DETERMINING THE ROLE OF MUC1 AND BETA-CATENIN ON THE EPIDERMAL RECEPTOR SIGNALING AND LOCALIZATION IN BREAST CANCER

Item Type text; Electronic Dissertation

Authors Bitler, Benjamin Guy

Publisher The University of Arizona.

Rights Copyright © is held by the author. Digital access to this material is made possible by the University Libraries, University of Arizona. Further transmission, reproduction or presentation (such as public display or performance) of protected items is prohibited except with permission of the author.

Download date 06/10/2021 03:40:01

Link to Item http://hdl.handle.net/10150/194738 1

DETERMINING THE ROLE OF MUC1 AND BETA-CATENIN ON THE EPIDERMAL SIGNALING AND LOCALIZATION IN BREAST CANCER

By

Benjamin Guy Bitler

A Dissertation Submitted to the Faculty of the

GRADUATE INTERDISCIPLINARY PROGRAM IN CANCER BIOLOGY

In Partial Fulfillment of the Requirements

For the Degree of

DOCTOR OF PHILOSOPHY

In the Graduate College

THE UNIVERSITY OF ARIZONA

2010 2

THE UNIVERSITY OF ARIZONA GRADUATE COLLEGE

As members of the Dissertation Committee, we certify that we have read the dissertation

prepared by Benjamin Guy Bitler

entitled Determining the role of MUC1 and beta-catenin on the receptor signaling and localization in breast cancer.

and recommend that it be accepted as fulfilling the dissertation requirement for the

Degree of Doctor of Philosophy

Dr. Joyce Schroeder ______Date: 05/07/2010

Dr. Eugene Gerner ______Date: 05/07/2010

Dr. Ron Heimark ______Date: 05/07/2010

Dr. Jesse Martinez ______Date: 05/07/2010

Dr. Raymond Nagle ______Date: 05/07/2010

Final approval and acceptance of this dissertation is contingent upon the candidate’s submission of the final copies of the dissertation to the Graduate College.

I hereby certify that I have read this dissertation prepared under my direction and recommend that it be accepted as fulfilling the dissertation requirement.

______Date: 05/07/2010 Dissertation Director: Dr. Joyce Schroeder

3

STATEMENT BY AUTHOR

This dissertation has been submitted in partial fulfillment of requirements for an advanced degree at The University of Arizona and is deposited in the University Library to be made available to borrowers under rules of the library.

Brief quotations from this dissertation are allowable without special permission, provided that accurate acknowledgement of source is made. Requests for permission for extended quotation from or reproduction of this manuscript in whole or in part may be granted by the head of the major department of the Dean of the Graduate College when in his or her judgment the proposed use of the material is in the interests of scholarship. In all other instances, however, permission must be obtained from the author.

SIGNED: Benjamin Guy Bitler

4

ACKNOWLEDGMENTS

First and foremost I would like to acknowledge my wife, Jamie Bitler. Her support in my pursuit of a PhD has been unwavering and constant. She is my best-friend and has been by my side every step of the way. We have experienced a lot in our short lives and I cannot imagine anyone else I would rather be with.

I would like to acknowledge my mentor, Dr. Joyce Schroeder, for her direction, encouragement and friendship. I can never begin to describe my appreciation for Joyce and all of the things she has done for me. I admire her appreciation and understanding of science and the constant drive to make me a better scientist.

I would like to acknowledge my family who have been my distractions in times experimental successes and failures. My dad and step-mother, my brothers, Brandon, John and Jack, and sisters, Stephanie and Savannah, have all supported me and given me the motivation to persevere. I am extremely proud of all of them.

I would like to acknowledge my lab family past and present for making my lab experiences enjoyable. I can honestly say for the past five years I have not been to “work” once, because I do not consider doing experiments, scientific discussions, and writing about my passions with people that share my same passions “work.” All of the friends I have made are lifelong friends and colleagues.

I would like to acknowledge the Cancer Biology program. More specifically I would like to thank Dr. Tim Bowden and Anne Cione for their guidance. I feel honored to have been a part of Dr. Bowden’s life and I feel as if I am a better person because of our relationship. There are not enough words to describe how much I appreciate all of the administrative assistance I have received from Anne Cione. She is an amazing person with a love and dedication to the Cancer Biology students that is unmatched.

I would like to acknowledge my committee members for their experimental suggestions and guidance. 5

DEDICATION

I am dedicating this work to my mother Lynne D. Bitler whose fight against breast

cancer was courageous and brave. I remember one incidence during a short period of

remission when my mom completed the 5K Susan G. Komen Race for the Cure. She was still bald from the chemotherapy, in agony from the cancer and was extremely exhausted and yet by her determination to defeat breast cancer she finished the race. Most people who have cancer and are undergoing treatment will sometimes have trouble just getting out bed or sitting up in a chair, but not my mom. I imagine when she completed the race she was telling the cancer that “I will not be defeated.” She was a warrior who wanted live to see her kids grow up, get married, graduate college, have kids of their own and be

successful. This was just one of example of her strength and perseverance, which

allowed her to battle cancer. The thought of my mom struggling with breast cancer has

motivated and pushed me to do research that will make a difference. When experiments

did not work or I had setbacks, I would refocus by reminding myself of my mom and all

the women that are dealing with this awful disease. I hope and pray that one day I will be

able to demonstrate the same courage as my mom in dealing with adversity. My mom is

my inspiration for my work and I hope that I have made her proud by continuing the fight

against breast cancer. 6

TABLE OF CONTENTS

LIST OF FIGURES ...... 10

LIST OF TABLES ...... 12

ABSTRACT ...... 13

I. INTRODUCTION...... 15

BREAST CANCER ...... 16 ERBB RECEPTORS...... 18 ERBB RECEPTOR TRAFFICKING ...... 24 ERBB RECEPTORS IN BREAST CANCER ...... 30 ERBB RECEPTOR THERAPEUTICS ...... 31 CELLULAR POLARITY...... 34 MUC1 ...... 37 MUC1 STRUCTURE ...... 38 MUC1 TRANSCRIPTIONAL REGULATION AND TRAFFICKING ...... 41 MUC1 CYTOPLASMIC TAIL PROTIEN INTERACTIONS ...... 43 BETA-CATENIN STRUCTURE FUNCTION ...... 46 BETA-CATENIN/CANONICAL WNT SIGNALING ...... 47 BETA-CATENIN IN BREAST CANCER ...... 50 PEPTIDE THERAPY ...... 51 STATEMENT OF THE PROBLEM...... 53

II. MATERIALS AND METHODS ...... 55

CELL LINES ...... 55 TRANSFECTION WITH SIRNA ...... 55 ANTIBODIES AND GROWTH FACTORS ...... 56 IMMUNOPRECIPITATION AND IMMUNOBLOTTING ...... 57 IMMUNOFLUORESCENCE ...... 58 GROWTH FACTOR TREATMENT ...... 58 DIFFERENTIAL DETERGENT FRACTIONATION ...... 58 CHROMATIN-BOUND PROTIEN ISOLATION ...... 60 HIGH-SALT PROTEIN EXTRACTION ...... 60 CHROMATIN IMMUNOPRECIPITATION ...... 61 INVASION ASSAY ...... 62 MTT ASSAY ...... 63

7

TABLE OF CONTENTS (Contd.)

