87
Comparison of CT and Optical Image-based Assessment of Liposome Distribution by Huang Huang A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Pharmaceutical Sciences University of Toronto © Copyright by Huang Huang 2012

Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

Comparison of CT and Optical Image-based Assessment of Liposome Distribution

by

Huang Huang

A thesis submitted in conformity with the requirements for the degree of Master of Science

Graduate Department of Pharmaceutical Sciences University of Toronto

© Copyright by Huang Huang 2012

Page 2: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

ii

Comparison of CT and Optical Image-based Assessment of

Liposome Distribution

Huang Huang

Master of Science

Department of Pharmaceutical Sciences

University of Toronto

2012

Abstract

The use of multimodal imaging as a tool to assess the in vivo pharmacokinetics and

biodistribution of nanoparticles is important in drug development and imaging-guided therapy.

The current study reports the use of combined computed tomography (micro-CT) and optical

imaging to quantitatively assess the whole body (macro) and intratumoural (micro) distribution

of a nano-sized liposome-based CT/optical imaging probe carrying iohexol and Cy5.5 over a

study period of eight days. The liposomes were found to have a vascular half-life of 30.3 ± 8.9 h

in mice bearing subcutaneous H520 non-small cell lung cancer (NSCLC) tumours with the

maximum liposome accumulation in tumour achieved 48 h post injection. The liposome

accumulation in tumour was quantitatively assessed using both micro-CT and fluorescence

molecular tomography (FMT), where micro-CT was used to guide the FMT tumour delineation

and signal correction. In situ confocal laser-scanning fluorescence microscopy analysis at the

tumor site revealed that most of the liposomes remain in the vasculature at early time points (24

h) with the majority having extravasated into the tumor interstitium at later time points (eight

days). This investigation demonstrates the critical role micro-CT can play in guiding FMT-based

Page 3: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

iii

quantification of distribution. As well the combination of CT and optical imaging enable

visualization of the liposomes at the whole body, tumor and cellular levels with high sensitivity

and excellent anatomical background. Such non-invasive assessment of therapeutic distribution

at the macro and micro scale is necessary for implementation of personalized medicine including

image-guided patient stratification and real-time adjustment of therapeutic dose.

Page 4: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

iv

Acknowledgments

It is a great pleasure to thank the many people who made this thesis possible.

Foremost, I would like to express my sincere gratitude to my supervisor, Dr. Christine Allen.

With her enthusiasm, her inspiration, and her continuous support for my study and research, she

helped me enjoy the research I did. I deeply appreciate all her contributions of time and patience

to make my M.Sc. experience exciting. Through my thesis writing, she provided encouragement,

sound advice, and good teaching. It has been a pleasure to be her student.

My committee member and unofficial co-supervisor, Dr. David Jaffray, has provided a lot of

guidance to me during this research. I am especially grateful for his unique thinking and diverse

knowledge, which provided me many ideas that have been very helpful. I appreciate Drs. Allen

and Jaffray for obtaining the funding that has supported my research.

I would like to thank my other committee member, Dr. Gang Zheng, for taking his time to

critically review my work and offer constructive suggestions.

My sincere thanks to all my colleagues and friends in the Allen lab and at STTARR, past and

present. In particular, I would like to express my special gratitude toward John Lo and Michael

Dunne. John took the time to train me as an undergraduate student, and provided collaborative

help that paved the way to this research. I thank Michael for using the H520 tumour model he

developed in our lab. His generous help on tumour cell preparation and animal inoculation

described in this research is also deeply appreciated. I would also like to thank Dr. Jinzi Zheng

for providing scientific support, and Debbie Squires for technical assistance with the

administration of the contrast agents.

Finally, I would like to thank my beloved family and friends for all their love, support and

encouragement throughout my studies. Thank you all.

Page 5: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

v

Table of Contents

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

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

List of Tables ................................................................................................................................ vii

List of Figures .............................................................................................................................. viii

List of Appendices ......................................................................................................................... xi

CHAPTER 1 GENERAL INTRODUCTION .............................................................................. 1

1.1 Goal, Objectives and Rationale........................................................................................... 1

1.2 Liposomes ........................................................................................................................... 3

2.2.1 Physico-chemical Properties................................................................................... 3

2.2.2 Liposomes as Drug Carriers.................................................................................... 6

1.3 Multimodal Imaging ......................................................................................................... 10

2.2.1 CT Imaging ........................................................................................................... 11

2.2.2 Optical Imaging .................................................................................................... 12

2.2.3 CT/Optical Multimodal Imaging .......................................................................... 14

1.4 Image-based Assessment of Nanoparticle Distribution.................................................... 15

CHAPTER 2 Comparison of CT and Optical Image-based Assessment of Liposome Distribution .............................................................................................................................. 19

2.1 Introduction....................................................................................................................... 20

2.2 Materials and Methods...................................................................................................... 23

2.2.1 Synthesis of Cy5.5-DSPE ..................................................................................... 23

2.2.2 Liposome Preparation ........................................................................................... 24

2.2.3 Liposome Characterization ................................................................................... 24

2.2.4 In vitro Stability and Agent Release ..................................................................... 25

Page 6: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

vi

2.2.5 In vivo CT/Optical Imaging .................................................................................. 26

2.2.6 Image Analysis...................................................................................................... 27

2.3 Results............................................................................................................................... 30

2.3.1 Synthesis and Characterization of Cy5.5-DSPE................................................... 30

2.3.2 Physicochemical Characterization of the Liposomes ........................................... 31

2.3.3 In vivo Pharmacokinetics, Biodistribution of the Liposomes ............................... 35

2.4 Discussion ......................................................................................................................... 46

CHAPTER 3 Conclusion............................................................................................................. 50

3.1 Conclusions....................................................................................................................... 50

3.2 Future Directions .............................................................................................................. 52

References..................................................................................................................................... 54

Appendix....................................................................................................................................... 67

Page 7: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

vii

List of Tables

Table 2.1 Physicochemical characteristics of CT and CT/optical liposomes (n=5). Data

are represented as the mean ± standard deviation .............................................................. 32

Table 2.2 Pharmacokinetic parameters for CT and CT/optical liposomes (n=7) in non-

tumour-bearing mice and CT/optical liposomes (n=5) in tumour-bearing mice. Statistics

were analyzed between the CT and CT/optical liposomes in non-tumour-bearing mice,

and between CT/optical liposomes in non-tumour and tumour-bearing mice .................. 36

Page 8: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

viii

List of Figures

Figure 2.1 Schematic of the CT/optical liposome-based contrast agent (not drawn to scale). The

average hydrodynamic diameter of the liposomes was found to be 86 ± 5 nm by dynamic light

scattering (DLS)............................................................................................................................ 22

Figure 2.2 The mass spectrum of Cy5.5-DSPE. The measured molecular weight of Cy5.5-DSPE

was determined to be 1645.7 Da, which is in good agreement with the theoretical molecular

weight 1646 Da. ............................................................................................................................ 30

Figure 2.3 Representative UPLC chromatograms for Cy5.5 and purified Cy5.5-DSPE. Cy5.5

elutes at ~3.5 min, Cy5.5-DSPE elutes at ~7.2 min. The distinction between the two sets of peaks

assures the purity achieved using the column chromatography method....................................... 31

Figure 2.4 Hydrodynamic diameter (obtained by DLS analysis) of the CT/optical liposomes over

a 2-week period with dialysis against a 250-fold volume excess of HBS containing 45mg/mL

BSA at 37°C. Data are represented as the mean ± standard deviation of three independent

batches of liposomes. .................................................................................................................... 33

Figure 2.5 (a) The in vitro release profile of iohexol and Cy5.5 from CT/optical liposomes (n=3)

and iohexol from CT liposomes (n=3) incubated in FBS at 37°C. (b) Ratio of iohexol to Cy5.5

(wt/wt) in the CT/optical liposomes following incubation in FBS at 37°C. The initial iohexol-to-

Cy5.5 ratio was normalized to 1. * indicates statistically significant difference (p < 0.01) from t =

0..................................................................................................................................................... 34

Figure 2.6 Micro-CT (top) and FMT (bottom) images of a non-tumour bearing mouse injected

with CT/optical liposomes. The same window and level were used for all images from each

modality. ....................................................................................................................................... 37

Figure 2.7 Micro-CT transverse (top), coronal (middle), and FMT images of a tumour bearing

mouse injected with CT/optical liposomes at various time points. The transverse micro-CT

images display the right side of the mouse. Arrow indicates the location of the tumour. The same

window and level was used for images from each modality. ....................................................... 38

Page 9: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

ix

Figure 2.8 Pharmacokinetics and biodistribution profiles of CT and CT/optical liposomes in

non-tumour bearing mice as determined using micro-CT image-based assessment. (a) The blood

iodine concentration. The biodistribution in (b) liver, (c) spleen, (d) left kidney and (e) right

kidney. * indicates statistically significant differences (p<0.05) between CT and CT/optical

liposomes in %ID/cm3 tissue. ....................................................................................................... 41

Figure 2.9 (a) Pharmacokinetics of CT/optical liposomes in tumour bearing mice as determined

using micro-CT image-based assessment of the blood iodine concentration. (b) Micro-CT and

FMT image-based determination of tumor distribution profile for CT/optical liposomes (n=5). *

indicates a statistically significant difference between micro-CT and FMT data at that time point.

(c) Ratio of iohexol to Cy5.5 (wt/wt) at the tumour site. The initial iohexol-to-Cy5.5 ratio was

normalized to 1. * indicates statistically significant difference from pre-injection. (d) Iohexol-to-

Cy5.5 ratio (wt/wt) determined from various concentric cubic VOIs over the tumour region in

both FMT and micro-CT. Data include liposome accumulation in tumours 24 to 72 h post-

injection. The tumour volumes are 25 to 30 mm3. The ratios were normalized to the iohexol-to-

Cy5.5 ratio pre-injection. .............................................................................................................. 44

Figure 2.10 In vivo microscopic images of the CT/optical liposomes in the tumour area at 24 h

(a, b) and 8 days (c) post-injection................................................................................................ 45

Figure A1. (a) Inside the FMT system. The imaging cassette (as seen in (b)) is placed in the

scanning stage prior to a scan. (b) A photo of the phantom gel inside the imaging cassette. The

gel insert (1 x 1 x 1 cm3) is fused inside the large gel (3.8 x 3.8 x 1.3 cm3), and the cassette

height is adjusted to 1.3 cm to sandwich and tightly immobilize the gel. (c) A schematic of the

large gel geometry with the gel inserted either toward or away from the source laser. Both

geometries were considered In this study. .................................................................................... 70

Figure A2. Effect of background concentration (a-f) on signal detected in ROI. The gel insert

(200 nM GH680) is facing the camera with various backgrounds (a) 20 nM, (b) 50 nM, (c) 80

nM, (d) 150 nM, (e) 200 nM, and (f) 300 nM. The dotted contours (5 mm x 5 mm) represent the

ROIs used for the analysis. The positions of the ROIs were located using both the FMT intensity

as well as the reflectance image. Slices shown have been averaged over 5mm in the z-dimension

for illustration purposes. ............................................................................................................... 71

Page 10: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

x

Figure A3. FMT GH680 concentrations vs. actual GH680 concentrations at various background

levels (n=3 per level). The dotted line represents the line of identity. The FMT concentration

increases substantially as the background level increases. At the lowest background of 20 nM,

the quantified concentrations are very close to the actual concentrations in the gel cubes.

Background below 20 nM was found to be equivalent to zero background. At any given

background, linearity is still preserved for varying GH680 concentrations in the gel cubes (R2 >

0.95 for all fits). ............................................................................................................................ 72

Figure A4. The quantification error of the FMT for the gel inserts with various background

levels before and after applying background subtraction method using the method reported here.

The signal-to-background (S/B) ratio has been used to express the strength of the FMT signal in

the gel cube compared to background (BKG). Data are expressed as mean ± standard deviation

(n=3 to 12). The dotted line indicates a S/B ratio of 1, representing that the GH680

concentrations in the gel cube and background are the same. As shown, the quantification error

before background subtraction followed an exponential growth as the S/B ratio decreased,

reaching 908 ± 89% at S/B ratio of 0.167. Overall, with background subtraction, the

quantification improved substantially (p < 0.05).......................................................................... 73

Page 11: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

xi

List of Appendices

Appendix……………………………………………………………………………...…………67

Page 12: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

xii

List of Abbreviations

AUC – area under the curve

BSA – bovine serum albumin

CH – cholesterol

CT – computed tomography

DICOM – digital imaging and communications in medicine

DLS – dynamic light scattering

DOPE – 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine

DOTA – 1,4,7,10-tetraazacyclododecane -1,4,7,10-tetraacetic acid

DPPC – dipalmitoylphosphatidylcholine

DSPE – Distearoylphosphatidylethanolamine

DTPA – diethylene triamine pentaacetic acid

EMPA – Poly(glycidyl methacrylate)poly(2,3,-epoxypropylmethacrylate)

EPR – enhanced permeation and retention

FBS – fetal bovine serum

FDA – food and drug administration

FMT – fluorescence molecular tomography

FRV – freeze-drying rehydration vesicles

Gd – Gadolinium

Page 13: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

xiii

HBS – HEPES buffered saline

HDL – high density lipoprotein

HEPES – hydroxyethylpiperazine ethanesulfonic acid

HU – Hounsfield unit

ID – injected dose

ke – elimination constant

LDL – low density lipoprotein

LUV – large unilamellar vesicles

MRI – magnetic resonance imaging

MWCO – molecular weight cut off

NSCLC – non-small cell lung cancer

PC – phosphatidylcholine

PE –Phosphatidylethanolamine

PEG – polyethylene glycol

PET – positron emission tomography

PS –Phosphatidylserine

RES – reticuloendothelial system

ROI – region of interest

SPECT – single photon emission computed tomography

SUV – small unilamellar vesicles

Page 14: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

xiv

t1/2 – half life

UPLC – ultra-performance liquid chromatography

US – ultrasound

UV – ultraviolet

VD – volume of distribution

Page 15: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

1

CHAPTER 1 GENERAL INTRODUCTION

1.1 Goal, Objectives and Rationale

The overall goal of this research is to evaluate and compare the in vivo distribution of liposomes

using CT and optical imaging modalities. The specific objectives include:

1. Synthesis and in vitro characterization of liposomes that support CT and optical imaging.

2. Quantitative in vivo comparison of whole body distribution and tumor accumulation

profiles of the CT/optical liposomes obtained by CT and optical imaging.

3. Assessment of the CT and optical imaging modalities in their ability to quantify liposome

distribution in vivo.

Medical imaging is one of the primary tools used to evaluate structure and function non-

invasively in a living subject [1-8]. The employment of imaging to assess the distribution of

nanoparticles and drug delivery systems in vivo has provided invaluable insight into the pathway

and fate of these systems [9-15]. This unique information enables the optimization of delivery

systems and may accelerate the development of therapeutic strategies. In cancer research, by

loading a drug delivery system with therapeutic and imaging agents, one enables image-guided

drug delivery for the real-time monitoring of pharmacokinetics and whole-body distribution as

well as tumour accumulation, and intratumoural distribution. Such multifunctional delivery

systems may ultimately contribute to the development of personalized therapies and lead to

improvements in treatment outcomes.

In recent years, multimodal imaging has received increasing interest as this provides a

means to exploit the unique strengths of each modality and to gain complementary information

[16-21]. In this study, CT and optical imaging were selected to assess the in vivo distribution of

liposomes. CT imaging provides anatomic information and has been shown by our laboratory to

reliably enable quantitative assessment of the distribution of liposomes in vivo [22-24].

Therefore CT may be considered a reference that enables assessment and validation of the

Page 16: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

2

performance of other imaging modalities in the quantitative evaluation of biodistribution of

liposomes. On the other hand, optical imaging offers high detection sensitivity and can provide

information on liposome distribution at the cellular level via microscopy or endoscopy

measurements. In addition, the development of FMT allows optical imaging to three-

dimensionally and quantitatively image the whole-body biodistribution of fluorophores in small

animals, enabling data registration and comparison with CT and other tomographical imaging

modalities [25, 26]. Therefore, the combination of CT and optical imaging, should enable

monitoring of the macro and microdistribution of liposomes in vivo, and allow for data

comparison and validation between CT and FMT.

Liposomes are the most established of the advanced drug delivery technologies. Several

drugs relying on formulation in liposomes have been approved by the FDA and used for the

treatment of cancer, infectious disease and autoimmune diseases [27-30]. As drug delivery

vehicles, liposomes have the ability to incorporate hydrophilic, hydrophobic and/or amphiphilic

molecules [16, 31]. Through the optimization of their physico-chemical properties such as size,

charge, and stability, liposomes can achieve the desired in vivo pharmacokinetic profile. In

oncology, properly designed liposomes can take advantage of the leaky tumour vasculature to

passively accumulate at tumour sites [32, 33]. The utilization of liposomes as carriers for

imaging agents has great potential in the medical imaging field as they can be easily labeled or

loaded with probes, contrast agents, or radionuclides to support imaging in single or multiple

modalities [34-37]. Image-based assessment of the in vivo distribution of liposomes can provide

unique information that is otherwise unavailable using traditional methods [38-42].

The experiments performed to accomplish the objectives outlind above are detailed in

Chapter 2 of this dissertation. Briefly, CT/optical liposomes were prepared with a CT contrast

agent encapsulated within the liposomes and a fluorophore was conjugated to the surface of the

liposomes. The liposome formulation was characterized in vitro in terms of size and size

distribution, zeta potential, agent loading levels, and agent release profiles. Quantitative micro-

CT, FMT and intravital microscopy were used at several time points to determine the

biodistribution of the liposomes for eight days following intravenous administration to mice

bearing subcutaneous H520 non-small cell lung cancer tumours.

