topical molecular beacons for in vivo image …...ii topical molecular beacons for in vivo...

80
Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma by Laura Allysa Jane Burgess A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Medical Biophysics University of Toronto © Copyright by Laura Burgess 2014

Upload: others

Post on 15-Mar-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma

by

Laura Allysa Jane Burgess

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

Department of Medical Biophysics University of Toronto

© Copyright by Laura Burgess 2014

Page 2: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

ii

Topical Molecular Beacons for In Vivo Image-Guided Resection of

Oral Carcinoma

Laura Allysa Jane Burgess

Master of Science

Department of Medical Biophysics

University of Toronto

2014

Abstract

Oral carcinoma has become a major health problem, with nearly 300,000 people diagnosed

worldwide annually. The 5-year survival rate is as low as 30%, mainly attributed to poor

delineation of lesions. Optical imaging approaches to identify oral carcinoma tissue during

surgery are currently in trial. While decreased recurrence rates are shown, high rates of false

positives occur. Expanding upon this, a molecular beacon strategy for oral carcinoma delineation

was devised.

The selected MMP molecular beacon consists of a fluorophore conjugated to a quencher via a

disease-specific linker, activated by MMPs. The activated beacon becomes fluorescent, guiding

resection. MMPs have been associated with oral tumors and several members as highly

upregulated in oral carcinoma, making them an ideal target.

Page 3: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

iii

I investigated, in vivo, the utility of this MMP-cleavable beacon in targeting oral carcinoma. Here

I demonstrate its high tumor specificity and potential for integration into the clinic to improve

patient outcome.

Page 4: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

iv

Acknowledgments

To my supervisors, Dr. Gang Zheng and Dr. Alex Vitkin, thank you for supporting me and

allowing this work to be possible. Gang, thank you for your encouragement over these past few

years, and for giving me the opportunity and the room to grow. To my committee members, Dr.

Brian Wilson and Dr. Ming Tsao, thank you for your guidance, I’ve learned so much from you

both.

This work would not have been possible without help from so many. Dr. Tracy Liu, very simply,

you were my first mentor here and you taught me how science should be done, all while being

one of the great friends I made here. Dr. Juan Chen, you’re always there to help for any problem

and I know you have nothing but the best interests of all your students in mind, thank you. Dr.

Nikolaus Wolter, you were amazing to work with and I’ll always appreciate your advice and

encouragement. Dr. Eduardo Moriyama, you’re so good at what you do and you’re always

willing to help everyone around you. Thank you for all the help you gave me. Everyone at ARC,

particularly Sadiya Yousef, who always goes above and beyond to help me, Dr. David Hanwell

and everyone else who worked with my hamsters, thank you. The illness and sacrifice of those

hamsters will never been forgotten. To everyone at the Zheng lab, I’ve learned so much and have

had a great time with all of you. Especially Lizzie, Moj, TD, Luby, Tracy, Neeshma, Ken, Stash,

and Danielle; you helped me when I needed it, listened to me whine, made me laugh, provided

frequent distractions and we had quite a few amazing PNPs. I’ll miss you all and I hope the

memories don’t stop now.

Mom and Dad, any ambition, work ethic, dedication and empathy in me are the result of you.

You always gave me every opportunity and supported all of dreams, from putting me into

engineering and science camps to trips on a whim to see David Beckham play and everything in

between. You have always been and always will be my biggest cheerleaders. Dad, it feels like

it’s been forever since you sat across from me while I did my homework every night, quizzed me

Page 5: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

v

for every test and annoyed me when I thought I couldn’t figure something out. Simply put, any

accomplishment now or in the future, wouldn’t have been possible without you and your

unwavering support. Samantha, Dayna, and Darrell, you always pushed me to do more and be

better, striving to be more like my big brother and sisters. To you, and Allan, Matt and Monique,

thanks for never treating me like your annoying little sister.

Lastly, Daniel, thank you for putting up with me when grad school literally made me crazy and

for making everything better when it all seemed to be too much. I wouldn’t have completed this

thesis now and be going to medical school in the fall if you hadn’t told me I’d be amazing at it,

until one day I finally believed you.

Page 6: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

vi

Table of Contents

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

Table of Contents ........................................................................................................................... vi

List of Tables ............................................................................................................................... viii

List of Figures ................................................................................................................................ ix

Chapter 1 Potential Value of Molecular Beacons for Treatment of Oral Carcinoma ..................... 1

1.1 Oral Cancer ......................................................................................................................... 1

1.1.1 Oral Carcinoma ....................................................................................................... 1

1.1.2 Current Treatments ................................................................................................. 4

1.2 Optical Imaging ................................................................................................................ 11

1.2.1 Fluorescence ......................................................................................................... 11

1.2.2 COOLS Trial ......................................................................................................... 14

1.3 Matrix Metalloproteinases ................................................................................................ 17

1.3.1 MMPs in Cancer ................................................................................................... 19

1.4 Molecular Beacons ............................................................................................................ 23

1.4.1 Peptide-Based Molecular Beacons ....................................................................... 25

1.4.2 MMP-Targeted Molecular Beacons ...................................................................... 26

Chapter 2 Topically Applied Molecular Beacons for Fluorescence-Guided Resection of Oral

Carcinoma ................................................................................................................................ 31

2.1 Introduction ....................................................................................................................... 31

2.2 Materials and Methods ...................................................................................................... 33

2.3 Results ............................................................................................................................... 37

2.3.1 In vivo validation of molecular beacon specificity in a murine head and neck

cancer model ......................................................................................................... 37

Page 7: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

vii

2.3.2 In vivo activation and specificity of the molecular beacon in a clinically

relevant animal model ........................................................................................... 39

2.3.3 Ex vivo HPLC Studies ........................................................................................... 43

2.4 Discussion ......................................................................................................................... 45

Chapter 3 Future Directions, Research and Development ............................................................ 48

3.1 Introduction ....................................................................................................................... 48

3.2 Immunostain-like Procedure To Identify MMPs Within Hamster Cheek Tissue ............. 49

3.3 Quantification of Fluorescence Microscopy ..................................................................... 51

3.4 Topical Gel or Spray ......................................................................................................... 52

3.5 Fluorescence-Guided Resection ........................................................................................ 54

3.6 Increased Tissue Penetration ............................................................................................. 55

3.7 Conclusions ....................................................................................................................... 57

References ..................................................................................................................................... 59

Page 8: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

viii

List of Tables

Table 1: Treatment modalities for oral carcinoma .......................................................................... 5

Table 2: The side effects associated with the different treatment modalities for OSCC 7,13. ....... 9

Page 9: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

ix

List of Figures

Figure 1: The different stages of disease progression. .................................................................... 3

Figure 2: Jablonski diagram illustrating absorption, fluorescence and phosphorescence. ........... 12

Figure 3: Patient with an occult recurrent carcinoma identified by the use of the autofluorescence

technique. ...................................................................................................................................... 15

Figure 4: The probability of local recurrence-free survival of the 60 patients enrolled in the

preliminary trial, with this optically-guided approach. ................................................................. 16

Figure 5: The domain structure of MMPs. .................................................................................... 18

Figure 6: The various functions of MMPs in cancer progression. ................................................ 20

Figure 7: Immunohistochemistry was performed on formalin-fixed human oral carcinoma tissue

using mouse monoclonal antibodies demonstrating MMP expression in oral cancer. ................. 23

Figure 8: Molecular beacon structure. .......................................................................................... 24

Figure 9: Validation of the MB's MMP-7 specificity. .................................................................. 27

Figure 10: MMP-7-specific activation validation in vitro with fluorescent and brightfield

confocal images. ........................................................................................................................... 28

Figure 11: In vivo images of a mouse bearing a KB flank tumor post-i.v. injection of MB. ....... 29

Figure 12: The HPLC traces of beacon incubated with various MMPs for 48 hours. .................. 30

Figure 13: Molecular beacon structure in the absence and presence of the specific protease

capable of activating the beacon. .................................................................................................. 32

Figure 14: In-house fluorescence imaging system. ....................................................................... 34

Page 10: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

x

Figure 15: The experimental protocol for all hamster cheek studies involved beginning with

tumor-bearing hamster cheeks. ..................................................................................................... 36

Figure 16: Representative xenograft model fluorescence images with PPMMPB and PPMMP

demonstrating tumor-associated fluorescence and whole tongue fluorescence, respectively.. .... 38

Figure 17: Representative hematoxylin and eosin (i) and confocal images (ii) of tumor-bearing

tongues. ......................................................................................................................................... 39

Figure 18: Representative images of a hamster cheek treated with PPMMPB... ............................ 40

Figure 19: Representative images of various control hamster cheeks: A) PPMMP-treated, B)

negative control-treated, C) healthy cheek having undergone surgery and treated with PPMMPB.

....................................................................................................................................................... 41

Figure 20: Representative hematoxylin and eosin and confocal images of cheeks treated with

PPMMPB.. ....................................................................................................................................... 42

Figure 21: Representative hematoxylin and eosin (i) and confocal images (ii) of hamster cheeks

treated with the various controls.. ................................................................................................. 43

Figure 22: Representative HPLC traces of A) beacon extracted from tumor showing that both

intact PPMMPB and PPMMP fragment are present within the tumor, B) beacon applied to the

tumor.. ........................................................................................................................................... 44

Figure 23: Representative images of a hamster cheek treated with PPMMPB and corresponding

PPMMPB cleavage or activation. .................................................................................................... 45

Figure 24: Tumor-bearing hamster cheeks that did not undergo PPMMPB application in vivo were

frozen and tissues slices obtained for the immunostain-like procedure on sequential slices. ...... 50

Figure 25: Confocal images of droplets of A) 1 mM, B) 10 M (n=2). ....................................... 52

Figure 26: Hamster cheek treated with the thermal-transitioning PPMMPB.. ................................ 54

Figure 27: Confocal and transmitted light images of MT-1 cells incubated with (A) pyro, (B)

pyro-5r and (C) pyro-8r. ............................................................................................................... 56

Page 11: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

1

Chapter 1 Potential Value of Molecular Beacons for Treatment of Oral

Carcinoma

1.1 Oral Cancer

Oral cancer is the sixth most common malignancy in the world, after lung, stomach, breast, colon

and rectum, and cervix and corpus uteri cancers. Worldwide, those diagnosed with oral cancer

have a 5-year survival rate of only 50% 1,2. Four thousand Canadians are diagnosed with oral

cancer every year 3,4 and even in developed countries like Canada, a third of patients diagnosed

with oral cancer will die as a result of their disease.

1.1.1 Oral Carcinoma

The epidermis, including that within the oral cavity, mainly consists of squamous cells.

Squamous cell carcinoma (SCC) occurs at many different sites in the body (including bladder,

skin, and the digestive tract) 5. While SCC at different sites in the body presents with different

symptoms and these sites are treated differently, SCC is histologically distinct from other forms

of cancer. Epithelial cells multiply uncontrollably and distinct architectural changes ensue,

including epithelial stratification and premature keratinization 6. Oral squamous cell carcinoma

(OSCC) represents more than 95% of all oral cancers 7,8. Most commonly, OSCC arises in the

mucous membrane of the tongue, floor of mouth and vermillion borders of lips and is

characterized by having a much greater linear extent than depth 9,10.

Oral cancer development is multistep and sequential, with oral cancer preceded by premalignant

lesions or dysplasia, often asymptomatic 11. Precancerous lesions consist of morphologically

altered tissues where cancer is more likely to occur, when compared to normal tissues 12.

Dysplastic cells are classified based on a number of changes, including irregular epithelial

stratification, premature keratinization, increased nuclear size and increased number of nucleoli

Page 12: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

2

9. Such lesions can be further classified as being either leukoplakia or erythroplakia. These

premalignant or early malignant lesions typically appear as painless white or red patches,

respectively, 11,13.

The most common precancerous lesion is leukoplakia. Leukoplakia is a predominantly white

lesion of the oral mucosa that cannot be classified as any other definable lesion 10,12,15. These

develop in 1-4% of the population and up to 40% of these transform into malignant lesions

within 5 years 10. Leukoplakias can either be smooth, homogeneous (see Figure 1a) or speckled,

pitted, non-homogeneous (see Figure 1b). There is increased risk of malignant transformation

with pitted, non-homogeneous lesions. Additionally, speckled leukoplakia often have

microscopic dysplasia and malignancy 16. Erythroplakias (see Figure 1c) are red and a rarer

precancerous lesion. These are typically thought to be of highest risk for cancer development,

with a 90% malignant transformation rate 10. Typically, dysplasia is graded based on

smoothness of leukoplakias and erythroplakias or based on the depth of architectural changes

within the epithelium.

Page 13: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

3

Figure 1: The different stages of disease progression with diseased regions circled: A) a typical

homogeneous leukoplakia, B) a typical speckled leukoplakia, C) a typical erythroplakia and D)

typical late stage oral carcinoma 11, reproduced with permission.

Globally, oral cancer incidence increases with age, with 98% of oral cancer patients in developed

countries being over forty years of age 1. This is because oral cancer occurrence is highly

correlated to a number of risk factors which have a mutagenic effect inducing genetic, epigenetic

and metabolic changes with increased exposure 17. The most significant of these risk factors are

heavy alcohol and tobacco consumption, particularly when used in conjunction; they have a

supermultiplicative effect in the mouth 1,18. Poor diet, exposure to UV light, human papilloma

virus, a slight familial correlation and transplants, where patients are immunosuppressed, are also

thought to be risk factors for oral cancer 19-27.

OSCC originates by “field cancerization”, a process where a carcinogenic agent preconditions an

area of epithelium. By definition, this carcinogenic agent is active with sufficient intensity over

long enough time that a change towards cancer of this preconditioned epithelium is inevitable 10.

Specifically, when examining tumors with a diameter less than 1 cm, separate foci of cancer can

Page 14: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

4

be found that later coalesce as they grow. The time between application of the carcinogen and the

development of cancer is varied. The field can break down at multiple points, thus multiple oral

cancers can arise from independent cell clones. This means that there are multifocal areas of

precancer that lead to OSCC 10. The specific mechanism is unclear, but could be due to the

spread of clonal stem cells, saliva inoculation or the presence of undetectable invasiveness. Field

cancerization helps to explain the high local-recurrence rate associated with oral cancer and must

be considered when developing treatments for OSCC.

1.1.2 Current Treatments

The goal when treating OSCC is complete tumor eradication, while either restoring or preserving

both anatomy and function and limiting recurrence or a second primary tumor 13,28. While

overall survival and disease-free survival are typical measures of success of cancer treatment,

patient quality of life is of great concern, particularly in a region of such functional and cosmetic

importance 29. Functionally, the oral cavity performs a variety of complex tasks, including

speech, chewing, tasting, swallowing and salivation 13,29. Aesthetically speaking, reconstruction

is very common, but can still lead to significant disfigurement.

The mainstay of OSCC treatment is surgical excision, but radiation therapy and chemotherapy

are also used in its treatment, either alone or in combination 5,13,30. Single modalities are

generally used in early stages of the disease, typically surgery or radiation therapy, and

combination therapies are used with more advanced disease 5,13,30. The type and the extent of

treatment are determined by tumor characteristics, side effects and co-morbidities, as described

in greater detail in Table 1.

Page 15: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

5

Table 1: Treatment modalities for oral carcinoma

Treatment

type

Applicable stage of

disease

Stand alone or

combination therapy

Method of action

Surgery Early stage and late

stage

Primary treatment modality Remove malignant tissue,

leaving remaining normal tissue

functionally intact

Radiation

therapy

Early stage and late

stage

Sometimes used as stand

alone, but most commonly

combination

Destroy DNA of malignant cells

in localized region, impacting

target and adjacent normal

tissue

Chemotherapy Late stage Combination therapy Inhibit rapidly dividing cells,

managing spread and metastasis

1.1.2.1 Surgery

Surgery, the most common form of treatment, attempts to remove all abnormal tissue while

leaving histologically normal tumor margins and trying to preserve tissue anatomy and function

31-34.

