tumor initiating cells in mesenchymal neoplasms · population cells to initiate tumor formation in...

153
Tumor Initiating Cells in Mesenchymal Neoplasms by Colleen Wu A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Institute of Medical Sciences University of Toronto © Copyright by Colleen Wu 2010

Upload: others

Post on 24-Aug-2020

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

Tumor Initiating Cells in Mesenchymal Neoplasms

by

Colleen Wu

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Institute of Medical Sciences University of Toronto

© Copyright by Colleen Wu 2010

Page 2: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

ii

Tumor Initiating Cells in Mesenchymal Neoplasms

Colleen Wu

Degree of Doctor of Philosophy

Institute of Medical Sciences University of Toronto

2010

Abstract

Despite the clonal origins of tumors, the majority of neoplasms are composed of a heterogeneous

population of cells. The origins of this phenotype these cells have the potential to get can be

associated with cancer stem cells or tumor initiating cells have the potential to self-renew and to

differentiate giving rise to all cell types compromising a heterogeneous malignancy. These cells

are clinically important as they preferentially give rise to tumors and are therefore hypothesized

to account for the longevity and recurrence of neoplastic lesions. Cancer stem cells have been

identified from a broad range of hematopoietic, neural and epithelia tumors; however, their

function in mesenchymal neoplasms is less well defined. Using the side population assay, we

identified a subpopulation of cells within mesenchymal neoplasms, referred to as side population

cells, which are enhanced for tumor initiating potential. Importantly, we show a correlation

between the percentage of side population cells and tumor grade suggesting clinical prognostic

value as the proportion of side population cells may be a predictor of patient outcome.

Interestingly side population cells show distinct molecular features when compared to non-side

population cells and manipulation of these molecular mechanisms reduces the ability of side

population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these

experiments, we also sought to determine the cellular origins of the mesenchymal neoplasm,

aggressive fibromatosis. Using mouse models we show the influence of a mesenchymal

precursor cells in the development of this malignancy. These results identify important biological

features of mesenchymal neoplasms from which the development of targeted treatment strategies

can begin.

Page 3: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

iii

Table of Contents

TableofContents ................................................................................................................................ iii

ListofFigures.......................................................................................................................................vii

ListofTables ...................................................................................................................................... viii

ListofAbbreviations .......................................................................................................................... ix

Chapter1 .................................................................................................................................................2

1 Theroleofcancerstemcellsintheinitiationandprogressionofmesenchymal

neoplasms ...............................................................................................................................................2

1.1 Abstract....................................................................................................................................................21.2 MesenchymalNeoplasms...................................................................................................................31.3 CancerStemCells..................................................................................................................................61.3.1 Overview..............................................................................................................................................................61.3.2 CSCsinsolidtumors........................................................................................................................................71.3.3 CharacteristicsofCSCs...................................................................................................................................81.3.4 ClinicalsignificanceandtherapeutictargetingofCSCs................................................................101.3.5 CSCs:ongoingcontroversies ....................................................................................................................121.3.6 Isolationtechniques.....................................................................................................................................14

1.4 SidePopulationCells ........................................................................................................................ 141.4.1 Sidepopulationcellsinhumanneoplasms........................................................................................151.4.2 Cellularphenotype .......................................................................................................................................161.4.3 Tumorigenicpotential.................................................................................................................................171.4.4 Expressionofstem‐likegenes .................................................................................................................171.4.5 Drugefflux........................................................................................................................................................18

1.5 TheOriginsofCSCs............................................................................................................................ 191.6 MesenchymalProgenitorCells ..................................................................................................... 201.6.1 Overview...........................................................................................................................................................201.6.2 Definingthemesenchymalstem/progenitorcell ...........................................................................211.6.3 Locationofmesenchymalstemcells ....................................................................................................221.6.4 Arepericytesmesenchymalstemcells?..............................................................................................22

1.7 MesenchymalProgenitorCellsastheSarcomaCellofOrigin ............................................ 23

Page 4: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

iv

1.8 AggressiveFibromatosis................................................................................................................. 231.8.1 Overview...........................................................................................................................................................231.8.2 Molecularetiologyofaggressivefibromatosis.................................................................................241.8.3 Mousemodelsofaggressivefibromatosis .........................................................................................25

1.9 Summaryandconclusions.............................................................................................................. 251.10 ThesisSummaryandRationale.................................................................................................. 261.11 References ......................................................................................................................................... 30

Chapter2 .............................................................................................................................................. 42

2 Sidepopulationcellsisolatedfrommesenchymalneoplasmscontaintumor

initiatingcells ..................................................................................................................................... 42

2.1 Abstract................................................................................................................................................. 422.2 Introduction ........................................................................................................................................ 432.3 Results ................................................................................................................................................... 452.3.1 Mesenchymaltumorscontainsidepopulation(SP)cells............................................................452.3.2 Theproportionofsidepopulationcellscorrelateswithaggressivenessofthetumor ..452.3.3 SPcellshavethecapacitytoformtumorsuponserialtransplantationinNOD/SCID

mice 452.3.4 SPCellsEffluxRhodamine‐123...............................................................................................................47

2.4 Discussion ............................................................................................................................................ 472.5 MaterialsandMethods .................................................................................................................... 632.5.1 PrimaryTumors ............................................................................................................................................632.5.2 Pathology ..........................................................................................................................................................632.5.3 FlowCytometry..............................................................................................................................................632.5.4 CellTransplantationintoNOD/SCIDmice .........................................................................................64

2.6 References ........................................................................................................................................... 65

Chapter3 .............................................................................................................................................. 71

3 Blockadeofhedgehogsignalinginhibitstheformationoftumorsderivedfrom

osteosarcomasidepopulationcells ............................................................................................ 71

3.1 Abstract................................................................................................................................................. 713.2 Introduction ........................................................................................................................................ 723.3 Results ................................................................................................................................................... 753.3.1 Sidepopulationcellsarepresentinosteosarcomacelllines.....................................................75

Page 5: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

v

3.3.2 Hedgehogsignalinginosteosarcomasidepopulationcells .......................................................753.3.3 Invivotumorigenicpotentialofosteosarcomasidepopulationandnon‐sidepopulation

cells 763.3.4 Blockadeofhedgehogsignalinginhibitsinvivotumorformationofsidepopulation

cells 763.3.5 BlockadeofHedgehogSignalingdecreasesthecellularityofsidepopulationderived

tumors773.4 Discussion ............................................................................................................................................ 773.5 Figures................................................................................................................................................... 813.6 MaterialsandMethods .................................................................................................................... 913.6.1 Celllines ............................................................................................................................................................913.6.2 Flowcytometry ..............................................................................................................................................913.6.3 RNAextractionandrealtime‐RT‐PCR.................................................................................................913.6.4 Xenograftmodels ..........................................................................................................................................923.6.5 Dissociationofxenograftedtumors ......................................................................................................923.6.6 Invitroandinvivoblockadeofhedgehogsignaling.......................................................................92

3.7 References ........................................................................................................................................... 94

Chapter4 .............................................................................................................................................. 98

4 Thedevelopmentofaggressivefibromatosis(desmoidtumor)isinfluencedby

mesenchymalprogenitorcells...................................................................................................... 98

4.1 Abstract................................................................................................................................................. 984.2 Introduction ........................................................................................................................................ 994.3 Results .................................................................................................................................................1014.3.1 Aggressivefibromatosiscontainasubpopulationofcellswithprogenitorproperties

1014.3.2 PositivecorrelationbetweennumbersofaggressivefibromatosisandCFU‐Fsin

Apcwt/1638nmice............................................................................................................................................................ 1014.3.3 Mesenchymalprogenitorsareinvolvedinthedevelopmentofaggressivefibromatosis

1024.3.4 MesenchymalbutnotepithelialderivedtumorsareimpactedbyalterationofMPCs 1024.3.5 MesenchymalprecursorsfromApcwt/1638Nhavethecapacitytoinitiatetumorformation

1034.4 Discussion ..........................................................................................................................................103

Page 6: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

vi

4.5 MaterialsandMethods ..................................................................................................................1184.5.1 Primarytumors........................................................................................................................................... 1184.5.2 Flowcytometry ........................................................................................................................................... 1184.5.3 Generationofgeneticallyengineeredmice..................................................................................... 1184.5.4 Scoringoftumors ....................................................................................................................................... 1194.5.5 CellCulture.................................................................................................................................................... 1194.5.6 Xenograftmodels ....................................................................................................................................... 1194.5.7 Geneprofiling .............................................................................................................................................. 120

4.6 References .........................................................................................................................................121

Chapter5 ............................................................................................................................................125

5 Summary,conclusions,andfuturedirections ................................................................125

5.1 Summary ............................................................................................................................................1265.2 Conclusions........................................................................................................................................1265.3 FutureDirections.............................................................................................................................1275.3.1 Thesidepopulationassay:considerationsforitsuseintheisolationofcancerstem

cells/tumorinitiatingcells .................................................................................................................................... 1275.3.2 Characterizationofsidepopulationcellsinmesenchymalneoplasms .............................. 1305.3.3 Clinicalsignificanceofsarcomasidepopulationcell ................................................................. 1335.3.4 Tumormicroenvironment ..................................................................................................................... 1345.3.5 Mesenchymalprogenitorcellsandtheirinvolvementinthedevelopmentofaggressive

fibromatosis ................................................................................................................................................................. 1375.4 ConcludingRemarks.......................................................................................................................1395.5 References .........................................................................................................................................140

Page 7: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

vii

List of Figures

Figure 1.1 Cellular origins of cancer stem cells ........................................................................... 28

Figure 2.1 Mesenchymal tumors contain side population cells .................................................... 51

Figure 2.2 High grade sarcomas have an increased prevalence of SP cells when compared to

lower grade lesions. ...................................................................................................................... 53

Figure 2.3 Histopathologic features of SP tumors ........................................................................ 57

Figure 2.4 Characteristics of tumors derived from SP and non-SP cells...................................... 59

Figure 2.5 SP cells efflux Rhodamine-123 ................................................................................... 61

Figure 3.1 Side population cells are present in osteosarcoma cell lines ....................................... 81

Figure 3.2 Hedgehog signaling in osteosarcoma side population cells ........................................ 83

Figure 3.3 Blockade of hedgehog signaling inhibits in vivo tumor formation of osteosarcoma side

population cells ............................................................................................................................. 87

Figure 3.4 Side population tumors treated with triparanol have decreased cellularity................. 89

Figure 4.1 Human AF tumors contain progenitor cells .............................................................. 106

Figure 4.2 Correlation of CFU-F with numbers of AF tumors................................................... 108

Figure 4.3 Modulation of MPCs impacts tumor development ................................................... 110

Figure 4.4 Progenitor statuses in Apcwt/1638N/Sca-1-/- Mice ......................................................... 112

Figure 4.5 Loss of Sca-1 does not impact the formation of epithelial lesions............................ 114

Figure 4.6 Stromal cells with oncogenic mutations have tumor initiating potential .................. 116

Page 8: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

viii

List of Tables

Table 2.1 The proportion of tumors that formed from injection of various numbers of cells from

each subpopulation into NOD/SCID mice.................................................................................... 55

Table 3.1 Proportion of tumors formed in NOD/SCID mice from injection of various numbers of

side population and non-side population cells. ............................................................................. 85

Page 9: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

ix

List of Abbreviations

ABC ATP binding cassette

APC adenomatous polyposis coli

AF aggressive fibromatosis

AML acute myeloid leukemia

CFU-F colony forming unit-fibroblastic

CML chronic myelogenous leukemia

CSC cancer stem cell

DMSO dimethyl sulfoxide

FAP familial adenomatous polyposis

FBS fetal bovine serum

FIF familial infiltrative fibromatosis

HSC hematopoietic stem cell

LSC leukemic stem cell

MPC mesenchymal progenitor cells

MSC mesenchymal stem cell

OS osteosarcoma

PTCH1 Patched 1

SP side population

SMO smoothened

TIC tumor initiating cell

Page 10: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

1

CHATPER 1

INTRODUCTION

The role of cancer stem cells in the initiation and progression of

mesenchymal neoplasms

Page 11: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

2

Chapter 1

Introduction

1 The role of cancer stem cells in the initiation and progression of mesenchymal neoplasms

1.1 Abstract

Cancer stem cells (CSCs) are found in multiple tumor types. While the presence of surface

markers selectively expressed on CSCs is used to isolate these cells, no marker or pattern of

makers is known to prospectively identify CSCs in many tumor types, including mesenchymal

neoplasms. In such cases exploitation of stem cell characteristics can be used to identify CSCs,

and one such characteristic is the capacity to extrude dyes such as Hoechst 33342. Cell that

exclude this dye are referred to as side population (SP) cells. These cells share characteristics of

CSCs, specifically, they are enriched for tumor initiating capacity, they express stem-like genes,

and they are resistant to chemotherapeutic drugs. Dye exclusion is a valuable technique as it

identifies a unique population of cells with stem-like characteristics.

Page 12: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

3

1.2 Mesenchymal Neoplasms

Mesenchymal neoplasms or sarcomas, unlike epithelial tumors, do not originate from a specific

organ, but rather develop from supporting tissues, such as muscle, fat, fibrous tissue and bone.

Mesenchymal neoplasms can be grouped into two general categories, soft tissue sarcomas and

primary bone sarcomas. In each class, based on histological lines of differentiation, there are

approximately 50 subgroups demonstrating the heterogeneity of this tumor type. In addition to

location of origin and histological appearance, sarcomas can also be broadly classified based on

molecular features into two categories, those with defined diagnostic molecular events and those

with variable complex histological genetic changes[1-3].

The defined molecular event can be a specific point mutation or a translocation event.

Chromosomal translocations constitute the majority of specific genetic alterations associated

with sarcomas. Often, they result in the expression of an oncogenic fusion protein that act as an

abnormal transcription factor deregulating the transcription of multiple downstream genes and

pathways[4]. Fusion gene related sarcomas may account for a third of all sarcomas[5, 6]. Many

specific recurrent chromosomal translocations have been cloned and the resulting fusion genes

identified. Sarcomas affiliated with specific translocation events include alveolar

rabdomyosarcoma, synovial sarcoma, myxoid lipsarcoma, and Ewing’s sarcoma.

The contribution of these fusion proteins in sarcomagenesis supported with the generation of

mouse models for this class of mesenchymal neoplasms. These models also reveal important

information regarding the cell of origin for certain sarcomas. For example, approximately eighty

five percent of alveolar rabdomyosarcoma are associated with a translocation that fuses the Pax

transcription factors, most often Pax3, to a Fkhr head transcription factor[7]. The resultant

protein is believed to act as an oncogene, forcing somatic cells down aberrant embryonic

differentiation. Generation of a Cre/loxP-mediated conditional “knock in” system to insert a

silenced portion of the fkhr gene at the Pax3 locus whereby Pax3 can be normally transcribed

until the Cre recombinase converts Pax3 into the fusion protein. Spatial and temporal regulating

the expression of Pax3:Fkhr reveals expression in late muscle progenitors and not embryonic

cells results in the formation of tumors in small numbers of mice. Tumor frequency could be

increased by the conditional ablation of either p53 or CDKN2A in these mice and produced

neoplasms that phenotypically recapitulated the human disease[8]. Synovial sarcomas are often

Page 13: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

4

marked by a translocation leading to the production of the chimeric fusion protein SYT-SSX[9].

Generation of a Cre/loxP-mediated conditional “knock in” whereby the ubiquitous Rosa 26

promoter regulates SYT-SSX expression and Myf5, a promoter specific to myoblasts, drives the

conditional expression of Cre; revealed that expression of the oncogene in committed muscle cell

progenitors results in the formation of synovial sarcoma with 100% penetrance. In contrast,

expression driven by a promoter specific to differentiated muscle cells induces myopathy without

tumor induction[10]. It should be noted that not all mouse models expressing sarcoma associated

translocations result in mesenchymal neoplasms that recapitulate the human disease. For

example, Ewing’s sarcomas are characterized by the presence of the fusion of a portion of the

EWS gene to a segment of one of the ets family of genes. The most common translocation

generating the EWS-FLI-1 fusion protein, believed to act as an aberrant transcriptional activator

contributing to the development of Ewing’s sarcoma[11-13]. Conditional expression of this

protein in the bone marrow and mesenchymal tissue results in the development of leukemia.

While the use of mouse models help in understanding the biologic significance of these fusion

proteins, it is important to note that, not only do these translocations contribute to the molecular

pathogenesis of the disease, they also serve as powerful diagnostic markers for classification[14].

The second broad category of sarcomas, characterized by complex karyotypes, can include

osteosarcomas, leiomyosarcomas, malignant fibrous histocytoma, embryonal

rhabdomyosarcoma, and chondrosarcomas. Inactivation of the p53 pathways appears to be a key

differentiating factor between the two sarcoma classes[15]. Despite their low prevalence,

secondary alterations in the p53 pathway impact the clinical behavior of sarcomas with specific

gene mutations. In contrast, sarcomas with unbalanced karyotypes, mutations in p53 have a

weaker prognostic value, despite a higher prevalence of mutations[16, 17]. It can be

hypothesized, that the p53 pathway is at least partially functional in most sarcomas with specific

translations, possibly acting in concert with the cellular effects of fusion oncoproteins. However,

in sarcomas with nonspecific genetic alterations, p53 pathway inactivation may be a common

early event needed to overcome checkpoints triggered by senescence, telomere erosion, or

double stranded DNA breaks in their progression[3]. The relative timing of common oncogenic

events remains unclear and cannot be readily extrapolated from mouse models.

Despite this, mouse models confirm the importance the p53 pathway in contributing to

sarcomagenesis with p53 knockout mice developing both sarcomas and lymphomas[18, 19].

Page 14: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

5

Also, mice harboring specific germline point mutations in p53 develop a spectra of tumors,

including osteosarcomas[20, 21]. Other signaling pathways can act in conjunction with an

aberrant p53 pathway to induce sarcoma formation. For example, expression of the Kras allele in

mice has the potential to induce lung cancer with a high penetrance; however this is not

sufficient to induce sarcoma formation[22]. In contrast, Kras expression in concert with loss of

p53 in muscle tissue results in the formation of poorly differentiated primary soft tissue

sarcomas[23]. Furthermore, in mice, over expression of the Gli2 transcription factor in

chondrocytes results in the development of benign cartilage lesions; however, these lesions

develop into neoplastic entities that resemble chondrosarcomas when combined with a p53

deficiency[24]. This data highlights the importance of the p53 inactivation in sarcomagenesis;

however, given the wide spectrum of sarcoma tumor types that can arise from p53 mutations,

elucidation of both temporal and spatial mechanisms that regulate this process will be required to

provide further insight into this neoplastic process. For example, it has been postulated that

mesenchymal precursors may strongly influence the development of certain sarcomas.

Supporting this notion is the observation in mouse models where the loss of p53 in osteoblast

precursors results in the formation of highly metastatic osteosarcomas with 100%

penetrance[25]. 63% of mice harboring heterozygous germline deletions in NF2 develop highly

differentiated osteosarcomas with high metastatic potential[26]. However, the role of NF2 in the

formation of mouse osteosarcomas is not clear and it should be noted that humans carrying

germline or somatic mutations in this gene do not acquire osteosarcomas, but rather develop

benign Schwann cell neoplasms[27].Also, deletion of PTEN in smooth muscle induced the

formation of leiomyosarcomas in 80% of mice found, in the abdominal wall however no tumors

were found in cardiac muscle despite loss of PTEN in their precursor cells[28]. It is clear given

the broad class and heterogeneity of these tumors that a common cell of origin may not be likely.

Interestingly, ectopic expression of these translocations in mesenchymal stem cells results in the

malignant transformation of these cells and they are able to generate tumors in immuno-deficient

mice suggesting some influence of mesenchymal precursors in the development of sarcomas.

Given the heterogeneity of sarcomas, treatment for those afflicted with these tumors is complex.

Primary presentation with metastasis of soft tissue sarcomas to the lymph nodes is uncommon,

however, when this occurs, prognosis is poor[29, 30]. With the exception of a few, such as

osteosarcoma and Ewing’s sarcoma, generally these tumors do not respond well to conventional

Page 15: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

6

chemotherapy. Small, low grade sarcomas, can be treated with surgery and larger or higher grade

lesions can be treated with surgery in conjunction with radiation therapy. While this approach

decreases local recurrence it does not effect overall survival[2]. Furthermore, fibrosis or

impairment in wound healing often result as an effect of radiation therapy[31, 32]. Customized

therapy for some sarcomas for has shown some experimental promise. For example, blockade of

the hedgehog signaling pathway reduces the size of tumor formation in xenografted

chondrosarcomas [33]. Given the cellular diversity within mesenchymal neoplasms,

identification a common modality within this tumor type may and help in developing effective

targeted treatments. As such, the identification of cancer stem cells (CSC) or tumor initiating

cells (TIC) within mesenchymal may be a promising avenue of investigation.

1.3 Cancer Stem Cells

1.3.1 Overview

Despite their monoclonal origins, tumors demonstrate remarkable heterogeneity as evidenced by

marked differences in cellular morphology, kinetic growth properties and expression of cell

surface markers. Perhaps the most striking demonstration of tumor heterogeneity is the differing

capacity of neoplastic cells to initiate de novo tumor formation. In one prominent study

performed in the 1960’s, 35 patients were injected with 1 billion of their own cancer cells.

Remarkably, only seven of the thirty-five patients developed tumors from these autotransplants

[34]. Since then the development of immuno-deficient mice that can tolerate the growth of

human cells has led to a series of experiments validating this initial observation. While

differences in in vivo tumor initiating potential is only one measure of tumor heterogeneity, it

effectively establishes that all cells within a pre-existing tumor are not functionally equivalent.

The cellular mechanisms that underlie tumor heterogeneity are a widely debated issue. The

stochastic model proposes each cell within a tumor is influenced by events, either intrinsic (i.e.

cellular mutations) or extrinsic (i.e. influence of the microenvironment) that confer a

deterministic phenotype to individual cells. In this model every cell within a neoplastic lesion is

biologically equivalent and is influenced by events in a completely random manner. Importantly,

within a tumor each cell has an equal opportunity to be impacted by these events to influence

their cellular phenotype. This random, non-static process results in tumor heterogeneity that can

be assessed in cell surface marker expression, growth kinetics, or the capacity to initiate tumor

Page 16: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

7

formation. In contrast, the cancer stem cell model postulates that tumors are similar to normal

tissues in that cells are organized into a cellular hierarchy at the top of which resides the CSC or

tumor initiating cell. These cells, like their normal counterparts, have the potential to self-renew

and to differentiate into all the cell types within heterogeneous a neoplastic lesion. In this model,

cells are not influenced by random events, but rather heterogeneity is conferred by the behavioral

similarities of CSCs to their normal counterparts. The cancer stem cell model proposes that cells

within a lesion are not functionally equivalent, but rather, only a specific subset of cells, the

CSCs, are capable of establishing the heterogeneous phenotype of a tumors. As CSC have the

potential to both self-renew and to differentiate, these cells become clinically important as it is

postulated that similar to normal stem cells that maintain tissue, CSCs are responsible for tumor

growth and maintenance.

1.3.2 CSCs in solid tumors

Human CSCs were originally characterized by their immunophenotype and their ability to

reconstitute whole tumors after serial xenotransplantaion into sub-lethally irradiated immuno-

deficient mice. The presence of CSCs was originally identified in acute myeloid leukemias

(AML). It was demonstrated that only the phenotypically distinct population of AML cells

marked by the expression of CD34+ /CD38- were able to transplant human disease into immuno-

deficient mice. The remaining AML cells were in various stages of differentiation and contained

multiple mature blood cell types with limited proliferative potential. Importantly the transplanted

leukemias closely resembled the disease of the original patients demonstrating the stem like

behavior of these cells [35, 36]. Since this original finding, CSCs have been prospectively

isolated from a broad range of solid tumors. For example, in breast cancer, CSCs have been

identified as a subpopulation of cells that selectively express CD 44high/CD24 low// Lin-. As few as

100 of these cells injected into the mammary gland of mice had the potential to initiate tumor

formation whereas tens of thousands of the negative fraction failed to instigate a malignant

growth when injected into the mammary gland. Importantly these cells had the capacity to form

tumors after secondary and tertiary transplants and the new tumors recapitulated the original

caner [37]. Following this work, CSCs in brain and colon cancers were identified and isolated

based on the selective expression of CD133. In both these malignancies, CD133+ cells have the

ability to initiate tumorigenesis, whereas CD133- cells are incapable of triggering neoplastic

lesions[38-41]. What has followed has been an explosion of similar data in melanoma[42],

Page 17: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

8

prostate[43-45], sarcomas[46, 47], head and neck[48], pancreas[49, 50], liver[51] and various

other malignancies thereby highlighting the importance and universality of CSCs in the

neoplastic process.

1.3.3 Characteristics of CSCs

As CSCs are postulated to share characteristics with their normal counterparts, it can be

hypothesized that the two cell types share common cellular and molecular features critical for

regulating self-renewal, proliferation, and differentiation. For example, one family of proteins

that have been shown to be involved in regulating the self-renewal of stem cells is the Polycomb

genes, which repress the expression of their target genes through chromatin modifications. Bmi1

is a member of the Polycomb group protein family and is crucial for the self-renewal of

hematopoietic stem cells and mouse leukemic stem cells[52-54]. Firstly, Bmi1 is highly

expressed in purified mouse and human HSCs[55]. Secondly, although hematopoietic stem cells

are present in normal numbers in the fetal liver of Bmi1-/- mice, they are depleted in the postnatal

bone marrow. Reconstitution experiments indicate the Bmi1 fetal liver cells were only able to

reconstitute primary recipient mice and not secondary recipient mice indicating that the

hematopoietic stem cells are impaired in the potential to self-renew[55]. Thirdly, Bmi-1-/- HSCs

transfected with genes known to induce AML in normal HSCs resulted in leukemias that could

not be serially transplanted suggesting that the polycomb gene is also important in leukemic stem

cell self-renewal[52]. Bmi1 also promotes the proliferation of leukemic stem cells in a mouse

model of AML, as Bmi1 expressing leukemic cells are able to induce leukemia when

transplanted into irradiated mice. Furthermore, in patients with AML, expression of Bmi1 is

higher in AML cells that in normal bone marrow[56]. Bmi1 has also been implicated in self-

renewal so mammary CSCs as down regulation of Bmi1 in mammosphere initiating cells results

in an impairment of these cells to form secondary mammospheres structures by 60% when

compared to control cells[57].

Signaling cascades, such as the Hedgehog and the canonical Wnt pathways, are utilized by both

normal stem cells and CSCs to modulate their behavior. For example, the Wnt/β-catenin

pathway is implicated in regulation of stem cell self-renewal for a variety of tissue systems.

Perhaps the best-characterized function of β-catenin is in the regulation of tissue specific stem

cells within the intestine. Deregulation of this pathway can result in aberrant stem/progenitor cell

Page 18: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

9

proliferation and differentiation, often leading to carcinoma formation. Mutations that activate

the Wnt signaling cascade can induce the hyper-proliferation of crypt progenitor cells, generating

benign polyps in which multi-lineage differentiation is evident[58, 59]. Furthermore inhibition of

the pathway induces cell cycle arrest and the expression of differentiation markers in colorectal

cancer cells. A dominant negative form of TCF4 induced cell cycle arrest and the expression of

differentiation markers in colorectal cancer cells in vitro. Taken together, this suggests that β-

catenin may play a role in colon carcinogenesis by initially by increasing the self-renewal of

intestinal crypt stem cells[60, 61]. Expression of stabilized β-catenin also promotes the self-

renewal of central nervous system stem cells keratinocyte stem cells and leads to the

tumorigenesis of the central nervous system and skin. Furthermore, CSCs isolated from

cutaneous cancers are dependent on β-catenin signaling. Using mouse models, Malanchi et al

identified a population of CSC in the mouse skin and ablation of β-catenin in this population of

cells resulted in complete tumor regression[62]. In the hematopoietic system, over-expression of

β-catenin in cultured bone-marrow from mice increase the numbers of stem cells as measured by

the enhanced ability to reconstitute the hematopoietic systems of irradiated mice[63]. Also,

purified WNT3a promotes HSC self-renewal and inhibits the differentiation of HSCs in

culture[64]. In the neoplastic process, In addition, in CML β-catenin has the capacity to

transform committed progenitors into leukemic stem cells[65]. As such it is possible that

deregulation of WNT signaling causes the neoplastic proliferation of normal stem cells by over

activation of their self-renewal program.

Hedgehog signaling also plays an important role in the regulation of both normal and malignant

stem cells. Initial in vitro studies demonstrated that in the presence of Hedgehog antibodies, the

cytokine induced proliferation of HSCs was inhibited. Conversely, activation of the hedgehog

pathway via addition of Sonic hedgehog resulted in the expansion of primitive hematopoeitc

repopulating cells[66]. These in vitro observations are supported by data generated using in vivo

mouse models of this signaling pathway. Ptch+/- mice, which exhibit elevated hedgehog

signaling, show an alteration the proliferation and self-renewal homeostasis of HSCs resulting in

the expansion of hematopoietic stem cells ultimately leading to the exhaustion of HSC numbers

as measured by a diminished long-term engraftment potential after bone marrow

transplantation[66, 67]. Furthermore, mice with the condition deletion of Smo in the

hematopoietic system demonstrate a defect in long term HSC function in primary and secondary

Page 19: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

10

transplants. This was not due to impairment in either homing or the loss a specific blood

lineage[68]. However, it should be noted, that the requirement of hedgehog signaling for

hematopoiesis remains controversial, as recent papers demonstrate that contrary to previous

reports, the signaling pathway is dispensable for HSC function. HSC isolated from wild type and

Smo null fetal livers show no differences in HSC function suggesting hedgehog signaling may

have different roles in embryonic HSC function in comparison to adult HSC function. In

hematopoietic malignancies hedgehog signaling was found to be required for the development of

BCR-ABL induced leukemias. Transduction of Smo-/- hematopoietic progenitors with BCR-

ABL1 resulted in a diminished capacity of these cells to induce leukemia. Importantly, this was

not due to effects in homing or engraftments but rather a subsequent loss in the number of

leukemic stem population was detected suggesting hedgehog activity is required for the initiation

of BCR-ABL1 induced leukemias[68, 69].