Page 17: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

3

1.2 Liposomes

Liposomes are vesicles with an internal aqueous volume surrounded by one or more concentric

bilayers of lipids. In an aqueous medium, lipids will be thermodynamically driven to form

liposomes, creating a hydrophilic outer surface and inner core, as well as a hydrophobic domain

within the bilayer. This property allows the encapsulation of molecules in either the hydrophobic

or hydrophilic compartments, as part of the lipid framework or free in the internal aqueous

volume, respectively. Since their discovery in 1965 [43], liposomes have been explored broadly

in the field of pharmaceutics for the encapsulation and controlled delivery of various

chemotherapeutic and anti-fungal drugs [29]. As well, liposomes have shown promises in cancer

imaging applications such as disease detection, characterization and staging (ref). Compared to

their unencapsulated counterparts, drugs administered in liposome formulations experience

longer circulation lifetimes and reduced systemic toxicities. Through the selection of their

composition and the method of preparation, one can control various parameters such as size,

drug loading and retention, in vivo circulation lifetime, as well as their in vivo distribution.

Several liposome formulations for chemotherapy have been approved by the FDA for human use

with many other formulations in preclinical and clinical evaluation. For example, Doxil, the

pegylated liposome formulation of doxorubicin, has been approved for the treatment of ovarian

cancer [44]. Another approved liposome formulation, DaunoXome, is used for treatment of

Kaposi’s scarcoma [45]. In addition to chemotherapy, Visudyne, a liposomal formulation, has

been approved by the FDA for the treatment of macular degeneration [46].

2.2.1 Physico-chemical Properties

A liposome consists of several components analogous to that of a cell membrane. The main

component of the liposome is phospholipid, which generally includes phosphatidylcholines (PC),

phosphatidylethanolamines (PE), and phosphatidylserines (PS). Phospholipids are amphiphilic

molecules consisting of a polar head group and two non-polar tail chains, which have various

lengths and degrees of saturation. These phospholipids comprise at least 40% of the liposome,

and are the main building blocks of the liposome membrane. A sterol, commonly cholesterol, is

another component in the liposome, and serves as structural support for the lipid bilayer by

increasing the fluidity of rigid membranes and rigidity of fluid membranes [47]. The presence of

Page 18: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

4

cholesterol in liposomes also increases the encapsulation efficiency of hydrophilic agents

through decreasing the permeability of the bilayer [48]. On the other hand, cholesterol has been

shown to decrease solubilization capacity for hydrophobic drugs, possibly due to that the fact

that cholesterol is a bulky and robust molecule intercalated within the hydrophobic region of the

bilayer, thus reducing the space available for loading hydrophobic agents [49-51]. In addition,

inclusion of cholesterol in the lipid bilayer has also been shown to inhibit the transfer of lipid

components to plasma lipoproteins, resulting in improved in vivo stability [52]. In addition to

cholesterol, poly(ethylene glycol) (PEG) has been widely exploited in liposome formulations to

improve their in vivo performance. PEG is a non-ionic, water-compatible, flexible polymer chain

that is usually conjugated to a lipid such as DSPE that serves to anchor the hydrophilic polymer

to the bilayer surface. The presence of PEG provides steric stability and such liposomes are

known to have enhanced circulation lifetimes and reduced accumulation in the liver and spleen,

in comparison to conventional liposomes [53]. This may be due to the theory that PEG

discourages liposome self-aggregation, reduces the binding of serum opsonins, and increases

surface hydrophilicity. Studies have shown that 5 mol% of PEG2000-lipid is optimal in prolonging

the circulation lifetime of liposomes [54]. Such steric stabilization is critical for targeting

liposomes to solid tumors, where a longer circulation life time leads to increased tumour

accumulation.

Liposomes can be classified into four different categories depending on their size and the

method of preparation employed: small unilamellar vesicles (SUV), multilamellar vesicles

(MLV), large unilamellar vesicles (LUV), and freeze-drying rehydration vesicles (FRV). There

are several steps in liposome preparation. The first step is lipid dispersion, which relies on the

dispersion of the lipid in a solution, so that the lipids may self-assemble into various forms of

lipid spheres with aqueous internal volumes. Any soluble molecule can be encapsulated into the

internal aqueous volume of the liposomes or inserted into the bilayer (if attached to a

phospholipid) by inclusion in the aqueous solution during liposome formation. Common

methods for lipid dispersion include mechanical dispersion, solvent dispersion, emulsion

preparation, or detergent solubilization [55]. Next, the liposomes are usually extruded to result in

a well-defined size and a narrow size distribution. The extrusion step uses high pressure to force

the liposomes through membranes with pores of a certain size [56]. Extrusion typically produces

SUVs with a unimodal size distribution [57, 58].

Page 19: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

5

The size of liposomes is critical in controlling their in vivo circulation time. It has been

shown that liposomes smaller than 100 nm have a longer circulation lifetime than liposomes

greater than 200 nm in size, since the smaller liposomes are cleared less rapidly by the RES [59].

Liposome size is also important in dictating the liposome’s ability to take advantage of the

enhanced permeability and retention (EPR) effect, which occurs in solid tumors and at sites of

inflammation. Tumour vasculature lacks a smooth muscle cell layer, and includes large

fenestrations (200-4000 nm) caused by poor alignment of endothelial cells [33]. This results in

enhanced permeation of liposomes smaller than 200 nm in diameter through the fenestrations

and into the tumour interstitial space. In addition, the poor lymphatic drainage of the tumour

tissue creates the enhanced retention of liposomes, leading to a high accumulation of liposomes

in tumour over time. It has been shown that liposomes up to 400 nm in diameter are able to take

advantage of the EPR effect, but smaller liposomes will do so to a greater extent [60]. Therefore,

size and circulation lifetime of the liposomes are crucial in determining the extent of the EPR

effect.

There are several vascular mediators that affect the EPR effect, including bradykinin,

nitric oxide (NO), prostaglandins, angiogensin-converting enzyme inhibitors, vascular

permeability factor and other cytokines. Two methods to artificially promote the EPR effect for

more efficient tumour-selective drug delivery was reported. One method is the elevation of

systemic blood pressure by iv infusion of angiotensin II [61-63]. Induction of systemic

hypertension leads to increased blood flow and vasoconstriction in normal tissues, resulting in no

change in blood flow volume. On the contrary, tumour blood vessels usually lack a smooth

muscle layer or pericytes needed for vasoconstriction. Therefore, during hypertension, tumour

blood vessels would open, leading to increased blood flow and increased drug delivery [62].

Elevated blood pressure was suggested as a strategy to increase tumor-targeted delivery of

SMANCS [64]. Another method is through the use nitroglycerin and similar nitric oxide (NO)

releasing agents. In hypoxic tumour tissue, nitroglycerin can be converted into nitrite, which can

then be reduced to NO [65]. NO is one of the potent mediators of vascular extravasation. It has

been shown that administration of nitroglycerin induced a two-fold increase in NO in tumor

tissue in a dose dependent manner, whereas normal tissues showed no significant increase [66].

In a study with tumour-bearing mice, nitroglycerin ointment was topically applied over the tumor

or on skin opposite or distal to the tumor site, at doses of 1.0 μg/mouse to 1.0 mg/mouse, the

Page 20: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

6

increased NO level led to significant elevation in blood flow in tumour tissue, and the EPR effect

was seen to increase substantially. This led to significantly augmented accumulation of both the

Evans blue/albumin complex and the macromolecular anticancer drug PEG conjugated zinc

protoporphyrin (PZP) in all tumors studied [67].

2.2.2 Liposomes as Drug Carriers

Due to their amphiphilic nature, liposomes have been widely explored to incorporate both

hydrophilic and hydrophobic drugs, such as doxorubicin, cisplatin, paclitaxel, camptothecin, and

vincristine [68, 69]. Hydrophilic drugs can be easily encapsulated in the internal aqueous volume

of the liposome, while hydrophobic drugs may be incorporated within the hydrophobic region of

the lipid bilayer. Drugs can be passively and actively loaded into the liposomes. In passive

loading, drugs and lipids are co-dispersed in an aqueous medium, thus achieving encapsulation

while liposomes are being formed. The loading efficiencies for passive encapsulation are usually

very low [70]. Active loading involves the creation of a gradient (i.e. pH, ionic) across the lipid

membrane, which causes the drug to enter the aqueous core. For example, Doxil® is prepared by

loading doxorubicin into liposomes using an ammonium sulphate gradient [71-73].

Taking advantage of the EPR effect, liposomes can exploit passive targeting to increase

the localization of anticancer drugs at solid tumours. Passive targeting can result in increases in

drug concentrations in solid tumours of several fold compared to levels obtained following

administration of free drug [74]. Liposomes can also be actively targeted to solid tumors by

conjugating targeting moieties to their surface. Over the last few decades, significant research

has been dedicated to the development of actively targeted liposome formulations [29, 75].

Targeting moieties may include antibody, small molecular weight, naturally occurring or

synthetic ligands like carbohydrates, glycoproteins, peptides, or receptor ligands, i.e. essentially

any molecule that selectively recognizes and binds to target antigens or receptors over-expressed

or specifically expressed on cancer cells [76]. There are several advantages of employing

actively targeted liposomes. First of all, relatively few ligand molecules are required per

liposome to deliver high amounts of drugs to target cells. Compared with other delivery systems

such as immunoconjugates, which can only deliver a few drugs (<10) per antibody molecule,

liposomes have the ability to deliver thousands of drug molecules using a few tens of antibody or

Page 21: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

7

ligand molecules on the liposomal surface [77-79]. In addition, the presence of multiple targeting

molecules on a single liposome can promote multivalent binding of monovalent antibody

fragments, thus greatly increasing their binding avidity for the targets. Another advantage of

targeted liposomes lies in the potential of enabling additive or synergistic effects between

signaling antibodies present on the liposome surface and the encapsulated drug. Antibodies have

been shown to exhibit additivity or synergy when used in combination with drugs [79-81]. For

example, clinical studies have demonstrated the additive benefits for an antibody against anti-

HER2 in combination with taxol or in combination with anthracycline drugs plus

cyclophosphamide [79].

In active targeting strategies using liposomes, the choice of the target receptor, targeting

ligand, and the drug are all important factors to be considered. The target receptor should be

either selectively expressed or over-expressed on malignant cells. These targeted cells should

possess minimum heterogeneity in their receptor expression with minimum shedding of the

receptors. The location of the receptors is also an important factor. Targets within the vasculature

or readily accessible from the vasculature should bind targeted liposomes more readily than

targets buried deep within tissues. Vascular targets are accessible to the liposomes as long as

they remain in the blood pool, while targeting to tumour cells requires the passive accumulation

of liposomes at the tumour site, followed by binding of the ligand to the target receptor on the

cell surface. Targeting ligands that have been explored most extensively include antibodies,

fragments of antibodies, peptides, aptamers, vitamins, and carbohydrates [82]. The factors

affecting ligand selection include binding affinity, avidity, immunogenicity, and ligand density

on liposomes [76]. In addition, since the ligands are surface bound, it is important to ensure that

these ligands do not seriously compromise the pharmacokinetics of the liposomes. In choosing

the drug to be encapsulated, one should consider the drug release rates and their bioavailability.

It is desired that when liposomes are at the tumour site or internalized into cells by receptor-

mediated endocytosis, the drugs are readily released. Strategies that enable remote drug release

at the tumour site include the design of pH and thermo sensitive liposomes [83, 84].

There have been several promising applications of antibody targeted liposomes in

research. For example, anti-HER2 immunoliposomes were developed to target mammary

carcinoma cells [79, 85, 86]. In these studies, anti-HER2 immunoliposomes were produced by

coupling the doxorubicin loaded liposomes with recombinant human mAb HER2-Fab’ or anti-

Page 22: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

8

HER2 scFv C6.5. After administration in nude mice bearing HER2-over-expressing tumor

xenografts, the anti-HER2 immunoliposomes resulted in efficient tumor localization, with

doxorubicin tumour-to-blood and tumour-to-muscle ratios greater than 22-fold at 67 h post

injection. Histological samples with immunoliposomes containing colloidal gold showed that the

immunoliposomes were present in both the perivascular areas and within cellular regions of the

tumour. In contrast, non-targeted liposomes were observed mainly in extracellular and

perivascular spaces, and were not observed within individual tumour cells. In multiple HER2-

overexpressing human breast tumor xenograft models, treatment with doxorubicin loaded anti-

HER2 immunoliposomes produces significantly increased efficacy as compared to free dox or

dox-loaded non-targeted liposomes and significantly less systemic toxicity than free dox [79]. It

was later demonstrated that the effect was due to the specific targeting of the liposomes, and not

due to effects of the targeting agent or free drug alone. Therefore, it was concluded that the

therapeutic advantage associated with doxorubicin-loaded anti-HER2 immunoliposomes was due

to specific intracellular drug delivery to the target cells. Anti-HER2 immunoliposomes are

currently undergoing scale up for clinical evaluation [76].

Therapeutic efficacy was also demonstrated for anti-CD19 targeted liposomes in a

murine model of human B-cell lymphoma. Anti-CD19 targeted liposomes were shown to

efficiently bind and internalize into maliganant B cells compared to non-targeted liposomes. In

vivo survival studies performed in SCID mice xenografts demonstrated a significantly increased

life span for mice treated with anti-CD19 targeted liposomes loaded with either doxorubicin or

vincristine, compared to mice treated with non-targeted liposomes or free drugs [87].

In addition to targeting cancer cells, liposomes can also be designed to carry targeting

moieties that target the tumour vasculature. Aside from greater accessibility of the liposomes to

the tumour vasculature, there is limited variation in the vasculature between tumour types. As a

result, vascular targeting is more generally applicable than specialized tumour cell-targeted

treatment [88]. RGD liposomes loaded with doxorubicin were designed to target the αvβ3

integrins that are over-expressed on neovasculature [89]. It was demonstrated that compared to

non-targeted liposomes and non-specifically targeted RAD liposomes, the RGD liposomes

exhibited faster blood clearance and increased spleen uptake, likely due to the presence of αvβ3

integrins on MPS cells in the spleen. However, tumour accumulation at 24 h was unchanged and

animals administered the RGD-liposomes experienced markedly decreased tumour growth. In

Page 23: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

9

another study, a novel peptide GPLPLR was attached to the liposomes to target membrane type

1-matrix metalloproteinase (MT1-MMP), which can be found on both vascular endothelial cells

and tumour cells [90]. PET imaging with liposome-encapsulated 18F-FDG demonstrated an

increase in tumour accumulation for the targeted formulation compared to the non-targeted

liposomes. Furthermore, incorporation of a cytotoxic peptide into the liposome resulted in a

significant increase in therapeutic efficacy.

Dunne M. et al attached the amino acid sequence asparagine-glycine-arginine (NGR) to

the liposome surface to target matrix metalloprotease aminopeptidase N (APN/CD13) expressed

on tumour vasculature [22]. This study showed a two fold increase in tumour accumulation of

the NGR liposomes compared with the non-targeted and control liposomes. In addition, it was

demonstrated that the inclusion of a longer PEG chain (i.e. PEG3400 versus PEG2000) at the

surface of the liposome was found to alter the shape of the tumour accumulation versus time

profile.

In addition to therapeutic applications, passively and actively targeted liposomes are

being explored for diagnostic applications. Liposomes labeled with various imaging agents have

already been shown to hold potential for detecting diseases, as well as for visualizing various

important aspects of the drug delivery process. In addition to this, liposome formulations are

being prepared with to contain both imaging and therapeutic agents. Compared to conventional

therapeutic liposomes, such imageable delivery systems take advantage of medical imaging and

can be used to noninvasively assess biodistribution and target site accumulation, to monitor and

quantify drug release, to facilitate therapeutic intervention, to predict therapeutic response, and to

longitudinally monitor the efficacy of therapeutic interventions [91]. Such image-guided therapy

will provide great potential in the development of personalized medicine.

Page 24: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

10

1.3 Multimodal Imaging

There has been an increase in the use of non-invasive imaging techniques for clinical diagnosis,

treatment guidance, as well as drug discovery and development research [92-94]. Examples

include computed tomography (CT), optical, magnetic resonance (MR), single photon emission

CT (SPECT), positron emission tomography (PET), and ultrasound (US) [95-100]. These

imaging modalities have been successfully employed to investigate anatomical or functional

information of tissues in the body. Each imaging modality has its own inherent strengths and

limitations and differs in terms of sensitivity of detection, spatial and temporal resolution, depth

of penetration into tissues, accuracy, cost and 3D tomography. For instance, CT and MR provide

a high degree of spatial resolution with 3D tomography but are limited by low sensitivity. On the

other hand, PET has good sensitivity but provides low spatial resolution. In contrast, US has high

spatial resolution but relatively low penetration and sensitivity, whereas optical imaging also has

good sensitivity but suffers from low tissue penetration. Since modalities with the highest

sensitivity have relatively poor resolution, while those with high resolution have poor sensitivity,

there is no single modality that can provide overall structural and functional information.

Therefore, it is often through the combination of multiple modalities that complementary

information is obtained [16].