Negative margins are crucial in effectively treating the disease. Patients having positive margins

are expected to have double the rate of local recurrence, compared to patients with pathologically

negative margins 35. Consequently, one to two centimeter margins of tissue that appears normal

should be removed to prevent microscopic disease from being left behind 13. As the margins are

of the greatest concern, and of high prognostic significance, in surgical treatment of OSCC,

intraoperative frozen tissue assessment of both mucosal and deep margins is often performed 36.

However, accurately identifying any positive surgical margins has been a source of treatment

failure; in a study of 25 patients, all of whom were diagnosed with pathologically negative

Page 16: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

6

surgical margins during intraoperative frozen tissue assessment, five of the patients had biopsy-

proven local recurrences within 7 months of their initial surgical resection 37. Many studies are

looking at molecular staging, for example, detecting p53 mutations common in many head and

neck cancers, and preliminary clinical results show promising results 37,38.

The surgical technique varies based on lesion size and access. With smaller tumors, surgeons can

often remove the tumors from within the oral cavity, but if access is limited or the tumor is very

large, the surgeon might have to approach from outside the cavity; removing soft tissue and bone

7. Cheek flaps are often removed to access the floor of the mouth and the mandible or they may

approach from under the eye to access the maxilla 34. In fact, often the mandible and/or maxilla

need to be fully excised, facilitating sufficient access to the oral cavity 34,39. More advanced

stages of disease are often associated with lymph node invasion and may require radical neck

dissection 28,31,34,39. Like treatment of the primary site, the extent of neck dissection is related

to the extent of lymph node invasion: size, site, number 28,34,39.

Following surgical excision, reconstruction surgery may be required to restore loss of functions

and/or to repair cosmetic defects 13,34. Small defects are often covered with split-thickness

grafts and larger defects with forearm grafts 40. Mandibular reconstructions, following removal

of a portion of the mandible, are most commonly performed with bone from the patient’s fibula

13.

In addition to possible neck dissection, advanced OSCC is also typically treated with

combination therapy: surgery and radiotherapy or surgery, radiotherapy and chemotherapy 11,13.

Page 17: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

7

1.1.2.2 Radiation Therapy

Radiation therapy (RT) aims to damage the DNA in malignant cells within a small localized

region while preserving both function and anatomy of normal tissue in adjacent regions 28,30. In

early stage OSCC, RT, specifically external beam radiation therapy possibly with brachytherapy,

is offered as an alternative to surgical resection, when surgical access proves difficult or the

patient refuses surgery 30,40. In early stages of the disease, RT has similar 5-year survival rates

and a similar 37% rate of local recurrence when compared to surgery 41,42. That being said, it is

rarely used as a primary treatment modality, with its main purposes being prevention of

recurrences and eliminating remnant tumor following an incomplete resection 11.

Patients with large tumors and those with positive margins post-resection are commonly treated

with postoperative RT. RT is typically administered postoperatively so as to minimize risk of

infection and maximize postoperative healing 13,28. The treatment is planned to maximize dose

in the region surrounding the tumor, where there is the greatest likelihood of genetic, epigenetic

and metabolic changes that could lead to a secondary malignancy 30. The standard RT protocol

involves administration of about 60 Gy total given as 2 Gy daily from Monday to Friday over 6

weeks 13. Avoidance of breaks is critical as several studies have shown there to be a significant

correlation between timing of treatment without breaks, and locoregional control and overall

survival rates 43.

Recent studies show that the use of altered fractionated treatments and combination

chemotherapy both improve survival, although neither have yet been widely adopted 44-46. The

most recent alteration to RT protocol involves the addition of concurrent chemotherapy to the

treatment protocol. Meant to improve radiosensitization, it is currently being explored in the

hopes that it will improve malignant cell sensitivity to RT compared to surrounding healthy

tissue 47,48. However, for patients with low risk of post-surgery recurrence, concurrent

chemotherapy is of no benefit 49.

Page 18: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

8

1.1.2.3 Chemotherapy

Chemotherapy (chemo), systemic anticancer drug therapy, is not a curative single modality for

OSCC. Chemo was previously thought to mainly be of use as a palliative treatment, however, it

is becoming an important adjunctive therapy in the battle against OSCC 40. Chemo targets

malignant, abnormal and rapidly dividing cells in the hopes of preventing spread and targeting

any metastases 50.

While chemo can be used before surgery, concurrently with postoperative RT, or after the

completion of both surgery and RT, the most promising results are associated with concurrent

RT 51-53. The combination of RT and chemo has proved to increase RT efficacy leading to

better locoregional tumor control, reduced mortality and improved survival 47,54,55.

1.1.2.4 Treatment Side Effects

OSCC patients can have a wide variety of side effects from the various treatment options and

morbidities must be considered when determining the treatment protocol for each patient, with

common side effects summarized in Table 2.

Page 19: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

9

Table 2: The side effects associated with the different treatment modalities for OSCC 7,13.

Treatment modality Short-term side effects Long-term side effects

Surgery Difficulty swallowing and speaking

Wound infection

Fistula

Functional problems

Cosmetic defects

Tissue and bone loss

Nerve pain

Radiation therapy Loss of energy

Dermatitis

Mucositis

Xerostomia

Altered taste

trismus

Persistent xerostomia

Fibrosis

Osteoradionecrosis

Increased risk of tooth decay and

periodontal disease

Hypothyroidism

Laryngaeal edema

Chemotherapy Nausea and vomiting

Diarrhea

Stomatitis

Bone marrow suppression

Neuropathy

OSCC, and its subsequent treatment, has profound impact on patient quality of life, in the long-

term and short-term, both physically and psychologically. Like other cancer survivors, patients

who have undergone OSCC treatment have a risk of recurrence, of secondary primary tumors

and can suffer from pulmonary, renal, cerebrovascular and cardiovascular complications 56,57.

Additionally, patients who have undergone surgery may suffer from impaired swallowing, eating

and speaking as a result of their disease or treatment, which may become permanent. It is critical

that patients undergo rehabilitation to improve swallowing, and speech, and have dental

rehabilitation. This can either begin preoperatively or postoperatively, but is crucial in

minimizing any long-term deficits. Following treatment, nerve pain and loss of sensation can

Page 20: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

10

occur 58,59. Additionally, the anatomical changes associated with surgery can lead to cosmetic

defects requiring extensive reconstruction and even with reconstruction, permanent

disfigurement, limited movement and loss of function can result 60.

RT has both acute and late side effects. In the short term, RT results in mucositis in more than

50% of patients, can lead to a lack of energy, a loss of taste, radiation-induced dermatitis and

burn, xerostomia (dry mouth caused by a change in saliva function or damage to the salivary

glands) and trismus (muscle spasm leading to limited mouth opening) 50,61. For more than 60%

of patients, xerostomia is caused by radiation-induced damage to the salivary glands and remains

permanent, significantly increasing the risk of tooth decay and periodontal disease 13,61. In

addition to permanent xerostomia, RT can also lead to long-term side effects including

permanent mucositis, laryngeal edema and hypothyroidism (like xerostomia, induced by a

significant radiation dose to the larynx and thyroid, respectively), difficulty swallowing, tissue

fibrosis and osteoradionecrosis 40. Osteoradionecrosis is a non- or slow-healing bone damage

that can result soon after, or many years post-completion of RT and results from radiation-

induced hypoxia or hypovascularity to the bone 62,63. The mandible is particularly susceptible

to osteoradionecrosis. RT destroys malignant cells, but also neighbouring healthy tissue, to a

certain extent, and can damage normal cells, limiting blood supply to the bone. Eventually, the

mucosa overlying the bone falls away and a necrotic bone is left exposed. Additionally, RT is

also associated with the possibility of radiation-induced secondary cancers 13.

While surgery and RT are associated with localized side effects, chemotherapy involves the

systemic administration of anticancer agents and is, thus, associated with side effects across the

whole body. Many chemotherapy agents are associated with nausea, vomiting, diarrhea,

mucositis and suppression of the bone marrow, leaving patient susceptible to infections 61 and

others are associated with renal problems and neuropathy 64,65. Additionally, the

cardiovascular, cerebrovascular, pulmonary and renal complications that most cancer patients

suffer from are generally associated with chemotherapy agents 57. With chemo, most side effects

are dose-dependent and dependent on patient age and overall health, and all these factors must be

considered when the patient is treated 50. There are a number of potential avenues to reduce the

Page 21: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

11

treatment-associated side effects, namely better identification of tumor boundaries during

surgery, which would facilitate the complete and accurate removal of diseased tissue and,

requiring less use of RT and chemo.

1.2 Optical Imaging

While there have been significant improvements to the therapeutic management of OSCC, there

hasn’t been any significant improvement in the prognosis of the disease over the last few

decades, with 5-year survival rates ranging between 30 and 60% depending on the region of the

world 66. Additionally, there is a very high rate of recurrence of OSCC at the primary site, in

fact, one of the highest rates of recurrence across all cancers 67-69. As discussed, the primary

treatment modality of OSCC is surgical excision where physicians estimate tumor boundaries

based on tissue texture and their own experience. This is relatively inaccurate, with high rates of

failure to achieve negative margins, contributing to the poor survival rates and high rates of

recurrence. Conversely, excessive tissue removal leads to unnecessary functional and cosmetic

damage, detrimental to patient quality of life. There is a need for real-time visualization of tumor

boundaries during surgery, facilitating complete removal of diseased tissue, while sparing

healthy oral tissue. A relatively new approach to facilitate the detection of disease boundaries

involves the use of tissue optics and fluorescence.

1.2.1 Fluorescence

Photoluminescence is the process of absorption and subsequent re-emission of photons that

occurs in certain molecules upon excitation by light from their ground state 70. This absorption

of light raises the molecule from its ground state to its excited state. Its subsequent return to

ground state is accompanied by the spontaneous emission of light. The Jablonski diagram, in

Figure 2, is convenient for visualizing the various processes that are possible with

photoluminescence, most commonly fluorescence and phosphorescence 70.

Page 22: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

12

Figure 2: Jablonski diagram illustrating absorption, fluorescence and phosphorescence. Straight

arrows represent radiative processes and wavy arrows represent non-radiative processes and S

represents singlet states and T triplet states 70, reproduced with permission.

Upon energy absorption, an electron of a fluorescent molecule, a fluorophore, is excited into the

first excited singlet state (S1), a higher vibrational level of this singlet state, or into a higher

singlet state. With excitation into higher states, some of the absorbed energy is frequently shed

through processes including vibrational relaxation and internal conversion (IC) causing the

electron to fall into the first excited singlet state’s lowest vibrational energy level. From the first

excited singlet state’s lowest vibrational energy level, fluorescence (S1 S0) may occur, as

shown in Figure 2 70,71. Alternatively, from the excited singlet state’s lowest vibrational energy,

the electron may undergo intersystem crossing (ISC) to produce a triplet state, see Figure 2,

potentially leading to phosphorescence 72.

Page 23: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

13

Fluorescence involves short-lived emission, within nanoseconds of absorption, from the lowest

vibrational level of a singlet state and involving spin-paired electrons 70. Fluorescence is made

very powerful, and has become a popular technique for the diagnostics and image-guidance in

cancer treatment, due to the Stokes shift: light emitted upon return to a molecule’s ground state

has a longer wavelength than the light used to excited the molecule 70. This allows for the

visualization of fluorescence, by blocking out exciting light with filters.

Excited state fluorophores don’t always have to undergo fluorescence, they may proceed through

ISC, changing from an excited singlet state to an excited triplet state by a change in the

electron’s spin 70. Emission of light from this triplet state down to ground state is

phosphorescence (T1 S0). But phosphorescence often requires microseconds up to tens of

seconds to occur.

1.2.1.1 Quenching

Quenching refers to the reversible loss of a fluorescent signal due to intramolecular interactions

between a fluorophore and its environment, or a quencher molecule 71. The specific mechanism

through which quenching occurs varies with quencher and fluorophore and some examples

include electron transfer, self-quenching and contact quenching.

Of particular interest, Forster resonance energy transfer (FRET) is a quantum mechanical process

involving nonradiative energy transfer from an excited state fluorophore, known as the donor,

and another chromophore, known as the acceptor 73,74. Generally, the extent of FRET between

the donor and the acceptor is determined by the proximity of the donor to the acceptor and the

extent of spectral overlap in the emission spectrum of the donor and the absorption spectrum of

the acceptor 72. When FRET occurs, the donor’s excited state energy is transferred to the

Page 24: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

14

acceptor, should they be in close proximity, resulting in an increase in fluorescence intensity of

the acceptor and a decrease in the fluorescence intensity of the donor. That is, the donor becomes

optically silent 75.

1.2.2 COOLS Trial

Recently, there has been a change in the way carcinogenesis is viewed with respect to OSCC.

With the emergence of the field cancerization model, surgeons try to target at risk fields of

tissue. Traditionally, surgeons have removed lesions with abnormal tissue texture and

appearance, but altered tissue extends beyond these borders, that is, all diseased tissue is not

clinically visible. The high rates of recurrence associated with OSCC may be attributed to the

inability to accurately delineate the boundaries of diseased or altered tissue. Autofluorescence

imaging of oral tissue may provide a means to identify the abnormal tissue that cannot currently

be seen clinically.

Autofluorescence imaging of the oral cavity consists of application of higher-energy light to

excite specific fluorophores within the tissue, which then emit lower-energy light. In some

countries, autofluorescence imaging is considered the standard of care for detection of early

stage lung cancer. Additionally, it is being used in various stages of clinical trials for cervical,

skin, esophageal, bladder and colon cancer 76-79. Autofluorescence by endogenous fluorophores

can be used in the detection of malignancy because their optical properties can be correlated with

disease progression 80-82. Normal oral tissue will autofluoresce a pale green colour when

excited by blue light (400-450 nm), but autofluorescence decreases with malignancy and tissue

appears brown or black 83, illustrated in Figure 3.

Page 25: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

15

Figure 3: Patient with an occult recurrent carcinoma identified by the use of the

autofluorescence technique; A) under white light no lesion is visible on the tongue, B) When

irradiated with blue light, a region with decreased autofluorescence can be seen (note the arrows

in A and B indicate the correlation between the images), C) A biopsy from the anterior portion of

the tongue showed dysplasia and epithelial thickening, while from the posterior portion of the

tongue there was carcinoma in situ D) Moderate inflammation can be identified by increased

stromal activity, E) Severe inflammation can be identified direct visualization 84, reproduced

with permission.

The loss of autofluorescence is the direct result of both fluorophore alterations and structural or

morphological changes in tissue associated with the transition from normal tissue to malignant

tissue. When the oral cavity is illuminated with blue light, the resulting fluorescence in normal

tissue mainly originates from collagen, its crosslinks and elastin. These are located in both the

stroma and the basement membrane 80,84,85. In addition to changes in these components,

progression towards malignancy is also associated with changes to nuclear morphology,

epithelial thickness and tissue vascularization, all impacting absorption and scattering of light

and contributing to the fluorescence properties of diseased tissue 85.

Page 26: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

16

This technology was used in a 20-patient study where patients underwent surgical removal of

their tumors. The study found that autofluorescence can identify lesions that cannot be seen

under white light conditions 84. Additionally, all tumors displayed a loss of autofluorescence and

in 19 of the 20 tumors, this autofluorescence loss extended between 4mm and 25 mm beyond the

clinically visible lesion 86. From these promising preliminary results, a longitudinal study

involving 60 patients was performed. Patients were either treated with white light surgical

resection or with this optically-guided approach to surgery. At least twelve months after surgery,

none of the patients who had this optically-guided approach to surgery showed any signs of

recurrence. Seven of the 28 patients in the white light surgery group, however, have shown

recurrence with severe dysplasia or malignancy 87. The probability of local recurrence-free

survival is shown in Figure 4. This has led to a multi-center clinical trial called the Canadian

Optically-Guided Approach to Oral Lesions Surgical Trial, COOLS, currently underway.