1.3.4 Clinical significance and therapeutic targeting of CSCs

The presence of embryonic stem cell-like gene expression signatures in human cancers is

associated with aggressive histopathology confirming the clinical-prognostic significance of the

suggests that these cells need to be eliminated in order for disease free survival to occur. As

such, developing treatment strategies that exploit pathways thought to be involved in CSCs self-

renewal and/or differentiation are beginning to emerge. For example, glioblastoma cells express

bone morphogenic proteins (BMP) and their cell surface receptors. BMP treatment of

undifferentiated glioblastoma cells results in reduced cell proliferation and induced

differentiation of cells into mature astrocytes. Moreover, glioma cancer stem cells can be

identified by the expression of the cell surface marker CD133. Importantly, the treatment of

CD133+ glioma cells with BMP reduces the size of tumors implanted into immuno-deficient

mice increasing animal survival. This suggests that BMPs can induce differentiation of CD 133+

glioma stem cells to astrocytes, markedly attenuating their tumor-forming ability[70]. L1CAM

has higher expression in CD133+ glioma cells than in CD133+ normal neural stem cells.

Inhibition of L1CAM exression CD133+ glioma cells resulted in the disruption of neurosphere

formation and the disruption of the growth of the glioma stem cells. Importantly, using shRNA

to reduce L1CAM expression in glioma cells prior to injection into immuno-deficient mice

resulted in decreased tumor growth and increased survival of tumor bearing animals[71].

Page 20: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

11

Targeting CSCs with monoclonal antibodies against cell surface markers differentially expressed

on cancer stem cells also shows clinical promise as a treatment strategy. In AML leukemic, but

not normal stem cells exhibit high levels of the IL3 receptor (CD123). Administration of a

CD123 antibody inhibits the engraftment of AML leukemic stem cells in NOD/SCID mice and

also diminishes engraftment of secondary leukemias; suggesting an impact on the self-renewal

capacity of the leukemic stem cells. Importantly, this did not confer cytotoxic effects to normal

stem cells demonstrating specificity to the cancer stem cell population[72]. The use of a

monoclonal antibody specific for the adhesion molecule CD44 in AML led to a marked delay in

the progression of leukemia in mouse models by altering the ability of leukemia stem cells to

traffic to their supportive microenvironments and by altering lineage commitment. Interestingly,

normal hematopoietic stem cells do not appear to rely on this adhesion molecule for their

function to the same extent as leukemais, once again suggesting tumor cell specificity[73, 74]. In

gliomas, the development of a neutralizing monoclonal antibody against the Delta-like 4 ligand

(DLL), which is a member of the notch pathway, inhibits the ability to colon cancer cells to

serial transplant into immuno-deficient mice suggesting that this targets the self-renewing cancer

stem cells[75]. Importantly, treatment with notch inhibitor decreased the frequency of colon

cancer stem cells and as such is believed to impair the capacity for these cells to self- renew.

Not surprisingly, CSCs seem particular well equipped to tolerate external insults making them

relatively resistant to conventional treatments that seemingly target bulk tumors cells. Glioma,

CSCs have an enhanced ability to activate DNA damage response pathways allowing for the

rapid repair of DNA damage caused by radiation which is a major therapeutic modality for this

malignancy. Exposure to non-ionizing radiation of glioma cell cultures and xenografted gliomas

tumors resulted in an increased proportion of CD133 expressing cells. Importantly serial

transplantation of irradiated tumors demonstrated decreased latency in tumor formation, showing

the importance of enrichment of the CD133+ cells. The decreased sensitivity to radiation could

be attributed in part to the increased activation of DNA checkpoint proteins in CD133+ cells in

response to radiation when compared to CD 133- cells. Importantly, in vitro inhibition of these

proteins resulted in a loss of radiation sensitivity in the CD133+ fraction suggesting possible

clinical importance of these findings but this observation was not shown in vivo[76]. However, in

MMTV mice that develop breast cancers, exposure to ionizing radiation results in the enrichment

of breast cancer stem cells as detected by flow cytometry against specific CSC markers[77]. It is

Page 21: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

12

important to consider that radioresistance of cancer cells is strongly influenced by the tumor

microenvironment. For example, hypoxic tumors cells are more radioresistant than well-

oxygenated ones[78]. Interestingly, brain cancer stem cells may be enriched in areas of hypoxia

as it has been demonstrated that the fraction of CD133+ medulloblastoma cell lines and primary

tumors increases when cultured at reduced oxygen levels[79, 80]. As hypoxia promotes stem cell

maintenance and blocks differentiation it may play a critical role in by defining both stem cell

maintenance and resistance to radiotherapy. However, other factors may also contribute to the

increased resistance of CSCs to radiation treatment. For example, reactive oxygen species (ROS)

are critical mediators of ionizing radiation induced killing. Examination of both primary human

and mouse breast cancer stem cells reveal that these cells contain lower levels of ROS when

compared to their non CSC counterparts. Treatment of cells to ionizing radiation revealed fewer

DNA strand breaks in CSCs when compared to bulk tumor cells. This observation was attributed

to lower levels of ROS in the CSCs population as depletion of ROS scavengers in the non CSCs

population resulted in increased survival of the cells when treated with ionizing radiation[77].

The impact of targeting CSCs as a means of treatment is in the initial stages of development.

Correlations between CSC activity and clinical outcome will be of critical importance in

determining the relevance of this treatment strategy. Furthermore, most studies have been

preformed by isolating of CSCs from untreated malignancies, however, the main clinical

problem is disease relapse after initial response to therapy, as such, testing of cells after

treatment failure may provide insight into the strength of this treatment strategy.

1.3.5 CSCs: ongoing controversies

The assumption that cancer stem cells represent only a small fraction of tumor cells is

controversial as some studies demonstrate that greater than 10% of tumor cells in transgenic

mouse models of leukemia and lymphoma are capable of initiating tumors in histocompatible

mice[81] . It should be noted that the majority of in vivo data previously generated has taken

advantage of xenograft models in which human tumor cells are injected into immuno-deficient

mice[36-38, 41]. The recent data generated by Kelly et al. suggests that previous work fails to

capture the true frequency of tumor initiating cells as the mouse microenvironment may only be

conducive for the growth of a very small fraction of cells derived from human neoplasms. For

example, using a melanoma model, the frequency of tumor initiating cells was estimated to be

Page 22: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

13

0.00083% within metastatic melanomas based on the selection of the ABCG5 transporter

protein. However, Quintana et al, demonstrate that manipulation of the host environment has a

dramatic impact on the capacity of melanoma tumor cells to engraft. Prolonged incubation time,

co-culture of cells with extra-cellular matrix components, and using strains of mice that have

higher degree of immunodeficiency dramatically increased the frequency of melanoma CSCs

[42, 82]. In this work, 27% of implanted, single, unselected melanoma cells had the capacity to

engraft and to form tumors in vivo. Furthermore, expression of cell surface markers that had

previously been shown to enrich for tumorigenic potential failed to do so within this modified

microenvironment, raising the possibility that markers that enrich for rare cells with tumorigenic

potential in NOD/SCID mice may fail to distinguish tumorigenic cells from non tumroigenic

cells in assays that detect higher frequencies of tumorigenic cells. This work suggests that only a

very small proportion of tumor initiating stem cells may be represented in current NOD/SCID

xenograft models. Thus, in some neoplasms, cancer stem cells may not be a small population of

the bulk tumor and these numbers may vary widely depending on the tumor type [83]. This work

highlights the importance of establishing assays that are optimized to capture the stem cell

potential within a neoplasm.

Another source of controversy surrounding the CSC field arises from the assumptions of the

behavior of cancer stem cells in relation to their normal counterparts. It is important to define a

CSC based not on their cell of origin, but rather, on their functional properties, as such, the term

CSC does not reflect the derivation of the cell or imply a normal cell of origin. Furthermore, as

CSCs arise from tumorigenic processes, these cells may not behave in a similar fashion to

normal stem cells. For example, a normal stem cell may respond in to normal external/internal

stimuli maintaining features such as immuophenotype and frequency thereby maintaining a

steady state of a particular stem cell component and their subsequent offspring. However, this

same stability may not hold true for CSCs during the course of a disease. For example, there is a

large variance in the prevalence of leukemia stem cells isolated from multiple independent

samples indicating that the size of the CSC compartment can be variable. In addition, analysis of

leukemia populations with respect to cell surface markers associated with a primitive phenotype

showed variability from patient to patient. As such it is important to define stem cells based on

their functionality and to develop assays that properly capture and read out these stem cell

functions as opposed to the expression of a relatively small set of surface markers which may or

Page 23: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

14

may not remain static. In addition, CSCs may themselves represent a heterogeneous population

with varying intrinsic capabilities for self-renewal and differentiation. As such frequency of

CSCs may not be an important feature, but rather, the ability for CSCs to be targeted for

effective treatment may be a more effective endpoint.

However, regardless of the absolute frequency of cancer stem cells present in a given tumor, the

capacity to prospectively identify a subpopulation of cells from a heterogeneous tumor that are

enriched for the capacity to initiate tumor growth, is important. Therefore, regardless of the rarity

of the stem cell population, these CSCs represent a biologically distinct population of cells as

such, their prospective identification may prove to be a means of successful target treatment in

specific tumor types.

1.3.6 Isolation techniques

Prospective identification of CSCs in solid tumors has been propelled by the discovery of

markers that are selectively expressed on CSCs and not on the bulk of the tumor cells. Although

the use of makers does confer specificity to the identification and the isolation of CSCs, this

strategy fails in the absence of known markers. Furthermore, different CSC markers are used for

different tumor types and markers of normal stem cells may not be necessarily successful in the

identification of cancer stem cells. Hence, the discovery of markers that will universally identify

a TIC seems unlikely, and as such, the identification of CSCs using this strategy may prove to be

elusive. An alternative means of CSC isolation exploits manipulation of the differing

characteristics between stem like cells and non-stem cells offers. Furthermore, the universality of

these assays makes them an alluring alternative means of isolation. One such assay, Hoechst dye

exclusion, has proven to be successful on this front.

1.4 Side Population Cells

Hoechst 33342 dye binds to the AT-rich regions of the minor groove of DNA. Fluorescence

intensity is dependant on many factors involved in DNA structure such as chromatin structure,

DNA content and position of the cell within the cell cycle[84, 85]. While uptake occurs

universally in all cells, efflux is less permissive. Cells with the capacity to efflux the dye were

first identified in the mouse bone marrow and they were referred to as side population cells as

they fell to the “side” of the bulk of the positively stained cells in FACS analysis plots[86].

Page 24: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

15

Mouse bone marrow SP cells are highly enriched for long term repopulating cells. Since this

original discovery, SP cells have been identified in a variety of tissues[87-89], including skin[90,

91], lung[92, 93], liver[94], heart[95], brain[96], mammary gland[97] and skeletal muscle[98]. In

normal tissues they express high levels of stem-like genes and possess multi-potent

differentiation potential, and as such, they are thought to behave in a similar fashion to stem

cells. In addition, SP cells isolated from mouse bone marrow and muscle share transcriptome

signatures and they under express genes representing tissue specific functions[99].

The mechanism regulating the efflux of Hoechst dye is conferred in part, through the expression

of ATP binding cassette protein (ABC) transporters[100]. Forced expression of these membrane

transporters has direct effects on murine stem cells[101, 102]. However, it should be noted that

Mdr1a/b/Bcrp1 triple knockout mice are viable and still retain some SP cells in the bone

marrow[103]. This suggests that there is either a redundancy in transporter function and/or the

mechanism in which the SP phenotype is determined is not solely conferred through the

expression of ABC transporter proteins. While the exact mechanism of Hoechst dye exclusion

has yet to be fully elucidated, non the less, in adult tissues, SP cells appear to share similar

features to stem like cells.

1.4.1 Side population cells in human neoplasms

SP cells have been identified in a large variety of cancer cell lines with their presence ranging

from 0-20% of the total cell population[46, 104-111]. Given the cancer stem cell hypothesis, it

would be interesting to postulate that the percentage of SP cells within a given cancer cell line

would correlate to its tumorigenicity and/or its aggressiveness; however to date, there has been

no data demonstrating this relationship in cell lines. Also, given that some cell lines lack SP

cells, tumorigenicity, is therefore unlikely to be solely dependant on their presence. In any case,

SP cells contribute to the maintenance and the tumorigenic potential in those cell lines in which

they are present[104, 105, 107-109, 112, 113]. For example, in the C6 glioma cell line, only SP

cells have the capacity to form both SP and non-SP populations suggesting that only SP cells

have the capacity to self-renew and recapitulate the original phenotype of the cell line. In

addition, only SP cells had the ability to grow as neurospheres, a hallmark of neuronal stem cells

and differentiate down the different neuronal lineages[113]. Similar observations have also been

demonstrated in the MCF-7 breast cancer cell line[112]. Interestingly, many tumor derived cell

Page 25: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

16

lines contain side population cells and it has been demonstrated that in these cell lines, SP cells

have an increased capacity for self-renewal as measured by the ability to form colonies from

single cells. This suggests that in cell lines, SP cells also have stem cell-like characteristics

similar to those of primary tumors. However, it should be noted, that not all cell lines contain SP

cells hence this population is not exclusively responsible for the prolonged in vitro lifespan of

cell lines. Taken together, this data suggests that in cell lines, SP cells do have characteristics

similar to stem cells.

While much of the work examining the presence of SP cells in neoplasms has been through the

use of existing cancer cell lines, they have been also discovered in primary tumors. Specifically,

they have been detected in primary neuroblastomas[111], the ascites of ovarian cancers[106],

and in a wide range of mesenchymal neoplasms[46]. Interestingly, in primary mesenchymal

tumors, there exists a correlation between tumor grade and the percentage of SP cells

present[46]. Hence, the SP percentage may potentially be a valuable prognostic indicator in this

tumor type.

Paradoxically, SP cells are present in primary mesenchymal neoplasms, including primary

osteosarcomas; however, both SaOS and U2OS osteosarcoma cell lines do not appear to contain

them[111]. This can be explained, in part, by the fact that cell lines only very crudely represent

the true in vivo nature of tumors. For example, culture conditions used in these assays may not

completely recapitulate the in vivo conditions required to detect SP cells. Notably, the presence

of the specific growth factors fibroblastic growth factor (FGF)and basic fibroblastic growth

factor (bFGF) and a lack of serum were required to enhance the detection SP cells in the C6

glioma cell line[113]. In addition, one hallmark of osteosarcoma tumors is their notoriously

heterogeneous karyotypes, both within cells of the same tumors and in comparison to cells from

different osteosarcoma tumors[114]. In contrast, cell lines lack this feature and as such, this may

account for the differences seen in the presence of SP cells between the primary tumors and that

of the cell lines.

1.4.2 Cellular phenotype

The cellular phenotype of SP cells in mice has been characterized in a variety of tissues,

including, but not exclusive to the bone marrow, skeletal muscle, mammary gland, testis, and

skin. SP cells from these tissues highly express stem cells markers such as Sca-1, and CD

Page 26: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

17

34[86]. Unfortunately, to date, data on the cellular phenotype of SP cells isolated from tumors

has not been extensively studied. In human neuroblastomas, SP cells were shown to be negative

for the hematopoietic marker, CD45. Also, compared to non-SP cells, SP cells from

neuroblatsomas had increased expression of C-kit/CD117 and had lowered expression of

AC133/CD71 and CD56[111]. This staining pattern is similar to the cellular phenotype of neural

crest progenitor cells, suggesting that SP cells from these tumors may have stem cell

characteristics[111].

1.4.3 Tumorigenic potential

Data from several independent laboratories have demonstrated that when compared to both the

bulk tumor cell populations and to the non-SP population, SP cells isolated from

hepatocellular[104], lung[107], gastric[109], and nasopharyngeal carcinoma[108] cell lines are

highly enriched for the capacity to initiate tumor formation when xenografted into NOD/SCID

mice. Importantly, this observation has also been demonstrated in primary mesenchymal tumors.

However, in this neoplasm non-SP cells initially do have the capacity to form tumors in

NOD/SCID mice, but only SP cells have the capacity to initiate tumors upon serial

transplantation. Interestingly, in primary mesenchymal tumors, cells derived from non-SP tumors

have increased DNA content when compared to cells derived from SP tumors. This observation

has also been noted in primary ovarian neoplasms[46]. Taken together, this data suggests that the

non-SP fraction may contain a population of transiently amplifying cells that have the ability to

initially form tumors through rapid proliferation. However, these cells do not have the capacity

to self-renew and therefore cannot sustain tumor initiation upon serial transplantation.

Importantly, in primary mesenchymal tumors cells from non-SP tumors only gave rise to non-SP

cells and were therefore unable to recapitulate the original tumor phenotype[46]. Thus, similar to

CSCs, only SP cells can self renew and differentiate.

1.4.4 Expression of stem-like genes

In comparison to non-SP cells, SP cells have increased expression of genes that that are believed

to be involved in the regulation of stem cell function. Using microarray analysis and validation

with RT-PCR analysis, SP cells from MCF-7 breast cancer[112], hepatocellular[104],

gastrointestinal[109], and thyroid[105] cancer cell lines have shown to be up-regulated in the

expression of the ABCG2 transporter when compared to the non-SP cells. In addition, SP cells

Page 27: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

18

from both colon carcinoma and breast carcinoma cell lines have increased expression of genes

involved in the WNT/beta-catenin signaling pathway when compared to non-SP cells[109]. This

pathway has been shown to be involved the self-renewal of hematopoietic stem cells, and as

such, may play a similar role in SP cells[63, 64]. Recent data from the MCF-7 breast carcinoma

cell line demonstrates that in comparison to the non-SP cells, SP cells show an increase in the

expression of genes involved in cell cycle regulation, including EXT1, INHBA and CCNT2.

Furthermore, SP cells were shown to have more cells in G1/G0 than non-SP cells. In MCF-7

cells, SP cells are also up regulated in genes belonging to PI3K/AKT pathway. Interestingly, in

the presence of the PI3K inhibitor LY4498 there was a decrease in the percentage of SP cells.

Furthermore, inhibition resulted in a decreased ability of in vitro colony formation and in vivo

tumor formation[115]. This suggests that identifying signaling pathways upregulated in SP cells

may prove to be useful strategies for therapy. However, it should be noted that these array

studies were based on cancer cell lines that have been maintained in culture over long periods of

time and as such, they may not recapitulate the true tumor phenotype. Microarray data stemming

from primary tumors should shed some insight into the expression of “stemness” genes in SP

cells.

Interestingly, recent work has shown that in breast and colon cancers a relatively small number

of signaling pathways are disrupted at a high frequency while a large number of pathways are

disrupted at a low frequency [116]. Given these findings, it would be interesting to determine if

signaling pathways disrupted in the SP fraction differ from those in the bulk of the tumor cells

and as such, this may account for the differences in tumor initiating potential between the two

populations of cells.

1.4.5 Drug efflux

Even when tumors appear to be eradicated by chemotherapy, relapse often occurs. One

hypothesis is that cancer stem cells have the capacity to elude such treatments and as such,

remain viable and are therefore responsible for disease reoccurrence. One mechanism by which

this may arise is through the expression of ABC transporter proteins, as they efflux lipophillic

chemotherapeutic agents such as doxorubicin[117]. SP cells from a variety of mouse tissues have

increased expression of ABC transporters in comparison to non-SP cells, as do cancer cell lines

and cells derived from primary tumors[104, 105, 109, 112]. Expression of these proteins may be

Page 28: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

19

one mechanism by which tumorigenic potential is conferred upon SP cells, as such; these

proteins would make ideal targets for cancer therapy. Unfortunately, to date, a correlation

between the tumor initiating potential of neoplasms and the expression of ABC transporters has

not been demonstrated. In fact, ABCG2 positive MCF-7 cells showed no more tumorigenic

potential than ABCG2 negative cells[112]. Also, in microarray analysis of over 500 soft tissue

sarcomas no correlation between the expression of ABCG1 transporter and tumor grade was

observed[118]. However, this data does not rule out the possibility of the existence of a

relationship between the two. As in the case with CSC markers, transporter expression and

function may be diverse amongst different tumor types. For example, in primary mesenchymal

tumors, while SP cells are capable of forming tumors in immunodeficient mice, Rhodaminelow

cells are not[46]. Efflux of Rhodamine-123 dye is largely conferred by the expression of the

ABCG1 transporter, and as such, ABCG2 may play the critical role in tumor initiation in soft

tissue sarcomas but not in breast carcinomas. Furthermore, Mdr1a/b/Bcrp1 triple knockout mice

are viable and although diminished, SP cells are still present in the bone marrow[100]. It is

therefore likely that multiple transporters are expressed and responsible for the SP phenotype in

any given tumor. In those tumors that are particularly resistant to chemotherapeutic agents,

transporter expression may strongly correlate to the percentage of SP cells present and these

tumors may be ideal candidates for targeted treatment of such transporters.

1.5 The Origins of CSCs

The prospective isolation of cancer stem cells demonstrates that within tumors there resides a

subpopulation of cells with stem-like characteristics; however, their cellular origins cannot be

extrapolated from these experiments. It is not clear whether cancer stem cells arise from

mutations in either normal stem cells or more differentiated progenitors. The cellular longevity

allows stem cells to be subjected to the acquisition of multiple genetic abnormalities required for

tumorigenesis; however, they are relatively quiescent they may not undergo sufficient number of

cell divisions to become a neoplastic entity. Alternatively, differentiated cells can acquire

mutations giving them the capacity to self-renew and differentiate, thereby developing stem-like

features (Figure 1.1).

Evidence from both leukemia and brain tumors suggests that mutations in both normal stem cells

and differentiated progenitors can result in tumorigenesis indicating both sources can act as

Page 29: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

20

potential candidates from which cancer stem cells can arise. In myoproliferative disorders,

overexpression of BCR-ABL will only induce tumorigenesis in stem-like cells and not

progenitors. As expression of oncogenes did not confer self-renewal properties to committed

progenitors it can be concluded that these mutations must impact a population of cells with the

pre-existing capacity to self renew[119]. However, the expression of MLL fusions in committed

myeloid progenitors can generate AML upon transplantation into immune-deficient mice[120].

Mouse models of neurofibromas indicate that loss of the tumor suppressor neurofibromin (NF1)

transforms committed progenitors rather than stem cells. Furthermore, conditional deletion of

NF1 at different stages of development demonstrates that committed Schwann cell progenitors,

proliferate and predominately contribute to neurofibroma formation [121, 122]. However,

another study demonstrates that NF-1 deficient skin derived precursors can give rise to

neurofibromas[123]. In prostate cancer, genetic lineage marking demonstrates that rare luminal

epithelial can act as stem cells and deletion of PTEN in these cells results in carcinoma

formation[119]. In the gastrointestinal tract, Lgr+ cells mark normal stem cell in both the

proximal and distal sites[124]. Conditional ablation of the APC tumor suppressor gene, which

mediates the development of colorectal cancer, from Lgr+ intestinal stem cells results in the

formation of adenomas in the small intestine and colon. In contrast ablation of APC in transiently

amplifying cells did not result in neoplastic transformation[125].

The cellular origin of mesenchymal neoplasms has not been identified; however, it has been

postulated that they may arise from mutations in mesenchymal progenitor cells.

1.6 Mesenchymal Progenitor Cells

1.6.1 Overview

The identification of cells with the potential to differentiate down the various lineages of

mesenchymal tissue was first described in the bone marrow. Originally these cells were used as

feeder layers to promote the growth of hematopoietic stem cells, however it became apparent

that these stromal cells had stem like capacities[126]. Specifically, these cells had capacity to

form adherent colonies in culture and were able to differentiate into cells of bone, cartilage, and

fat demonstrating multipotent potential[127]. Since the discovery of multi-potent mesenchymal

progenitors in the bone marrow, cells with similar characteristics have been identified from

various adult and fetal tissues. However, it has yet to be determined whether these tissues arise

Page 30: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

21

from a common precursor such as a mesenchymal stem/progenitor cell as the isolation of a single

clonogenic self-renewing cell that can generate on one or more of the specialized cell types that

constitutes mesenchymal tissue has yet to be definitively isolated. This work is largely hampered

by the lack of cell surface markers or pattern of markers known to identify a mesenchymal stem

cell (MSC).

1.6.2 Defining the mesenchymal stem/progenitor cell

The ambiguity regarding the existence of the MSC stems from the lack of identifying markers,

various locations of isolation, and disparity in methods used to culture and expand cells. This has

resulted in the isolation of a heterogeneous population of “MSC” that exhibit variable

phenotypes, and as such, hampers the precision of defining such a population of cells. Currently,

the commonly accepted tests for the identification of MSCs include the capacity to form colony

forming unit fibroblastic (CFU-F) in culture, analysis of surface marker profiles, and multi-

lineage potential, particularly oteogenesis, chondrogenesis and adipogeneiss[128]. As mentioned,

to date no single marker or pattern of markers is known to isolate a clonogenic MSC. Profiling of

MSC surface antigen expression demonstrates some consistency amongst the differing

populations of MSCs. To date, the consensus for marker profile by the International Society for

Cellular Therapy establishes that by flow cytometry, MSC express CD73, CD90 (Thy-1) and

CD105. Other markers know to be expressed on MSCS are CD49a and STRO-1. While STRO-1

is the best known MSC marker, it is not exclusive to these cells and is lost during culture[129].

CD146 in conjunction with NG2 and PDGF2, identifies pericytes, a population of sub-

endothelial cells ability to differentiate down mesodermal lineages and is another putative

marker that shows promise in the identification of MSCs.

The majority of work identifying MSCs has focuses on the multi-potent abilities of isolated cells.

However, by definition, a stem cell is required to self-renew to create more stem cells. While

CFU-F are capable of forming colonies, clonal expansion does not equate to self-renewing

potential. In vitro work demonstrating the self-renewal potential of clonogenic MSC reveal that

these cell are organized into a cellular hierarchy in which differentiation down multi lineages

occurs in a regulated fashion. These clones had the capacity to self-renew, demonstrating in

vitro, isolation of a single clonogenic MSC[130].

Page 31: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

22

One challenge of identifying MSC such cells is that by definition these cells must have the

capacity to regenerate their tissue of origin. This has best been exemplified in the hematopoietic

system in which a single HSC has the capacity to reconstitute the entire blood system. However,

in other tissue types this becomes a more challenging question. Currently, the strongest evidence

for the existence of a mesenchymal progenitor cells is provided by experiments demonstrating

muscle satellite cells, which have the capacity to differentiate into cells of various mesenchymal

lineages, also have the capacity to regenerate in vivo muscle tissue in mouse models of

Duchanne’s muscular dystrophy [131].

1.6.3 Location of mesenchymal stem cells

Adult, tissue specific stem cells are found in specialized niches in their corresponding tissues of

origin. For example, hematopoietic stem cells can be found in the bone marrow, epidermal stem

cells in mammalian hair follicles, intestinal stem cell in the intestinal crypts[132]. However, by

current definitions, MSCs can be found throughout an entire organism[133]. This phenomenon

can be occur by several mechanisms. For example, MSCs may reach all these areas by

circulating through the blood system from a primary source; however, the difficulty in isolating

of MSCs from peripheral blood argues against this possibility[134, 135]. A second possibility,

based on the fact that postnatal MSC have been isolated from different tissues, is that each tissue

type possesses tissue specific MSC with intrinsic stem cell like properties when characterized in

vitro. Thirdly, there has been increasing evidence that an intimate relationship between MSCs

and perivascular cells[136]. Furthermore, this would explain the ability to isolate mesenchymal

progenitor cells from a wide range of tissues and the establishment of MSC-like cultures from

blood vessels supports this hypothesis[133]. As such the pericyte may be an in vivo source of

MSCs.

1.6.4 Are pericytes mesenchymal stem cells?

Pericytes are defined morphologically on their basis of their location in relation to endothelial

cells. They are located on the abluminal side of blood vessels, immediately opposed to

endothelial cells[137]. Tissue sections stained with an antibody specific to periciytes

demonstrates their presence in both small and large blood vessels, suggesting that they may form

a subendothelial network spanning the vasculature[138]. Both pericytes and MSC express similar

cell surface markers, and interestingly, there is also evidence that suggesting that pericytes have

Page 32: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

23

the in vivo potential to act as progenitors for adipocytes, cartilage and bone after injury. Further

supporting this relationship between MSCs and pericytes, is the identification of markers that

allows for the prospective isolation of pericyets from a variety of tissue types. Using these

markers, Crisan et al were able to identify clones that were multi-potent for osteogenic,

chondrogenic, and myogenic lineages in vitro from multiple organs including skeletal muscle,

pancreas, and adipose tissue. Thus blood vessel walls may harbor a reserve of progenitor cells

that may be central to the origins of MSC[139].

1.7 Mesenchymal Progenitor Cells as the Sarcoma Cell of Origin

Given the cellular diversity of mesenchymal neoplasms, it has been postulated that multi-potent

MSCs are their cell of origin. The demonstration that some neoplasms can arise from oncogenic

mutations in normal stem cells, aids in supporting this hypothesis. Recent publications have

begun to address this question in Ewing’s sarcoma and in malignant fibrous histiocytoma

(MFH), both of which are mesenchymal tumors. Specifically, it has been shown that over-

expression of the EWS-FL-1 fusion protein, the transforming event in Ewing’s sarcoma, results

in the transformation of primary bone marrow derived mesenchymal progenitor cells.

Furthermore, these cells generate tumors that display the hallmarks of Ewing’s sarcoma [140].

Tirode et al. demonstrate shRNA silencing of the EWS-FL1 gene in Ewing’s sarcoma cell lines

results in the generation of a transcriptional profile similar to mesenchymal stem cells. These

silenced cells also recovered the phenotype of mesenchymal stem cells in that they had the

potential to differentiate into both adipocytes and osteoblasts[141]. Finally, it has been shown

that human mesenchymal stem cells can be transformed, via inhibition of the Wnt/β-catenin

signaling pathway, to form MFHs[142]. Taken together this suggests that mesenchymal

neoplasms may indeed originate from mesenchymal progenitor cells.