The idea of combining multiple imaging modalities moved to the mainstream with the

successful development of commercial fused instruments. PET/CT [17, 101] and SPECT/CT

[102] fused systems have been successfully developed and widely adopted in the clinic, and the

PET/MRI system was recently introduced [103, 104]. With hybrid technology clearly on the rise,

there has been significant progress made toward the development of multimodal imaging probes

as researchers and investigators look to increase the clinical benefits of such hybrid instrument

technology [105-110].Ideally, the combined imaging modalities and imaging probes work

synergistically to provide high-resolution and high-sensitivity. For example, with dual function

probes for MRI/optical [111, 112], both MRI and whole body optical imaging can be used to

assess the probe distribution in the body. Although MRI provides more biodistribution

information at the whole body level than whole body optical imaging, the presence of a

fluorophore allows confirmation of probe labeling in subsequent histology or in vivo fluorescent

Page 25: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

11

endoscopy. Moreover, such a system could be translated into clinical settings, where MR

imaging is used for preoperative scanning to achieve disease localization, identification and to

guide the surgical procedure, while optical imaging may be used for intraoperative target

delineation.

2.2.1 CT Imaging

CT is widely used in clinical settings for diagnosis because radiography has the ability to go

through the body and provide external visualization of the internal anatomical structures [113].

In oncology, CT is used to detect or confirm the presence of a tumour, to help plan therapy or

surgery, and to assist in monitoring treatment response [114-116]. Contrast in CT is generated by

the differential attenuation of the X-ray beam in neighbouring tissues or materials. This

attenuation is dependent on the density of the tissue and is represented by the Hounsfield unit

(HU). Conventional contrast agents for CT include iodine and barium, which have high atomic

numbers and provide high X-ray attenuation [24].

Volumetric CT imaging allows for extremely fast data acquisition (a few seconds) in

submillimeter isotropic voxels, providing high spatial and temporal resolution. Moreover, CT

provides a linear response to increasing concentrations of contrast agent without being affected

by environment [34]. When combined with 3D image analysis tools, not only is the volumetric

quantification of an organ or tissue possible, but the amount of contrast agent distributed within

the organ or tissue can also be quantified. This makes CT very attractive for evaluation of

biodistribution and pharmacokinetics of contrast agents.

Micro-CT imaging has gained popularity in preclinical CT imaging involving small

animals such as mice, rats and rabbits. Over the past decade, the number of publications using

micro-CT imaging in preclinical in vivo studies has increased exponentially [117]. Micro-CT is

equipped with higher spatial and temporal resolution and allows researchers to capture

increasingly detailed anatomical images of small animals and to monitor the progression of

disease in small animal models. The first applications of micro-CT were to evaluate bone

anatomy and density [118, 119]. In the 1990s, micro-CT was further applied to study the

vasculature of small animals such as angiogenesis [120, 121] and neovascularization [122]. To

Page 26: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

12

date, micro-CT has been popular in cardiothoracic imaging [123] and imaging of various organs

of small animals [124-126].

2.2.2 Optical Imaging

Optical imaging involves the detection of light photons transmitted through tissues. It utilizes

bioluminescent and fluorescent endogenous reporters or exogenous probes to monitor molecular

and biological processes [127]. Exogenous fluorescent probes have been developed in the near-

infrared (NIR) region (700–1000 nm) where optical imaging is optimal due to minimal tissue

absorption and low autofluorescence at these wavelengths. These probes offer a number of

advantages over radionuclides in that they are relatively inexpensive, have straightforward

synthetic and conjugation chemistries, and emit detectable, non-ionizing photons on excitation

[128]. Indocyanine green (ICG) is one such NIR probe that has been approved by the FDA and

used extensively for monitoring hepatic function, angiography, and cardiac physiology [129,

130]. Cy5.5 is another NIR fluorophore in the cyanine dye family that has been widely used in

studies to label targeting moieties [131, 132] or form activatable in vivo fluorescent probes that

utilize the concept of FRET to quench probe fluorescence upon protease cleavage [130, 133].

Studies involving fluorescent compounds include gene expression profiling, elucidation

of cellular pathways, and protein function determination [134, 135]. Currently, there is a number

of high-resolution microscopic fluorescence imaging techniques developed to study molecular

events in vivo. In particular, intravital fluorescence microscopy [136-138], confocal laser

scanning microscopy [139, 140], multi-photon laser scanning microscopy [141], and in situ

scanning force microscopy[142] have been introduced within the past years. Such techniques are

preferred for visualization at the cellular or intracellular levels and generally operate with small,

two-dimensional fields of view (~1mm2) [143].

Macroscopic fluorescence imaging systems employ photographic principles to collect

images in low light and enable whole-body imaging of small animals. Fluorescence reflectance

and tomographic fluorescence are the two main types of macroscopic imaging. Fluorescence

reflectance imaging (FRI) systems consist of an excitation source, filters, and a charge-coupled-

device (CCD) camera to obtain two-dimensional images. This is useful for imaging of surface

Page 27: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

13

events, such as xenograft tumours and surgically exposed organs, and for intra-operative use

[144]. However, FRI has a limited depth penetration and is thus not inherently quantitative. On

the other hand, tomographic fluorescence systems are able to image the biodistribution of

fluorescence in small animals and tissues three-dimensionally and quantitatively, with a

penetration depth of up to several centimeters [133]. Fluorescence molecular tomography (FMT)

systems illuminate a tissue of interest over multiple angles and collect photons that have

propagated through tissue. Mathematical processing of the raw data yields three-dimensional

quantitative images of the fluorophore distribution in tissue [145]. Early FMT systems require

the use of index-matching fluids and the need to immerse mice or tissues in the fluid to simplify

the mathematics. Recent development has eliminated the need for the fluid [144]. While photon

scattering is the dominating limitation for fluorescent microscopy in terms of image depth, it also

reduces the image resolution for fluorescent microscopy. In FMT, photon scattering leads to

challenging image reconstruction problems and effectively limits the image resolution (1 mm - 1

cm) as well as the accuracy of quantification [143, 144]. The penetration depth of FMT is limited

by light attenuation in tissue, which depends on both tissue scattering and absorption. In muscle

or brain, the penetration depth is 3-6 cm, while in less absorbing organs such as the breast, the

penetration depth is 10-12 cm [143]. Therefore, macroscopic optical imaging can be used to

image small animals or certain tissues/organs of larger animals and humans, or used in

endoscopic and intraoperative applications

As with most optical imaging methods, FMT is highly sensitive and is able to detect

fluorophores present in the picomolar concentration range [133]. A single FMT scan usually

takes 3-5 minutes. FMT has been used in studies evaluating protease activity [133], angiogenesis

and the effects of chemotherapy on tumours [146]. In the latter study [146], Cy5.5 was

conjugated to annexin V, and the complex was used to image Lewis lung carcinomas in a mouse

model. Based on the FMT fluorescence intensity, an apoptotic index ratio was calculated and

found to be in agreement with results found using ex vivo staining of excised tumour tissues.

This demonstrates the capability of FMT to noninvasively assess the tumour accumlation of the

fluorescent probe and tumour response to chemotherapy. In addition, the multi-channel feature

allows simultaneous imaging of fluorophores emitting at different wavelengths [147, 148].

Overall, FMT’s high sensitivity, operational safety, and short image acquisition times makes it

an attractive choice for bench-top quantitative in vivo optical imaging.

Page 28: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

14

2.2.3 CT/Optical Multimodal Imaging

The lack of a commercial multimodal scanner to incorporate whole body optical imaging makes

multimodal imaging more difficult as simultaneous imaging in the distinct modalities is not

possible. This makes image registration across modalities more difficult, especially at early time

points.. Therefore, to achieve near-simultaneous imaging across modalities, the scanners for each

modality must be in close proximity, and the scanning time for each modality must be fast. In

this case, CT imaging is the best candidate to combine with optical imaging as CT imaging

provides both high resolution and good anatomical reference, while also offering fast scanning

time (a few seconds for micro-CT). FMT as an optical imaging modality offers scanning time in

the range of 3-5 minutes, which makes near simultaneous multimodal CT/optical imaging

feasible. However, it can be argued that at early timepoints (i.e. less than 1 hr after

administration of contrast agents), image co-registration creates a larger relative temporal error.

But as the study period is prolonged (i.e. scanning a few hours post administration), this error

becomes negligible.

As mentioned before, owing to intrinsic fluorescent properties and photon scattering, the

reconstructed FMT images have a low spatial resolution (1mm) and poor anatomical reference.

For these reasons, CT [149-151] and MR [25, 26] are being used increasingly in conjunction

with FMT to provide the necessary anatomic information. Moreover, the quantitative nature of

CT enables the assessment of pharmacokinetics and biodistribution of contrast agents in vivo

[24], and can be used as a tool to evaluate the true extent that FMT can be used for quantitative

imaging. On the other hand, FMT and microscopic fluorescence imaging offers high sensitivity

and the latter enables the monitoring of the contrast agents at a microscopic level. Therefore,

combining CT and FMT should provide confirmation of the consistent transportation of the

contrast agents in vivo, as well as provide a quantitative monitoring of whole-body distribution

of the probes, and enable monitoring of the probes in a localized region. Currently, efforts are

underway to improve multimodal imaging involving optical imaging, and hybrid imaging

systems using MR or CT with FMT have already been proposed (28-32).

Page 29: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

15

1.4 Image-based Assessment of Nanoparticle Distribution

The characterization of the pharmacokinetics and in vivo distribution of novel imaging and

therapeutic agents is crucial for understanding their in vivo performance and effectiveness. The

employment of imaging techniques to assess the in vivo distribution of the nanoparticles

provides unique insight into the in vivo pathway and fate of the nanoparticles. The noninvasive

nature of image-based assessments enables repeated in vivo data acquisition from the same

subject over multiple time points. This increases the accuracy of measurements. In addition, if

the imaging technique has high resolution, the intra-organ and tissue distribution of the agent can

be resolved. In cancer research, the ability to noninvasively quantify the nanoparticles

accumulated at tumor sites and to noninvasively map their intratumoral distribution with respect

to functional and physiological parameters of the tumor microenvironment provides the potential

to assist in the development of effective therapies.

Nanoparticle formulations are designed to improve the biodistribution and the target site

accumulation of systematically administered therapeutic agents. Many drug loaded nanoparticles

have been labeled with contrast agents in order to assess their in vivo pharmacokinetics and

biodistribution using imaging. In 2001, a preliminary clinical study evaluated the biodistribution

and pharmacokinetics of 111In-DTPA-liposomes in 17 patients with a wide range of cancers (i.e.

breast, head and neck, bronchus, glioma, cervix) [40]. Scintigraphy was used to visualize the

biodistribution of the liposomes. SPECT scans were used to help better identify the tumour.

Strong blood pool images were obtained 30 min post injection. Normal organ uptake was seen

prominently in the RES of the spleen and liver. The clearance half life of the liposomes was

determined to be 76.1 h, though it varied from 40 to 100 h from patient to patient. Overall,

through imaging, the tumour was seen in 15 out of the 17 patients. Clear visualization of the

tumours was obtained only at 48-72 h after injection due to the high blood background at earlier

time points. It was also found that there was considerable heterogeneity in the uptake of the

liposomes both between different tumor types and between different patients with the same

tumor type. For example, the levels of liposome uptake seen in the breast tumors (5.3 ± 2.6%

ID/kg) were considerably lower than those seen in the lung (18.3 ± 5.7% ID/kg) and head and

neck tumours (33.0 ± 15.8% ID/kg). In patients with head and neck tumours, the liposome

uptake in tumour varied from less than 10% ID/kg to over 50% ID/kg. This study highlights the

importance of image-guided therapy in identifying whether a patient is able to benefit from the

Page 30: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

16

treatment. In another study, γ-scintigraphy was used to image the distribution galactosamine-

modified liver-targeted pHPMA-GFLG-doxorubicin labeled with 123I [152]. Through imaging, it

was shown that this targeted formulation effectively localized in the liver, while a comparable

formulation lacking galactosamine failed to show liver localization. Image superimposition of

the anatomical CT and functional SPECT images revealed that the majority of the targeted

formulation was associated with areas of normal liver (16.9 ± 3.9% ID, determined through

radioactivity distribution) with low accumulation in areas of hepatic tumour tissue (3.3 ± 5.6%

ID) This study highlights the importance of monitoring drug distribution, and also exemplifies

the advantages of multimodal imaging.

Visualizing drug distribution at the tumour site has been studied using gadolinium and

fluorescently labeled liposomes [153]. In this research, MRI and fluorescence microscopy were

used to monitor the intratumoural distribution of RGD-modified endothelial cell-targeted

liposomes and RAD-modified control liposomes. MRI was used for localization of the liposomes

at the whole body level, and fluorescence microscopy was used for liposome localization at the

sub-cellular level. Imaging results showed that liposomes targeted to endothelial cells

accumulated preferentially in the angiogenic rim of subcutaneously transplanted B16 tumors,

whereas control liposomes distributed non-specifically throughout the tumors. Zheng et al [24]

used quantitative CT imaging to assess the in vivo distribution of a combined liposome-based CT

and MR imaging agent This study reported the bulk organ/tissue (liver, kidneys, spleen, tumor

and blood) and intratumoral distribution of liposomes containing iohexol and gadoteridol over a

14-day period in VX2 sarcoma-bearing New Zealand White rabbits using CT imaging. It was

demonstrated that through quantitative CT based assessment, the pharmacokinetics and

biodistribution profiles of the agent were determined. It was concluded that such noninvasive,

quantitative image-guided pharmacokinetics and biodistribution assessments in the development

and preclinical testing of novel nanocarriers has the potential to greatly facilitate their clinical

translation. A later study [23] evaluated the performance of a similar CT liposome formulation to

detect tumor and inflammatory lesions in a rabbit model relative to 18F-fluorodeoxyglucose PET

(FDG-PET). This study showed that CT imaging was able to detect the differential liposome

accumulation at sites of tumor and inflammation. In addition, the use of the liposome increased

the CT contrast of neoplastic and inflammatory lesions, which was able to detect more soft-tissue

inflammatory lesions in comparison to FDG-PET.

Page 31: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

17

Another important application of image-guided drug delivery is the potential to use

imaging to visualize drug release from the nanoparticles. As the vast majority of therapeutic

agents used in nanotechnology are inactive when conjugated to or entrapped in the delivery

system, it is important to ensure that the agents are actually being released. Conventional

methods to determine drug release require the harvesting and homogenization of target organ or

tissue, which has the problem of destabilizing carrier material as well as, leading to difficulty in

discrimination between released and retained drug. The employment of imaging can overcome

this problem and enables the noninvasive analyses of in vivo drug release through the simple co-

incorporation of the drugs and contrast agents in one delivery vehicle. MR contrast agents such

as gadolinium and manganese are highly suitable for monitoring drug release as they depend on

the interaction with surrounding water molecules to generate a signal, which varies substantially

when these agents are present within versus outside of water-impermeable vesicles such as

liposomes. In an interesting study, manganese sulfate (MnSO4) was used both to load

doxorubicin into liposomes and to generate a significant increase in MR signal upon drug and

contrast agent release [42]. This temperature sensitive liposome formulation (TSL) was shown to

have comparable relaxivity to non-temperature sensitive liposomes (NTSL) below its transition

temperature, but its relaxivity substantially increased upon heating to temperatures exceeding the

transition temperature, indicating release of Mn2+ from the liposomes [42]. A follow up study

[154] showed through MRI determineation, that the contrast agent release correlated well with

doxorubicin release, which was analyzed invasively using HPLC and fluorescence.

Employment of multimodal imaging to provide complementary information on the

distribution of nanoparticles has received increased interest among researchers. In one study,

liposomes were produced carrying rhodamine-labeled lipid on the surface and encapsulated

Ferridex particles and fluorescently labeled siRNA [155]. In this liposome formulation, Ferridex,

an MRI contrast agent, and siRNA were encapsulated during liposome formation, while 0.2%

rhodamine B-DOPE, a fluorescent dye conjugated to a phospholipid, were mixed together to

form a part of the lipid bilayer. This formulation served as a method to deliver therapeutic

siRNA to silence COX-2, a key enzyme in the inflammatory pathway that is upregulated in

several cancers. MRI was used to image the siRNA delivery to tumors in mouse models, while

ex vivo fluorescence microscopy was performed in histological samples to confirm the MRI

results and determine the siRNA delivery into cells [155]. MR/optical liposomes have also been

Page 32: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

18

prepared through initial chelation of MRI contrast agents such as Gd with a chelator such as

DTPA or DOTA [156, 157]. For example, liposomes containing Gd-lipid and rhodamine-DOPE

were formed by sonication and studied in a T47D breast cancer mouse xenograft model [112].

The T47D cells were incubated along with the liposomes prior to inoculation. T1-weighted MRI

and whole body optical imaging (Maestro) were performed seven days after inoculation, and

clear contrast enhancement in the tumour was observed. Tumour sections showed fluorescence

consistent with rhodamine localization, confirming the in vivo imaging results. In another study

[158], 99mTc-chelates and Gd-DTPA were incorporated into perfluorocarbon-containing

liposomes. This formulation produced liposomes of 270 nm in size, and was used to target αvβ3

integrin in a tumour model. Three imaging modalities were used to monitor the liposomes, with

MRI to assess the extent of vascularization, CT for soft tissue contrast, and SPECT for probe

sensitivity detection.

Page 33: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

19

CHAPTER 2 Comparison of CT and Optical Image-based Assessment of

Liposome Distribution

This chapter has been submitted for publication in Molecular Imaging as “Comparison of CT and

Optical Image-based Assessment of Liposome Distribution” Huang H, Dunne M, Lo J, Jaffray

DA, Allen C.

Contribution statement by the author:

In this thesis, the following studies and analyses have been performed by Huang Huang:

1. Synthesis and characterization of Cy5.5-DSPE (section 2.2.1), excluding Mass Spectrometry.

2. Preparation and characterization of CT/optical liposomes (section 2.2.2 – 2.2.4)

3. In vivo CT/optical imaging (section 2.2.5), excluding the tumour cell preparation and

inoculation, as well as the IV injection.

