxel

encapsulated within micelles. Furthermore, the level of expression of transporters may

vary over time, may change with repetitive exposure to a drug, or may be different when

cells are cultured as spheroids relative to monolayers [261, 262]. However, further

investigation is required to fully elucidate the mechanism of cytotoxicity that lead to

enhanced therapeutic effects of BCM+DTX relative to Taxotere® in vitro.

Page 88: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

73

3.6 Conclusions

Overall, as outlined in Figure 3.2, the three assays employed in this study provide

complementary information on the therapeutic potential of drug formulations.

Importantly, comparison of results obtained in monolayer and spheroid cultures

demonstrated the important influence of the microenvironment and 3-D tissue structure

on formulation efficacy. Therefore, 3-D cultures such as MCTS may serve as important

tools for investigating the performance of nanomedicines in environments that more

closely mimic intratumoral conditions in vivo. However, while spheroids share several

important structural and microenvironmental properties with native tumors, there are

important differences which may limit the extent to which this in vitro model can be used

to predict drug efficacy in vivo. Notably, the MCTS model does not account for the

potential influence of convective flow or presence of stromal cells on drug and

nanoparticle transport. Despite these limitations, evaluation of formulation efficacy in

spheroids rather than monolayer cultures is expected to more accurately reflect

therapeutic performance in vivo.

Page 89: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

74

4 Chapter 4

Image-based Analysis of the Time-dependent Penetration of

Polymeric Micelles in Multicellular Tumor Spheroids and Tumor

Xenografts

Chapter 4

Page 90: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

75

This chapter is a copy of: “AS. Mikhail, S. Eetezadi, J. Stewart and C. Allen. “Image-

based Analysis of the Time-dependent Penetration of Polymeric Micelles in Multicellular

Tumor Spheroids and Tumor Xenografts”. Submitted, 2012.

All experiments and data analyses were performed by AS. Mikhail. J. Stewart wrote the

MATLAB® algorithms. S. Eetezadi illustrated Figure 4.1 and Figure 4.3.

Immunohistochemical staining was performed by the Pathology Research Program

(PRP) at the University Health Network (Toronto, ON).

This chapter was written by AS. Mikhail and edited by Dr. C. Allen.

Page 91: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

76

4.1 Abstract

Although the selective tumor accumulation of systemically administered nanomedicines

has been well documented, corresponding improvements in therapeutic efficacy have

often been incommensurate. A significant reason for this is their heterogeneous

intratumoral distribution and poor interstitial penetration which limits the exposure of

cancer cells to therapy. In the present work, fluorescence-based methodologies were

used for the computational assessment of polymeric micelle penetration in multicellular

tumor spheroids (MCTS) and tumor xenografts originating from human cervical (HeLa)

and colon (HT29) cancer cells. The influence of micelle size on tissue penetration was

examined at multiple time points in vitro and in vivo. Penetration of 15 nm micelles in

MCTS was rapid and resulted in a homogeneous distribution from the periphery to the

core while 55 nm micelles underwent delayed, time-dependent penetration of MCTS.

Following i.v administration of micelles to mice bearing tumor xenografts, penetration of

55 nm micelles into the tumor interstitium was slow but progressive while 15 nm

micelles exhibited rapid penetration with visible clearance by 24 h post injection. Higher

concentrations of penetrating micelles in HeLa MCTS and tumor xenografts relative to

HT29 models were attributed to differences in tissue architecture. These results show

that micelle penetration is size-, tumor type- and time-dependent. Together, these

findings demonstrate the potential applicability of 3-dimensional MCTS models for the

high-throughput assessment of nanoparticle tissue transport in vitro.

Page 92: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

77

4.2 Introduction

Poor penetration of chemotherapeutic agents in solid tumors has been recognized as

one of the major challenges limiting the efficacy of macromolecular and nanoparticle-

based cancer therapies [122]. The successful distribution of therapeutic agents

throughout a tumor remains a challenge due to the heterogeneous architecture of the

vascular network, abnormal blood flow, dense nature of the extracellular matrix and

interstitial hypertension [73, 122, 263]. Given that lymphatic drainage is compromised

throughout much of the tumor due to compression of lymphatic vessels by cancer cells

and leakage of excess fluid from the tumor vasculature, interstitial fluid pressure (IFP) is

elevated particularly within the tumor core [264]. Since high IFP levels can approach or

exceed intra-capillary pressure, diffusion is believed to be the primary mechanism of

transvascular and interstitial transport throughout much of the tumor mass [90].

Supramolecular assemblies of amphiphilic copolymers known as block

copolymer micelles (BCMs) constitute a promising class of nanoparticles for the

systemic delivery of chemotherapy [6, 22, 23, 198, 265, 266]. In contrast to conventional

small molecule surfactants that are commonly used as solubilizing excipients,

biocompatible copolymers such as poly(ethylene glycol)-b-poly(ε-caprolactone) (PEG-b-

PCL) result in micelles with a greater degree of thermodynamic and kinetic stability

[242]. Stable, long-circulating BCMs can accumulate preferentially within a tumor via the

enhanced permeability and retention (EPR) effect, resulting in tumor-specific delivery of

the encapsulated drug [69, 267].

Indeed, the systemic delivery of anti-cancer drugs via nanoparticle-encapsulation

has been shown to increase their tumor accumulation relative to conventional

chemotherapeutic agents [3, 6, 268, 269]. However, seminal research pertaining to the

intratumoral fate of macromolecules and nanoparticles suggests that they are

heterogeneously distributed and display limited penetration into avascular tumor

compartments [80, 84, 95, 120–122, 132]. While evaluating chemotherapeutic

nanomedicines in conventional tissue culture provides insight into therapeutic

responses at the cellular level, this approach often fails to accurately predict a

formulation’s efficacy in vivo. This is largely due to the simplified structure of monolayer

cultures which does not account for multicellular mechanisms of drug resistance and

Page 93: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

78

transport restrictions commonly associated with 3-D tumor tissues. In contrast, 3-D

cultures such as multicellular tumor spheroids possess an extracellular matrix as well as

pH, oxygen, metabolic and proliferative gradients analogous to the microenvironment in

hypovascular and avascular regions of solid tumors [234–236]. MCTS are composed of

an outer layer of proliferating cells surrounding a quiescent cell layer and necrotic core,

mimicking the radial organization of tissues adjacent to a tumor blood vessel (Figure

4.1) [29]. As such, MCTS provide a useful platform for studying diffusion-based

transport of nanoparticles with respect to the structural and microenvironmental

heterogeneity commonly associated with solid tumors.

Figure 4.1. Schematic representation of tumor and MCTS cross-sections. MCTS are

composed of a peripheral region of proliferating cells surrounding an intermediate

region of quiescent cells and a nutrient deprived or necrotic core mimicking the radial

organization of tissues adjacent to a tumor blood vessel.

Herein we directly compare the spatial and temporal penetration of block copolymer

micelles using a 3-D in vitro tissue model and corresponding human tumor xenografts.

The influences of BCM size and tumor tissue type on interstitial transport are also

assessed. The capacity of the spheroid model to predict the in vivo tissue penetration

of BCMs is evaluated with the objective of validating its potential as a high-throughput in

Page 94: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

79

vitro platform for optimization of nanoparticle transport properties. Computational image

analysis is used to assess BCM penetration in MCTS and tumor xenografts with the

ultimate goal of developing rational design criteria for improving the transport and

therapeutic efficacy of drug-loaded nanoparticles.

4.3 Methods

4.3.1 Materials

Methoxy poly(ethylene glycol) of different molecular weights (CH3O-PEG-OH; Mn =

5000, Mw/Mn = 1.06; Mn = 2000, Mw/Mn = 1.06) were obtained from Sigma-Aldrich

(Oakville, ON, Canada). ε-Caprolactone and dichloromethane were purchased from

Sigma-Aldrich and dried using calcium hydride prior to use. Hydrogen chloride (1.0 M in

diethyl ether), N,N-dimethylformamide (DMF), diethyl ether, hexane and acetonitrile

were also purchased from Sigma-Aldrich and used without further purification. Alexa

Fluor® 488 (AF488) carboxylic acid succinimidyl ester was purchased from Molecular

Probes (Eugene, OR).

4.3.2 Synthesis of CH3O-PEG-b-PCL (PEG-b-PCL) and Alexa Fluor® 488-PEG-b-PCL (AF488-PEG-b-PCL) copolymers

PEG-b-PCL copolymers were prepared by metal-free cationic ring opening

polymerization of ε-CL with CH3O-PEG-OH as macroinitiator in the presence of HCl

using an established method previously reported [242]. For the synthesis of

fluorescently labelled copolymer (AF488-PEG-b-PCL), NH2-PEG-OH was used in place

of CH3O-PEG-OH for polymerization. Alexa Fluor® carboxylic acid succinimidyl ester

(AF488) was then conjugated to NH2-PEG-b-PCL (2x molar excess with respect to

AF488) in DMSO with TEA (30x molar excess with respect to NH2-PEG-b-PCL) for 24 h

at room temperature. AF488-PEG-b-PCL was purified using a silica gel 100 C8- reverse

phase column (Sigma-Aldrich, Oakville, ON, CA) packed in 50/50 acetonitrile/water and

equilibrated with water prior to injection of the reaction mixture. Unbound AF488 was

eluted using water followed by elution of the AF488-PEG-b-PCL conjugate using a

90/10 solution of acetonitrile/water. The purity of the conjugate was assessed by high

performance liquid chromatography (HPLC) using a Styragel HR2 column (effective

molecular-weight range of 500 to 20,000; Waters, MA) connected to a fluorescence

Page 95: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

80

detector (Agilent Technologies Inc., CA) with excitation and emission wavelengths of

λex=495 nm and λem=519 nm, respectively. Filtered HPLC grade DMF was used as the

mobile phase with a flow rate of 0.5 mL/min.

4.3.3 Preparation and characterization of BCMs

Micelles were prepared from PEG2000-b-PCL1000 or PEG5000-b-PCL5000 copolymers by

hydration of thin films. First, copolymers were dissolved in DMF and stirred overnight.

For generation of fluorescent BCMs, AF488-conjugated PEG2000-b-PCL1000 or PEG5000-

b-PCL5000 copolymers dissolved in DMF were added to the unlabeled copolymer in

order to achieve a final AF488 concentration of 1 µM for in vitro studies and 10 µM for in

vivo studies in each BCM sample. DMF was evaporated under N2 at 30oC and trace

solvent was removed under vacuum. Dry copolymer films were then heated to 60 °C in

a water bath prior to the addition of PBS buffer (pH 7.4) at the same temperature with

thorough vortexing. Resultant micelle solutions were stirred for 24 h and then sonicated

for 1 h (Laboratory Supplies Co., NY). The critical micelle concentration (CMC) of the

copolymers was determined by an established fluorescence-based method as

previously described [242].

4.3.4 Transmission electron microscopy (TEM)

The size and morphology of the BCMs were determined by TEM using a Hitachi 7000

microscope (Schaumburg, IL) operating at an acceleration voltage of 75 kV. BCM

solutions (10 mg/mL) were diluted in double distilled water immediately prior to analysis

and negatively stained with a 1 % uranyl acetate (UA) solution. The negative stain

provided an electron dense layer resulting in reverse-contrast, negative electron

images. The samples mixed with UA were deposited on copper grids (Ted Pella Inc.,

Redding, CA) that had been pre-coated with carbon and negatively charged. The final

copolymer concentration on the grid was 0.5 mg/mL. The copper grids were briefly left

to stand to allow the solvent to evaporate. The average sizes of BCMs were obtained by

measuring the diameter of a minimum of 20 BCMs using TEM images and SigmaScan

Pro software (Jandel Scientific).

Page 96: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

81

4.3.5 Tissue culture and growth of MCTS

Human cervical (HeLa) and colon (HT29) cancer cells were cultured in DMEM

containing 1% penicillin-streptomycin solution supplemented with 10% FBS. Cells were

incubated at 37 °C and 5% CO2 and harvested by trypsinization with trypsin-EDTA. To

initiate growth of MCTS, monolayer cells were trypsinized and seeded onto non-

adherent 96-well round-bottomed Sumilon PrimeSurface™ plates (MS-9096U;

Sumitomo Bakelite, Tokyo, Japan). Following cellular aggregation and growth, each

well contained a single spheroid. During growth, 50% of the culture medium was

exchanged every other day.

4.3.6 Penetration of BCMs in MCTS

MCTS were incubated with 15 (BCM-15) or 55 (BCM-55) nm fluorescently labeled

BCMs in separate wells for 1 and 24 h at a copolymer concentration of 1 mg/mL in each

well. Following incubation with BCMs, MCTS were washed in PBS and transferred onto

a vinyl specimen mold (Cryomold®, Tissue-Tek, Sakura Finetek, CA). Samples were air-

dried for 5 minutes prior to the addition of Tissue-Tek® O.C.T. compound (Sakura

Finetek, Torrance, CA). MCTS were frozen by submersion in an isopentane bath cooled

by liquid nitrogen. MCTS samples were then cut into 5 μm thick sections and mounted

on glass slides. Images were acquired at 10x magnification using an Olympus BX50

upright fluorescence microscope (Olympus, PA) equipped with an EXFO fluorescence

illumination source and Semrock Quad Sedat filter set. A Photometrics CoolSnap HQ2

CCD camera was used to capture images with fixed exposure and contrast settings.

The fluorescence intensity per unit area was calculated with respect to the distance

from the MCTS surface using a custom MATLAB (MathWorks Inc., MA) algorithm. The

total area-normalized signal intensity in arbitrary units is then calculated within three

concentric regions of the MCTS: periphery, intermediate and core (Figure 4.1).

4.3.7 Animals and growth of tumor xenografts

Four week-old nude female CD1 mice (Charles River, MA) were housed at the Animal

Resource Centre of the University Health Network (Toronto, ON, Canada) where they

were given water and food ad libitum. All procedures were carried out under a protocol

approved by the Animal Care Committee of the University Health Network. HeLa

Page 97: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

82

(7.5x106) or HT29 (1.25x106) cells were suspended in DMEM medium and injected

subcutaneously into the right hind flank of the mice. The tumor volume was determined

using calipers to measure the diameter of each tumor and applying the formula of an

ellipsoid v = a x b2 / 2, where v is the tumor volume and a and b are the largest and

smallest diameters, respectively. Tumors were allowed to grow for two weeks until

reaching a size of ~ 60-80 mm3 prior to use of the animals in the studies outlined below.

4.3.8 Penetration of BCMs in tumor xenografts

Tumor-bearing animals were administered BCM-15 or BCM-55 formulations i.v.

(copolymer concentration of 10 mg/mL (10 µM conjugated AF488)) and sacrificed at 1,

6, or 24 h post injection. In order to identify perfused blood vessels within the tumors,

each animal received a 100 µL injection of Hoechst 33342 (10 mg/mL) (Sigma-Aldrich)

1 minute prior to sacrifice. Tumors were excised, embedded in Tissue-Tek® O.C.T.

compound and frozen immediately in a bath of isopentane cooled by liquid nitrogen

prior to tissue sectioning and analysis. Frozen tumor blocks were cut into 5 µm-thick

sections separated by 200 µm within each tumor. Fluorescence images were captured

as described above. Images of entire tumor sections were generated by tiling and

stitching using MetaMorph imaging software (Molecular Devices Inc., CA).

First, images identifying the distribution of BCMs and Hoechst were captured

using fluorescein isothiocyanate (FITC) and 4′,6-diamidino-2-phenylindole (DAPI) filter

sets, respectively. Sections were then stained for identification of both functional and

non-functional blood vessels using primary rat anti-CD31 and secondary Cy3-

conjugated goat anti-rat IgG antibodies (BD Biosciences, Toronto, Canada). Images

were subsequently captured using a rhodamine (TRITC) filter set. Composite images of

complete tumor sections containing fluorescence signals representing BCMs, blood

vessels, and blood perfusion were overlayed and registered (Image Pro PLUS, v6.0). In

preparation for image analysis, regions of necrosis and artifacts resulting from

sectioning and staining were excluded. Background tissue autofluorescence was

accounted for by subtraction of average intensity values from regions in the tumor

sections with no fluorescence signal. Mean pixel intensities at specific distances from

the nearest CD31-positive pixel (endothelial cell) within whole tumor sections were

determined using a custom MATLAB algorithm as described further below. Results were

Page 98: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

83

reported as background-subtracted mean AF488 fluorescence pixel intensity at given

distances from CD31-positive pixels in a minimum of 4 tumor sections from 3 animals

+/- SD. The AUC was calculated by integration of mean pixel intensity vs. distance

curves using the trapezoid rule. Microvessel density was measured as the total area of

CD31-positive signal divided by the total surface area of the tumor section, n = 8 +/- SD.

Cell packing density was determined by measuring the total area of nuclear staining

divided by the total surface area in the field of view, n = 5 +/- SD.