1.8 Aggressive Fibromatosis

1.8.1 Overview

Aggressive fibromatosis (AF: also called desmoids tumor) is a rare, benign neoplasm

representing 0.03 to 0.1% of all tumors and 3.5% of fibrous tissue tumors with an occurrence

frequency of approximately 2-4 cases per million population[143]. Tumors are characteristically

Page 33: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

24

slow growing, although a wide variation exists, and is often associated with local invasion of fat

and muscle with infiltration of neurovascular structures. Metastasis is exceedingly rare[144].

Aggressive fibromatosis are benign lesions of fibrous growth that are believed to arise from

deregulation of connective tissue growth[145]. The lesions lack a capsule and are characterized

by poorly demarcated basses that blend into the surrounding tissue[146, 147]. Histopatholocially,

these tumors are composed of bipolar cells that stain positively for the intermediate filament

vimentin but do not express the epithelial marker keratin; strongly resembling well differentiated

fibroblasts[148]. The anatomical location, cellular morphology, and histological profile of these

lesions suggest that they are of mesenchymal origins.

There are several therapeutic options for the management of AF including surgical excision,

radiation and pharmacological therapy. Overall, however, the effectiveness of these approaches

is limited and recurrence rates are high[149]. The primary therapy of choice is wide local

surgical excision to a normal tissue margins, but his approach is limited to mainly extremity and

small mesenteric AF[150]. Complete excision of large intra-abdominal AF is more difficult as it

may involve sacrificing critical structures. Local recurrence rates are high, despite

microscopically clear surgical margins[151].

1.8.2 Molecular etiology of aggressive fibromatosis

In children, AF is found at the highest frequency in the extremities, but can also be located in

intro abdominal regions. Most AF in children occurs sporadically. The molecular etiology of

aggressive fibromatosis is well characterized. In adults, patients with autosomal dominant

hereditary condition know as familial adenomatous polyposis (FAP) have a 1000-fold higher

susceptibility to developing AF[152, 153]. The adenomatous polyposis coli (APC) tumor

suppressor gene is mutated in FAP patients leading to the truncation and loss of function of the

APC protein. The frequency of occurrence of AF in FAP patients varies in adults between

different studies ranging from 3.6-34%[143, 152, 154]. Mutations in β-catenin protein that result

in the up-regulation of the β-catenin/WNT signaling pathway is a consistent feature of both

sporadic and FAP-associated AF tumors. In sporadic cases, most tumors contain a mutation in

CTNNB1, the gene that codes for β-catenin, resulting in protein elevation are present in 75% of

cases[145, 155, 156]. Regardless of the causative mutation, one unifying factor in AF

pathogenesis is the elevation of the β-catenin/WNT signaling pathway.

Page 34: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

25

1.8.3 Mouse models of aggressive fibromatosis

The Apc1638N mouse is a well-characterized mouse that closely approximates human FAP. The

APC1683N mice carries a targeted mutation at codon 1638 of the mouse Apc gene. This

mutation, a targeted frameshift at codon 1638 of the mouse Apc gene, represents a null allele and

leads to haploinsufficiency in heterozygous animals. These mice begin to develop

gastrointestinal and desmoids tumors by two months of age[157, 158]. As such this mouse model

provides an excellent tool from which we can investigate the role of mesenchymal progenitor

cells in the development and progression of aggressive fibromatosis.

1.9 Summary and conclusions

Cancer stem cells have been identified in many human malignancies. Traditionally, isolation is

based on the selective expression of cell surface markers; however, alternative methods founded

on functional characteristics of stem cells can also be used. For example, the side population

assay, based on the active efflux of the fluorescent dye Hoechst 33342, allows for the

prospective isolation of CSCs from many neoplasms. Importantly, identification of cells with

stem like characteristics within neoplastic lesions does not suggest that they arise from normal

stem cells harboring oncogenic mutations. The use of mouse models have begun to address the

cellular origins of neoplasms with recent evidence suggests both stem cells and committed

progenitors have the potential to induce tumorigenesis.

The mechanisms underlying the pathogenesis of mesenchymal neoplasm is poorly understood.

These malignancies may model the characteristics described in hematological, neuronal, and

epithelial tumors. The identification of the biological processes that give rise to mesenchymal

lesions can potentially aid in the development of new therapies.

Page 35: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

26

1.10 Thesis Summary and Rationale

The goal of this research is to investigate the development and progression of mesenchymal

neoplasms. We hypothesize that, similar to other solid malignancies, mesenchymal neoplasms

contain a subpopulation of cells that are enriched for tumor initiating potential. Furthermore, as

these tumors may be of mesodermal origins, we postulate that normal mesenchymal progenitor

cells can influence the development of certain mesenchymal neoplasms.

To examine these hypotheses, the following questions will be addressed:

1) Do mesenchymal neoplasms contain a subpopulation of cells with enhanced tumor

initiating potential? As no known markers, or pattern of markers are known to

distinguish a mesenchymal progenitor cell, we use the side population assay to identify

and prospectively isolate tumor-initiating cells within mesenchymal neoplasms (Chapter

2).

2) Can we identify a biological feature that can be exploited and used as a therapeutic

target to diminish the tumorigenic potential of mesenchymal side population cells?

Deregulation of the hedgehog signaling pathway is associated with a broad range of

cancers, including mesenchymal neoplasms. Furthermore, blockade of this pathway is an

effective therapeutic treatment modality for a variety of tumor types. Given these

observations, we investigated how chemical modulation of hedgehog signaling would

impact the behavior of tumors derived from SP cells isolated from osteosarcoma cell

lines (Chapter 3).

3) Can normal mesenchymal progenitor cells influence the development of the

mesenchymal neoplasm, aggressive fibromatosis? The cellular origins of tumors are

poorly defined. Through the use of mouse models we sought to determine if altering the

numbers of mesenchymal progenitor cells would impact the development of aggressive

fibromatosis in mice predisposed to forming these tumors. Furthermore we questioned

whether mesenchymal progenitor cells harboring a mutation associated with aggressive

fibromatosis would have tumorigenic potential. (Chapter 4).

Page 36: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

27

The identification of a subpopulation of cells within mesenchymal neoplasms enriched for tumor

initiating potential, reveals a previously unknown aspect of this tumor type and provides a

powerful tool for further investigation into mesenchymal tumorigenesis. Further elucidation of

the molecular mechanisms, conferring this phenotype will uncover important information and

will aid in the development of effective treatment modalities targeted against what may be the

most potent malignant cells. Finally, the identification of progenitor cells with the capacity to

influence the development of mesenchymal neoplasms such as aggressive fibromatosis raises the

intriguing possibility that protecting these cells in patients with known genetic predispositions to

tumor development, such as familial adenomatous polyposis patients, can prevent the

development of these neoplastic lesions. These studies highlight the importance of understanding

mesenchymal progenitor cell biology in the development of potential new treatments.

Page 37: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

28

Figure 1.1 Cellular origins of cancer stem cells

Tumors are composed of a heterogeneous population of cells from which cancer stem cells can

be prospectively isolated; however, the origins of these cells remain unclear. It has been

postulated that cancer stem cells can arise from normal stem cells harboring oncogenic

mutations. Conversely, it is hypothesized that arise from committed progenitor cells that that

acquire stem like characteristics such as the ability to self-renew.

Page 38: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

29

!"#$%&'()*+*(,&-.//&

0)1.%.(*+2.3&-.//&4$%#+/&52.#&&

-.//&

Page 39: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

30

1.11 References

1. Pollock, R.E., The seminal role of Cancer in our understanding of sarcoma. Cancer, 2008. 113(7 Suppl): p. 1969-79.

2. Skubitz, K.M. and D.R. D'Adamo, Sarcoma. Mayo Clin Proc, 2007. 82(11): p. 1409-32.

3. Borden, E.C., et al., Soft tissue sarcomas of adults: state of the translational science. Clin Cancer Res, 2003. 9(6): p. 1941-56.

4. Mitelman, F., B. Johansson, and F. Mertens, The impact of translocations and gene fusions on cancer causation. Nat Rev Cancer, 2007. 7(4): p. 233-45.

5. Mitelman, F., Recurrent chromosome aberrations in cancer. Mutat Res, 2000. 462(2-3): p. 247-53.

6. Bennicelli, J.L. and F.G. Barr, Genetics and the biologic basis of sarcomas. Curr Opin Oncol, 1999. 11(4): p. 267-74.

7. Sorensen, P.H., et al., PAX3-FKHR and PAX7-FKHR gene fusions are prognostic indicators in alveolar rhabdomyosarcoma: a report from the children's oncology group. J Clin Oncol, 2002. 20(11): p. 2672-9.

8. Keller, C., et al., Pax3:Fkhr interferes with embryonic Pax3 and Pax7 function: implications for alveolar rhabdomyosarcoma cell of origin. Genes Dev, 2004. 18(21): p. 2608-13.

9. Clark, J., et al., Identification of novel genes, SYT and SSX, involved in the t(X;18)(p11.2;q11.2) translocation found in human synovial sarcoma. Nat Genet, 1994. 7(4): p. 502-8.

10. Haldar, M., et al., A conditional mouse model of synovial sarcoma: insights into a myogenic origin. Cancer Cell, 2007. 11(4): p. 375-88.

11. Arvand, A. and C.T. Denny, Biology of EWS/ETS fusions in Ewing's family tumors. Oncogene, 2001. 20(40): p. 5747-54.

12. Delattre, O., et al., Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours. Nature, 1992. 359(6391): p. 162-5.

13. May, W.A., et al., The Ewing's sarcoma EWS/FLI-1 fusion gene encodes a more potent transcriptional activator and is a more powerful transforming gene than FLI-1. Mol Cell Biol, 1993. 13(12): p. 7393-8.

14. Ladanyi, M. and J.A. Bridge, Contribution of molecular genetic data to the classification of sarcomas. Hum Pathol, 2000. 31(5): p. 532-8.

Page 40: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

31

15. Marina, N., et al., Biology and therapeutic advances for pediatric osteosarcoma. Oncologist, 2004. 9(4): p. 422-41.

16. Drobnjak, M., et al., Prognostic implications of p53 nuclear overexpression and high proliferation index of Ki-67 in adult soft-tissue sarcomas. J Natl Cancer Inst, 1994. 86(7): p. 549-54.

17. Wurl, P., et al., Clinical relevance of pRb and p53 co-overexpression in soft tissue sarcomas. Cancer Lett, 1999. 139(2): p. 159-65.

18. Williams, B.O., et al., Cooperative tumorigenic effects of germline mutations in Rb and p53. Nat Genet, 1994. 7(4): p. 480-4.

19. Purdie, C.A., et al., Tumour incidence, spectrum and ploidy in mice with a large deletion in the p53 gene. Oncogene, 1994. 9(2): p. 603-9.

20. Olive, K.P., et al., Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome. Cell, 2004. 119(6): p. 847-60.

21. Lang, G.A., et al., Gain of function of a p53 hot spot mutation in a mouse model of Li-Fraumeni syndrome. Cell, 2004. 119(6): p. 861-72.

22. Tuveson, D.A., et al., Endogenous oncogenic K-ras(G12D) stimulates proliferation and widespread neoplastic and developmental defects. Cancer Cell, 2004. 5(4): p. 375-87.

23. Kirsch, D.G., et al., A spatially and temporally restricted mouse model of soft tissue sarcoma. Nat Med, 2007. 13(8): p. 992-7.

24. Ho, L., et al., Gli2 and p53 cooperate to regulate IGFBP-3- mediated chondrocyte apoptosis in the progression from benign to malignant cartilage tumors. Cancer Cell, 2009. 16(2): p. 126-36.

25. Berman, S.D., et al., Metastatic osteosarcoma induced by inactivation of Rb and p53 in the osteoblast lineage. Proc Natl Acad Sci U S A, 2008. 105(33): p. 11851-6.

26. McClatchey, A.I., et al., Mice heterozygous for a mutation at the Nf2 tumor suppressor locus develop a range of highly metastatic tumors. Genes Dev, 1998. 12(8): p. 1121-33.

27. Gutmann, D.H. and M. Giovannini, Mouse models of neurofibromatosis 1 and 2. Neoplasia, 2002. 4(4): p. 279-90.

28. Hernando, E., et al., The AKT-mTOR pathway plays a critical role in the development of leiomyosarcomas. Nat Med, 2007. 13(6): p. 748-53.

29. Fong, Y., et al., Lymph node metastasis from soft tissue sarcoma in adults. Analysis of data from a prospective database of 1772 sarcoma patients. Ann Surg, 1993. 217(1): p. 72-7.

Page 41: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

32

30. Wunder, J.S., et al., Opportunities for improving the therapeutic ratio for patients with sarcoma. Lancet Oncol, 2007. 8(6): p. 513-24.

31. Davis, A.M., et al., Late radiation morbidity following randomization to preoperative versus postoperative radiotherapy in extremity soft tissue sarcoma. Radiother Oncol, 2005. 75(1): p. 48-53.

32. Akudugu, J.M., et al., Wound healing morbidity in STS patients treated with preoperative radiotherapy in relation to in vitro skin fibroblast radiosensitivity, proliferative capacity and TGF-beta activity. Radiother Oncol, 2006. 78(1): p. 17-26.

33. Tiet, T.D., et al., Constitutive hedgehog signaling in chondrosarcoma up-regulates tumor cell proliferation. Am J Pathol, 2006. 168(1): p. 321-30.

34. Southam, C.M., et al., Effect of leukocytes on transplantability of human cancer. Cancer, 1966. 19(11): p. 1743-53.

35. Lapidot, T., et al., A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature, 1994. 367(6464): p. 645-8.

36. Bonnet, D. and J.E. Dick, Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med, 1997. 3(7): p. 730-7.

37. Al-Hajj, M., et al., Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A, 2003. 100(7): p. 3983-8.

38. O'Brien, C.A., et al., A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature, 2007. 445(7123): p. 106-10.

39. Ricci-Vitiani, L., et al., Identification and expansion of human colon-cancer-initiating cells. Nature, 2007. 445(7123): p. 111-5.

40. Singh, S.K., et al., Identification of a cancer stem cell in human brain tumors. Cancer Res, 2003. 63(18): p. 5821-8.

41. Singh, S.K., et al., Identification of human brain tumour initiating cells. Nature, 2004. 432(7015): p. 396-401.

42. Schatton, T., et al., Identification of cells initiating human melanomas. Nature, 2008. 451(7176): p. 345-9.

43. Patrawala, L., et al., Highly purified CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic and metastatic progenitor cells. Oncogene, 2006. 25(12): p. 1696-708.

44. Patrawala, L., et al., Hierarchical organization of prostate cancer cells in xenograft tumors: the CD44+alpha2beta1+ cell population is enriched in tumor-initiating cells. Cancer Res, 2007. 67(14): p. 6796-805.

Page 42: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

33

45. Collins, A.T., et al., Prospective identification of tumorigenic prostate cancer stem cells. Cancer Res, 2005. 65(23): p. 10946-51.

46. Wu, C., et al., Side population cells isolated from mesenchymal neoplasms have tumor initiating potential. Cancer Res, 2007. 67(17): p. 8216-22.

47. Levings, P.P., et al., Expression of an exogenous human Oct-4 promoter identifies tumor-initiating cells in osteosarcoma. Cancer Res, 2009. 69(14): p. 5648-55.

48. Prince, M.E., et al., Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci U S A, 2007. 104(3): p. 973-8.

49. Li, C., et al., Identification of pancreatic cancer stem cells. Cancer Res, 2007. 67(3): p. 1030-7.

50. Hermann, P.C., et al., Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer. Cell Stem Cell, 2007. 1(3): p. 313-23.

51. Yang, Z.F., et al., Significance of CD90+ cancer stem cells in human liver cancer. Cancer Cell, 2008. 13(2): p. 153-66.

52. Lessard, J. and G. Sauvageau, Bmi-1 determines the proliferative capacity of normal and leukaemic stem cells. Nature, 2003. 423(6937): p. 255-60.

53. Lessard, J. and G. Sauvageau, Polycomb group genes as epigenetic regulators of normal and leukemic hemopoiesis. Exp Hematol, 2003. 31(7): p. 567-85.

54. Leung, C., et al., Bmi1 is essential for cerebellar development and is overexpressed in human medulloblastomas. Nature, 2004. 428(6980): p. 337-41.

55. Park, I.K., et al., Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature, 2003. 423(6937): p. 302-5.

56. Sawa, M., et al., BMI-1 is highly expressed in M0-subtype acute myeloid leukemia. Int J Hematol, 2005. 82(1): p. 42-7.

57. Liu, S., et al., Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res, 2006. 66(12): p. 6063-71.

58. Powell, S.M., et al., APC mutations occur early during colorectal tumorigenesis. Nature, 1992. 359(6392): p. 235-7.

59. Moser, A.R., et al., The Min (multiple intestinal neoplasia) mutation: its effect on gut epithelial cell differentiation and interaction with a modifier system. J Cell Biol, 1992. 116(6): p. 1517-26.

Page 43: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

34

60. Kinzler, K.W. and B. Vogelstein, Lessons from hereditary colorectal cancer. Cell, 1996. 87(2): p. 159-70.

61. Bienz, M. and H. Clevers, Linking colorectal cancer to Wnt signaling. Cell, 2000. 103(2): p. 311-20.

62. Malanchi, I., et al., Cutaneous cancer stem cell maintenance is dependent on beta-catenin signalling. Nature, 2008. 452(7187): p. 650-3.

63. Reya, T., et al., A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature, 2003. 423(6938): p. 409-14.

64. Willert, K., et al., Wnt proteins are lipid-modified and can act as stem cell growth factors. Nature, 2003. 423(6938): p. 448-52.

65. Jamieson, C.H., et al., Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med, 2004. 351(7): p. 657-67.

66. Bhardwaj, G., et al., Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation. Nat Immunol, 2001. 2(2): p. 172-80.

67. Trowbridge, J.J., M.P. Scott, and M. Bhatia, Hedgehog modulates cell cycle regulators in stem cells to control hematopoietic regeneration. Proc Natl Acad Sci U S A, 2006. 103(38): p. 14134-9.

68. Zhao, C., et al., Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature, 2009. 458(7239): p. 776-9.

69. Dierks, C., et al., Expansion of Bcr-Abl-positive leukemic stem cells is dependent on Hedgehog pathway activation. Cancer Cell, 2008. 14(3): p. 238-49.

70. Piccirillo, S.G., et al., Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells. Nature, 2006. 444(7120): p. 761-5.

71. Bao, S., et al., Targeting cancer stem cells through L1CAM suppresses glioma growth. Cancer Res, 2008. 68(15): p. 6043-8.

72. Jin, L., et al., Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell, 2009. 5(1): p. 31-42.

73. Krause, D.S., et al., Requirement for CD44 in homing and engraftment of BCR-ABL-expressing leukemic stem cells. Nat Med, 2006. 12(10): p. 1175-80.

74. Jin, L., et al., Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nat Med, 2006. 12(10): p. 1167-74.

75. Hoey, T., et al., DLL4 blockade inhibits tumor growth and reduces tumor-initiating cell frequency. Cell Stem Cell, 2009. 5(2): p. 168-77.

Page 44: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

35

76. Bao, S., et al., Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature, 2006. 444(7120): p. 756-60.

77. Diehn, M., et al., Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature, 2009. 458(7239): p. 780-3.

78. Wright, E.A. and P. Howard-Flanders, The influence of oxygen on the radiosensitivity of mammalian tissues. Acta radiol, 1957. 48(1): p. 26-32.

79. Platet, N., et al., Influence of oxygen tension on CD133 phenotype in human glioma cell cultures. Cancer Lett, 2007. 258(2): p. 286-90.

80. Blazek, E.R., J.L. Foutch, and G. Maki, Daoy medulloblastoma cells that express CD133 are radioresistant relative to CD133- cells, and the CD133+ sector is enlarged by hypoxia. Int J Radiat Oncol Biol Phys, 2007. 67(1): p. 1-5.

81. Kelly, P.N., et al., Tumor growth need not be driven by rare cancer stem cells. Science, 2007. 317(5836): p. 337.

82. Quintana, E., et al., Efficient tumour formation by single human melanoma cells. Nature, 2008. 456(7222): p. 593-8.

83. Kennedy, J.A., et al., Comment on "Tumor growth need not be driven by rare cancer stem cells". Science, 2007. 318(5857): p. 1722; author reply 1722.

84. Arndt-Jovin, D.J. and T.M. Jovin, Analysis and sorting of living cells according to deoxyribonucleic acid content. J Histochem Cytochem, 1977. 25(7): p. 585-9.

85. Lalande, M.E. and R.G. Miller, Fluorescence flow analysis of lymphocyte activation using Hoechst 33342 dye. J Histochem Cytochem, 1979. 27(1): p. 394-7.

86. Challen, G.A. and M.H. Little, A side order of stem cells: the SP phenotype. Stem Cells, 2006. 24(1): p. 3-12.

87. Goodell, M.A., et al., Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med, 1996. 183(4): p. 1797-806.

88. Goodell, M.A., et al., Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species. Nat Med, 1997. 3(12): p. 1337-45.

89. Pearce, D.J., et al., Multiparameter analysis of murine bone marrow side population cells. Blood, 2004. 103(7): p. 2541-6.

90. Larderet, G., et al., Human side population keratinocytes exhibit long-term proliferative potential and a specific gene expression profile and can form a pluristratified epidermis. Stem Cells, 2006. 24(4): p. 965-74.

Page 45: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

36

91. Yano, S., et al., Characterization and localization of side population cells in mouse skin. Stem Cells, 2005. 23(6): p. 834-41.

92. Summer, R., et al., Side population cells and Bcrp1 expression in lung. Am J Physiol Lung Cell Mol Physiol, 2003. 285(1): p. L97-104.

93. Majka, S.M., et al., Identification of novel resident pulmonary stem cells: form and function of the lung side population. Stem Cells, 2005. 23(8): p. 1073-81.

94. Shimano, K., et al., Hepatic oval cells have the side population phenotype defined by expression of ATP-binding cassette transporter ABCG2/BCRP1. Am J Pathol, 2003. 163(1): p. 3-9.

95. Martin, C.M., et al., Persistent expression of the ATP-binding cassette transporter, Abcg2, identifies cardiac SP cells in the developing and adult heart. Dev Biol, 2004. 265(1): p. 262-75.

96. Kim, M. and C.M. Morshead, Distinct populations of forebrain neural stem and progenitor cells can be isolated using side-population analysis. J Neurosci, 2003. 23(33): p. 10703-9.

97. Behbod, F., et al., Transcriptional profiling of mammary gland side population cells. Stem Cells, 2006. 24(4): p. 1065-74.

98. Meeson, A.P., et al., Cellular and molecular regulation of skeletal muscle side population cells. Stem Cells, 2004. 22(7): p. 1305-20.

99. Liadaki, K., et al., Side population cells isolated from different tissues share transcriptome signatures and express tissue-specific markers. Exp Cell Res, 2005. 303(2): p. 360-74.

100. Zhou, S., et al., The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med, 2001. 7(9): p. 1028-34.

101. Bunting, K.D., et al., Transduction of murine bone marrow cells with an MDR1 vector enables ex vivo stem cell expansion, but these expanded grafts cause a myeloproliferative syndrome in transplanted mice. Blood, 1998. 92(7): p. 2269-79.

102. Bunting, K.D., et al., Enforced P-glycoprotein pump function in murine bone marrow cells results in expansion of side population stem cells in vitro and repopulating cells in vivo. Blood, 2000. 96(3): p. 902-9.

103. Jonker, J.W., et al., Contribution of the ABC transporters Bcrp1 and Mdr1a/1b to the side population phenotype in mammary gland and bone marrow of mice. Stem Cells, 2005. 23(8): p. 1059-65.

104. Chiba, T., et al., Side population purified from hepatocellular carcinoma cells harbors cancer stem cell-like properties. Hepatology, 2006. 44(1): p. 240-51.

Page 46: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

37

105. Mitsutake, N., et al., Characterization of side population in thyroid cancer cell lines: cancer stem-like cells are enriched partly but not exclusively. Endocrinology, 2007.

106. Szotek, P.P., et al., Ovarian cancer side population defines cells with stem cell-like characteristics and Mullerian Inhibiting Substance responsiveness. Proc Natl Acad Sci U S A, 2006. 103(30): p. 11154-9.

107. Ho, M.M., et al., Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Res, 2007. 67(10): p. 4827-33.

108. Wang, J., et al., Identification of cancer stem cell-like side population cells in human nasopharyngeal carcinoma cell line. Cancer Res, 2007. 67(8): p. 3716-24.

109. Haraguchi, N., et al., Characterization of a side population of cancer cells from human gastrointestinal system. Stem Cells, 2006. 24(3): p. 506-13.

110. Hadnagy, A., et al., SP analysis may be used to identify cancer stem cell populations. Exp Cell Res, 2006. 312(19): p. 3701-10.

111. Hirschmann-Jax, C., et al., A distinct "side population" of cells with high drug efflux capacity in human tumor cells. Proc Natl Acad Sci U S A, 2004. 101(39): p. 14228-33.

112. Patrawala, L., et al., Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+ and ABCG2- cancer cells are similarly tumorigenic. Cancer Res, 2005. 65(14): p. 6207-19.

113. Kondo, T., T. Setoguchi, and T. Taga, Persistence of a small subpopulation of cancer stem-like cells in the C6 glioma cell line. Proc Natl Acad Sci U S A, 2004. 101(3): p. 781-6.

114. Hiddemann, W., et al., Tumor heterogeneity in osteosarcoma as identified by flow cytometry. Cancer, 1987. 59(2): p. 324-8.

115. Zhou, J., et al., Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc Natl Acad Sci U S A, 2007. 104(41): p. 16158-63.

116. Wood, L.D., et al., The genomic landscapes of human breast and colorectal cancers. Science, 2007. 318(5853): p. 1108-13.

117. Doyle, L.A. and D.D. Ross, Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2). Oncogene, 2003. 22(47): p. 7340-58.

118. Wunder, J.S., et al., A comparison of staging systems for localized extremity soft tissue sarcoma. Cancer, 2000. 88(12): p. 2721-30.

119. Wang, X., et al., A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature, 2009. 461(7263): p. 495-500.

Page 47: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

38

120. Huntly, B.J., et al., MOZ-TIF2, but not BCR-ABL, confers properties of leukemic stem cells to committed murine hematopoietic progenitors. Cancer Cell, 2004. 6(6): p. 587-96.

121. Joseph, N.M., et al., The loss of Nf1 transiently promotes self-renewal but not tumorigenesis by neural crest stem cells. Cancer Cell, 2008. 13(2): p. 129-40.

122. Zheng, H., et al., Induction of abnormal proliferation by nonmyelinating schwann cells triggers neurofibroma formation. Cancer Cell, 2008. 13(2): p. 117-28.

123. Le, L.Q., et al., Cell of origin and microenvironment contribution for NF1-associated dermal neurofibromas. Cell Stem Cell, 2009. 4(5): p. 453-63.

124. Barker, N., et al., Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature, 2007. 449(7165): p. 1003-7.

125. Barker, N., et al., Crypt stem cells as the cells-of-origin of intestinal cancer. Nature, 2009. 457(7229): p. 608-11.

126. Dexter, T.M., T.D. Allen, and L.G. Lajtha, Conditions controlling the proliferation of haemopoietic stem cells in vitro. J Cell Physiol, 1977. 91(3): p. 335-44.

127. Bianco, P. and A. Boyde, Confocal images of marrow stromal (Westen-Bainton) cells. Histochemistry, 1993. 100(2): p. 93-9.

128. Dominici, M., et al., Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy, 2006. 8(4): p. 315-7.

129. Kolf, C.M., E. Cho, and R.S. Tuan, Mesenchymal stromal cells. Biology of adult mesenchymal stem cells: regulation of niche, self-renewal and differentiation. Arthritis Res Ther, 2007. 9(1): p. 204.

130. Sarugaser, R., et al., Human mesenchymal stem cells self-renew and differentiate according to a deterministic hierarchy. PLoS One, 2009. 4(8): p. e6498.

131. Dellavalle, A., et al., Pericytes of human skeletal muscle are myogenic precursors distinct from satellite cells. Nat Cell Biol, 2007. 9(3): p. 255-67.

132. Watt, F.M. and B.L. Hogan, Out of Eden: stem cells and their niches. Science, 2000. 287(5457): p. 1427-30.

133. da Silva Meirelles, L., P.C. Chagastelles, and N.B. Nardi, Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci, 2006. 119(Pt 11): p. 2204-13.

134. Lazarus, H.M., et al., Human bone marrow-derived mesenchymal (stromal) progenitor cells (MPCs) cannot be recovered from peripheral blood progenitor cell collections. J Hematother, 1997. 6(5): p. 447-55.

Page 48: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

39

135. Wexler, S.A., et al., Adult bone marrow is a rich source of human mesenchymal 'stem' cells but umbilical cord and mobilized adult blood are not. Br J Haematol, 2003. 121(2): p. 368-74.

136. Farrington-Rock, C., et al., Chondrogenic and adipogenic potential of microvascular pericytes. Circulation, 2004. 110(15): p. 2226-32.

137. Hirschi, K.K. and P.A. D'Amore, Pericytes in the microvasculature. Cardiovasc Res, 1996. 32(4): p. 687-98.

138. Andreeva, E.R., et al., Continuous subendothelial network formed by pericyte-like cells in human vascular bed. Tissue Cell, 1998. 30(1): p. 127-35.

139. Crisan, M., et al., A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell, 2008. 3(3): p. 301-13.

140. Riggi, N., et al., Development of Ewing's sarcoma from primary bone marrow-derived mesenchymal progenitor cells. Cancer Res, 2005. 65(24): p. 11459-68.

141. Tirode, F., et al., Mesenchymal stem cell features of Ewing tumors. Cancer Cell, 2007. 11(5): p. 421-9.

142. Matushansky, I., et al., Derivation of sarcomas from mesenchymal stem cells via inactivation of the Wnt pathway. J Clin Invest, 2007. 117(11): p. 3248-57.

143. McAdam, W.A. and J.C. Goligher, The occurrence of desmoids in patients with familial polyposis coli. Br J Surg, 1970. 57(8): p. 618-31.