4. Image and data analyses (section 2.2.6).

Page 34: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

20

2.1 Introduction

Image-based assessment of the in vivo pathway and fate of nanoparticles has received

widespread interest [92, 159-162]. This is made possible by labeling the nanoparticles with one

or more contrast agents or probes which enable visualization and measurement of the spatio-

temporal distribution of the nanoparticles in vivo using single or combined imaging modalities.

Imaging modalities that are commonly used in clinical and pre-clinical settings include computed

tomography (CT), optical, magnetic resonance (MR), positron emission tomography (PET),

single photon emission computed tomography (SPECT) and ultrasound (US) [18, 163-167].

Each of these modalities has inherent strengths and weaknesses as they differ in terms of

sensitivity, resolution, speed, and/or cost [16]. For example, optical and radionuclide imaging

(i.e. PET and SPECT) are associated with high sensitivity, however, the former presents low

tissue penetration while the latter is limited by relatively poor spatial resolution. MR and CT can

provide detailed anatomic information but are constrained by their relatively low sensitivity. US

provides high spatial resolution but it has limited field of view, and its performance is highly

user dependent. Furthermore, its application is greatly limited by its inability to access biological

targets enclosed by bony structures or air. Multimodal imaging combines the strengths of

different modalities to provide complementary information on structural, functional, and

molecular processes in vivo. Efforts to combine modalities have led to the successful

development of integrated PET/CT and SPECT/CT scanners, which have been widely adopted in

clinical settings for the detection and diagnosis of cancer [19, 168]. Prototype PET/MR systems

have also recently been developed and successfully implemented for both small animal and

human imaging [169, 170]. Furthermore, images acquired on different scanners can be registered

together to provide more complete and accurate information [1].

Multimodal imaging involving optical imaging has been studied in the past [25, 26, 149-

151, 171, 172]. The development of fluorescence molecular tomography (FMT) in optical

imaging provides the capability to three-dimensionally image the biodistribution of fluorophores

in small animals and tissues with an imaging depth of several centimeters [133, 144, 173, 174].

Hybrid imaging systems using MR or CT with FMT have also been proposed [175-179]. Owing

to intrinsic fluorescent properties and photon scattering, reconstructed FMT images present

relatively low spatial resolution (1 mm). This in turn leads to questions regarding the accuracy

Page 35: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

21

associated with the quantification of accumulation of fluorophores in localized tissues such as

tumour using FMT. Therefore, comparison and co-registration of the quantitative data extracted

from the images in FMT with other modalities is of interest yet remains challenging.

Liposomes are the most established of the advanced drug delivery technologies and can

easily be labeled or loaded with probes, contrast agents, or radionuclides to support imaging in

single or multiple modalities [34, 37, 153, 155, 156, 180, 181]. Labeled liposomes have been

used to self-report on their fate in vivo. Image-based assessment of the distribution of drug

delivery systems can provide unique information that is otherwise unavailable using

conventional techniques [13, 182]. Such labeled delivery systems or formulations may be

employed in the design and implementation of personalized medicine. Numerous studies have

reported on the in vivo distribution of liposomes as determined using nuclear imaging techniques

[40, 183]. However, the imaging time window in PET and SPECT is limited by radioactive

decay of the radioisotopes, and thus is not ideal for long study periods [24]. Alternatively, CT

contrast agents such as iodine and barium provide strong and persistent X-ray attenuation, which

makes CT imaging an attractive tool for monitoring the fate of long-circulating nanoparticles

over prolonged periods of time. In addition, CT imaging allows for fast 3D data acquisition with

sub-millimeter resolution. This then permits sequential imaging with other fast 3D imaging tools

such as fluorescence molecular tomography (FMT), and consequently, increases the temporal

resolution of data obtained across multiple imaging modalities. Previous studies in our laboratory

have employed CT imaging to quantitatively assess the distribution of liposomes containing the

iodinated agent iohexol over prolonged periods of time in mouse and rabbit models [24, 184].

The current study extends the use of a liposome formulation to support optical imaging (Figure

2.1) and compares the information that can be gained on the macro- and microscopic distribution

of liposomes in vivo using micro-CT, FMT and confocal laser-scanning fluorescence

microscopy.

Page 36: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

22

Figure 2.1 Schematic of the CT/optical liposome-based contrast agent (not drawn to scale).

The average hydrodynamic diameter of the liposomes was found to be 86 ± 5 nm by

dynamic light scattering (DLS).

Page 37: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

23

2.2 Materials and Methods

Anhydrous dimethyl sulfoxide (DMSO), anhydrous chloroform, triethylamine (TEA), C8

reverse-phase silica gel, and bovine serum albumin (BSA) were purchased from Sigma-Aldrich

Canada (Oakville, Canada). 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC, MW 734) and

1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-poly(ethylene glycol) 2000 (DSPE-

PEG2000, MW 2774) were purchased from Genzyme Pharmaceuticals (Cambridge, USA). 1,2-

distearoyl-sn-glycero-3-phosphoethanolamine (DSPE, MW 748.09) and cholesterol (CH, MW

387) were obtained from Avanti Lipids Inc. (Alabaster, USA). Omnipaque™, a commercially

available iodinated CT contrast agent that contains iohexol (M.W. 821.14) at 300 mg/mL iodine,

was purchased from GE Healthcare (New Jersey, USA). Cy5.5 mono-NHS ester (Cy5.5-NHS,

molar extinction coefficient 250,000 M-1cm-1, MW 1128.4) was obtained from Amersham

Sciences (Buckinghamshire, UK). NCI-H520 human non-small cell lung cancer cells and RPMI

1640 medium (2 mM L-glutamine, 10 mM HEPES, 1 mM sodium pyruvate) were supplied by

ATCC (Manassas, USA). Female nude athymic CD-1 mice were provided by Charles River

(Wilmington, USA). 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) was

purchased from BioShop Canada (Burlington, Canada).

2.2.1 Synthesis of Cy5.5-DSPE

The synthesis of Cy5.5-DSPE was modified from previously published studies [185, 186]. In this

method, DSPE dissolved in anhydrous chloroform mixed with TEA was added to Cy5.5-NHS

dissolved in anhydrous DMSO at a DSPE:Cy5.5 molar ratio of 1:1. The solution was stirred at

22ºC for 18 h with protection from light and was then dried and re-dissolved in methanol.

Unreacted reagents were removed by reverse phase chromatography using a column packed with

C8 reverse-phase silica gel. Deionized water, methanol and methanol/chloroform (80/20, % v/v)

were used to elute Cy5.5, Cy5.5-DSPE, and DSPE, respectively. The Cy5.5-DSPE product was

confirmed using electrospray mass spectroscopy (QStarXL mass spectrometer with electrospray

(ESI) source (MDS Sciex, Concord, Canada)) with methanol/H2O (50/50, % v/v) as solvent. The

Page 38: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

24

purity of Cy5.5-DSPE was determined by ultra performance liquid chromatography (UPLC)

separation of Cy5.5-DSPE and Cy5.5 on a 10 cm BEH C18 column (Waters, Milford, USA). A

constant flow rate of 0.1 mL/min was employed with a gradient mobile phase of 50/50 (% v/v)

0.05M Triethylammonium acetate (TEAA) buffer (pH 7.0)/methanol to 100% methanol in 6

minutes. Both compounds were detected using a fluorescence detector (Waters, Milford, USA).

The yield of Cy5.5-DSPE was measured by ultraviolet (UV) absorbance at a wavelength of 674

nm using a Cary 50 UV-visible spectrophotometer (Varian, Palo Alto, USA).

2.2.2 Liposome Preparation

The method used for liposome preparation was adapted from a previously published report [34].

Briefly, lipid components for the CT/optical liposome formulation (i.e. DPPC, CH, DSPE-

PEG2000, and Cy5.5-DSPE) were dissolved in anhydrous ethanol at 70 °C at a molar ratio of

54.98:40:5:0.02 DPPC:CH:DSPE-PEG2000:Cy5.5-DSPE. OmnipaqueTM (300 mg/mL iodine) was

added to the solution with a lipid concentration of 100mM following ethanol removal. The

solution was kept at 70 °C for 4 h with intermittent vortexing. Unilamellar vesicles were formed

via extrusion at 70 °C using a 10-mL Lipex Extruder (Northern Lipids Inc, Vancouver, Canada)

with five passages through two stacked 200 nm pore size Track-Etch polycarbonate membranes

(Whatman Inc., Clifton, NJ) followed by ten passages through two stacked 80 nm membranes.

The unincorporated contrast agents were removed by 18 h of dialysis (MWCO 8kDa) against a

250-fold volume excess of 0.02mM HEPES-buffered saline solution (HBS, pH 7.4). The

liposome formulation was then concentrated to a final iodine concentration of approximately 40

mg/mL, with a final Cy5.5 concentration ranging between 35 μg/mL and 45 μg/mL. Liposomes

without Cy5.5-DSPE (CT liposomes) were also made using the same method with a molar lipid

ratio of 55:40:5 DPPC, CH, DSPE-PEG2000.

2.2.3 Liposome Characterization

To evaluate their hydrodynamic diameter and zeta potential, the liposomes were diluted to a lipid

concentration of 2.5 mM in deionized water. The hydrodynamic diameter was measured by

Page 39: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

25

dynamic light scattering (DLS) at an angle of 90 degrees and a temperature of 25 °C using a

90Plus particle size analyzer (Brookhaven, Holtsville, NY). Zeta potential was measured by

photon correlation spectroscopy (PCS) using a ZetaPALS zeta potential analyzer (Brookhaven,

Holtsville, NY). The average of 10 runs consisting of 25 measurement cycles per run was

reported. For iodine and Cy5.5 concentration measurements, the liposomes were ruptured with a

10-fold volume excess of ethanol and diluted with HBS. The iodine concentration was

determined by measuring UV absorbance at a wavelength of 245 nm, and that of Cy5.5 was

determined by measuring UV absorbance at a wavelength of 674 nm. The loading efficiencies

for both iohexol and Cy5.5 were calculated using the following equation:

Agent loading efficiency (%) = (Amount of agent in final product) / (Amount of agent added

during preparation) x 100

2.2.4 In vitro Stability and Agent Release

Liposome stability was evaluated by monitoring their hydrodynamic diameter by DLS analysis

over a 14-day incubation period. Specifically, the liposomes were placed in a dialysis bag

(MWCO 8 kDa) and immersed in 250-fold volume excess of HBS containing 45 mg/mL BSA.

The sample was incubated at 37 °C. Over the course of 14 days, aliquots of the liposome solution

were taken from the dialysis bag and their hydrodynamic diameter was measured using DLS.

The release of contrast agents from the liposomes was also evaluated. Briefly, liposomes were

mixed with FBS such that the final lipid concentration was 6.5 mM and incubated at 37 °C. At 0,

2, 6, 24, 48, 72, and 96 h post-incubation, aliquots were collected and a Sepharose CL-4B

column was used to separate the liposomes from the free agents and protein. The liposome

fraction was collected, and the encapsulated amounts of iohexol and Cy5.5 were determined by

measuring the UV absorbance at 245 nm and 674 nm, respectively.

Page 40: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

26

2.2.5 In vivo CT/Optical Imaging

The in vivo imaging studies were performed under protocols approved by the University Health

Network Animal Care and Use Committee. Athymic female CD-1 mice (6 weeks, avg. wt. 27 g)

were injected via the tail vein with a liposome formulation (CT or CT/optical) such that each

animal received 1.8 g lipid/kg body weight, 0.35 g iodine/kg body weight, and for the CT/optical

liposomes, 0.3 mg Cy5.5/kg body weight. The animals were anaesthetized using 2% isoflurane

and full body 16-second anatomical micro-CT scans (GE Locus Ultra micro-CT, GE Healthcare,

Waunakee, WI) were performed pre-injection and 0.083, 1, 4, 8, 24, 48, 72, 150, and 168 h post-

injection. CT images were acquired at 80 kVp and 70 mA with a voxel size of 0.15 mm x 0.15

mm x 0.15 mm and a field of view of 15.7 cm (transaxial) and 10.2 cm (long). Immediately after

each CT scan, the animal was fixed in the scanning chamber of the FMT system (VisEn Medical

Inc., Bedford, MA). A reflectance image of each animal was obtained and the chamber was filled

with the index matching fluid (VisEn Medical Inc., Bedford, MA) up to the animal’s upper chest.

The scanning window was adjusted to its maximum size. Each animal was scanned at excitation

and emission wavelengths of 670 nm and 700 nm, respectively. FMT images were acquired with

a nominal voxel size of 1 mm x 1 mmx 0.5 mm and a tomographic nominal field of view of 3.45

cm x 3.45 cm.

The same protocol was used for imaging tumour-bearing animals. H520 NSCLC cells

were cultured in RPMI 1640 medium supplemented with 10%vol FBS and 1%vol penicillin-

streptomycin in an atmosphere containing 5% CO2. Cells were inspected for consistent

morphology and growth rate and were passaged no more than 10 times prior to mouse

inoculation. Athymic female CD-1 mice (4 weeks, avg. wt. 23 g) were inoculated

subcutaneously with 5 x 106 H520 cells in the right hind flank. The CT/optical liposomes (0.35

g/kg iodine, 0.3 mg/kg Cy5.5, and 1.8 g/kg lipid) were injected into the animals 9 days after cell

inoculation. CT scans were performed pre-injection and 0.083, 24, 50, 73, 97, 122, 146, 169, and

193 h post-injection. Immediately after each CT scan, an FMT scan was performed, with the

scanning window adjusted to surround the right hind flank. At 24 h and 193 h post-injection, one

animal was scanned using the confocal laser-scanning fluorescence microscope (Leica

Microsystems Inc., Bannockburn, IL). Specifically, the skin above the tumour was removed, and

the microscopic probe (excitation and emission wavelengths of 650 nm and 700 nm,

Page 41: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

27

respectively) was stabilized at various locations inside and on the surface of the tumour. Real-

time confocal videos of the regions were recorded.

2.2.6 Image Analysis

CT images were exported via DICOM and analyzed using MicroView v2.2 (GE Healthcare,

Waunakee, WI) according to a reported method [24]. Briefly, the mean iodine concentrations in

various organs were determined by contouring the region of interest (ROI) on multiple 2D slices

to generate a 3D volume, and calculating the mean voxel intensity (expressed in CT attenuation

Hounsfield Units, or HU) for each volume. For the non-tumour bearing animals, iodine

concentrations in blood (aorta), liver, spleen, and kidneys were measured. For the tumour-

bearing animals, iodine concentrations in blood, tumour, and muscle surrounding the tumour

were measured. Signal enhancement for each tissue was calculated as the difference between the

mean voxel intensity at each time point post-injection and the mean voxel intensity pre-injection.

For the tumour, the corrected mean voxel intensity was determined by subtracting the muscle

mean voxel intensity from the tumour mean voxel intensity. The intensity values were then

converted to iodine concentrations using a CT calibration curve of HU versus iodine

concentration, and the results were expressed as percent injected dose per volume tissue

(%ID/cm3). Pharmacokinetic parameters were calculated by fitting the blood iodine

concentration profiles with a one-compartment model using Scientist®3.0 (Micromath®, St.

Louis, MO). Initial parameter values were estimated through curve stripping of the blood iodine

concentration versus time plot. The fitting model used a mono-exponential decay function (C =

C0e-Ke*t, where Ke is the elimination constant) and method of weighted least squares with

weightings of 0, 1, and 2. The best fit was chosen based on highest coefficient of determination,

model selection criterion, and lowest coefficient of variation. All best fits possessed an R2 >

0.98. The vascular half-life was calculated as t1/2 = ln(2)/Ke. The area under the blood-iodine

concentration versus time curve (AUC) was calculated using the trapezoidal rule. Volume of

distribution (VD) was calculated as VD = Dose/C0. Clearance (Cl) was calculated as Cl =

Dose/AUC.

Page 42: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

28

FMT images for non-tumour bearing animals were not analyzed due to the lack of

defined tissue boundaries, which are needed for accurate delineation of the organ and tissue

structures (Figure 4). The FMT data for the tumour-bearing animals were analyzed for the

tumour, where tissue boundaries were better defined due to isolation of the tumour from highly

perfused tissues. The mean Cy5.5 concentration in the tumour was determined using the FMT

analysis software (VisEn Medical Inc., Bedford, MA). Briefly, a 3D contour was initially laid

out to include the tumour region. An intensity threshold was adjusted to exclude zero or low

fluorescent voxels in the contour until the contour volume matched the tumour volume

determined by micro-CT analysis. A contour was also applied to the muscle region surrounding

the tumour to obtain the background signal. The corrected mean voxel intensity was determined

by subtracting the muscle mean voxel intensity from the tumour mean voxel intensity. The

corrected mean voxel intensity was converted to %ID/cm3 tumour using an FMT calibration

curve of fluorescence intensity versus Cy5.5 concentration.

The iohexol-to-Cy5.5 ratio (wt/wt) for the tumour was calculated for all time points by

dividing the concentration of iohexol (quantified by micro-CT) by the concentration of Cy5.5

(quantified by FMT) at the tumour region and normalizing to the iohexol-to-Cy5.5 ratio prior to

in vivo administration.