Figure 4: The probability of local recurrence-free survival of the 60 patients enrolled in the

preliminary trial, with this optically-guided approach. There is a significant decrease in the

probability of local recurrence using this optically-guided approach when compared to white

light visualization alone 87, reproduced with permission.

Despite the very positive results associated with this autofluorescence-based technique, there are

still some potential issues with this technique. Several studies have shown that autofluorescence

is unable to differentiate between benign lesions, early stage disease and malignancy. This could

Page 27: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

17

lead to high rates of false positives and unnecessary removal of functionally and cosmetically

important tissue; an unacceptable morbidity.

1.3 Matrix Metalloproteinases

One of the defining characteristics of malignant lesions is their ability to invade tissues and

subsequently establish metastases 88. Such tumor invasion is a dynamic and complex process

where tumor cells must detach from their point of origin, traverse both the extracellular

membranes and basement membranes and enter into lymphovascular channels 89. Thus, the

extracellular matrix (ECM) of tumors and the non-malignant tumor-associated stroma cells are

critical in tumor progression 90. Proteolytic degradation of basement membrane and ECM

components requires specific proteases, including matrix metalloproteinases (MMPs). This

makes MMPs crucial to the regulation of tumor microenvironment. In fact, there is increased

MMP expression in most human cancers, when compared to normal tissue 91.

MMPs are a family of zinc-dependent endopeptidases, with more than 21 members of human

MMPs, collectively capable of cleaving almost any ECM component 89,91. They are either

secreted or anchored to the plasma membrane and are generally divided into five distinct groups:

collagenases, gelatinases, stromelysins, matrilysins and membrane-type MMPs 89,91. There is a

high degree of similarity between the enzymes within each group (roughly 80%) and between

enzymes in different groups (roughly 50%) 92. MMPs consist of an N-terminal propetide

domain, a catalytic domain, a hinge region and a C-terminal haemopexin-like domain, as seen in

Figure 5 93.

Page 28: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

18

Figure 5: The domain structure of MMPs where S is signal peptide; Pro is propeptide domain;

Cat is catalytic domain; Zn is zinc active site; Hpx is haemopexin-like domain 93, reproduced

with permission.

Under normal physiological conditions, MMP activities are tightly regulated. But a loss of MMP

regulation can lead to any number of diseases, including cancer, arthritis, atherosclerosis,

nephrititis and fibrosis 94. MMP regulation occurs through both transcriptional and post-

transcriptional mechanisms 95. This regulation is influenced by several factors including

hormones, growth factors, oncogenes and cytokines 89. Additionally, MMPs are synthesized as

zymogens, an inactive form, requiring extracellular activation. They remain inactive due to the

interaction between the zinc ion bound to the catalytic domain and a cysteine-sulphydryl group

in the propeptide domain. Activation occurs through proteolytic removal of the propeptide

domain 96. Most MMPs are activated extracellularly either by other activated MMPs or by serine

proteinases. There are exceptions, for example MMP-11, MMP-28 and MT-MMPs can be

activated intracellularly 96. Several inhibitors, including endogenous inhibitors, also affect MMP

activity. For example, in tissue fluids the main inhibitor of MMPs is 2-macroglobulin, a plasma

protein 97. Upon binding to MMPs, a 2-macroglobulin-MMP complex forms which then binds

to a scavenger receptor, facilitating clearance by endocytosis. Similarly, thrombospondin-2

complexes with MMP-2 leading to clearance by the same mechanism 98. The most well-studied

Page 29: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

19

endogenous MMP inhibitors, though, are tissue inhibitors of metalloproteinases (TIMPs) -1, -2, -

3 and -4. TIMPs reversibly inhibit MMPs and each TIMP displays different tissue-specific

expression and MMP inhibition 99.

1.3.1 MMPs in Cancer

Cancer progression is associated with several distinct changes to normal cell physiology, known

as the hallmarks of cancer: self-supported growth through its own growth signals; insensitivity to

signals inhibitory to growth; ability to escape apoptosis; ability to replicate infinitely;

angiogenesis; and tissue invasion and metastasis 88. Initially, it was thought that MMPs were

important exclusively in invasion and metastasis, but it has been found that MMPs play a pivotal

role in many of these hallmarks of cancer, summarized in Figure 6.

Page 30: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

20

Figure 6: The various functions of MMPs in cancer progression. Various MMPs play critical

roles in cancer cell growth and survival, in angiogenesis, in invasion, in differentiation and in the

body’s immune response to cancer 91, reproduced with permission.

MMPs have been shown to promote cancer cell proliferation through three different mechanisms,

illustrated in Figure 6a. MMPs release precursors of various growth factors including TGF-

100, MMPs allow any growth factors within the ECM to become bioavailable, for example

insulin growth factor (IGF) is released when MMPs cleave insulin growth factor binding protein

101,102, and MMPs can indirectly regulate proliferative signals through integrins 103.

MMPs also play a role in apoptosis, a process critical for cancer cells to avoid so they can

survive even with significant genetic instability. Specifically MMP-3, -7, -9, and -11 regulate

apoptosis and MMPs have both apoptotic and anti-apoptotic roles, illustrated in Figure 6b. When

Page 31: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

21

MMP-3 is overexpressed in mammary epithelial cells, it induces apoptosis, likely through

degradation of laminin 104,105. MMP-7 possesses both apoptotic and anti-apoptotic functions; it

releases membrane-bound FASL, a stimulator of death receptor FAS. Once released, FASL

either induces apoptosis of surrounding cells or decreases cancer cell apoptosis 106,107.

Conversely, MMP-7 also inhibits apoptosis by cleaving pro-heparin-binding epidermal growth

factor (pro-HB-EGF), producing HB-EGF, a promoter of cell survival 108. MMP-11 inhibits

cancer cell apoptosis, with its overexpression decreasing spontaneous apoptosis in xenograft

models 109. It has been hypothesized that MMP-11 inhibits apoptosis through the release of

IGFs, which act as survival factors 102,110.

MMPs appear to be important contributors to tumor angiogenesis, blood vessels sprouting from

previously existing blood vessels to support tumor tissue, with MMP inhibitors decreasing tumor

angiogenesis in various animal experiments 111-113. It is possible that MMPs contribute to

tumor angiogenesis purely through degradation of ECM, enabling endothelial cells to invade

tumor stroma. It has in fact been shown that cleavage of collagen type I is necessary for

endothelial cells to invade the ECM and for blood vessels to subsequently form 114. Specific

MMPs shown to regulate angiogenesis include MMP-2, -9, -14 and -19 114-117. Additionally,

MMPs produce anti-angiogenesis fragments, shown in Figure 6c. MMP-2, -3, -7, -9 and -12 can

cleave plasminogen to produce angiostatin 118,119 and MMP-3, -9, -12, -13 and -20 may

produce endostatin 120, both of which reduce endothelial cell proliferation 121,122.

In order for a cancer cell to metastasize, it must first cross the epithelial basement membrane,

invade tumor-associated stroma, enter blood vessels and/or lymphatics, extravasate and establish

proliferating metastases 91. The role of MMPs in this process has been investigated in vitro

through invasion assays and in vivo through xenograft metastasis assays. The overexpression of

MMP-2, -3, -13 and -14 or inhibition of TIMPS all promote the invasion of cell lines through

collagen type I 123-126. In a metastasis assay, the downregulation of MMP-9 in mice led to

significantly reduced numbers of colonies formed in mice lungs 127. Another important step in

invasion is migration and MMP-2 and -14 cleavage of laminin-5 reveals an important site that

triggers motility, shown in Figure 6d, 128,129. This cleaved form of laminin-5 is found in

Page 32: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

22

experimental tumors, and MMP-14 and laminin-5 are co-localized in human cancers 128,129.

For migration, a cancer cell must detach from its surrounding matrix and its neighbouring cells.

CD44 is cleaved by MMP-14, leading to the release of the extracellular domain, but when the

cleavage site is mutated, cell migration is inhibited, illustrating the importance of MMP

activation for tumor cell motility 130. MMPs, specifically MMP-3 and -7, also play a critical role

in the downregulation of cell-cell adhesion, freeing the cells to migrate, and in the transition from

epithelial cells to mesenchymal cells, separated by the basement membrane (see Figure 6e),

associated with a more aggressive malignancy 131,132.

1.3.1.1 MMPs in Oral Carcinoma

OSCC is a highly invasive malignancy that begins in the oral cavity epithelium and progresses

by degradation of both the ECM and the basement membrane. MMPs are critical to such ECM

and basement membrane remodelling, so it is expected that they play a role in OSCC. In fact,

there are many studies that have shown MMP-1, -2, -3, -9, -10, 11, and -13 to be expressed in

oral carcinoma and to be critical in the oral tumor progression 133-139.

MMP-1 mRNA has been found to be expressed in epithelial cells within tumor nests and in

surrounding tumor-associated connective tissue 133,134. MMP-2 has been found in in vivo

OSCC, within small tumor islands 135, but recently there has been evidence that it is derived

from tumor-associated stroma, mainly fibroblasts, rather than the cancer cells themselves

140,141. Additionally, MMP-2 expression in OSCC has been positively correlated to lymph

node metastases, illustrating the critical role that tumor microenvironment plays in tumor

invasion. Kusukawa et al. 136 found many oral carcinoma tumors that expressed MMP-3 at their

invasive margin and they later showed a positive correlation between MMP-3 expression and

tumor size, tumor thickness and invasion. MMP-9 has been shown to be expressed at the tumor-

stroma interface of malignant keratinocytes 142,143. The overexpression of MMP-10 and -11

has been correlated with tumor differentiation and overall invasiveness 141. MMP-13 is

expressed both by fibroblasts in the stromal region and by the invading tumor front 139,144.

Page 33: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

23

A recent study investigated the MMPs present in various patient oral tissue samples, including

samples with OSCC 145. The patient tissue samples were tested for expression of various

MMPs, including some (MMP-7, 19 and -26) never or rarely tested in OSCC. The expression of

most MMPs tested, MMP-2, -3, -7, -9, -10, -12, -13, -26, were upregulated in OSCC samples and

in 90% of the samples MMP-7, -9, -10, and -13 were expressed on the cancer cells, while

expression of MMP-2, -3 and -12 was mostly on the stromal cells 145, see Figure 7.

Figure 7: Immunohistochemistry was performed on formalin-fixed human oral carcinoma tissue

using mouse monoclonal antibodies demonstrating: A) MMP-7 expression is high in cancer cells

in OSCC samples, with occasional expression in stromal cells, B) MMP-13 is highly expressed

in invasive OSCC, C) MMP-9 is expressed in highly invasive OSCC 145, reproduced with

permission.

It is clear that MMPs play a critical role in OSCC and any number of them could provide a target

for specific treatment of OSCC.

1.4 Molecular Beacons

In the post-genomic era, there has been great focus on developing agents that can specifically

target particular genomic sequences or proteomic sequences overexpressed in diseased tissue,

including disease-associated enzymes like MMPs 146-148. These agents would facilitate specific

Page 34: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

24

treatment of diseased tissue, sparing healthy tissue. Molecular beacons are one such class of

probes with high sensitivity and selectivity biomolecular recognition 146.

Conventional molecular beacons (MBs) consist of a stable stem-loop oligonucleotide, forming a

hairpin, with a fluorophore on one end, typically the 5’-end, and a quencher on the other end, as

seen in Figure 8a 146,149. In the hairpin conformation, the MBs show no fluorescence, as the

close proximity of the quencher to the fluorophore facilitates efficient energy transfer from the

fluorophore to the quencher, quenching fluorescence 146. The MB is designed such that the

hairpin’s loop is complementary to the target of interest, that is, the genomic sequence

overexpressed in the diseased tissue of interest. In the presence of the target, the MB undergoes a

conformational change and the structure from hairpin to linear sequence. In this new linear

conformation, the fluorophore and quencher are too far from one another for efficient FRET and

the fluorophore is free to fluoresce. Consequently, the conformational change is characterized by

a sharp increase in fluorescence, illustrated in Figure 8b.

Figure 8: A) The conventional molecular beacon with a hairpin stem formed by complementary

sequences and when the probe hybridizes with its target, the stem loop structure cannot exist,

resulting in a conformational change and B) MB specificity is illustrated with the MB incubated

with complementary target (a) leading to a strong increase in fluorescence intensity, whereas a

single mismatch (b) and a single deletion (c) led to no significant fluorescence increase 146,

reproduced with permission.

Page 35: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

25

Conventional MBs display high specificity, capable of differentiating between two DNA

sequence targets with as little as one differing nucleotide, as demonstrated in Figure 8b

146,150,151. Additionally, with the appropriate selection of fluorophores and quenchers, very

high signal-to-background ratios (with 200-fold increases often occurring under optimal

conditions) are possible, as they are non-fluorescent in their native state 152. Both of these have

led to MBs being used for a number of bioanalysis applications including DNA arrays and

monitoring of real-time polymerase chain reaction 151,153,154.

1.4.1 Peptide-Based Molecular Beacons

While MBs were originally designed to specifically recognize nuclei acid sequences, they have

also been designed to be specifically activated by certain protein sequences 148. These can be

useful for imaging purposes, with MB activation and fluorescence confined to tissues

overexpressing the protein target of interest and remaining quenched in all other tissues. These

peptide-based MBs are cleaved by proteases, catalytic enzymes that hydrolyze peptide bonds

155,156. MBs have been widely explored for cancer imaging, with their increased protease

expression, and typically fall into one of three categories: classic peptide probes; polymer-based

peptide beacons; nanoparticle-based peptide beacons.

Classic peptide beacons consist of a fluorophore and quencher molecule conjugated to a short

protease-cleavable linker. Due to the close proximity of the fluorophore and quencher molecules,

there is no fluorescence when the linker remains intact. Enzymatic cleavage of the MB by a

protease leads to an increase in fluorescence by the fluorophore. These classic MBs have been

used in vivo for imaging of tumors (providing image guidance and the ability to discern healthy

and tumor tissue), imaging of tumor-associated protease activity, assessment of protease inhibitor

therapies and to provide greater understanding of the role proteases play in tumorigenesis,

angiogenesis and metastasis 158,159.

Page 36: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

26

However, there are a few issues associated with these classic peptide MBs. Small peptides tend

to have poor pharmacokinetic properties, with the main concern being poor tumor accumulation

caused by their short plasma half-life and limited tumor penetration 160. Consequently, a number

of strategies were developed to try and improve peptide-based MBs for in vivo applications,

namely conjugation to a polymer and encapsulation in nanoparticles 158,160-162.

1.4.2 MMP-Targeted Molecular Beacons

With heightened MMP expression in almost every human cancer and given its expression is

correlated with advanced tumor stage, increased invasion and metastasis, and shorter survival 91,

it seems only natural that MMPs have become a protease target for molecular beacons.

Our lab previously designed a MB to be cleaved in the presence of MMP-7 163, targeted because

of its epithelial origin and high expression in a number of malignancies 164,165. The

fluorophore, pyropheophorbide (pyro), was selected for its near infrared emission and high tumor

affinity 166. Near-infrared (650-900 nm) fluorescence imaging is considered optimal due to the

greater tissue penetration, as a result of low tissue absorption, and low autofluorescence 167,168.

Pyro was separated from the quencher, black hole quencher 3 (BHQ3), via a short MMP-7-

cleavable peptide sequence, GPLGLARK; specifically cleaved between G and L, indicated in

italics 166. We hypothesized that upon entrance into MMP-7-expressing cells, MMP-7 would

specifically cleave the peptide sequence, separating pyro from BHQ3, thus restoring pyro’s

fluorescence.

MMP-7-triggered MB activation was first validated in solution studies. The MB was incubated at

37C, with MMP-7, with MMP-7 and its inhibitor, with MMP-2 and an uncleavable MB was

incubated with MMP-7 and the fluorescence intensity was monitored over time, see Figure 9. An

immediate increase in fluorescence intensity was observed in the MB incubated with MMP-7,

but no fluorescence increase was observed in the other samples. Samples were subsequently

Page 37: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

27

analyzed by high-performance liquid chromatography (HPLC) and this confirmed that MMP-7

was able to specifically cleave the MB at the known cleavage site between G and L 169,170.