144. Burke, A.P., et al., Intra-abdominal fibromatosis. A pathologic analysis of 130 tumors with comparison of clinical subgroups. Am J Surg Pathol, 1990. 14(4): p. 335-41.

145. Alman, B.A., et al., Aggressive fibromatosis (desmoid tumor) is a monoclonal disorder. Diagn Mol Pathol, 1997. 6(2): p. 98-101.

146. Lewis, J.J., et al., The enigma of desmoid tumors. Ann Surg, 1999. 229(6): p. 866-72; discussion 872-3.

147. Shields, C.J., et al., Desmoid tumours. Eur J Surg Oncol, 2001. 27(8): p. 701-6.

148. Alman, B.A., et al., Aggressive fibromatosis. J Pediatr Orthop, 1992. 12(1): p. 1-10.

149. Reitamo, J.J., T.M. Scheinin, and P. Hayry, The desmoid syndrome. New aspects in the cause, pathogenesis and treatment of the desmoid tumor. Am J Surg, 1986. 151(2): p. 230-7.

150. Khorsand, J. and C.P. Karakousis, Desmoid tumors and their management. Am J Surg, 1985. 149(2): p. 215-8.

151. Faulkner, L.B., et al., Pediatric desmoid tumor: retrospective analysis of 63 cases. J Clin Oncol, 1995. 13(11): p. 2813-8.

Page 49: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

40

152. Gurbuz, A.K., et al., Desmoid tumours in familial adenomatous polyposis. Gut, 1994. 35(3): p. 377-81.

153. Hizawa, K., et al., Desmoid tumors in familial adenomatous polyposis/Gardner's syndrome. J Clin Gastroenterol, 1997. 25(1): p. 334-7.

154. Rodriguez-Bigas, M.A., et al., Desmoid tumors in patients with familial adenomatous polyposis. Cancer, 1994. 74(4): p. 1270-4.

155. Alman, B.A., et al., Increased beta-catenin protein and somatic APC mutations in sporadic aggressive fibromatoses (desmoid tumors). Am J Pathol, 1997. 151(2): p. 329-34.

156. Tejpar, S., et al., Predominance of beta-catenin mutations and beta-catenin dysregulation in sporadic aggressive fibromatosis (desmoid tumor). Oncogene, 1999. 18(47): p. 6615-20.

157. Smits, R., et al., Apc1638N: a mouse model for familial adenomatous polyposis-associated desmoid tumors and cutaneous cysts. Gastroenterology, 1998. 114(2): p. 275-83.

158. Fodde, R., et al., A targeted chain-termination mutation in the mouse Apc gene results in multiple intestinal tumors. Proc Natl Acad Sci U S A, 1994. 91(19): p. 8969-73.

Page 50: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

41

CHATPER 2

Side population cells isolated from mesenchymal neoplasms have

tumor initiating potential

I preformed all experiments describe in the paper with equal contribution from Qingxia Wei with the following exceptions:

• Velani Utomo helped in processing primary tumor samples • Puviindran Nadesan helped in managing the mouse colony • Heather Whetstone preformed the hemotoxylin and eosin staining (Figure 2.3) • Rita Kandel preformed pathological analysis of the tumor samples (Figure 2.3) • Jay Wunder provided human tumor samples for the side population experiments

Page 51: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

42

Chapter 2

2 Side population cells isolated from mesenchymal neoplasms contain tumor initiating cells

2.1 Abstract

Although many cancers are maintained by tumor initiating cells, this has not been demonstrated

for mesenchymal tumors, in part due to the lack of unique surface markers that identify

mesenchymal progenitors. An alternative technique to isolate stem-like cells is to isolate side

population cells based on efflux of Hoechst 33342 dye. We examined 29 mesenchymal tumors

ranging from benign to high grade sarcomas, and identified a side population of cells in all but

six samples. There was a positive correlation between the percentage of side population cells and

the grade of the tumor. Side population cells preferentially formed tumors when grafted into

immunodeficient mice, and only cells from tumors that developed from the side population cells

had the ability to initiate tumor formation upon serial transplantation. Although the side

population cells are able to efflux Rhodamine dye in addition to Hoechst 33342, we found that

the ability to efflux Rhodamine dye did not identify a population of cells enriched for tumor

initiating capacity. Here we identify a subpopulation of cells within a broad range of benign and

malignant mesenchymal tumors with tumor initiating capacity. In addition, our data suggests that

the proportion of side population cells could be used as a prognostic factor, and that

therapeutically targeting this subpopulation of cells could be used to improve patient outcome.

Page 52: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

43

2.2 Introduction

Solid tumors are composed of a heterogeneous population of cells. These cells have different in

vitro proliferative capacities; only a minority have the ability to initiate tumor formation in

immuno-deficient mice. This observation led to the concept of cancer stem cells (CSC) which

have the ability to self-renew and differentiate. By manipulating these characteristics, CSCs

have been postulated to be responsible for driving the growth of tumors and the recurrence of

neoplasms after therapy (1, 2). Although these cells have been identified in a variety of

malignancies, such as hematologic, neural and epithelial cancers, they have not been identified in

neoplasms of mesenchymal origin. The solid tumors from which CSCs have been isolated are

not of mesenchymal origin. As such, cell surface markers used in these tumors may not be

successful for the isolation of these CSCs from mesenchymal neoplasms (3-11). Furthermore,

there are no unique markers for mesenchymal stem cells. Fortunately, other properties of stem

cells can be used to isolate cells with progenitor characteristics.

One such property is the ability of stem cells to efflux chemotherapeutic drugs and certain dyes

such as Hoechst 33342 (12, 13). This feature is conferred partly by high levels of expression of

ABC transporter proteins on primitive progenitor cells (14, 15). During flow cytometry analysis,

negatively stained cells fall to the “side” of the majority, hence they are commonly referred to as

side population (SP) cells (16). They enrich for progenitor cells from a variety of tissue types

(17-20); furthermore, data has proven that some cancer cell lines and primary tumors contain SP

cells. It is the SP cells that confer tumorgenicity of the tumor or cancer cell line (21, 22). This

technique provides an alternative means to isolate progenitor cells other than through the use of

specific surface markers, and may be one way to identify putative tumor initiating cells.

Primary musculoskeletal tumors are of mesenchymal origins. They range from benign lesions,

such as the soft tissue aggressive fibromatosis (desmoid), to very aggressive malignancies such

as synovial sarcomas. Mesenchymal progenitors express a variety of markers, such as Stro-1,

CD105, and CD44; however, to date no marker or pattern of markers universally defines a

consensus phenotype for a mesenchymal stem cell. This has hampered research into the cell of

origin of these lesions (23-26). Recent in vitro evidence suggests that mesenchymal tumors may

contain cells with stem-like characteristics. Cells within these tumors had the capacity to form

spheres similar to those formed by neural stem cells. Furthermore, these spheres preferentially

Page 53: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

44

expressed genes that are involved in regulating stem cell fate(27). Despite this, the ability to

identify and isolate tumor initiating cells (TICs) within mesenchymal tumors has proved to be

elusive.

The initiation and progression of these tumor types is poorly understood. These lesions can have

a high rate of recurrence suggesting that a small population of cells have the potential to escape

treatment and may have tumor initiating cell characteristics (28). SP cells, which are enriched for

TICs, efflux dye through a mechanism that is similar to that used by chemotherapeutic resistant

cells. Therefore, TICs may be relatively resistant to chemotherapeutic drugs and may confer the

malignant phenotype to tumors (13, 29, 30). As such, these cells could be responsible for

sarcoma recurrence after chemotherapy. Here we report, for the first time, the identification of

SP cells from mesenchymal tumors. These cells have the capacity to initiate tumor formation.

Further investigation of these TICs cells may lead to the development of more effective and

efficient targeted treatment modalities.

Page 54: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

45

2.3 Results

2.3.1 Mesenchymal tumors contain side population (SP) cells

We sought to determine whether we could isolate tumor initiating cells from primary

mesenchymal tumors based on the observation that stem-like cells have the ability to efflux

Hoecsht 33342 dye (16). Cells from 29 primary mesenchymal tumors, ranging from locally

invasive lesions to high grade sarcomas were incubated with the fluorescent dye, Hoechst 33342.

Cells were also incubated in the presence of verapamil, a chemical inhibitor of the ABC protein

family of transporters, which inhibits the efflux of Hoechst (15). A distinct SP was found in all

of the 29 tumors examined except for one dermatofibrosarcoma protuberans, one myxoid

chondrosarcoma, one of three chondrosarcomas, and one of two leiomyosarcomas. With the

addition of verapamil, the presence of SP cells was abolished, indicating that dye efflux

occurred, in part, through an ABC transporter regulated mechanism (Figure 2.1). The presence of

a SP within these tumors, raises the possibility this represents a subpopulation of cells with

tumor initiating characteristics.

2.3.2 The proportion of side population cells correlates with aggressiveness of the tumor

Mesenchymal tumors are highly heterogeneous groups of neoplasms with varying levels of

histologic and clinical aggressiveness. (28). We observed a trend between the proportion of SP

cells present in a given tumor and the relative aggressiveness of the tumor (Figure. 2.2). In

general, higher grade tumors had an increased prevalence of SP cells. This suggests that the

proportion of SP cells may be a predictor of patient outcome. Furthermore, cells in the side

population may be responsible for the maintenance and propagation of mesenchymal tumors

giving the high grade tumors a more aggressive behavior. However numbers of individual tumor

types were too small to determine if within a particular tumor type the percentage of cells sorting

to the SP correlated with grade, aggressiveness, or clinical outcome.

2.3.3 SP cells have the capacity to form tumors upon serial transplantation in NOD/SCID mice

We tested the ability of SP cells to initiate tumor formation when grafted into NOD/SCID mice.

Cells from one osteosarcoma, two malignant fibrous histocytomas, and one synovial sarcoma

Page 55: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

46

were stained with Hoechst 33342 dye and sorted into SP and non-SP fractions. Both fractions

were then subcutaneously injected into NOD/SCID mice. Initially, both SP and non-SP fractions

had the capacity to form tumors, however, significantly smaller numbers of SP cells formed

tumors at a significantly higher frequency when compared to non-SP cells (Table 1.1). The SP

from each of the individual tumors examined showed the same enhanced ability to form tumors

in NOD/SCID mice. Furthermore, tumors that did form from the non-SP fraction, even though

they formed from larger numbers of injected cells, were significantly smaller when compared to

tumors from the SP fraction (0.30 –vs- 1.08 gm, p<0.01). Tumors expressed human GAPDH

and had a nearly identical cytology to the primary sarcoma (Figure 2.3).

To test for self-renewal (34), SP and non-SP tumors were dissociated and cells from both tumors

were re-stained with Hoechst 33342 dye. SP re-analysis demonstrated that only cells derived

from SP tumors were able to repopulate both SP and NSP fractions (Figure 2.4A). The

percentage of SP present from xenografted tumors was similar to that of the parental tumor cells

suggesting that SP cells had the ability to recapitulate the pheonotypic distribution of the original

tumor. When labeled with propidium iodide, there was significant decrease in viable cells in the

tumors derived from the non-SP compared to the cells derived from SP tumors (Figure 2.4B).

Viable cells from non-SP tumors had an increase in DNA content when compared to the SP

tumor cells suggesting a higher mitotic rate (Figure 2.4 C). Furthermore even after 32 weeks,

non-SP tumor cells injected into NOD/SCID failed to initiate tumor growth. Taken together, this

suggests non-SP tumors represent a population of cells with only transient amplifying potential.

To determine the tumorigenic potential of the cells from the xenograft tumors, we next serially

transplanted SP and non-SP cells from the SP tumors and non-SP tumors. In the secondary

transplant, cells from non-SP tumors did not initiate tumors in the NOD/SCID mice. Only the

SP fraction from tumors from the initial SP fraction had the capacity to initiate tumor formation.

Furthermore, as few as 100 SP cells resulted in the formation of tumors whereas 10,000 non-SP

cells failed to form tumors (Table 1.1). These secondary xenografted tumors had an identically

histological appearance to both primary xenografted tumors and to original parental tumors

(Figure 2.3 C). Hence, only tumors derived from SP cells can self-renew and differentiate in a in

a manner that can be propagated through serial transplantation into NOD/SCID mice.

Page 56: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

47

2.3.4 SP Cells Efflux Rhodamine-123

The phenomenon of dye efflux is not exclusive to Hoechst 33342. For instance, Rhodamine-123

efflux can be used to identify populations enriched for hematopoetic stem cells from mouse bone

marrow. We next examined the ability of Rhodamine-123 efflux to recapitulate the results

demonstrated with Hoechst efflux. Staining with Rhodamine-123 resulted in a broad range of

florescent uptake. Two populations were sorted based on gating of the upper and lower 10% of

live cells, hereafter referred to as Rholow and Rhohigh. Cells were also dual stained with both

Rhodamine-123 and Hoechst 33342. We noted that the majority of SP cells did not stain highly

for Rhodamine-123 while the staining pattern of non-sp cells was identical to the general

population of cells (Figure 2.5 A). Thus, SP cells have the capacity to efflux Rhodamine-123 and

the two populations were not mutually exclusive. Rholow, Rhohigh , Rhohigh /non-SP, Rholow/SP

cells were sorted and injected into NOD/SCID mice (Figure 2.5 B). While both Rhodamine-123

populations failed to form tumors after 12 weeks, as few as 375 Rholow/SP cells did initiate

tumor formation in 2/2 mice. This corresponded to the number of cells required to form tumors

from SP cells, suggesting that the capacity to efflux Rhodamine-123 does not enhance the

tumorigenic potential of SP cells.

2.4 Discussion

Here we demonstrate for the first time that mesenchymal tumors contain a subpopulation of cells

with tumor initiating capacity, which can be identified based on their exclusion of Hoescht

33342 dye. Intriguingly, there are higher proportions of SP cells in high grade mesenchymal

malignancies than in less aggressive benign lesions. While tumor initiating cells (TICs) have

been identified in hematologic, neural, and epithelial cancer (3-11), to date, this has not been

proven for mesenchymal tumors whose origins differ from other solid tumor. Our data suggests

that TICs are present in a broad range of benign and malignant neoplastic processes, and as such,

are a general phenomenon in tumorigenesis.

The identification of TICs in mesenchymal tumors has proven to be more elusive than in tumors

that originate from other tissue types, in part due to the lack of universal markers for

mesenchymal progenitor cells (23, 25, 26). Other techniques to isolate stem cells include in-vitro

functional techniques. Indeed, recent work has shown that in an in vitro culture system, cells

derived from mesenchymal tumors have the capacity to form spheres similar to those derived

Page 57: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

48

from neural stem cells(27). While these “sarcospheres” preferentially express “stemness” genes

in comparison to their adherent counterparts, it has not been demonstrated that these cells have a

preferential ability to form tumors when grafted into immuno-deficient mice.

Given these obstacles, we chose to use Hoecsht dye exclusion as an alternative method of stem

cell isolation. While this method has been successfully utilized to isolate stem-like cells in a

variety of cell lines(21), it is infrequently used in primary tumors(22). We found that cells from

most primary mesenchymal tumors contained a side population and that these cells have the

capacity to initiate tumors when transplanted into immuno-deficient mice. Furthermore, upon

serial transplantation, only SP tumors had the ability to re-initiate the tumor in immuno-deficient,

suggesting that only this population is able to self-renew in vivo.

Large numbers of non-SP cells also formed tumors in NOD/SCID mice, and others demonstrated

that in breast, prostate, and thyroid cancer cell lines, large numbers of non-SP cells have the

capacity to initiate tumor formation. It has been suggested that this finding is due to

contamination of small numbers of SP cells in the non-SP fraction(35-37). However in our

experiments, while cells from the non-SP fraction were initially able to form tumors in

NOD/SCID mice, when re-stained with Hoechst dye they did not contain an SP fraction. They

also exhibited high levels of cell death as demonstrated by a high level of propridium iodide

staining. Furthermore, analysis of DNA content demonstrated that these cells have an increase in

DNA content when compared to cells derived from SP tumors suggesting that non-SP tumor

cells have an increased proliferation rate. Importantly, non-SP tumor cells were not able to

initiate tumors after serial transplantation even after 32 weeks post injection. This suggests non-

SP cells are fundamentally different from their SP counterparts. Given these observations, it is

possible that cells from the non-SP fraction represent a more differentiated subpopulation,

characterized by a short term proliferative potential, such as a transient amplifying cell. Taken

together, this strongly suggests that Hoescht dye exclusion will enrich for cells with the capacity

to divide asymmetrically and to self-renew, a key feature of stem cells.

As Hoescht dye exclusion selects for cells with the capacity to exclude dye via the expression of

protein transporters(15, 38), we cannot exclude the possibility that SP cells are more mature

tumor cells that have acquired the ability to increase the expression of genes responsible for this

pumping mechanism. Such cells might also be resistant to chemotherapy and may be responsible

Page 58: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

49

for the relapse of disease (13). To address this issue, we isolated cells that effluxed the

fluorescent dye, Rhodamine-123. Unlike SP cells, neither Rholow nor Rhohigh cells had the

capacity to initiate tumor formation, however, we cannot exclude the possibility that our gating

strategy was too generous for enrichment of tumor initiating cells and that with increased

stringency there may be tumor initiating cells in the Rholow population. Despite this, our data

suggests that the capacity to efflux materials may not the sole determinant of tumor initiating

cells since SP but not Rholow cells had the capacity enrich for TICs. In any event, we have shown

that SP cells are distinct from their non-SP counterparts, and are enriched for TICs. A more in-

depth analysis of this population may provide important clues into sarcoma biology.

A striking result of our study is the correlation seen between the percentage of SP cells and the

aggressiveness of the mesenchymal tumor examined. We found that benign, locally invasive

low-grade tumors had a low SP population whereas high-grade malignant tumors had high SP

populations. This data implies that SP is a predictor of patient outcome, however, prospective

studies are needed to determine if this hypothesis will be proven to be correct.

The high proportion of SP cells in malignant tumors might also correlate with chemotherapeutic

resistance, as this resistance may be due in part to the over-expression of transporter proteins

which efflux drugs. As we have shown that SP cells have the capacity to efflux material and have

enhanced tumorigenic potential when compared to non-SP cells, it is interesting to hypothesize

that the efflux mechanism plays a role in chemotherapeutic resistance. Perhaps the expression of

transporter proteins would correlate with both outcome and the proportion of SP cells. Hoechst

dye exclusion is conferred in part by the expression of the ABCG2 transporter protein while

Rhodamine dye exclusion is mediated by ABCB1/P-gp protein (15). Previous data has shown no

correlation between the expression of ABCB1/P-gp in soft tissue sarcomas and tumor grade and

not surprisingly, Rholow cells failed to initiate tumor formation(39, 40). To date the role of

ABCG2 transporter protein in soft tissue sarcomas has yet to be elucidated. However, it should

be noted that Bcrp1/Mdr1a/b triple knock-out mice still exhibit SP and it is likely multiple

transporters are involved in this process(41).

Our studies demonstrate for the first time, the existence of SP cells in primary mesenchymal

tumors. Furthermore, we found that these cells are enriched for tumor initiating cells. There

appears to be a direct correlation between the number of SP cells present in a tumor and the

Page 59: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

50

aggressiveness of the tumor. Targeting this side population has the potential to be developed into

an effective treatment modality for mesenchymal tumors.

Page 60: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

51

Figures

Figure 2.1 Mesenchymal tumors contain side population cells

(A) Cells from a representative grade two primary sarcoma were stained with Hoechst 33342 and

analyzed by flow cytometry. The SP from a representative tumor is shown. The SP cells are

outlined and shown as a percentage of the total cell population. (B) This cell population

disappears in the presence of verapamil.

Page 61: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

52

Page 62: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

53

Figure 2.2 High grade sarcomas have an increased prevalence of SP cells when compared to lower grade lesions.

(A) SP analysis from a benign but locally aggressive aggressive fibromatosis, a grade two

liposarcoma and a grade three malignant fibrous histiocytoma are shown. SP cells are outlined

and shown as a percentage of the total cell population. (B) Mean and 95% confidence intervals

for the percent of SP cells from benign and various grade lesions. A 95% confidence interval that

does not cross the mean of a comparison is a statistically significant difference at p<0.05. There

are significantly higher proportions of SP cells in higher grade tumors.

Page 63: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

54

Page 64: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

55

Table 2.1 The proportion of tumors that formed from injection of various numbers of cells

from each subpopulation into NOD/SCID mice.

Primary mice were injected with cells sorted from the primary tumor, and secondary mice are

injected with cells sorted from the tumor that developed in primary mice. Side population cells

from each tumor type examined formed tumors from the side population fraction at each cell

number examined. In contrast, only in a few instances did non-side population cells formed

tumors in primary mice. Larger numbers of injected cells were required for the non-side

population cells to form tumors. The side population fraction from tumors that formed in the

primary mice were able to form tumors in secondary mice, while in only one case would cells

from the non-side population form a tumor. None of the cells from the primary tumors that

formed from non-side population cells were able to form tumors in secondary mice.

Abbreviation: nt not tested

Page 65: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

56

Cell Type Cell Dose

(number of primary mice with tumors)/(total number injected)

(number of secondary mice with tumors)/(total number injected)

Total number of mice with tumors (percent)

Side Population 1x102 11/16 5/8 16/24 (67)

5 x 102 5/5 nt 5/5 (100)

1x103 8/10 9/12 17/24 (71)

1x104 8/10 4/4 12/14 (86)

1x105 nt nt

Non Side Population 1x102 0/8 0/8 0/16 (0)

5 x 102 0/4 nt 0/4 (0)

1x103 3/14 0/12 3/26 (12)

1x104 1/10 0/10 1/20 (5)

1x105 4/16 1/10 5/26 (19)

Page 66: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

57

Figure 2.3 Histopathologic features of SP tumors

A to C) SP cells from (A) parental, (B) primary, and (C) secondary xenografted tumors were

injected into NOD/SCID mice. After 12 weeks tumors were collected formalin fixed, paraffin

embedded and stained with H&E. The grafted tumors (B and C) formed had nearly identical

cytological appearances to the (A) primary tumors. (D) A representative NSP tumor also showed

similar cytologic appearances to the primary tumor. A representative malignant fibrous

histiocytoma is shown, and the size bar is 200um.

Page 67: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

58

Page 68: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

59

Figure 2.4 Characteristics of tumors derived from SP and non-SP cells.

Cells from derived from both SP and non-SP (NSP) primary xenograft tumors were reanalyzed

for SP cells . (A) Only SP tumors contained cells that had the capacity to reform both SP and

non-SP fractions . (B) Cell viability was determined by propidium iodide staining. 23% of cells

derived from SP tumors were viable, in contrast, only 8% of cells derived from non-SP tumors

were viable. (C) Cells from non-SP tumors had an increase in DNA content when compared to

the SP tumor cells suggesting a higher mitotic rate.

Page 69: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

60

0 1000 2000 3000 4000

FL9

0

1000

2000

3000

4000

FL6

1.79

0 1000 2000 3000 4000

FL9

0

1000

2000

3000

4000

FL6

0.16

0 1000 2000 3000 4000

FSC

100

101

102

103

104

FL4

23

0 1000 2000 3000 4000

FSC

100

101

102

103

104

FL4

7.95

SPN

SP

Hoechst 33342 Propidum Iodide DNA ContentA B C

0 1000 2000 3000 4000

PE-A

0

100

200

300

# Cells

0 1000 2000 3000 4000

PE-A

0

50

100

150

200

250

# Cells

Page 70: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

61

Figure 2.5 SP cells efflux Rhodamine-123

Cells derived from a primary MFH sample were dual stained with both Hoechst 33342 and

Rhodamine-123. (A) Twenty percent cutoffs were used to sort Rhohigh and Rholow fractions.

Shared profiles indicate SP cells (blue) have decreased Rhodamine-123 staining when compared

to the total population of cells (green). The non-SP cells (red) have a staining patter similar to

that of the total population of cells. (B) Cells were sorted into SP, non-SP, Rhohigh, Rholow,

SP/Rholow, non-SP/Rhohigh fractions and varying cell numbers were injected into NOD/SCID

mice. Tumor formation was assessed 12 weeks post injection. Numbers indicate the number of

tumors that formed/number of injections.

Page 71: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

62

0 1000 2000 3000 4000

FL9

0

1000

2000

3000

4000

FL6

0.11

68.3

100

101

102

103

104

DSP(Comp FL1)

0

1000

2000

3000

4000

# Cells

20.2 20.6

100

101

102

103

104

DSP(Comp FL1)

0

20

40

60

80

100

% of Max

Hoechst 33342 Rhodamine 123

Cells injected

Sample Population 100 375 1000 5000

T-1 MFH SP 3/8 6/8NSP 0/8 0/8

T-1 MFH Rholow 0/6 0/4Rhohigh 0/5 0/4

T-1 MFH SP Rholow 2/2NSPRhohigh 0/4

A

B

Page 72: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

63

2.5 Materials and Methods

2.5.1 Primary Tumors

29 mesenchymal tumors were processed at the time of surgical excision. Local ethical approval

was obtained for all tissue samples collected. The samples included 7 aggressive fibromatosis, 5

osteosarcomas, 3 chondrosarcomas, 3 synovial sarcomas, 2 leiomyosarcoma, 4 malignant fibrous

histocytomas, 1 myxoid liposarcoma, 1 pleomorphic liposarcoma, 1 dermatofibrosarcoma

protuberans, 1 myxoid chondrosarcoma, and 1 chordoma. Primary tumor samples were manually

minced and all visible clumps removed. Enzymatic digestion followed at 37oC for 45 minutes

with constant rotation using 10mg/ml of collagenase IV (Worthington), 2.4 U/ml of Dispase

(Becton Dickinson), 0.05% trypsin (Wisent). Further manual dissociation was performed by

passing the cell slurry through an 18 gauge needle. Cells were then centrifuged at 1400 rpm for 5

minutes and washed three times in PBS. After washing, cells were strained through 70µm filters

to remove remaining clumps. Collected cells were plated in alpha-MEM supplemented with L-

glutamine and containing 18% fetal bovine serum (Wisent) and cultured at 37oC with 5% CO2 in

a humidified chamber until subjected to fluorescent activated cells sorting (FACS).

2.5.2 Pathology

Formalin fixed, paraffin embedded samples were stained with hematoxylin and eosin (H&E) and

observed in a blinded manner by an experienced pathologist. Tumors were graded into benign or

malignant groups. Each tumor was sub-classified according to the WHO classification and

graded using the standard 3 scale American Joint Commission on Staging (31-33).

2.5.3 Flow Cytometry

Cells were trypsinized and resuspended in PBS supplemented with 2% FBS at a concentration of

1X106 cells/ml. For SP assays, cells were treated either alone with 2.5 µg/ml of Hoechst 33342

dye (Sigma) for 90 minutes or with 0.1ug/ml at 37oC, or in combination with 50 µM verapamil

(Sigma), an inhibitor of ABC transporters. For Rhodamine-123 staining, 1X106 cells/ml were

incubated with 0.1µg/ml of Rhodamine-123 (Molecular Probes) for 30 minutes at 37oC. For

analysis of Rhodamine-123 and Hoechst 33342 efflux activity, cells were initially incubated with

0.1µg/ml of Rhodamine-123 for 30 minutes at 37oC, washed and then re-suspended in the same

cell concentration with 2.5µg/ml of Hoechst dye for 90 minutes at 37oC. Cells were

Page 73: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

64

counterstained with 1µg/ml of propidium iodide (Molecular Probes), and non-viable cells were

excluded from both analysis and sorting assays. To detect for SP, cells were analyzed by using a

dual wavelength analysis (blue, 424-444nm; red, 675nM) after excitation with 350nm UV light

(MoFlow, Cytomation). SP and non-SP cells were collected and either grown in culture medium

for in vitro studies, or injected into NOD/SCID mice. For Rhodamine-123 staining, ten percent

cutoffs were used to sort Rhohigh and Rholow fractions.

2.5.4 Cell Transplantation into NOD/SCID mice

Sorted SP and non-SP cells were collected and cells were resuspended in PBS at concentrations

ranging from 200-20,000 cells/50µl. Cells were then mixed with 50µl of Matrigel (Becton

Dickinson). This cell:matrigel suspension was then subcutaneously injected in to eight to ten

week old NOD/SCID mice. Mice were observed for up to 12 weeks after which they were

euthanized and tumor formation was assessed. Tumors that formed were removed and samples

from each tumor were harvested for FACS and histology. For FACS, tumors were dissociated, as

for the primary tumors and resorted into SP and NSP fractions. These secondary sorted cells

were then re-injected into eight to ten week old NOD/SCID mice and tumor formation was

assessed after 12 weeks. Samples of tumors were formalin fixed and processed for histology.

Page 74: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

65

2.6 References

1. Al-Hajj, M. and Clarke, M. F. Self-renewal and solid tumor stem cells. Oncogene, 23:

7274-7282, 2004.

2. Pardal, R., Clarke, M. F., and Morrison, S. J. Applying the principles of stem-cell biology

to cancer. Nat Rev Cancer, 3: 895-902, 2003.

3. Bonnet, D. and Dick, J. E. Human acute myeloid leukemia is organized as a hierarchy

that originates from a primitive hematopoietic cell. Nat Med, 3: 730-737, 1997.

4. Singh, S. K., Hawkins, C., Clarke, I. D., Squire, J. A., Bayani, J., Hide, T., Henkelman,

R. M., Cusimano, M. D., and Dirks, P. B. Identification of human brain tumour initiating

cells. Nature, 432: 396-401, 2004.

5. Singh, S. K., Clarke, I. D., Terasaki, M., Bonn, V. E., Hawkins, C., Squire, J., and Dirks,

P. B. Identification of a cancer stem cell in human brain tumors. Cancer Res, 63: 5821-

5828, 2003.

6. Al-Hajj, M., Wicha, M. S., Benito-Hernandez, A., Morrison, S. J., and Clarke, M. F.

Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A,

100: 3983-3988, 2003.

7. Li, C., Heidt, D. G., Dalerba, P., Burant, C. F., Zhang, L., Adsay, V., Wicha, M., Clarke,

M. F., and Simeone, D. M. Identification of pancreatic cancer stem cells. Cancer Res, 67:

1030-1037, 2007.