Results obtained by micro-CT image-based quantification of liposome accumulation in

tumour were compared to those obtained by FMT image analysis. Data selected for comparison

were between 24 to 72 h post-injection of the formulation wherein the liposome accumulation in

the tumours was high. Within this timeframe, micro-CT defined tumour volumes ranged between

25 to 30 mm3. For FMT analysis, a 2 mm3 cubic VOI was placed at the centre of the tumour, the

mean Cy5.5 concentration of the VOI was quantified, and the minimum and maximum FMT

intensity of the ROI was recorded. The VOI was then increased, without changing its centre, to

4, 6, 9, 18, 24, 32, 50 and 80 mm3. For each VOI, the Cy5.5 concentration was quantified and the

minimum and maximum FMT intensity were recorded. All FMT VOIs remained within the

boundary of the FMT observed tumour. Similarly in micro-CT, cubic VOIs were generated to

match the VOI volumes of FMT, and the mean iodine concentration was measured for each VOI.

For FMT VOIs that were larger than the micro-CT defined tumour volume, the micro-CT VOIs

remained the same as the tumour size. The iohexol-to-Cy5.5 ratio was obtained for all VOIs. For

FMT, the percentage window threshold level for each VOI was calculated as follows:

Page 43: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

29

% FMT window threshold level = min. FMT intensity / max. FMT intensity x 100%

Page 44: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

30

2.3 Results

2.3.1 Synthesis and Characterization of Cy5.5-DSPE

The mass spectrum for Cy5.5-DSPE is shown in Figure 2.2. The theoretical molecular

weight of Cy5.5-DSPE (1646 Da) and the molecular weight determined by MS analysis was in

good agreement. Figure 2.3 shows the UPLC chromatograms demonstrating the separation of

Cy5.5 (~3.5 min) and the more hydrophobic Cy5.5-DSPE (~7.2 min). The purity of Cy5.5-DSPE

was determined to be 99.6 ± 0.3% and the yield of Cy5.5-DSPE was 15 ± 5%.

Figure 2.2 The mass spectrum of Cy5.5-DSPE. The measured

molecular weight of Cy5.5-DSPE was determined to be 1645.7

Da, which is in good agreement with the theoretical molecular

weight 1646 Da.

Page 45: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

31

Figure 2.3 Representative UPLC chromatograms for Cy5.5 and

purified Cy5.5-DSPE. Cy5.5 elutes at ~3.5 min, Cy5.5-DSPE

elutes at ~7.2 min. The distinction between the two sets of peaks

assures the purity achieved using the column chromatography

method.

2.3.2 Physicochemical Characterization of the Liposomes

The physicochemical properties of the CT and CT/optical liposomes are summarized in

Table 2.1. Following concentration of the liposome formulation to ~40mg/mL iodine (35 –

45μg/mL Cy5.5), the agent-to-lipid ratio (w/w) was ~0.53:1 for iohexol, and ~1.4x10-4:1 for

Cy5.5. DLS analysis of the CT/optical liposomes revealed a narrow, unimodal size distribution,

with the incorporation of Cy5.5 increasing the hydrodynamic diameter slightly. The zeta

potential of the CT/optical liposomes (-34 mV) was more negative than that of the CT liposomes

(-28 mV), which is attributed to the negatively-charged Cy5.5 molecules at the surface of the

vesicles [187, 188]. The CT/optical liposomes were shown to be stable with no change in their

size over the two week incubation period in the presence of BSA (Figure 2.4). Figure 2.5a shows

Page 46: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

32

the in vitro release of iohexol and Cy5.5 from the CT/optical liposomes and of only iohexol from

CT liposomes in FBS at 37°C. After 96 hours, ~15% iohexol was released from both CT and

CT/optical liposomes, while ~25% Cy5.5 was released from the CT/optical liposomes.

Therefore, the ratio of iohexol to Cy5.5 (w/w) in the CT/optical liposomes increased during the

course of the incubation period such that it was significantly higher (p < 0.05) than that in the

pre-incubation formulation at all time points except that of 2 h (Figure 2.5b).

Table 2.1 Physicochemical characteristics of CT and CT/optical liposomes (n=5). Data are represented

as the mean ± standard deviation

Liposome

formulation

Hydrodynami

c diameter

(nm)

Zeta

potential

(mV)

Number of

iohexol

molecules per

liposome*

Iohexol

loading

efficiency (%)

Number of

Cy5.5

molecules per

liposome*

Cy5.5 loading

efficiency (%)

CT/optical 86 ± 5 -34 ± 3** (3.9 ± 0.8) x 104 5.8 ± 0.4 12 ± 2 68 ± 8%

CT 82 ± 2 -28 ± 3** (3.6 ± 0.4) x 104 5.9 ± 0.5 N/A N/A

* based on (8.1 ± 0.9) x 104 lipids/liposome, 4.5nm bilayer thickness [189], and a homogeneous distribution of Iohexol and Cy5.5

** Indicates statistically significant differences with p < 0.05

Page 47: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

33

Figure 2.4 Hydrodynamic diameter (obtained by DLS analysis)

of the CT/optical liposomes over a 2-week period with dialysis

against a 250-fold volume excess of HBS containing 45mg/mL

BSA at 37°C. Data are represented as the mean ± standard

deviation of three independent batches of liposomes.

Page 48: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

34

Figure 2.5 (a) The in vitro release profile of iohexol and Cy5.5 from CT/optical liposomes (n=3) and

iohexol from CT liposomes (n=3) incubated in FBS at 37°C. (b) Ratio of iohexol to Cy5.5 (wt/wt) in

the CT/optical liposomes following incubation in FBS at 37°C. The initial iohexol-to-Cy5.5 ratio was

normalized to 1. * indicates statistically significant difference (p < 0.01) from t = 0.

Page 49: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

35

2.3.3 In vivo Pharmacokinetics, Biodistribution of the Liposomes

Figure 2.6 displays representative single coronal micro-CT and FMT image slices from a

non-tumour bearing mouse at various time points post-administration of the CT/optical

liposomes, illustrating the distribution of the liposomes in vivo. As shown, tissues and organs can

be distinguished in the CT images while FMT fails to enable individual tissues to be identified.

Each FMT slice contains a reflectance image of the animal and the fluorescence in the scanning

window (depicted in the square in each slice). Micro-CT and FMT image slices representing the

accumulation of liposomes in the tumour are shown in Figure 2.7. Both the transverse and

coronal CT images are displayed. The CT/optical liposomes possess a slightly increased in vivo

half-life (p < 0.05) compared to the CT liposomes in non-tumour-bearing mice (Figure 2.8a,

Table 2.2). The data obtained from analysis of biodistribution revealed a slight increase in

liposome accumulation in the liver and kidneys for the CT/optical liposomes at certain time

points, with statistically significant differences at 0.083 and 24 h post injection for the liver, and

48, 72, and 96 h post-injection for both kidneys (Figure 2.8b,d,e). Liposome accumulation in the

spleen was equivalent for both formulations (Figure 2.8c). The differential tumour-to-muscle

accumulation profile for the CT/optical liposomes over the eight-day study period is shown in

Figure 2.9b, which compares data obtained from both micro-CT and FMT.

As shown in Figure 2.9b, both analyses by micro-CT and FMT imaging reveal similar

tumour accumulation profiles with maximum liposome accumulation in tumour at 48 h post-

injection (i.e.~10 %ID iohexol/cm3 tumour and ~7 %ID Cy5.5/cm3 tumour). However, FMT-

based determination of tumor accumulation shows a higher %ID/cm3 tumour for the liposomes at

5 minutes post-injection in comparison to the accumulation observed by micro-CT. At all other

time points, micro-CT-based determination showed higher liposome accumulation than that

determined by FMT. These phenomena are partially attributed to the increased rate of release of

Cy5.5 in comparison to iohexol from the liposomes, which were demonstrated in vitro (Figure

2.5b). Figure 2.9c plots the in vivo iohexol-to-Cy5.5 ratio over the entire tumour volume wherein

the iohexol and Cy5.5 concentrations are determined by micro-CT and FMT analyses,

respectively (Figure 2.9b). Figure 2.9d shows the iohexol-to-Cy5.5 ratio over various concentric

ROIs from the observed tumour in both micro-CT and FMT. As shown, the ratio remained

constant when the ROIs were less or equal to the tumour volume, but increased drastically when

Page 50: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

36

the ROI was greater than the true tumour volume, demonstrating a sharp decrease in the Cy5.5

concentration. The lowest FMT threshold where the iohexol-to-Cy5.5 ratio remained constant

was determined to be 66 ± 7%.

Figure 2.10 shows the images taken using the confocal microscopic probe. As shown in

Figures 2.10 a and b, most liposomes still reside within the tumour vasculature at 24 h post-

injection. However, eight days post-injection (Fig. 2.10 c), liposomes can be seen scattered

within the tumour tissue, as opposed to the tumour vasculature.

Table 2.2 Pharmacokinetic parameters for CT and CT/optical liposomes (n=7) in non-tumour-

bearing mice and CT/optical liposomes (n=5) in tumour-bearing mice. Statistics were analyzed

between the CT and CT/optical liposomes in non-tumour-bearing mice, and between

CT/optical liposomes in non-tumour and tumour-bearing mice

Formulation t1/2

(hrs)

Vd

(mL)

Blood AUC

(mg·mL-1·hr)

CL

(mL·hr-1)

CT 29.2 ± 3.7** 2.1 ± 0.3 175 ± 29** 0.039 ± 0.003*

CT/optical 36.2 ± 2.1** 2.3 ± 0.3 244 ± 46* ** 0.055 ± 0.004*

CT/optical (tumour

bearing)

30.3 ± 8.9 2.0 ± 0.2 157 ± 50* 0.054 ± 0.015

* Indicates statistically significant differences with p < 0.05

**Indicates statistically significant differences with p < 0.01

Page 51: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

37

Figure 2.6 Micro-CT (top) and FMT (bottom) images of a non-tumour bearing mouse injected

with CT/optical liposomes. The same window and level were used for all images from each

modality.

Page 52: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

38

Figure 2.7 Micro-CT transverse (top), coronal (middle), and FMT images of a tumour bearing

mouse injected with CT/optical liposomes at various time points. The transverse micro-CT

images display the right side of the mouse. Arrow indicates the location of the tumour. The same

window and level was used for images from each modality.

Page 53: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

39

Page 54: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

40

Page 55: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

41

Figure 2.8 Pharmacokinetics and biodistribution profiles of CT and CT/optical liposomes in

non-tumour bearing mice as determined using micro-CT image-based assessment. (a) The blood

iodine concentration. The biodistribution in (b) liver, (c) spleen, (d) left kidney and (e) right

kidney. * indicates statistically significant differences (p<0.05) between CT and CT/optical

liposomes in %ID/cm3 tissue.

Page 56: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

42

Page 57: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

43

Page 58: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

44

Figure 2.9 (a) Pharmacokinetics of CT/optical liposomes in tumour bearing mice as determined

using micro-CT image-based assessment of the blood iodine concentration. (b) Micro-CT and

FMT image-based determination of tumor distribution profile for CT/optical liposomes (n=5). *

indicates a statistically significant difference between micro-CT and FMT data at that time point.

(c) Ratio of iohexol to Cy5.5 (wt/wt) at the tumour site. The initial iohexol-to-Cy5.5 ratio was

normalized to 1. * indicates statistically significant difference from pre-injection. (d) Iohexol-to-

Cy5.5 ratio (wt/wt) determined from various concentric cubic VOIs over the tumour region in

both FMT and micro-CT. Data include liposome accumulation in tumours 24 to 72 h post-

injection. The tumour volumes are 25 to 30 mm3. The ratios were normalized to the iohexol-to-

Cy5.5 ratio pre-injection.

Page 59: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

45

Figure 2.10 In vivo microscopic images of the CT/optical liposomes in the tumour area at 24 h

(a, b) and 8 days (c) post-injection.

Page 60: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

46

2.4 Discussion

Optical imaging enables both micro- and macroscopic monitoring of nanoparticles.

Intravital microscopy provides information with cellular or intracellular resolution within a small

imageable area (~1 mm2) [143], therefore offering localized information on nanoparticle

distribution. Conventional whole-body optical imaging techniques such as fluorescence

reflectance imaging (FRI) are more limited by penetration depth and provide planar images,

preventing reporting of underlying biological processes [190]. Conversely, fluorescence

molecular tomography (FMT) provides the capability to three-dimensionally and quantitatively

image the biodistribution of fluorophores in small animals and tissues with an imaging depth of

several centimeters [133, 144, 173]. However, owing to intrinsic fluorescent properties and

photon scattering, reconstructed FMT images present low spatial resolution (1mm) and poor

anatomical reference. For these reasons, CT [149-151] and MR [25, 26] are being used

increasingly in conjunction with FMT to deliver indispensable anatomic information.

In the current study, iohexol and Cy5.5 were co-incorporated in a liposome formulation

to enable CT and FMT imaging of the biodistribution and tumour accumulation of the

nanoparticles following administration. Iohexol was passively encapsulated within the liposomes

while Cy5.5 was conjugated to the bilayer surface via insertion of a phospholipid-Cy5.5

construct. The levels of the CT and optical agents incorporated in the CT/optical liposome

formulation were significantly different due to the distinct differences between the intrinsic

sensitivity and resolution of the micro-CT and FMT imaging modalities. Micro-CT requires an

iodine concentration of at least 0.5 mg/mL [191] for detection while FMT can detect fluorescent

probe concentrations in the low picomolar range (ng/mL of Cy5.5) [133]. Our CT/optical

liposomes contain ~40000 iohexol molecules and only ~12 Cy5.5 molecules (Table 1) per

liposome, constituting a 1000-fold difference in the dose of agents administered (i.e. 0.35g/kg

iodine compared to 0.3mg/kg Cy5.5). The unique capacity of the liposomes to incorporate such

significantly different amounts of CT and optical contrast agents allows a single dose of

liposomes to be administered with the desired contrast enhancement achieved in both CT and

optical imaging modalities.

Development of a liposome formulation that is stable in vitro (Figure 2.4) with minimal

release of the incorporated contrast agents (Figure 2.5a) is shown to translate into prolonged in

Page 61: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

47

vivo stability. This stability results in extended circulation lifetimes and provides prolonged

contrast enhancement in both the CT and FMT images (Figures 2.6–2.9). The tumour

accumulation profile (Figure 2.9b) clearly demonstrates the EPR effect, with a maximum

liposome accumulation in tumour at 48 h. This shows that prolonged circulation is important to

achieve the optimal contrast enhancement level in the tumour. The lower %ID/cm3 values from

FMT compared with that from micro-CT (Figure 2.9b) reveal that Cy5.5 is released at a more

rapid rate than iohexol in vivo, correlating well with the in vitro observations (Figure 2.5a).

Micro-CT has been shown to provide an accurate delineation of organs/tissues and a

quantitative measure of liposome distribution in vivo and is therefore useful in guiding the FMT

delineation of the tumour [24]. FMT detects the tumour through estimation of location from the

reflectance image of the animal and presence of the fluorescence intensity in that region (Figure

2.7). However, photon scattering leads to an intrinsic blur around the tumour, which increases

the observed tumour volume and makes delineation of the tumour difficult. The smaller the

actual tumour, the greater the relative increase in volume observed by FMT. In addition,

quantification error arises in FMT with inaccurate delineation (Figure 2.9d). Therefore, tumour

volumes obtained from micro-CT were employed as references to guide the accurate FMT

delineation of the tumour and subsequent measurement of liposome accumulation at the site.

Several FMT-based studies have used CT to guide FMT reconstruction (54-57). Specifically,

Garofalakis et al. (54) demonstrated that anatomical information provided by the CT data set can

be utilized during the FMT reconstruction process to improve its ability to accurately report the

concentration of fluorophores in vivo through application of geometric constrains. In the current

study, a different approach was employed where the CT data set was used as a guide for

delineation of the VOIs in the FMT data set. This method serves as a means to evaluate the

FMT’s consistency in quantification. As shown in Figure 2.9d, the iohexol-to-Cy5.5 ratio

remained constant (~1.72) only when the VOI was within 10% of the true tumour volume (as

determined through micro-CT). In order to reach the true tumour volume, the FMT VOI of the

tumour was reduced through increasing the FMT signal threshold level to exclude voxels with

lower fluorescence. The minimum FMT threshold that was needed to match the true tumour

volume as well as to keep the iohexol-to-Cy5.5 ratio constant was found to be 66 ± 7% of the

maximum intensity within the VOI. Similar adaptive thresholding methods have been proposed

and applied to PET imaging [192, 193]. In these studies, thresholds (% of the maximum

Page 62: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

48

intensity) were applied to PET images and ROIs were contoured and compared to those

determined by CT imaging. The PET threshold that gave rise to the smallest difference in VOI

volumes was identified as optimal. This threshold was reported to be around 40% [192] and has

been adopted in several clinical studies [194-196]. However, it was noted that the threshold

depends on the volume of interest [193] as well as the background signal [197] and therefore

should not be a fixed value. Indeed, in our study, the constant FMT threshold only holds for

tumours of a certain volume (25-30 mm3), and varied based on the micro-CT-defined tumour

volume. In addition, the presence of heterogeneous background fluorescence contributes to the

quantification error of FMT [198]. It has been reported that this error can be as large as 50%.

Nevertheless, subtraction of the background fluorescence prior to image reconstruction can lead

to improved image quality and quantification accuracy. In spite of this subtraction, quantification

error was still reported to be over 30% [198]. Such quantification error leads to differences in

micro-CT and FMT-based quantification results (Figure 2.9b). In fact, as shown in Figure 7b, the

background fluorescence greatly affected the FMT signal at early time points, most notably at 5

min post injection, where the quantified Cy5.5 concentration in the tumour is abnormally high

when compared with the micro-CT result. The extent to which background fluorescence can

affect FMT quantification is currently the goal of a separate study.