PEPTIDE SYNTHESIS ...... 63 MICE ...... 64 MOUSE GENOTYPING ...... 64 MOUSE PROTEIN LYSATES ...... 64 MAMMARY GLAND WHOLE MOUNTS ...... 65 XENOGRAFT TUMOR STUDIES ...... 65 GENETICALLY-DRIVEN TUMOR STUDIES ...... 66 MMTV-WNT-1 TRANSGENIC MOUSE STUDY ...... 66 STATISTICAL ANALYSIS ...... 67

III. MUC1 REGULATES NUCLEAR LOCALIZATION AND FUNCTION OF THE EPIDERMAL GROWTH FACTOR RECEPTOR ...... 68 INTRODUCTION ...... 68 RESULTS ...... 70 MUC1 PROMOTES ACCUMULATION OF NUCLEAR EGFR ...... 71 INHIBITING MUC1/EGFR INTERACTIONS DECREASES NUCLEAR EGFR ...... 77 MUC1 AND EGFR INTERACT IN THE NUCLEUS ...... 79 EGFR COLOCALIZES WITH PHOSPHORYLATED-RNA POLYMERASE II ...... 82 MUC1 PROMOTES EGFR-MEDIATED CYCLIN D1 EXPRESSION ...... 85 DISCUSSION ...... 88

IV. EGFR ACTIVITY PROMOTES TUMORIGENESIS IN MMTV-WNT-1 MOUSE MODEL OF BREAST CANCER ...... 93 INTRODUCTION ...... 93 RESULTS ...... 97 EGFR KINASE ACTIVITY PROMOTES WNT-1 DEPENDENT TUMORIGENESIS ...... 97 BETA-CATENIN AND E-CADHERIN PROTEIN EXPRESSION IS EGFR KINASE INDEPENDENT ...... 98 BETA-CATENIN LOCALIZATION IN MMTV-WNT-1 TUMORS IS EGFR KINASE INDEPENDENT ...... 98 EGFR KINASE ACTIVITY ALTERS MAMMARY GLAND DEVELOPMENT ...... 101 DISCUSSION ...... 102

8

TABLE OF CONTENTS (Contd.) V. INTRACELLULAR MUC1 PEPTIDES INHIBIT CANCER PROGRESSION ...... 105 INTRODUCTION ...... 105 RESULTS ...... 108 PMIP EFFICIENTLY ENTERS THE CELLS, REDUCES EGFR PROTEIN LEVELS AND INHIBITS MUC1 COLOCALIZATION WITH BETA- CATENIN ...... 108 PMIP INHIBITS CELLULAR INVASION AND PROLIFERATION IN VITRO ...... 113 PMIP INHIBITS TUMOR GROWTH AND RECURRENCE IN A XENOGRAFT BREAST CANCER MODEL ...... 115 PMIP INHIBITS TUMOR GROWTH AND INDUCES REGRESSION IN A GENETICALLY-DRIVEN BREAST CANCER ...... 118 MSPMIP TREATMENT RESULTS IN A REDUCTION OF EGFR AND MUC1 LEVELS ...... 123 PMIP DOWNREGULATES MET EXPRESSION...... 125 DISCUSSION ...... 128

VI. ANTI-CANCER THERAPIES THAT UTILIZE CELL PENETRATING PEPTIDES ...... 131 INTRODUCTION ...... 131 CELL PENETRATING PEPTIDES ...... 132 MECHANISM OF CELL PENETRATING PEPTIDES ...... 135 NON-IMMUNOGENIC ...... 138 ANTI-CANCER CELL PENETRATING PEPTIDES ...... 138 NUCLEAR TRANSLOCATION ...... 141 ONCOGENIC SIGNALING MODULATION ...... 145 MODULATION OF ECM INTERACTIONS ...... 150 NOVEL THERAPEUTIC TARGETS ...... 152 CURRENT & FUTURE DEVELOPMENTS ...... 152 ENDOSOMAL ESCAPE ...... 153 CPP STABILITY AND MODIFICATIONS ...... 153

VII. CONCLUDING STATEMENTS ...... 156 MUC1 AND EGFR’S ROLE IN AUTOPHAGY ...... 158 TARGETING MUC1/EGFR INTERACTION AND EGFR ACTIVITY IN COMBINATION ...... 159 EGFR AS A BIOMARKER ...... 161 PREVENTING LOSS OF CELL POLARITY ...... 161

9

TABLE OF CONTENTS (Contd.) ROLE OF HEAT SHOCK PROTIENS IN EGFR AND MUC1-DEPENDENT TUMORIGENESIS ...... 162

MUC1’S ROLE IN EGFR CYTOSOLIC RELEASE ...... 163 NUCLEAR EGFR TRAFFICKING: ANTI-CANCER TARGET ...... 164

IIX. REFERENCES ...... 168

10

LIST OF FIGURES

Figure 1.1: Cellular Origin and Subtypes of Breast Cancer ...... 17

Figure 1.2: ErbB Dimers and Adaptor Protiens ...... 20

Figure 1.3: EGFR Signaling Pathways and Biological Outputs ...... 22

Figure 1.4: EGFR Trafficking ...... 27

Figure 1.5: EGFR Oncogenic Function ...... 29

Figure 1.6: Maintenance of Cell Polarity ...... 35

Figure 1.7: Structure of MUC1 ...... 40

Figure 1.8: MUC1’s Cytoplasmic Tail and it Protein Interaction ...... 43

Figure 1.9: Canonical Wnt Signaling ...... 49

Figure 1.10: Working Model ...... 54

Figure 3.1: MUC1 Promotes Nuclear EGFR Localization ...... 74

Figure 3.2: MUC1 Promotes Nuclear EGFR Accumulation ...... 76

Figure 3.3: Inhibiting MUC1/EGFR Interaction Inhibitt Nuclear EGFR Accumulation . 78

Figure 3.4: MUC1/EGFR Interaction in the Nucleus Promotes EGFR/Chromatin

Interaction ...... 81

Figure 3.5: EGFR Colocalizes with Phosphorylated RNA Polymerase II ...... 84

Figure 3.6: EGFR Interaction with the cyclin D1 promoter is MUC1-dependent ...... 87

Figure 3.7: MUC1 Promotes Cyclin D1 Expression ...... 87

Figure 3.8: Model ...... 92

Figure 4.1: Loss of EGFR kinase activity significantly delays Wnt-1 dependent

tumorigenesis ...... 99 11

Figure 4.2: MMTV-Wnt-1 Egfr waved-2 tumors grow significantly slower ...... 99

Figure 4.3: Total β-catenin protein is independent of EGFR activity ... 100

Figure 4.4: β-catenin localization is independent of EGFR tyrosine kinase activity ...... 100

Figure 4.5: EGFR kinase activity is required for normal branching morphogenesis ...... 101

Figure 5.1: MUC1Peptide can effeciently enter cells ...... 111

Figure 5.2: PMIP blocks MUC1/beta-catenin and MUC1/EGFR Interactions ...... 112

Figure 5.3: PMIP inhibits cell proliferation and invasion of breast cancer cells ...... 114

Figure 5.4: PMIP significantly inhibits tumor growth and recurrence in vivo ...... 117