8. O'Brien, C. A., Pollett, A., Gallinger, S., and Dick, J. E. A human colon cancer cell

capable of initiating tumour growth in immunodeficient mice. Nature, 445: 106-110,

2007.

9. Prince, M. E., Sivanandan, R., Kaczorowski, A., Wolf, G. T., Kaplan, M. J., Dalerba, P.,

Weissman, I. L., Clarke, M. F., and Ailles, L. E. Identification of a subpopulation of cells

with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl

Acad Sci U S A, 104: 973-978, 2007.

Page 75: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

66

10. Ricci-Vitiani, L., Lombardi, D. G., Pilozzi, E., Biffoni, M., Todaro, M., Peschle, C., and

De Maria, R. Identification and expansion of human colon-cancer-initiating cells. Nature,

445: 111-115, 2007.

11. Patrawala, L., Calhoun, T., Schneider-Broussard, R., Li, H., Bhatia, B., Tang, S., Reilly,

J. G., Chandra, D., Zhou, J., Claypool, K., Coghlan, L., and Tang, D. G. Highly purified

CD44+ prostate cancer cells from xenograft human tumors are enriched in tumorigenic

and metastatic progenitor cells. Oncogene, 25: 1696-1708, 2006.

12. Challen, G. A. and Little, M. H. A side order of stem cells: the SP phenotype. Stem Cells,

24: 3-12, 2006.

13. Dean, M., Fojo, T., and Bates, S. Tumour stem cells and drug resistance. Nat Rev Cancer,

5: 275-284, 2005.

14. Bunting, K. D. ABC transporters as phenotypic markers and functional regulators of stem

cells. Stem Cells, 20: 11-20, 2002.

15. Zhou, S., Schuetz, J. D., Bunting, K. D., Colapietro, A. M., Sampath, J., Morris, J. J.,

Lagutina, I., Grosveld, G. C., Osawa, M., Nakauchi, H., and Sorrentino, B. P. The ABC

transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular

determinant of the side-population phenotype. Nat Med, 7: 1028-1034, 2001.

16. Goodell, M. A., Brose, K., Paradis, G., Conner, A. S., and Mulligan, R. C. Isolation and

functional properties of murine hematopoietic stem cells that are replicating in vivo. J

Exp Med, 183: 1797-1806, 1996.

17. Chiba, T., Kita, K., Zheng, Y. W., Yokosuka, O., Saisho, H., Iwama, A., Nakauchi, H.,

and Taniguchi, H. Side population purified from hepatocellular carcinoma cells harbors

cancer stem cell-like properties. Hepatology, 44: 240-251, 2006.

18. Redvers, R. P., Li, A., and Kaur, P. Side population in adult murine epidermis exhibits

phenotypic and functional characteristics of keratinocyte stem cells. Proc Natl Acad Sci

U S A, 103: 13168-13173, 2006.

Page 76: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

67

19. Jackson, K. A., Mi, T., and Goodell, M. A. Hematopoietic potential of stem cells isolated

from murine skeletal muscle. Proc Natl Acad Sci U S A, 96: 14482-14486, 1999.

20. Asakura, A. and Rudnicki, M. A. Side population cells from diverse adult tissues are

capable of in vitro hematopoietic differentiation. Exp Hematol, 30: 1339-1345, 2002.

21. Kondo, T., Setoguchi, T., and Taga, T. Persistence of a small subpopulation of cancer

stem-like cells in the C6 glioma cell line. Proc Natl Acad Sci U S A, 101: 781-786, 2004.

22. Hirschmann-Jax, C., Foster, A. E., Wulf, G. G., Nuchtern, J. G., Jax, T. W., Gobel, U.,

Goodell, M. A., and Brenner, M. K. A distinct "side population" of cells with high drug

efflux capacity in human tumor cells. Proc Natl Acad Sci U S A, 101: 14228-14233,

2004.

23. Zannettino, A. C., Harrison, K., Joyner, C. J., Triffitt, J. T., and Simmons, P. J. Molecular

cloning of the cell surface antigen identified by the osteoprogenitor-specific monoclonal

antibody, HOP-26. J Cell Biochem, 89: 56-66, 2003.

24. Haynesworth, S. E., Baber, M. A., and Caplan, A. I. Cell surface antigens on human

marrow-derived mesenchymal cells are detected by monoclonal antibodies. Bone, 13: 69-

80, 1992.

25. Tondreau, T., Meuleman, N., Delforge, A., Dejeneffe, M., Leroy, R., Massy, M., Mortier,

C., Bron, D., and Lagneaux, L. Mesenchymal stem cells derived from CD133-positive

cells in mobilized peripheral blood and cord blood: proliferation, Oct4 expression, and

plasticity. Stem Cells, 23: 1105-1112, 2005.

26. Simmons, P. J. and Torok-Storb, B. Identification of stromal cell precursors in human

bone marrow by a novel monoclonal antibody, STRO-1. Blood, 78: 55-62, 1991.

27. Gibbs, C. P., Kukekov, V. G., Reith, J. D., Tchigrinova, O., Suslov, O. N., Scott, E. W.,

Ghivizzani, S. C., Ignatova, T. N., and Steindler, D. A. Stem-like cells in bone sarcomas:

implications for tumorigenesis. Neoplasia, 7: 967-976, 2005.

28. Olstad, O. K., Gautvik, V. T., Reppe, S., Rian, E., Jemtland, R., Ohlsson, C., Bruland, O.

S., and Gautvik, K. M. Molecular heterogeneity in human osteosarcoma demonstrated by

Page 77: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

68

enriched mRNAs isolated by directional tag PCR subtraction cloning. Anticancer Res,

23: 2201-2216, 2003.

29. Doyle, L. A., Yang, W., Abruzzo, L. V., Krogmann, T., Gao, Y., Rishi, A. K., and Ross,

D. D. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc

Natl Acad Sci U S A, 95: 15665-15670, 1998.

30. Kim, M., Turnquist, H., Jackson, J., Sgagias, M., Yan, Y., Gong, M., Dean, M., Sharp, J.

G., and Cowan, K. The multidrug resistance transporter ABCG2 (breast cancer resistance

protein 1) effluxes Hoechst 33342 and is overexpressed in hematopoietic stem cells. Clin

Cancer Res, 8: 22-28, 2002.

31. Fletcher, C. D. The evolving classification of soft tissue tumours: an update based on the

new WHO classification. Histopathology, 48: 3-12, 2006.

32. Levine, E. A. Prognostic factors in soft tissue sarcoma. Semin Surg Oncol, 17: 23-32,

1999.

33. Wunder, J. S., Healey, J. H., Davis, A. M., and Brennan, M. F. A comparison of staging

systems for localized extremity soft tissue sarcoma. Cancer, 88: 2721-2730, 2000.

34. Reya, T., Morrison, S. J., Clarke, M. F., and Weissman, I. L. Stem cells, cancer, and

cancer stem cells. Nature, 414: 105-111, 2001.

35. Patrawala, L., Calhoun, T., Schneider-Broussard, R., Zhou, J., Claypool, K., and Tang, D.

G. Side population is enriched in tumorigenic, stem-like cancer cells, whereas ABCG2+

and ABCG2- cancer cells are similarly tumorigenic. Cancer Res, 65: 6207-6219, 2005.

36. Mitsutake, N., Iwao, A., Nagai, K., Namba, H., Ohtsuru, A., Saenko, V., and Yamashita,

S. Characterization of side population in thyroid cancer cell lines: cancer stem-like cells

are enriched partly but not exclusively. Endocrinology, 2007.

37. Szotek, P. P., Pieretti-Vanmarcke, R., Masiakos, P. T., Dinulescu, D. M., Connolly, D.,

Foster, R., Dombkowski, D., Preffer, F., Maclaughlin, D. T., and Donahoe, P. K. Ovarian

cancer side population defines cells with stem cell-like characteristics and Mullerian

Page 78: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

69

Inhibiting Substance responsiveness. Proc Natl Acad Sci U S A, 103: 11154-11159,

2006.

38. Zhou, S., Morris, J. J., Barnes, Y., Lan, L., Schuetz, J. D., and Sorrentino, B. P. Bcrp1

gene expression is required for normal numbers of side population stem cells in mice, and

confers relative protection to mitoxantrone in hematopoietic cells in vivo. Proc Natl Acad

Sci U S A, 99: 12339-12344, 2002.

39. Oda, Y., Saito, T., Tateishi, N., Ohishi, Y., Tamiya, S., Yamamoto, H., Yokoyama, R.,

Uchiumi, T., Iwamoto, Y., Kuwano, M., and Tsuneyoshi, M. ATP-binding cassette

superfamily transporter gene expression in human soft tissue sarcomas. Int J Cancer, 114:

854-862, 2005.

40. Noonan, K. E., Beck, C., Holzmayer, T. A., Chin, J. E., Wunder, J. S., Andrulis, I. L.,

Gazdar, A. F., Willman, C. L., Griffith, B., Von Hoff, D. D., and et al. Quantitative

analysis of MDR1 (multidrug resistance) gene expression in human tumors by

polymerase chain reaction. Proc Natl Acad Sci U S A, 87: 7160-7164, 1990.

41. Jonker, J. W., Freeman, J., Bolscher, E., Musters, S., Alvi, A. J., Titley, I., Schinkel, A.

H., and Dale, T. C. Contribution of the ABC transporters Bcrp1 and Mdr1a/1b to the side

population phenotype in mammary gland and bone marrow of mice. Stem Cells, 23:

1059-1065, 2005.

Page 79: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

70

CHATPER 3

Blockade of hedgehog signaling inhibits the formation of tumors

derived from osteosarcoma side population cells

I preformed all experiments described in this chapter.

Page 80: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

71

Chapter 3

3 Blockade of hedgehog signaling inhibits the formation of tumors derived from osteosarcoma side population cells

3.1 Abstract

Our previous work demonstrates that side population (SP) cells are enriched for tumorigenic

potential in a broad range of primary mesenchymal neoplasms. Identification and manipulation

of the biological features underlying side population tumorigenesis may result in the

development of more effective treatment modalities against these neoplasms. Using

osteosarcoma cell lines as a model system, we showed that osteosarcoma side population cells

were up-regulated in the expression of the downstream Hedgehog targets genes Gli-1 and Ptch

when compared to their non-side population counterparts. In vitro blockade of the hedgehog

pathway with the chemical agent triparanol diminished the proportion of side population cells

when compared to vehicle control. Importantly, in primary transplants, triparanol treatment

resulted in an impairment of growth in side population derived tumors but not non-side

population derived tumors. Furthermore, treatment with triparanol decreased tumor cellularity

and the percentage of side population cells within SP derived tumors. Taken together, we

indentified a biologically distinguishing property between osteosarcoma side population and

non-side population cells that has the potential to be exploited and used as a treatment modality

for this mesenchymal neoplasm.

Page 81: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

72

3.2 Introduction

Osteosarcoma (OS) is the most common primary bone cancer in children and adolescence

representing 15% of all primary bone tumors and 0.2% of all malignant tumors in children[1, 2].

While survival rates have improved over the last decade with patients presenting with non-

metastatic disease having a 70% chance of long term survival, patients presenting with

metastases at time of diagnosis or with recurrent disease have only 20% chance of survival at 5

years[3, 4]. Osteosarcoma is a highly aggressive tumor with approximately 10% of patients

presenting with lung metastases at diagnosis[5]. The mechanisms underlying osteosarcoma

initiation and progression are poorly understood; however, mutations in both p53 and Rb are

associated with the disease. Patients with Li-freumeni syndrome, an autosomal dominant

disorder characterized by germline mutations in p53, develop a wide variety of tumor including

osteosarcomas[6, 7]. Also, genetic alterations of RB have been found in up to 70% of sporadic

cases of osteosarcoma[8, 9]. Conventional treatment for this malignancy includes a combination

of chemotherapy and surgery has significantly improved the survival rate of certain patients,

however, the overall long-term disease-free survival for patients diagnosed with osteosarcoma is

still low, indicating the need for exploration of new treatment options.

During normal fetal development various cell signaling pathways are regulated in a coordinated

manner that allows for stem cells to proliferate, differentiate, move and die to allow for normal

organ formation. Later in life cells in neoplastic process can utilize these same pathways to drive

tumorigenesis. Understanding the manipulation of these pathways by malignant cells may reveal

novel therapeutic approaches based on modulating the activities of these pathways in neoplastic

entities. One potential pathway for exploitation in the treatment of malignancies is the hedgehog

pathway, as it controls cell growth and differentiation during embryonic development and

deregulation of it is associated with the development of a variety of epithelial neoplasms[10, 11].

Briefly, the mechanism of signal transduction in this pathway involves the binding of secreted

hedgehog ligand to the Patched 1 (PTCH1) receptor on target cells. This interaction relieves the

inhibition of Smoothened (SMO) by PTCH1, and SMO sends signals through a series of

interacting proteins, including suppressor of fused, resulting in activation of the downstream Gli

family of transcription factors: GLI1, GLI2 and GLI3[10]. Patients with Gorlin’s syndrome have

germline mutations in the PTCH1 gene predisposing them to the development of basal cell

carcinoma and medulloblastoma[12, 13]. Also, both sporadically occurring basal cell carcinomas

Page 82: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

73

and medulloblastoma show up-regulation of various components of the hedgehog signaling

pathway[14, 15]. Deregulation of this pathway is also implicated in cartilaginous lesions, with

inhibition leading to a decrease in tumor volume, cellularity, and proliferation rates of

xenografted chondrosarcomas [16-18]. Osteosarcoma cell lines show elevated expression of

hedgehog signaling components and chemical blockade of cell lines with high level of GLI1 and

PTCH1 leds to impaired in vitro cellular proliferation[19]. However, the means by which

hedgehog signaling hinders the advancement of both epithelial and mesenchymal tumorigenesis

has yet to be fully elucidated.

The cancer stem cell theory is based on the observation that tumors are composed of a

heterogeneous population of cells organized into a cellular hierarchy. At the top of this hierarchy,

resides the cancer stem cell (CSC) or tumor initiating cell (TIC) that has the capacity to self-

renew and to differentiate into all the cells composing the heterogeneous tumor. The prospective

identification of TICs from a broad range of hematopoietic malignancies and solid tumors

strongly supports this hypothesis. Notably, only CSCs have the potential to induce tumor

formation when injected into immuno-deficient mice and a such, the CSC theory has important

clinical implications as it suggests that these cells need to be eliminated in order for disease free

survival to occur. As CSCs are thought to behave in a fashion similar to normal stem cells, it can

be reasoned that CSCs utilize similar signaling pathways to regulate self-renewal and

differentiation. As such, development of treatment strategies that specifically target these

pathways have begun to emerge[20]. Hedgehog signaling plays a critical role in the development

of both normal and malignant stem cells. It can be postulated that inhibition of this signaling

pathway may reduce tumor burden by effectively targeting the most potent malignant cells for

destruction. Treatment of glioma CSC with the hedgehog inhibitor, cyclopamine inhibits

proliferation and self-renewal while increasing apoptosis in the CSC population[21]. In breast

CSCs hedgehog signaling is also active[22], and treatment with cyclopamine or anti-hedgehog

blockade antibody reduces the clonogenicity of CD19+CD37+ multiple myeloma CSCs[23].

We previously demonstrated that primary mesenchymal tumors contain a subpopulation of cells,

SP cells, that preferentially form tumors when subcutaneously injected into immuno-deficient

mice[24]. We sought to identify a biological feature of osteosarcoma side population cells that

could be exploited to reduce their tumor forming capacity. As hedgehog signaling is implicated

Page 83: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

74

in osteosarcoma pathogenesis, we sought to determine if chemical inhibition of this pathway in

osteosarcoma side population cells would reduce their tumorigenic potential.

Page 84: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

75

3.3 Results

3.3.1 Side population cells are present in osteosarcoma cell lines

We previously identified the presence of SP cells in a broad range of primary mesenchymal

neoplasms, including osteosarcomas. To determine if osteosarcoma cell lines would recapitulate

this phenotype, we screened four well-characterized osteosarcoma cell lines; MG63, HOS, HOS-

MN, and KHOS for the presence of side population cells. Cells from each line were stained with

the fluorescent dye, Hoechst 33342, and staining was quantified using a fluorescence activated

cytometer. With the exception of the MG63, all other cell lines contained side population cells

with the percentage ranging from 0.58%-0.73% of the total cell population (Figure 3.1A&B).

This percentage falls within the range found in primary mesenchymal neoplasms[34], suggesting

that osteosarcoma cell lines maintain some features of primary mesenchymal neoplasms.

Staining in the presence of verapamil, an inhibitor of the ATP-binding cassette (ABC) family of

transporters, resulted in the loss of the side population fraction, indicating that the presence of

side populations cells are due in part, to the expression of the ABC family of transporters (Figure

3.1A). Neither passage number nor confluency of the cell lines at the time of staining impacted

the percentage of side population cells (data not shown).

3.3.2 Hedgehog signaling in osteosarcoma side population cells

Components of the hedgehog pathway are activated in mesenchymal neoplasms[20, 21, 35]. In

light of these findings, we sought to determine if differential activation of the hedgehog pathway

would be detected between osteosarcoma side population and non side population cells. KHOS

and HOS-MN cells were sorted into side population and non-side population fractions, RNA was

isolated, and real- time RT-PCR was performed to assess the levels of the hedgehog downstream

targets GLI-1 and PTCH-1. When compared to non side population cells, side population cells

showed a 3 fold higher expression in the hedgehog downstream target GLI-1 and a two fold

higher expression of the downstream target PTCH1 (Figure 3.2 A, B). Furthermore, KHOS cells

treated with 11.25µM of triparanol, a chemical inhibitor of the hedgehog pathway[36], exhibited

a diminished percentage of side population cells (Figure 3.2 C), suggesting that hedgehog

signaling may be important in regulating the behavior of these cells in osteosarcomas.

Page 85: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

76

3.3.3 In vivo tumorigenic potential of osteosarcoma side population and non-side population cells

As blockade of the hedgehog signaling pathway is an effective therapeutic treatment for a variety

of tumor types[14, 37, 38], we next investigated the potential of exploiting this pathway to

therapeutic target side population cells in osteosarcomas. Initially, we ascertained the

tumorigenic potential of osteosarcoma side population and non-side population cells. 5000 and

10,000 side population and non-side population cells isolated from HOS-MN cell lines were

injected subcutaneously into NOD/SCID mice. For the KHOS cell line 100, 1000, 5000, and

10,000 cells were injected into immuno-compromised mice. As with primary mesenchymal

tumors, both HOS and KHOS side population cells and non-side population cells had the

potential to form tumors upon primary transplantation. In the KHOS cell line, as cell dosage

increased, the frequency of tumor formation also increased (Table 3.1). However, unlike primary

mesenchymal neoplasms, no statistically significant differences were seen in tumor formation

frequency between the side population cells and the non-side population cells in either of the two

cell lines.

3.3.4 Blockade of hedgehog signaling inhibits in vivo tumor formation of side population cells

Next, KHOS cells were sorted into side population and non side population fractions and 1000

cells of each population were subcutaneously injected into immuno-compromised mice. Mice

were treated with 400mg/kg of triparanol for four weeks after which they were sacrificed and

tumor formation was assessed. Blockade of the hedgehog pathway reduced both the frequency

and size of tumors derived from side population cells when compared to tumors derived from

non-SP cells (Figure 3.3 A, B). No statistically significant differences in the weight of the mice

were detected (treated versus control), suggesting that effects were not due to a cytotoxic impact

on the animals (Figure 3.3 C). Tumors exposed to triparanol showed decreased expression of

GLI-1 when compared to untreated tumors (Figure 3.3 D). Interestingly, despite the lack of

differences seen in tumor formation frequency between the two populations of cells, only side

population cells were sensitive to chemical blockade of the hedgehog pathway, suggesting that

exploitation of this pathway may preferentially target side population cells.

Page 86: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

77

3.3.5 Blockade of Hedgehog Signaling decreases the cellularity of side population derived tumors

Xenografted tumors were formalin fixed, paraffin embedded and stained with hematoxylin and

eosin. The grafted tumors had nearly identical cytological appearances; however, SP tumors

exposed to triparanol showed decreased cellularity when compared to DMSO controls. No

differences were seen between treated and control groups in the non-side population cell derived

tumors (Figure 3.4 A, B). Single cell suspensions generated from harvested tumors were stained

with Hoechst 33342 and analyzed for side population cells by flow cytometery. Similar to our in

vitro findings, in vivo blockade of the hedgehog pathway resulted in diminishment of the

proportion of side population cells when compared to untreated controls (Figure 3.4 C).

3.4 Discussion

We sought to identify a biological property that could be exploited to therapeutically target the

tumorigenic potential of mesenchymal side population cells. To do so, we demonstrated that

osteosarcoma side population cells have increased expression of the downstream hedgehog

signaling target genes, Gli-1 and PTCH1 when compared to non-side population cells. In vitro

blockade of hedgehog signaling with the chemical agent triparanol diminished the proportion of

side population cells found in osteosarcoma cell lines. Both side population and non-side

population cells were able to induce tumor formation in primary transplants in immuo-deficient

NOD/SCID mice; however, only side population derived tumors were sensitive to inhibition of

hedgehog signaling. Specifically, triparanol treatment resulted in a decrease in both the tumor

frequency and size of side population derived tumors. Importantly, inhibition of hedgehog

signaling had no impact on tumors derived from non-side population cells. Furthermore, SP

tumors exposed to triparanol showed decreased cellularity and flow cytometry analysis revealed

diminished numbers of side population cells within these tumors when compared to untreated

lesions. Taken together, this suggests that the hedgehog pathway may influence the development

of side population derived osteosarcoma tumors; and as such, targeting this pathway may be an

effective treatment modality for this mesenchymal neoplasm.

The mechanism by which hedgehog blockade suppresses tumor growth in osteosarcoma side

population cells is currently being investigated. We can postulate that triparanol treatment has

the potential to inhibit self-renewal, alter proliferation/differentiation, increase apoptosis, or any

Page 87: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

78

combination of these three events within the side population. The capacity of SP cells to serially

engraft when treated with triparanol would focus on the influence of hedgehog blockade on the

self-renewal potential of these cells. Flow cytometry cell cycle analysis of DNA content coupled

with immunohistochemistry of tumor tissue sections (ki-67, tunnel staining) could reveal

differences in proliferative capacity and/or apoptotic status of side population cells treated with

triparanol. Both in vitro and in vivo treatment of cells treated with triparanol resulted in a

diminished proportion of side population cells. As mentioned, one possible explanation for this

observation is the impairment of SP cell proliferation and/or self-renewal. Alternatively,

inhibition of hedgehog signaling may impact the expression of the ABCG family of transporters,

a key determinant of the side population phenotype[29, 30]. Expression analysis of these

transporters will yield important information to explain the diminished proportion of SP cells in

the presence of triparanol. However, regardless of the mechanism, it is important to note that

tumors derived from non-side population cells were not effected by hedgehog blockade

demonstrating a biologically distinct property of side population cells.

The most common location of osteosarcomas in young adults is in areas of rapid bone

growth[31]. In these areas, osteoblasts, which are thought to derive from an early mesenchymal

precursor, undergo a well-regulated terminal differentiation process. Given the location, cellular

origins, and histological heterogeneity of osteosarcoma; it can be postulated that they arise from

the aberrant differentiation of mesenchymal or osteoblastic precursor[32]. Furthermore, in mice,

conditional loss of p53 within osteoblast precursors resulted in the formation of osteosarcomas

with 100% penetrance[33]. Also, as side population cells isolated from primary mesenchymal

neoplasms have stem-like characteristics[24], it is possible, that osteosarcoma side population

cells may also share this feature. It would be interesting to speculate whether triparanol

specifically inhibits the growth of a stem-like population within osteosarcomas. However, one

major caveat to our stud, is the use of osteosarcoma cell lines as a model system to investigate

the tumorigenic phenotype of side population cells in relation to non-side population cells within

osteosarcomas. The KHOS and HOS-MN cell lines contained a similar percentage of SP cells to

primary osteosarcoma tumors. Furthermore, both populations formed tumors upon primary

transplantation into NOD/SCID mice, thereby demonstrating some similarity of the cell lines to

their primary counterparts[24]. However, unlike primary mesenchymal neoplasms, no

differences were detected in either tumor frequency or tumor size (data not shown) between the

Page 88: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

79

side population and non-side population derived tumors, highlighting important behavioral

differences between cell lines and primary neoplastic tumors. In glioblastomas, tumor stem cells

isolated from primary human tumors showed marked differences in gene expression patterns and

in vivo tumor biology compared to matched glioma cell lines[34]. As such, it will be important to

determine our observations can be recapitulated in primary osteosarcoma tumors.

While studies examining a broad range of epithelial carcinomas demonstrate hedgehog blockade

results in the inhibition of tumor size the exact cause of this phenomenon is currently being

debated. As oncogenic cells are not independent entities, but rather act in concert with the tissue

microenvironment, it can be hypothesized that the impact of hedgehog blockade on tumor

formation being acts on extrinsic cellular influences, rather than intrinsic cell signaling. In some

studies, the decrease in tumor size seen in epithelial carcinomas exposed to hedeghog blockade

was attributed to an inhibition of signal responsiveness in the surrounding stromal cells rather

than of the epithelial derived cancers[35, 36]. The exact means by which the inhibition of

hedgehog signaling in the murine stroma impacts the development of human carcinomas has yet

to be elucidated. Regardless of the mechanism of action, this study raises the intriguing

possibility that in xenografted studies the surrounding stroma, may be at least in part, more

sensitive to the loss of hedgehog signaling and this results in the subsequent inhibition of tumor

growth. Given this observation, it can be postulated that osteosarcoma side population cells have

a heavier reliance on the surrounding stroma and blockade of hedgehog signaling may result in

the diminished tumor volumes seen in the side population but not non-side population derived

tumors. Examination of the expression of GLI-1 in the surrounding stroma through the use of

mouse specific primers may help to support this hypothesis.

Blockade of the hedgehog pathway has proved to be successful in the treatment of

medulloblastomas; specifically, chemical blockade of the Smoothened receptor results in a

dramatic reduction of metastatic tumor burden[28, 37]. However, the use of hedgehog inhibitors

for the treatment of osteosarcomas may prove to be more elusive. As hedgehog signaling is

critical mediator of normal bone development during endochondral ossification[38], the use of

hedgehog blocking agents in young patients whose growth plates are still active may prove to be

problematic. Studies demonstrate that transient inhibition of this pathway in young mice causes

permanent defects in bone structures[39]. Given these findings, the use of hedgehog blockade in

Page 89: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

80

young osteosarcoma patients may not be ideal; however, in it may be an option for older patients

whose the growth plates are no longer in their most active state.

Page 90: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

81

3.5 Figures

Figure 3.1 Side population cells are present in osteosarcoma cell lines

(A) MG63, HOS, HOS-MN, and KHOS cell lines were stained with Hoechst 33342 dye. To

detect for SP, cells were analyzed by using dual wavelength analysis (blue, 424-444nm; red,

675nM) after excitation with 350nm UV light. Representative staining samples of each cell line

are shown. The SP cells are outlined in red and shown as a percentage of the total cell

population. In the presence of verapamil, the percentage of SP cells is greatly diminished. (B)

Graphical data represents the mean percentage from 3 independent trials and error bars represent

95% confidence intervals. A 95% confidence interval that does not cross the mean of a

comparison is a statistically significant difference at p<0.05.

Page 91: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

82

HO Blue-A

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

0.61

HO Blue-A

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

0.044

HO Blue-A

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

0.017

HO Blue-A

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

9.63e-3

HO Blue-A

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

0.58

HO Blue-A

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

0.099

HO Blue-A

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

0.73

HO Blue-A

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

0.39

!"#$ %&' %&'(!) *%&'

Hoechst Red

Hoe

chst

Blu

e

0.0

0.5

1.0

1.5

2.0

A

B

!"#$

%&'

%&'(!

)

*%&'

% S

ide

Popu

latio

n

Page 92: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

83

Figure 3.2 Hedgehog signaling in osteosarcoma side population cells

(A, B) HOS-MN and KHOS cells were stained with Hoechst 33342 dye and sorted into side

population and non-side population cells. RNA was isolated and real time RT-PCR was

performed using primers specific to human GLI-1 and PTCH. There is a 3 fold increase in

expression of GLI-1 and a 2 fold increase in expression of PTCH in side population cells when

compared to non-side population cells. Expression of GLI-1 and PTCH was determined by

taking the ratio of gene expression over the expression of perspective housekeeping genes (data

not shown). Fold increases were then determined by taking the ratio of GLI-1 and PTCH

expression of side population cells over their expression in non-side population cells. Data

shown is the mean percentage from 3 independent trials and error bars represent 95% confidence

intervals. A 95% confidence interval that does not cross the mean of a comparison is a

statistically significant difference at p<0.05. (C) KHOS cells were treated with 11.25µM of

triparanol in DMSO for 24 hours. Treatment group exhibited a decrease in the proportion of side

population when compared to DMSO carrier control. Data shown is a representative sample from

three independent trials.

Page 93: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

84

0 1000 2000 3000 4000

FL 7 Lin: FL 7 Hst-Red

0

1000

2000

3000

4000

FL 6 Lin: FL 6 Hst-Blue

0.34

0 1000 2000 3000 4000

FL 7 Lin: FL 7 Hst-Red

0

1000

2000

3000

4000

FL 6 Lin: FL 6 Hst-Blue

0.025

DMSO Triparanol

Hoechst Red

Hoe

chst

Blu

e

0

1

2

3

4

5

Fold

Incr

ease

SP/

nonS

P G

li-1

KHOS

KHOS

HOS-MN

! "

Fold

Incr

ease

SP/

nonS

P PT

CH

#

0.0

0.5

1.0

1.5

2.0

2.5

3.0

KHOS HOS-MN

Page 94: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

85

Table 3.1 Proportion of tumors formed in NOD/SCID mice from injection of various

numbers of side population and non-side population cells.

HOS-MN and KHOS side population and non-side population cells were subcutaneously

injected into immuno-deficient NOD/SCID mice. After 6 weeks mice were euthanized and tumor

formation was assessed. The number of cells that were injected into each mouse is denoted. The

number of tumors formed and the number of injections that were performed are indicated for

each population. The frequency of tumor formation is indicated as percentages. Chi-squared

analysis demonstrated no statistical significance (p value < 0.05) in the frequency of tumor

formation between side population and non-side population cells.