4.3.9 Computational analysis of BCM distribution in tumor sections

A method was developed to determine the mean AF488 pixel intensity (BCMs) with

respect to distance from the nearest CD31-positive pixel (blood vessel) using a custom

MATLAB algorithm. Registered images of whole tumor sections stained for blood

vessels were thresholded in order to generate binarized images containing only CD31-

positive or CD31-negative pixels (Figure 4.7a). Binarized images were used to generate

distance maps (Figure 4.7b) in which gradients in pixel intensity surround each CD31-

positive pixel. In a distance map, the intensity of each pixel is analogous to its distance

from the nearest blood vessel. Using both a distance map and registered tumor section

image containing AF488-BCMs, the mean BCM pixel intensities with background

subtracted and associated distances from the CD31-positive pixels in bins of 5 µm

increments were calculated.

4.4 Results

4.4.1 Synthesis and characterization of PEG-b-PCL copolymers and BCMs

Synthesis of the diblock copolymer, PEG-b-PCL, was achieved via metal-free cationic

ring opening polymerization, as previously described [242]. Table 4.1 summarizes the

composition of the copolymers and the physicochemical properties of the resulting

BCMs. PEG-b-PCL copolymers comprised of different block lengths were generated by

altering the feed ratio of ε-CL to CH3O-PEG-OH used as starting materials. In the case

of fluorescently labeled copolymers, NH2-PEG-OH was used to initiate the

Page 99: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

84

polymerization resulting in the synthesis of NH2-PEG-b-PCL copolymers. Alexa Fluor

488 carboxylic acid succinimidyl ester was conjugated to the amino group of the

copolymer forming a stable amide bond. Complete separation of unconjugated AF488-

PEG-b-PCL was achieved using a silica gel 100 C8- reversed phase column.

Table 4.1. Composition of block copolymers and properties of block copolymer micelles.

BCMs generated from PEG2000-b-PCL1000 or PEG5000-b-PCL5000 had diameters of 15.8

+/- 2.2 nm (BCM-15) and 53.9 (+/- 5.1) nm (BCM-55), respectively, as measured by

TEM and were primarily spherical in shape with a few rod-like structures observed in the

BCM-15 samples (Figure 2).

Figure 4.2. TEM images of a) PEG5000-b-PCL5000 and b) PEG2000-b-PCL1000 BCMs.

4.4.2 Growth of MCTS

Individual MCTS were grown from HT29 or HeLa cells in non-adherent U-bottom well

plates using a modified liquid overlay technique (Figure 4.3) [270]. Both HeLa and HT29

cells formed spherical MCTS and demonstrated sigmoidal growth profiles (Chapter 3)

[271]. By appropriate selection of the initial number of cells seeded in each well, MCTS

a) b)

100 nm 100 nm

Page 100: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

85

of both cell lines reached a volume of approximately 5x107 µm3 (~ 500 µm in diameter)

following 7 days of culture. No significant difference was found in collagen content

between the HeLa and HT29 spheroids (Appendix 2, Figure A1). However, examination

of spheroid microstructure by light microscopy revealed greater porosity in the HeLa

spheroids in comparison to the more tightly packed tissue architecture within the HT29

spheroids (Figure 4.4).

Figure 4.3. Growth of MCTS using the liquid overlay method. Images of growing

spheroids were captured using a light microscope and 10x objective lens. Scale bars

represent 200 µm.

4.4.3 Penetration of BCMs in MCTS

In this study, the diffusive transport of fluorescently labeled BCMs of different sizes was

examined in MCTS. Representative images of MCTS cross-sections following

incubation with 15 and 55 nm BCMs for 1 h and 24 h are shown in Figure 4.5 and

results of computational analysis are shown in

Figure 4.6. The signal intensity in HeLa MCTS was much greater at all radial positions

in comparison to HT29 MCTS after both 1 h and 24 h incubation periods. After

incubation for 1 h, computational image analysis confirmed that BCM-15 was distributed

homogeneously in the MCTS while BCM-55 demonstrated significantly less penetration

into the core of both HeLa and HT29 MCTS. By 24 h, both BCM-15 and BCM-55

achieved a homogeneous distribution throughout HeLa and HT29 MCTS, although

dramatically lower total fluorescence intensity was observed in HT29 MCTS.

a) b) c) d)

Page 101: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

86

Figure 4.4. H&E staining of a) HT29 and b) HeLa xenografts as well as c) HT29 and d)

HeLa MCTS. Scale bars represent 100 µm in a) and b) and 50 µm in c) and d).

Transmitted light, cross-sectional images of e) HT29 and f) HeLa MCTS at 40x

magnification. Arrows indicate large intercellular channels. Scale bars represent 10 µm

in e) and f).

4.4.4 Penetration of BCMs in tumor xenografts

The penetration of BCMs in xenografts was evaluated by fluorescence imaging of

tumor cross-sections at 1, 6, and 24 h following i.v administration of fluorescently

labelled BCMs. Computational image analysis was conducted using a customized

algorithm to determine the mean fluorescence pixel intensity at a given distance from

the nearest tumor blood vessel (CD31-positive pixel) (Figure 4.7).

a)

e) f)

a) b) c) d)

Page 102: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

87

Figure 4.5. BCM fluorescence in representative cross-sections of HeLa MCTS following

incubation of MCTS for 1 h with a) 15 nm and b) 55 nm BCMs; and 24 h with e) 15 nm

and f) 55 nm BCMs. HT29 MCTS cross-sections following incubation of MCTS for 1 h

with c) 15 nm and d) 55 nm BCMs; and 24 h with g) 15 nm and h) 55 nm BCMs. Scale

bars represent 100 µm.

Representative regions of HeLa and HT29 tumor xenograft cross-sections following

administration of BCM-15 and BCM-55 are shown in Figure 4.8 along with the results of

computational image analysis. The green, red, and blue signals correspond to

fluorescence of AF488-BCMs, blood vessels (anti-CD31), and blood vessel perfusion

(Hoechst), respectively. These results demonstrate that BCM-55 localized more closely

to the tumor vasculature relative to BCM-15 in both xenograft models. The areas under

the mean signal intensity curves (AUC) were lower in HT29 xenografts indicating lower

concentrations of penetrating BCMs relative to HeLa xenografts (Appendix 2, Table T1).

BCM-15

24h

HT29

BCM-55 BCM-55 BCM-15

1h

HeLa

a) b) c) d)

e) f) g) h)

Page 103: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

88

Figure 4.6. Area-normalized distribution of 15 (BCM-15) and 55 (BCM-55) nm BCMs in

HeLa (top row) and HT29 (bottom row) MCTS following 1 or 24 h exposures. “*”

indicates statistically significant difference from unmarked groups (top left). “*” indicates

statistically significant differences between groups with the same mark (bottom left).

Bars indicate differences between groups marked “*” and “**”. In all cases, p < 0.05.

In addition, minimum and maximum mean fluorescent pixel intensities at a depth of 100

µm from the nearest blood vessel occurred at 24 h p.i. for BCM-15 and BCM-55,

respectively, indicating that the smaller micelles had begun to clear from the tumor

tissues when the larger micelles were reaching their maximum depth of penetration

(Figure 4.9). Hoechst staining identified vessel perfusion at the time of animal sacrifice

a) b)

BCM-55 BCM-15

* *

**

* *

HeL

a

HT

29

1 h 24 h

*

Page 104: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

89

but not necessarily vessels that may have been perfused at an earlier time. In this study

Hoechst staining was only used for qualitative identification of perfused vessels. The

microvessel density of HeLa and HT29 tumor xenografts was 6.01+/-1.04 and 4.15+/-

1.17, respectively. The cell packing density of HT29 tumor xenografts was found to be

55.4 +/- 1.2 while HeLa tumor xenografts had a cell packing density of 36.3 +/- 3.1.

Figure 4.7. Computational image-based analysis was employed to determine the mean

fluorescence pixel intensity at a given distance from the nearest tumor blood vessel. a)

Representative binary image showing CD31-positive pixels obtained from a whole HT29

tumor section stained for blood vessels. b) Representative distance map (generated

from the CD-31 binary image) in which the distance of each pixel from the nearest blood

vessel is analogous to its intensity with black pixels being nearest to a blood vessel and

white pixels being farthest away. c) Overlay of CD31-positive and BCM-15 fluorescence

images pseudocoloured in red and green, respectively. Scale bar represents 500 µm.

a) b) c)

Page 105: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

90

Figure 4.8. Representative images and corresponding fluorescence intensity profiles (p.

91) demonstrating the interstitial penetration of BCMs (green) relative to tumor blood

vessels (red) in HeLa and HT29 tumor xenografts. A perfusion marker (blue) was

injected immediately prior to animal sacrifice. Scale bars represent 50 µm.

BC

M-1

5

BC

M-5

5

BC

M-5

5

BC

M-1

5

HeL

a

HT

29

1 h 6 h 24 h

Page 106: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

91

Figure 4.8 (continued from p. 90). Fluorescence intensity profiles.

Figure 4.9. Mean fluorescence pixel intensities in arbitrary units of 15 and 55 nm BCMs

at 5 µm (top left), 25 µm (top right), 50 µm (bottom left) and 100 µm (bottom right) from

the nearest blood vessel in HeLa and HT29 tumor xenografts at 1, 6, and 24 h p.i.

BCM-15 BCM-55

15 nm

HT

29

H

eL

a

HT29 BCM-15 HT29 BCM-55 HeLa BCM-55 HeLa BCM-15

Page 107: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

92

4.5 Discussion

The heterogeneous intratumoral distribution of nano-scale drug delivery systems poses

a significant challenge to their successful application in the treatment of cancer. For

maximum efficacy, anticancer drugs delivered via the systemic circulation must travel

deep into the extravascular space in order to reach cancer cells located in regions of

limited vascular density or function [26]. It has been recognized that conventional

chemotherapeutic agents penetrate poorly within the tumor interstitium resulting in the

exposure of tumor cells to sub-therapeutic concentrations which may promote drug

resistance and tumor repopulation [272]. This problem is exacerbated by the presence

of quiescent cells which often reside within hypoxic regions of the tumor distant from

blood vessels and that are intrinsically less sensitive to chemotherapy [228]. As such,

many anticancer formulations which are shown to be highly cytotoxic in monolayer cell

cultures often elicit an unsatisfactory therapeutic response within the complex

microenvironment of solid tumors.

3-D tissue cultures are useful intermediates for bridging the gap between the

over-simplified structure of monolayer cultures and pre-clinical tumor models [29, 139,

233, 252, 273]. While 3-D cell cultures have been instrumental in exposing limitations in

the interstitial penetration of conventional chemotherapeutic agents, there remains

limited application of these models in the design of nanomedicines [150, 241, 274]. To

date, our understanding of nanoparticle intratumoral transport has been largely based

on assumptions drawn from studies which have evaluated the intratumoral fate of drugs

and other non-colloidal macromolecules. However, distinguishing properties of

nanoparticles, including their size, shape, and surface properties, are likely to limit the

precise application of these findings in the design of nanomedicines.

A recent study of micelle transport by Cabral et al. found that while polymeric

micelles below 50 nm in diameter could penetrate the extravascular space and inhibit

the growth of poorly permeable pancreatic tumor xenografts, larger micelles remained

concentrated in the perivascular space and exerted no therapeutic effect [95]. Similarly,

our group has reported that a reduction in nanoparticle size enhances BCM penetration

in breast tumor xenografts [275]. In the current study, both spatial and temporal

changes in micelle penetration are reported using in vitro and in vivo tumor models. It

should be noted that we have previously demonstrated the high thermodynamic and

Page 108: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

93

kinetic stability of PEG-b-PCL micelles both in vitro and during extended circulation in

the blood [37]. Following a 1 h incubation with both HeLa and HT29 MCTS, the relative

accumulation of 55 nm micelles was greatest at the spheroid periphery while 15 nm

micelles were distributed homogeneously from the periphery to the core. However,

following a 24 h incubation with MCTS, 55 nm micelles achieved a homogeneous

distribution that paralleled that of 15 nm micelles. Similar size-dependent tissue

penetration profiles were observed in tumor xenografts at early time points following i.v

administration of BCMs. However, while the depth of penetration of 55 nm micelles

increased over time, the mean fluorescence intensity of 15 nm micelles appeared to

diminish in both HeLa and HT29 tumors by 24 h p.i presumably due to their limited

vascular supply and rapid tissue clearance (Figure 4.9). Although rod-like nanoparticles

have been shown to penetrate tumor tissues to a greater extent than spherical

nanoparticles, the influence of morphology on tissue transport in the present study is

limited due to the presence of only a small number of rod-like aggregated in the BCM-

15 sample [276]. Our group has previously demonstrated that block copolymer micelles

with diameters in the range of 15-25 nm have short circulation half-lives relative to 60

nm micelles [96], [275]. Therefore, the concentration gradient responsible for diffusion of

15 nm micelles within the tumor interstitium may diminish relatively quickly. By contrast,

larger, longer-circulating micelles with prolonged and elevated plasma concentrations

may undergo slower tumor accumulation and delayed penetration of the tumor

interstitium [100, 277, 278]. Furthermore, larger micelles are likely to be better retained

in the tumor tissue even following a decline in plasma concentration while smaller

micelles are susceptible to more rapid clearance [279].

At 24 h p.i., tumor xenograft sections demonstrated a somewhat heterogeneous,

punctate distribution of BCMs in agreement with previous reports of nanoparticle

intratumoral distribution [80]. This may be due to the internalization of BCMs by tumor

cells or sequestration by phagocytic cells of the immune system. In addition,

heterogeneous BCM extravasation is apparent as some blood vessels appear to not be

surrounded by BCMs while other vessels appear to have significant BCM accumulation

in the surrounding extravascular space. This phenomenon may be attributed to

differences in vascular permeability, particularly in the case of BCM-55, or due to

variations in vascular perfusion. In some cases, there is an absence of BCMs around

Page 109: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

94

vessels which appear to be perfused based on the extravasation of Hoechst while other,

apparently non-perfused vessels, are surrounded by BCMs. This finding is in agreement

with the notion that tumor vascular perfusion is transient and therefore vessels which

are observed to be perfused at the time of animal sacrifice may not have been perfused

previously, and vice versa.

Irrespective of micelle size, higher fluorescence signal intensities were apparent

in MCTS composed of HeLa cells relative to those composed of HT29 cells. This

phenomenon is likely attributed to the presence of large intercellular channels in HeLa

MCTS (Figure 4.4f). Pun’s group has previously demonstrated that the penetration of

nanoparticles in spheroids was greater following treatment with collagenase primarily

due to an increase in spheroid porosity [280]. It is postulated that the lower cell density

and greater extracellular volume of HeLa MCTS relative to HT29 MCTS resulted in

significantly higher amounts of penetrating BCMs. Greater fluorescence signal

intensities were also observed in HeLa xenografts in comparison to the more densely

structured HT29 xenografts. This observation is consistent with several reports of

greater drug penetration in tumors with a higher proportion of interstitial fluid space and

in tumor tissues of low cell density [94, 135, 281, 282]. For example, McGuire et al.

recently demonstrated greater ex vivo penetration of dextrans into tumor tissue samples

with enlarged and interconnected interstitial channels following exposure to chemical

agents that reduce cellular volume or induce cytotoxicity [282].

The similarities in BCM penetration between MCTS and tumor xenograft models

serves to establish the capacity of the MCTS model to predict trends in nanoparticle

interstitial transport in vivo. Specifically, BCM penetration in both models was found to

depend on nanoparticle size, time and tumor or cell type. However, MCTS lack several

important components possessed by tumor xenografts which may limit the predictive

capacity of this in vitro model. Whereas MCTS are, in most cases, composed entirely of

a single epithelial cell line, some tumors contain a large proportion of stromal cells and

fibrous tissues. Although the extent of the influence of tumor stroma on tissue transport

may vary, it is likely that tumors with high stromal content may be better modeled by

MCTS cocultures or other multi-component platforms. In addition, the MCTS model

does not account for the clearance of nanoparticles from tumor tissues in vivo. As such,

the use of MCTS is most suitable for the prediction of trends in nanoparticle tumor

Page 110: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

95

penetration during their accumulation in the tumor rather than at later stages following

significant influence by processes of nanoparticle retention or clearance.

Few methods have been reported for quantifying penetration of small molecules,

macromolecules or nanoparticles in solid tumors [26, 95, 275, 283, 284]. In the present

study, the distribution of fluorescently labeled BCMs relative to tumor blood vessels was

examined using an image-based computational methodology. In order to reduce

potential bias and to achieve a more comprehensive evaluation of tissue distribution, an

efficient procedure was implemented in which entire tumor cross-sections were

analyzed rather than selected sample regions within sections. However, the in vivo

computation of BCM penetration was limited to an analysis of 2-D images

representative of 3-D tumors. Therefore, the presence of out-of-plane vessels that could

potentially contribute to the accumulation of in-plane BCMs may serve to overestimate

the depth of penetration of the micelles. However, this effect, as well as any potential

influence of small differences in MVD between tumor models, was minimized by only

including data in the analysis acquired within a distance of 100 µm from the nearest

CD31-positive pixel.