Figure 5.5: PMIP significantly slows progression of MMTV-pyV mT induced mammary

glands tumors ...... 121

Figure 5.6: MMTV-pyV mT tumors have differential response to msPMIP ...... 122

Figure 5.7: PMIP is associated with reduced Muc1 Expression ...... 124

Figure 5.8: hPMIP treatment results in reduced levels of phosphorylated EGFR and

dpERK ...... 125

Figure 5.9: MET is significantly downregulated in response to PMIP ...... 127

Figure 6.1: Mechanism of Peptide Membrane Transduction ...... 137

Figure 6.2: Mechanism of CTT Anti-Cancer Therapy ...... 140

Figure 7.1: EGFR Tyrosine kinase inhibitor (TKI) and PMIP combination significantly

inhibit proliferation ...... 160

Figure 7.2: Blocking nuclear EGFR translocation inhibits cell proliferation ...... 165

Figure 7.3: Working Model with PMIP Treatment ...... 167

12

LIST OF TABLES

Table 1.1: Anti-Erb Receptor Therapies ...... 33

Table 1.2: Role of MUC1 in mouse models of breast cancer ...... 37

Table 2.1: Antibodies ...... 56

Table 6.1: Commonly Utilized Cationic and Amphipathic Cell Penetrating Peptides ...... 134

13

ABSTRACT

The epidermal growth factor family of receptors is important in the development

and progression of many types of cancers including, breast, lung, and glioblastoma. The

family consists of 4 members (EGFR/erbB1, Her2/erbB2, erbB3, and erbB4). In all

breast cancer cases, EGFR expression is deregulated 20 to 30% of the time; however in

the most aggressive form of breast cancer (basal-like) EGFR expression is upregulated in

60% of cases. EGFR’s expression and activity can be altered in transformed cells

through a variety of mechanisms, such as novel protein-protein interactions, gene amplification, mutations, and loss of regulatory proteins. In this work we have examined the role of cancer specific protein interactions of EGFR with MUC1 and beta-catenin in the progression of breast cancer.

Herein I report that the interaction of MUC1 and EGFR in breast cancer cells alters EGFR localization by promoting EGFR nuclear translocation. Importantly, I discovered that the presence of MUC1 mediates EGFR’s interaction with chromatin.

More specifically, I found that EGFR interacts with the cyclin D1 promoter region in a

MUC1-dependent fashion which resulted in a significant increase in cyclin D1 protein expression. Nuclear EGFR localization has been shown to correlate with resistance to anti-EGFR therapies, which indicates that MUC1’s interaction with EGFR could be a mechanism of resistance.

MUC1’s interaction with both EGFR and beta-catenin can promote transformation therefore a peptide therapy was developed, PMIP, which mimics the 14

hypothesized interaction domains of MUC1’s cytoplasmic tail. PMIP was designed to

inhibit the interaction of MUC1/EGFR and MUC1/beta-catenin thereby regulating EGFR

expression and promoting beta-catenin localization to adherens junctions. PMIP

effectively enters the cytosol of cells and inhibits the target interactions. Importantly,

PMIP inhibited invasion and proliferation of breast cancer cells and in mice significantly reduced the growth rate of breast cancer xenograft and genetically-driven tumors. This study demonstrated that the use of peptides to inhibit intracellular protein interactions is a viable option that would have limited toxic side-effects. Overall, this work reveals a new regulatory role of EGFR localization and activity by MUC1 and that this mechanism is

viable therapeutic breast cancer target.

Lastly, in a mouse model of breast cancer I examined the role of EGFR

tyrosine kinase activity in beta-catenin dependent tumorigenesis. A transgenic mouse

model of breast cancer, MMTV-Wnt-1, was bred onto an EGFR kinase deficient

background. I discovered that the loss of EGFR kinase activity in this model resulted in a

significant delay in tumor onset and inhibited tumor growth. These findings indicate a

cooperation of EGFR and beta-catenin dependent signaling pathways, which promote

transformation of glandular epithelial cells.

15

I. INTRODUCTION

Aberrant cellular localization and activity of proteins is often observed in human

cancers. The mislocalization of these proteins is frequently the result of other mutations

or cellular events, but the novel role for mislocalized proteins in promoting disease

progression and therapeutic resistance is starting to be appreciated. In cancer, abnormal

trafficking of proteins will result in atypical protein-protein interactions that will support

cell growth, metastases, and inhibit chemotherapy efficacy. Specifically, I was interested

in the how aberrant localization and interactions of the oncoproteins, MUC1, EGFR, and

β-catenin contribute to the progression of breast cancer.

MUC1, EGFR, and β-catenin have breast cancer specific interactions, which have

been determined to promote cancer progression. I discovered that EGFR/MUC1 interactions enhance atypical EGFR nuclear localization and transcriptional activation.

In addition, by reducing MUC1 expression or EGFR activity I could significantly inhibit

β-catenin-dependent tumorigenesis. These findings prompted the development of a mimetic peptide that could inhibit these cancer-specific protein-protein interactions.

Importantly, inhibition of MUC1/EGFR and MUC1/β-catenin interactions resulted in a significant anti-tumor effect both in in vitro and in vivo models of breast cancer.

Breast Cancer 16

Breast cancer is the second most common cancer in women and accounts for 27%

of all cancers cases diagnosed in American women. In 2009, there were approximately

41,000 deaths due to breast cancer, which makes breast the second leading cause of cancer deaths in women. Additionally, women that do respond to treatment have a 30 to

50% chance of recurring within the first ten years. Recurrence of the cancer is highly dependent on the stage and subtype of the primary cancer [9]. There are several subtypes of breast cancer that are important in the determining the appropriate clinical approach.

These subtypes are determined by their genetic and proteomic signatures. The subtypes include luminal A and B, Her-2/erbB2 overexpressing, claudin-low, basal-like, and normal-breast-like [10]. Figure 1.1 depicts the hypothesized cellular linage of the subtypes of breast cancer [Reviewed in [7]] (Figure 1.1). Subtypes are most often distinguished by their estrogen/progesterone receptor and Her-2/erbB2 expression.

Approximatley 65% of breast cancers diagnosed are part of luminal A or B subtypes,

which are normally characterized by smaller less invasive tumors that are estrogen and progesterone receptor positive and erbB2 negative [10-12]. In contrast, basal-like breast cancers account for only 13-28% of all breast cancer cases, but the 5-year survival rate

for this subtype is significantly shorter [11]. This a result of the increase incidence of

metastases observed in the basal-like subtype, which is frequently the cause of cancer

mortalities. 17

Figure 1.1. Cellular Origin and Subtypes (blue) of Breast Cancer. Percentage incidence of subtype (red). Modified from [8].

Basal-like breast cancer is the most aggressive and metastatic subtype, mainly due

to the limited therapeutic options and advanced nature of the disease. Basal-like breast

cancers are often characterized by being triple negative (ER-, PR-, and HER2/erbB2-)

however it is important to note that not all basal-like breast cancers are triple negatives.

In addition to the hormone receptors and erbB2 other molecular markers have been positively correlated such as epidermal growth factor receptor (EGFR), β-catenin, p63 and high molecular weight keratins [11-13]. Interestingly, in the basal-subtype there is a

strong correlation of aberrant EGFR expression and mutated breast cancer 1 gene

(BRCA1) [14, 15]. BRCA1 is a tumor suppressor protein that is critical in the repair of double-stranded DNA breaks and limits genetic aberrations [16-19]. The correlation of

BRCA1 mutations and EGFR overexpression is hypothesized to be linked, because of the 18

high frequency of EGFR amplifications seen in the basal-like subtype [Reviewed in

[20]]. Expression of EGFR in basal-like breast cancers has made it a significant therapeutic target, however efficacy of anti-EGFR drugs has not translated from the lab bench to the clinical setting [Reviewed in [21]]. This demonstrates the need for more understanding on how EGFR expression and therapeutic efficacy are linked.