Page 95: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

86

Cell Type

HOS-MN

Cell

Dose

(number of primary

mice with

tumors)/(total number

injected)

Frequency of

tumor formation

(percent)

Side Population 5x103 11/15 73%

1x 104 16/16 100%

Non Side Population 5x103 11/14 78%

1x 104 12/16 75%

Cell Type

KHOS

Cell

Dose

(number of primary

mice with

tumors)/(total number

injected)

Total number of

mice with

tumors (percent)

Side population 1x102 3/8 37.5%

1x103 5/9 55%

5x103 9/13 69%

1x 104 16/16 100%

Non Side Population 1x102 1/8 12.5%

1x103 6/10 60%

5x103 13/17 76%

1x 104 14/16 87%

Page 96: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

87

Figure 3.3 Blockade of hedgehog signaling inhibits in vivo tumor formation of osteosarcoma side population cells

(A) Mice injected with 1000 KHOS side population and 1000 KHOS non side population cells

were treated with 400mg/kg of triparanol or DMSO by oral gauvage for four weeks after which

they were sacrificed and tumor formation was assessed. Fewer tumors formed in triparanol

treated mice when compared to control mice (DMSO). No difference in tumor frequency was

detected between treated and control groups of mice injected with non-side population cells.

Differences in tumor frequency was determined using a Fisher’s exact test with two tailed p

values <0.05 considered statistically significant. (B) Triparanol treated side population tumors

also weighed less when compared to control group. No difference in weight was detected in

treated or untreated tumors that formed from the non-side population cells. Data shown is the

mean percentage of tumor weight and error bars represent 95% confidence intervals. A 95%

confidence interval that does not cross the mean of a comparison is a statistically significant

difference at p<0.05. (C) No difference in mice weight of mice detected between the two groups

at the time of sacrifice.

Page 97: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

88

Number of Cells

SP Non-SP

Tumor Formation (frequency)

1000 5/7 (71%)6/8 (75%)

KHOS

Number of Cells

SP Non-SP

Tumor Formation (frequency)

1000 5/7 (71%)2/7 (28%)

KHOS

0

200

400

600

800

1000

Tum

or w

eigh

t (m

g)

DM

SO

Trip

aran

ol

DM

SO

Trip

aran

ol

0

5

10

15

20

25

30

35

mou

se w

eigh

t (g)

DM

SO

Trip

aran

ol

SP non-SP SP

0.0

0.2

0.4

0.6

0.8

1.0

SP non-SP

Fold

dec

reas

e Tr

ipar

anol

/DM

SO G

li-1

!"

#

$

Page 98: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

89

Figure 3.4 Side population tumors treated with triparanol have decreased cellularity

(A) Harvested tumors were formalin fixed, stained with hematoxylin and eosin, and nuclei were

counted. Representative slides of the tumors are shown. (B) SP tumors subjected to triparanol

had fewer numbers of nuclei when compared to DMSO controls. No difference in cellularity was

detected between treated and control groups from tumors derived from non-side population cells.

Graphical data represents as the mean number of nuclei counted in each field of vision at 400X

magnification. Error bars represent 95% confidence intervals. A 95% confidence interval that

does not cross the mean of a comparison is a statistically significant difference at p<0.05. (C)

Real time RT-PCR revealed that treatment with triparanol resulted in the decrease in expression

of GLI-1 in SP derived tumors compared to untreated controls. Data is represented as a ratio of

gene expression of triparanol treated tumors relative to DMSO treated tumors. (D) Harvested

tumors derived from SP cells were dissociated, stained with Hoechst 33342 dye, and analyzed

with a flow cytometer. SP tumors exposed to triparanol had a lower proportion of SP cells when

compared to DMSO controls.

Page 99: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

90

0

50

100

150

200

250

side

pop

ulat

ion

non

-sid

e po

pula

tion

DMSO Triparanol

Num

ber o

f Cel

ls

DM

SO

Trip

aran

ol

DM

SO

Trip

aran

ol

SP non-SP

!

"

Hoechst Red

Hoe

chst

Blu

e

DMSO Triparanol#

0 1000 2000 3000 4000

FL 7 Lin: FL 7 Hst-Red

0

1000

2000

3000

4000

FL 6 Lin: FL 6 Hst-Blue

0.38

0 1000 2000 3000 4000

FL 7 Lin: FL 7 Hst-Red

0

1000

2000

3000

4000

FL 6 Lin: FL 6 Hst-Blue1.12

Page 100: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

91

3.6 Materials and Methods

3.6.1 Cell lines

MG63, HOS, HOS-MN, and KHOS cells were all obtained from the American Type Culture

Collection (Rockville, MD). Cells were routinely maintained in DMEM (Wisent) supplemented

with 10% fetal bovine serum (Wisent). Cells were routinely maintained and cultured at 37oC

with 5% CO2 in a humidified chamber until subjected to fluorescent activated cells sorting

(FACS).

3.6.2 Flow cytometry

For side population analysis 1.0 x 106 cells/mL were treated either alone or with 2.5 µg/mL of

Hoechst 33342 dye (Sigma) for 90 min at 37oC, or in combination with 50µmol/L verapamil

(Sigma) as previously reported[21]. To detect for SP, cells were analyzed by using a dual

wavelength analysis (blue, 424-444nm; red, 675nM) after excitation with 350nm UV light

(MoFlow, Cytomation). After staining all cells were then counterstained with 1µg/mL of

propidium iodide (Molecular Probes) and PI positive (non-viable) cells were excluded from

analysis. Cell staining was quantified using LSRII flow cytometer (Becton Dikinson).

3.6.3 RNA extraction and real time-RT-PCR

For sorted cells, after staining with Hoechst 33342 dye, cells were sorted in SP and non-SP

fractions. Sorted cells were rinsed one with 1X PBS and centrifuged at 1200rpm for 10 minutes.

RNA was extracted from cell pellets using RNeasy Mirco Kit (Qiagen) according to

manufacturer’s instructions. RNA quality was assessed with a Bioanalyzer (Agilent

Technologies) For xenografted tumor tissues; tumors were flash frozen with liquid nitrogen and

disociated with a mortar and pestle. RNA was then extracted using Trizol (Invitrogen) according

to manufacturer’s instructions and quality was assessed with a Bioanalyzer (Agilent

Technologies). cDNA was generated using 1st Strand Synthesis Kit (Invitrogen) according to

manufacturer’s instructions. For real-time RT-PCR gene specific Taq-Man fluorogenic probes

for human Gli-1 and Ptch was used. We performed standard quantitative RT-PCR reactions for

GLI-1 and PTCH on the ABI prism (Applied Biosystems) sequence detection system.

Asparagine synthetase (AS) and glyceraldehyde-3-phosphate (GAPDH) were used as internal

Page 101: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

92

control genes. The reactions were performed in triplicate in 20ul reaction volume using TaqMan

Universal PCR Master Mix (ABI) on a 96 well plate format. Expression of GLI-1 and PTCH in

individual populations of cells was first determined by taking the ratio of gene expression over

their perspective housekeeping gene. Fold increases between the two groups were determined by

taking the ratio of side population expression over non-side population expression.

3.6.4 Xenograft models

Sorted SP and non-SP cells were collected and cells were re-suspended in PBS at concentrations

ranging from 100-10,000 cells/50µl. Cells were then mixed with 50µl of Matrigel (Becton

Dickinson). This cell:matrigel suspension was then subcutaneously injected in to eight to ten

week old NOD/SCID mice. Mice were observed for up to 6 weeks after which they were

euthanized and tumor formation was assessed. Tumors that formed were removed and samples

from each tumor were harvested for FACS and histology. For histology, tumors were removed

and samples were paraffin embedded, formalin fixed, sectioned, and stained for hemotoxylin and

eosin. Staining was visualized using a Leica light microscope

3.6.5 Dissociation of xenografted tumors

Primary tumor samples were manually minced and all visible clumps removed. Enzymatic

digestion followed at 37oC for 45 minutes with constant rotation using 10mg/ml of collagenase

IV (Worthington), 2.4 U/ml of Dispase (Becton Dickinson), 0.05% trypsin (Wisent). Further

manual dissociation was performed by passing the cell slurry through an 18 gauge needle. Cells

were then centrifuged at 1400 rpm for 5 minutes and washed three times in PBS. After washing,

cells were strained through 70µm filters to remove remaining clumps. Single cell suspensions

were then stained with Hoechst 33342 dye and subjected to FACS

3.6.6 In vitro and in vivo blockade of hedgehog signaling

For in vitro experiments, cells were treated with 11.25uM of triparanol and control cells were

treated with carrier, DMSO. After 24 hours cells were detached with 0.05% trypsin in PBS

(Wisent), stained with Hoechst 33342 dye and subjected to FACS analysis (see above). Two

weeks prior to the initial injections treatment of mice began. For in vivo experiments, triparanol

was dissolved in olive oil and mice were orally gauvaged with 10mg/kg of triparanol every other

day for 4 weeks. By gauvage, one group of mice was treated with triparanol, three times a week,

Page 102: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

93

at a dosage of 400mg/kg. The control mice were treated with the carrier, olive oil. Mice were

treated for four weeks after with they were euthanized and tumors were harvested. Harvested

tumors were either dissociated to single cell suspension were generated (see below), paraffin

embedded, formalin fixed, sectioned, and stained for hemotoxylin and eosin (staining was

visualized using a Leica light microscope ) or 3) frozen for RNA extraction (see above).

Page 103: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

94

3.7 References

1. Zhou, B.B., et al., Tumour-initiating cells: challenges and opportunities for anticancer

drug discovery. Nat Rev Drug Discov, 2009. 8(10): p. 806-23.

2. Piccirillo, S.G., et al., Bone morphogenetic proteins inhibit the tumorigenic potential of

human brain tumour-initiating cells. Nature, 2006. 444(7120): p. 761-5.

3. Hoey, T., et al., DLL4 blockade inhibits tumor growth and reduces tumor-initiating cell

frequency. Cell Stem Cell, 2009. 5(2): p. 168-77.

4. Jiang, J. and C.C. Hui, Hedgehog signaling in development and cancer. Dev Cell, 2008.

15(6): p. 801-12.

5. Taipale, J. and P.A. Beachy, The Hedgehog and Wnt signalling pathways in cancer.

Nature, 2001. 411(6835): p. 349-54.

6. Hahn, H., et al., Mutations of the human homolog of Drosophila patched in the nevoid

basal cell carcinoma syndrome. Cell, 1996. 85(6): p. 841-51.

7. Johnson, R.L., et al., Human homolog of patched, a candidate gene for the basal cell

nevus syndrome. Science, 1996. 272(5268): p. 1668-71.

8. Dahmane, N., et al., Activation of the transcription factor Gli1 and the Sonic hedgehog

signalling pathway in skin tumours. Nature, 1997. 389(6653): p. 876-81.

9. Unden, A.B., et al., Human patched (PTCH) mRNA is overexpressed consistently in

tumor cells of both familial and sporadic basal cell carcinoma. Cancer Res, 1997.

57(12): p. 2336-40.

10. Bhardwaj, G., et al., Sonic hedgehog induces the proliferation of primitive human

hematopoietic cells via BMP regulation. Nat Immunol, 2001. 2(2): p. 172-80.

11. Trowbridge, J.J., M.P. Scott, and M. Bhatia, Hedgehog modulates cell cycle regulators in

stem cells to control hematopoietic regeneration. Proc Natl Acad Sci U S A, 2006.

103(38): p. 14134-9.

Page 104: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

95

12. Zhao, C., et al., Hedgehog signalling is essential for maintenance of cancer stem cells in

myeloid leukaemia. Nature, 2009. 458(7239): p. 776-9.

13. Dierks, C., et al., Expansion of Bcr-Abl-positive leukemic stem cells is dependent on

Hedgehog pathway activation. Cancer Cell, 2008. 14(3): p. 238-49.

14. Bar, E.E., et al., Cyclopamine-mediated hedgehog pathway inhibition depletes stem-like

cancer cells in glioblastoma. Stem Cells, 2007. 25(10): p. 2524-33.

15. Liu, S., et al., Hedgehog signaling and Bmi-1 regulate self-renewal of normal and

malignant human mammary stem cells. Cancer Res, 2006. 66(12): p. 6063-71.

16. Peacock, C.D., et al., Hedgehog signaling maintains a tumor stem cell compartment in

multiple myeloma. Proc Natl Acad Sci U S A, 2007. 104(10): p. 4048-53.

17. Tiet, T.D., et al., Constitutive hedgehog signaling in chondrosarcoma up-regulates tumor

cell proliferation. Am J Pathol, 2006. 168(1): p. 321-30.

18. Hopyan, S., et al., Dysregulation of hedgehog signalling predisposes to synovial

chondromatosis. J Pathol, 2005. 206(2): p. 143-50.

19. Hopyan, S., et al., A mutant PTH/PTHrP type I receptor in enchondromatosis. Nat Genet,

2002. 30(3): p. 306-10.

20. Warzecha, J., et al., Inhibition of osteosarcoma cell proliferation by the Hedgehog-

inhibitor cyclopamine. J Chemother, 2007. 19(5): p. 554-61.

21. Wu, C., et al., Side population cells isolated from mesenchymal neoplasms have tumor

initiating potential. Cancer Res, 2007. 67(17): p. 8216-22.

22. Cooper, M.K., et al., Teratogen-mediated inhibition of target tissue response to Shh

signaling. Science, 1998. 280(5369): p. 1603-7.

23. Skubitz, K.M. and D.R. D'Adamo, Sarcoma. Mayo Clin Proc, 2007. 82(11): p. 1409-32.

24. Tang, N., et al., Osteosarcoma development and stem cell differentiation. Clin Orthop

Relat Res, 2008. 466(9): p. 2114-30.

Page 105: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

96

25. Yauch, R.L., et al., A paracrine requirement for hedgehog signalling in cancer. Nature,

2008. 455(7211): p. 406-10.

26. Shaw, A., J. Gipp, and W. Bushman, The Sonic Hedgehog pathway stimulates prostate

tumor growth by paracrine signaling and recapitulates embryonic gene expression in

tumor myofibroblasts. Oncogene, 2009.

27. Rudin, C.M., et al., Treatment of medulloblastoma with hedgehog pathway inhibitor

GDC-0449. N Engl J Med, 2009. 361(12): p. 1173-8.

28. Yauch, R.L., et al., Smoothened mutation confers resistance to a Hedgehog pathway

inhibitor in medulloblastoma. Science, 2009. 326(5952): p. 572-4.

29. de Crombrugghe, B., V. Lefebvre, and K. Nakashima, Regulatory mechanisms in the

pathways of cartilage and bone formation. Curr Opin Cell Biol, 2001. 13(6): p. 721-7.

30. Kimura, H., J.M. Ng, and T. Curran, Transient inhibition of the Hedgehog pathway in

young mice causes permanent defects in bone structure. Cancer Cell, 2008. 13(3): p. 249-

60.

31. Lee, J., et al., Tumor stem cells derived from glioblastomas cultured in bFGF and EGF

more closely mirror the phenotype and genotype of primary tumors than do serum-

cultured cell lines. Cancer Cell, 2006. 9(5): p. 391-403.

Page 106: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

97

CHATPER 4

The development of aggressive fibromatosis (desmoid tumor) is

influenced by mesenchymal progenitor cells

I preformed all experiments described in the chapter with the following exceptions:

• Puviindran Nadesan helped in managing the mouse colony • William Stanford generated the Sca-1-/- mice

Page 107: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

98

Chapter 4

4 The development of aggressive fibromatosis (desmoid tumor) is influenced by mesenchymal progenitor cells

4.1 Abstract

The cellular origins from which most tumors arise are poorly defined. In pre-neoplastic

conditions, this knowledge can lead to the development of strategies to suppress tumor

development. Aggressive fibromatosis, also known as desmoid tumor, is a locally invasive soft

tissue tumor, whose cellular origin is undefined, but has mesenchymal cell characteristics. We

found that aggressive fibromatosis contain a subpopulation of cells that exclude Hoescht dye and

express cell surface markers found on mesenchymal progenitor cells (MPCs), demonstrating

mesenchymal progenitor features within a subset of aggressive fibromatosis cells. Using a mouse

model genetically predisposed to aggressive fibromatosis (Apcwt/1638N), we found that the number

of tumors that developed was directly proportional to the number of MPCs present as measured

by colony forming units-fibroblastic (CFU-F). Sca1-/- mice, which develop fewer MPCs, were

crossed with Apcwt/1638N mice. Apcwt/1638N/Sca1-/- mice developed substantially fewer aggressive

fibromatosis tumors. In contrast, Sca-1 deficiency had no affect on the formation of epithelial

derived intestinal polyps. Finally, MPCs isolated from Apcwt/1638N mice had the capacity to induce

aberrant cellular growth when injected into immuno-compromised mice. Taken together, this

suggests that MPCs can influence in the formation and development of aggressive fibromatosis.

Page 108: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

99

4.2 Introduction

Mesenchymal tumors display a great deal of cellular heterogeneity with a subpopulation of cells,

side population cells (SP) showing enhanced tumor initiating potential. These observations

suggest that this tumor type may be organized into a cellular hierarchy with SP cells behaving

like cancer stem cells driving tumorigenesis in established lesions[1, 2]. However, these findings

neither prove nor insinuate that mesenchymal neoplasms are derived from a normal

mesenchymal progenitor that has sustained oncogenic mutations nor do they suggest that cells

with stem-like characteristics contribute to de novo tumor formation.

Some mesenchymal neoplasms are associated with specific translocations that result in the

expression of functional fusion proteins that often contribute to oncogenesis. For instance,

rearrangement of the EWSR1 gene on chromosome 22 to the ETS gene family member, FLI-1, on

chromosome 11 generates the EWS-FLI-1 fusion protein commonly found in Ewing’s sarcoma.

Also, chromosomal translocations resulting in the generation of the FUS-CHOP fusion protein

are observed in myxoid lipsarcomas. Interestingly, over-expression of both EWS-FLI-1 and

FUS-CHOP proteins in murine mesenchymal progenitor cells results in the induction of tumors

strongly resembling the neoplastic lesions from which they are affiliated[4, 5]. Importantly,

tumorigenesis is only driven by super physiologic expression of these fusion proteins in

mesenchymal progenitor cells, indicating cell type specificity in this malignant process. Data

also demonstrates that after long-term culture, both murine and human mesenchymal progenitor

cells can undergo spontaneous transformation producing tumors resembling fibrosarcomas[6, 7].

Also, conditional expression of a translocation specific to synovial sarcomas in muscle

progenitors, but not mature myoblasts has the capacity to induce the formation of synovial

sarcomas in mice[8], further supporting the notion that that mesenchymal precursors give rise to

mesenchymal neoplasms.

Aggressive fibromatosis, also know as desmoid tumor, are a locally invasive soft tissue tumors,

generally arising in connective tissues. While these lesions infiltrate into surrounding normal

tissues, they do not metastasize to distant sites. Histological and cytologic analysis of the tumors

reveal that they are composed of bipolar fibroblastic cells that expresses the intermediate

filament, vimentin; but lack expression of epithelial markers such as E-cadherin. The location,

Page 109: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

100

cellular morphology and histological profile of these tumors suggest that they derive from

mesenchymal sources; however the cellular origins of aggressive fibromatosis have yet to be

elucidated[3, 9, 10].

The molecular etiology of aggressive fibromatosis is well known[11, 12]. These lesions can

occur as sporadic tumors or as part of pre-neoplastic conditions, such as familial adenomatous

polyposis (FAP) and familial infiltrative fibromatosis (FIF). Patients with familial adenomatous

polyposis develop gastrointestinal lesions that typically progress to cancer by the third decade of

life [13, 14]. In both familial adenomatous polyposis and familial infiltrative fibromatosis, the

lesions are associated with a mutation in the APC gene, while in sporadic aggressive

fibromatosis, most tumors contain mutations in CTNNB1, the gene that codes for β-catenin[15,

16]. In both cases, genetic aberrations ultimately result in the stabilization of β-catenin and the

up-regulation of β-catenin/TCF/LEF-1 dependant transcriptional activity[3, 17]. In mice,

mutations in the WNT/β-catenin pathway can also result in the formation of murine aggressive

fibromatosis. For example, the Apcwt/1638N mouse harbors a targeted mutation in the Apc gene

resulting in the expression of a truncated non-functional version of the APC protein consequently

leading to the up-regulation of β-catenin/TCF/LEF-1 dependant transcriptional activity. These

mice development a large numbers of aggressive fibromatosis tumors as well as gastrointestinal

lesions and as such, are a well-characterized animal model that closely approximates the human

disease [18, 19].

Here we use a mouse model of familial adenomatous polyposis (Apcwt/1638N) to show the

influence of normal mesenchymal progenitor cells on the formation of aggressive fibromatosis.

Importantly, we demonstrate that modification of the numbers of mesenchymal progenitor cells

directly impacts the number of tumors that were found in Apcwt/1638N mice. Furthermore, we

demonstrate that genetic the signature of aggressive fibromatosis side population cells is similar

to that of mesenchymal progenitor cells.

Page 110: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

101

4.3 Results

4.3.1 Aggressive fibromatosis contain a subpopulation of cells with progenitor properties

Previous reports demonstrate side population cells isolated from various tissues, including

mesenchymal neoplasms, have stem like characteristics[20]. Within aggressive fibromatosis

tumors, SP cells compose 0.2-1.8% of the total population (Figure 4.1 A); suggesting the

presence of stem-like cells within this tumor type. To further verify this finding, we examined

tumors for the presence of cell surface makers known to help in the identification of

mesenchymal progenitor cells (Section 1.6.3). Cells from primary aggressive fibromatosis

tumors were stained for CD146 and Stro-1 and expression was analyzed using flow cytometry.

Staining of cells from three independent tumors revealed the presence of both markers on

aggressive fibromatosis cells (Figure 4.1B, C). Furthermore, when co-stained with Hoechst

33342 dye, we found an enrichment of CD 146 cells within the SP, suggesting that within the SP

fraction, there is an enrichment of progenitor like cells (Figure 4.1 D).

4.3.2 Positive correlation between numbers of aggressive fibromatosis and CFU-Fs in Apcwt/1638n mice

We next sought to determine if there was a relationship between the number of mesenchymal

progenitors and the development of AF tumors. To study the in vivo role of MPCs in tumor

development we utilized a well-established mouse model of AF. Apcwt/1638n mice carry a targeted

mutation at codon 1638 of the murine APC gene. Mice heterozygous for the Apc1638N mutation

(Apcwt/1638N) develop high numbers of aggressive fibromatosis with complete penetrance [18].

The inherent variability in the number of AF tumors that form in each individual Apcwt/1638n mice

allowed us to determine if a relationship existed between the numbers of tumors and the number

of MPCs. Colony forming units-fibroblastic (CFU-F), which have the potential to differentiate

down various mesenchymal lineages, is a well established measure of MPCs present in the bone

marrow, and as such, we used the number of CFU-Fs as a surrogate for MPCs. 12 Apcwt/1638n

male mice were sacrificed at 6 months of age and tumors were counted. Tumors from individual

mice ranged from 10-35. Femurs from each mouse were removed and bone marrow cells were

plated to assess CFU-F formation. We found that mice with low numbers of tumors had fewer

CFU-Fs when compared to with high numbers of tumors (Figure 4.2 A). We have previously

reported than we can modulate the number of AF tumors in APC 1638n mice by mating the mice

Page 111: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

102

to either Timp Tg or Rhamm deficient mice[22, 23]. To confirm our findings that the CFU-F

alter with number of tumors, we measured the CFU-F numbers in these mice. Consistent with

our findings in APC 1638n mice we found is a positive correlation between numbers of CFU-Fs

and number of AF that develop (Figure 4.2 B,C) suggesting that there is a relationship between

mesenchymal progenitor cells, as measured by CFU-F, and tumor formation.

4.3.3 Mesenchymal progenitors are involved in the development of aggressive fibromatosis

Mice lacking the Sca-1 antigen develop fewer numbers of mesenchymal progenitor cells over

time, as marked by progressively fewer CFU-F[24]. Using a previously described breeding

strategy we mated Apcwt/1638n mice to Sca-1-/- mice and generated Apcwt/1638n mice that were either

wildtype, heterozygous, or completely lacking the Sca-1 gene[22]. At 6 months of age mice were

sacrificed and tumor development was assessed. We found that Apcwt/1638N/Sca-1+/+ mice had the

greatest numbers of tumors, averaging 24 tumors per male mice. In contrast, Apcwt/1638N/Sca-1-/-

littermate controls developed significantly fewer tumors averaging 12 tumors per male mouse

(Figure 4.3 A). Consistent with previously published reports male mice developed more tumors

than female mice (6,9,10). However, as with the males, Apcwt/1638n/Sca-1+/+ female mice formed

more tumors than Apcwt/1638n/Sca-1-/- littermate controls (Figure 4.3 A). Tumors from each

genotype were also stained with hematoxylin and eosin. We found that tumors derived from

Apcwt/1638N/Sca-1-/- had fewer cells compared to Apcwt/1638N/Sca-1+/+ mice (Figure 4.3 B,C)

suggesting that modulation of the number of mesenchymal progenitor cells impacts both the

number of aggressive fibromatosis that form as well as the cellularity of the tumors.

4.3.4 Mesenchymal but not epithelial derived tumors are impacted by alteration of MPCs

In addition to the formation of mesenchymal neoplasms, Apcwt/1638N mice also develop intestinal

polyps and skin cysts [19]. Interestingly, while the numbers of aggressive fibromatosis were

reduced in Apcwt/1638N/Sca-1-/- mice when compared to Apcwt/1638N/Sca-1+/+ littermate controls, no

differences were observed in the numbers of intestinal polyps or skin cysts (Figure 4.4) in either

male or female mice. Thus demonstrating that in Apcwt/1638N epithelial neoplasms were not

affected by the ablation of Sca-1.

Page 112: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

103

4.3.5 Mesenchymal precursors from Apcwt/1638N have the capacity to initiate tumor formation

We next determined if MPCs with an oncogenic mutations within murine AFs had tumorigenic

potential. To address this question, we isolated bone marrow stromal cells from 8 week old

Apcwt/1638N and Apcwt mice. 3.0 X 106 cells from both genotypes were subcutaneously injected

into immuno-deficient NOD/SCID mice. After 12 weeks mice were sacrificed and tumor

formation was assessed. We detected the formation of aberrant cellular growth in all mice

injected with cells derived from Apcwt/1638N (Figure 4.5). In contrast, none of the mice injected

with cells derived from wild type mice generated tumors. Histological examination of the lesions

revealed the presence cells of mesenchymal origins, specifically, cells resembling those found in

the bone marrow. Interestingly, cells on the outer edge of the lesions (arrowhead) appear to be

spindle shaped, resembling fibroblasts found within aggressive fibromatosis. We observed that

mesenchymal progenitors with oncogenic mutations have the capacity to deregulate cellular

growth and the resulting tumors appear to contain cells with mesenchymal origins.

4.4 Discussion

The presence of stem-like cells in aggressive fibromatosis tumors is evidenced by the expression

of MPC surface markers and the ability of a subset of cells (SP) to exclude Hoechst dye. This

suggests that within pre-existing AF tumors, there is a population of cells with stem-like features.

However, this observation does not imply that tumors arise from normal stem cells and as such,

we sought to identify the cells responsible for oncogenesis prior to tumor formation[3, 4]. We

observed a positive correlation between MPCs numbers and AF tumor formation and we

demonstrated that the loss of MPCs caused a reduction in the numbers of AF tumors in mature

mice. We also showed that MPCs derived form mice predisposed to AF tumor formation have

the capacity to initiate aberrant cellular growth when subcutaneously injected into immuno-

compromised mice. Taken together, these findings suggest that development of aggressive

fibromatosis is influenced by a mesenchymal precursor. The observation that MPCs numbers did

not impact the formation of intestinal neoplasms, which derive from epithelial precursors,

strengthens the specificity of the association between aggressive fibromatosis and MPCs.

Not only did we demonstrate a reduction in the number of tumors from Apcwt/1638n /Sca-1-/-, but

we also showed that tumors derived from these mice contain fewer numbers of cells when

Page 113: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

104

compared to Apcwt/1638n/Sca-1+/+ mice. Mice lacking Sca-1 develop age dependant osteoporosis

and this phenotype is caused not by the loss of differentiated osteoprogenitors, but rather by a

deficit in the numbers of MPCs. Interestingly, no differences in MPC frequency is detected in

young Sca-1-/- mice; however aged Sca-1-/- mice have much lower numbers of MPCs when

compared to their wildtype counterparts. This deficit and the subsequent development of age

dependant osteoporosis is attributed to a impairment in the capacity of MPCs cells to self-

renew[25]. It has been postulated that Sca-1 may play a role in balancing the signals between

differentiation and self-renewal in stem cells[26]. Given these observations, decreased tumor

cellularity in Apcwt/1638n /Sca-1-/- mice may be attributed to a diminished capacity of oncogenic

MPCs to self-renew.

Alternatively, aggressive fibromatosis, defined as a fibro-proliferative disorder, may be

attributed to a deregulation of MPC differentiation. MPCs are multi-potent with the capacity to

differentiate into muscle, fat, bone, cartilage, and fibroblastic cells. Isolation of single cell

derived clonal populations reveals that lineage commitment is not a random process, but rather,

organized into a cellular hierarchy, with the quinti-potent, self-renewing MPC at the apex and the

restricted fibroblast at the base[26]. Deregulation of these well-orchestrated events can alter the

lineage commitment of cells within this hierarchy[27-29]. For example, conditional deletion of

β-catenin in limb and head mesenchyme during early embryonic development results in an arrest

of osteoblastic differentiation as osteochondroprogentiors preferentially differentiate into

chondrocytes as opposed to osteoblasts[29]. Conversely, ectopic canonical Wnt signaling

enhances osteoblastic differentiation of these progenitor cells[27]. While this demonstrates that

deregulation of canonical Wnt signaling is implicated in the regulation of

osoteochondroprogenitors, the impact of this signaling pathway on cells higher up in the MPC

hierarchy has not been fully elucidated. As altered β-catenin signaling is a known molecular

determinant of AF tumor formation, it can be postulated that distortion of this pathway may tilt a

large proportion of mesenchymal precursors to forfeit towards the committed fibroblast, a

defining feature and cell population of AF tumors. The reduction in the number of MPCs in

Apcwt/1638n /Sca-1-/-, may therefore lower the numbers of cells mandated down an impaired

differentiation pathway.