Figure 4.10. Potential influence of micelles on the intratumoral distribution of the drug.

As illustrated in Figure 4.10, the influence of BCM intratumoral distribution on the

distribution of the drug will depend on the extent to which the drug remains associated

with the carrier. For slow release nanocarriers, the heterogeneous distribution of the

Page 111: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

96

delivery system may negatively influence the distribution of the drug and treatment

efficacy. For drugs that are rapidly released following nanoparticle accumulation, the

distribution of the drug may depend strongly on its ability to penetrate within the tumor

interstitium. It is possible that the distribution of poorly penetrating drugs, due to

significant cellular uptake and binding, may be improved by encapsulation in

nanocarriers which demonstrate limited interaction with cells. Slow releasing

nanocarriers that are well retained in the tumor may also serve as drug depots which

can provide extended drug exposure and ameliorated penetration of the free drug by

establishing localized elevated drug concentrations that serve to drive diffusive transport

in the interstitium.

4.6 Conclusions

The in vitro assessment of BCM tissue penetration using MCTS provided a useful

prediction of relative BCM penetration in HeLa and HT29 tumor xenografts and the

influence of nanoparticle size on interstitial transport. Both in MCTS and tumor

xenografts, the amount of penetrating nanoparticles was limited in tissues with a high

cell packing density while the rate of penetration was promoted by a reduction in

nanoparticle size. Smaller nanoparticles achieved a more rapid penetration into tumor

tissues but began to clear by 24 h p.i while larger nanoparticles achieved slow but

similar depth of penetration and prolonged retention in the tumor. Therefore, the design

of effective delivery systems will require careful adjustment of nanoparticle properties,

such as size, in order to help circumvent the unique transport restrictions imposed by

tumor tissues and to optimize their performance for specific therapeutic applications.

Page 112: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

97

4.7 Acknowledgements

A. Mikhail is the recipient of post-graduate scholarships from NSERC and the

Government of Ontario. S. Eetezadi is funded by the NSERC CREATE Biointerfaces

training program and holds an Ontario Trillium scholarship. The authors thank Dr. I.

Tannock’s Laboratory (Princess Margaret Hospital) and J. Jonkman (Advanced Optical

Microscopy Facility, University Health Network) for helpful discussions on image

analysis as well as P. Zahedi for technical assistance with animal studies.

Page 113: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

98

5 Chapter 5. Summary and Future Directions

5.1 Conclusions and Summary of Findings

This thesis has explored potential determinants of nanocarrier intratumoral penetration

and evaluated the influence of important properties of the 3-D tumor microenvironment

on the therapeutic efficacy of nanomedicines. The initial objective of this research was

to design, synthesize and characterize a model BCM-based drug delivery system for the

chemotherapeutic agent, DTX. Chapter 2 described the incorporation of DTX into PEG-

b-PCL BCMs by means of two methods of drug encapsulation: physical entrapment and

chemical conjugation. Physical entrapment of DTX resulted in > 90% drug release from

the BCMs over 12 h while chemical conjugation resulted in extended drug release over

the course of 7 days. Incorporation of DTX by both chemical and physical entrapment

resulted in significantly greater loading and slower release of the physically entrapped

drug relative to BCMs containing no chemically conjugated drug. This phenomenon was

attributed to improved compatibility between DTX and the core-forming block containing

conjugated DTX (PCL-DTX) relative to unmodified BCMs (PCL alone) and

demonstrated the potential to customize both drug loading and release properties of the

nanocarrier system. In addition, due to a high drug loading capacity, the BCMs were

capable of dramatically enhancing the solubility of DTX. Interestingly, it was also found

that chemical conjugation of DTX to the core of the micelles resulted in profound

changes in micelle morphology. In fact, conversions between spherical and rod-like

morphologies were controlled simply by altering the ratio of unmodified copolymer to

DTX-conjugated copolymer incorporated into the BCMs. This was an important finding

as the shape of nanocarriers has recently been shown to have significant implications in

drug delivery in terms of its influence on drug loading and circulation longevity. The role

of BCM morphology in determining intratumoral distribution and penetration is also of

great interest and warrants future investigation.

In the next phase of research, the therapeutic efficacy of the BCM+DTX

formulation was assessed in MCTS cultures as a means for evaluating the influence of

the 3-D tumor microenvironment on the performance of nanomedicines. Both

BCM+DTX and Taxotere® demonstrated significantly less cytotoxic activity against

Chapter 5. Summary and Future Directions

Page 114: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

99

MCTS cultures relative to monolayers. The degree to which culturing cells as MCTS

increased their resistance to treatment relative to monolayers was different for each cell

line due to cell line-specific differences in MCTS structure and microenvironment. For

instance, culturing cells as MCTS imparted a greater enhancement in therapeutic

resistance to HT29 cells than to HeLa cells. Histological examination revealed that

HT29 MCTS contained fewer proliferating cells and a greater degree of hypoxia and

necrosis relative to HeLa MCTS. It was also found that cells treated immediately

following their recovery from disaggregated spheroids retained residual heightened drug

resistance relative to cells grown and treated as monolayers. These findings

demonstrated that drug resistance in 3-D tumor tissues is due in part to mechanisms of

resistance associated with properties of the tissue microenvironment. Interestingly, the

DTX-loaded micelles demonstrated greater cytotoxicity than Taxotere® both in

monolayer and spheroid cultures. Heightened in vitro cytotoxicity of nanoparticle-

encapsulated chemotherapies has been attributed elsewhere to differences in the rate

of cellular uptake and efflux relative to free drug, among other potential mechanisms

[256, 285–287]. This promising result warrants further study, including examination of

cellular uptake and efflux kinetics as well as comparative investigations of efficacy in

tumor xenograft models.

Having identified microenvironmental parameters which may limit the efficacy of

therapy, the focus of Chapter 4 was to investigate the ability of nanoparticles to

penetrate 3-D tumor tissues. A computational method was implemented for the direct

comparison of the spatial and temporal penetration of BCMs in both a 3-D in vitro tissue

model and human tumor xenografts. Specifically, the in vivo penetration of BCM-55 into

the tumor interstitium was found to be slower than BCM-15 and reached a maximum

depth of penetration at 24 h p.i. BCM-15 exhibited more rapid and greater interstitial

transport with its maximum signal intensity occurring at 6 h p.i. Furthermore, significant

clearance of BCM-15 from the tumors had occurred by 24 h post injection while BCM-55

was well retained. Greater permeability of HeLa MCTS and xenografts relative to HT29

were attributed to differences in cell packing density and the size of intercellular

channels. Overall it was determined that the penetration of nanocarriers in tumor tissues

was nanoparticle-size-, tumor tissue type-, and time-dependent. In the future this

method of analysis can be applied to track the independent fate of the drug and

Page 115: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

100

nanocarrier with respect to cellular markers of toxicity in order to evaluate the influence

of nanoparticle properties on drug distribution and therapeutic efficacy.

This research has demonstrated potential limitations in the therapeutic efficacy of

nanomedicines due to both transport limitations associated with 3-D tumor tissues as

well as resistance associated with properties of the tumor microenvironment.

Importantly, a series of assays was established and validated for the comprehensive

evaluation of nanomedicines using a 3-D in vitro tissue model that approximates

important conditions associated with drug resistance in solid tumors. Computational,

image-based methods were successfully applied for evaluating nanoparticle penetration

in tumor tissues at the sub-micron level using both in vitro and in vivo tumor models.

Results obtained in MCTS cultures reflected trends in the tumor tissue penetration of

nanoparticles in vivo thus establishing the model as a valuable tool for optimizing

transport properties of nanomedicines in vitro. Future work will seek to evaluate the

predictive capacity of MCTS in terms of therapeutic efficacy in tumor xenografts.

5.2 Future Directions

5.2.1 Improving drug retention and tumor-specific drug delivery

The degree to which a nanoparticle can influence the in vivo disposition of a drug

depends upon the extent to which the drug is retained following administration. Drugs

that remain encapsulated within the nanoparticle will adopt the transport,

pharmacokinetics, and clearance properties of the delivery system [33–35, 288]. In

contrast, drugs that are rapidly released from the nanoparticle will acquire a similar in

vivo fate to that of free drug [31, 32, 289]. Ideally, the drug should remain encapsulated

until the nanoparticles have sufficiently accumulated in the tumor. In reality, drug

delivery systems must strike a balance between drug retention and the bioavailability of

the drug at the tumor site. This balance will require careful consideration of the

demands of a specific therapy which may benefit from more rapid or more sustained

release of the therapeutic agent. In some cases achieving significant improvements in

drug solubility by replacing toxic solubilizing excipients with inert synthetic copolymers is

sufficient to reduce the severity of side effects or allow for the implementation of a more

Page 116: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

101

aggressive dosing strategy. However, truly tumor-specific drug delivery systems hold

the greatest promise in terms of achieving significant advancements in treatment

outcomes.

In the case of BCMs, efforts have been made to improve the retention of the drug

in the micelle core in order to increase tumor-specific drug delivery. Several BCM-based

formulations are undergoing clinical trial evaluation based on this research [197]. In this

thesis, chemical conjugation of the drug to the micelle core was demonstrated as one

possible approach for improving the retention of freely encapsulated drug due to an

improvement in core-drug compatibility. The conjugated drug itself was released very

slowly via hydrolysis of the ester linkage between DTX and the hydrophobic block of the

copolymer. In the future, it is worth exploring the development of so called “triggered

release” or “environmentally responsive” systems. In these systems, the drug is

released following exposure to environmental stimuli unique to the physiology of tumors.

Alternatively, exposure to externally applied stimuli, such as heat, ultrasonic waves and

magnetic fields may be applied to the tumor in order to trigger or guide drug release. In

the case of the BCM+DTX formulation developed in this thesis, inclusion of pH sensitive

or enzymatically cleavable chemical linkers may be an important next step for improving

the tumor-specific delivery of DTX.

5.2.2 Therapeutic efficacy and the intratumoral fate of DTX

Although the findings in this thesis provide important insight into the transport of BCMs

at the intratumoral level, the analysis was limited to assessment of delivery vehicle

distribution and did not report the fate of DTX itself. Yet, since we are ultimately

interested in the access of tumor cells to therapeutic concentrations of DTX rather than

to the carrier itself, evaluating the independent fate of DTX and the BCMs is an

important future direction for this research. For example, it is plausible that drug carriers

confined to the perivascular space may act as drug reservoirs for sustained release of

the drug over an extended period of time. In this scenario, the efficacy of treatment may

depend on the rate of release and interstitial penetration of the drug rather than solely

on the distribution of the delivery vehicle itself. The image-based methodology for

evaluating intratumoral penetration described in Chapter 4 can be used to analyze the

relative distribution of any two (or more) fluorescent (or chromogenic, autoradiographic)

Page 117: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

102

signals in the tumor. Therefore, this procedure is expected to serve as the basis for

future investigations into the relationship between the intratumoral distribution of drug

and nanocarrier, the tumor microenvironment and therapeutic efficacy.

Since the response of tumor cells to a drug is highly dependent on conditions

within the local tumor microenvironment, it would be beneficial to evaluate the

intratumoral distribution of downstream markers of cytotoxic effects rather than directly

assessing the distribution of the drug. For example, identification of cleaved caspase-3

or gamma-H2AX, markers of apoptosis and DNA double-strand breaks, respectively,

would provide invaluable insight into therapeutic response within the context of the local

tumor microenvironment [26, 243]. This analysis can be correlated to therapeutic

efficacy observed in traditional growth delay experiments in mice bearing human tumor

xenografts. In fact, tumor sections can be analyzed at various time points throughout

the trial, providing insight into changes in the distribution of therapeutic activity in the

tumor that may occur during the course of treatment. Changes in important properties of

the tumor microenvironment (ex. hypoxia) or proliferative activity of cells can also be

evaluated following treatment with nanomedicines. It is also of particular interest to

apply these analyses following administration of tumor “modulators” [290, 291] in order

to investigate the potential to actively circumvent barriers to nanoparticle penetration

and therapeutic efficacy associated with the 3-D tumor microenvironment, discussed in

greater detail below.

5.2.3 Overcoming transport barriers by modulating the tumor microenvironment

Effective delivery of a chemotherapeutic agent from the site of administration to its

cellular target is a multi-step process that requires accumulation at the tumor, adequate

distribution via the tumor vascular network, extravasation and permeation of the

interstitial space. However, each of these steps is associated with a number of

physiological barriers which limit the transport of drugs at the intratumoral level, as

discussed in Chapters 3 and 4. Moreover, even if a therapeutic agent reaches its target,

the drug’s efficacy may be greatly diminished in regions of hypoxia and/or limited

cellular metabolic and proliferative activity. Therefore, future studies may include

attempts to circumvent these barriers by altering properties of the tumor

Page 118: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

103

microenvironment for the purpose of improving the intratumoral distribution and

penetration of nanomedicines. For example, Kataoka`s group demonstrated an

enhancement in nanoparticle extravasation and penetration following administration of a

TGF-β inhibitor resulting in a reduction in tumor fibrosis and pericyte coverage of the

endothelium in a hypovascular, fibrotic pancreatic tumor model [292, 293]. Similarly,

Jain’s group demonstrated enhanced penetration and efficacy of a nanomedicine

following inhibition of collagen I synthesis by administration of losartan, a clinically

approved compound with recognized antifibrotic activity [294]. In another approach,

conjugation or coating of collagenase to nanoparticles induced ECM-penetrating

properties that enhanced their transport in ECM gels and tumor spheroids [295, 296].

Enhanced penetration in the spheroids was attributed to an increase in the accessible

space (volume fraction) and a decrease in frictional resistance to diffusion [296].

Alterations to the composition of the ECM have also been shown to facilitate

extravasation by altering tumor IFP. Eikenes et al. reported a reduction in tumor IFP

following intratumoral injection of hyaluronidase, an ECM-degrading enzyme, resulting

in an increase in the transcapillary pressure gradient which promoted the tumor

accumulation and penetration of Caelyx™ (liposomal doxorubicin) [297]. Modulators of

tumor vascular permeability and systemic blood pressure have also been investigated

for the purpose of enhancing transvascular transport and the EPR effect [68].

The theory of “vascular normalization”, largely advanced by Jain and coworkers,

describes the administration of anti-angiogenic therapies to return the leaky, tortuous

tumor vasculature to its more natural architecture [298, 299]. Consequently, normal IFP

is restored, enhancing convective flow in the interstitium resulting in improved transport

of therapeutic agents. However, this approach must be applied with care, as anti-

angiogenic agents are also known to reduce the density or permeability of tumor-

associated vessels leading to a potential reduction in the tumor uptake and penetration

of subsequently administered nanomedicines [300]. Recently, promising alternative

approaches for enhancing drug accumulation and penetration have been reported

including use of hyperthermia [301], high-intensity focused ultrasound [302] and

radiation [303]. The co-administration of nanomedicines and external modulators of the

tumor microenvironment remains an exciting potential area of research, worthy of

further investigation.

Page 119: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

104

References

1. Zuylen, L. van; Verweij, J.; Sparreboom, A. Role of formulation vehicles in taxane

pharmacology. Investigational New Drugs. 2001, 19, 125–141.

2. Tije, A. J. Ten; Verweij, J.; Loos, W. J.; Sparreboom, A. Pharmacological effects

of formulation vehicles: implications for cancer chemotherapy. Clinical

Pharmacokinetics. 2003, 42, 665–685.

3. Nishiyama, N.; Kataoka, K. Current state, achievements, and future prospects of

polymeric micelles as nanocarriers for drug and gene delivery. Pharmacology &

Therapeutics. 2006, 112, 630–48.

4. Liu, J.; Lee, H.; Allen, C. Formulation of drugs in block copolymer micelles: drug

loading and release. Current Pharmaceutical Design. 2006, 12, 4685–701.

5. Torchilin, V. P. Targeted polymeric micelles for delivery of poorly soluble drugs.

Cellular and Molecular Life Sciences. 2004, 61, 2549–59.

6. Miyata, K.; Christie, R. J.; Kataoka, K. Polymeric micelles for nano-scale drug

delivery. Reactive and Functional Polymers. 2011, 71, 227–234.

7. Yokoyama, M. Clinical applications of polymeric micelle carrier systems in

chemotherapy and image diagnosis of solid tumors. Journal of Experimental &

Clinical Medicine. 2011, 3, 151–158.

8. Kwon, G. S.; Kataoka, K. Block copolymer micelles as long-circulating drug

vehicles. Advanced Drug Delivery Reviews. 2012, 1–9.

9. Kataoka, K.; Harada, A.; Nagasaki, Y. Block copolymer micelles for drug delivery:

Design, characterization and biological significance. Advanced Drug Delivery

Reviews. 2012, 47.

Page 120: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

105

10. Satchi-Fainaro, R.; Duncan, R.; Barnes, C. M. Polymer therapeutics for cancer :

current status and future challenges. Matrix. 2006, 1–65.