ErbB Receptors

The erbB family (EGFR/erbB1, Her2/Neu/erbB2, erbB3, and erbB4) of receptor tyrosine kinases (RTKs) is a well studied group of tyrosine kinases mainly because of their role in development and cancer progression. Loss of EGFR in the mouse results in embryonic lethality because of severe developmental defects [22]. In contrast, EGFR was also the first associated with cancer and has since become a

major focus in anti-cancer therapeutics [23].

The 170 kDa type I receptor tyrosine kinase ErbB1/EGFR was first described by

Carpenter et al. in 1978 when treatment with EGF resulted in an increase in plasma membrane associated phosphorylation [24]. In 1980 the egfr gene was mapped to chromosome 7, which allowed for cloning and ultimately genetic manipulation of the gene [25]. Cloning and sequencing of egfr led to its description as the first receptor tyrosine kinase to play a significant role in cancer [23]. Transcriptional regulation of

EGFR occurs in a non-classical fashion, the lack of a TATA and CAAT box in the 5’

promoter region results in multiple transcriptional initiation sites [26]. However, several 19

transcription factors, such as Sp1 and c-Jun, have been discovered to promote egfr

mRNA production [27, 28]. Following transcription the mRNA is translated and the

resulting EGFR protein is inserted into the endoplasmic reticulum membrane. Mediated

by a signal peptide, EGFR is then trafficked through the golgi to the basolateral

membrane [29]. Trafficking of EGFR and other erbB receptors to the plasma membrane

allows the interaction of receptor and extracellular ligand.

Normally, these membrane bound receptors function by binding ligand and there

are three different groups of ligands: 1) EGFR-specific (Epidermal Growth Factor, EGF;

Transforming Growth Factor α, TGFα; and , AR) 2) EGFR and ErbB4-

specfic (beta-cellulin, BTC; heparin-binding EGF, HB-EGF; and , EPR) 3)

ErbB3 and ErbB4-specfic ( 1, 2, 3 and 4, NRG1-4) (Figure 1.2a). Following

ligand binding, the receptor undergoes a conformational change which accommodates

either homo or heterodimerizations with a neighboring erbB receptor (eg. EGFR/EGFR

or EGFR/erbB2) [30, 31]. The erbB2 receptor has no known soluble ligand and its

ability to bind ligand is controversial. However erbB2’s native conformation is

conducive for dimer formation, which will allow dimer formation in the absence of

ligand [32]. The dimerization of the receptors leads to trans-phosphorylation of the

cytoplasmic tails, with the exception of erbB3. ErbB3 lacks a kinase domain and its

activation is dependent on the dimerization with another erbB receptor [33]. Upon

dimerization and phosphorylation the receptors mediate several downstream signaling

events, which are important in development and tumorigenesis. 20

Figure 1.2. ErbB Dimers and Adaptor Protein Binding Sites. Taken from [7]. 21

The trans-phosphorylation event is responsible for the initiation of many signaling

pathways that include: the Mitogen Activated Protein Kinase (MAPK), Akt/

Phosphoinositide 3-Kinases (PI3K), mammalian Target of Rapamycin (mTOR) and Janus

Kinase/Signal transducers and activators of transcription (JAK/STAT) pathways

[Reviewed in [5]] (Figure 1.3). The MAPK signaling pathway is important in cellular

proliferation by propagating a signal from the cell surface to the nucleus, where it

promotes the transcription of c-Jun and c-fos [34]. EGFR induced activation of the

Akt/PI3K signaling pathway can inhibit apoptosis by indirectly blocking the release of

the pro-apoptotic cytochrome C [35]. Inhibiting the release of cytochrome C prevents the

assembly of the pro-apoptotic caspase-9 dependent apoptosome and ultimately prevents

the cell from undergoing programmed cell death. It is important to note that EGFR lacks

a binding domain for p85 (subunit of PI3K), but is able to activate the pathway indirectly

through erbB3 dimers or the adaptor molecule Gab1 [36, 37]. Akt/PI3K signaling is

repressed by phosphatase and tensin homolog (PTEN) protein, which is responsible for

removing phosphates from membrane associated phosphotidylinositols. Removal of

phosphates prevents the interaction of PI3K with Akt by altering their spatial localization.

mTOR acts downstream of the PI3K/Akt pathway to promote cell proliferation and

survival by upregulating translation via eIF4E of survival and anti-apoptotic proteins

[38]. JAK/STAT signaling plays role in development and cancer progression by

promoting activating the transcription of genes important in cellular differentiation,

proliferation, and apoptosis [Reviewed in [39]]. Activated EGFR promotes the kinase

activity of JAK, which results in the phosphorylation and dimerization of STATs. STAT 22

dimers translocate to the nucleus and directly activate transcription of genes such as,

cyclin D1 (cell cycle regulator/proliferation), MMPs (invasion/metastases), VEGF

(angiogenesis) and cytokines (immune modulation) [Reviewed in [40]]. Aberrant

activation of all these signaling cascades can further promote normal cell transformation

and cancer progression.

Figure 1.3. EGFR Signaling Pathways and Biological Outputs. Taken from [1] 23

Most of these signaling pathways are initiated as result of post-translational

modifications, such as phosphorylation (tyrosines, serines and theronines), ubiquitination

(lysines), and N-glycosylation. The variety of modifications and different dimer pairs

(EGFR/erbB2, EGFR/erbB3, etc) allows for the propagation of specific signaling

pathways. For example, the tyrosines that are phosphorylated on EGFR can mediate a

specific signal to proliferate (MAPK; Y992, Y1068, and Y1148) or inhibit apoptosis

(Akt/PI3K, Y1101) [Reviewed in [41]] (Figure 1.2b). Frequently, erbB receptors will

form a dimer with a neighboring erbB2 receptor, because of erbB2 native conformation.

This demonstrates that different dimer formations, phosphorylation events and specific adaptor/receptor interactions can promote or inhibit a diverse number downstream signaling event.

In breast cancer, the overexpression of erbB2 and its constitutively active conformation allows it to act as a potent oncoprotein in promoting tumorigenesis [42,

43]. Therefore erbB2 is a preferential dimer because it promotes receptor activation in a

ligand-independent fashion. This has made erbB2 a highly effective anti-cancer target in

breast cancers that overexpress it, but in the basal-like subtype erbB2 expression is commonly lost. In basal-like breast cancers, EGFR is overexpressed 60% of cases, but anti-EGFR tyrosine kinase inhibitors have shown limited efficacy in the treatment of basal-like breast cancers. Aberrant EGFR trafficking to the nucleus can promote cell proliferation, radioresistance, and metastases [44-47]. This indicates that although kinase activity is important the oncogenic function of EGFR is not limited to its effect on 24

signaling but aberrant trafficking of EGFR also is important in conveying a tumor-

promoting phenotype.

ErbB Receptor Trafficking

Regulation of total erbB receptor levels and downstream signaling is heavily dependent on the trafficking of the receptors following ligand stimulation. Normally prior to ligand stimulation EGFR is frequently found associated with portions of the

plasma membrane rich in lipids and protein (caveolae) [48]. Following EGF binding to

EGFR the resulting dimerization and receptor activation leads to the recruitment of the

regulatory protein, Casitas B-lineage lymphoma (Cbl) protein. Recruitment of Cbl to

EGFR is mediated by the phosphorylated tyrosine on EGFR (Y1045) and the subsequent

Src-homolgy domain (SH2) found on Cbl. Following recruitment, Cbl’s E3 ubiquitin

ligase activity mediates the addition of ubiquitins to lysines found on EGFR’s

cytoplasmic tail. EGFR ubiquitination promotes the recruitment of the Epidermal growth

factor receptor Substrate-15 (Eps15) through its ubiquitin interacting motif (UIM) and

will mediate it translocation to a clathrin-coated pit which promotes internalization [49].