Figure 9: Validation of the MB's MMP-7 specificity: the fluorescence intensity was monitored

over time with MMP-7, MMP-7 and an inhibitor, MMP-2 and of an uncleavable MB, C-PPB,

and MMP-7 163, reproduced with permission.

MMP-7 activation of the MB was then assessed in cancer cells in vitro. Confocal fluorescence

microscopy studies were performed using KB cells (MMP-7 positive) and BT20 cells (MMP-7

negative) 171 incubated with either the MB, or its uncleavable counterpart. The confocal images

show a strong fluorescence signal in KB cells incubated with the MB (see Figure 10a 2), but

minimal fluorescence is observed in BT20 cells incubated with the MB or either cell line

incubated with the uncleavable beacon (see Figure 10a 3,5,6).

Page 38: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

28

Figure 10: MMP-7-specific activation validation in vitro with fluorescent and brightfield

confocal images of 1) KB cells alone, 2) KB cells with 60 uM MB, 3) KB cells plus 60 uM

uncleavable beacon, 4) BT20 cells alone, 5) BT20 cells with 60 uM MB, 6) BT20 cells with 60

uM uncleavable beacon 163, reproduced with permission.

In vivo validation was also performed. A mouse bearing a KB tumor had a tail vein injection of

80 nmol of MMP-7-specific MB and its fluorescence intensity was monitored over time, as

shown in Figure 11. There is no fluorescent signal prior to injection and there is a strong increase

in fluorescence in the tumor following MB injection, reaching maximum intensity by 3 hours

post-injection, as seen in Figure 11, illustrating MMP-7 activation of the beacon in vivo.

Subsequent studies have been performed with other cell lines and more clinically relevant animal

models, including vertebral metastases and femur metastases, all showing this MB to be highly

effective and specific in identifying and combating malignancy 172,173.

Page 39: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

29

Figure 11: In vivo images of a mouse bearing a KB flank tumor: a) prescan image, b) 10 min

post-i.v. injection, c) 3 hrs post-i.v. injection, d) 5 hrs post-i.v. injection 163, reproduced with

permission.

Additional studies were also performed examining the specificity of the beacon to MMP-7,

compared to other MMPs expressed in various malignancies, as MMPs have overlapping

cleavage sites and often MMP-specific peptide sequences can be specifically cleaved by multiple

MMPs. The beacon was incubated in solution with MMP-9, -8, -7, -3, -1, -13, -12, -11, -10,

respectively, for 48 hours and then analyzed with the HPLC. Based on the HPLC traces, shown

in Figure 12, MMP-3, -7, -9, -10 and -12 all show specific beacon cleavage, all of which are

implicated in OSCC.

Page 40: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

30

Figure 12: The HPLC traces of beacon incubated with various MMPs for 48 hours; the traces

show that MMP-3, -7, -9, -10, and -12 all specifically cleave the beacon 173, reproduced with

permission.

Page 41: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

31

Chapter 2 Topically Applied Molecular Beacons for Fluorescence-Guided

Resection of Oral Carcinoma

2.1 Introduction

Optical imaging-based approaches, specifically using autofluorescence, have been widely

investigated for the identification of a variety of malignant diseases including lung, skin, cervical

and esophageal cancers 76-78. This technique has also recently been evaluated for use in the

detection of oral carcinoma. The 5-year survival rate for patients with oral carcinoma is as low as

30% 174. Additionally, it has one of the highest rates of recurrence of all cancers, mainly

attributed to the inability to accurately identify tumor boundaries. An ongoing clinical trial, the

COOLS study, uses autofluorescence to aid in the identification of oral carcinoma during

surgical resection. Preliminary results from this trial show decreased recurrence rates using this

optically-guided approach when compared to white light surgery, the standard of care for

patients with oral carcinoma, alone 84. However, autofluorescence cannot discern malignant

lesions from benign lesions or very early stage premalignant lesions, potentially leading to high

rates of false positives and the unnecessary disfiguration of patients. As an alternative, optical

imaging with exogenous contrast agents, offer promise for improvement in outcome.

Our group had previously reported molecular beacons for specific fluorescence imaging of

malignant lesions 163. These beacons consist of a fluorophore conjugated to a quencher

molecule by a short, disease-specific peptide linker (Figure 13). The beacons were designed for

the peptide linker to be specifically cleaved by enzymes highly expressed in diseased tissue, with

only minimal expression in healthy tissue. Consequently, in healthy tissue the linker would

remain intact and should the beacon be irradiated with excitation light, there would be no

fluorescence, as the fluorophore would be quenched. In diseased tissue, though, the enzymes

Page 42: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

32

capable of cleaving the linker were prevalent. They cleaved the linker and the fluorophore was

unquenched. Thus, there was fluorescence when beacons in diseased tissue were illuminated

with excitation light.

Figure 13: Molecular beacons consist of a fluorophore (F) conjugated to quencher (Q) via a

short disease-specific linker. In healthy tissue, the linker remains intact and no fluorescence is

observed, but in diseased tissue, the linker is specifically cleaved, activating the beacon and

allowing for fluorescence.

The success of other optical imaging strategies combined with the high specificity associated

with molecular beacons, led to the concept of using molecular beacons to effectively identify

oral carcinoma for the purposes of real-time surgical guidance. The beacons had been used to

effectively identify regions of disease in other malignancies 173,175, but its ability to identify

regions of oral carcinoma is dependent upon its linker being specifically cleaved by enzymes

much more highly expressed in oral carcinoma than in surrounding healthy oral tissue. MMPs

are proteolytic enzymes that have long been associated with various stages of tumor progression

and have more recently been associated with oral carcinoma 136,141. With the overexpression of

MMPs in oral carcinoma, it was thought that a beacon that was specifically cleaved by MMPs

could serve as an effective probe for the detection of oral carcinoma at the molecular level,

Page 43: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

33

providing faster, more effective delineation of oral carcinoma boundaries. This could facilitate

and complete and accurate removal of diseased tissue, while sparing healthy tissue.

The potential of an MMP-specific molecular beacon as an image-guided approach to surgery is

evaluated herein. The molecular beacon used consists of pyropheophorbide (pyro) is linked to its

quencher molecule black hole quencher 3 (BHQ3) via an MMP-cleavable peptide sequence,

GPLGLARK and is referred to as PPMMPB. The ability of PPMMPB to effectively and specifically

activate and fluoresce in oral carcinoma is evaluated, providing the first step towards the

implementation of PPMMPB as a strategy to identify oral carcinoma for the purposes of real-time

surgical guidance

2.2 Materials and Methods

PPMMPB Synthesis: The PPMMPB consists of pyro conjugated to black hole quencher 3 (BHQ3)

via an MMP-cleavable peptide sequence, GPLGLARK, with the cleavage site is indicated in

italics. This PPMMPB was synthesized as described previously 163. The positive control PPMMP

was synthesized by the same standard protocol, but without the addition of BHQ3.

Cell Line: UM-SCC-1, a human oral carcinoma cell line, was kindly provided by Dr. Thomas

Carey at the University of Michigan, USA 176. The cells were grown and maintained in

Dulbecco’s Modified Eagles’ Medium (D-MEM) supplemented with 10% fetal bovine serum,

100 U/ml penicillin, 100 g/ml streptomycin, 100 M non-essential amino acids and 2 mM L-

glutamine, at 37C in a humidified incubator with 5% CO2.

Page 44: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

34

In vivo Xenograft Model: All animal studies were carried out with institutional approval

(University Healthy Network, Toronto, Canada). Adult male athymic nude mice (athymic,

Charles River, Wilmington, USA) were inoculated with 1x105 UM-SCC-1 cells in 20 L of

media at the tip of the tongue. All animals were maintained in pathogen-free conditions in

autoclaved microisolator cages. Two weeks after injection, the tumors reached 2-3mm in

diameter. A 50 nmol dose of either PPMMPB or PPMMP was formulated in 20 L of aqueous

solution with 5% dimethyl sulfoxide (DMSO, Sigma Aldrich) and 1.5% Tween-80. Under

general inhalation anaesthesia (isofluorane in oxygen), the tongues were imaged using an in-

house fluorescence imaging endoscopy system (650 20 nm excitation, 700 25 nm detection,

various integration times, set manually), similar to those described previously 177 and illustrated

in Figure 14. The PPMMPB or PPMMP was then injected into the tumor and surrounding tongue

tissue using a 31G needle. 15 minutes post-injection the tongues were imaged again, using the

same imaging system.

Figure 14: In-house fluorescence imaging system; A) the various components to the

fluorescence imaging system, adapted from 177 with permission, B) The imaging set up with an

animal under the endoscope, in position for imaging.

Page 45: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

35

Ex vivo Xenograft Studies: Tongues were harvested from the mice immediately following

fluorescence imaging. They were snap-frozen and stored at -80C. Frozen sections were cut on a

cryostat (6m slices). The slices were immersed in phosphate buffered saline (PBS) for 5

minutes, dried and 5 L of mounting solution with DAPI (4’,6-diamidino-2-phenylindole from

Vector Laboratories Inc.) was added as a nuclear stain. The sections were covered with a

coverslip and imaged by confocal microscopy (Olympus, 460 nm excitation, 690 40 nm

emission).

In vivo Hamster Model: Male 6-8 week old hamsters (Syrian, Harlan, Indianapolis, USA) were

used as a model to mimic oral carcinoma in humans. Under general inhalation anaesthesia, 0.5%

DMBA (7,12-dimethylbenz(a)anthracene) in DMSO was applied to both sides of the hamster

cheeks three times every week over the course of 16-20 weeks. KimWipes (Kimberly-Clark)

were packed into the cheek pouch prior to application, to minimize any spills down the throat,

and a non-absorbent material (5mm in diameter) was used to apply the DMBA to their cheeks for

6 seconds at a time. The KimWipes were removed after the application was completed. After 16-

20 weeks of application, the tumors reached 5-10 mm in size. A 50 nmol dose of PPMMPB or

PPMMP was formulated in 100 L of aqueous solution with 5% DMSO and 1.5% Tween-80.

Under general injectable anaesthesia (80 mg/kg ketamine and 5 mg/kg xylazine), a 90-95%

resection of the tumor of interest was performed (shown in Figure 15), to mimic the clinical

scenario. After this initial resection, the cheek was imaged using the in-house fluorescence

imaging system (prescan). Then either the PPMMPB or PPMMP solution was applied topically to

the tumors and surrounding healthy cheek tissue for 15 minutes. After 15 minutes, the solution

was removed and the cheek was washed with sterile saline solution and imaged again with the

in-house imaging system. Hamsters were euthanized by pentobarbital overdose (Euthanyl®

Bimeda-MTC Animal Health Inc., Cambridge, ON, Canada).

Page 46: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

36

Figure 15: The experimental protocol for all hamster cheek studies involved beginning with

tumor-bearing hamster cheeks (A) and 90-95% of the tumor would be resected (B) leaving

remnant tumor tissue, illustrated by the arrows in B, and fluorescence imaging would be used to

try and detect this tumor tissue.

Ex vivo HPLC studies: HPLC studies to confirm the specificity of PPMMPB activation were

performed on hamster cheek pouch tumors. Whole tumors were either topically incubated in

PPMMPB solution in vivo (while still in the hamster cheek) or ex vivo (immediately following

surgical removal of the tumor) for 15 minutes. 200 L of DMSO was added to the tumors

(enough to ensure they were completely submerged) to extract any PPMMPB for 2 hours. The

extracted solution was then run through the HPLC-MS.

Ex vivo Hamster Cheek Studies: Whole cheeks were harvested from the hamsters immediately

following fluorescence imaging. They were snap-frozen and stored at -80C. Frozen sections

were cut on a cryostat (6m slices). The slices were immersed in PBS for 5 minutes, dried and

30 L of mounting solution with DAPI (4’,6-diamidino-2-phenylindole from Vector

Laboratories Inc.) was added as a nuclear stain. The sections were covered with a coverslip and

imaged by confocal microscopy (Olympus, 460 nm excitation, 690 40 nm emission).

Page 47: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

37

2.3 Results

2.3.1 In vivo validation of molecular beacon specificity in a murine head and neck cancer model

The PPMMPB activation in a murine head and neck cancer model was examined. Following

injection of 50 nmol of PPMMPB into the tongue tumors and surrounding healthy tongue tissue,

fluorescence signal was detected within the tumor but not in surrounding healthy tissue,

indicating selective PPMMPB activation by cancerous tissue (Figure 16Av). To further validate

the specificity, the same was performed with PPMMP. Tongues treated with PPMMP became

fluorescent in tongue tumor as well as in healthy tongue within 15 minutes of injection (Figure

16Bv). This was anticipated as, with no quencher present, no specificity should be seen with

PPMMP. When tongue tumors were treated with a negative control, no pyro was present, no

fluorescence observed over the 15 minutes. Additionally, no fluorescence was observed when

PPMMPB was injected into healthy tongues.

Page 48: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

38

Figure 16: Representative xenograft model fluorescence images with PPMMPB and PPMMP

demonstrating tumor-associated fluorescence and whole tongue fluorescence, respectively. A)

Tongues injected with PPMMPB: i) pre-injection colour white light image, ii) pre-injection

monochrome white light image, iii) pre-injection fluorescence image, iv) 15 min post-injection

monochrome white light image, v) 15 min post-injection image showing fluorescence localized

to the tip of the tongue, tumor tissue. A) Tongues injected with PPMMP: i) pre-injection colour

white light image, ii) pre-injection monochrome white light image, iii) pre-injection fluorescence

image, iv) 15 min post-injection monochrome white light image, v) 15 min post-injection image

showing fluorescence throughout tongue, in healthy and tumor tissue (n=3 animals).

The fluorescence images obtained within 15 minutes post-tongue injections suggested that

specific PPMMPB activation was obtained within the tongue tumors, but this needed to be

validated with histology and fluorescence microscopy. Confocal images of the frozen sections of

harvested tongues confirmed that the PPMMPB activation and fluorescence was confined within

cancerous tissue (Figure 17A). No activation was observed in healthy tissue, whereas in tongues

treated with PPMMP, there was fluorescence in both tumor and healthy tissue alike (Figure 17B).

Page 49: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

39

Figure 17: Representative hematoxylin and eosin (i) and confocal images (ii) where red is

activated beacon fluorescence and blue is Dapi stain. A) tongue injected with PPMMPB, validating

that PPMMPB activation leading to fluorescence is confined to cancerous tissues, B) tongue

injected with PPMMP, confirming that injection of PPMMP leads to fluorescence within cancerous

and healthy tissue alike, C) tongue injected with negative control, confirming that no

fluorescence is due to the aqueous solution in which PPMMPB is brought up (n=3 animals).

These data confirm the specific activation of PPMMPB and subsequent fluorescence within tumor

tissue in vivo in a murine head and neck cancer model

2.3.2 In vivo activation and specificity of the molecular beacon in a clinically relevant animal model

While there was great promise to the in vivo xenograft model data, in order to validate the

effectiveness of MMP-specific molecular beacons for this clinical application, a study in a more

clinically relevant animal model was required. Hamster cheek pouch oral tumors are the model

of choice when studying oral carcinoma as the disease progression closely mimics that in

humans 178, and so were used to evaluate MMP-specific molecular beacons. Clinically, I

envision that surgeons would first remove all oral tissue they believe to be diseased under white

light conditions before the MMP-specific molecular beacon would be topically applied to

identify any remnant disease. With this in mind, once the oral tumors were induced by DMBA

(over the 16-20 week period), 90-95% of the tumors were resected. After this resection, PPMMPB

was topically applied to the cheeks for 15 minutes and the cheeks imaged. There was some

Page 50: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

40

background autofluorescence observed prior to PPMMPB application, but it does not allow

diseased and healthy tissue to be discerned. Following the topical application, there was a

significant increase in tumor-associated fluorescence. The resection area, intentionally left

remnant tumor, and an additional tumor also left intentionally could all be easily identified

(Figure 18). Diseased tissue could be visualized by fluorescence, whereas the surrounding

healthy tissue showed no fluorescent signal.