The main benefit provided by multimodal imaging is the acquisition of complementary

information. In our study, micro-CT imaging enabled the clear identification of anatomical

features (Figure 2.6) with good spatial resolution and permitted the quantitative and longitudinal

assessment of the pharmacokinetics and biodistribution of the liposomes in vivo. This

compensated for the inability of the FMT to distinguish between different organs (Figure 2.6)

due to poor image resolution and considerable photon scattering. These characteristics

effectively blur the boundaries of organs and result in severely limited information regarding the

intratumoral distribution of the liposomes (Figure 2.7). In contrast, as shown in Figure 2.7,

micro-CT provides information on the intratumoural heterogeneity associated with liposome

accumulation. Moreover, FMT can only provide a reflectance image of the animal prior to

scanning as a crude and planar estimation of its anatomical features, and therefore greatly relies

on micro-CT to provide an accurate delineation of organs or tissues. In this study, each animal

was scanned in different positions in micro-CT and FMT owing to scanning limitations. When

animals are scanned in the same position in the two modalities, the images from both micro-CT

Page 63: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

49

and FMT can be co-registered. In that case, micro-CT would not only complement FMT in

providing accurate volume information, but also offer the precise location of organs/tissues that

would otherwise only be estimated from FMT. Nonetheless, the greater sensitivity associated

with FMT, and optical imaging in general, not only enabled a very low fluorescent probe dose to

be administered, but also provided sufficient signal when only low levels of liposome were

present in the tumour. For example, the FMT image in Figure 2.7 exhibits a distinguishable

signal in the tumour region eight days after liposome administration, confirming the presence of

liposomes, whereas the micro-CT image did not visually show contrast at this time point.

Furthermore, by utilizing the confocal microscopic endoscope, the distribution of liposomes in

localized regions of the tumour can be visualized in real time (Figure 2.10). Therefore, by

exploiting CT/optical dual-modality liposomes, one can employ CT imaging to monitor and

quantify whole-body distribution and clearance of the liposomes, as well as provide adequate

anatomical background with good spatial resolution. While optical imaging modalities can be

used to confirm liposomal distribution using whole-body imaging, localized monitoring of the

distribution of the liposomes (i.e. within the tumour microenvironment) is also made possible via

microscopic imaging. Ultimately, the high sensitivity attributed to optical imaging may be

exploited in monitoring and ensuring the clearance of liposomes from the body.

Page 64: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

50

CHAPTER 3 Conclusion

3.1 Conclusions

This thesis has demonstrated the feasibility and effectiveness of combining CT and optical

imaging to obtain qualitative and quantitative measurements of the pharmacokinetics and in vivo

biodistribution of a CT/optical liposome formulation. Through the use of multimodality FMT

and micro-CT imaging, the whole-body and tumour distribution of liposomes were monitored

and quantitatively assessed (Chapter 2). Micro-CT provided quantitative data on the distribution

of liposomes in all organs including tumour, while FMT was exclusively employed to assess the

liposome accumulation in tumour. At the tumor site, FMT demonstrated higher sensitivity than

micro-CT to detect the presence of liposomes. These findings demonstrated the complementary

nature of CT and optical imaging. Specifically, FMT and micro-CT were shown to provide

consistent assessments of liposome accumulation in tumours. A major limitation associated with

FMT is that the presence of high background fluorescence at early imaging time points due to

high systemic levels of liposomes affected the ability of FMT to provide quantitative assessment

of tumor-specific fluorescence signal. Nevertheless, it was concluded that when combined with

CT, FMT can serve as a valuable quantitative 3D optical imaging tool.

In an attempt to improve FMT quantification, the micro-CT data was used to guide the

FMT analysis of liposome accumulation in tumours The tumour volume information obtained

from the CT data set was used during the FMT analysis such that the FMT VOIs matched the

tumour volume determined using micro-CT. Results showed that the employment of micro-CT

greatly increased the delineation and quantification accuracy of FMT. This work provided

valuable insight on the use of CT to assist with FMT image analysis, which will undoubtedly

serve in the ongoing translation of 3D optical imaging techniques into the clinical setting.

Overall, this thesis provided two main innovations. The first is the development of

purification and characterization methodology for the Cy5.5-DSPE optical probe. The second is

the demonstration of the feasibility of using a registered CT data set to improve the accuracy of

quantitative FMT image analysis. This thesis provided significant contribution to the field of

Page 65: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

51

multimodal imaging, through the development of a new CT/optical imaging agent and an

improved dual-modality imaging data quantification method.

Page 66: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

52

3.2 Future Directions

In this report, CT was used to assist FMT-based quantification of the tumour accumulation of a

liposome imaging probe. The accuracy of FMT-based quantification alone has been reported to

vary widely (20 – 300%) for home-built FMT systems [146, 198, 199]. In these reports, the

FMT’s sensitivity and imaging performance has been shown to drop in the presence of non-

specific background fluorescence that reduces contrast [198, 199]. In vivo fluorescence

background is caused by a combination of tissue auto-fluorescence and non-specific distribution

of the administered fluorescent probe. Therefore, there is a need to study the effect of

background fluorescence on the linearity and accuracy of quantification of FMT. In an ongoing

study (see Appendix), the ability of FMT to quantify a fluorescent target embedded in a 3D

tissue-like gel phantom containing known fluorophore and background signal levels is assessed.

In addition, a simple background subtraction method is used to improve the quantification

performance of the FMT system.

Continued development of the FMT system has led to a new generation of imaging

systems, such as the FMT 1500 and FMT 2500 series by Perkin Elmer, which eliminates the

upright suspension of the mouse and the use of IMF fluid that were originally required for the

first generation FMT systems. In the new FMT systems, the anaesthetized mouse can be

immobilized in an imaging cassette in the prone position during scanning, and the imaging

cassette can be transferred to other scanners for easy multimodality image registration. In this

thesis, a first generation FMT system was employed, thus the registration of the FMT images to

the micro-CT images was challenging. Errors in image registration (i.e. location of the FMT

tumour volume within the 3D CT data set) may have contributed to the inaccuracies in

quantification. With the advent of the new generation of FMT systems, the co-registration of

Page 67: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

53

FMT images and micro-CT images should be much more accurate, thereby also likely improving

the quantitative nature of the FMT data. In addition, accurate whole-body co-registration would

also allow CT-based contouring of various organs and tissues of interest, such as the liver, heart,

spleen, and the kidneys, and the subsequent FMT based assessment of the biodistribution of

liposomal agents. Therefore, for future studies involving FMT, it is recommended that the new

generation FMT systems be used to allow proper image registration with 3D imaging modalities

such as CT, MRI, or PET.

The work in this thesis has demonstrated great potential in the development of CT/optical

probes for clinical applications such as image-guided surgery. However, the CT/optical

liposome formulation designed in this work was not found to provide adequate retention of the

Cy5.5-DSPE lipid conjugate, in comparison to the retention of the physically entrapped agent

iohexol. Future studies should focus on the design of a CT/optical liposome formulation wherein

the optical agent is also entrapped within the internal aqueous volume of the vesicles. This will

then ensure that the ratio of the CT to optical agent remains relatively constant over time and will

facilitate more accurate in vivo data comparison between the CT and optical imaging modalities.

As a result, this will greatly increase the confidence level of quantitative optical imaging data,

which will offer significant value in the comprehensive CT-guided assessment of quantitative

optical imaging modalities such as the FMT.

Page 68: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

54

References

1. Lee, S. and X. Chen, Dual-modality probes for in vivo molecular imaging. Molecular Imaging, 2009. 8(2): p. 87-100.

2. Henschke, C.I., et al., Early Lung Cancer Action Project: Overall design and findings from baseline screening. Lancet, 1999. 354(9173): p. 99-105.

3. Harisinghani, M.G., et al., Noninvasive detection of clinically occult lymph-node metastases in prostate cancer. New England Journal of Medicine, 2003. 348(25): p. 2491-2499.

4. Pieterman, R.M., et al., Preoperative staging of non-small-cell lung cancer with positron- emission tomography. New England Journal of Medicine, 2000. 343(4): p. 254-261.

5. Strauss, L.G. and P.S. Conti, The applications of PET in clinical oncology. Journal of Nuclear Medicine, 1991. 32(4): p. 623-648.

6. Tjuvajev, J.G., et al., Noninvasive imaging of herpes virus thymidine kinase gene transfer and expression: A potential method for monitoring clinical gene therapy. Cancer Research, 1996. 56(18): p. 4087-4095.

7. Toloza, E.M., L. Harpole, and D.C. McCrory, Noninvasive staging of non-small cell lung cancer: A review of the current evidence. Chest, 2003. 123(1 SUPPL.): p. 137S-146S.

8. Yang, M., et al., Whole-body optical imaging of green fluorescent protein-expressing tumors and metastases. Proceedings of the National Academy of Sciences of the United States of America, 2000. 97(3): p. 1206-1211.

9. Hwang, D.W., et al., A nucleolin-targeted multimodal nanoparticle imaging probe for tracking cancer cells using an aptamer. Journal of Nuclear Medicine, 2010. 51(1): p. 98-105.

10. Gao, X., et al., In vivo cancer targeting and imaging with semiconductor quantum dots. Nature Biotechnology, 2004. 22(8): p. 969-976.

11. Kumar, R., et al., In vivo biodistribution and clearance studies using multimodal organically modified silica nanoparticles. ACS Nano, 2010. 4(2): p. 699-708.

12. Bartlett, D.W., et al., Impact of tumor-specific targeting on the biodistribution and efficacy of siRNA nanoparticles measured by multimodality in vivo imaging. Proceedings of the National Academy of Sciences of the United States of America, 2007. 104(39): p. 15549-15554.

13. Hoang, B., et al., Noninvasive monitoring of the fate of<sup>111</sup>in-labeled block copolymer micelles by high resolution and high sensitivity microSPECT/CT imaging. Molecular Pharmaceutics, 2009. 6(2): p. 581-592.

14. Yuan, F., et al., Microvascular permeability and interstitial penetration of sterically stabilized (stealth) liposomes in a human tumor xenograft. Cancer Research, 1994. 54(13): p. 3352-3356.

15. Chen, Q., et al., Targeting tumor microvessels using doxorubicin encapsulated in a novel thermosensitive liposome. Molecular Cancer Therapeutics, 2004. 3(10): p. 1311-1317.

Page 69: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

55

16. Louie, A., Multimodality imaging probes: Design and challenges. Chemical Reviews, 2010. 110(5): p. 3146-3195.

17. Beyer, T., D.W. Townsend, and T.M. Blodgett, Dual-modality PET/CT tomography for clinical oncology. Quarterly Journal of Nuclear Medicine, 2002. 46(1): p. 24-34.

18. Snyder, C.S., et al., Complementarity of ultrasound and fluorescence imaging in an orthotopic mouse model of pancreatic cancer. BMC Cancer, 2009. 9.

19. Patton, J.A., D.W. Townsend, and B.F. Hutton, Hybrid Imaging Technology: From Dreams and Vision to Clinical Devices. Seminars in Nuclear Medicine, 2009. 39(4): p. 247-263.

20. Mulder, W.J.M., et al., Nanoparticulate assemblies of amphiphiles and diagnostically active materials for multimodality imaging. Accounts of Chemical Research, 2009. 42(7): p. 904-914.

21. Devaraj, N.K., et al., <sup>18</sup>F labeled nanoparticles for in Vivo PET-CT imaging. Bioconjugate Chemistry, 2009. 20(2): p. 397-401.

22. Dunne, M., et al., APN/CD13-targeting as a strategy to alter the tumor accumulation of liposomes. Journal of Controlled Release.

23. Zheng, J., et al., Liposome contrast agent for CT-based detection and localization of neoplastic and inflammatory lesions in rabbits: Validation with FDG-PET and histology. Contrast Media and Molecular Imaging, 2010. 5(3): p. 147-154.

24. Zheng, J., D. Jaffray, and C. Allen, Quantitative CT imaging of the spatial and temporal distribution of liposomes in a rabbit tumor model. Molecular Pharmaceutics, 2009. 6(2): p. 571-580.

25. Swirski, F.K., et al., Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science, 2009. 325(5940): p. 612-616.

26. McCann, C.M., et al., Combined magnetic resonance and fluorescence imaging of the living mouse brain reveals glioma response to chemotherapy. NeuroImage, 2009. 45(2): p. 360-369.

27. Brigger, I., C. Dubernet, and P. Couvreur, Nanoparticles in cancer therapy and diagnosis. Advanced Drug Delivery Reviews, 2002. 54(5): p. 631-651.

28. Couvreur, P. and C. Vauthier, Nanotechnology: Intelligent design to treat complex disease. Pharmaceutical Research, 2006. 23(7): p. 1417-1450.

29. Torchilin, V.P., Recent advances with liposomes as pharmaceutical carriers. Nature Reviews Drug Discovery, 2005. 4(2): p. 145-160.

30. Danhier, F., O. Feron, and V. Préat, To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery. Journal of Controlled Release, 2010. 148(2): p. 135-146.

31. Torchilin, V., Antibody-modified liposomes for cancer chemotherapy. Expert Opinion on Drug Delivery, 2008. 5(9): p. 1003-1025.

32. Meda, H., Strategy for Designing Anticancer Agents and Consideration of EPR-Effect of Polymeric Drugs in Solid Tumor. Biotherapy, 2004. 18(2): p. 167-173.

Page 70: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

56

33. Greish, K., Enhanced permeability and retention of macromolecular drugs in solid tumors: A royal gate for targeted anticancer nanomedicines. Journal of Drug Targeting, 2007. 15(7-8): p. 457-464.

34. Zheng, J., et al., Multimodal contrast agent for combined computed tomography and magnetic resonance imaging applications. Investigative Radiology, 2006. 41(3): p. 339-348.

35. Oku, N., Delivery of contrast agents for positron emission tomography imaging by liposomes. Advanced Drug Delivery Reviews, 1999. 37(1-3): p. 53-61.

36. Weng, K.C., et al., Targeted tumor cell internalization and imaging of multifunctional quantum dot-conjugated immunoliposomes in vitro and in vivo. Nano Letters, 2008. 8(9): p. 2851-2857.

37. Mulder, W.J.M., et al., A liposomal system for contrast-enhanced magnetic resonance imaging of molecular targets. Bioconjugate Chemistry, 2004. 15(4): p. 799-806.

38. Harrington, K.J., et al., Biodistribution and pharmacokinetics of <sup>111</sup>In-DTPA-labelled pegylated liposomes in a human tumour xenograft model: Implications for novel targeting strategies. British Journal of Cancer, 2000. 83(2): p. 232-238.

39. Van Tilborg, G.A.F., et al., Improved magnetic resonance molecular imaging of tumor angiogenesis by avidin-induced clearance of nonbound bimodal liposomes. Neoplasia, 2008. 10(12): p. 1459-1469.

40. Harrington, K.J., et al., Effective targeting of solid tumors in patients with locally advanced cancers by radiolabeled pegylated liposomes. Clinical Cancer Research, 2001. 7(2): p. 243-254.

41. Saito, R., et al., Distribution of Liposomes into Brain and Rat Brain Tumor Models by Convection-Enhanced Delivery Monitored with Magnetic Resonance Imaging. Cancer Research, 2004. 64(7): p. 2572-2579.

42. Viglianti, B.L., et al., In vivo monitoring of tissue pharmacokinetics of liposome/drug using MRI: Illustration of targeted delivery. Magnetic Resonance in Medicine, 2004. 51(6): p. 1153-1162.

43. Bangham, A.D., M.M. Standish, and J.C. Watkins, Diffusion of univalent ions across the lamellae of swollen phospholipids. Journal of Molecular Biology, 1965. 13(1): p. 238-252.

44. Gabizon, A.A., Pegylated liposomal doxorubicin: Metamorphosis of an old drug into a new form of chemotherapy. Cancer Investigation, 2001. 19(4): p. 424-436.

45. Forssen, E.A., The design and development of DaunoXome® for solid tumor targeting in vivo. Advanced Drug Delivery Reviews, 1997. 24(2-3): p. 133-150.

46. Brown, D.M., et al., Ranibizumab versus verteporfin for neovascular age-related macular degeneration. New England Journal of Medicine, 2006. 355(14): p. 1432-1444.

47. Mayhew, E., et al., Role of cholesterol in enhancing the antitumor activity of cytosine arabinoside entrapped in liposomes. Cancer Treatment Reports, 1979. 63(11-12): p. 1923-1928.

Page 71: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

57

48. Kulkarni, S.B., G.V. Betageri, and M. Singh, Factors affecting microencapsulation of drugs in liposomes. Journal of Microencapsulation, 1995. 12(3): p. 229-246.

49. Zhang, J.A., et al., Development and characterization of a novel Cremophor® EL free liposome-based paclitaxel (LEP-ETU) formulation. European Journal of Pharmaceutics and Biopharmaceutics, 2005. 59(1): p. 177-187.

50. Crosasso, P., et al., Preparation, characterization and properties of sterically stabilized paclitaxel-containing liposomes. Journal of Controlled Release, 2000. 63(1-2): p. 19-30.

51. Moribe, K., K. Maruyama, and M. Iwatsuru, Encapsulation characteristics of nystatin in liposomes: Effects of cholesterol and polyethylene glycol derivatives. International Journal of Pharmaceutics, 1999. 188(2): p. 193-202.

52. Drummond, D.C., et al., Optimizing liposomes for delivery of chemotherapeutic agents to solid tumors. Pharmacological Reviews, 1999. 51(4): p. 691-743.

53. Allen, T.M., et al., Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivo. Biochimica et Biophysica Acta - Biomembranes, 1991. 1066(1): p. 29-36.

54. Simard, P., J.C. Leroux, C. Allen, O. Meyer, Liposomes for drug delivery., in Nanoparticles for pharmaceutical applications. 2007, American Scientific Publishers.