Recently, the ring-finger domain of Cbl has been demonstrated to be vital in the

regulation of EGFR internalization and subsequent trafficking to the early endosome

[50]. Dependent on the poly or multi-ubiquitination status of EGFR, the receptor is then

trafficked to late endosome and is ultimately degraded in the lysosomal compartment.

Trafficking and degradation of EGFR in the lysosome are major regulatory mechanisms 25

for controlling total EGFR levels and signaling [51]. Mutations and alterations in the

EGFR trafficking pathway can lead to overexpression and overactivation of the receptor

which can promote aberrant signaling and oncogenic function (Figure 1.4A).

However, in the last decade the trafficking of EGFR to novel intracellular

compartments has been described. The presence of transmembrane proteins in the

nucleus has been established for all of the ErbB receptors, but the biological significance

of this altered trafficking is still being investigated [52-55]. Retrograde trafficking of

EGFR from the plasma membrane to the nucleus has recently been described [52, 56].

Following EGFR internalization, the receptor undergoes retrograde trafficking and is

transported back to golgi or directly to the endoplasmic reticulum (ER) where it interacts

with the Sec61 translocon. Normally, the Sec61 translocon is responsible for moving

mis-folded proteins from the ER membrane to cytosol for subsequent proteosomal

degradation [57]. Once in endoplasmic reticulum EGFR interacts with the Sec61

translocon, which results in EGFR’s release into the cytosol. In the cytoplasm, the

hydrophobic regions of EGFR interact with Hsp70 to prevent protein aggregation. EGFR

then interacts with Importin-β1 through its tri-partite nuclear localization signal (NLS,

RRRHIVRKRTLRR) [56]. Importantly, the NLS located on the juxtamembrane domain

on EGFR, ErbB2, and ErbB4 and the C-terminus of ErbB3 is well-conserved [58].

Importin-β1 mediates the trafficking of EGFR into the nucleus and the EGFR/Importin-

β1 interaction is necessary for EGFR nuclear translocation, however the fundamental

interaction with nuclear pore complex has not yet been described for EGFR [59].

However, nuclear translocation of erbB2 also requires Importin-β1 interaction and this 26

complex interacts with Nucleoporin-358 (Nup358) and the small GTPase, Ran [60].

Because of the similarities in the NLS of EGFR and erbB2 it is hypothesized that Nup358

is also important in EGFR’s entry into the nucleus (Figure 1.4B). Chromosome region

maintenance 1 (CRM1) has been proposed as the protein responsible for nuclear export

of both EGFR and erbB2 [59, 60]. An increase of nuclear EGFR trafficking is often

observed following ligand stimulation of the receptor [56]. Indicating that

overexpression of a ligand (EGF) or hyperactivation of EGFR could promote aberrant

trafficking.

A recent report demonstrated that EGFR being trafficked to the nucleus is bound

to ligand and that nuclear EGFR remains tyrosine phosphorylated. The authors utilized a

radioactive (I125) labeled EGF and subsequently cross-linked the labled EGF to

membrane EGFR and following high-salt nuclear extraction the EGFR-EGF-I125 complex

was detected in the nucleus [13]. The presence of EGFR in the nucleus is a poor

prognostic indicator in several types of cancers including, breast and ovarian [44, 47].

Once in the nucleus EGFR directly or indirectly interacts with promoter regions to

activate transcription of genes such as cyclin D1, b-myb, iNOS, Aurora kinase A, and

COX-2 [52, 61-64]. All of these genes and their resulting proteins are important in the

development and progression of breast cancer. 27

Figure 1.4. EGFR Trafficking. A) In non-transformed cells, EGFR (green) is trafficked and degraded in lysosome. B) In cancer cells, EGFR interacts with other protein, such as MUC1 (red), and this results in aberrant trafficking of EGFR to the nucleus and increased membrane recycling. 28

Recently, altered EGFR trafficking leading to its accumulation and activity in the

nucleus has been demonstrated to be important in conveying chemotherapeutic

resistance. Hsu et al. discovered that following treatment with a DNA damaging agent

() and ionizing radiation the presence of EGFR in the nucleus activated DNA protein kinase (DNA-PK). This activation lead to the increase of DNA repair and resulted in increased cell survival [45]. Another report found MDA-MB-468 breast

cancer cell line was resistant to an EGFR kinase inhibitor and that nuclear EGFR in these

cells was unchanged [65]. The altered trafficking of EGFR to the nucleus commonly found in cancer could be playing a significant role in chemoresistance and cancer

progression (Figure 1.5). However, the function of EGFR in the nucleus to act as a

transcription factor and other proteins that could be modulating this process has yet to be

fully elucidated. 29

Figure 1.5. EGFR oncogenic functions: tyrosine kinase-dependent signaling (left) and direct transcriptional activator (right). Modified from [6]. 30

ErbB Receptors in Breast Cancer

The transformation of a normal cell to cancerous cell requires the acquisition of

several hallmarks of cancer, which include sustained angiogenesis, self-sufficiency in

growth signals, metastatic potential, limitless replicative potential, insensitivity to anti-

growth signals, and evading apoptosis [66]. ErbB receptors have become favored

therapeutic targets because of their prominent role in all of these hallmarks of cancer. In

addition, transgenic mouse models of breast cancer have been established that are

dependent on erbB receptors for tumorigenesis demonstrating their oncogenic potency.

These mouse models of breast cancer function by overexpression of the receptor

(MMTV-Neu) or receptor hyperactivation (WAP-TGFα) in the mammary gland [67, 68].

Several anti-EGFR therapeutics have been developed and most show efficacy in mouse

models of breast cancer, but this efficacy has not significantly translated to human breast

cancers setting [Reviewed in [21]]. In some cases targeting both EGFR and erbB2 at the same time was shown to accelerate cancer progression [69]. The limited success of these targeted therapies indicates the need for a better understanding of how EGFR functions in cancerous conditions.

EGFR is overexpressed in approximately 30% of all breast cancers, but in the basal-like subtype it is closer to 60%. This overexpression can be a result of several

different events including gene amplification, activating mutations and altered

trafficking. EGFR (7p12) gene amplification has been found in approximately 6% of

breast cancers and results in increased protein expression [70]. Loss of EGFR regulation 31

can also be a result of mutation. For example in glioma, non-small lung, and breast

cancer a mutant EGFR (EGFRvIII) which has an in-frame deletion of the extracellular

domain results in a constitutively active receptor [71]. Proteins that are important in

EGFR trafficking have also been implicated in overexpression of the receptor. For

example, the tumor suppressor, Sprouty, is important in the normal trafficking of EGFR,

but loss of this protein results in increased EGFR expression [72]. In several cancers, the

aberrant trafficking can lead to EGFR overexpression or accumulation of EGFR in the

nucleus, where it directly promotes the transcription of other oncogenes [52]. Up until

recently the sole focus of anti-EGFR therapies has been the signaling pathways downstream of EGFR activation, but in breast cancers these therapies have had limited

success.