11. Torchilin, V. P. Recent advances with liposomes as pharmaceutical carriers.

Nature Reviews. Drug Discovery. 2005, 4, 145–60.

12. Moghimi, S. M.; Hunter, a C.; Murray, J. C. Long-circulating and target-specific

nanoparticles: theory to practice. Pharmacological Reviews. 2001, 53, 283–318.

13. Moghimi, S. M. Nanoparticles: Structure and Biological

Targeting/Pharmacokinetics. Annual Review of Pharmacology and Toxicology.

2011, 52, 481–503.

14. Aliabadi, H. M.; Brocks, D. R.; Lavasanifar, A.; Shahin., M. Disposition of drugs in

block copolymer micelle delivery systems: from discovery to recovery. Clinical

Pharmacokinetics. 2008, 47, 619–634.

15. Allen, C.; Maysinger, D.; Eisenberg, A. Nano-engineering block copolymer

aggregates for drug delivery. Colloids and Surfaces B: Biointerfaces. 1999, 16, 3–

27.

16. Alexis, F.; Pridgen, E.; Molnar, L. K.; Farokhzad, O. C. Factors affecting the

clearance and biodistribution of polymeric nanoparticles. Molecular

Pharmaceutics. 2008, 5, 505–15.

17. Matsumura, Y.; Hamaguchi, T.; Ura, T.; Muro, K.; Yamada, Y.; Shimada, Y.;

Shirao, K.; Okusaka, T.; Ueno, H.; Ikeda, M. et al. Phase I clinical trial and

pharmacokinetic evaluation of NK911, a micelle-encapsulated doxorubicin. British

Journal of Cancer. 2004, 91, 1775–81.

18. Hamaguchi, T.; Kato, K.; Yasui, H.; Morizane, C.; Ikeda, M.; Ueno, H.; Muro, K.;

Yamada, Y.; Okusaka, T.; Shirao, K. et al. A phase I and pharmacokinetic study

of NK105, a paclitaxel-incorporating micellar nanoparticle formulation. British

Journal of Cancer. 2007, 97, 170–6.

Page 121: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

106

19. Kim, T.-Y.; Kim, D.-W.; Chung, J.-Y.; Shin, S. G.; Kim, S.-C.; Heo, D. S.; Kim, N.

K.; Bang, Y.-J. Phase I and pharmacokinetic study of Genexol-PM, a cremophor-

free, polymeric micelle-formulated paclitaxel, in patients with advanced

malignancies. Clinical Cancer Research. 2004, 10, 3708–16.

20. Danson, S.; Ferry, D.; Alakhov, V.; Margison, J.; Kerr, D.; Jowle, D.; Brampton,

M.; Halbert, G.; Ranson, M. Phase I dose escalation and pharmacokinetic study

of pluronic polymer-bound doxorubicin (SP1049C) in patients with advanced

cancer. British Journal of Cancer. 2004, 90, 2085–91.

21. Saif, M. W.; Podoltsev, N. a; Rubin, M. S.; Figueroa, J. a; Lee, M. Y.; Kwon, J.;

Rowen, E.; Yu, J.; Kerr, R. O. Phase II clinical trial of paclitaxel loaded polymeric

micelle in patients with advanced pancreatic cancer. Cancer Investigation. 2010,

28, 186–94.

22. Matsumura, Y. Polymeric micellar delivery systems in oncology. Japanese

Journal of Clinical Oncology. 2008, 38, 793–802.

23. Yokoyama, M. Polymeric micelles as a new drug carrier system and their required

considerations for clinical trials. Expert opinion on drug delivery. 2010, 7, 145–58.

24. Ehrlich, P. Collected Studies on Immunity; John Wiley and Sons: New York, 1906.

25. Tannock, I. F.; Lee, C. M.; Tunggal, J. K.; Cowan, D. S. M.; Egorin, M. J. Limited

penetration of anticancer drugs through tumor tissue: a potential cause of

resistance of solid tumors to chemotherapy. Clinical Cancer Research. 2002, 8,

878–84.

26. Fung, A.; Jonkman, J.; Tannock, I. F. Quantitative immunohistochemistry for

evaluating the distribution of Ki67 and other biomarkers in tumor sections and use

of the method to study repopulation in xenografts after treatment. Neoplasia.

2012, 14, 324–334.

27. Vaupel, P.; Mayer, A. Hypoxia in cancer: significance and impact on clinical

outcome. Cancer Metastasis Reviews. 2007, 26, 225–39.

Page 122: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

107

28. Vinci, M.; Gowan, S.; Boxall, F.; Patterson, L.; Zimmermann, M.; Court, W.;

Lomas, C.; Mendiola, M.; Hardisson, D.; Eccles, S. a Advances in establishment

and analysis of 3D tumour spheroid-based functional assays for target validation

and drug evaluation. BMC Biology. 2012, 10.

29. Hirschhaeuser, F.; Menne, H.; Dittfeld, C.; West, J.; Mueller-Klieser, W.; Kunz-

Schughart, L. A. Multicellular tumor spheroids: an underestimated tool is catching

up again. Journal of Biotechnology. 2010, 148, 3–15.

30. Achilli, T.-M.; Meyer, J.; Morgan, J. R. Advances in the formation, use and

understanding of multi-cellular spheroids. Expert Opinion on Biological Therapy.

2012, 12, 1347–60.

31. Kim, S. C.; Kim, D. W.; Shim, Y. H.; Bang, J. S.; Oh, H. S.; Wan Kim, S.; Seo, M.

H. In vivo evaluation of polymeric micellar paclitaxel formulation: toxicity and

efficacy. Journal of Controlled Release. 2001, 72, 191–202.

32. Burt, H. M.; Zhang, X.; Toleikis, P.; Embree, L.; Hunter, W. L. Development of

copolymers of poly (D, L-lactide ) and methoxypolyethylene glycol as micellar

carriers of paclitaxel. Colloids and Surfaces. 1999, 16, 161 – 171.

33. Nakanishi, T.; Fukushima, S.; Okamoto, K.; Suzuki, M.; Matsumura, Y.;

Yokoyama, M.; Okano, T.; Sakurai, Y.; Kataoka, K. Development of the polymer

micelle carrier system for doxorubicin. Journal of Controlled Release. 2001, 74,

295–302.

34. Hamaguchi, T.; Matsumura, Y.; Suzuki, M.; Shimizu, K.; Goda, R.; Nakamura, I.;

Nakatomi, I.; Yokoyama, M.; Kataoka, K.; Kakizoe, T. NK105, a paclitaxel-

incorporating micellar nanoparticle formulation, can extend in vivo antitumour

activity and reduce the neurotoxicity of paclitaxel. British Journal of Cancer. 2005,

92, 1240–6.

35. Nishiyama, N.; Okazaki, S.; Cabral, H.; Miyamoto, M.; Kato, Y.; Sugiyama, Y.;

Nishio, K.; Matsumura, Y.; Kataoka, K. Novel cisplatin-incorporated polymeric

Page 123: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

108

micelles can eradicate solid tumors in mice. Cancer Research. 2003, 63, 8977–

83.

36. Kang, N.; Perron, M.; Prud’homme, R. E.; Zhang, Y.; Gaucher, G.; Leroux, J.-C.

Stereocomplex Block Copolymer Micelles: Core−Shell nanostructures with

enhanced stability. Nano Letters. 2005, 5, 315–319.

37. Liu, J.; Zeng, F.; Allen, C. In vivo fate of unimers and micelles of a poly(ethylene

glycol)-block-poly(caprolactone) copolymer in mice following intravenous

administration. European Journal of Pharmaceutics and Biopharmaceutics. 2007,

65, 309–19.

38. Novakova, K.; Laznicek, M.; Rypacek, F.; Machova, L. Pharmacokinetics and

distribution of 125I-PLA-b-PEO block copolymers in rats. Pharmaceutical

Development and Technology. 2003, 8, 153–61.

39. Bae, Y. H.; Yin, H. Stability issues of polymeric micelles. Journal of Controlled

Release. 2008, 131, 2–4.

40. Novakova, K.; Laznicek, M.; Rypacek, F.; Machova, L. 125I-Labeled PLA/PEO

block copolymer: biodistribution studies in rats. Journal of Bioactive and

Compatible Polymers. 2002, 17, 285–296.

41. Batrakova, E. V; Li, S.; Li, Y.; Alakhov, V. Y.; Elmquist, W. F.; Kabanov, A. V

Distribution kinetics of a micelle-forming block copolymer Pluronic P85. Journal of

Controlled Release. 2004, 100, 389–97.

42. Liu, J.; Xiao, Y.; Allen, C. Polymer-drug compatibility: a guide to the development

of delivery systems for the anticancer agent, ellipticine. Journal of Pharmaceutical

Sciences. 2004, 93, 132–43.

43. Adams, M. L.; Lavasanifar, A.; Kwon, G. S. Amphiphilic block copolymers for drug

delivery. Journal of Pharmaceutical Sciences. 2003, 92, 1343–1355.

44. Carstens, M. G.; de Jong, P. H.; van Nostrum, C. F.; Kemmink, J.; Verrijk, R.; de

Leede, L. G.; Crommelin, D. J.; Hennink, W. E. The effect of core composition in

Page 124: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

109

biodegradable oligomeric micelles as taxane formulations. European Journal of

Pharmaceutics and Biopharmaceutics . 2008, 68, 596–606.

45. Kato, K.; Chin, K.; Yoshikawa, T.; Yamaguchi, K.; Tsuji, Y.; Esaki, T.; Sakai, K.;

Kimura, M.; Hamaguchi, T.; Shimada, Y. et al. Phase II study of NK105, a

paclitaxel-incorporating micellar nanoparticle, for previously treated advanced or

recurrent gastric cancer. Investigational New Drugs. 2012, 30, 1621–7.

46. Yokoyama, M.; Fukushima, S.; Uehara, R. Characterization of physical

entrapment and chemical conjugation of adriamycin in polymeric micelles and

their design for in vivo delivery to a solid tumor. Journal of Controlled Release.

1998, 50, 79–92.

47. Dwan’Isa, J. L.; Rouxhet, L.; Pre, V.; Arie, A. Prediction of drug solubility in

amphiphilic di-block copolymer micelles : the role of polymer-drug compatibility.

Pharmazie. 2007, 62, 499–504.

48. Huynh, L.; Grant, J.; Leroux, J.-C.; Delmas, P.; Allen, C. Predicting the solubility

of the anti-cancer agent docetaxel in small molecule excipients using

computational methods. Pharmaceutical Research. 2008, 25, 147–57.

49. Moghimi, S. M.; Hunter, a C.; Andresen, T. L. Factors controlling nanoparticle

pharmacokinetics: an integrated analysis and perspective. Annual Review of

Pharmacology and Toxicology. 2012, 52, 481–503.

50. Hunter, a C.; Moghimi, S. M. Therapeutic synthetic polymers: a game of Russian

roulette? Drug Discovery Today. 2002, 7, 998–1001.

51. Yamamoto, Y.; Nagasaki, Y.; Kato, Y.; Sugiyama, Y.; Kataoka, K. Long-circulating

poly(ethylene glycol)-poly(D,L-lactide) block copolymer micelles with modulated

surface charge. Journal of Controlled Release. 2001, 77, 27–38.

52. Bazile, D.; Prud’homme, C.; Bassoullet, M. T.; Marlard, M.; Spenlehauer, G.;

Veillard, M. Stealth Me.PEG-PLA nanoparticles avoid uptake by the mononuclear

phagocytes system. Journal of Pharmaceutical Sciences. 1995, 84, 493–8.

Page 125: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

110

53. Nakada, Y.; Tudomi, R.; Sakurai, K.; Takahashi, Y. Evaluation of long-circulating

nanoparticles using biodegradable ABA triblock copolymers containing of poly(L -

lactic acid) A-blocks attached to central poly(oxyethylene) B-blocks in vivo.

International Journal of Pharmaceutics. 1998, 175, 109 – 117.

54. Liu, J.; Zeng, F.; Allen, C. Influence of serum protein on polycarbonate-based

copolymer micelles as a delivery system for a hydrophobic anti-cancer agent.

Journal of Controlled Release. 2005, 103, 481–97.

55. Roerdink, F.; Regts, J.; Leeuwen, B. Van; Scherphof, G. Intrahepatic uptake and

processing of intravenously injected small unilamellar phopholipd vesicles in rats.

Biochimica Et Biophysica Acta - Biomembranes. 1984, 770, 195–202.

56. Sherphof, G.; Roerdink, F.; Dijkstra, J. Uptake of liposomes by rat and mouse

hepatocytes and Kupffer cells. Biology of the Cell. 1983, 47, 47.

57. Braet, F.; de Zanger, R.; Baekeland, M.; Crabbé, E.; van der Smissen, P.; Wisse,

E. Structure and dynamics of the fenestrae-associated cytoskeleton of rat liver

sinusoidal endothelial cells. Hepatology. 1995, 21, 180–189.

58. Jeon, S.; Lee, J.; Andrade, J.; Degennes, P. Protein—surface interactions in the

presence of polyethylene oxideI. Simplified theory. Journal of Colloid and

Interface Science. 1991, 142, 149–158.

59. Owens, D. E.; Peppas, N. A. Opsonization, biodistribution, and pharmacokinetics

of polymeric nanoparticles. International Journal of Pharmaceutics. 2006, 307,

93–102.

60. Hagan, S.; Coombes, A.; Garnett, M.; Dunn, S.; Davies, M.; Illum, L.; Davis, S.;

Harding, S.; Purkiss, S.; Gellert, P. Polylactide−poly(ethylene glycol) copolymers

as drug delivery systems. 1. Characterization of Water Dispersible Micelle-

Forming Systems. Langmuir. 1996, 12, 2153–2161.

61. Santos, N. Dos; Allen, C.; Doppen, A.-M.; Anantha, M.; Cox, K. a K.; Gallagher,

R. C.; Karlsson, G.; Edwards, K.; Kenner, G.; Samuels, L. et al. Influence of

Page 126: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

111

poly(ethylene glycol) grafting density and polymer length on liposomes: relating

plasma circulation lifetimes to protein binding. Biochimica Et Biophysica Acta.

2007, 1768, 1367–77.

62. Kwon, G. S.; Yokoyama, M.; Okano, T.; Sakurai, Y.; Kataoka, K. Biodistribution of

micelle-forming polymer-drug conjugates. Pharmaceutical Research. 1993, 10,

970–974.

63. Grindel, J. M.; Jaworski, T.; Piraner, O.; Emanuele, R. M.; Balasubramanian, M.

Distribution, metabolism, and excretion of a novel surface-active agent, purified

poloxamer 188, in rats, dogs, and humans. Journal of Pharmaceutical Sciences.

2002, 91, 1936–47.

64. Seymour, L. W.; Duncan, R.; Strohalm, J.; Kopecek, J. Effect of molecular weight

of N-(2-hydroxypropyl)methacrylamide cpolymers on body distribution and rate of

excretion after subcutaneous, intraperitoneal, and intravenous administration to

rats. Journal of Biomedical Materials Research. 1987, 21, 1341–1358.

65. Takakura, Y.; Hashida, M. Macromolecular carrier systems for targeted drug

delivery: pharmacokinetic considerations on biodistribution. Pharmaceutical

Research. 1996, 13, 820–31.

66. Choi, H. S.; Liu, W.; Misra, P.; Tanaka, E.; Zimmer, J. P.; Itty Ipe, B.; Bawendi, M.

G.; Frangioni, J. V. Renal clearance of quantum dots. Nature Biotechnology.

2007, 25, 1165–70.

67. Matsumura, Y.; Maeda, H. A new concept for macromolecular therapeutics in

cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and

the antitumor agent smancs. Cancer Research. 1986, 46, 6387–92.

68. Maeda, H. Tumor-Selective Delivery of Macromolecular Drugs via the EPR

Effect : Background and Future Prospects. Bioconjugate Chemistry. 2010, 797–

802.

Page 127: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

112

69. Iyer, A. K.; Khaled, G.; Fang, J.; Maeda, H. Exploiting the enhanced permeability

and retention effect for tumor targeting. Drug Discovery Today. 2006, 11, 812–8.

70. Kwon, G. S.; Suwa, S.; Yokoyama, M.; Okano, T.; Sakurai, Y.; Kataoka, K.

Enhanced tumor accumulation and prolonged circulation times of micelle-forming

poly(ethylene oxide-aspartate) block conjugates. Journal of Controlled Release.

1994, 29, 17–23.

71. Rafi, M.; Cabral, H.; Kano, M. R.; Mi, P.; Iwata, C.; Yashiro, M.; Hirakawa, K.;

Miyazono, K.; Nishiyama, N.; Kataoka, K. Polymeric micelles incorporating (1,2-

diaminocyclohexane)platinum (II) suppress the growth of orthotopic scirrhous

gastric tumors and their lymph node metastasis. Journal of Controlled Release.

2012, 159, 189–96.