Figure 18: Representative images of a hamster cheek treated with PPMMPB. A) Hamster cheek

following 90-95% resection and before any PPMMPB is applied: i) colour white light image, ii)

monochrome white light image, iii) fluorescence image showing some background fluorescence

is detected, but not sufficient to accurately identify lesions. B) Hamster cheek following 15 min

topical application of PPMMPB: i) colour white light image, ii) monochrome white light image,

iii) fluorescence image showing that the resected area, the remnant tumor and an additional

tumor intentionally left behind can all be easily visualized (n=5 animals).

To examine the beacon’s tumor specificity more closely, tumor-bearing hamsters were also

treated with PPMMP or with a negative control, containing no fluorophore. Additionally healthy

hamster cheeks underwent the same surgery as tumor-bearing cheeks would and were similarly

treated with PPMMPB. Similarly to the xenograft results, hamster cheeks treated with PPMMP

Page 51: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

41

showed no specific fluorescence: healthy and tumor tissue alike fluoresced after the 15 minute

application (Figure 19A). There was no observed fluorescence in the tumor-bearing cheeks

treated with the negative control (Figure 19B) or in the healthy cheeks treated with PPMMPB

(Figure 19C).

Figure 19: Representative images of various control hamster cheeks: A) PPMMP-treated, B)

negative control-treated, C) healthy cheek having undergone surgery and treated with PPMMPB

where i) prescan colour white light image, ii) prescan monochrome white light image, iii)

prescan fluorescence white light image, iv) post-application colour white light image, v) post-

application monochrome white light image, vi) post-application fluorescence image. Looking at

Avi) it is clear that both healthy and cancerous tissue is fluorescent post-PPMMP application. In

Bvi) and Cvi) it can be seen that there is no fluorescence associated with the negative control or

with PPMMPB applied to surgically-treated healthy tissue, respectively (n=5 animals).

To ensure that the observed fluorescence in tumor-bearing cheeks treated with PPMMPB was

indeed the result of tumor-specific PPMMPB activation, the cheeks were imaged ex vivo. Using

confocal microscopy, activated PPMMPB is confined to tumor tissue (Figure 20); healthy

surrounding tissue does not activate PPMMPB. Consequently, fluorescence is confined and

specific to oral carcinoma tissue. It is also clear, though, that PPMMPB displays limited depth

penetration: it is only present at the outermost layers of the tumor tissue (Figure 20B and D).

This does not prevent identification of the boundaries of the disease, critical for complete

Page 52: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

42

surgical removal of the disease. Additionally, oral carcinoma is characterized as being much

greater in linear extent than depth 10,174, meaning identification of lateral boundaries is the

most critical metric of successful surgical resection.

Figure 20: Representative hematoxylin and eosin and confocal images, where red is activated

beacon fluorescence and blue is Dapi stain. A) hematoxylin and eosin image of a depth-wise

slice through the tissue and B) corresponding confocal image showing activated PPMMPB signal

only in tumor tissue, at the outermost layer. C) hematoxylin and eosin image of an en-face slice

of tissue and D) corresponding confocal image showing activated PPMMPB at the outer layer of

tumor tissue (n=5 animals).

Similarly, the PPMMP and negative control treated tissue, as well as the surgery-treated healthy

cheeks were also imaged using confocal microscopy, again validating the in vivo fluorescence

data. PPMMP-treated cheeks show no specificity: PPMMP is equally distributed within healthy and

tumor tissue alike (Figure 21A). There is also no activated beacon signal within tissue treated

with the fluorophore-lacking negative control (Figure 21C) or the surgery-treated healthy cheek

treated with PPMMPB (Figure 21B).

Page 53: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

43

Figure 21: Representative hematoxylin and eosin (i) and confocal images (ii), where red is

activated beacon fluorescence and blue is Dapi stain. A) Tissue treated with PPMMP shows non-

specific fluorescence in healthy and cancerous tissue with limited penetration. B) Healthy cheek

treated that underwent surgery and were subsequently treated with PPMMPB shows no PPMMPB

activation or fluorescence, further validating PPMMPB’s tumor specificity. C) Tissue treated with

negative control shows no activated PPMMPB or fluorescence, confirming that fluorescence is not

the result of the aqueous solution in which PPMMPB is brought up (n=3 animals).

Combined, the fluorescence microscopy data confirms the tumor-specific PPMMPB activation

following a 15-minute topical application, which may facilitate the complete and accurate

identification of oral carcinoma in vivo.

2.3.3 Ex vivo HPLC Studies

Tumor-specific PPMMPB activation was confirmed by in vivo fluorescence and subsequent

fluorescence microscopy. In an attempt to further validate this and also examine whether the

Page 54: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

44

activation was in fact the result of MMP-specific cleavage, HPLC-MS of the PPMMPB extracted

from the hamster cheek pouch tumors was performed. Topical application of PPMMPB to the

tumors occurred both ex vivo and in vivo (but not jointly for the same sample). For the ex vivo

application, whole tumors were removed and incubated in PPMMPB and all beacon was extracted.

The corresponding HPLC traces confirmed tumor-specific cleavage: both intact PPMMPB and the

cleaved fragment (pyro-GPLG), a cleavage site characteristic of MMP 179, were observed in all

treated tumors (Figure 22A). The PPMMPB solution that had been applied to these tumors only

had intact PPMMPB (Figure 22B), confirming that PPMMPB cleavage and activation occurred in

the tumor.

Figure 22: Representative HPLC traces of A) beacon extracted from tumor showing that both

intact PPMMPB and PPMMP fragment are present within the tumor, B) beacon applied to the tumor,

where only intact PPMMPB is present, confirming PPMMPB activation occurs in the tumor (n=3

animals).

For the in vivo application, hamster cheeks were imaged prior to tumor removal, facilitating the

direct comparison between in vivo fluorescence images and PPMMPB activation. All tumors

showed some level of fluorescence and PPMMPB activation (Figure 23), with cleavage of the

peptide sequence occurring at the site characteristic of MMP-mediated activation 179. However,

the extent of activation varied between tumors, even within the same cheek (with every tumor

displaying 10-60% cleavage). The variation in activation can be attributed to any number of

reasons. The most likely explanations are that this variation is the result of tumor heterogeneity

or the tumors may not all have been fully submerged in PPMMPB solution, due to difficulty in

applying an aqueous solution to all of the tumors. In addition to the tumors applied with PPMMPB,

Page 55: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

45

surrounding tissue that had been immersed in PPMMPB was also analyzed and showed no

activated PPMMPB, although intact PPMMPB was present. This corresponds well with the

fluorescence images (Figure 23) and provides further support for tumor-specific molecular

beacon activation and its utility in the detection of oral carcinoma.

Figure 23: Representative images of a hamster cheek treated with PPMMPB and corresponding

PPMMPB cleavage or activation. A) Hamster cheek before any PPMMPB is applied: i) colour white

light image, ii) monochrome white light image, iii) fluorescence image showing some

background fluorescence, mainly caused by bacteria. B) Hamster cheek following 15 min topical

application of PPMMPB: i) colour white light image, ii) monochrome white light image, iii)

fluorescence image showing each of the three tumors become fluorescent, although to different

extents (n=3 animals).

2.4 Discussion

These data demonstrate the specific activation of PPMMPB, its enzymatic cleavage, potentially by

MMP, resulting in unquenching and subsequent fluorescence, in oral carcinoma tissue. Specific

PPMMPB activation was validated in a xenograft model for oral carcinoma (Figure 16 and Figure

17). Its activation was then validated with topical application in a hamster cheek pouch model: a

model that closely mimics the disease in patients (Figure 18 - Figure 21). HPLC was used to

confirm that oral carcinoma was in fact activating PPMMPB at a site characteristic of MMP-

Page 56: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

46

mediated cleavage (Figure 22 and Figure 23). The lateral boundaries of oral carcinoma can be

effectively identified in a very short time scale following topical application, allowing for ease of

integration into current clinical practices; and it would not significantly delay surgical resection.

The combination of PPMMPB’s high tumor specificity combined with this fast activation

following topical application can see PPMMPB fluorescence-guided resection improving treatment

of oral carcinoma; in combination with current clinical methods, improving survival and

recurrence rates.

That being said, there are limitations to this molecular beacon image-guided resection approach

to oral carcinoma treatment, as well as critical studies left to be performed. MMP expression is

heterogeneous across not only different oral carcinoma tumors, but also the various cells within a

given tumor. This differential expression will lead to varying fluorescence across tumors and

tumor cells. Should a tumor or certain cells within the tumor not express any of the MMPs

capable of activating the molecular beacon, they will not fluoresce or be identified by this

approach. This could lead to diseased tissue going undetected and consequently being left

behind. Additionally, it is critical to know the minimum concentration of activated beacon to see

fluorescence and the number of tumor cells required to activate the beacon.

On the opposite side of things, this MMP-specific molecular being is able to be cleaved by five

different MMPs (MMP-3, -7, -9, -10, -12) located within both tumor cells and stromal cells,

alike. This can lead to more than just tumor tissue being identified by this fluorescence-based

approach and, like the COOLS study, to some unnecessary loss of functionally and cosmetically

important tissue.

All things being considered, it is likely that this MMP-specific molecular beacon image-guided

treatment may need to be considered with other approaches. For example, image endogeneous

fluorophores characteristic with the disease using the COOLS autofluorescence-based approach

and then exogenous fluorophores with the MMP-specific molecular beacon fluorescence. They

Page 57: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

47

each provide different information about the tissue that can be combined to establish optimal

treatment protocol.

Page 58: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

48

Chapter 3 Future Directions, Research and Development

3.1 Introduction

Based upon the successes of optical imaging-based approaches for the identification of a number

of malignancies 77,78 and the high specificity provided by peptide-cleavable molecular beacons,

a molecular beacon approach to the detection and delineation of oral carcinoma for the purposes

of real-time surgical guidance was devised. Several members of the MMP family are highly

upregulated in oral carcinoma 135,136,141, and so an MMP-cleavable molecular beacon,

PPMMPB, was thought to be appropriate. This PPMMPB was designed to remain quenched in

healthy tissue, but MMP would specifically cleave its peptide linker in oral carcinoma, activating

PPMMPB, producing fluorescence. Thus, it has the potential to guide surgical resection by

specifically fluorescing in and, therefore, identifying oral carcinoma

This PPMMPB was first validated in vivo in a murine head and neck cancer model: PPMMPB

activation was found to be tumor-specific, with fluorescence confined to cancerous tissue.

Moving into a more clinically relevant model, better representing oral carcinoma in patients,

tumor tissue from incomplete resections and whole tumors were readily identified based on the

tumor-specific activation and subsequent fluorescence of PPMMPB following a short topical

application. To further examine PPMMPB’s specificity with respect to oral carcinoma, HPLC-MS

was used to identify beacon, whether intact or activated, in healthy and cancerous hamster cheek

tissue. This confirmed that PPMMPB is activated in oral carcinoma and remains fully quenched,

producing no fluorescent signal, in surrounding healthy tissue. Additionally, the site of cleavage

on PPMMPB was characteristic of MMP-mediated cleavage, suggesting that tumor-specific

activation could be MMP-mediated. The potential of an MMP-specific molecular beacon as an

Page 59: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

49

image-guided approach to surgery has been demonstrated, but additional steps must be taken

prior to any clinical integration. Some of the next critical studies are described herein.

3.2 Immunostain-like Procedure To Identify MMPs Within Hamster Cheek Tissue

In vivo fluorescence imaging with the in-house fluorescence endoscopy system, combined with

fluorescence microscopy and histological validation, confirmed that PPMMPB was specifically

activated (enzymatically cleaved and unquenched, producing fluorescence) in oral carcinoma

(Figure 18 - Figure 21). Additionally, HPLC-MS confirmed that PPMMPB activation occurred

through cleavage of the peptide sequence at a site that is characteristic for MMP-mediated

cleavage (Figure 22 and Figure 23). However, immunostaining could not be performed to

identify MMPs within the tissue, due to a lack of hamster MMP antibodies, to confirm that

PPMMPB activation was in fact MMP-mediated. Ideally, the fluorescence microscopy data could

be overlaid with histology and tissue slices immunostained, illustrating any MMPs and

potentially attributing PPMMPB activation to the MMPs.

In an attempt to mimic immunostaining procedures and elucidate the MMPs within the cheek

tissue, a beacon activation approach to tissue staining was conceived. In this pilot study, whole

cheeks were harvested from the hamsters immediately following fluorescence imaging. They

were snap-frozen and stored at -80C. Frozen sections were cut on a cryostat (6m slices). The

slices were immersed in PBS for 5 minutes, and dried. Then 50 M of PPMMPB or PPMMP in

aqueous solution with 5% DMSO and 1.5% Tween-80 was applied to the tissue for 15 minutes.

The solution was washed off with PBS, and 30 L of mounting solution with DAPI (4’,6-

diamidino-2-phenylindole from Vector Laboratories. Inc.) was added as a nuclear stain. The

sections were covered with a coverslip and imaged by confocal microscopy (Olympus, 460 nm

excitation, 690 40 nm emission).

Page 60: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

50

Following the application of PPMMPB to the frozen tissue slice, there was activation and

associated fluorescence within certain structures within the cheek tissue (Figure 24Aii). This can

be compared to PPMMP stained frozen tissue, which has non-specific fluorescence to the point of

saturation in all tissue structures (Figure 24Aiii). Additionally, unstained frozen tissue and tissue

stained with negative control (no fluorophore) exhibited no fluorescence (Figure 24Ai and Figure

24Aiv, respectively). Looking more closely at the frozen tissue stained with PPMMPB, it appears

that PPMMPB is being activated within tumor tissue and, potentially, normal epithelial tissue

(Figure 24Bi), but the fluorescence is fairly minimal, making accurate differentiation between

regions of activated PPMMPB and intact PPMMPB challenging.

Figure 24: Tumor-bearing hamster cheeks that did not undergo PPMMPB application in vivo were

frozen and tissues slices obtained for the immunostain-like procedure on sequential slices at

different magnifications (A and B). A) i) confocal image of an unstained tissue slice showing no

fluorescence, ii) confocal image of a slice stained with 50 M PPMMPB for 15 min with

activation and fluorescence in some parts of the tissue, but difficult to discern, iii) confocal

image of a slice stained with 50 M PPMMP for 15 min with non-specific fluorescence, iv)

Page 61: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

51

confocal image of a slice stained with negative control for 15 min, confirming no staining is

accomplished by the DMSO and Tween-80 used to solubilize PPMMPB, v) hematoxylin and eosin

image representative of the tissue in the confocal images. B) Looking more closely at the tumor

region in the slice stained with 50 M PPMMPB, i) confocal image of the tumor region shows

activation and fluorescence are localized to tumor and epithelial cells, but the fluorescence is not

strong, ii) corresponding hematoxylin and eosin image showing the tissue in the tumor region of

interest (n=3).

Attempting to potentially increase the signal difference between regions of activated PPMMPB

and intact PPMMPB, the staining protocol was modified: various protocols with increased PPMMPB

concentration and/or increased staining time were tested. However, it appeared that both of these

led to increased non-specific PPMMPB activation. This suggests a difference in MMP activity or

localization is possible in the frozen tissue. Future directions will investigate the activity and

localization of the MMPs in the frozen tissue and optimize the staining concentration and

staining incubation accordingly. Additionally, studies using MMP inhibitors in vivo can be

performed to assess MMP-specific activity.

3.3 Quantification of Fluorescence Microscopy

The fluorescence microscopy data of the xenograft tongues and hamster cheeks demonstrate

specific PPMMPB activation within oral carcinoma (Figure 17 and Figure 20, respectively). The

extent of activation within specific structures and regions can be deduced by comparing the

relative fluorescence intensity within these images, under the same imaging conditions.