ErbB Receptor Therapeutics

Several anti-EGFR therapeutics have been developed for the treatment of a

variety of cancers and the target of most of these drugs is the receptor’s tyrosine kinase

activity (Table 1.1). For example, (Iressa) is a small molecule tyrosine kinase

inhibitor, which attenuates downstream signaling. Gefitinib’s efficacy as an anti-tumor

agent was first demonstrated in a mouse xenograft models of breast, lung, and prostate cancers [73]. However, resistance to gefitinib has been determined to be the result of egfr gene mutations, compensation from other receptor tyrosine kinases (IGFR), upregulation of downstream effector proteins (Akt) or downregulation of inhibitory 32

proteins (PTEN) [74]. Additionally, most patients that initially respond to gefitinib will

develop resistance within 6-12 months [Reviewed in [75]]. Another anti-EGFR therapy

recently developed was the , (Erbitux), directed against

EGFR’s ectodomain, which prevents dimerization and subsequent downstream signaling.

Preclinical in vitro and in vivo studies for cetuximab show significant efficacy in inhibiting metastases, proliferation, and angiogenesis [Reviewed in [76]]. However, the results observed in preclinical trials have not translated to the clinic. Interestingly, the expression level of EGFR in the tumor cells is a poor indicator of drug response for both the tyrosine kinase inhibitor and monoclonal antibody [77, 78]. Although it is important to target EGFR’s kinase activity, recently a kinase-dead mutant of EGFR was able to inhibit autophagic cell death in cancer cells [79]. Authors discovered that EGFR stabilized glucose transporters to promote cell survival under cytotoxic conditions. This indicates an EGFR oncogenic function that is independent of its kinase activity and a possible mechanism of resistance. Although some mechanisms of anti-EGFR therapy resistance have been established, evidence for altered EGFR trafficking to the nucleus as mode of resistance is becoming apparent [45, 80]. An initial step in the progression of breast cancer that results in this aberrant trafficking of EGFR to the nucleus is the loss of cellular polarity. Maintenance of the apical-basolateral border is essential in normal cell function and deterioration of this border will result in aberrant protein localization,

regulation, expression, and activity.

33

Compound Type Target Company (Herceptin) Humanized mAb ErbB2 Genentech/Roche (Omnitarg) Humanized mAb ErbB2 Genentech Cetuximab (Erbitux) Chimeris mAb EGFR ImClone/Merck

Matuzumab Humanized mAb EGFR Merck

Panitumumab Humanized mAb EGFR Abgenix Gefitinib (Iressa) Tyrosine Kinase Inhibitor EGFR AstraZeneca (Tarceva) Tyrosine Kinase Inhibitor EGFR Genentech

Lapatinib Tyrosine Kinase Inhibitor EGFR/ErbB2 GlaxoSmithKline

AEE788 Tyrosine Kinase Inhibitor EGFR/ErbB2/VEGFR Novartis Irreversible Tyrosine CI-1033 Kinase Inhibitor EGFR/ErbB2 Pfizer Irreversible Tyrosine EKB-569 Kinase Inhibitor EGFR/ErbB2 Wyeth-Ayerst EXEL 7647/EXEL 0999 Tyrosine Kinase Inhibitor EGFR/ErbB2/VEGFR EXELIXIS

Table 1.1. Anti-ErbB Therapies. Tyrosine Kinase Inhibitors, TKI and monoclonal antibody, mAb.

Modified from [7]. 34

Cellular Polarity

Maintenance of the apical and basolateral borders has recently been described as a

“non-canonical tumor suppressor” because loss of this border promotes cellular

transformation. Normally, the cell polarity is maintained by three different complexes of

protein: the Pars complex (Par3-Par6-atypical PKC (aPKC), apical), Crumbs complex

(Crb, PALS1, and PATJ), and the Scribble complex proteins (Discs large, Dlg and lethal giant larvae, Lgl) [Reviewed in [81]] (Figure 1.6). The apical membrane is maintained by the Crumbs and Pars complexes as a result of aPKC (Pars complex) phosphorylation

of the Scribble (Scr) complex [82]. Phosphorylation of the Scribble complex promotes

its localization to the basolateral membrane. As a result of this event the Crumbs/Pars

and Scribble complexes distinguishes the apical and basolateral membranes, which is

vital in normal cellular function. Additionally, cellular polarity is also maintained by the

presence of tight junctions and adherens junctions, but the localization of these junctions is highly dependent on the activity of the Pars/Crumbs/Scribble interactions [Reviewed in

[81]]. Overall, regulation of polarity is a vital component in cellular homeostasis, which

upon disruption could promote catastrophic side-effects. 35

Figure 1.6. Maintenance of Cell Polarity. Several complexes of proteins are important in maintaining polarity, including the Pars, Scribble, and Crumbs complexes. Adherens and Tight junctions (AJC) also a play significant role in this process. Taken from [4]. 36

The relationship of aberrant EGFR trafficking and cell polarity in cancer has been

hypothesized to be a major component in EGFR-dependent breast cancers. Under normal

circumstance, EGFR basolateral trafficking is highly regulated, but following cellular

transformation cell polarity is lost and EGFR is localized throughout the cell [8]. In a

kidney epithelial cell line, there is altered downstream signaling as a result of

mislocalized apical EGFR [83]. Interestingly, EGFR and other erbB receptors have been

determined to directly affect cellular polarity by several different aspects including

inhibiting the function of the Pars complex and promoting the dissociation of β-catenin

from adherens junctions [84-86]. These events will ultimately results in the degradation

of cellular polarity thereby promoting initiation and transformation. Importantly, the loss

of polarity promotes novel EGFR-protein interactions, which results in a more severe

oncogenic phenotype [8]. For example, EGFR interacts with another oncogene, MUC1,

in a cancer-dependent fashion and this interaction promotes altered EGFR trafficking and

function [8, 87]. Inhibition of EGFR’s interaction with MUC1 inhibited the growth of

tumors in vivo, which demonstrates the importance of cell polarity maintenance and the

consequence of its deterioration.

37

MUC1

MUC1 expression has been correlated with poor prognosis in several different

cancers including cholangiocarcinoma [88], breast [89], colon [90], gastric [91], lung

[92], and renal cell carcinoma [93]. MUC1 was first characterized when a laboratory

generated mouse antibodies against human milk-fat globules, which could be utilized for

diagnosing breast cancer [94]. MUC1 has been observed to be overexpressed in about

90% of all breast cancers and its loss of apical localization is a poor prognostic indicator

[95, 96]. Overexpression of MUC1 in the 3Y1 rat fibroblast cell line and in the

mammary gland of mice (MMTV-MUC1) both resulted in transformation and

tumorigenesis [97, 98]. Importantly, MUC1 expression is important in the development

of tumors in EGFR and Wnt-dependent tumorigenesis mouse models [8, 98]. Table 1.2

provides the description of MUC1’s role in several different transgenic mouse models.