72. Jain, R. K. Vascular and interstitial barriers to delivery of therapeutic agents in

tumors. Cancer Metastasis Reviews. 1990, 9, 253–66.

73. Chauhan, V. P.; Stylianopoulos, T.; Boucher, Y.; Jain, R. K. Delivery of molecular

and nanoscale medicine to tumors: transport barriers and strategies. Annual

Review of Chemical and Biomolecular Engineering. 2011, 2, 281–298.

74. Lammers, T.; Kiessling, F.; Hennink, W. E.; Storm, G. Drug targeting to tumors:

Principles, pitfalls and (pre-) clinical progress. Journal of Controlled Release.

2012, 161, 175–87.

75. Bae, Y. H. Drug targeting and tumor heterogeneity. Journal of Controlled Release.

2009, 133, 2–3.

76. Bae, Y. H.; Park, K. Targeted drug delivery to tumors: Myths, reality and

possibility. Journal of Controlled Release. 2011, 153, 198–205.

77. Harrington, K. J.; Mohammadtaghi, S.; Uster, P. S.; Glass, D.; Peters, A. M.; Vile,

R. G.; Stewart, J. S. W. Effective targeting of solid tumors in patients with locally

advanced cancers by radiolabeled pegylated liposomes. Clinical Cancer

Research. 2001, 243–254.

Page 128: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

113

78. Karathanasis, E.; Chan, L.; Karumbaiah, L.; McNeeley, K.; D’Orsi, C. J.;

Annapragada, A. V; Sechopoulos, I.; Bellamkonda, R. V. Tumor vascular

permeability to a nanoprobe correlates to tumor-specific expression levels of

angiogenic markers. PloS One. 2009, 4, e5843.

79. Jain, R. K. Determinants of tumor blood flow: a review. Cancer Research. 1988,

48, 2641–58.

80. Yuan, F.; Leunig, M.; Huang, S. K.; Berk, D. A.; Papahadjopoulos, D.; Jain, R. K.

Mirovascular permeability and interstitial penetration of sterically stabilized

(Stealth) Liposomes in a Human Tumor Xenograft. Cancer Research. 1994, 54,

3352–3356.

81. Fukumura, D.; Yuan, F.; Monsky, W. L.; Chen, Y.; Jain, R. K. Effect of host

microenvironment on the microcirculation of human colon adenocarcinoma. The

American Journal of Pathology. 1997, 151, 679–88.

82. Yuan, F.; Dellian, M.; Fukumura, D.; Leunig, M.; Berk, D. a; Torchilin, V. P.; Jain,

R. K. Vascular permeability in a human tumor xenograft: molecular size

dependence and cutoff size. Cancer Research. 1995, 55, 3752–6.

83. Hobbs, S. K.; Monsky, W. L.; Yuan, F.; Roberts, W. G.; Griffith, L.; Torchilin, V. P.;

Jain, R. K. Regulation of transport pathways in tumor vessels: role of tumor type

and microenvironment. Proceedings of the National Academy of Sciences of the

United States of America. 1998, 95, 4607–12.

84. Dreher, M. R.; Liu, W.; Michelich, C. R.; Dewhirst, M. W.; Yuan, F.; Chilkoti, A.

Tumor vascular permeability, accumulation, and penetration of macromolecular

drug carriers. Journal of the National Cancer Institute. 2006, 98, 335–44.

85. Monsky, W. L.; Fukumura, D.; Gohongi, T.; Ancukiewcz, M.; Weich, H. a;

Torchilin, V. P.; Yuan, F.; Jain, R. K. Augmentation of transvascular transport of

macromolecules and nanoparticles in tumors using vascular endothelial growth

factor. Cancer Research. 1999, 59, 4129–35.

Page 129: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

114

86. Ogawara, K.-I.; Un, K.; Minato, K.; Tanaka, K.-I.; Higaki, K.; Kimura, T.

Determinants for in vivo anti-tumor effects of PEG liposomal doxorubicin:

importance of vascular permeability within tumors. International Journal of

Pharmaceutics. 2008, 359, 234–40.

87. Seymour, L. W.; Miyamoto Y; Maeda, H.; M, B.; J, S.; K, U.; Duncan, R. Influence

of molecular weight on passive tumour accumulation. European Journal of

Cancer. 1995, 5, 766–770.

88. Heldin, C.; Rubin, K.; Pietras, K.; Östman, A. High interstitial fluid pressue — an

obstacle in cancer therapy. Nature Reviews Cancer. 2004, 4, 806–813.

89. Holback, H.; Yeo, Y. Intratumoral drug delivery with nanoparticulate carriers.

Pharmaceutical Research. 2011, 28, 1819–1830.

90. Boucher, Y.; Baxter, L. T.; Jain, R. K. Interstitial pressure gradients in tissue-

isolated and subcutaneous tumors: implications for therapy. Cancer Research.

1990, 50, 4478–84.

91. Jain, R. K.; Baxter, L. T. Mechanisms of heterogeneous distribution of monoclonal

antibodies and other macromolecules in tumors: significance of elevated

interstitial pressure. Cancer Research. 1988, 48, 7022–32.

92. Swartz, M. a; Fleury, M. E. Interstitial flow and its effects in soft tissues. Annual

Review of Biomedical Engineering. 2007, 9, 229–56.

93. Netti, P. A.; Berk, D. A.; Swartz, M. A.; Grodzinsky, A. J.; Jain, R. K. Role of

extracellular matrix assembly in interstitial transport in solid tumors. Cancer

Research. 2000, 2497–2503.

94. Krol, A.; Maresca, J.; Dewhirst, M. W.; Yuan, F. Available volume fraction of

macromolecules in the extravascular space of a fibrosarcoma: implications for

drug delivery. Cancer Research. 1999, 59, 4136–4141.

95. Cabral, H.; Matsumoto, Y.; Mizuno, K.; Chen, Q.; Murakami, M.; Kimura, M.;

Terada, Y.; Kano, M. R.; Miyazono, K.; Uesaka, M. et al. Accumulation of sub-100

Page 130: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

115

nm polymeric micelles in poorly permeable tumours depends on size. Nature

Nanotechnology. 2011, 6, 815–823.

96. Fonge, H.; Huang, H.; Scollard, D.; Reilly, R. M.; Allen, C. Influence of formulation

variables on the biodistribution of multifunctional block copolymer micelles.

Journal of Controlled Release. 2012, 157, 366–74.

97. Tabata, Y.; Murakami, Y.; Ikada, Y. Tumor Accumulation of poly(vinyl Alcohol) of

different sizes after intravenous injection. Journal of controlled release. 1998, 50,

123–33.

98. Moghimi, S. M.; Porter, C. J. H.; Muir I.S., I. L.; S.S., D. Non-phagocytic uptake of

intravenously injected microspheres in rat spleen: Influence of particle size and

hydrophilic coating. Biochemical and Biophysical Research Communications.

1991, 177, 861–866.

99. Pirollo, K. F.; Chang, E. H. Does a targeting ligand influence nanoparticle tumor

localization or uptake? Trends in Biotechnology. 2008, 26, 552–8.

100. Charrois, G. J. R.; Allen, T. M. Rate of biodistribution of STEALTH liposomes to

tumor and skin: influence of liposome diameter and implications for toxicity and

therapeutic activity. Biochimica Et Biophysica Acta. 2003, 1609, 102–8.

101. Byrne, J. D.; Betancourt, T.; Brannon-Peppas, L. Active targeting schemes for

nanoparticle systems in cancer therapeutics. Advanced Drug Delivery Reviews.

2008, 60, 1615–26.

102. Jeong, Y.-I.; Seo, S.-J.; Park, I.-K.; Lee, H.-C.; Kang, I.-C.; Akaike, T.; Cho, C.-S.

Cellular recognition of paclitaxel-loaded polymeric nanoparticles composed of

poly(gamma-benzyl L-glutamate) and poly(ethylene glycol) diblock copolymer

endcapped with galactose moiety. International Journal of Pharmaceutics. 2005,

296, 151–61.

Page 131: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

116

103. Lee, H.; Hu, M.; Reilly, R. M.; Allen, C. Apoptotic epidermal growth factor (EGF)-

conjugated block copolymer micelles as a nanotechnology platform for targeted

combination therapy. Molecular Pharmaceutics. 2007, 4, 769–81.

104. You, J.; Li, X.; Cui, F. de; Du, Y.-Z.; Yuan, H.; Hu, F. Q. Folate-conjugated

polymer micelles for active targeting to cancer cells: preparation, in vitro

evaluation of targeting ability and cytotoxicity. Nanotechnology. 2008, 19, art. no.

045102.

105. Noh, T.; Kook, Y. H. E. E.; Park, C.; Youn, H.; Kim, H.; Oh, E. U. N. T. A. X.; Choi,

E. U. N. K.; Park, H. J. O. O.; Kim, C. Block copolymer micelles conjugated with

anti-EGFR antibody for targeted delivery of anticancer drug. Polymer. 2008,

7321–7331.

106. Nasongkla, N.; Shuai, X.; Ai, H.; Weinberg, B. D.; Pink, J.; Boothman, D. a; Gao,

J. cRGD-functionalized polymer micelles for targeted doxorubicin delivery.

Angewandte Chemie. 2004, 43, 6323–7.

107. Xiong, X.-B.; Mahmud, A.; Uludağ, H.; Lavasanifar, A. Multifunctional polymeric

micelles for enhanced intracellular delivery of doxorubicin to metastatic cancer

cells. Pharmaceutical Research. 2008, 25, 2555–66.

108. Zeng, F.; Lee, H.; Allen, C. Epidermal growth factor-conjugated poly(ethylene

glycol)-block- poly(delta-valerolactone) copolymer micelles for targeted delivery of

chemotherapeutics. Bioconjugate Chemistry. 2006, 17, 399–409.

109. Rapoport, N.; Marin, A.; Luo, Y.; Prestwich, G. D.; Muniruzzaman, M. D.

Intracellular uptake and trafficking of Pluronic micelles in drug-sensitive and MDR

cells: effect on the intracellular drug localization. Journal of Pharmaceutical

Sciences. 2002, 91, 157–70.

110. Kim, D.; Lee, E. S.; Oh, K. T.; Gao, Z. G.; Bae, Y. H. Doxorubicin-loaded

polymeric micelle overcomes multidrug resistance of cancer by double-targeting

folate receptor and early endosomal pH. Small. 2008, 4, 2043–50.

Page 132: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

117

111. Yoo, H. S.; Park, T. G. Folate-receptor-targeted delivery of doxorubicin nano-

aggregates stabilized by doxorubicin-PEG-folate conjugate. Journal of Controlled

Release. 2004, 100, 247–56.

112. Bae, Y.; Nishiyama, N.; Kataoka, K. In vivo antitumor activity of the folate-

conjugated pH-sensitive polymeric micelle selectively releasing adriamycin in the

intracellular acidic compartments. Bioconjugate Chemistry. 2007, 18, 1131–9.

113. Mamot, C.; Drummond, D. C.; Noble, C. O.; Kallab, V.; Guo, Z.; Hong, K.;

Kirpotin, D. B.; Park, J. W. Epidermal growth factor receptor-targeted

immunoliposomes significantly enhance the efficacy of multiple anticancer drugs

in vivo. Cancer Research. 2005, 65, 11631–8.

114. Hussain, S.; Plückthun, A.; Allen, T. M.; Zangemeister-Wittke, U. Antitumor

activity of an epithelial cell adhesion molecule targeted nanovesicular drug

delivery system. Molecular Cancer Therapeutics. 2007, 6, 3019–27.

115. Torchilin, P.; Lukyanov, A.; Gao, Z.; Papahadjopoulos-Sternberg, B.

Immunomicelles: Targeted pharmaceutical carriers for poorly soluble drugs.

Proceedings of the National Academy of Sciences of the United States of

America. 2003, 100, 6039–6044.

116. Wu, D.-Q.; Lu, B.; Chang, C.; Chen, C.-S.; Wang, T.; Zhang, Y.-Y.; Cheng, S.-X.;

Jiang, X.-J.; Zhang, X.-Z.; Zhuo, R.-X. Galactosylated fluorescent labeled micelles

as a liver targeting drug carrier. Biomaterials. 2009, 30, 1363–71.

117. Kirpotin, D. B.; Drummond, D. C.; Shao, Y.; Shalaby, M. R.; Hong, K.; Nielsen, U.

B.; Marks, J. D.; Benz, C. C.; Park, J. W. Antibody targeting of long-circulating

lipidic nanoparticles does not increase tumor localization but does increase

internalization in animal models. Cancer Research. 2006, 66, 6732–40.

118. Pun, S. H.; Tack, F.; Bellocq, N. C.; Cheng, J.; Grubbs, B. H.; Jensen, G. S.;

Davis, M. E.; Brewster, M.; Janicot, M.; Janssens, B. et al. Targeted delivery of

RNA-cleaving DNA enzyme (DNAzyme) to tumor tissue by transferrin-modified,

cyclodextrin-based particles. Therapy. 2004, 641–650.

Page 133: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

118

119. Bae, Y.; Nishiyama, N.; Kataoka, K. In vivo antitumor activity of the folate-

conjugated pH-sensitive polymeric micelle selectively releasing adriamycin in the

intracellular acidic compartments. Bioconjugate Chemistry. 2007, 18, 1131–9.

120. Tunggal, J. K.; Cowan, D. S.; Shaikh, H.; Tannock, I. F. Penetration of anticancer

drugs through solid tissue: a factor that limits the effectiveness of chemotherapy

for solid tumors. Clinical Cancer Research. 1999, 5, 1583–6.

121. Primeau, A. J.; Rendon, A.; Hedley, D.; Lilge, L.; Tannock, I. F. The distribution of

the anticancer drug Doxorubicin in relation to blood vessels in solid tumors.

Clinical Cancer Research. 2005, 11, 8782–8.

122. Minchinton, A. I.; Tannock, I. F. Drug penetration in solid tumours. Nature

Reviews. Cancer. 2006, 6, 583–92.

123. Jain, R. K.; Stylianopoulos, T. Delivering nanomedicine to solid tumors. Nature

reviews. Clinical oncology. 2010, 7, 653–64.

124. Jain, R. K. Transport of molecules in the tumor interstitium: a review. Cancer

Research. 1987, 3039–3051.

125. Cowan, D. S.; Tannock, I. F. Factors that influence the penetration of

methotrexate through solid tissue. International Journal of Cancer. 2001, 91, 120–

5.

126. Ackerman, M. E.; Pawlowski, D.; Wittrup, K. D. Effect of antigen turnover rate and

expression level on antibody penetration into tumor spheroids. Molecular Cancer

Therapeutics. 2008, 7, 2233–2240.

127. Pluen, a; Boucher, Y.; Ramanujan, S.; McKee, T. D.; Gohongi, T.; Tomaso, E. di;

Brown, E. B.; Izumi, Y.; Campbell, R. B.; Berk, D. a; et al. Role of tumor-host

interactions in interstitial diffusion of macromolecules: cranial vs. subcutaneous

tumors. Proceedings of the National Academy of Sciences of the United States of

America. 2001, 98, 4628–33.

Page 134: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

119

128. Nomoto, T.; Matsumoto, Y.; Miyata, K.; Oba, M.; Fukushima, S.; Nishiyama, N.;

Yamasoba, T.; Kataoka, K. In situ quantitative monitoring of polyplexes and

polyplex micelles in the blood circulation using intravital real-time confocal laser

scanning microscopy. Journal of Controlled Release. 2011, 151, 104–109.

129. Murakami, M.; Cabral, H.; Matsumoto, Y.; Wu, S.; Kano, M. R.; Yamori, T.;

Nishiyama, N.; Kataoka, K. Improving drug potency and efficacy by nanocarrier-

mediated subcellular targeting. Science Translational Medicine. 2011, 3, 64ra2.

130. Zheng, J.; Liu, J.; Dunne, M.; Jaffray, D. a; Allen, C. In vivo performance of a

liposomal vascular contrast agent for CT and MR-based image guidance

applications. Pharmaceutical Research. 2007, 24, 1193–201.

131. Zheng, J.; Jaffray, D.; Allen, C. Quantitative CT imaging of the spatial and

temporal distribution of liposomes in a rabbit tumor model. Molecular

Pharmaceutics. 2009, 6, 571–580.

132. Hoang, B.; Lee, H.; Reilly, R.; Allen, C. Non-Invasive monitoring of the fate of

111In-labeled block copolymer micelles by high resolution and high sensitivity

microSPECT/CT imaging. Molecular Pharmaceutics. 2009, 6, 581–592.

133. Cowan, D. S. M.; Hicks, K. O.; Wilson, W. Multicellular membranes as an in vitro

model for extravascular diffusion in tumours. British Journal of Cancer. 1996, 74,

28–31.

134. Hicks, K. O.; Ohms, S. J.; Zijl, P. L. Van; Denny, W. A.; Hunter, P. J.; Wilson, W.

R. An experimental and mathematical model for the extravascular transport of a

DNA intercalator in tumours. British Journal of Cancer. 1997, 76, 894–903.