55. Torchilin, V.P., V.Weissig V, Liposomes: A practical approach. 2003: Oxford University Press.

56. Hope, M.J., et al., Production of large unilamellar vesicles by a rapid extrusion procedure. Characterization of size distribution, trapped volume and ability to maintain a membrane potential. Biochimica et Biophysica Acta - Biomembranes, 1985. 812(1): p. 55-65.

57. Hernández, J., A. Martí, and J. Estelrich, Interaction of doxorubicin with lipid systems. Bioconjugate Chemistry, 1991. 2(6): p. 398-402.

58. Olson, F., C.A. Hunt, and F.C. Szoka, Preparation of liposomes of defined size distribution by extrusion through polycarbonate membranes. Biochimica et Biophysica Acta, 1979. 557(1): p. 9-23.

59. Senior, J., J.C.W. Crawley, and G. Gregoriadis, Tissue distribution of liposomes exhibiting long half-lives in the circulation after intravenous injection. Biochimica et Biophysica Acta - General Subjects, 1985. 839(1): p. 1-8.

60. Yuan, F., et al., Vascular permeability in a human tumor xenograft: Molecular size dependence and cutoff size. Cancer Research, 1995. 55(17): p. 3752-3756.

61. Suzuki, M., K. Hori, and I. Abe, A new approach to cancer chemotherapy: Selective enhancement of tumor blood flow with angiotensin II. Journal of the National Cancer Institute, 1981. 67(3): p. 663-669.

62. Fang, J., H. Nakamura, and H. Maeda, The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Advanced Drug Delivery Reviews, 2011. 63(3): p. 136-151.

63. Maeda, H., Tumor-selective delivery of macromolecular drugs via the EPR effect: Background and future prospects. Bioconjugate Chemistry, 2010. 21(5): p. 797-802.

Page 72: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

58

64. Nagamitsu, A., K. Greish, and H. Maeda, Elevating blood pressure as a strategy to increase tumor-targeted delivery of macromolecular drug SMANCS: Cases of advanced solid tumors. Japanese Journal of Clinical Oncology, 2009. 39(11): p. 756-766.

65. Feelisch, M. and E.A. Noack, Correlation between nitric oxide formation during degradation of organic nitrates and activation of guanylate cyclase. European Journal of Pharmacology, 1987. 139(1): p. 19-30.

66. Kojima, H., et al., Fluorescent indicators for imaging nitric oxide production. Angewandte Chemie - International Edition, 1999. 38(21): p. 3209-3212.

67. Pasut, G., et al., Polymer-drug conjugates for combination anticancer therapy: Investigating the mechanism of action. Journal of Medicinal Chemistry, 2009. 52(20): p. 6499-6502.

68. Allen, T.M., Liposomal drug formulations: Rationale for development and what we can expect for the future. Drugs, 1998. 56(5): p. 747-756.

69. Torchilin, V.P., Polymer-coated long-circulating microparticulate pharmaceuticals. Journal of Microencapsulation, 1998. 15(1): p. 1-19.

70. Cullis, P.R., et al., Generating and loading of liposomal systems for drug-delivery applications. Advanced Drug Delivery Reviews, 1989. 3(3): p. 267-282.

71. Haran, G., et al., Transmembrane ammonium sulfate gradients in liposomes produce efficient and stable entrapment of amphipathic weak bases. Biochimica et Biophysica Acta - Biomembranes, 1993. 1151(2): p. 201-215.

72. Bolotin, E.M., et al., Ammonium sulfate gradients for efficient and stable remote loading of amphipathic weak bases into liposomes and ligandoliposomes. Journal of Liposome Research, 1994. 4(1): p. 455-479.

73. Lasic, D.D., et al., Gelation of liposome interior. A novel method for drug encapsulation. FEBS Letters, 1992. 312(2-3): p. 255-258.

74. Northfelt, D.W., et al., Doxorubicin encapsulated in liposomes containing surface-bound polyethylene glycol: Pharmacokinetics, tumor localization, and safety in patients with AIDS-related Kaposi's sarcoma. Journal of Clinical Pharmacology, 1996. 36(1): p. 55-63.

75. Torchilin, V., Multifunctional and stimuli-sensitive pharmaceutical nanocarriers. European Journal of Pharmaceutics and Biopharmaceutics, 2009. 71(3): p. 431-444.

76. Sapra, P. and T.M. Allen, Ligand-targeted liposomal anticancer drugs. Progress in Lipid Research, 2003. 42(5): p. 439-462.

77. Ahmad, I., et al., Antibody-targeted delivery of doxorubicin entrapped in sterically stabilized liposomes can eradicate lung cancer in mice. Cancer Research, 1993. 53(7): p. 1484-1488.

78. Carter, P., Improving the efficacy of antibody-based cancer therapies. Nature Reviews Cancer, 2001. 1(2): p. 118-129.

79. Park, J.W., et al., Anti-HER2 immunoliposomes for targeted therapy of human tumors. Cancer Letters, 1997. 118(2): p. 153-160.

Page 73: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

59

80. Coiffier, B., et al., Chop chemotherapy plus rituximab compared with chop alone in elderly patients with diffuse large-B-cell lymphoma. New England Journal of Medicine, 2002. 346(4): p. 235-242.

81. Slamon, D.J., et al., Use of chemotherapy plus a monoclonal antibody against her2 for metastatic breast cancer that overexpresses HER2. New England Journal of Medicine, 2001. 344(11): p. 783-792.

82. Peer, D., et al., Nanocarriers as an emerging platform for cancer therapy. Nature Nanotechnology, 2007. 2(12): p. 751-760.

83. Drummond, D.C., M. Zignani, and J.C. Leroux, Current status of pH-sensitive liposomes in drug delivery. Progress in Lipid Research, 2000. 39(5): p. 409-460.

84. Pradhan, P., et al., Targeted temperature sensitive magnetic liposomes for thermo-chemotherapy. Journal of Controlled Release, 2010. 142(1): p. 108-121.

85. Park, J.W., et al., Anti-HER2 immunoliposomes: Enhanced efficacy attributable to targeted delivery. Clinical Cancer Research, 2002. 8(4): p. 1172-1181.

86. Park, J.W., et al., Tumor targeting using anti-her2 immunoliposomes. Journal of Controlled Release, 2001. 74(1-3): p. 95-113.

87. Lopes De Menezes, D.E., L.M. Pilarski, and T.M. Allen, In vitro and in vivo targeting of immunoliposomal doxorubicin to human B-cell lymphoma. Cancer Research, 1998. 58(15): p. 3320-3330.

88. Griffioen, A.W., et al., Angiogenesis, a target for tumor therapy. Journal of Laboratory and Clinical Medicine, 1998. 132(5): p. 363-368.

89. Schiffelers, R.M., et al., Anti-tumor efficacy of tumor vasculature-targeted liposomal doxorubicin. Journal of Controlled Release, 2003. 91(1-2): p. 115-122.

90. Kondo, M., et al., Anti-neovascular therapy by liposomal drug targeted to membrane type-1 matrix metalloproteinase. International Journal of Cancer, 2004. 108(2): p. 301-306.

91. Lammers, T., et al., Nanotheranostics and image-guided drug delivery: Current concepts and future directions. Molecular Pharmaceutics, 2010. 7(6): p. 1899-1912.

92. Tashjian, J.A., et al., Rationale for and measurement of liposomal drug delivery with hyperthermia using non-invasive imaging techniques. International Journal of Hyperthermia, 2008. 24(1): p. 79-90.

93. Nieman, K., et al., Reliable noninvasive coronary angiography with fast submillimeter multislice spiral computed tomography. Circulation, 2002. 106(16): p. 2051-2054.

94. Jaffer, F.A. and R. Weissleder, Molecular imaging in the clinical arena. Journal of the American Medical Association, 2005. 293(7): p. 855-862.

95. Bach, P.B., et al., Computed tomography screening and lung cancer outcomes. Journal of the American Medical Association, 2007. 297(9): p. 953-961.

96. Altinoǧlu, E.I., et al., Near-infrared emitting fluorophore-doped calcium phosphate nanoparticles for in vivo imaging of human breast cancer. ACS Nano, 2008. 2(10): p. 2075-2084.

Page 74: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

60

97. Heesakkers, R.A., et al., MRI with a lymph-node-specific contrast agent as an alternative to CT scan and lymph-node dissection in patients with prostate cancer: a prospective multicohort study. The Lancet Oncology, 2008. 9(9): p. 850-856.

98. Müller, C., et al., SPECT study of folate receptor-positive malignant and normal tissues in mice using a novel <sup>99m</sup>Tc-radiofolate. Journal of Nuclear Medicine, 2008. 49(2): p. 310-317.

99. Lordick, F., et al., PET to assess early metabolic response and to guide treatment of adenocarcinoma of the oesophagogastric junction: the MUNICON phase II trial. Lancet Oncology, 2007. 8(9): p. 797-805.

100. Kennedy, J.E., High-intensity focused ultrasound in the treatment of solid tumours. Nature Reviews Cancer, 2005. 5(4): p. 321-327.

101. Townsend, D.W., T. Beyer, and T.M. Blodgett, PET/CT scanners: A hardware approach to image fusion. Seminars in Nuclear Medicine, 2003. 33(3): p. 193-204.

102. Hasegawa, B.H., et al. Dual-modality imaging of function and physiology. 2002. San Diego, CA.

103. Slates, R.B., et al., A study of artefacts in simultaneous PET and MR imaging using a prototype MR compatible PET scanner. Physics in Medicine and Biology, 1999. 44(8): p. 2015-2027.

104. Cho, Z.H., et al., A fusion PET-MRI system with a high-resolution research tomograph-PET and ultra-high field 7.0 T-MRI for the molecular-genetic imaging of the brain. Proteomics, 2008. 8(6): p. 1302-1323.

105. Mulder, W.J.M., et al., Lipid-based nanoparticles for contrast-enhanced MRI and molecular imaging. NMR in Biomedicine, 2006. 19(1): p. 142-164.

106. Tallury, P., K. Payton, and S. Santra, Silica-based multimodal/multifunctional nanoparticles for bioimaging and biosensing applications. Nanomedicine, 2008. 3(4): p. 579-592.

107. McCarthy, J.R., et al., Multimodal nanoagents for the detection of intravascular thrombi. Bioconjugate Chemistry, 2009. 20(6): p. 1251-1255.

108. Rodriguez-Porcel, M., et al., Imaging of VEGF receptor in a rat myocardial infarction model using PET. Journal of Nuclear Medicine, 2008. 49(4): p. 667-673.

109. Frullano, L. and T.J. Meade, Multimodal MRI contrast agents. Journal of Biological Inorganic Chemistry, 2007. 12(7): p. 939-949.

110. Kim, J., Y. Piao, and T. Hyeon, Multifunctional nanostructured materials for multimodal imaging, and simultaneous imaging and therapy. Chemical Society Reviews, 2009. 38(2): p. 372-390.

111. Storrs, R.W., et al., Paramagnetic polymerized liposomes as new recirculating MR contrast agents. Journal of magnetic resonance imaging : JMRI, 1995. 5(6): p. 719-724.

112. Storrs, R.W., et al., Paramagnetic polymerized liposomes: Synthesis, characterization, and applications for magnetic resonance imaging. Journal of the American Chemical Society, 1995. 117(28): p. 7301-7306.

Page 75: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

61

113. Yu, S.B. and A.D. Watson, Metal-based X-ray contrast media. Chemical Reviews, 1999. 99(9): p. 2353-2377.

114. Webb, W.R., et al., CT and MR imaging in staging non-small cell bronchogenic carcinoma: Report of the Radiologic Diagnostic Oncology Group. Radiology, 1991. 178(3): p. 705-713.

115. Uematsu, M., et al., Computed tomography-guided frameless stereotactic radiotherapy for stage I non-small cell lung cancer: A 5-year experience. International Journal of Radiation Oncology Biology Physics, 2001. 51(3): p. 666-670.

116. Lagerwaard, F.J., et al., Identification of prognostic factors in patients with brain metastases: A review of 1292 patients. International Journal of Radiation Oncology Biology Physics, 1999. 43(4): p. 795-803.

117. Schambach, S.J., et al., Application of micro-CT in small animal imaging. Methods, 2010. 50(1): p. 2-13.

118. Feldkamp, L.A., et al., The direct examination of three-dimensional bone architecture in vitro by computed tomography. Journal of Bone and Mineral Research, 1989. 4(1): p. 3-11.

119. Ruegsegger, P., B. Koller, and R. Muller, A microtomographic system for the nondestructive evaluation of bone architecture. Calcified Tissue International, 1996. 58(1): p. 24-29.

120. Wietholt, C., et al., Bronchial circulation angiogenesis in the rat quantified with SPECT and micro-CT. European Journal of Nuclear Medicine and Molecular Imaging, 2008. 35(6): p. 1124-1132.

121. Drinane, M., et al., The antiangiogenic activity of rPAI-123 inhibits vasa vasorum and growth of atherosclerotic plaque. Circulation Research, 2009. 104(3): p. 337-345.

122. Langheinrich, A.C., et al., 3-Deazaadenosine inhibits vasa vasorum neovascularization in aortas of ApoE<sup>-/-</sup>/LDL<sup>-/-</sup> double knockout mice. Atherosclerosis, 2009. 202(1): p. 103-110.

123. Cavanaugh, D., et al., In vivo respiratory-gated micro-CT imaging in small-animal oncology models. Molecular Imaging, 2004. 3(1): p. 55-62.

124. Almajdub, M., et al., Kidney volume quantification using contrast-enhanced in vivo X-ray micro-CT in mice. Contrast Media and Molecular Imaging, 2008. 3(3): p. 120-126.

125. Graham, K.C., et al., Noninvasive quantification of tumor volume in preclinical liver metastasis models using contrast-enhanced X-ray computed tomography. Investigative Radiology, 2008. 43(2): p. 92-99.

126. Detombe, S.A., et al., Longitudinal follow-up of cardiac structure and functional changes in an infarct mouse model using retrospectively gated micro-computed tomography. Investigative Radiology, 2008. 43(7): p. 520-529.

127. Hickson, J., In vivo optical imaging: Preclinical applications and considerations. Urologic Oncology: Seminars and Original Investigations, 2009. 27(3): p. 295-297.

128. Graves, E.E., et al., Validation of in vivo fluorochrome concentrations measured using fluorescence molecular tomography. Journal of biomedical optics., 2005. 10(4): p. 44019.

Page 76: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

62

129. Frangioni, J.V., In vivo near-infrared fluorescence imaging. Current Opinion in Chemical Biology, 2003. 7(5): p. 626-634.

130. Klohs, J., A. Wunder, and K. Licha, Near-infrared fluorescent probes for imaging vascular pathophysiology. Basic Research in Cardiology, 2008. 103(2): p. 144-151.

131. von Wallbrunn, A., et al., In vivo optical imaging of CD13/APN-expression in tumor xenografts. Journal of biomedical optics, 2008. 13(1): p. 011007.

132. Von Wallbrunn, A., et al., In vivo imaging of integrin ανβ<sub>3</sub> expression using fluorescence-mediated tomography. European Journal of Nuclear Medicine and Molecular Imaging, 2007. 34(5): p. 745-754.

133. Ntziachristos, V., et al., Fluorescence molecular tomography resolves protease activity in vivo. Nature Medicine, 2002. 8(7): p. 757-760.

134. Han, M., et al., Quantum-dot-tagged microbeads for multiplexed optical coding of biomolecules. Nature Biotechnology, 2001. 19(7): p. 631-635.

135. Wouters, F.S., P.J. Verveer, and P.I.H. Bastiaens, Imaging biochemistry inside cells. Trends in Cell Biology, 2001. 11(5): p. 203-211.

136. Dewhirst, M.W., et al., Intravital fluorescence facilitates measurement of multiple physiologic functions and gene expression in tumors of live animals. Disease Markers, 2002. 18(5-6): p. 293-311.

137. Farkas, S., et al., Quantification of mucosal leucocyte endothelial cell interaction by in vivo fluorescence microscopy in experimental colitis in mice. Clinical and Experimental Immunology, 2001. 126(2): p. 250-258.

138. Jain, R.K., L.L. Munn, and D. Fukumura, Dissecting tumour pathophysiology using intravital microscopy. Nature Reviews Cancer, 2002. 2(4): p. 266-276.

139. Hadjantonakis, A.K., et al., Technicolour transgenics: Imaging tools for functional genomics in the mouse. Nature Reviews Genetics, 2003. 4(8): p. 613-625.

140. Halbhuber, K.J. and K. König, Modern laser scanning microscopy in biology, biotechnology and medicine. Annals of Anatomy, 2003. 185(1): p. 1-20.

141. Zipfel, W.R., R.M. Williams, and W.W. Webb, Nonlinear magic: Multiphoton microscopy in the biosciences. Nature Biotechnology, 2003. 21(11): p. 1369-1377.

142. Künneke, S. and A. Janshoff, Visualization of molecular recognition events on microstructured lipid-membrane compartments by in situ scanning force microscopy. Angewandte Chemie - International Edition, 2002. 41(2): p. 314-316.

143. Ntziachristos, V., Going deeper than microscopy: The optical imaging frontier in biology. Nature Methods, 2010. 7(8): p. 603-614.

144. Weissleder, R. and M.J. Pittet, Imaging in the era of molecular oncology. Nature, 2008. 452(7187): p. 580-589.

145. Ntziachristos, V., Fluorescence molecular imaging, M.L. Yarmush, Editor. 2006. p. 1-33.

146. Ntziachristos, V., et al., Visualization of antitumor treatment by means of fluorescence molecular tomography with an annexin V-Cy5.5 conjugate. Proceedings of the National Academy of Sciences of the United States of America, 2004. 101(33): p. 12294-12299.