Additionally, the extent of activation was quantified in various hamster cheek tumors with

HPLC-MS (Figure 22 and Figure 23). It is desirable, though, to be able to quantify the

concentration of activated PPMMPB within the various tissues. This is possible by quantifying the

fluorescence intensity within the microscopy images.

Page 62: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

52

Activated PPMMPB behaves as PPMMP. Consequently, PPMMP was used for a pilot study

investigating the potential to quantify fluorescence microscopy images. A series of 1 L droplets

of PPMMP was formulated in aqueous solution with 5% DMSO and 1.5% Tween-80 and with

varying concentrations: 1 mM, 100 M, 10 M and 1 M. The droplets were covered with a

coverslip and imaged by confocal microscopy (Olympus, 460 nm excitation, 690 40 nm

emission).

The fluorescence intensity can be correlated to different concentrations of PPMMP (Figure 25).

However, more concentrations need to be assessed, particularly between 1 mM and 10 M

(Figure 25) to truly quantify the activated PPMMPB within the different regions of oral tissues.

These investigations are currently underway.

Figure 25: Confocal images of droplets of A) 1 mM, B) 10 M (n=2).

3.4 Topical Gel or Spray

Topical application is the safest and most desirable method for treatment of epithelial lesions and

it also requires fewer approval guidelines, aiding in clinical translatability. PPMMPB is currently

applied topically as an aqueous solution and there are a number of drawbacks associated with the

Page 63: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

53

application of a liquid. A liquid provides minimal control over the location of topical application,

as it can leak away from regions of interest and down the patient’s throat. Additionally, it is not

feasible to apply a liquid to all structures within the oral cavity. For example, a large percentage

of oral carcinoma patients have lesions on their tongues 64, but a liquid cannot easily be applied

to this region, limiting the usefulness of PPMMPB for fluorescence-guided resection of oral

carcinoma.

With this in mind, a thermal-transitioning PPMMPB gel was created. The formulation was

identical to the liquid PPMMPB, but also had 20% mass/volume Pluronics™, creating a PPMMPB

that was liquid at 4C, but a gel at 37C. This allowed for the PPMMPB to be applied as a liquid to

the region of interest within the oral cavity, where it would immediately gel and stay in place

(Figure 26A). This thermal transitioning PPMMPB was used to identify remaining diseased tissue

following an initial resection.

The tumor left intentionally can be better visualized following the administration of the topical

thermal-transitioning PPMMPB (Figure 26C). However, a more significant increase in

fluorescence is preferred to ease identification of diseased tissue and a stronger fluorescence is

associated with liquid PPMMPB (Figure 18). It is possible that the gel prevents migration of

PPMMPB outside of the gel, limiting the amount exposed to the hamster cheek tissue. Future

directions should optimize the gel with respect to PPMMPB migration into tissue, while still

forming a solution that can be specifically applied to regions of interest and stay in place over the

course of desired application. Alternatively, a spray, a cream or other substance could be created

and subsequently optimized.

Page 64: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

54

Figure 26: Hamster cheek treated with the thermal-transitioning PPMMPB. A) The appearance of

the thermal-transitioning PPMMPB, which stays in place as soon as applied to the tissue. B)

Images of the hamster cheek before thermal-transitioning PPMMPB was applied: i) colour white

light image, ii) monochrome white light image, iii) fluorescence white light image showing

minimal background fluorescence. B) Images of the hamster cheek after 15 min application of

thermal-transitioning PPMMPB: i) colour white light image, ii) monochrome white light image,

iii) fluorescence white light image showing a small increase in fluorescence and an easier

identification of resection area and remnant lesions. However, the fluorescence increase is not

significant, which may be attributed to the complete tumor resection or the inability of the

PPMMPB to penetrate into the tissue when in this gel.

3.5 Fluorescence-Guided Resection

The PPMMPB has been shown to specifically elucidate regions of oral carcinoma in vivo,

demonstrating its potential clinical utility in facilitating complete surgical resections. However,

PPMMPB fluorescence-guided resection has yet to be performed. The studies need to be expanded

to include, not only the identification of oral carcinoma in hamster cheek pouches, but also the

subsequent surgical resection of all fluorescent tissue, which would contain the activated

PPMMPB. Post-resection, whole cheeks could be harvested and analyzed to see if PPMMPB

fluorescence facilitates complete removal of oral carcinoma, a critical next step in the evaluation

of this strategy to improve treatment of oral carcinoma.

Page 65: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

55

3.6 Increased Tissue Penetration

Throughout the studies performed to date, PPMMPB has been shown to only penetrate into the

outer layers of tissue (Figure 20 and Figure 21). This should not hinder the ability of PPMMPB to

delineate the lateral boundaries of oral carcinoma, critical to the complete and accurate removal

of diseased oral tissue. Oral carcinoma has a much greater linear extent than depth 10,174 and

current clinical practices involve taking 1-2 cm margins of healthy tissue at the boundaries of

disease. Consequently, PPMMPB-guided surgical resection could have a much greater immediate

clinical impact on the amount of healthy surrounding tissue that is removed laterally from the

lesions, rather than depth-wise. That being said, identification of the lower boundaries of disease

could spare healthy tissue and also provides a possibility for surgical bed clean up, rather than

additional resections.

One strategy to improve PPMMPB penetration that has been investigated involves the use of a

zipper molecular beacon (ZMB). This ZMB forms through electrostatic attraction between a

polycationic peptide and polyanionic peptide, separated by the same MMP-cleavable peptide

linker that was used for all studies. At the end of the polyanion is the BHQ3 and of the

polycation is the pyro. The polycation increases cellular uptake of the fluorophore 158,180,181,

thus polycation-fluorophore complexes are referred to as cell penetrating peptides, which has the

potential to significantly improve penetration of PPMMPB within oral carcinoma.

The ZMB was optimized to maximize cellular uptake. It was believed that longer polycation

sequences would be more easily internalized than their shorter counterparts, as it is reported in

literature that cell internalization increases with increasing length of cationic arm, until a certain

point where a maximum is reached and cellular toxicity begins to increase 182. The cell

penetrating peptide was tested with pyro conjugated to five (pyro-5r) and eight (pyro-8r) arginine

(Arg) residues, respectively. These were analyzed by confocal microscopy and pyro-5r was

Page 66: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

56

internalized just as effectively as its eight-Arg counterpart and both much more effectively than

pyro alone (Figure 27).

Figure 27: Confocal and transmitted light images of MT-1 cells incubated with (A) pyro, (B)

pyro-5r and (C) pyro-8r for (i) 30 minutes and (ii) 1 hour. Both pyro-5r and pyro-8r show

significantly enhanced cell penetration compared to pyro alone.

An enhanced cell penetrating ability was demonstrated using this arginine-conjugated pyro,

when compared to pyro alone as used in PPMMPB for all studies described in Chapter 2. This

suggests that greater delivery of activated PPMMPB might be possible using this ZMB, rather than

the classical PPMMPB. This should be further investigated, potentially in conjunction with a spray

or gel to improve topical application and tissue penetration. This could allow for surgical bed

clean up to be used, rather than additional surgical resection. For example, photodynamic therapy

(PDT) could be used. In addition to being a fluorophore, pyro is also a photosensitizer (PS),

Page 67: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

57

meaning it induces cytotoxicity due to a series of photooxidative reactions, specifically through

the generation of singlet oxygen. PDT has generated significant interest because it allows for the

destruction of tumors with limited and reversible damage to surrounding healthy tissue 183. It is

minimally invasion, does not generate heat and has minimal impact on connective tissue 184.

The feasibility of using PDT for treatment of oral carcinoma is well established, with superficial

lesions and accessible light delivery 183. Should improved penetration of PPMMPB into tumors be

achieved, PDT should be investigated for surgical bed clean up, further minimizing any damage

to healthy tissue.

3.7 Conclusions

Every year, almost 300,000 individuals worldwide, including four thousand Canadians, are

diagnosed with oral cancer, more than 90% of these being oral carcinoma 4,185. The standard

treatment for these patients is surgical resection. However, accurate delineation of these tumor

boundaries is the greatest challenge facing complete and accurate removal of diseased tissue.

Currently, tissue texture is used to identify tumor boundaries, but this is fairly inaccurate and is

attributed as a cause of one of the highest rates of recurrence among cancers 42,174. In an

attempt to improve the likelihood of negative margins, surgeons aim for 1-2 cm margins when

removing oral carcinoma, leading to a huge loss of functionally and cosmetically important

tissue, often leading to permanent disfigurement, even with reconstructive surgery. Even with

these measures, positive surgical margins are common. For the patients with positive surgical

margins, radiation therapy is required and chemotherapy may also be necessary. These have a

long list of side effects including xerostomia, dermatitis, and nausea and vomiting, in the short

term. More permanent side effects include osteoradionecrosis, fibrosis, tooth decay and

neuropathy (Table 2). Even after suffering through surgery, potential disfigurement and the side

effects of radiation therapy and chemotherapy, the disease may very likely recur, with a 5-year

rate of recurrence of 30% 3.

Page 68: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

58

The ability to accurately identify oral carcinoma boundaries during surgery could significantly

improve patient prognosis. Negative surgical margins could be more easily achieved, potentially

facilitating the removal of smaller margins of healthy tissue, minimizing the debilitating effects

of surgery on quality of life. Additionally, it could minimize or even remove the need for

radiation therapy and chemotherapy, minimizing treatment procedures and leaving patients with

fewer side effects. That is, the development and clinical integration of PPMMPB fluorescence-

guided resection of oral carcinoma could change the prognosis and quality of life of patients with

oral carcinoma.

If instead of current clinical practices, surgeons could first remove all oral tissue they believe to

be diseased and then have PPMMPB fluorescence confirm the accuracy of the resections, patient

prognosis and quality of life could be improved. Following that initial white light resection,

PPMMPB could be topically applied for 15 minutes and subsequent fluorescence imaging could

effectively identify any remaining oral carcinoma. This could then be surgically removed, and if

need be, another PPMMPB topical application, followed by fluorescence imaging, given until we

could be confident in negative surgical margins. Patients wouldn’t have to suffer through

additional therapy and the associated side effects. Importantly, the rate of recurrence would be

expected to decrease significantly. This also has the potential to impact survival rates for patients

with oral carcinoma.

The work outlined involves the first steps, and the next steps, towards this goal. Disease-specific

PPMMPB activation facilitates the identification of oral carcinoma, and with continued work, we

may see integration of this molecule into the clinic, improving patient treatment and quality of

life.

Page 69: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

59

References

1 Johnson, N., Tobacco and Oral Cancer: A Global Perspective. Journal of Dental

Education 65 (4), 12 (2001).

2 Boyle, P. et al., Recent advances in epidemiology of head and neck cancer. Curr Opin

Oncol 4 (3), 471-477 (1992).

3 Society, C.C., 2013.

4 Parkin, D.M., Bray, F., Ferlay, J., & Pisani, P., Global cancer statistics, 2002. CA Cancer

J Clin 55 (2), 74-108 (2005).

5 Muller, M.G. et al., Spectroscopic detection and evaluation of morphologic and

biochemical changes in early human oral carcinoma. Cancer 97 (7), 1681-1692 (2003).

6 Reibel, J., Prognosis of oral pre-malignant lesions: significance of clinical,

histopathological, and molecular biological characteristics. Crit Rev Oral Biol Med 14

(1), 47-62 (2003).

7 Prelec, J. & Laronde, D.M., Treatment modalities of oral cancer. Can J Dent Hyg 48 (1),

8 (2014).

8 Muir, C. & Welland, L., Upper aerodigestive tract cancers. Cancer 75 (1), Suppl: 147-

153 (1995).

9 Betz, C.S. et al., A comparative study of normal inspection, autofluorescence and 5-

ALA-induced PPIX fluorescence for oral cancer diagnosis. Int J Cancer 97 (2), 245-252

(2002).

10 Slaughter, D.P., Southwick, H.W., & Smejkal, W., Field cancerization in oral stratified

squamous epithelium; clinical implications of multicentric origin. Cancer 6 (5), 963-968

(1953).

11 Zakrzewska, J.M., Fortnightly review: oral cancer. BMJ 318 (7190), 1051-1054 (1999).

12 Strong, M.S., Vaughan, C.W., & Incze, J.S., Toluidine blue in the management of

carcinoma of the oral cavity. Arch Otolaryngol 87 (5), 527-531 (1968).

13 Day, T.A. et al., Oral cancer treatment. Curr Treat Options Oncol 4 (1), 27-41 (2003).

14 Bouquot, J.E. & Whitaker, S.B., Oral leukoplakia--rationale for diagnosis and prognosis

of its clinical subtypes or "phases". Quintessence Int 25 (2), 133-140 (1994).

15 Axell, T., Pindborg, J.J., Smith, C.J., & van der Waal, I., Oral white lesions with special

reference to precancerous and tobacco- related lesions: conclusions of an international

symposium held in Uppsala, Sweden, May 18-21 1994. International Collaborative

Group on Oral White Lesions. J Oral Pathol Med 25 (2), 49-54 (1996).

Page 70: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

60

16 Silverman, S., Jr., Gorsky, M., & Lozada, F., Oral leukoplakia and malignant

transformation. A follow-up study of 257 patients. Cancer 53 (3), 563-568 (1984).

17 Lippman, S.M. & Hong, W.K., Molecular markers of the risk of oral cancer. N Engl J

Med 344 (17), 1323-1326 (2001).

18 IARC, 1989.

19 Cannon-Albright, L.A. et al., Familiality of cancer in Utah. Cancer Res 54 (9), 2378-

2385 (1994).

20 Ankathil, R., Mathew, A., Joseph, F., & Nair, M.K., Is oral cancer susceptibility

inherited? Report of five oral cancer families. Eur J Cancer B Oral Oncol 32B (1), 63-67

(1996).

21 Pukkala, E. & Notkola, V., Cancer incidence among Finnish farmers, 1979-93. Cancer

Causes Control 8 (1), 25-33 (1997).

22 La Vecchia, C. et al., Epidemiology and prevention of oral cancer. Oral Oncol 33 (5),

302-312 (1997).

23 Block, G., Patterson, B., & Subar, A., Fruit, vegetables, and cancer prevention: a review

of the epidemiological evidence. Nutr Cancer 18 (1), 1-29 (1992).

24 Potter, J.D. & Steinmetz, K., Vegetables, fruit and phytoestrogens as preventive agents.

IARC Sci Publ (139), 61-90 (1996).

25 Woods, K.V., Shillitoe, E.J., Spitz, M.R., Schantz, S.P., & Adler-Storthz, K., Analysis of

human papillomavirus DNA in oral squamous cell carcinomas. J Oral Pathol Med 22 (3),

101-108 (1993).

26 Goldstein, A.M. et al., Familial risk in oral and pharyngeal cancer. Eur J Cancer B Oral

Oncol 30B (5), 319-322 (1994).

27 King, G.N. et al., Increased prevalence of dysplastic and malignant lip lesions in renal-

transplant recipients. N Engl J Med 332 (16), 1052-1057 (1995).

28 Deng, H., Sambrook, P.J., & Logan, R.M., The treatment of oral cancer: an overview for

dental professionals. Aust Dent J 56 (3), 244-252, 341 (2011).

29 Chandu, A., Smith, A.C., & Rogers, S.N., Health-related quality of life in oral cancer: a

review. J Oral Maxillofac Surg 64 (3), 495-502 (2006).

30 Haddad, R., Annino, D., & Tishler, R.B., Multidisciplinary approach to cancer treatment:

focus on head and neck cancer. Dent Clin North Am 52 (1), 1-17, vii (2008).

31 Woolgar, J.A. et al., Survival and patterns of recurrence in 200 oral cancer patients

treated by radical surgery and neck dissection. Oral Oncol 35 (3), 257-265 (1999).

32 Rogers, S.N., Fisher, S.E., & Woolgar, J.A., A review of quality of life assessment in oral

cancer. Int J Oral Maxillofac Surg 28 (2), 99-117 (1999).