135. Grantab, R.; Sivananthan, S.; Tannock, I. F. The penetration of anticancer drugs

through tumor tissue as a function of cellular adhesion and packing density of

tumor cells. Cancer Research. 2006, 66, 1033–9.

Page 135: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

120

136. Dhiman, H. K.; Ray, A. R.; Panda, A. K. Three-dimensional chitosan scaffold-

based MCF-7 cell culture for the determination of the cytotoxicity of tamoxifen.

Biomaterials. 2005, 26, 979–86.

137. Kievit, F. M.; Florczyk, S. J.; Leung, M. C.; Veiseh, O.; Park, J. O.; Disis, M. L.;

Zhang, M. Chitosan-alginate 3D scaffolds as a mimic of the glioma tumor

microenvironment. Biomaterials. 2010, 31, 5903–10.

138. Chen, L.; Xiao, Z.; Meng, Y.; Zhao, Y.; Han, J.; Su, G.; Chen, B.; Dai, J. The

enhancement of cancer stem cell properties of MCF-7 cells in 3D collagen

scaffolds for modeling of cancer and anti-cancer drugs. Biomaterials. 2012, 33,

1437–44.

139. Nyga, A.; Cheema, U.; Loizidou, M. 3D tumour models: novel in vitro approaches

to cancer studies. Journal of Cell Communication and Signaling. 2011, 5, 239–48.

140. Kurioka, D.; Takagi, A.; Yoneda, M.; Hirokawa, Y.; Shiraishi, T. Multicellular

spheroid culture models: applications in prostate cancer research and

therapeutics. Journal of Cancer Science & Therapy. 2011, 03, 060–065.

141. Durand, R. E.; Sutherland, R. M. Effects of intercellular contact on repair of

radiation damage. Experimental Cell Research. 1972, 71, 75–80.

142. Sutherland, R. M.; Durand, R. E. Radiation response of multicell spheroids--an in

vitro tumour model. Current Topics in Radiation Research Quarterly. 1976, 11,

87–139.

143. Sutherland, R. M.; McCredie, J. a; Inch, W. R. Growth of multicell spheroids in

tissue culture as a model of nodular carcinomas. Journal of the National Cancer

Institute. 1971, 46, 113–20.

144. Sutherland, R. M. Cell and environment interactions in tumor microregions: the

multicell spheroid model. Science. 1988, 240, 177–84.

Page 136: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

121

145. Mueller-klieser, W. Three- dimensional cell cultures: from molecular mechanisms

to clinical applications. The Journal of the Operational Research Society. 1997,

48, 1037.

146. Carlsson, J.; Nederman, T. Tumour spheroid technology in cancer therapy

research. European Journal of Cancer & Clinical Oncology. 1989, 25, 1127–1133.

147. West, G. W.; Weichselbaum, R.; Little, J. B. Limited penetration of methotrexate

into human osteosarcoma spheroids as a proposed model for solid tumor

resistance to adjuvant chemotherapy. Cancer Research. 1980, 40, 3665–8.

148. Mellor, H. R.; Callaghan, R. Accumulation and distribution of doxorubicin in

tumour spheroids: the influence of acidity and expression of P-glycoprotein.

Cancer Chemotherapy and Pharmacology. 2011, 68, 1179–90.

149. Durand, R. E.; Olive, P. L. Resistance of tumor cells to chemo- and radiotherapy

modulated by the three-dimensional architecture of solid tumors and spheroids.

Methods in Cell Biology. 2001, 64, 211–33.

150. Kim, T.-H.; Mount, C. W.; Gombotz, W. R.; Pun, S. H. The delivery of doxorubicin

to 3-D multicellular spheroids and tumors in a murine xenograft model using

tumor-penetrating triblock polymeric micelles. Biomaterials. 2010, 31, 7386–97.

151. Perche, F.; Patel, N. R.; Torchilin, V. P. Accumulation and toxicity of antibody-

targeted doxorubicin-loaded PEG-PE micelles in ovarian cancer cell spheroid

model. Journal of Controlled Release. 2012, 164, 95–102.

152. Tsukioka, Y.; Matsumura, Y.; Hamaguchi, T.; Koike, H.; Moriyasu, F.; Kakizoe, T.

Pharmaceutical and biomedical differences between micellar doxorubicin (NK911)

and liposomal doxorubicin (Doxil). Japanese Journal of Cancer Research. 2002,

93, 1145–53.

153. Santini, M. T.; Rainaldi, G. Three-dimensional spheroid model in tumor biology.

Pathobiology : Journal of Immunopathology, Molecular and Cellular Biology. 2008,

67, 148–57.

Page 137: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

122

154. Goodman, T. T.; Ng, C. P.; Pun, S. H. 3-D tissue culture systems for the

evaluation and optimization of nanoparticle-based drug carriers. Bioconjugate

Chemistry. 2008, 19, 1951–9.

155. Maysinger, D.; Berezovska, O.; Savic, R.; Lim Soo, P.; Eisenberg, A. Block

copolymers modify the internalization of micelle-incorporated probes into neural

cells. Biochimica Et Biophysica Acta. 2001, 1539, 205–217.

156. Allen, C.; Yu, Y.; Eisenberg, A.; Maysinger, D. Cellular internalization of PCL(20)-

b-PEO(44) block copolymer micelles. Biochimica Et Biophysica Acta. 1999, 1421,

32–8.

157. Mahmud, A.; Lavasanifar, A. The effect of block copolymer structure on the

internalization of polymeric micelles by human breast cancer cells. Colloids and

Surfaces. B, Biointerfaces. 2005, 45, 82–9.

158. Luo, L.; Tam, J.; Maysinger, D.; Eisenberg, A. Cellular internalization of

poly(ethylene oxide)-b-poly(ε-caprolactone) diblock copolymer micelles.

Bioconjugate Chemistry. 2002, 13, 1259–1265.

159. Ghosh, N.; Maxfield, R. Endocytosis. Physiological Reviews. 1997, 77, 759–803.

160. Bareford, L. M.; Swaan, P. W. Endocytic mechanisms for targeted drug delivery.

Advanced Drug Delivery Reviews. 2007, 59, 748–58.

161. Prokop, A.; Davidson, J. M. Nanovehicular Intracellular Delivery Systems. Journal

of Pharmaceutical Sciences. 2008, 97, 3518–3590.

162. Conner, S. D.; Schmid, S. L. Regulated portals of entry into the cell. Nature. 2003,

422, 37–44.

163. Liaw, J.; Aoyagi, T.; Kataoka, K.; Sakurai, Y.; Okano, T. Permeation of PEO-

PBLA-FITC Polymeric Micelles in Aortic Endothelial Cells. Pharmaceutical

research 1999, 16, 213–20.

Page 138: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

123

164. Du, W.; Xu, Z.; Nyström, A. M.; Zhang, K.; Leonard, J. R.; Wooley, K. L. 19F- and

fluorescently labeled micelles as nanoscopic assemblies for chemotherapeutic

delivery. Bioconjugate Chemistry. 2008, 19, 2492–8.

165. Yoo, H. S.; Park, T. G. Biodegradable polymeric micelles composed of

doxorubicin conjugated PLGA-PEG block copolymer. Journal of Controlled

Release. 2001, 70, 63–70.

166. Nam, Y.; Kanga, H. S.; Parka, J. Y.; Parkb, T. G.; Hana, S.-H.; Changa, I.-S. New

micelle-like polymer aggregates made from PEI–PLGA diblock copolymers:

micellar characteristics and cellular uptake. Biomaterials. 2003, 24, 2053–2059.

167. Sawant, R. M.; Hurley, J. P.; Salmaso, S.; Kale, a; Tolcheva, E.; Levchenko, T.

S.; Torchilin, V. P. “SMART” drug delivery systems: double-targeted pH-

responsive pharmaceutical nanocarriers. Bioconjugate Chemistry. 2006, 17, 943–

9.

168. Bae, Y.; Nishiyama, N.; Fukushima, S.; Koyama, H.; Yasuhiro, M.; Kataoka, K.

Preparation and biological characterization of polymeric micelle drug carriers with

intracellular pH-triggered drug release property: tumor permeability, controlled

subcellular drug distribution, and enhanced in vivo antitumor efficacy.

Bioconjugate Chemistry. 2005, 16, 122–30.

169. Bae, Y.; Jang, W.-D.; Nishiyama, N.; Fukushima, S.; Kataoka, K. Multifunctional

polymeric micelles with folate-mediated cancer cell targeting and pH-triggered

drug releasing properties for active intracellular drug delivery. Molecular

bioSystems. 2005, 1, 242–50.

170. Lee, Y.; Park, S. Y.; Mok, H.; Park, T. G. Synthesis, characterization, antitumor

activity of pluronic mimicking copolymer micelles conjugated with doxorubicin via

acid-cleavable linkage. Bioconjugate Chemistry. 2008, 19, 525–31.

171. Lee, E. S.; Na, K.; Bae, Y. H. Polymeric micelle for tumor pH and folate-mediated

targeting. Journal of Controlled Release. 2003, 91, 103–13.

Page 139: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

124

172. Ko, J.; Park, K.; Kim, Y.-S.; Kim, M. S.; Han, J. K.; Kim, K.; Park, R.-W.; Kim, I.-S.;

Song, H. K.; Lee, D. S. et al. Tumoral acidic extracellular pH targeting of pH-

responsive MPEG-poly(β-amino ester) block copolymer micelles for cancer

therapy. Journal of Controlled Release. 2007, 123, 109–15.

173. Miyata, K.; Oba, M.; Kano, M. R.; Fukushima, S.; Vachutinsky, Y.; Han, M.;

Koyama, H.; Miyazono, K.; Nishiyama, N.; Kataoka, K. Polyplex micelles from

triblock copolymers composed of tandemly aligned segments with biocompatible,

endosomal escaping, and DNA-condensing functions for systemic gene delivery

to pancreatic tumor tissue. Pharmaceutical Research. 2008, 25, 2924–36.

174. Kang, H. C.; Bae, Y. H. pH-Tunable endosomolytic oligomers for enhanced

nucleic acid delivery. Advanced Functional Materials. 2007, 17, 1263–1272.

175. Lee, E. S.; Gao, Z.; Kim, D.; Park, K.; Kwon, I. C.; Bae, Y. H. Super pH-sensitive

multifunctional polymeric micelle for tumor pHe specific TAT exposure and

multidrug resistance. Journal of Controlled Release. 2008, 129, 228–36.

176. Yessine, M.-A.; Leroux, J.-C. Membrane-destabilizing polyanions: interaction with

lipid bilayers and endosomal escape of biomacromolecules. Advanced Drug

Delivery Reviews. 2004, 56, 999–1021.

177. Yessine, M.-A.; Dufresne, M.-H.; Meier, C.; Petereit, H.-U.; Leroux, J.-C. Proton-

actuated membrane-destabilizing polyion complex micelles. Bioconjugate

Chemistry. 2007, 18, 1010–4.

178. Boussif, O.; Lezoualc’h, F.; Zanta, M. a; Mergny, M. D.; Scherman, D.; Demeneix,

B.; Behr, J. P. A versatile vector for gene and oligonucleotide transfer into cells in

culture and in vivo: polyethylenimine. Proceedings of the National Academy of

Sciences of the United States of America. 1995, 92, 7297–301.

179. Kim, D.; Lee, E. S.; Park, K.; Kwon, I. C.; Bae, Y. H. Doxorubicin loaded pH-

sensitive micelle: antitumoral efficacy against ovarian A2780/DOXR tumor.

Pharmaceutical Research. 2008, 25, 2074–82.

Page 140: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

125

180. Batrakova, E. V; Kabanov, A. V Pluronic block copolymers: evolution of drug

delivery concept from inert nanocarriers to biological response modifiers. Journal

of Controlled Release. 2008, 130, 98–106.

181. Zastre, J. a; Jackson, J. K.; Wong, W.; Burt, H. M. P-glycoprotein efflux inhibition

by amphiphilic diblock copolymers: relationship between copolymer concentration

and substrate hydrophobicity. Molecular Pharmaceutics. 2008, 5, 643–53.

182. Prescribing Information, Taxotere http://products.sanofi.us/Taxotere/taxotere.html.

183. Herbst, R.; Khuri, F. Mode of action of docetaxel – a basis for combination with

novel anticancer agents. Cancer Treatment Reviews. 2003, 29, 407–415.

184. Montero, A.; Fossella, F.; Hortobagyi, G.; Valero, V. Docetaxel for treatment of

solid tumours: a systematic review of clinical data. The Lancet Oncology. 2005, 6,

229–39.

185. Clarke, S.; Rivory, L. Clinical pharmacokinetics of docetaxel. Clinical

Pharmacokinetics. 2006, 45, 235–52.

186. Jones, S. E.; Erban, J.; Overmoyer, B.; Budd, G. T.; Hutchins, L.; Lower, E.;

Laufman, L.; Sundaram, S.; Urba, W. J.; Pritchard, K. I. et al. Randomized phase

III study of docetaxel compared with paclitaxel in metastatic breast cancer.

Journal of Clinical Oncology. 2005, 23, 5542–51.

187. Chan, S.; Friedrichs, K.; Noel, D.; Pintér, T.; Belle, S. Van; Vorobiof, D.; Duarte,

R.; Gil Gil, M.; Bodrogi, I.; Murray, E. et al. Prospective randomized trial of

docetaxel versus doxorubicin in patients with metastatic breast cancer. Journal of

Clinical Oncology. 1999, 17, 2341–54.

188. Trudeau, M. E.; Eisenhauer, E.; Higgins, B.; Letendre, F.; Lofters, W.; Norris, B.;

Vandenberg, T.; Delorme, F.; Muldal, A. Docetaxel in patients with metastatic

breast cancer: a phase II study of the National Cancer Institute of Canada-Clinical

Trials Group. Journal of Clinical Oncology. 1996, 14, 422–428.

Page 141: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

126

189. Sjo, J.; Blomqvist, C.; Mouridsen, H.; Pluzanska, A.; Bengtsson, N.; Mjaaland, I.;

Wist, E.; Valvere, V.; Anderson, H.; Bergh, J. Docetaxel compared with sequential

methotrexate and 5-fluorouracil in patients with advanced breast cancer after

anthracycline failure : a randomised Phase III study with crossover on progression

by the Scandinavian Breast Group. European Journal of Cancer. 1999, 35, 1194–

1201.

190. Urien, S.; Barré, J.; Morin, C.; Paccaly, a; Montay, G.; Tillement, J. P. Docetaxel

serum protein binding with high affinity to alpha 1-acid glycoprotein.

Investigational New Drugs. 1996, 14, 147–51.

191. Extra, J.; Rousseau, F.; Bruno, R.; Clavel, M.; Bail, N. Le; Marty, M. Phase I and

pharmacokinetic study of Taxotere (RP 56976; NSC 628503 ) given as a short

intravenous infusion. 1993, 53, 1037–1042.

192. Baur, M.; Oosterom, A. T. van; Diéras, V.; Tubiana-Hulin, M.; Coombes, R. C.;

Hatschek, T.; Murawsky, M.; Klink-Alakl, M.; Hudec, M.; Dittrich, C. A phase II trial

of docetaxel (Taxotere) as second-line chemotherapy in patients with metastatic

breast cancer. Journal of Cancer Research and Clinical Oncology. 2008, 134,

125–35.

193. World Health Organization http://www.who.int/cancer/en/.

194. Canadian Cancer Statistics 2008 www.cancer.ca.

195. Lyman, G. H. Impact of chemotherapy dose intensity on cancer patient outcomes.

The Journal of the National Comprehensive Cancer Network. 2009, 7, 99–108.

196. Dodwell, D. J.; Gurney, H.; Thatcher, N. Dose intensity in cancer chemotherapy.

British Journal of Cancer. 1990, 61, 789–94.

197. Matsumura, Y.; Kataoka, K. Preclinical and clinical studies of anticancer agent-

incorporating polymer micelles. Cancer Science. 2009, 100, 572–579.

Page 142: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

127

198. Mikhail, A.; Allen, C. Block copolymer micelles for delivery of cancer therapy:

transport at the whole body, tissue and cellular levels. Journal of Controlled

Release. 2009, 138, 214–23.

199. Letchford, K.; Liggins, R.; Burt, H. Solubilization of hydrophobic drugs by methoxy

poly(ethylene glycol)-b-polycaprolactone diblock copolymer micelles : theoretical

and experimental data and correlations. Journal of Pharmaceutical Sciences.

2008, 97, 1179–1190.

200. Soo, P. L.; Luo, L.; Maysinger, D.; Eisenberg, A. Micelles : implications for drug

delivery. Drug Delivery. 2002, 9996–10004.

201. Adams, M.; Kwon, G. S. Relative aggregation state and hemolytic activity of

amphotericin B encapsulated by poly(ethylene oxide)-b–poly(N-hexyl-L-

aspartamide)-acyl conjugate micelles: effects of acyl chain length. Journal of

Controlled Release. 2003, 87, 23–32.