Mouse Genotype Transformation Phenotype Reference MMTV-MUC1 Mammary Gland Tumor Formation {Schroeder, 2004 #42}

MMTV-Wnt-1/Muc1-/- Delayed Tumor Progression {Schroeder, 2003 #41}

WAP-TGFα/Muc1 -/- Delayed Tumor Onset/Reduced Lung Lesions {Pochampalli, 2007 #35}

MMTV-pyMT/Muc1-/- Delayed Tumor Progression/ Reduction of Lung Metastases {Spicer, 1995 #44} No Change (Note: Found that erbB2 overexpression lead to MMTV-ErbB2/Muc1-/- {Adriance, 2004 #245} transcriptional repression of Muc1) Table 1.2. Role of MUC1 in mouse model of breast cancer. Mouse mammary tumor virus –MMTV, Whey acid protein-WAP, and polyoma virus middle T antigen- pyMT

Additionally, knock-down of MUC1 in several breast cancer lines resulted in the differential transcription of genes that are important in both proliferation and invasion. 38

Authors discovered that loss of MUC1 expression resulted in a decrease in genes

involved in MAP- kinase (MAP2K1) signaling and extracellular matrix interactions

(ITGAV) resulting in a significant decrease in proliferation and invasion [99]. Recently,

aberrant trafficking of MUC1 into nucleus promoted the interaction of MUC1 and the

transcription factor, NF-kB, resulting in transcriptional activation [100]. This

demonstrates that MUC1 function as an oncogene is not only dependent on it signaling

effector role, but its role in the nucleus as a transcriptional regulator. These studies

demonstrate MUC1’s impact on tumorigenesis and its importance in further

understanding the mechanism driving its oncogenic function.

MUC1 Structure

Mucin1 (MUC1) is part of a larger family which is composed of three subtypes:

gel-forming, soluble, and transmembrane mucins. There are approximately 20 genes

(MUC1-MUC20) which make up this larger family, but the majority of mucins are in the

gel-forming and transmembrane subtypes. MUC1 is >300kDa heterodimeric

transmembrane mucin and consist of a single pass hydrophobic region, which anchors the

protein to the plasma membrane (Figure 1.7). MUC1’s extracellular domain (ED) consists of a SEA (Sea urchin, Enterokinase, and Agrin) domain, which allows for the interaction of carbohydrates [101]. Additionally, MUC1’s ED is composed of several serine/threonine tandem repeat regions and the number of tandem repeat regions is highly variable as a result of genetic polymorphisms (Variable Number Tandem Repetition, 39

VNTR). The abundance of serine and threonines allows for extensive O-glycosylation of

the ED, which can account of up to 50% of the mucin’s mass [Reviewed in [102]].

Glycosylation of MUC1’s VNTR is important in its intracellular trafficking to the apical

membrane and following proper trafficking glycosylation aids in MUC1’s function.

Importantly, MUC1 expression is not isolated to glandular epithelial of the breast, but

expression has been associated with over 36 different tissue and cell types [Reviewed in

[103]]. Normal functions of MUC1’s ED consist of hydration and lubrication of the

lumen and microbial protection, but it is also able to interact with other extracellular

proteins [104]. The extracellular MUC1-protein interactions include galectin-3,

intercellular adhesion molecule-1 (ICAM-1), and selectins [105-107]. Galectin-3 is a

membrane associated galactoside-binding protein, which interacts with various O-glycans

on MUC1’s ED to promote adhesion. Recently, the over-expression of MUC1 has been

shown to upregulate galectin-3 expression, which promoted cancer-endothelial cell

interactions, a step that is vital in the metastatic process [108, 109]. Interestingly,

galectin-3 is also responsible for mediating the EGF-dependent extracellular interaction

of MUC1 and EGFR [108]. MUC1’s ED also interacts with ICAM-1 on endothelial cells

which provides another possible insight into MUC1’s role in intravasation and cancer

metastases [106]. Although MUC1’s extracellular interactions are important, its

intracellular protein interactions play a significant role in MUC1’s oncogenic function. 40

Figure 1.7. Structure of MUC1. MUC1 has a heavily O- glycosylated extrace llular domain (ecto-domain), a single pass transmembrane domain (TM) and a short cytoplasmic tail (CT) [2]. 41

MUC1 consist of a 72 amino acids cytoplasmic tail (CT), which has a plethora of

MUC1-protien interaction domains (Figure 1.8). Although the ED is important in the normal function the CT has been demonstrated to the main mediator of MUC1’s oncogenic function [110, 111]. The CT interacts with several proteins including, p53, estrogen receptor alpha, IKKβ, IKKγ, Src, Hsp90, Hsp70, β-catenin, EGFR, GSK3-β,

PKCδ, and Lck (Figure 1.8) [Reviewed in [4]]. Although it is important to note that some of these interactions are cancer-dependent as a result of disrupted cellular distribution and polarity. MUC1’s interaction with these others proteins is further discussed on page 43. The CT also has several domains important in post-translation trafficking such as the apical targeting, nuclear localization and oligomerization [112,

113].

MUC1 Transcriptional Regulation And Trafficking

MUC1’s transcription is regulated by several different transcription factors

including the estrogen receptor (ERα), signal transducer and activator of transcription

(STAT), nuclear factor –kappa B (NF-κB), and GATA3 [100, 114-116]. Following

transcription of the MUC1 gene, the mRNA is translocates to the endoplasmic reticulum,

where the protein is translated and inserted into the plasma membrane. Still in the

endoplasmic reticulum MUC1 is cleaved into an extracellular and membrane product,

following this proteolysis the two non-covalently linked subunits are trafficked through

the golgli to the apical membrane [117]. Apical membrane trafficking is achieved by the 42

presence of an apical targeting sequence (Cys-Gln-Cys) on MUC1’s juxtamembrane

domain [113]. During golgi trafficking, the extracellular domain is heavily O-

glycosylated, which will allow MUC1 to lubricate and hydrate the lumen [2, 118]. While

at the plasma membrane MUC1 will undergo constitutive internalization in order to

maintain O-glycosylation [119]. However in cancer MUC1’s level of O-glycosylation is

greatly reduced and its cellular distribution is altered. Recently, it has been determined

that following cleavage of the ED, the CT of MUC1 can translocate to the nucleus by

interacting with importin-β1 and nucleoporin p62 (Nup62) [112]. Additionally, this

nuclear translocation is dependent on MUC1’s ability to oligiomerize with Nup62 and

mutation of the amino acids important in this oligomerization results in a reduction of

transcriptional co-activation and tumorgenicity [112].

The most important attribute of MUC1’s cellular processes and function is it

cellular localization. Normally, MUC1’s apical localization is regulated by the apical

targeting sequence, but in cancer this regulation is lost and MUC1 can be detected in

several cellular compartments (basolateral plasma membrane, mitochondria, nucleus and

cytosol) [8, 120, 121]. Aberrant cellular localization promotes novel protein-protein

interactions that have been determined to be vital for MUC1’s oncogenic function.

Importantly, the loss of apical localization and accumulation of MUC1 in the cytosol

significantly correlates with a poor prognosis [122]. Although the importance of

maintaining cellular polarity is well-established the role of MUC1expression in this

maintenance has yet to be determined. 43

MUC1 Cytoplasmic Tail-Protein Interactions

The loss of MUC1 apical localization is poor prognostic indicator and is

commonly seen in breast cancer [122]. Aberrant cellular localization allows for novel

MUC1-protein interactions, which have been demonstrated to be important in MUC1’s

oncogenic function [123]. The cytoplasmic tail of MUC1 has been shown to contain

binding sites for many proteins that are implicated in transformation and progression of

cancer. These proteins include: GSK3β, c-Src, EGFR, β-catenin, and PKCδ [Reviewed

in [4] and [3]] (Figure 1.8).

Figure 1.8. MUC1’s Cytoplasmic Tail and its Protein Interactions. Taken from [3].