33 Sutton, D.N., Brown, J.S., Rogers, S.N., Vaughan, E.D., & Woolgar, J.A., The prognostic

implications of the surgical margin in oral squamous cell carcinoma. Int J Oral

Maxillofac Surg 32 (1), 30-34 (2003).

Page 71: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

61

34 Shah, J.P. & Gil, Z., Current concepts in management of oral cancer--surgery. Oral

Oncol 45 (4-5), 394-401 (2009).

35 Loree, T.R. & Strong, E.W., Significance of positive margins in oral cavity squamous

carcinoma. Am J Surg 160 (4), 410-414 (1990).

36 Ord, R.A. & Aisner, S., Accuracy of frozen sections in assessing margins in oral cancer

resection. J Oral Maxillofac Surg 55 (7), 663-669; discussion 669-671 (1997).

37 Forastiere, A., Koch, W., Trotti, A., & Sidransky, D., Head and neck cancer. N Engl J

Med 345 (26), 1890-1900 (2001).

38 Partridge, M. et al., Detection of minimal residual cancer to investigate why oral tumors

recur despite seemingly adequate treatment. Clin Cancer Res 6 (7), 2718-2725 (2000).

39 Haddad, R.I. & Shin, D.M., Recent advances in head and neck cancer. N Engl J Med 359

(11), 1143-1154 (2008).

40 Argiris, A., Karamouzis, M.V., Raben, D., & Ferris, R.L., Head and neck cancer. Lancet

371 (9625), 1695-1709 (2008).

41 Mazeron, R., Tao, Y., Lusinchi, A., & Bourhis, J., Current concepts of management in

radiotherapy for head and neck squamous-cell cancer. Oral Oncol 45 (4-5), 402-408

(2009).

42 Schwartz, G.J., Mehta, R.H., Wenig, B.L., Shaligram, C., & Portugal, L.G., Salvage

treatment for recurrent squamous cell carcinoma of the oral cavity. Head Neck 22 (1), 34-

41 (2000).

43 Parsons, J.T., Mendenhall, W.M., Stringer, S.P., Cassisi, N.J., & Million, R.R., An

analysis of factors influencing the outcome of postoperative irradiation for squamous cell

carcinoma of the oral cavity. Int J Radiat Oncol Biol Phys 39 (1), 137-148 (1997).

44 Ang, K.K., Altered fractionation trials in head and neck cancer. Semin Radiat Oncol 8

(4), 230-236 (1998).

45 Adelstein, D.J. et al., Maximizing local control and organ preservation in stage IV

squamous cell head and neck cancer With hyperfractionated radiation and concurrent

chemotherapy. J Clin Oncol 20 (5), 1405-1410 (2002).

46 Calais, G. et al., Randomized trial of radiation therapy versus concomitant chemotherapy

and radiation therapy for advanced-stage oropharynx carcinoma. J Natl Cancer Inst 91

(24), 2081-2086 (1999).

47 Cooper, J.S. et al., Postoperative concurrent radiotherapy and chemotherapy for high-risk

squamous-cell carcinoma of the head and neck. N Engl J Med 350 (19), 1937-1944

(2004).

48 Zackrisson, B., Mercke, C., Strander, H., Wennerberg, J., & Cavallin-Stahl, E., A

systematic overview of radiation therapy effects in head and neck cancer. Acta Oncol 42

(5-6), 443-461 (2003).

49 Laramore, G.E. et al., Adjuvant chemotherapy for resectable squamous cell carcinomas

of the head and neck: report on Intergroup Study 0034. Int J Radiat Oncol Biol Phys 23

(4), 705-713 (1992).

Page 72: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

62

50 Logan, R.M., Advances in understanding of toxicities of treatment for head and neck

cancer. Oral Oncol 45 (10), 844-848 (2009).

51 Pignon, J.P., le Maitre, A., & Bourhis, J., Meta-Analyses of Chemotherapy in Head and

Neck Cancer (MACH-NC): an update. Int J Radiat Oncol Biol Phys 69 (2 Suppl), S112-

114 (2007).

52 Adelstein, D.J. et al., Mature results of a phase III randomized trial comparing concurrent

chemoradiotherapy with radiation therapy alone in patients with stage III and IV

squamous cell carcinoma of the head and neck. Cancer 88 (4), 876-883 (2000).

53 Lamont, E.B. & Vokes, E.E., Chemotherapy in the management of squamous-cell

carcinoma of the head and neck. Lancet Oncol 2 (5), 261-269 (2001).

54 Pignon, J.P., Bourhis, J., Domenge, C., & Designe, L., Chemotherapy added to

locoregional treatment for head and neck squamous-cell carcinoma: three meta-analyses

of updated individual data. MACH-NC Collaborative Group. Meta-Analysis of

Chemotherapy on Head and Neck Cancer. Lancet 355 (9208), 949-955 (2000).

55 Pignon, J.P., le Maitre, A., Maillard, E., & Bourhis, J., Meta-analysis of chemotherapy in

head and neck cancer (MACH-NC): an update on 93 randomised trials and 17,346

patients. Radiother Oncol 92 (1), 4-14 (2009).

56 Khan, N.F., Mant, D., Carpenter, L., Forman, D., & Rose, P.W., Long-term health

outcomes in a British cohort of breast, colorectal and prostate cancer survivors: a

database study. Br J Cancer 105 Suppl 1, S29-37 (2011).

57 Hewitt, M., Greenfield, S., & Stovall, E. Committee on Cancer Survivorship: Improving

Care Quality of Life, National Cancer Policy Board, edited by Institute of Medicine and

National Research Council (The National Academies Press, Washington, DC, 2005).

58 List, M.A. & Bilir, S.P., Functional outcomes in head and neck cancer. Semin Radiat

Oncol 14 (2), 178-189 (2004).

59 Buckwalter, A.E., Karnell, L.H., Smith, R.B., Christensen, A.J., & Funk, G.F., Patient-

reported factors associated with discontinuing employment following head and neck

cancer treatment. Arch Otolaryngol Head Neck Surg 133 (5), 464-470 (2007).

60 Taylor, J.C. et al., Disability in patients with head and neck cancer. Arch Otolaryngol

Head Neck Surg 130 (6), 764-769 (2004).

61 Seiwert, T.Y., Salama, J.K., & Vokes, E.E., The chemoradiation paradigm in head and

neck cancer. Nat Clin Pract Oncol 4 (3), 156-171 (2007).

62 Cooper, J.S., Fu, K., Marks, J., & Silverman, S., Late effects of radiation therapy in the

head and neck region. Int J Radiat Oncol Biol Phys 31 (5), 1141-1164 (1995).

63 Chrcanovic, B.R., Reher, P., Sousa, A.A., & Harris, M., Osteoradionecrosis of the jaws--

a current overview--part 1: Physiopathology and risk and predisposing factors. Oral

Maxillofac Surg 14 (1), 3-16.

64 Funk, G.F. et al., Presentation, treatment, and outcome of oral cavity cancer: a National

Cancer Data Base report. Head Neck 24 (2), 165-180 (2002).

Page 73: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

63

65 Shintani, S. et al., Association of preoperative radiation effect with tumor angiogenesis

and vascular endothelial growth factor in oral squamous cell carcinoma. Jpn J Cancer

Res 91 (10), 1051-1057 (2000).

66 Poh, C.F. et al., Canadian Optically-guided approach for Oral Lesions Surgical (COOLS)

trial: study protocol for a randomized controlled trial. BMC Cancer 11, 462 (2011).

67 Leemans, C.R., Tiwari, R., Nauta, J.J., van der Waal, I., & Snow, G.B., Recurrence at the

primary site in head and neck cancer and the significance of neck lymph node metastases

as a prognostic factor. Cancer 73 (1), 187-190 (1994).

68 Brennan, J.A. et al., Molecular assessment of histopathological staging in squamous-cell

carcinoma of the head and neck. N Engl J Med 332 (7), 429-435 (1995).

69 Tabor, M.P. et al., Genetically altered fields as origin of locally recurrent head and neck

cancer: a retrospective study. Clin Cancer Res 10 (11), 3607-3613 (2004).

70 Valeur, B. & Berberan-Santos, M.N., Characteristics of Fluorescence Emission. (John

Wiley, 2012).

71 Lichtman, J.W. & Conchello, J.A., Fluorescence microscopy. Nat Methods 2 (12), 910-

919 (2005).

72 Lakowicz, J., Fluorescence Spectroscopy. (Springer, New York, 2006).

73 Periasamy, A., Fluorescence resonance energy transfer microscopy: a mini review. J

Biomed Opt 6 (3), 287-291 (2001).

74 Jares-Erijman, E.A. & Jovin, T.M., FRET imaging. Nat Biotechnol 21 (11), 1387-1395

(2003).

75 Selvin, P.R., The renaissance of fluorescence resonance energy transfer. Nat Struct Biol 7

(9), 730-734 (2000).

76 Ramanujam, N. et al., In vivo diagnosis of cervical intraepithelial neoplasia using 337-

nm-excited laser-induced fluorescence. Proc Natl Acad Sci U S A 91 (21), 10193-10197

(1994).

77 de Leeuw, J., van der Beek, N., Neugebauer, W.D., Bjerring, P., & Neumann, H.A.,

Fluorescence detection and diagnosis of non-melanoma skin cancer at an early stage.

Lasers Surg Med 41 (2), 96-103 (2009).

78 Lam, S. et al., Localization of bronchial intraepithelial neoplastic lesions by fluorescence

bronchoscopy. Chest 113 (3), 696-702 (1998).

79 D'Hallewin, M.A., Bezdetnaya, L., & Guillemin, F., Fluorescence detection of bladder

cancer: a review. Eur Urol 42 (5), 417-425 (2002).

80 Drezek, R. et al., Understanding the contributions of NADH and collagen to cervical

tissue fluorescence spectra: modeling, measurements, and implications. J Biomed Opt 6

(4), 385-396 (2001).

81 Pavlova, I. et al., Microanatomical and biochemical origins of normal and precancerous

cervical autofluorescence using laser-scanning fluorescence confocal microscopy.

Photochem Photobiol 77 (5), 550-555 (2003).

Page 74: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

64

82 Richards-Kortum, R. & Sevick-Muraca, E., Quantitative optical spectroscopy for tissue

diagnosis. Annu Rev Phys Chem 47, 555-606 (1996).

83 Lane, P.M. et al., Simple device for the direct visualization of oral-cavity tissue

fluorescence. J Biomed Opt 11 (2), 024006 (2006).

84 Poh, C.F. et al., Direct fluorescence visualization of clinically occult high-risk oral

premalignant disease using a simple hand-held device. Head Neck 29 (1), 71-76 (2007).

85 Poh, C.F., MacAulay, C.E., Zhang, L., & Rosin, M.P., Tracing the "at-risk" oral mucosa

field with autofluorescence: steps toward clinical impact. Cancer Prev Res (Phila) 2 (5),

401-404 (2009).

86 Poh, C.F. et al., Fluorescence visualization detection of field alterations in tumor margins

of oral cancer patients. Clin Cancer Res 12 (22), 6716-6722 (2006).

87 Lane, P. et al., Fluorescence-guided surgical resection of oral cancer reduces recurrence.

Proc. Of SPIE 7883.

88 Hanahan, D. & Weinberg, R.A., The hallmarks of cancer. Cell 100 (1), 57-70 (2000).

89 Curran, S. & Murray, G.I., Matrix metalloproteinases in tumour invasion and metastasis.

J Pathol 189 (3), 300-308 (1999).

90 Bissell, M.J. & Radisky, D., Putting tumours in context. Nat Rev Cancer 1 (1), 46-54

(2001).

91 Egeblad, M. & Werb, Z., New functions for the matrix metalloproteinases in cancer

progression. Nat Rev Cancer 2 (3), 161-174 (2002).

92 Thomas, G.T., Lewis, M.P., & Speight, P.M., Matrix metalloproteinases and oral cancer.

Oral Oncol 35 (3), 227-233 (1999).

93 Visse, R. & Nagase, H., Matrix metalloproteinases and tissue inhibitors of

metalloproteinases: structure, function, and biochemistry. Circ Res 92 (8), 827-839

(2003).

94 Woessner, J.F., The matrix metalloproteinase family in Matrix Metalloproteinases, edited

by W. C. Parks & R. P. Mecham (Academic Press, San Diego, California, 1998), pp. 1-

13.

95 Jones, J.L. & Walker, R.A., Control of matrix metalloproteinase activity in cancer. J

Pathol 183 (4), 377-379 (1997).

96 Sternlicht, M.D. & Werb, Z., How matrix metalloproteinases regulate cell behavior. Annu

Rev Cell Dev Biol 17, 463-516 (2001).

97 Sottrup-Jensen, L. & Birkedal-Hansen, H., Human fibroblast collagenase-alpha-

macroglobulin interactions. Localization of cleavage sites in the bait regions of five

mammalian alpha-macroglobulins. J Biol Chem 264 (1), 393-401 (1989).

98 Yang, Z., Strickland, D.K., & Bornstein, P., Extracellular matrix metalloproteinase 2

levels are regulated by the low density lipoprotein-related scavenger receptor and

thrombospondin 2. J Biol Chem 276 (11), 8403-8408 (2001).

Page 75: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

65

99 Edwards, D.R., Tissue Inhibitors Metalloproteinases in Matrix Metalloproteinase

Inhibitors in Cancer Therapy, edited by N. J. Clendeninn & K. Appelt (Humana Press,

Totowa, New Jersey, 2001), pp. 67-84.

100 Peschon, J.J. et al., An essential role for ectodomain shedding in mammalian

development. Science 282 (5392), 1281-1284 (1998).

101 Manes, S. et al., The matrix metalloproteinase-9 regulates the insulin-like growth factor-

triggered autocrine response in DU-145 carcinoma cells. J Biol Chem 274 (11), 6935-

6945 (1999).

102 Manes, S. et al., Identification of insulin-like growth factor-binding protein-1 as a

potential physiological substrate for human stromelysin-3. J Biol Chem 272 (41), 25706-

25712 (1997).

103 Boulay, A. et al., High cancer cell death in syngeneic tumors developed in host mice

deficient for the stromelysin-3 matrix metalloproteinase. Cancer Res 61 (5), 2189-2193

(2001).

104 Alexander, C.M., Howard, E.W., Bissell, M.J., & Werb, Z., Rescue of mammary

epithelial cell apoptosis and entactin degradation by a tissue inhibitor of

metalloproteinases-1 transgene. J Cell Biol 135 (6 Pt 1), 1669-1677 (1996).

105 Witty, J.P., Lempka, T., Coffey, R.J., Jr., & Matrisian, L.M., Decreased tumor formation

in 7,12-dimethylbenzanthracene-treated stromelysin-1 transgenic mice is associated with

alterations in mammary epithelial cell apoptosis. Cancer Res 55 (7), 1401-1406 (1995).

106 Powell, W.C., Fingleton, B., Wilson, C.L., Boothby, M., & Matrisian, L.M., The

metalloproteinase matrilysin proteolytically generates active soluble Fas ligand and

potentiates epithelial cell apoptosis. Curr Biol 9 (24), 1441-1447 (1999).

107 Mitsiades, N., Yu, W.H., Poulaki, V., Tsokos, M., & Stamenkovic, I., Matrix

metalloproteinase-7-mediated cleavage of Fas ligand protects tumor cells from

chemotherapeutic drug cytotoxicity. Cancer Res 61 (2), 577-581 (2001).

108 Yu, W.H., Woessner, J.F., Jr., McNeish, J.D., & Stamenkovic, I., CD44 anchors the

assembly of matrilysin/MMP-7 with heparin-binding epidermal growth factor precursor

and ErbB4 and regulates female reproductive organ remodeling. Genes Dev 16 (3), 307-

323 (2002).

109 Wu, E. et al., Stromelysin-3 suppresses tumor cell apoptosis in a murine model. J Cell

Biochem 82 (4), 549-555 (2001).

110 Baserga, R., The contradictions of the insulin-like growth factor 1 receptor. Oncogene 19

(49), 5574-5581 (2000).