202. Yokoyama, M.; Opanasopit, P.; Okano, T.; Kawano, K.; Maitani, Y. Polymer

design and incorporation methods for polymeric micelle carrier system containing

water-insoluble anti-cancer agent camptothecin. Journal of Drug Targeting. 2004,

12, 373–84.

203. Yokoyama, M.; Satoh, a; Sakurai, Y.; Okano, T.; Matsumura, Y.; Kakizoe, T.;

Kataoka, K. Incorporation of water-insoluble anticancer drug into polymeric

micelles and control of their particle size. Journal of Controlled Release. 1998, 55,

219–29.

204. Nishiyama, N. Nanocarriers shape up for long life. Nature Nanotechnology. 2007,

2, 203–204.

205. Champion, J. a; Katare, Y. K.; Mitragotri, S. Particle shape: a new design

parameter for micro- and nanoscale drug delivery carriers. Journal of Controlled

Release. 2007, 121, 3–9.

Page 143: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

128

206. Geng, Y.; Discher, D. E. Visualization of degradable worm micelle breakdown in

relation to drug release. Polymer. 2006, 47, 2519–2525.

207. Christian, D. a; Cai, S.; Garbuzenko, O. B.; Harada, T.; Zajac, A. L.; Minko, T.;

Discher, D. E. Flexible filaments for in vivo imaging and delivery: persistent

circulation of filomicelles opens the dosage window for sustained tumor

shrinkage. Molecular Pharmaceutics. 2009, 6, 1343–52.

208. Geng, Y.; Dalhaimer, P.; Cai, S.; Tsai, R.; Tewari, M.; Minko, T.; Discher, D. E.

Shape effects of filaments versus spherical particles in flow and drug delivery.

Nature Nanotechnology. 2007, 2, 249–55.

209. Fairley, N.; Hoang, B.; Allen, C. Morphological control of poly(ethylene glycol)-

block-poly(ε-caprolactone) copolymer aggregates in aqueous solution.

Biomacromolecules. 2008, 9, 2283–2291.

210. Adams, D. J.; Kitchen, C.; Adams, S.; Furzeland, S.; Atkins, D.; Schuetz, P.;

Fernyhough, C. M.; Tzokova, N.; Ryan, A. J.; Butler, M. F. On the mechanism of

formation of vesicles from poly (ethylene oxide)-block-poly(caprolactone)

copolymers. Soft Matter. 2009, 5, 3086–3096.

211. Lee, S.; Kang, S.; Kim, C.; Kwon, I.; Jeong, S. Synthesis and characterization of

amphiphilic poly(2-ethyl-2-oxazoline)/poly(ε-caprolactone) alternating multiblock

copolymers. Polymer. 2000, 41, 7091–7097.

212. Zhang, X.; Jackson, J. K.; Burt, H. M. Determination of surfactant critical micelle

concentration by a novel fluorescence depolarization technique. Journal of

Biochemical and Biophysical Methods. 1996, 31, 145–150.

213. Liu, J.; Zahedi, P.; Zeng, F.; Allen, C. Nano-Sized assemblies of a PEG-docetaxel

conjugate as a formulation strategy for docetaxel. Journal of Pharmaceutical

Sciences. 2008, 97, 3274–3290.

214. Deutsch, H. M.; Glinski, J. a; Hernandez, M.; Haugwitz, R. D.; Narayanan, V. L.;

Suffness, M.; Zalkow, L. H. Synthesis of congeners and prodrugs. 3. Water-

Page 144: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

129

soluble prodrugs of taxol with potent antitumor activity. Journal of Medicinal

Chemistry. 1989, 32, 788–92.

215. Kingston, D. G. I.; Chordia, M. D.; Jagtap, P. G.; Liang, J.; Shen, Y.-C.; Long, B.

H.; Fairchild, C. R.; Johnston, K. A. Synthesis and biological evaluation of 1-

deoxypaclitaxel analogues. The Journal of Organic Chemistry. 1999, 64, 1814–

1822.

216. Greenwald, R. B.; Gilbert, C. W.; Pendri, a; Conover, C. D.; Xia, J.; Martinez, a

Drug delivery systems: water soluble taxol 2’-poly(ethylene glycol) ester prodrugs-

design and in vivo effectiveness. Journal of Medicinal Chemistry. 1996, 39, 424–

31.

217. Greenwald, R. B.; Pendri, A.; Bolikal, D.; Gilbert, C. W. Highly water soluble Taxol

derivatives: 2’-polyethyleneglycol esters as potential prodrugs. Bioorganic &

Medicinal Chemistry Letters. 1994, 4, 2465–2470.

218. Ali, S. M.; Hoemann, M. Z.; Aubé, J.; Georg, G. I.; Mitscher, L. a; Jayasinghe, L.

R. Butitaxel analogues: synthesis and structure-activity relationships. Journal of

Medicinal Chemistry. 1997, 40, 236–41.

219. Zhang, L.; Eisenberg, A. Formation of crew-cut aggregates of various

morphologies from amphiphilic block copolymers in solution. Polymers for

Advanced Technologies. 1998, 9, 677–699.

220. Halperin, A.; Alexander, S. Polymeric micelles: their relaxation kinetics.

Macromolecules. 1989, 22, 2403–2412.

221. Cameron, N. S.; Corbierre, M. K.; Eisenberg, A. 1998 E.W.R. Steacie Award

Lecture. Asymmetric amphiphilic block copolymers in solution: a morphological

wonderland. Canadian Journal of Chemistry. 1999, 77, 1311–1326.

222. Zhulina, E. B.; Adam, M.; LaRue, I.; Sheiko, S. S.; Rubinstein, M. Diblock

copolymer micelles in a dilute solution. Macromolecules. 2005, 38, 5330–5351.

Page 145: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

130

223. Richter, D.; Schneiders, D.; Monkenbusch, M.; Willner, L. Polymer aggregates

with crystalline cores : the system polyethylene-poly(ethylenepropylene).

Macromolecules. 1997, 9297, 1053–1068.

224. Lin, E. K.; Gast, A. P. Semicrystalline diblock copolymer platelets in dilute

solution. Macromolecules. 1996, 29, 4432–4441.

225. Du, Z.; Xu, J.; Fan, Z. Micellar morphologies of poly(e-caprolactone)-b-

poly(ethylene oxide) block copolymers in water with a crystalline core.

Macromolecules. 2007, 40, 7633–7637.

226. Zhang, J.; Wang, L.-Q.; Wang, H.; Tu, K. Micellization phenomena of amphiphilic

block copolymers based on methoxy poly(ethylene glycol) and either crystalline or

amorphous poly(caprolactone-b-lactide). Biomacromolecules. 2006, 7, 2492–500.

227. Elsabahy, M.; Perron, M.-E.; Bertrand, N.; Yu, G.-E.; Leroux, J.-C. Solubilization

of docetaxel in poly(ethylene oxide)-block-poly(butylene/styrene oxide) micelles.

Biomacromolecules. 2007, 8, 2250–7.

228. Trédan, O.; Galmarini, C. M.; Patel, K.; Tannock, I. F. Drug resistance and the

solid tumor microenvironment. Journal of the National Cancer Institute. 2007, 99,

1441–54.

229. Swartz, M. A.; Iida, N.; Roberts, E. W. Tumor microenvironment complexity :

emerging roles in cancer therapy. Cancer Research. 2012, 72, 2473–2480.

230. Kim, J. W.; Ho, W. J.; Wu, B. M. The role of the 3D environment in hypoxia-

induced drug and apoptosis resistance. Anticancer Research. 2011, 31, 3237–45.

231. Johnson, J. I.; Decker, S.; Zaharevitz, D.; Rubinstein, L. V; Venditti, J. M.;

Schepartz, S.; Kalyandrug, S.; Christian, M. Relationships between drug activity in

NCI preclinical in vitro and in vivo models and early clinical trials. British Journal of

Cancer. 2001, 84, 1424–1431.

Page 146: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

131

232. Phillips, R.; Bibby, M.; Double, J. A critical appraisal of the predictive value of in

vitro chemosensitivity assays. Journal of the National Cancer Institute. 1990, 82,

1457–1463.

233. Elliott, N. A review of three-dimensional in vitro tissue models for drug discovery

and transport studies. Journal of Pharmaceutical Sciences. 2010, 100, 2–8.

234. Rotin, D.; Robinson, B.; Tannock, I. F. Influence of hypoxia and an acidic

environment on the metabolism and viability of cultured cells: potential

implications for cell death in tumors. Cancer Research. 1986, 46, 2821–2826.

235. Nederman, T.; Norling, B.; Glimelius, B.; Carlsson, J.; Brunk, U. Demonstration of

an extracellular matrix in multicellular tumor spheroids. Cancer Research. 1984,

44, 3090–3097.

236. Acker, H.; Carlsson, J.; Mueller-Klieser, W.; Sutherland, R. M. Comparative pO2

measurements in cell spheroids cultured with different techniques. British Journal

of Cancer. 1987, 56, 325–7.

237. Fischbach, C.; Chen, R.; Matsumoto, T.; Schmelzle, T.; Brugge, J. S.; Polverini,

P. J.; Mooney, D. J. Engineering tumors with 3D scaffolds. Nature Methods. 2007,

4, 6–11.

238. Horning, J. L.; Sahoo, S. K.; Vijayaraghavalu, S.; Dimitrijevic, S.; Vasir, J. K.; Jain,

T. K.; Panda, A. K.; Labhasetwar, V. 3-D tumor model for in vitro evaluation of

anticancer drugs. Molecular Pharmaceutics. 2008, 5, 849–62.

239. Chetprayoon, P.; Kadowaki, K.; Matsusaki, M.; Akashi, M. Survival and structural

evaluations of three-dimensional tissues fabricated by the hierarchical cell

manipulation technique. Acta Biomaterialia. 2012.

240. Hosoya, H.; Kadowaki, K.; Matsusaki, M.; Cabral, H.; Nishihara, H.; Ijichi, H.;

Koike, K.; Kataoka, K.; Miyazono, K.; Akashi, M. et al. Engineering fibrotic tissue

in pancreatic cancer: A novel three-dimensional model to investigate nanoparticle

Page 147: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

132

delivery. Biochemical and Biophysical Research Communications. 2012, 419, 32–

7.

241. Oishi, M.; Nagasaki, Y.; Nishiyama, N.; Itaka, K.; Takagi, M.; Shimamoto, A.;

Furuichi, Y.; Kataoka, K. Enhanced growth inhibition of hepatic multicellular tumor

spheroids by lactosylated poly(ethylene glycol)-siRNA conjugate formulated in

PEGylated polyplexes. ChemMedChem. 2007, 2, 1290–7.

242. Mikhail, A.; Allen, C. Poly(ethylene glycol)-b-poly(epsilon-caprolactone) micelles

containing chemically conjugated and physically entrapped docetaxel: synthesis,

characterization, and the influence of the drug on micelle morphology.

Biomacromolecules. 2010, 11, 1273–80.

243. Zahedi, P.; Stewart, J.; Souza, R. De; Piquette-Miller, M.; Allen, C. An injectable

depot system for sustained intraperitoneal chemotherapy of ovarian cancer

results in favorable drug distribution at the whole body, peritoneal and

intratumoral levels. Journal of Controlled Release. 2012, 158, 379–85.

244. Marusić, M.; Bajzer, Z.; Freyer, J. P.; Vuk-Pavlović, S. Analysis of growth of

multicellular tumour spheroids by mathematical models. Cell Proliferation. 1994,

27, 73–94.

245. Friedrich, J.; Eder, W.; Castaneda, J.; Doss, M.; Huber, E.; Ebner, R.; Kunz-

Schughart, L. A. A reliable tool to determine cell viability in complex 3-D culture:

the acid phosphatase assay. Journal of Biomolecular Screening. 2007, 12, 925–

37.

246. Joyce, J. a; Pollard, J. W. Microenvironmental regulation of metastasis. Nature

Reviews. Cancer. 2009, 9, 239–52.

247. Hanahan, D.; Weinberg, R. a Hallmarks of cancer: the next generation. Cell.

2011, 144, 646–74.

Page 148: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

133

248. Okubo, H.; Matsushita, M.; Kamachi, H.; Kawai, T.; Takahashi, M.; Fujimoto, T.;

Nishikawa, K.; Todo, S. A novel method for faster formation of rat liver cell

spheroids. Artificial Organs. 2002, 26, 497–505.

249. Duca, D. Del; Werbowetski, T.; Maestro, R. F. Del Spheroid preparation from

hanging drops: characterization of a model of brain tumor invasion. Journal of

Neuro-oncology. 2004, 67, 295–303.

250. Friedrich, J.; Seidel, C.; Ebner, R.; Kunz-Schughart, L. A. Spheroid-based drug

screen: considerations and practical approach. Nature Protocols. 2009, 4, 309–

24.

251. Hardelauf, H.; Frimat, J.-P.; Stewart, J. D.; Schormann, W.; Chiang, Y.-Y.;

Lampen, P.; Franzke, J.; Hengstler, J. G.; Cadenas, C.; Kunz-Schughart, L. A.; et

al. Microarrays for the scalable production of metabolically relevant tumour

spheroids: a tool for modulating chemosensitivity traits. Lab on a Chip. 2011, 11,

419–28.

252. Ong, S.-M.; Zhao, Z.; Arooz, T.; Zhao, D.; Zhang, S.; Du, T.; Wasser, M.; Noort,

D. van; Yu, H. Engineering a scaffold-free 3D tumor model for in vitro drug

penetration studies. Biomaterials. 2010, 31, 1180–90.

253. Kyle, A. H.; Huxham, L. a; Yeoman, D. M.; Minchinton, A. I. Limited tissue

penetration of taxanes: a mechanism for resistance in solid tumors. Clinical

Cancer Research. 2007, 13, 2804–10.

254. Han, M.; Oba, M.; Nishiyama, N.; Kano, M. R.; Kizaka-Kondoh, S.; Kataoka, K.

Enhanced percolation and gene expression in tumor hypoxia by PEGylated

polyplex micelles. Molecular Therapy. 2009, 17, 1404–10.

255. Mikhail, A.; Eetezadi, S.; Stewart, J.; Allen, C. Image-based analysis of the time-

dependent penetration of polymeric micelles in multicellular tumor spheroids and

tumor xenografts. 2012, submitted.

Page 149: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

134

256. Fonseca, C.; Simões, S.; Gaspar, R. Paclitaxel-loaded PLGA nanoparticles:

preparation, physicochemical characterization and in vitro anti-tumoral activity.

Journal of Controlled Release. 2002, 83, 273–286.

257. Nemati, F.; Dubernet, C.; Poupon, M.; Couvreur, P. Reversion of multidrug

resistance using nanoparticles in vitro: influence of the nature of the polymer.

International Journal of Pharmaceutics. 1996, 237–246.

258. Cavallaro, G.; Fresta, M.; Giammona, G.; Puglisi, G.; Villari, A. Entrapment of β-

lactams antibiotics in polyethylcyanoacrylate nanoparticles: studies on the

possible in vivo application of this colloidal delivery system. International Journal

of Pharmaceutics. 1994, 111, 31–41.

259. Panyam, J.; Labhasetwar, V. Dynamics of endocytosis and exocytosis of

poly(D,L-lactide-co-glycolide) nanoparticles in vascular smooth muscle cells.

Pharmaceutical Research. 2003, 20, 212–20.

260. Yoo, H. S.; Park, T. G. Folate receptor targeted biodegradable polymeric

doxorubicin micelles. Journal of Controlled Release. 2004, 96, 273–83.

261. Atalay, C. Multi-drug resistance and cancer. Expert Opinion on Therapeutic

Patents. 2007, 17, 511–520.

262. Klein, F.; Wartenberg, M.; Ling, F. C.; Mu, M. Regulation of the multidrug

resistance transporter P-glycoprotein in multicellular tumor spheroids by hypoxia-

inducible factor (HIF-1) and reactive oxygen species 1. The FASEB Journal.

2003, 503–505.

263. Jang, S. H.; Wientjes, M. G.; Lu, D.; Au, J. L.-S. Drug delivery and transport to

solid tumors. Pharmaceutical Research. 2003, 20, 1337–50.

264. Baxter, L. T.; Jain, R. K. Transport of fluid and macromolecules in tumors. I. Role

of interstitial pressure and convection. Microvascular Research. 1989, 37, 77–

104.

Page 150: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

135

265. Blanco, E.; Hsiao, A.; Mann, A. P.; Landry, M. G.; Meric-Bernstam, F.; Ferrari, M.

Nanomedicine in cancer therapy: innovative trends and prospects. Cancer

Science. 2011, 102, 1247–52.

266. Vicent, M. J.; Ringsdorf, H.; Duncan, R. Polymer therapeutics: clinical applications

and challenges for development. Advanced Drug Delivery Reviews. 2009, 61,

1117–20.