Depletion of Sca-1+ cells in the mammary gland, results in the loss of regeneration potential in

mammary gland reconstitution experiments demonstrating the ability of Sca-1 to prospectively

Page 114: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

105

identify a population of mammary epithelial cells. [30]. Interestingly, in mouse models of breast

cancer, over-expression of components of the Wnt signaling pathway results is an expansion of

Sca-1/keratin 6 progenitor cells. In contrast, in mouse models where tumorigenesis is driven by

the over-expression of H-ras, Neu, or middle T antigen, no expansion of Sca-1/keratin 6

progenitor cells is detected[31, 32]. The molecular etiology of AF is well known where the up-

regulation of β-catenin signaling is a hallmark of these neoplasms. Taken together, this suggests

that the β-catenin pathway may contribute, in part, to tumorigenesis by expanding the progenitor

cell compartment allowing for an increase in the number of cells that may be responsible for

tumor maintenance. As such, loss of Sca-1 results in fewer numbers of progenitors susceptible to

this aberrant expansion.

In summary, the loss of Sca-1 may diminish the number of aggressive fibromatosis tumors

through multiple mechanisms. Firstly, the reduction in the number of mesenchymal progenitors,

which act as a potential candidate cell of origin for these neoplasms, results in fewer cells with

the potential to form tumors. This may be particularly important in mesenchymal neoplasms

susceptible to β-catenin signaling such as AF for this loss is amplified in the tumorigenic process

by reducing the number of progenitor cells available for both cellular expansion and distorted

lineage commitment. Secondly, in Sca-1 null mice, the remaining MPCs have an impaired ability

to self-renew, resulting in the eventual loss of tumors by targeting those cells responsible for the

maintenance of the malignant tissue. This provides strong evidence that functional mesenchymal

progenitor cells are important in the development and progression of aggressive fibromatosis. .

This data strongly suggests the influence of progenitor cells in this neoplasm. The identification

of the primary cell of origin in aggressive fibromatosis is a key step towards an understanding of

the pathology of this disease. Here we provide evidence to support a model in which cells with

mesenchymal stem cell like characteristics play a role on both play a role in both the initiation

and maintenance of aggressive fibromatosis tumors. Raising the intriguing possibility that

protecting the stem cells in patients with FAP can prevent AF and understanding mesenchymal

stem cell biology can be employed to develop new treatments.

Page 115: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

106

Figure 4.1 Human AF tumors contain progenitor cells

(A) Cells from primary human AF tumors were stained with Hoechst 33342 dye and were

analyzed by flow cytometry. The representative sample depicted shows the SP cells as outlined

and shown as a percentage of the total cell population. In the presence of verapamil, the

percentage of SP cells is greatly diminished. (B,C) Cells from representative aggressive

fibromatosis samples were stained for CD146 and Stro-1. Positively stained cells are outlined

and shown as a percentage of the total cell population. Isotype controls are shown to the left of

each experimental plot. (D) CD146 is enriched in the SP fraction. Cells from a representative

aggressive fibromatosis were stained co-stained with Hoechst 33342 dye and with an antibody

against CD146 antigen. SP cells are outlined and CD146 staining was examined in this fraction.

43% of SP cells express CD146.

Page 116: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

107

0 1000 2000 3000 4000

FL 7 Lin: FL 7 Hst Red

0

1000

2000

3000

4000

FL 6 Lin: FL 6 Hst Blue

1.8

0 1000 2000 3000 4000

FL 7 Lin: FL 7 Hst Red

0

1000

2000

3000

4000

FL 6 Lin: FL 6 Hst Blue

0.45

Hoechst Red

Hoechst R

ed

Hoecsht Hoecsht+Verapamil

Stro-1Isotype

A

101

102

103

104

105

PE-A

0

1000

2000

3000

4000

SSC-H0

101

102

103

104

105

<PE-A>

0

1000

2000

3000

4000

SSC-H 12.2

101

102

103

104

105

<PE-A>

0

1000

2000

3000

4000

FSC-H 43.7

<DAPI-A>

CD146Isotype

0 1000 2000 3000 4000

<Hoechst Red-A>

0

1000

2000

3000

4000

<DAPI-A>

0.2

CD146Hoechst Red

Ho

ech

st

Re

d

B

C

D

Page 117: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

108

Figure 4.2 Correlation of CFU-F with numbers of AF tumors.

A) Apcwt/1638n with fewer numbers of CFU-Fs develop fewer numbers of AF. Apcwt/1638N mice

were sacrificed at 6 months of age and tumors were counted. In tandem, colony forming units-

fibroblastic from each sacrificed mouse was assessed. Data shown represents a total of 12 mice

with calculated linear regression line and correlation coefficient displayed. B) CFU-F were

assessed from different genetically engineered strain of mice. Mice that form more tumors

(Apcwt/1638n/Timp-Tg) have higher numbers of CFU-F when compared to mice that develop

fewer numbers of AF (Apcwt/1638n/Rhamm-/-). Data represents the mean number of CFU-F per

mouse. 6 mice per given genotype were used. Error bars represent 95% confidence intervals. A

95% confidence interval that does not cross the mean of a comparison is a statistically significant

difference at p<0.05. C) Representative pictures demonstrating the differences in the number of

CFU-F between Apcwt/1638n/Rhamm-/-, Apcwt/1638n, Apcwt/1638n/Timp-Tg mice

Page 118: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

109

Increasing Number of Aggressive Fibromatosis in Mice

CF

U-F

Apc/Apc 1638N

Rhamm-/-Apc/Apc 1638N Apc/Apc 1638N

Timp-1 Tg

CF

U-F

Apc

/Apc

163

8N

Rha

mm

+/+

Apc

/Apc

163

8N

Rha

mm

-/-

Apc

/Apc

163

8N

Ti

mp-

1 Tg

Apc

/Apc

163

8N

Ti

mp-

1 W

T

Number of AF tumors

0

50

100

150

200

0

50

100

150

200

A B

C

#C

FU

-F

APC 1638N

Page 119: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

110

Figure 4.3 Modulation of MPCs impacts tumor development

A) Loss of Sca-1 reduces numbers of AF tumors in Apcwt/1638N mice. Apcwt/1638N/Sca-1+/+,

Apcwt/1638N/Sca-1+/-, and Apcwt/1638N/Sca-1-/- mice were sacrificed at 6 months of age and AF were

scored. In both males and females, Apcwt/1638N/Sca-1-/- mice formed fewer tumors than

Apcwt/1638N/Sca-1+/+ mice. Data represents mean number of tumors per mice (total of 15 mice per

given genotype). Error bars represent 95% confidence intervals. A 95% confidence interval that

does not cross the mean of a comparison is a statistically significant difference at p<0.05. B)

Tumors derived from Apcwt/1638N/Sca-1-/- mice have decreased cellularity when compared to

Apcwt/1638N/Sca-1+/+ littermate controls. Graphical data represents as the mean number of nuclei

counted in each field of vision at 400X magnification 3 mice for each given genotype and 4

fields for each section were counted. Error bars represent 95% confidence intervals. C)

Representative H & E slides of the tumors from each genotype show diminished numbers of

nuclei in tumors derived from Apcwt/1638N/Sca-1-/- when compared to Apcwt/1638N/Sca-1+/+ mice.

Page 120: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

111

0

5

10

15

20

25

30

35

40

0

2

4

6

8

10

12

0

30

60

90

120

150

Num

ber o

f AF

Tum

ors

Num

ber o

f AF

Tum

ors

Num

ber o

f Cel

ls

WT +/- -/-

Sca-1 Expression

WT +/- -/-

Sca-1 Expression

WT +/- -/-

Sca-1 Expression

Male Mice Female Mice

Sca-1 WT Sca-1 +/- Sca-1 -/-

H&

E

A

B

C

Page 121: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

112

Figure 4.4 Progenitor statuses in Apcwt/1638N/Sca-1-/- Mice

A) Apcwt/1638N/Sca-1-/- have fewer MPCs than Apcwt/1638N/Sca-1+/+ mice. There were fewer

numbers of CFU-F in Apcwt/1638N/Sca-1-/- mice when compared to Apcwt/1638N/Sca-1+/+ . Data

represents mean number of colony-forming units-fibroblastic isolated from three independent

mice from each genotype and error bars represent 95% confidence intervals. A 95% confidence

interval that does not cross the mean of a comparison is a statistically significant difference at

p<0.05. B) Side population analysis of whole bone marrow revealed a similar decrease in the

percentage of SP cells in Apcwt/1638N/Sca-1-/- compared to Apcwt/1638N/Sca-1-/- mice. Treatment of

cells with verapamil results in the inhibition of the SP population. Representative FACS plots

for each of the three genotypes are shown. C) Graphical data represents the mean number

percentage of SP cells from 3 independent mice (3 mice per given genotype). Error bars

represent 95% confidence intervals.

Page 122: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

113

CF

U-F

Apc/Apc

1638N

Sca

-1+/+

Apc/Apc

1638N

Sca

-1-/-

0.0

0.5

1.0

1.5

2.0

WT +/- -/-

Sca-1 Expression

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

HO Blue-A

1.09

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

HO Blue-A

0.38

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

HO Blue-A

1.82

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

HO Blue-A

0.2

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

HO Blue-A

0.54

0 1000 2000 3000 4000

HO Red-A

0

1000

2000

3000

4000

HO Blue-A

0.033

0

20

40

60

80

100

120

% S

P C

ells in B

M

Hoechst R

ed

Hoechst Red

A

B

Page 123: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

114

Figure 4.5 Loss of Sca-1 does not impact the formation of epithelial lesions

A) Apcwt/1638N/Sca-1+/+ and Apcwt/1638N/Sca-1-/- develop similar numbers of epithelial derived

lesions. Mice from each of the three genotypes were sacrificed at 6 months of age and the

number of intestinal polyps and skin cysts were counted. No significant differences were

observed in the numbers of intestinal polyps or skin cyst between Apcwt/1638N/Sca-1+/+,

Apcwt/1638N/Sca-1+/-, Apcwt/1638N/Sca-1-/- mice. Data represents the mean number of tumors per

mice (15 mice per given genotype) and error bars represent 95% confidence intervals. A 95%

confidence interval that does not cross the mean of a comparison is a statistically significant

difference at p<0.05.

Page 124: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

115

0

1

2

3

4

5

6

7

8

0

1

2

3

4

5

6

7

8

WT +/- -/-

Sca-1 Expression

WT +/- -/-

Sca-1 Expression

Male Mice Female Mice

# o

f In

testinal P

oly

ps

# o

f In

testinal P

oly

ps

Page 125: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

116

Figure 4.6 Stromal cells with oncogenic mutations have tumor initiating potential

A) Bone marrow stromal cells isolated from Apcwt/1638N and Apc wt/wt mice were cultured for 2

weeks, after which they were harvested and subcutaneously injected into NOD/SCID mice. After

3 months, mice were sacrificed and tumor formation assessed. Only stromal cells isolated from

Apcwt/1638N mice had the potential to induce aberrant cellular growth. B) Representative H&E

staining of lesions was performed for histological examination. Cells within the centre of the

lesions resemble bone marrow cells while cells lining the outer edge of the lesions appear to be

bipolar spindle shaped cells similar to fibroblasts (arrowheads).

Page 126: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

117

APC 1638NWT

H&E

5X 10X

!

"

Page 127: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

118

4.5 Materials and Methods

4.5.1 Primary tumors

Local ethical approval was obtained for all human tissue samples collected. Human and mouse

AF tumors were dissociated into single cells as previously reported [1]. Dissociated cells were

not cultured, but rather, used immediately afterwards for flow cytometry.

4.5.2 Flow cytometry

For side population analysis 1.0 x 106 cells/mL were treated either alone or with 2.5 µg/mL of

Hoechst 33342 dye (Sigma) for 90 min at 37oC, or in combination with 50µmol/L verapamil

(Sigma) as previously reported[1]. To detect for SP, cells were analyzed by using a dual

wavelength analysis (blue, 424-444nm; red, 675nM) after excitation with 350nm UV light

(MoFlow, Cytomation). For staining of mesenchymal progenitor markers, 1.0 X 106 cells

dissociated cells were re-suspended in 100µl of PBS supplemented with 2% fetal bovine serum

(Wisent) PE-conjugated CD146 (Becton Dickinson) and 0.1 µg/µl of Stro-1 (R&D Systems). For

visualization of Stro-1 staining, after incubation with primary antibody, cells were stained for 30

min at 4oC with FITC-conjugated IgM (Jackson labs). For co-staining of SP and CD146, cells

were initially incubated with Hoechst dye (see above) after which they were washed 2X with

PBS and then incubated with PE-conjugated CD146 (see above). After staining, cells were

washed 2X with PBS and then counterstained with 1µg/mL of propidium iodide (Molecular

Probes). PI positive (non-viable) cells were excluded from analysis. Cell staining was quantified

using LSRII flow cytometer (Becton Dikinson).

4.5.3 Generation of genetically engineered mice

The generation and phenotype of the APCWT/1638n and Sca-1-/- mice have been previously reported

([18], [24]. These mice were crossed to produce Apcwt/1638n/Sca-1+/+, Apcwt/1638n /Sca-1+/-, and

Apcwt/1638n /Sca-1-/- mice using a previously reported breeding strategy [22]. The generation and

phenotype of Apcwt/1638n/Rhamm-/-and Apcwt/1638n/Timp(Tg) mice has been previously reported

[22, 23].

Page 128: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

119

4.5.4 Scoring of tumors

Generation of these crosses allowed for the comparison of littermate controls between the

various genotypes. 15 male and 15 female mice of each genotype were sacrificed at 6 months of

age and the number of AF tumors, intestinal polyps, and skin cysts were scored as previously

reported [22]. For immunohistochemistry, representative tumor samples were formalin fixed,

paraffin embedded, sectioned, and stained with hematoxalin and eosin. To determine the number

of cells present in the tumors, a blinded study was performed counting the number of nuclei

present in stained sections. Specifically, stained sections were viewed under a light microscope

(Leica) and 3 representative samples from each genotype were used to count the number of

nuclei within the field of view (400X magnification).

4.5.5 Cell Culture

Mesenchymal stromal cells were isolated as previously described[30]. Briefly, mice were

euthanized at 8 weeks of age, femurs and tibias were removed, bone marrow was aspirated and

cells were plated in MesenCult® MSC Basal Medium for Mouse Mesenchymal Stem Cells

supplemented with Mesenchymal Stem Cell Stimulatory Supplements (StemCell Technologies).

After 72 hours the medium was changed to remove non-adherent cells. For colony forming units

fibroblastic, stromal cells were cultured for 7 days after which they were stained with crystal

violet (Sigma) (.05% w/v in methanol) and colonies greater than 1mm were counted[31].

4.5.6 Xenograft models

For xenografted tumors stromal cells were grown for 14 days after which the cells were

trypsinized and harvested for injections. 3.0 X 106 cells were re-suspended in 50 µl of 1XPBS

supplemented with 2% FBS. This suspension was then mixed with 50µl of Matrigel (Becton

Dickinson) and subcutaneously injected into immuno-deficient NOD-SCID mice with a 25

gauge needle as previously described[1, 32]. Mice were observed for 12 weeks after which they

were euthanized and tumor formation was assessed. Tumors were removed and samples were

paraffin embedded, formalin fixed, sectioned, and stained for hemotoxylin and eosin. Staining

was visualized using a Leica light microscope.

Page 129: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

120

4.5.7 Gene profiling

For human samples, after staining with Hoechst 33342 dye, cells were sorted in SP and non-SP

fractions. Sorted cells were rinsed one with 1X PBS and centrifuged at 1200rpm for 10 minutes.

RNA was extracted from cell pellets using RNeasy Mirco Kit (Qiagen) according to

manufacturer’s instructions. RNA quality was assessed with a Bioanalyzer (Agilent

Technologies) and cDNA was generated and hybridized onto Affymetrix Human Genome

U133.0 2.0 gene chips. We normalized the raw data using robust multi-array average (RMA)

algorithm (Irizarry et al. 2000). LPE (local-pooled-error test) (Jain et. al. 2003) was used to

identify differentially expressed genes between the side population and non-side population

fractions. Benjamini and Hochberg (BH) multiple testing procedure was used to evaluate false

discovery rate (FDR) (Benjamini and Hochberg, 1995). After which, analysis of gene expression

was performed using Parktec Genotyping Suite and Ingenuity Systems Software.

Page 130: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

121

4.6 References

1. Wu, C., et al., Side population cells isolated from mesenchymal neoplasms have tumor

initiating potential. Cancer Res, 2007. 67(17): p. 8216-22.

2. Wu, C. and B.A. Alman, Side population cells in human cancers. Cancer Lett, 2008.

268(1): p. 1-9.

3. Riggi, N., et al., Development of Ewing's sarcoma from primary bone marrow-derived

mesenchymal progenitor cells. Cancer Res, 2005. 65(24): p. 11459-68.

4. Riggi, N., et al., Expression of the FUS-CHOP fusion protein in primary mesenchymal

progenitor cells gives rise to a model of myxoid liposarcoma. Cancer Res, 2006. 66(14):

p. 7016-23.

5. Rosland, G.V., et al., Long-term cultures of bone marrow-derived human mesenchymal

stem cells frequently undergo spontaneous malignant transformation. Cancer Res, 2009.

69(13): p. 5331-9.

6. Li, H., et al., Spontaneous expression of embryonic factors and p53 point mutations in

aged mesenchymal stem cells: a model of age-related tumorigenesis in mice. Cancer Res,

2007. 67(22): p. 10889-98.

7. Haldar, M., et al., A conditional mouse model of synovial sarcoma: insights into a

myogenic origin. Cancer Cell, 2007. 11(4): p. 375-88.

8. Shields, C.J., et al., Desmoid tumours. Eur J Surg Oncol, 2001. 27(8): p. 701-6.

9. Lewis, J.J., et al., The enigma of desmoid tumors. Ann Surg, 1999. 229(6): p. 866-72;

discussion 872-3.

10. Alman, B.A., et al., Increased beta-catenin protein and somatic APC mutations in

sporadic aggressive fibromatoses (desmoid tumors). Am J Pathol, 1997. 151(2): p. 329-

34.

Page 131: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

122

11. Lips, D.J., et al., The role of APC and beta-catenin in the aetiology of aggressive

fibromatosis (desmoid tumors). Eur J Surg Oncol, 2009. 35(1): p. 3-10.

12. Kotiligam, D., et al., Desmoid tumor: a disease opportune for molecular insights. Histol

Histopathol, 2008. 23(1): p. 117-26.

13. Galiatsatos, P. and W.D. Foulkes, Familial adenomatous polyposis. Am J Gastroenterol,

2006. 101(2): p. 385-98.

14. Scott, R.J., et al., Familial infiltrative fibromatosis (desmoid tumours) (MIM135290)

caused by a recurrent 3' APC gene mutation. Hum Mol Genet, 1996. 5(12): p. 1921-4.

15. Hegde, M.R. and B.B. Roa, Detecting mutations in the APC gene in familial

adenomatous polyposis (FAP). Curr Protoc Hum Genet, 2006. Chapter 10: p. Unit 10 8.

16. Tejpar, S., et al., Predominance of beta-catenin mutations and beta-catenin dysregulation

in sporadic aggressive fibromatosis (desmoid tumor). Oncogene, 1999. 18(47): p. 6615-

20.

17. Tejpar, S., et al., Tcf-3 expression and beta-catenin mediated transcriptional activation in

aggressive fibromatosis (desmoid tumour). Br J Cancer, 2001. 85(1): p. 98-101.

18. Fodde, R., et al., A targeted chain-termination mutation in the mouse Apc gene results in

multiple intestinal tumors. Proc Natl Acad Sci U S A, 1994. 91(19): p. 8969-73.

19. Smits, R., et al., Apc1638N: a mouse model for familial adenomatous polyposis-

associated desmoid tumors and cutaneous cysts. Gastroenterology, 1998. 114(2): p. 275-

83.

20. Challen, G.A. and M.H. Little, A side order of stem cells: the SP phenotype. Stem Cells,

2006. 24(1): p. 3-12.

21. Tolg, C., et al., Genetic deletion of receptor for hyaluronan-mediated motility (Rhamm)

attenuates the formation of aggressive fibromatosis (desmoid tumor). Oncogene, 2003.

22(44): p. 6873-82.

Page 132: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

123

22. Kong, Y., et al., Matrix metalloproteinase activity modulates tumor size, cell motility,

and cell invasiveness in murine aggressive fibromatosis. Cancer Res, 2004. 64(16): p.

5795-803.

23. Bonyadi, M., et al., Mesenchymal progenitor self-renewal deficiency leads to age-

dependent osteoporosis in Sca-1/Ly-6A null mice. Proc Natl Acad Sci U S A, 2003.

100(10): p. 5840-5.

24. Holmes, C., et al., Longitudinal analysis of mesenchymal progenitors and bone quality in

the stem cell antigen-1-null osteoporotic mouse. J Bone Miner Res, 2007. 22(9): p. 1373-

86.

25. Holmes, C. and W.L. Stanford, Concise review: stem cell antigen-1: expression, function,

and enigma. Stem Cells, 2007. 25(6): p. 1339-47.

26. Sarugaser, R., et al., Human mesenchymal stem cells self-renew and differentiate

according to a deterministic hierarchy. PLoS One, 2009. 4(8): p. e6498.

27. Day, T.F., et al., Wnt/beta-catenin signaling in mesenchymal progenitors controls

osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell,

2005. 8(5): p. 739-50.

28. Glass, D.A., 2nd, et al., Canonical Wnt signaling in differentiated osteoblasts controls

osteoclast differentiation. Dev Cell, 2005. 8(5): p. 751-64.

29. Hill, T.P., et al., Canonical Wnt/beta-catenin signaling prevents osteoblasts from

differentiating into chondrocytes. Dev Cell, 2005. 8(5): p. 727-38.

30. Li, Y., et al., Evidence that transgenes encoding components of the Wnt signaling

pathway preferentially induce mammary cancers from progenitor cells. Proc Natl Acad

Sci U S A, 2003. 100(26): p. 15853-8.

31. Welm, B.E., et al., Sca-1(pos) cells in the mouse mammary gland represent an enriched

progenitor cell population. Dev Biol, 2002. 245(1): p. 42-56.

Page 133: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

124

32. Shackleton, M., et al., Generation of a functional mammary gland from a single stem cell.

Nature, 2006. 439(7072): p. 84-8.

33. Peister, A., et al., Adult stem cells from bone marrow (MSCs) isolated from different

strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation

potential. Blood, 2004. 103(5): p. 1662-8.

34. Phinney, D.G., et al., Plastic adherent stromal cells from the bone marrow of commonly

used strains of inbred mice: variations in yield, growth, and differentiation. J Cell

Biochem, 1999. 72(4): p. 570-85.

35. Cheon, S.S., et al., beta-Catenin stabilization dysregulates mesenchymal cell

proliferation, motility, and invasiveness and causes aggressive fibromatosis and

hyperplastic cutaneous wounds. Proc Natl Acad Sci U S A, 2002. 99(10): p. 6973-8.

Page 134: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

125

Chapter 5

5 Summary, conclusions, and future directions

Page 135: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

126

5.1 Summary

The prospective isolation of a tumor initiating cells from mesenchymal neoplasms and the

characterization of these cells is described in Chapters 2 and 3. Specifically, we demonstrated

that side population cells are present in a broad range of mesenchymal neoplasms and we

observed a positive correlation between the percentage of SP cells and tumor grade. In vivo

xenograft assays demonstrated that these cells were enriched for tumor initiating potential when

compared to their non-SP counterparts. Furthermore, only SP cells had the capacity to initiate

tumor formation upon serial transplantation (Chapter 2). In osteosarcomas, a specific

mesenchymal neoplasm, only SP cells were sensitive to chemical blockade of the Hedgehog

pathway. Specifically, treatment of osteosarcoma cell lines with an inhibitor to this pathway

resulted in the diminished capacity for SP cells to induce tumors in immuno-deficient mice

(Chapter 3).

The identification of a candidate cell of origin for the mesenchymal neoplasm, aggressive

fibromatosis is described in Chapter 4. We established that aggressive fibromatosis contain SP

cells and these cells have a genetic signature similar to mesenchymal progenitor cells. Genetic

alteration of the number of mesenchymal progenitor cells in mouse models of the tumor directly

influenced the numbers of mesenchymal tumors found in mature mice, but not in epithelial

derived lesions. In addition, mesenchymal progenitor cells with oncogenic mutations had the

capacity to induce aberrant cellular growth with a phenotype resembling cells of the

mesenchymal lineage (Chapter 4).

5.2 Conclusions

The existence of over 100 different subclasses of sarcomas highlights the large degree of

diversity within this tumor type and contributes to its poorly understood disease mechanism. The

inherent heterogeneous nature of these tumors coupled with the prospective identification of SP

cells with enhanced tumor initiating potential provides strong support that cells within these

tumors are organized in a cellular hierarchy with the mesenchymal tumor stem cell at the apex

and its differentiated progeny composing the heterogeneous tissue of the tumor body. The

biologically distinct properties of these cells, which include a stem-like phenotype, further

supports this hypothesis. Identification of the signaling pathways in the SP population that can be

Page 136: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

127

manipulated to inhibit tumor growth demonstrates one biologically discreet feature of these cells

with important clinical implications for tumor treatment.

While the prospective identification of stem like cells from mesenchymal neoplasms

provides evidence for the presence of progenitor cells in pre-existing tumors; this observation

does not identify the cell of origin for these neoplasms. Mouse models used in our studies

demonstrate the influence of mesenchymal progenitor cells in etiology of aggressive

fibromatosis. No impact was detected in epithelial derived tumors, showing cell type specificity

for this class of tumors. Furthermore, the oncogenic capacity of MPCs to induce aberrant

growths containing cells with histological features of mesenchymal cell types strengthens this

cellular association and helps to shed insight into the fundamental differences between

mesenchymal neoplasms and their epithelial counterparts.

5.3 Future Directions

5.3.1 The side population assay: considerations for its use in the isolation of cancer stem cells/tumor initiating cells

5.3.1.1 Criticisms and limitations of the side population assay

Several criticisms have been raised concerning the use of Hoechst dye as a means of isolating

stem like cells. Firstly, as Hoechst dye binds to DNA, staining renders the assay toxic to live

cells. Therefore, it can be argued that SP cells are not stem-like cells, but rather, only a

population of cells that are able to escape the lethal effects of Hoechst. Also, as dye efflux is a

dynamic process, minute variables in staining times, dye concentration, and cellular

concentrations can vastly affect the SP phenotype. Furthermore, cytometry gating strategies used

to isolate SP cells lack the consistency of gating strategies used when staining with markers[1,

2]. Taken together, these problems can lead to cross contamination of the SP and the non-SP

fractions ultimately resulting in the production of confounding data.

Studies using Hoechst dye to isolate tumor initiating cells have attempted to address such

concerns. For example, in primary mesenchymal tumors non-SP cells have the ability to form

tumors in the primary round of injections, however, these tumors fail to engraft after secondary

transplantation; presumably due to the inability of the non-SP cells to self-renew. However, the

initial capacity of tumor formation indicates that non-SP cells are indeed viable even after

Page 137: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

128

retaining Hoechst dye[3]. In further support of this finding, tumor initiating potential of

unstained but sorted MCF-7 breast carcinoma cells were compared to positively stained/non-SP

cells. Both populations of cells had similar in vivo tumorigenic potential and in vitro colony

forming potential. Taken together, this suggests dye toxicity does not account for the increased

“stemness” of SP cells[4].

There has been conflicting data regarding the capacity of SP cells to enrich for TICs. This may

be due, in part, to variation in gating strategies used by individual labs. While inhibitors against

ABC transporters, such as verapamil, are used as a control and ensure the capture of true SP

cells, this methodology does not always yield ideal results as positively stained cells can remain

within the negative or SP gate. Interestingly, gating within different regions of the SP fraction

has been shown to give rise to cells with differing “stemness” potential. That is cells in the

lowest quadrant have increased “stemness” potential when compared to cells in the upper

quadrant[1]. Taken together, this suggests that more stringent gating strategies are necessary for

elucidating the true nature of SP cells. However, it should be noted that all isolation strategies

have their shortfalls and perhaps the combination of different isolation methods are required to

enhance the purity of cancer stem cells.

SP cells isolated from tumors are enriched for cells with tumor initiating potential, however, the

exact nature of these cells have yet to be elucidated and it is unlikely that this population is

exclusively composed of tumor initiating cells. More likely, SP cells either represent a small

population of tumor initiating cells or conversely, tumor initiating cells represent a small fraction

of SP cells. While as few as 100 SP cell from primary mesenchymal neoplasms can initiate

tumor formation, ideally, a single SP cell would have the potential to form a tumor in order to

definitively determine the relationship between the two populations. However, this does not

mean that SP cells only represent a fraction of the total tumor initiating population, as mouse

xenograft assays may not capture the tumorigenic potential of all human SP cells.

Regardless of the unknown mechanisms by which SP cells initiate tumor formation, they

represent a population of clinically relevant cells as not only are they enriched for cells with

tumor initiating potential, but also, they are resistant to chemotherapeutic drugs. As such, this

population likely plays a critical role in tumor maintenance and reoccurrence. Unlike cell surface

markers used to identify cancer stem cells, SP cells are present in tumors from different cellular

Page 138: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

129

origins. Further knowledge of this population will therefore be important for the advancement

effective treatment modalities for wide variety of neoplasms.

5.3.1.2 An alternative hypothesis for defining side population cells

Hoechst 33342 staining intensity is not solely based on active dye removal, but rather on a broad

range of factors. For instance, before efflux begins, Hoechst 33342 binds to AT rich regions

within the minor groove of DNA; therefore, chromatin structure, conformation, and DNA

content all contribute to a given SP profile[5]. The dynamic nature of DNA structure, controlled

by both cell cycle and epigenetic events, raises the possibility that dye efflux may be a minor

contributor to the SP profile with the kinetic properties of dye to DNA binding playing a larger

role than currently credited for this phenotype[6].