Page 77: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

63

147. Graves, E.E., et al., A submillimeter resolution fluorescence molecular imaging system for small animal imaging. Medical Physics, 2003. 30(5): p. 901-911.

148. Ntziachristos, V. and R. Weissleder, Charge-coupled-device based scanner for tomography of fluorescent near-infrared probes in turbid media. Medical Physics, 2002. 29(5): p. 803-809.

149. Grimm, J., et al., Use of gene expression profiling to direct in vivo molecular imaging of lung cancer. Proceedings of the National Academy of Sciences of the United States of America, 2005. 102(40): p. 14404-14409.

150. Nahrendorf, M., et al., Hybrid in vivo FMT-CT imaging of protease activity in atherosclerosis with customized nanosensors. Arteriosclerosis, Thrombosis, and Vascular Biology, 2009. 29(10): p. 1444-1451.

151. Cortez-Retamozo, V., et al., Real-time assessment of inflammation and treatment response in a mouse model of allergic airway inflammation. Journal of Clinical Investigation, 2008. 118(12): p. 4058-4066.

152. Seymour, L.W., et al., Hepatic drug targeting: Phase I evaluation of polymer-bound doxorubicin. Journal of Clinical Oncology, 2002. 20(6): p. 1668-1676.

153. Mulder, W.J.M., et al., MR molecular imaging and fluorescence microscopy for identification of activated tumor endothelium using a bimodal lipidic nanoparticle. FASEB Journal, 2005. 19(14): p. 2008-2010.

154. Viglianti, B.L., et al., Chemodosimetry of in vivo tumor liposomal drug concentration using MRI. Magnetic Resonance in Medicine, 2006. 56(5): p. 1011-1018.

155. Mikhaylova, M., et al., Imaging of cationic multifunctional liposome-mediated delivery of COX-2 siRNA. Cancer Gene Therapy, 2009. 16(3): p. 217-226.

156. Kamaly, N., et al., Bimodal paramagnetic and fluorescent liposomes for cellular and tumor magnetic resonance imaging. Bioconjugate Chemistry, 2008. 19(1): p. 118-129.

157. Vucic, E., et al., Well-defined, multifunctional nanostructures of a paramagnetic lipid and a lipopeptide for macrophage imaging. Journal of the American Chemical Society, 2009. 131(2): p. 406-407.

158. Lijowski, M., et al., High sensitivity: High-resolution SPECT-CT/MR molecular imaging of angiogenesis in the Vx2 model. Investigative Radiology, 2009. 44(1): p. 15-22.

159. Torchilin, V.P., Targeted pharmaceutical nanocarriers for cancer therapy and imaging. AAPS Journal, 2007. 9(2).

160. Palko, H.A., J.Y. Fung, and A.Y. Louie, Positron emission tomography: A novel technique for investigating the biodistribution and transport of nanoparticles. Inhalation Toxicology, 2010. 22(8): p. 657-668.

161. Chrastina, A. and J.E. Schnitzer, Iodine-125 radiolabeling of silver nanoparticles for in vivo SPECT imaging. Int J Nanomedicine, 2010. 5: p. 653-9.

162. Beckmann, N., et al., In vivo visualization of macrophage infiltration and activity in inflammation using magnetic resonance imaging. Wiley interdisciplinary reviews. Nanomedicine and nanobiotechnology, 2009. 1(3): p. 272-298.

Page 78: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

64

163. Kaijzel, E.L., et al., Multimodal imaging and treatment of bone metastasis. Clinical and Experimental Metastasis, 2009. 26(4): p. 371-379.

164. Kamaya, A., et al., Imaging and diagnosis of postpartum complications: Sonography and other imaging modalities. Ultrasound Quarterly, 2009. 25(3): p. 151-162.

165. Daldrup, H.E., G.H. Simon, and R.C. Brasch, Imaging of tumor angiogenesis: Current approaches and future prospects. Current Pharmaceutical Design, 2006. 12(21): p. 2661-2672.

166. Phelps, M.E., PET: The merging of biology and imaging into molecular imaging. Journal of Nuclear Medicine, 2000. 41(4): p. 661-681.

167. Belanger, H.G., et al., Recent neuroimaging techniques in mild traumatic brain injury. Journal of Neuropsychiatry and Clinical Neurosciences, 2007. 19(1): p. 5-20.

168. Townsend, D.W., Dual-modality imaging: Combining anatomy and function. Journal of Nuclear Medicine, 2008. 49(6): p. 938-955.

169. Heiss, W.D., et al., Simultaneous functional and morphological brain imaging with an integrated MR/PET scanner. Journal of Cerebral Blood Flow and Metabolism, 2007. 27(SUPPL. 1).

170. Antoch, G. and A. Bockisch, Combined PET/MRI: A new dimension in whole-body oncology imaging? European Journal of Nuclear Medicine and Molecular Imaging, 2009. 36(SUPPL. 1): p. 113-120.

171. Srinivasan, S., et al., Developments in quantitative oxygen-saturation imaging of breast tissue in vivo using multispectral near-infrared tomography. Antioxidants and Redox Signaling, 2007. 9(8): p. 1143-1156.

172. Hyde, D., et al., Hybrid FMT-CT imaging of amyloid-β plaques in a murine Alzheimer's disease model. NeuroImage, 2009. 44(4): p. 1304-1311.

173. Rudin, M. and R. Weissleder, Molecular imaging in drug discovery and development. Nature Reviews Drug Discovery, 2003. 2(2): p. 123-131.

174. Bremer, C., et al., Optical imaging of spontaneous breast tumors using protease sensing 'smart' optical probes. Investigative Radiology, 2005. 40(6): p. 321-327.

175. Schulz, R.B., et al., Hybrid system for simultaneous fluorescence and X-ray computed tomography. IEEE Transactions on Medical Imaging, 2010. 29(2): p. 465-473.

176. Guo, X., et al., A combined fluorescence and microcomputed tomography system for small animal imaging. IEEE Transactions on Biomedical Engineering, 2010. 57(12): p. 2876-2883.

177. Davis, S.C., et al., Magnetic resonance-coupled fluorescence tomography scanner for molecular imaging of tissue. Review of Scientific Instruments, 2008. 79(6).

178. Ntziachristos, V., et al., Concurrent MRI and diffuse optical tomography of breast after indocyanine green enhancement. Proceedings of the National Academy of Sciences of the United States of America, 2000. 97(6): p. 2767-2772.

179. Ntziachristos, V., X.H. Ma, and B. Chance, Time-correlated single photon counting imager for simultaneous magnetic resonance and near-infrared mammography. Review of Scientific Instruments, 1998. 69(12): p. 4221-4233.

Page 79: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

65

180. Zielhuis, S.W., et al., Lanthanide-loaded liposomes for multimodality imaging and therapy. Cancer Biotherapy and Radiopharmaceuticals, 2006. 21(5): p. 520-527.

181. Gopalakrishnan, G., et al., Multifunctional lipid/quantum dot hybrid nanocontainers for controlled targeting of live cells. Angewandte Chemie - International Edition, 2006. 45(33): p. 5478-5483.

182. Mikhail, A.S. and C. Allen, Block copolymer micelles for delivery of cancer therapy: Transport at the whole body, tissue and cellular levels. Journal of Controlled Release, 2009. 138(3): p. 214-223.

183. Goins, B.A. and W.T. Phillips, The use of scintigraphic imaging during liposome drug development. Journal of Pharmacy Practice, 2001. 14(5): p. 397-406.

184. Zheng, J., et al., In vivo performance of a liposomal vascular contrast agent for CT and MR-based image guidance applications. Pharmaceutical Research, 2007. 24(6): p. 1193-1201.

185. Song, S., et al., Novel peptide ligand directs liposomes toward EGF-R high-expressing cancer cells in vitro and in vivo. FASEB Journal, 2009. 23(5): p. 1396-1404.

186. Song, S., et al., Peptide ligand-mediated liposome distribution and targeting to EGFR expressing tumor in vivo. International Journal of Pharmaceutics, 2008. 363(1-2): p. 155-161.

187. Southwick, P.L., et al., Cyanine dye labeling reagents - Carboxymethylindocyanine succinimidyl esters. Cytometry, 1990. 11(3): p. 418-430.

188. Mujumdar, R.B., et al., Cyanine dye labeling reagents: Sulfoindocyanine succinimidyl esters. Bioconjugate Chemistry, 1993. 4(2): p. 105-111.

189. Johnson, S.M., et al., Single bilayer liposomes. BBA - Biomembranes, 1971. 223(3): p. 820-826.

190. Ntziachristos, V., et al., Looking and listening to light: The evolution of whole-body photonic imaging. Nature Biotechnology, 2005. 23(3): p. 313-320.

191. Speck, U., Contrast agents: X-ray contrast agents and molecular imaging--a contradiction? Handbook of experimental pharmacology, 2008(185 Pt 1): p. 167-175.

192. Wu, K., et al., PET CT Thresholds for Radiotherapy Target Definition in Non-Small-Cell Lung Cancer: How Close are we to the Pathologic Findings? International Journal of Radiation Oncology Biology Physics, 2010. 77(3): p. 699-706.

193. Erdi, Y.E., et al., Segmentation of lung lesion volume by adaptive positron emission tomography image thresholding. Cancer, 1997. 80(12 SUPPL.): p. 2505-2509.

194. Erdi, Y.E., et al., Radiotherapy treatment planning for patients with non-small cell lung cancer using positron emission tomography (PET). Radiotherapy and Oncology, 2002. 62(1): p. 51-60.

195. Bradley, J., et al., Impact of FDG-PET on radiation therapy volume delineation in non-small-cell lung cancer. International Journal of Radiation Oncology Biology Physics, 2004. 59(1): p. 78-86.

Page 80: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

66

196. Miller, T.R. and P.W. Grigsby, Measurement of tumor volume by PET to evaluate prognosis in patients with advanced cervical cancer treated by radiation therapy. International Journal of Radiation Oncology Biology Physics, 2002. 53(2): p. 353-359.

197. Vines, D.C., et al., Quantitative PET comparing gated with nongated acquisitions using a NEMA phantom with respiratory-simulated motion. Journal of Nuclear Medicine Technology, 2007. 35(4): p. 246-251.

198. Gao, M., et al., Effects of background fluorescence in fluorescence molecular tomography. Applied Optics, 2005. 44(26): p. 5468-5474.

199. Soubret, A. and V. Ntziachristos, Fluorescence molecular tomography in the presence of background fluorescence. Physics in Medicine and Biology, 2006. 51(16): p. 3983-4001.

Page 81: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

67

Appendix

Included herein are preliminary results obtained from studies examining the effect of background

fluorescence on the linearity and accuracy of quantification of FMT as previously described in

Chapter 3.2.

The gel phantoms were prepared using agarose and index matching fluid (IMF, Perkin

Elmers). Briefly, agarose was dissolved in IMF (2% w/w) at 80°C. The fluorophore GenhanceTM

680 (GH680), which has similar excitation and emission wavelengths to those of Cy5.5, was

then added into the solutions to achieve concentrations of 0, 20, 50, 80, 100, 150, 200 and 300

nM GH680. Each individual solution was then cooled inside a 1cc cubic mold (1x1x1 cm3)

resulting in 1cc cubic gels that varied in terms of GH680 concentration. Each of these small

cubic gels was placed at the centre of an 18cc mold (3.8x3.8x1.3 cm3), and the mold was filled

with the GH680 agarose solution (prepared above), which engulfed the small 1cc cubic gel. The

solution was cooled down until the larger 18cc gel formed, which was then removed from the

mold. This results in a set of gel phantoms wherein the small gel was considered the “insert” and

the larger the “background” (Figure A1c). These gel phantoms were the immobilized inside an

FMT imaging cassette (Figure A1b), which was placed on the scanning stage of the FMT 2500

system (Figure A1a) and FMT scanning was performed. As shown in Figure A1c, the phantoms

were scanned with two different orientations, with the gel insert facing toward and away from

the CCD camera.

Quantitative image analysis was performed with zero FMT window threshold. The

contoured volume of interest (VOI) was adjusted to approximately a third of the actual gel

insert’s volume and engulfed its centre. The mean FMT intensity within the contour was

measured. Similarly, the background intensity was measured. A calibration curve was

constructed by contouring the gel insert with zero background fluorescence to convert mean

Page 82: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

68

FMT intensities to concentrations of GH680. The background subtraction method was then

applied to the gel insert as follows:

Corrected insert intensity = mean insert FMT intensity – mean background FMT intensity

Quantification error in each gel insert is determined by the following equation:

Quantification Error = (Quantified concentration (with or without background subtraction) –

Actual concentration) / Actual concentration x 100%

The obtained images (Figure A2) showed that the gel inserts can be identified visually

and accurately delineated at high signal to background ratios (S/B ratios > 1, Figure A2a, b, c,

and d), but that this worsens as the S/B ratio decreased (i.e. S/B ratios ≤ 1) (Figure A2e, f).

Therefore, a higher S/B ratio is preferred for the successful detection and delineation of the

target. The FMT quantification results also demonstrated that it is necessary to construct a

custom FMT calibration curve using phantoms with sizes and an environment that is similar to

that of the target to provide more accurate quantification. The internal FMT quantification,

which contained calibrations provided by the vendor, gave rise to ~100% quantification error at

zero background compared to our own calibration, which gave less than 5% error at zero

background.

The FMT signal increased linearly with increased GH680 concentrations in the gel insert

(R2 > 0.95 for all linear fits in Figure A3) regardless of the background level. However, the

quantification accuracy decreased with increasing background levels (Figure A3), leading to an

exponential increase in quantification error (Figure A4). As shown in Figure A4, at the lowest

Page 83: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

69

S/B ratio of 0.167, the quantification error was 908 ± 89%. At the highest S/B ratio of 15, the

quantification error was 6 ± 4%. Background subtraction led to an overall reduction in the

quantification error. Specifically, after background subtraction, at the lowest S/B ratio of 0.167,

the quantification error was reduced to 104 ± 89%, while at the highest S/B ratio of 15, the

quantification error remained at 6 ± 4%. Such results are consistent with studies performed by

Gao and Stobiet [198], where the background fluorescence was found to significantly affect the

accuracy of quantification using FMT when the S/B ratio is below 3 with homogeneous

background fluorescence, with a quantification error of ~150% at a S/B ratio of ~2 [198]. These

results demonstrate that the FMT’s quantification results need to be carefully interpreted for

studies involving a comparison between target concentrations with different background levels.

Assuming that acceptable quantification error is ≤ 30%, it was observed that without

background subtraction, acceptable quantification error is reached only when S/B ≥ 3.75, while

with background subtraction, acceptable quantification error was reached when S/B ≥ 1.5. This

demonstrates that applying such a background subtraction when analyzing FMT images greatly

improves the accuracy of quantification. However, it is still required that the target signal be at

least 1.5 times greater than the background signal. Thus, for an intravenously administered

fluorescent probe to be quantitatively assessed in FMT, it should be required that the target of

interest be isolated from organs responsible for the agent’s distribution and clearance; the target

should be isolated from major blood vessels; and the time of imaging post administration should

be optimized to provide the optimum signal-to-background ratio.

Page 84: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

70

Figure 11. (a) Inside the FMT system. The imaging cassette (as seen in (b)) is placed in the

scanning stage prior to a scan. (b) A photo of the phantom gel inside the imaging cassette. The

gel insert (1 x 1 x 1 cm3) is fused inside the large gel (3.8 x 3.8 x 1.3 cm3), and the cassette

height is adjusted to 1.3 cm to sandwich and tightly immobilize the gel. (c) A schematic of the

large gel geometry with the gel inserted either toward or away from the source laser. Both

geometries were considered In this study.

Page 85: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

71

Figure 12. Effect of background concentration (a-f) on signal detected in ROI. The gel insert

(200 nM GH680) is facing the camera with various backgrounds (a) 20 nM, (b) 50 nM, (c) 80

nM, (d) 150 nM, (e) 200 nM, and (f) 300 nM. The dotted contours (5 mm x 5 mm) represent the

ROIs used for the analysis. The positions of the ROIs were located using both the FMT intensity

as well as the reflectance image. Slices shown have been averaged over 5mm in the z-dimension

for illustration purposes.

Page 86: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

72

Figure 13. FMT GH680 concentrations vs. actual GH680 concentrations at various background

levels (n=3 per level). The dotted line represents the line of identity. The FMT concentration

increases substantially as the background level increases. At the lowest background of 20 nM,

the quantified concentrations are very close to the actual concentrations in the gel cubes.

Background below 20 nM was found to be equivalent to zero background. At any given

background, linearity is still preserved for varying GH680 concentrations in the gel cubes (R2 >

0.95 for all fits).

Page 87: Comparison of CT and Optical Image-based …...indicates a statistically significant difference between micro-CT and FMT data at that time point. (c) Ratio of iohexol to Cy5.5 (wt/wt)

73

Figure 14. The quantification error of the FMT for the gel inserts with various background levels

before and after applying background subtraction method using the method reported here. The

signal-to-background (S/B) ratio has been used to express the strength of the FMT signal in the

gel cube compared to background (BKG). Data are expressed as mean ± standard deviation (n=3

to 12). The dotted line indicates a S/B ratio of 1, representing that the GH680 concentrations in

the gel cube and background are the same. As shown, the quantification error before background

subtraction followed an exponential growth as the S/B ratio decreased, reaching 908 ± 89% at

S/B ratio of 0.167. Overall, with background subtraction, the quantification improved

substantially (p < 0.05).