267. Maeda, H.; Bharate, G. Y.; Daruwalla, J. Polymeric drugs for efficient tumor-

targeted drug delivery based on EPR-effect. European Journal of Pharmaceutics

and Biopharmaceutics. 2009, 71, 409–19.

268. Torchilin, V. P. Tumor delivery of macromolecular drugs based on the EPR effect.

Advanced Drug Delivery Reviews. 2010, 63, 131–135.

269. Alexis, F.; Pridgen, E. M.; Langer, R.; Farokhzad, O. C. Nanoparticle technologies

for cancer therapy. Handbook of Experimental Pharmacology. 2010, 197, 55–86.

270. Yuhas, J. M.; Li, A. P.; Martinez, A. O.; Martinez, A.; Ladman, A. J. A simplified

method for production and growth of multicellular tumor spheroids. Cancer

Research. 1977, 37, 3639–3643.

271. Chignola, R.; Schenetti, a; Andrighetto, G.; Chiesa, E.; Foroni, R.; Sartoris, S.;

Tridente, G.; Liberati, D. Forecasting the growth of multicell tumour spheroids:

implications for the dynamic growth of solid tumours. Cell Proliferation. 2000, 33,

219–29.

272. Kim, J. J.; Tannock, I. F. Repopulation of cancer cells during therapy: an

important cause of treatment failure. Nature Reviews. Cancer. 2005, 5, 516–25.

273. Wong, H. L.; Shen, Z.; Lu, Z.; Wientjes, M. G.; Au, J. L.-S. Paclitaxel tumor-

priming enhances siRNA delivery and transfection in 3-dimensional tumor

cultures. Molecular Pharmaceutics. 2011, 8, 833–40.

Page 151: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

136

274. Bandekar, A.; Karve, S.; Chang, M.-Y.; Mu, Q.; Rotolo, J.; Sofou, S. Antitumor

efficacy following the intracellular and interstitial release of liposomal doxorubicin.

Biomaterials. 2012, 2–9.

275. Lee, H.; Fonge, H.; Hoang, B.; Reilly, R. M.; Allen, C. The effects of particle size

and molecular targeting on the intratumoral and subcellular distribution of

polymeric nanoparticles. Molecular Pharmaceutics. 2010, 7, 1195–208.

276. Chauhan, V. P.; Popović, Z.; Chen, O.; Cui, J.; Fukumura, D.; Bawendi, M. G.;

Jain, R. K. Fluorescent nanorods and nanospheres for real-time in vivo probing of

nanoparticle shape-dependent tumor penetration. Angewandte Chemie. 2011, 50,

11417–20.

277. Dreher, M. R.; Liu, W.; Michelich, C. R.; Dewhirst, M. W.; Yuan, F.; Chilkoti, A.

Tumor vascular permeability, accumulation, and penetration of macromolecular

drug carriers. Journal of the National Cancer Institute. 2006, 98, 335–44.

278. Lee, C. M.; Tannock, I. F. The distribution of the therapeutic monoclonal

antibodies cetuximab and trastuzumab within solid tumors. BMC Cancer. 2010,

10, 1–11.

279. Maeda, H.; Wu, J.; Sawa, T.; Matsumura, Y.; Hori, K. Tumor vascular permeability

and the EPR effect in macromolecular therapeutics: a review. Journal of

Controlled Release. 2000, 65, 271–84.

280. Goodman, T. T.; Chen, J.; Matveev, K. Spatio-temporal modeling of nanoparticle

delivery to multicellular tumor spheroids. Biotechnology and Bioengineering.

2008, 101, 388–399.

281. Zheng, J. H.; Chen, C. T.; Au, J. L.-S.; Wientjes, M. G. Time and concentration

dependent penetration of doxorubicin in prostate tumors. AAPS pharmSci. 2001,

3, E15.

Page 152: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

137

282. McGuire, S.; Yuan, F. Improving interstitial transport of macromolecules through

reduction in cell volume fraction in tumor tissues. Nanomedicine :

Nanotechnology, Biology, and Medicine. 2012, 8, 1088–1095.

283. Rhoden, J. J.; Wittrup, K. D. Dose dependence of intratumoral perivascular

distribution of monoclonal antibodies. Journal of Pharmaceutical Sciences. 2012,

101, 860–867.

284. Jang, B.-S.; Lee, S.-M.; Kim, H. S.; Shin, I. S.; Razjouyan, F.; Wang, S.; Yao, Z.;

Pastan, I.; Dreher, M. R.; Paik, C. H. Combined-modality radioimmunotherapy:

synergistic effect of paclitaxel and additive effect of bevacizumab. Nuclear

Medicine and Biology. 2011, 39, 472–483.

285. Luo, Y.; Ling, Y.; Guo, W.; Pang, J.; Liu, W.; Fang, Y.; Wen, X.; Wei, K.; Gao, X.

Docetaxel loaded oleic acid-coated hydroxyapatite nanoparticles enhance the

docetaxel-induced apoptosis through activation of caspase-2 in androgen

independent prostate cancer cells. Journal of Controlled Release. 2010, 147,

278–88.

286. Ostacolo, L.; Marra, M.; Ungaro, F.; Zappavigna, S.; Maglio, G.; Quaglia, F.;

Abbruzzese, A.; Caraglia, M. In vitro anticancer activity of docetaxel-loaded

micelles based on poly(ethylene oxide)-poly(epsilon-caprolactone) block

copolymers: Do nanocarrier properties have a role? Journal of Controlled

Release. 2010, 148, 255–63.

287. Sanna, V.; Roggio, A. M.; Posadino, A. M.; Cossu, A.; Marceddu, S.; Mariani, A.;

Alzari, V.; Uzzau, S.; Pintus, G.; Sechi, M. Novel docetaxel-loaded nanoparticles

based on poly(lactide-co-caprolactone) and poly(lactide-co-glycolide-co-

caprolactone) for prostate cancer treatment: formulation, characterization, and

cytotoxicity studies. Nanoscale Research Letters. 2011, 6, 260.

288. Kataoka, K.; Matsumoto, T.; Yokoyama, M.; Okano, T.; Sakurai, Y.; Fukushima,

S.; Okamoto, K.; Kwon, G. S. Doxorubicin-loaded poly(ethylene glycol)–poly(β-

Page 153: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

138

benzyl-L-aspartate) copolymer micelles : their pharmaceutical characteristics and

biological significance. Journal of Controlled Release. 2000, 64, 143–153.

289. Lee, S.-W.; Yun, M.-H.; Jeong, S. W.; In, C.-H.; Kim, J.-Y.; Seo, M.-H.; Pai, C.-M.;

Kim, S.-O. Development of docetaxel-loaded intravenous formulation, Nanoxel-

PMTM using polymer-based delivery system. Journal of Controlled Release. 2011,

155, 262–271.

290. Marcucci, F.; Corti, A. How to improve exposure of tumor cells to drugs: promoter

drugs increase tumor uptake and penetration of effector drugs. Advanced Drug

Delivery Reviews. 2012, 64, 53–68.

291. Marcucci, F.; Corti, A. Improving drug penetration to curb tumor drug resistance.

Drug Discovery Today. 2012, 17, 1139–46.

292. Kano, M. R.; Bae, Y.; Iwata, C.; Morishita, Y.; Yashiro, M.; Oka, M.; Fujii, T.;

Komuro, A.; Kiyono, K.; Kaminishi, M. et al. Improvement of cancer-targeting

therapy, using nanocarriers for intractable solid tumors by inhibition of TGF-beta

signaling. Proceedings of the National Academy of Sciences of the United States

of America. 2007, 104, 3460–5.

293. Kumagai, M.; Kano, M. R.; Morishita, Y.; Ota, M.; Imai, Y.; Nishiyama, N.; Sekino,

M.; Ueno, S.; Miyazono, K.; Kataoka, K. Enhanced magnetic resonance imaging

of experimental pancreatic tumor in vivo by block copolymer-coated magnetite

nanoparticles with TGF-beta inhibitor. Journal of Controlled Release. 2009, 140,

306–11.

294. Diop-Frimpong, B.; Chauhan, V. P.; Krane, S.; Boucher, Y.; Jain, R. K. Losartan

inhibits collagen I synthesis and improves the distribution and efficacy of

nanotherapeutics in tumors. Proceedings of the National Academy of Sciences of

the United States of America. 2011, 108, 2909–14.

295. Kuhn, S. J.; Finch, S. K.; Hallahan, D. E.; Giorgio, T. D. Proteolytic surface

functionalization enhances in vitro magnetic nanoparticle mobility through

extracellular matrix. Nano Letters. 2006, 6, 306–12.

Page 154: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

139

296. Goodman, T. T.; Olive, P. L.; Pun, S. H. Increased nanoparticle penetration in

collagenase-treated multicellular spheroids. International Journal of

Nanomedicine. 2007, 2, 265–274.

297. Eikenes, L.; Tari, M.; Tufto, I.; Bruland, O. S.; de Lange Davies, C. Hyaluronidase

induces a transcapillary pressure gradient and improves the distribution and

uptake of liposomal doxorubicin (Caelyx) in human osteosarcoma xenografts.

British Journal of Cancer. 2005, 93, 81–8.

298. Fukumura, D.; Jain, R. K. Tumor microvasculature and microenvironment: targets

for anti-angiogenesis and normalization. Microvascular Research. 2007, 74, 72–

84.

299. Chauhan, V. P.; Stylianopoulos, T.; Martin, J. D.; Popović, Z.; Chen, O.; Kamoun,

W. S.; Bawendi, M. G.; Fukumura, D.; Jain, R. K. Normalization of tumour blood

vessels improves the delivery of nanomedicines in a size-dependent manner.

Nature Nanotechnology. 2012, 7, 8–13.

300. Tailor, T. D.; Hanna, G.; Yarmolenko, P. S.; Dreher, M. R.; Betof, A. S.; Nixon, A.

B.; Spasojevic, I.; Dewhirst, M. W. Effect of pazopanib on tumor

microenvironment and liposome delivery. Molecular Cancer Therapeutics. 2010,

9, 1798–808.

301. Hauck, M. L.; Zalutsky, M. R. Enhanced tumour uptake of radiolabelled antibodies

by hyperthermia. Part II: Application of the thermal equivalency equation.

International Journal of Hyperthermia. 2005, 21, 13–27.

302. Khaibullina, A.; Jang, B.-S.; Sun, H.; Le, N.; Yu, S.; Frenkel, V.; Carrasquillo, J. a;

Pastan, I.; Li, K. C. P.; Paik, C. H. Pulsed high-intensity focused ultrasound

enhances uptake of radiolabeled monoclonal antibody to human epidermoid

tumor in nude mice. Journal of Nuclear Medicine. 2008, 49, 295–302.

303. Davies, C. D. L. Radiation Improves the Distribution and uptake of liposomal

doxorubicin (Caelyx) in human osteosarcoma xenografts. Cancer Research.

2004, 64, 547–553.

Page 155: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

140

Appendix 1: Chapter 3 Supplemental Data

Figure A1: a) Schematic representation of the analysis process using a macro

developed for ImageJ, version 1.44m. The volume was derived from the area of the

MCTS assuming a spherical shape: . b) Correlation between manual and

automated volume measurements of HeLa MCTS. 1000 cells were seeded and the

volume of the MCTS was determined every 2 days for 10 days by manually measuring

the diameter and automatically by image recognition using a macro for ImageJ.

Methods

MCTS were imaged at selected intervals of growth and their size was determined by

measuring their 2-D cross-sectional area using an automated image analysis macro

developed for use with the ImageJ software package (Version 1.44m). Images were

converted into 8-bit greyscale, the perimeter of an individual MCTS was recognized by

an automated threshold function and the area of the 2-D MCTS mask was recorded and

HeLa Volume

0 2.010 7 4.010 7 6.010 7

0

2.010 7

4.010 7

6.010 7

Volume of MCTS Manual Measure (m3)

Volu

me o

f M

CT

S A

uto

Measure

(m

3)

a)

b)

Page 156: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

141

converted to μm2 by calibration using an image of known scale. Finally, the volume of

the MCTS was calculated by assuming a spherical shape as follows:

where V is the volume and A is the measured area. Growth curves were plotted as

mean spheroid volume of 30 MCTS vs. time and fit using the Gompertz growth equation

[243] .

0 5000 10000 15000 200000.0

0.2

0.4

0.6HT29

HeLa

Cells per well

OD

405

0 10000 20000 300000.0

0.5

1.0

1.5

2.0

2.5

HeLa

HT29

Cells per spheroid

OD

405

0 200 400 600 8000.0

0.5

1.0

1.5

2.0

2.5HT29

HeLa

Spheroid diameter

OD

405

Figure A2: Validation of the acid phosphatase assay (APH) for HeLa and HT29 cells

grown in monolayer (left) and spheroid culture (center and right) demonstrating a linear

relationship between cell number and UV absorption at 405 nm. Each data point

represents the mean of three independent experiments ± SD (n = 3)

Figure A3. WST-8 assay showing non-linear correlation between the number of cells

and OD450 in spheroid culture.

Page 157: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

142

Figure A4. Fluroescence images of HT29 (a, b) and HeLa (c, d) tumor xenografts

displaying markers of hypoxia (EF5, blue) and blood vessels (CD31, red). Fluroescence

signal of 55 nm BCMs is shown in green at 1 h p.i. Scale bars represent 100 µm.

a) b)

c) d)

Page 158: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

143

Appendix 2: Chapter 4 Supplemental Data

Table T1. Area under the curve (AUC), Dmean (mean penetration distance), and intensity

values for fluorescent BCMs in HeLa and HT29 tumor xenografts at selected distances

from the nearest blood vessel.

Cell Line Sample

Time post injection, h AUC

Dmean, µm

Fluorescence Intensity at 5 µm

Fluorescence Intensity at 50 µm

Fluorescence Intensity at 100 µm

HeLa BCM55 1 148.51 27.2 9.78 +/- 1.74 1.40 +/- 0.58 0.40 +/- 0.12

6 160.92 30.4 9.70 +/- 2.27 2.03 +/- 0.56 0.62 +/- 0.44

24 154.38 36.8 5.20 +/- 1.14 3.28 +/- 0.48 0.67 +/- 0.30

BCM15 1 176.09 33.9 9.76 +/- 0.98 2.16 +/- 0.39 1.11 +/- 0.12

6 150.72 40.2 6.00 +/- 1.17 2.59 +/- 0.64 1.33 +/- 0.61

24 117.38 37.2 4.19 +/- 0.62 1.90 +/- 0.46 0.32 +/- 0.03

HT29 BCM55 1 61.32 18.7 6.36 +/- 0.72 0.26 +/- 0.24 0.09 +/- 0.06

6 82.60 26.3 7.06 +/- 1.19 0.61 +/- 0.30 0.26 +/- 0.23

24 112.76 32.6 6.51 +/- 0.79 1.24 +/- 0.69 0.48 +/- 0.02

BCM15 1 142.37 35.9 6.31 +/- 1.48 1.83 +/- 0.51 0.86 +/- 0.06

6 134.68 37.8 5.65 +/- 1.37 1.83 +/- 0.95 0.81 +/- 0.31

24 68.89 29.3 5.24 +/- 0.69 0.41 +/- 0.22 0.17 +/- 0.09

0.00

0.05

0.10

0.15

0.20

HeLa

HT29

co

lla

ge

n

g/m

g w

et

tissu

e

Figure A1. Collagen content in HeLa and HT29 MCTS after 7 days of growth. Collagen

content is reported per mg of wet tissue ± SD.

Page 159: Development and Intratumoral Distribution of Block Copolymer Micelles … · 2014-07-01 · iii critical to the development of optimized delivery vehicles capable of fully exploiting

144

Method

Collagen content was determined by first incubating MCTS in papain digest buffer (125

µg/ml papain, 0.1 M Na-Phosphate, 5 mM Na2EDTA and 5 mM Cystein-HCL) at 60 °C

overnight. Solubilized samples were hydrolyzed in 6 M HCL at 110 °C for 24 h prior to

evaporation of the acid at 65 °C and further drying under vacuum conditions. The

residue was dissolved and neutralized in a 1:10 dilution of the stock buffer (stock buffer:

18.08 g sodium acetate, 12.5 g citric acid, 3 ml glacial acetic acid, 8.5 g sodium

hydroxide in 250ml ddH2O; pH 6.5). Chloramine-T solution (70 mg chloroamine T in 1

ml of ddH2O, 1.5 ml Methyl cellosolve and 2.5 ml stock buffer) was added to the sample

and incubated for 20 min at RT. Following disintegration of chloroamine T by 3.15 M

perchloric acid, Ehrlichs reagent was added to the sample (200 mg of dimethyle-

aminobenzaldehyde dissolved in 1ml of Methyl cellosolve) and incubated 20 min at 60

°C. After cooling in cold water the absorbance was measured within 30 min using a UV

spectrometer (SpectraMax Plus 384, Molecular Devices, Sunnyvale, CA) at 560 nm.

Concentration of the hydroxyproline was determined from a standard curve and

collagen content was calculated assuming 13% hydroxyproline content.