Our studies focused on characterization of SP cells in primary mesenchymal neoplasms.

However, in some samples, we observed differences in the non-SP staining profile, specifically

the presence of two distinct populations, non-SPlow and non-SPhigh was detected. This phenotype

was noted in a few primary osteosarcoma samples and in the transformed osteosarcoma cell

lines, KHOS and HOS-MN. Interestingly, although the percentage of SP cells in these samples

was relatively low (below 1%), the primary tumors that exhibited this non-SP profile had more

aggressive tumors as measured by poor patient outcome (unpublished data). Given this

observation, it can be hypothesized that tumors containing dual non-SP high fractions may

represent aggressive neoplasms with characteristics similar to the transformed osteosarcoma cell

lines. It is interesting to speculate how the non-SP may be involved in this phenotype.

Presumably, the non-SP high cells have increased DNA content, and represent highly

proliferating cells that contribute acutely to the aggressiveness of a particular sample. However,

based on the cancer stem cell hypothesis the non-SP high cells should represent transiently

amplifying cells with no capacity to self-renew, and should therefore, not contribute to chronic

maintenance of the disease.

An alternative hypothesis could be formulated by examining DNA content in relation to genomic

stability rather than cell cycle. Genomic stability is a defining factor in the development and

progression of cancer[7, 8]. Non-SP cells may have increased genomic instability resulting in

chromosomal aberrations that may manifest as two distinct non-SP populations that represent

varying karyotypes with the tumor. Samples that exhibited this non-SP phenotype were derived

Page 139: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

130

from either primary osteosarcomas or osteosarcoma cell lines. Importantly, this mesenchymal

neoplasm displays a high degree of chromosomal abnormalities with ploidy number ranging

from haploidy to near hexaploidy[9, 10]. As such the non-SPhigh fraction may represent a

population with a high ploidy number relative to the non-SPlow fraction.

Based on this hypothesis, the non SP fraction high cells may be less genetically stable and

therefore lack the cellular longevity of their SP counterparts, however, they given the poor

prognosis of the patients, points to another theory. It is also possible that within the non-SP high

cells, a small fraction of cells may acquire mutations giving them a stem cell like phenotype

thereby helping to replenish the stem cell compartment and thus contribute to poor disease

outcome[11]. Serial transplantation of the non-SPhigh population into immuno-compromised mice

and examination of the capacity of non-SPhigh cells to generate SP cells could easily address these

questions. Regardless of the mechanism, this suggests future experiments examining the non-SP

profile may reveal important the molecular mechanisms that confer the biological properties of

these cells.

5.3.2 Characterization of side population cells in mesenchymal neoplasms

5.3.2.1 Identifying molecular determinants of sarcoma side population cells

To date the molecular mechanisms that confer stem like characteristics to SP cells is not well

defined. Many experimental approaches could be used to address this question, initially to

further characterize these cells array analysis may allow for the identification of novel proteins

that are involved in conferring the SP phenotype.

miRNAs are involved in maintaining both embryonic and tissue stem cells with changes in

specific miRNAs associated with ES cell self-renewal and differentiation[12-14]. Furthermore,

not only are miRNAs involved in normal developmental processes, but they can also function as

either tumor suppressors or oncogenes, thereby regulating the tumorigenic processes[15]. Given

the broad range of their functions, a comparison of miRNA expression between SP and non-SP

cells may provide a global indication of how SP cells function and also identify novel

mechanisms by which this phenomenon occurs.

Page 140: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

131

5.3.2.2 Identifying novel sarcoma stem cell markers

Theoretically, the ability to manipulate the intrinsic characteristics that differentiate stem-like

cells from non stem cells as a means of isolation appears to be tantalizingly simple. In the case of

cancer stem cells where markers vary not only between different tumor types, but also within

different subclasses of tumors, the universality of these assays makes them highly appealing,

effectively alleviating the dependency on identifying markers. Unfortunately, to date, data using

these strategies as a means of isolating stem cells remains relatively sparse and the majority of

the CSC literature is based on the selective expression of cell surface markers[16], however,

these two strategies need not be mutually exclusive. In the absence of known CSC markers, the

universality of the SP phenotype in tumor tissues can be used as a means for identifying such

markers. For example, as the SP enriches for cells with tumorigenic potential, these same cells

may also express novel TIC antigens. As such, SP cells can be used to generate novel antibodies

against potential CSC markers. Alternatively, combining markers that weakly enrich for TICs

with SP cells may further purify the fraction beyond that of each individual assay. Finally, unlike

sphere forming assays, which may only be a measure of anchorage independence, a hallmark of

cancer cells, the SP assay confers more specificity. Importantly, the identification of a novel

marker may not only help in the prospective isolation of a single mesenchymal cancer stem cell,

but it may also identify a marker that can be used in the isolation of normal mesenchymal

progenitor cell for which there is currently no marker or pattern of markers known to

successfully prospectively identify and isolate which has long hindered the field.

5.3.2.3 Metastatic potential of sarcoma side population cells

Epithelial to mesenchymal transformation (EMT) occurs when epithelial cells acquire a cellular

phenotype usually associated with mesenchymal cells. This process occurs during normal

embryonic development and in normal physiological processes such as wound healing[17]. EMT

entails a wide range of cellular changes, notably; epithelial cells undergo gross changes in their

cytoskeleton including the loss of adherens junctions, the loss of cortical actin, and the induction

of stress fibers[18]. The ability of malignant cells to disseminate from the primary tumor is one

hallmark of tumor cells and the subversion of the transcriptional regulation of EMT by neoplastic

cells allows for the acquisition of the migratory phenotype required for metastasis[19]. While the

Page 141: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

132

role of EMT in metastasis is well documented, its involvement in cancer stem cells is less well

defined.

It can be postulated that in order metastatic entities to occur, they must be initially seeded by a

cancer stem cell with the potential to self-renew and differentiate thereby generating a new

colony at a distant anatomical site[20]. Herman et al also demonstrated that within a population

of human pancreatic cancer stem cells, there exists a subpopulation of cells with increased

potential to metastasis[21]. Also, in an immortalized breast cancer cell line, cells with

mesenchymal phenotypes had more stem like properties defined by an increased expression of

stem cell markers associated with breast cancer stem cells an increased capacity to form

mammary spheres, an increased ability to grow in soft agar colonies and importantly, an

increased efficiency to form tumors in immuno-compromised mice. Furthermore, forced

expression of the transcription factor snail, the master regulator of EMT results in similar

observations indicating the importance of EMT demonstrating a link between cancer stem cells

and metastatic potential[22].

In light of these findings, it would be interesting to determine if metastatic tumors had a higher

percentage of SP cells or conversely, if SP cells have an increased potential to metastasize.

Microarray analysis to screen for markers associated with a mobile or invasive phenotype could

be undertaken. In addition in vitro experiments, to measure both mobility and cellular phenotype

could be performed. However, given the loss of the SP cells during prolonged culture periods,

presumably through the induction of cellular differentiation, these experiments would be limited

to assays with short incubation times. Although, it is important to note that sarcomas are derived

from mesenchymal cells and these studies would therefore, differ from their epithelia

counterparts. As such in vitro experiments may not yield biologically significant results in this

context.

In vivo experiments to determine if SP cells have and increased capacity to metastasize would

also address this question. This would be particularity well modeled in the osteosarcomas in

which metastasize to the lungs is common and contributes to poor patient prognosis. As non-SP

cells have the potential to form tumors upon primary injections, albeit at a lower frequency than

SP cells, tail vein injections could be performed and lung colonies could be measured to

determine the metastatic potential of SP cells compared to non-SP cells. However, to definitively

Page 142: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

133

address this question, ideal, if we could mark a single SP cells to determine the metastatic

potential.

5.3.3 Clinical significance of sarcoma side population cell

5.3.3.1 Prognostic value of sarcoma side population cells

The focus of cancer stem cell research relies on the biology of tumor initiating cells; however,

whether this biological phenomena will be clinically significant has yet to be definitively

elucidated. Studies examining the brain tumor cancer stem cell marker CD133 show promise as

expression of the antigen correlates with patient survival in gliomas [23, 24] To date, there is no

know prognostic marker to predict clinical outcome in sarcomas. In osteosarcoma, size of the

tumor upon discovery is still the best know predictive indicator[25]. This highlights the

importance of the findings that there was a positive correlation between percentage of SP cells

and tumor grade for SP cells may have the potential to be used as a prognostic determinant in

sarcoma patients. In this particular tumor type screening for the presence of these cells prior to

patient treatment coupled with long term follow up of these patients would yield invaluable data

regarding the importance of SP as a prognostic tool. For example, long term prospective studies

on the SP will determine if those tumors with a high percentage of these cells are more resistant

to conventional chemotherapy as these cells are not only enriched for cells with tumor initiating

potential but they also have the capacity to efflux such drugs. Hence, altering treatment strategies

based on the presence or absence of SP cells may ultimately result in more efficient treatment

and improved patient outcomes. Alternatively, diminishing the population of SP cells within a

given tumor may also be a potential treatment strategy. In the mesenchymal neoplasm,

aggressive fibromatosis, the presence of interferon results in an increase in the number of SP

cells demonstrating chemical agents are capable of shifting SP numbers within a given

tumor[26]. As such, agents capable of this may be used in conjunction with conventional

chemotherapy thereby decreasing or eliminating tumorigenic cells and preventing tumor

reoccurrence.

Given the broad range of mesenchymal neoplasms examined for our side population studies,

examination of individual subclasses in isolation may yield allow for the more subtle

information.

Page 143: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

134

5.3.4 Tumor microenvironment

Tumors reside in a specialized microenvironment composed of normal cells and extracellular

matrix, both of which provide regulatory input governing the development and progression of

neoplastic lesions. While the role of the tissue microenvironment on malignant cell proliferation

and metastasis is well established, less is known about its impact on neoplastic cells organized in

a cellular hierarchy[27]. Certainly, in normal tissue, the surrounding ecosystem deeply alters the

fate of stem cells playing a critical role in orchestrating the signals required for cell fate

determination and self-renewal[28, 29]. As the normal stem niche directly impacts its cellular

residents, it can be reasoned that the tumor niche also influences the behavior of cancer stem

cells[30, 31].The observation that SP cells are enriched for tumor initiating potential in

mesenchymal neoplasms provides a useful means to examine the role of the niche on the

behavior of malignant stem-like cells

5.3.4.1 The influence of the sarcoma niche on its side population cell residents

Work in the hematopoietic field suggests a possible role for the niche in regulating CSC

maintenance, for example, specialized microenvironments of bone marrow endothelial cells

appear to be required for homing and engraftment of leukemic stem cells[32]. Furthermore, both

extracellular matrix components and signaling molecules in the HSC microenvironment can

promote cell survival in AML, providing resistance to chemotherapeutic agents[33]. In addition,

brain cancer stem cells have been shown to reside in a vascular niche that secretes factors that

promote their long-term survival; furthermore, increasing the number of endothelial cells in brain

tumor xenografts expands the proportion of self-renewing cells in the tumor[34].

We demonstrated that chemical blockade of Hedgehog signaling inhibits SP cell tumor formation

in osteosarcomas (Chapter 3). However, whether this phenomenon is due to intrinsic or extrinsic

influences of the signaling pathway cannot be elucidated from our studies. Work from other labs

examined a broad range of epithelial carcinomas and demonstrated hedgehog blockade resulted

in the inhibition of tumor size due to an inhibition of signal responsiveness in the surrounding

stromal cells rather than of the epithelial derived cancers[35, 36]. This raises the possibility that

alteration of the niche can impact the behavior of sarcoma side population cells.

Page 144: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

135

However, it should be noted that transplantation of human cells into mouse recipients has

limitations as there is increasing speculation regarding the efficiency of the murine stroma to

support the growth of human cells. For example studies demonstrate that alteration of variables

such as the use of matrigel, strain of NOD/SCID mice, and length of incubation time can

dramatically increase the frequency of tumor initiating cells in human melanomas[37]. This

observation raises speculation regarding the efficiency of the murine stroma to support the

growth of human cells and brings into question in the use of xenograted models to study CSCs.

As such, mouse models may be a more efficient means to examine the role of the niche on the

behavior of tumor initiating cells. Furthermore, this strategy allows for the use of a wide range of

pre-existing genetically modified mice. For example, in breast cancer, sustained expression in

vivo in the mammary gland of mice of matrix metalloproteinase-3, which destroys the basement

membrane can lead to epithelial tumorigenesis. While the isolation of a murine sarcoma stem

cell has yet to be identified, Sca-1 prospectively identifies a population of cells from murine

osteosarcomas that are enriched for tumor initiating potential[38]. Injection of these cells into

genetically engineered mice lacking hedgehog expression in the stroma may begin to address the

impact of the tumor environment of sarcoma stem cells.

Importantly, the sarcoma niche has yet to be identified. As mentioned, there are 100 different

sarcoma subtypes, which in turn, are localized to various locations within the body. For example,

soft tissue sarcomas are found in surrounding connective tissue while sarcomas of the bone are

generally found in bone/ cartilage. As such, while subcutaneous injection of SP cells does allow

for tumor engraftment, this site of injection might not accurately recapitulate the true sarcoma

niche and examination within the context of the different subclasses of tumors may yield more

valuable information. Manipulation of the tumor niche may inhibit the growth of neoplastic

lesions by altering the phenotype of cancer stem cell, ultimately inducing reversion of

tumorigenesis [39].

5.3.4.2 Influence of hypoxia on side population cells

Just as the surrounding normal tissue and extracellular milieu establishes a niche with the

capacity to alter neoplastic progression, tumors themselves generate a unique microenvironment

that influences its cellular constituents. Specifically, as malignant tissue expands, the distance

between the center of a tumor mass and the surrounding normal stroma containing capillary

Page 145: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

136

vessels increases, thereby generating an oxygen gradient with cells furthest from blood vessels

exposed to the lowest levels of oxygen. Tumors exposed to hypoxic environments, or low O2,

created from this physical distance, are more resistant to radiation, highlighting the importance

of this phenomenon in tumor biology.

Recent studies have begun to address the influence of a hypoxic environment on biologically

distinct populations of cells that constitute a heterogeneous neoplastic lesion. Given the

observation that low oxygen levels maintain the pluripotency of embryonic stem cell by blocking

differentiation, it can be reasoned that cancer stem cells would also be impacted from similar

conditions. Not surprisingly, an in vitro hypoxic environment was found to increase the number

of cells expressing cancer stem cell markers in brain tumors. The mechanisms that confer these

properties have yet to be fully elucidated, however, current studies reveal that the hypoxia

inducible family (HIF) of proteins are key regulators in this process. For example, hypoxic

inducible factors were found to regulate the tumorigenic capacity of glioma stem cells[40]. Less

is known about the impact of hypoxia in mesenchymal neoplasms, however in normal

mesenchymal stem cells, a hypoxic environment improves the success isolation of these cells,

suggesting that it plays a role in stem cell maitenance. This may be due to the hypoxic

environment created in the bone marrow where mesenchymal stem cells reside. In light of these

observations, it would be interesting to determine if a hypoxic environment would alter the

cellular hierarchy in mesenchymal neoplasms by impacting the behavior of SP cells.

The hypoxic environment may promote self-renewal and block differentiation of mesenchymal

tumor initiating cells. Alternatively, the intrinsic properties of these cells may allow for survival

of external insults generated by a hypoxic niche. SP cells by definition have the potential to

efflux hydrophilic substances, which may include the acidic substances created in the tumor

microenvironment. Not surprisingly, a hypoxic environment leads to the increased expression of

ABCG2 transporters; therefore, as tumor tissues grow beyond their blood supply, SP cells are

better equipped to sustain themselves within the toxic surroundings. This protective

characteristic likely contributes to the stem cell like phenotype of not only sarcoma SP cells, but

to all SP cells.

Initial experiments to test this hypothesis could be performed by culturing SP cells under

hypoxic conditions to determine if this alters the percentage of SP cells. Importantly, if hypoxic

Page 146: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

137

conditions do indeed increase, or maintain SP cells this would provide a useful tool to

maintaining cells in culture as one current drawback to primary cultures is the loss of stem cells

after prolonged exposure to culture conditions. This would therefore, not only provide useful

information regarding the biology of sarcomas, would help to overcome technical difficulties

within this field.

5.3.5 Mesenchymal progenitor cells and their involvement in the development of aggressive fibromatosis

5.3.5.1 Cell of origin?

At six months of age, a decrease in the numbers of mesenchymal progenitor cells in

Apcwt/1638N/Sca-1-/- mice correlated to diminished numbers of aggressive fibromatosis tumors

(Chapter 4); however, it is unclear if this result was due to impaired de novo tumorigenesis or

altered fate determination of pre-existing tumor cells. As Sca-1 null mice have normal numbers

of mesenchymal progenitor cells at two months of age, when aggressive fibromatosis begin to

develop[41], this suggests that de novo tumor formation should not be impacted. It has been

postulated that Sca-1 may play a role in balancing the signals between differentiation and self-

renewal in stem cells[42]. Work with primary myoblasts isolated from Sca-1 null mice

demonstrates that these mice exhibit increased proliferation and reduced numbers of

undifferentiated cells[43]. Taken together, it can be hypothesized that mice lacking Sca-1 have a

signaling defect that results in increased differentiation in conjunction with an inhibition of self-

renewal of stem cells ultimately leading to the depletion in the number of stem-like cells as mice

age. Given the role Sca-1 plays in the self –renewal of MPCs, decreased tumor cellularity may be

attributed to a diminished capacity of onogenic MPCs to self renew. As such, it would be

interesting to observe the Apcwt/1638N/Sca-1-/- mice over a longer period of time to determine if

tumor numbers would be further diminished when compared to younger mice. In addition,

examination the differentiation potential of progenitors within the aggressive fibromatosis in

Apcwt/1638N/Sca-1-/-would reveal any differences in linage commitment as increased

differentiation may leave diminish the number of progenitor cells remaining to maintain the

neoplastic lesion

We demonstrate that mesenchymal progenitor cells isolated from Apcwt/1638N have the potential to

induce aberrant cellular growth when transplanted into immuno-deficient mice (Chapter 4).

Page 147: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

138

While this suggests that MPCs may be the cellular origin of aggressive fibromatosis; these

transplantation experiments may not represent the tumorigenic process that occurs under normal

physiologic conditions. To determine if aggressive fibromatosis tumors arise from MPCs, in vivo

linage tracking of MPCs from in Apcwt/1638N mice would be required. This has been effectively

demonstrated in colon cancer. However, one again, the limitation to this experiment is the lack of

markers known to identify a mesenchymal stem cell, unlike intestinal stem cells in which BMI1,

and Lgr5 mark intestinal stem cells[44, 45]. However, conditional expression of a translocation

specific to synovial sarcomas in muscle progenitors, but not mature myoblasts has the capacity to

induce the formation of synovial sarcomas in mice[46], aids in supporting the notion that that

mesenchymal precursors give rise to mesenchymal neoplasms.

5.3.5.2 Involvement of early progenitors or differentiated progeny?

Currently, the commonly accepted tests for the identification of MSCs include the capacity to

form colony forming unit fibroblastic (CFU-F) in culture, analysis of surface marker profiles,

and multi-lineage potential, particularly osteogenesis, chondrogenesis and adipogeneiss[47]. In

the hematopoietic system the identification of cell surface markers on both stem and committed

progeny has let to the stratification of cells into well-established hierarchy. Recent in vitro data

suggests that clonogenic MSC are organized into a cellular hierarchy in which differentiation

down multiple lineages occurs in a regulated fashion [48]. In both the hematopoietic and

neuronal systems, it has been demonstrated that both stem cells and committed progenitors have

the capacity to induce tumorigenesis (See Section 1.5). To address this question in our model

system, forced differentiation of Apc1638n/wt stromal cells down various mesenchymal lineages

pathways would produce committed progenitors. Injection of these cells into immuno-

compromised mice to assess the tumor initiating abilities of these cells relative to earlier

progenitor would reveal valuable information regarding the which cells are capable of

oncogeniesis. Importantly, this technique would circumvent the need for identifying cell marker

on committed progenitors. The ambiguity regarding the existence of the MSC stems from the

lack of identifying markers, various locations of isolation, and disparity in methods used to

culture and expand cells. This has resulted in the isolation of a heterogeneous population of

“MSC” that exhibit variable phenotypes, and as such, hampers the precision of defining such a

population of cells. However, regardless of the state of the progenitor cells, we have identified a

Page 148: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

139

mesenchymal precursor that has the potential to induce aberrant cellular growth, demonstrating

the influence of these cells in the oncogenic process.

5.4 Concluding Remarks

We identified a population of cells within mesenchymal neoplasms that are enriched for tumor

initiating potential. This observation begins to unravel the mechanisms by which mesenchymal

tumorigenesis occurs. The studies demonstrating that osteosarcoma side population cells

differentially expresses components of the hedgehog signaling prove that these cells are indeed

biological distinct from non-side population cells and provides a useful foundation from which

further investigation on the characterization of side population cells can begin. This work

provides valuable information on the pathogenesis of mesenchymal neoplasm and validates

future exploration in studying the role of mesenchymal progenitor cells in this tumor type.

Page 149: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

140

5.5 References

1. Montanaro,F.,etal.,DemystifyingSPcellpurification:viability,yield,andphenotype

aredefinedbyisolationparameters.ExpCellRes,2004.298(1):p.144‐54.

2. Challen,G.A.andM.H.Little,Asideorderofstemcells:theSPphenotype.StemCells,

2006.24(1):p.3‐12.

3. Wu,C.,etal.,Sidepopulationcellsisolatedfrommesenchymalneoplasmshavetumor

initiatingpotential.CancerRes,2007.67(17):p.8216‐22.

4. Zhou,J.,etal.,ActivationofthePTEN/mTOR/STAT3pathwayinbreastcancerstem­

likecellsisrequiredforviabilityandmaintenance.ProcNatlAcadSciUSA,2007.

104(41):p.16158‐63.

5. Lalande,M.E.andR.G.Miller,Fluorescenceflowanalysisoflymphocyteactivation

usingHoechst33342dye.JHistochemCytochem,1979.27(1):p.394‐7.

6. Ibrahim,S.F.,etal.,Kineticanalysesasacriticalparameterindefiningtheside

population(SP)phenotype.ExpCellRes,2007.313(9):p.1921‐6.

7. Grady,W.M.andS.Markowitz,Genomicinstabilityandcolorectalcancer.CurrOpin

Gastroenterol,2000.16(1):p.62‐7.

8. Heng,H.H.,etal.,Stochasticcancerprogressiondrivenbynon­clonalchromosome

aberrations.JCellPhysiol,2006.208(2):p.461‐72.

9. Sandberg,A.A.andJ.A.Bridge,Updatesonthecytogeneticsandmoleculargeneticsof

boneandsofttissuetumors:osteosarcomaandrelatedtumors.CancerGenet

Cytogenet,2003.145(1):p.1‐30.

10. Marina,N.,etal.,Biologyandtherapeuticadvancesforpediatricosteosarcoma.

Oncologist,2004.9(4):p.422‐41.

Page 150: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

141

11. Shackleton,M.,etal.,Heterogeneityincancer:cancerstemcellsversusclonal

evolution.Cell,2009.138(5):p.822‐9.

12. Hatfield,S.D.,etal.,StemcelldivisionisregulatedbythemicroRNApathway.Nature,

2005.435(7044):p.974‐8.

13. Shcherbata,H.R.,etal.,TheMicroRNApathwayplaysaregulatoryroleinstemcell

division.CellCycle,2006.5(2):p.172‐5.

14. Croce,C.M.andG.A.Calin,miRNAs,cancer,andstemcelldivision.Cell,2005.122(1):

p.6‐7.

15. Lu,J.,etal.,MicroRNAexpressionprofilesclassifyhumancancers.Nature,2005.

435(7043):p.834‐8.

16. Lobo,N.A.,etal.,Thebiologyofcancerstemcells.AnnuRevCellDevBiol,2007.23:p.

675‐99.

17. Chaffer,C.L.,E.W.Thompson,andE.D.Williams,Mesenchymaltoepithelialtransition

indevelopmentanddisease.CellsTissuesOrgans,2007.185(1‐3):p.7‐19.

18. Hay,E.D.,Anoverviewofepithelio­mesenchymaltransformation.ActaAnat(Basel),

1995.154(1):p.8‐20.

19. Thiery,J.P.,Epithelial­mesenchymaltransitionsintumourprogression.NatRev

Cancer,2002.2(6):p.442‐54.

20. Dalerba,P.,R.W.Cho,andM.F.Clarke,Cancerstemcells:modelsandconcepts.Annu

RevMed,2007.58:p.267‐84.

21. Hermann,P.C.,etal.,Distinctpopulationsofcancerstemcellsdeterminetumor

growthandmetastaticactivityinhumanpancreaticcancer.CellStemCell,2007.

1(3):p.313‐23.

22. Mani,S.A.,etal.,Theepithelial­mesenchymaltransitiongeneratescellswithproperties

ofstemcells.Cell,2008.133(4):p.704‐15.

Page 151: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

142

23. Zeppernick,F.,etal.,StemcellmarkerCD133affectsclinicaloutcomeinglioma

patients.ClinCancerRes,2008.14(1):p.123‐9.

24. Pallini,R.,etal.,Cancerstemcellanalysisandclinicaloutcomeinpatientswith

glioblastomamultiforme.ClinCancerRes,2008.14(24):p.8205‐12.

25. Bieling,P.,etal.,Tumorsizeandprognosisinaggressivelytreatedosteosarcoma.JClin

Oncol,1996.14(3):p.848‐58.

26. Tjandra,S.S.,etal.,IFN­{beta}signalingpositivelyregulatestumorigenesisin

aggressivefibromatosis,potentiallybymodulatingmesenchymalprogenitors.Cancer

Res,2007.67(15):p.7124‐31.

27. Tlsty,T.D.andL.M.Coussens,Tumorstromaandregulationofcancerdevelopment.

AnnuRevPathol,2006.1:p.119‐50.

28. Xie,T.andL.Li,Stemcellsandtheirniche:aninseparablerelationship.Development,

2007.134(11):p.2001‐6.

29. Li,L.andT.Xie,Stemcellniche:structureandfunction.AnnuRevCellDevBiol,2005.

21:p.605‐31.

30. Bissell,M.J.andM.A.Labarge,Context,tissueplasticity,andcancer:aretumorstem

cellsalsoregulatedbythemicroenvironment?CancerCell,2005.7(1):p.17‐23.

31. Sneddon,J.B.andZ.Werb,Location,location,location:thecancerstemcellniche.Cell

StemCell,2007.1(6):p.607‐11.

32. Sipkins,D.A.,etal.,Invivoimagingofspecializedbonemarrowendothelial

microdomainsfortumourengraftment.Nature,2005.435(7044):p.969‐73.

33. DeToni,F.,etal.,AcrosstalkbetweentheWntandtheadhesion­dependentsignaling

pathwaysgovernsthechemosensitivityofacutemyeloidleukemia.Oncogene,2006.

25(22):p.3113‐22.

Page 152: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

143

34. Calabrese,C.,etal.,Aperivascularnicheforbraintumorstemcells.CancerCell,2007.

11(1):p.69‐82.

35. Yauch,R.L.,etal.,Aparacrinerequirementforhedgehogsignallingincancer.Nature,

2008.455(7211):p.406‐10.

36. Shaw,A.,J.Gipp,andW.Bushman,TheSonicHedgehogpathwaystimulatesprostate

tumorgrowthbyparacrinesignalingandrecapitulatesembryonicgeneexpressionin

tumormyofibroblasts.Oncogene,2009.

37. Quintana,E.,etal.,Efficienttumourformationbysinglehumanmelanomacells.

Nature,2008.456(7222):p.593‐8.

38. Berman,S.D.,etal.,MetastaticosteosarcomainducedbyinactivationofRbandp53in

theosteoblastlineage.ProcNatlAcadSciUSA,2008.105(33):p.11851‐6.

39. Kenny,P.A.andM.J.Bissell,Tumorreversion:correctionofmalignantbehaviorby

microenvironmentalcues.IntJCancer,2003.107(5):p.688‐95.

40. Li,Z.,etal.,Hypoxia­induciblefactorsregulatetumorigeniccapacityofgliomastem

cells.CancerCell,2009.15(6):p.501‐13.

41. Smits,R.,etal.,Apc1638N:amousemodelforfamilialadenomatouspolyposis­

associateddesmoidtumorsandcutaneouscysts.Gastroenterology,1998.114(2):p.

275‐83.

42. Holmes,C.andW.L.Stanford,Concisereview:stemcellantigen­1:expression,

function,andenigma.StemCells,2007.25(6):p.1339‐47.

43. Mitchell,P.O.,etal.,Sca­1negativelyregulatesproliferationanddifferentiationof

musclecells.DevBiol,2005.283(1):p.240‐52.

44. Barker,N.,etal.,Identificationofstemcellsinsmallintestineandcolonbymarker

geneLgr5.Nature,2007.449(7165):p.1003‐7.

Page 153: Tumor Initiating Cells in Mesenchymal Neoplasms · population cells to initiate tumor formation in osteosarcoma cell lines. In conjunction with these experiments, we also sought to

144

45. Sangiorgi,E.andM.R.Capecchi,Bmi1isexpressedinvivoinintestinalstemcells.Nat

Genet,2008.40(7):p.915‐20.

46. Haldar,M.,etal.,Aconditionalmousemodelofsynovialsarcoma:insightsintoa

myogenicorigin.CancerCell,2007.11(4):p.375‐88.

47. Dominici,M.,etal.,Minimalcriteriafordefiningmultipotentmesenchymalstromal

cells.TheInternationalSocietyforCellularTherapypositionstatement.Cytotherapy,

2006.8(4):p.315‐7.

48. Sarugaser,R.,etal.,Humanmesenchymalstemcellsself­renewanddifferentiate

accordingtoadeterministichierarchy.PLoSOne,2009.4(8):p.e6498.