111 Martin, D.C. et al., Transgenic TIMP-1 inhibits simian virus 40 T antigen-induced

hepatocarcinogenesis by impairment of hepatocellular proliferation and tumor

angiogenesis. Lab Invest 79 (2), 225-234 (1999).

112 Li, H. et al., AdTIMP-2 inhibits tumor growth, angiogenesis, and metastasis, and

prolongs survival in mice. Hum Gene Ther 12 (5), 515-526 (2001).

Page 76: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

66

113 Gatto, C. et al., BAY 12-9566, a novel inhibitor of matrix metalloproteinases with

antiangiogenic activity. Clin Cancer Res 5 (11), 3603-3607 (1999).

114 Seandel, M., Noack-Kunnmann, K., Zhu, D., Aimes, R.T., & Quigley, J.P., Growth

factor-induced angiogenesis in vivo requires specific cleavage of fibrillar type I collagen.

Blood 97 (8), 2323-2332 (2001).

115 Fang, J. et al., Matrix metalloproteinase-2 is required for the switch to the angiogenic

phenotype in a tumor model. Proc Natl Acad Sci U S A 97 (8), 3884-3889 (2000).

116 Hiraoka, N., Allen, E., Apel, I.J., Gyetko, M.R., & Weiss, S.J., Matrix metalloproteinases

regulate neovascularization by acting as pericellular fibrinolysins. Cell 95 (3), 365-377

(1998).

117 Zhou, Z. et al., Impaired endochondral ossification and angiogenesis in mice deficient in

membrane-type matrix metalloproteinase I. Proc Natl Acad Sci U S A 97 (8), 4052-4057

(2000).

118 Dong, Z., Kumar, R., Yang, X., & Fidler, I.J., Macrophage-derived metalloelastase is

responsible for the generation of angiostatin in Lewis lung carcinoma. Cell 88 (6), 801-

810 (1997).

119 Cornelius, L.A. et al., Matrix metalloproteinases generate angiostatin: effects on

neovascularization. J Immunol 161 (12), 6845-6852 (1998).

120 Ferreras, M., Felbor, U., Lenhard, T., Olsen, B.R., & Delaisse, J., Generation and

degradation of human endostatin proteins by various proteinases. FEBS Lett 486 (3), 247-

251 (2000).

121 O'Reilly, M.S. et al., Angiostatin: a novel angiogenesis inhibitor that mediates the

suppression of metastases by a Lewis lung carcinoma. Cell 79 (2), 315-328 (1994).

122 O'Reilly, M.S. et al., Endostatin: an endogenous inhibitor of angiogenesis and tumor

growth. Cell 88 (2), 277-285 (1997).

123 Deryugina, E.I., Luo, G.X., Reisfeld, R.A., Bourdon, M.A., & Strongin, A., Tumor cell

invasion through matrigel is regulated by activated matrix metalloproteinase-2.

Anticancer Res 17 (5A), 3201-3210 (1997).

124 Ahonen, M., Baker, A.H., & Kahari, V.M., Adenovirus-mediated gene delivery of tissue

inhibitor of metalloproteinases-3 inhibits invasion and induces apoptosis in melanoma

cells. Cancer Res 58 (11), 2310-2315 (1998).

125 Ala-Aho, R., Johansson, N., Baker, A.H., & Kahari, V.M., Expression of collagenase-3

(MMP-13) enhances invasion of human fibrosarcoma HT-1080 cells. Int J Cancer 97 (3),

283-289 (2002).

126 Belien, A.T., Paganetti, P.A., & Schwab, M.E., Membrane-type 1 matrix metalloprotease

(MT1-MMP) enables invasive migration of glioma cells in central nervous system white

matter. J Cell Biol 144 (2), 373-384 (1999).

127 Hua, J. & Muschel, R.J., Inhibition of matrix metalloproteinase 9 expression by a

ribozyme blocks metastasis in a rat sarcoma model system. Cancer Res 56 (22), 5279-

5284 (1996).

Page 77: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

67

128 Giannelli, G., Falk-Marzillier, J., Schiraldi, O., Stetler-Stevenson, W.G., & Quaranta, V.,

Induction of cell migration by matrix metalloprotease-2 cleavage of laminin-5. Science

277 (5323), 225-228 (1997).

129 Koshikawa, N., Giannelli, G., Cirulli, V., Miyazaki, K., & Quaranta, V., Role of cell

surface metalloprotease MT1-MMP in epithelial cell migration over laminin-5. J Cell

Biol 148 (3), 615-624 (2000).

130 Kajita, M. et al., Membrane-type 1 matrix metalloproteinase cleaves CD44 and promotes

cell migration. J Cell Biol 153 (5), 893-904 (2001).

131 Noe, V. et al., Release of an invasion promoter E-cadherin fragment by matrilysin and

stromelysin-1. J Cell Sci 114 (Pt 1), 111-118 (2001).

132 Birchmeier, C., Birchmeier, W., & Brand-Saberi, B., Epithelial-mesenchymal transitions

in cancer progression. Acta Anat (Basel) 156 (3), 217-226 (1996).

133 Polette, M. et al., Detection of mRNAs encoding collagenase I and stromelysin 2 in

carcinomas of the head and neck by in situ hybridization. Invasion Metastasis 11 (2), 76-

83 (1991).

134 Gray, S.T., Wilkins, R.J., & Yun, K., Interstitial collagenase gene expression in oral

squamous cell carcinoma. Am J Pathol 141 (2), 301-306 (1992).

135 Kusukawa, J., Sasaguri, Y., Shima, I., Kameyama, T., & Morimatsu, M., Expression of

matrix metalloproteinase-2 related to lymph node metastasis of oral squamous cell

carcinoma. A clinicopathologic study. Am J Clin Pathol 99 (1), 18-23 (1993).

136 Kusukawa, J., Sasaguri, Y., Morimatsu, M., & Kameyama, T., Expression of matrix

metalloproteinase-3 in stage I and II squamous cell carcinoma of the oral cavity. J Oral

Maxillofac Surg 53 (5), 530-534 (1995).

137 Okada, A. et al., Membrane-type matrix metalloproteinase (MT-MMP) gene is expressed

in stromal cells of human colon, breast, and head and neck carcinomas. Proc Natl Acad

Sci U S A 92 (7), 2730-2734 (1995).

138 Yoshizaki, T. et al., Increased expression of membrane type 1-matrix metalloproteinase

in head and neck carcinoma. Cancer 79 (1), 139-144 (1997).

139 Johansson, N. et al., Expression of collagenase-3 (matrix metalloproteinase-13) in

squamous cell carcinomas of the head and neck. Am J Pathol 151 (2), 499-508 (1997).

140 Charous, S.J., Stricklin, G.P., Nanney, L.B., Netterville, J.L., & Burkey, B.B., Expression

of matrix metalloproteinases and tissue inhibitor of metalloproteinases in head and neck

squamous cell carcinoma. Ann Otol Rhinol Laryngol 106 (4), 271-278 (1997).

141 Muller, D. et al., Increased stromelysin 3 gene expression is associated with increased

local invasiveness in head and neck squamous cell carcinomas. Cancer Res 53 (1), 165-

169 (1993).

142 Stahle-Backdahl, M. & Parks, W.C., 92-kd gelatinase is actively expressed by

eosinophils and stored by neutrophils in squamous cell carcinoma. Am J Pathol 142 (4),

995-1000 (1993).

Page 78: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

68

143 Pyke, C. et al., Localization of messenger RNA for Mr 72,000 and 92,000 type IV

collagenases in human skin cancers by in situ hybridization. Cancer Res 52 (5), 1336-

1341 (1992).

144 Airola, K., Johansson, N., Kariniemi, A.L., Kahari, V.M., & Saarialho-Kere, U.K.,

Human collagenase-3 is expressed in malignant squamous epithelium of the skin. J Invest

Dermatol 109 (2), 225-231 (1997).

145 Impola, U. et al., Differential expression of matrilysin-1 (MMP-7), 92 kD gelatinase

(MMP-9), and metalloelastase (MMP-12) in oral verrucous and squamous cell cancer. J

Pathol 202 (1), 14-22 (2004).

146 Tyagi, S. & Kramer, F.R., Molecular beacons: probes that fluoresce upon hybridization.

Nat Biotechnol 14 (3), 303-308 (1996).

147 Venkatesan, N., Seo, Y.J., & Kim, B.H., Quencher-free molecular beacons: a new

strategy in fluorescence based nucleic acid analysis. Chem Soc Rev 37 (4), 648-663

(2008).

148 Tan, W., Wang, K., & Drake, T.J., Molecular beacons. Curr Opin Chem Biol 8 (5), 547-

553 (2004).

149 Kostrikis, L.G., Tyagi, S., Mhlanga, M.M., Ho, D.D., & Kramer, F.R., Spectral

genotyping of human alleles. Science 279 (5354), 1228-1229 (1998).

150 Wang, H. et al., Label-free hybridization detection of a single nucleotide mismatch by

immobilization of molecular beacons on an agarose film. Nucleic Acids Res 30 (12), e61

(2002).

151 Manganelli, R., Tyagi, S., & Smith, I., Real-time PCR using molecular beacons: a new

tool to identify point mutations and to analyze gene expression in mycobacterium

tuberculosis in Methods in Molecular Medicine vol. 54, Mycobacterium tuberculosis,

edited by T. Parish & N. G. Stoker (Humana Press, 2001), pp. 295-310.

152 Tyagi, S., Bratu, D.P., & Kramer, F.R., Multicolor molecular beacons for allele

discrimination. Nat Biotechnol 16 (1), 49-53 (1998).

153 Broude, N.E., Stem-loop oligonucleotides: a robust tool for molecular biology and

biotechnology. Trends Biotechnol 20 (6), 249-256 (2002).

154 Vet, J.A., Van der Rijt, B.J., & Blom, H.J., Molecular beacons: colorful analysis of

nucleic acids. Expert Rev Mol Diagn 2 (1), 77-86 (2002).

155 Turk, B., Targeting proteases: successes, failures and future prospects. Nat Rev Drug

Discov 5 (9), 785-799 (2006).

156 Law, B. & Tung, C.H., Proteolysis: a biological process adapted in drug delivery,

therapy, and imaging. Bioconjug Chem 20 (9), 1683-1695 (2009).

157 Liu, T.W., Chen, J., & Zheng, G., Peptide-based molecular beacons for cancer imaging

and therapy. Amino Acids 41 (5), 1123-1134 (2010).

158 Weissleder, R., Tung, C.H., Mahmood, U., & Bogdanov, A., Jr., In vivo imaging of

tumors with protease-activated near-infrared fluorescent probes. Nat Biotechnol 17 (4),

375-378 (1999).

Page 79: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

69

159 Bremer, C., Tung, C.H., & Weissleder, R., In vivo molecular target assessment of matrix

metalloproteinase inhibition. Nat Med 7 (6), 743-748 (2001).

160 Funovics, M., Weissleder, R., & Tung, C.H., Protease sensors for bioimaging. Anal

Bioanal Chem 377 (6), 956-963 (2003).

161 Krinick, N.L. et al., A polymeric drug delivery system for the simultaneous delivery of

drugs activatable by enzymes and/or light. J Biomater Sci Polym Ed 5 (4), 303-324

(1994).

162 Law, B., Weissleder, R., & Tung, C.H., Protease-sensitive fluorescent nanofibers.

Bioconjug Chem 18 (6), 1701-1704 (2007).

163 Zheng, G. et al., Photodynamic molecular beacon as an activatable photosensitizer based

on protease-controlled singlet oxygen quenching and activation. Proc Natl Acad Sci U S

A 104 (21), 8989-8994 (2007).

164 Shiomi, T. & Okada, Y., MT1-MMP and MMP-7 in invasion and metastasis of human

cancers. Cancer Metastasis Rev 22 (2-3), 145-152 (2003).

165 Leinonen, T. et al., Expression of matrix metalloproteinases 7 and 9 in non-small cell

lung cancer. Relation to clinicopathological factors, beta-catenin and prognosis. Lung

Cancer 51 (3), 313-321 (2006).

166 MacDonald, I.J. et al., Subcellular localization patterns and their relationship to

photodynamic activity of pyropheophorbide-a derivatives. Photochem Photobiol 70 (5),

789-797 (1999).

167 Weissleder, R. & Ntziachristos, V., Shedding light onto live molecular targets. Nat Med 9

(1), 123-128 (2003).

168 Frangioni, J.V., In vivo near-infrared fluorescence imaging. Curr Opin Chem Biol 7 (5),

626-634 (2003).

169 Weingarten, H. & Feder, J., Spectrophotometric assay for vertebrate collagenase. Anal

Biochem 147 (2), 437-440 (1985).

170 Weingarten, H., Martin, R., & Feder, J., Synthetic substrates of vertebrate collagenase.

Biochemistry 24 (23), 6730-6734 (1985).

171 Giambernardi, T.A. et al., Overview of matrix metalloproteinase expression in cultured

human cells. Matrix Biol 16 (8), 483-496 (1998).

172 Liu, T.W. et al., Imaging of specific activation of photodynamic molecular beacons in

breast cancer vertebral metastases. Bioconjug Chem 22 (6), 1021-1030.

173 Liu, T., University of Toronto, 2013.

174 Speight, P.M., Farthing, P.M., & Bouquot, J.E., The pathology of oral cancer and

precancer. Current Diagnostic Pathology 3, 165-176 (1996).

175 Liu, T.W. et al., Imaging of specific activation of photodynamic molecular beacons in

breast cancer vertebral metastases. Bioconjug Chem 22 (6), 1021-1030 (2011).

176 Brenner, J.C. et al., Genotyping of 73 UM-SCC head and neck squamous cell carcinoma

cell lines. Head Neck 32 (4), 417-426.

Page 80: Topical Molecular Beacons for In Vivo Image …...ii Topical Molecular Beacons for In Vivo Image-Guided Resection of Oral Carcinoma Laura Allysa Jane Burgess Master of Science Department

70

177 Moriyama, E.H., Kim, A., Bogaards, A., Lilge, L., & Wilson, B.C., A Ratiometric

Fluorescence Imaging System for Surgical Guidance. Advances in Optical Technologies,

1-11 (2008).

178 Shklar, G., Schwartz, J., Grau, D., Trickler, D.P., & Wallace, K.D., Inhibition of hamster

buccal pouch carcinogenesis by 13-cis-retinoic acid. Oral Surgery 50 (1), 45-52 (1980).

179 Knight, C.G., Willenbrock, F., & Murphy, G., A novel coumarin-labelled peptide for

sensitive continuous assays of the matrix metalloproteinases. FEBS Lett 296 (3), 263-266

(1992).

180 Jiang, T. et al., Tumor imaging by means of proteolytic activation of cell-penetrating

peptides. Proc Natl Acad Sci U S A 101 (51), 17867-17872 (2004).

181 Potocky, T.B., Menon, A.K., & Gellman, S.H., Cytoplasmic and nuclear delivery of a

TAT-derived peptide and a beta-peptide after endocytic uptake into HeLa cells. J Biol

Chem 278 (50), 50188-50194 (2003).

182 Mitchell, D.J., Kim, D.T., Steinman, L., Fathman, C.G., & Rothbard, J.B., Polyarginine

enters cells more efficiently than other polycationic homopolymers. J Pept Res 56 (5),

318-325 (2000).

183 Nauta, J.M. et al., Photodynamic therapy of oral cancer. A review of basic mechanisms

and clinical applications. Eur J Oral Sci 104 (2 ( Pt 1)), 69-81 (1996).

184 Woodhams, J.H., MacRobert, A.J., Novelli, M., & Bown, S.G., Photodynamic therapy

with WST09 (Tookad): quantitative studies in normal colon and transplanted tumours. Int

J Cancer 118 (2), 477-482 (2006).

185 Vokes, E.E., Weichselbaum, R.R., Lippman, S.M., & Hong, W.K., Head and neck

cancer. N Engl J Med 328 (3), 184-194 (1993).