44

Loss of MUC1 in Wnt-1/β-catenin-dependent tumorigenesis significantly delays

tumor onset, which indicates that MUC1 is able to directly alter β-catenin function [124].

In vitro studies have concluded that MUC1’s cytoplasmic domain can promote β-catenin

dissociation from adherens junctions, which could also play a role in the breakdown of

cell polarity. Additionally, MUC1’s interaction with β-catenin blocks the degradation of

cytosolic β-catenin by inhibiting its GSK3β mediated serine/threonine phosphorylation

[124, 125]. Additionally, GSK3β interactions and serine phosphorylation of MUC1’s

cytoplasmic tail inhibits β-catenin/MUC1 interactions and initiates β-catenin degradation.

This indicates that the presence of MUC1 could act as switch to promote or inhibit b-

catenin stabilization, but this is highly dependent on other MUC1-protein interactions

(eg. MUC1-EGFR). Additionally, tyrosine phosphorylation on the MUC1 cytoplasmic

domain tyrosine, Y46, has been shown to increase the binding of β-catenin, Hsp70,

Hsp90 and c-Src [120, 126]. Additionally, the MUC1/β-catenin interactions have also

been detected in the nucleus, which promotes β-catenin dependent transcription [121].

The regulation of MUC1/β-catenin interaction that is mediated by MUC1’s tyrosine

phosphorylation is a possible mechanism to explain how activated EGFR promotes

MUC1/β-catenin interactions.

MUC1’s cytoplasmic tail also interacts with another oncogene, EGFR, in a

cancer-dependent fashion. Importantly, this interaction promotes the tyrosine

phosphorylation of MUC1’s CT (YEKV) and an increase in MAPK signaling [110].

Additionally, expression of MUC1 in an EGFR-dependent mouse model of tumorigenesis 45

promoted the formation tumors and lesions in the lung [8]. The authors discovered that

that expression of a vital cell cycle regulator, cyclin-D1, in these tumors was MUC1-

dependent. In vitro the presence of MUC1 following EGFR stimulation with EGF

resulted in a significant reduction in EGFR ubiquitination and increased recycling [87].

Additionally, inhibiting MUC1/EGFR interactions in both in vitro and in vivo models of breast cancer resulted in significant anti-proliferative and anti-oncogenic effects [123]

(Refer to chapter V).

Another oncogene, c-Src, interacts and tyrosine phosphorylates MUC1’s cytoplasmic tail and this interaction is important in tumorigenesis. In a c-Src dependent mouse model of breast cancer (MMTV-pyMT) the loss of Muc1 resulted in a significant delay in tumor onset and a reduction of pulmonary metastases [127]. The authors determined that expression of Muc1 promoted c-Src stability which promoted the interaction of c-Src with β-catenin and PI3K. Recently, a study using transgenic mice that over express MUC1 demonstrated that MUC1 was responsible for the development of mouse mammary tumors [98]. One mechanism driving MUC1-dependent transformation was discovered to be an increase in anti-apoptotic signaling such as, the

PI3K/Akt pathway [128]. Also, it is proposed that MUC1 inhibits the intrinsic apoptotic pathway through the interactions with Hsp70/90, which promote mitochondrial trafficking of MUC1’s CT [120, 129].

Overexpression of MUC1 alone is sufficient to drive tumorigenesis in a mouse model of breast cancer and the loss of MUC1 in other models (WAP-TGFα and MMTV- 46

Wnt-1) significantly delays tumor onset. In addition, our laboratory demonstrated that

the presence of MUC1 in an EGFR-dependent tumorigenesis mouse model of breast

cancer promoted tumor formation. Pochampalli et al found that in these tumors the expression of the cell cycle regulator/oncogene, cyclin D1, was dependent on MUC1 [8].

Additionally, these studies demonstrated that MUC1’s interaction with other known oncogenes (EGFR and β-catenin) is a cancer-specific event which promotes tumorigenesis and progression.

β-catenin Structure and Function

β-catenin signaling has been reported to be upregulated in greater than 50% of all breast tumors [Reviewed in [130]]. β-catenin is a 92kDa protein that has been shown to be conserved among several different organisms. The protein is made up of 12 armadillo domains, which are 42 amino acid repeats that form a superhelix [131, 132]. Normally, β-

catenin interacts with epithelial-cadherin (E-Cadherin) and α-catenin to form adherens

junctions which maintain cell-cell interactions [133]. E-Cadherin is calcium-dependent

transmembrane protein that functions by interacting with E-Cadherin from neighboring

cells to promote cell-cell adhesions. Additionally, E-Cadherin also functions to inhibit

receptor tyrosine kinases, by preventing dimerization and ligand binding [134]. β-catenin

has three highly conserved tyrosines that are critical in maintaining cell-cell adhesion

function. Tyrosines 142 and 654 are important in the interactions with α-catenin and E-

Cadherin. Tyrosine 489 is not in a known protein binding domain, although 47

phosphorylation of any three of these tyrosine results in β-catenin dissociation from

adherens junction [Reviewed in [135]]. The β-catenin/E-Cadherin complex of proteins

is responsible for binding the actin cytoskeleton, which promotes maintenance of cellular

and tissue organization. Adherens junctions can be stabilized through several serine

phosphorylations on E-Cadherin and p120 catenin [132]. p120 catenin is associated with

the juxtamembrane domain of E-Cadherin and loss of p120 results in the total loss of cell-cell adhesion [136].

In normal cells, once β-catenin dissociates from E-Cadherin it forms a complex with Axin and Adenomatous Polyposis Coli (APC) and then β-catenin is serine or

threonine phosphorylated by the Glycogen Synthase Kinase 3β (GSK3β).

Serine/threonine phosphorylation of β-catenin promotes the recruitment of the ubiquitin

ligase β-transducing repeat-containing protein (βTrCP). Ubiquitination of β-catenin

marks it for degradation by promoting its proteosomal trafficking, which degrades β-

catenin and prevents it from accumulating in the cytoplasm [137]. Both in normal and

cancer cells, Wnt signaling stabilizes cytoplasmic β-catenin by inhibiting the action of

the GSK3β/Axin/APC complex [138].

β-catenin/Canonical Wnt Signaling

The canonical Wnt/β-catenin signaling pathway is mediated by the binding of a

Wnt ligand (eg. Wnt-1) to the Frizzled (Frz) receptor/ Lipoprotein Receptor-Related 48

Protein (LRP) complex. Following ligand interaction with receptor complex, casein

kinase I/II is recruited, which activates the dishevelled (Dsh) protein. Activated Dsh will

inhibit GSK3β activity and prevent the serine/theronine phosphorylation of β-catenin

[139] (Figure 1.9). Normally, the is important in development

and is reported to induce differentiation and cell survival [Reviewed in [140]]. However,

in most transformed tissues β-catenin is not degraded, because of specific mutations

within β-catenin, upregulation of Wnt ligands, downregulation of Wnt inhibitors,

mutations in the GSK3β/Axin/ APC degradation pathway and/or deregulation of critical

signaling pathways [141, 142].

49

Figure 1.9. Canonical Wnt Signaling. Taken from [5].

50

β-Catenin in Breast Cancer

In breast cancer, EGFR is able to directly tyrosine phosphorylate β-catenin, which

results in a 6-fold reduction in E-Cadherin binding [143]. Recently, an indirect

mechanism of an EGFR-dependent β-catenin/E-Cadherin release was described, where

following transactivation of EGFR through treatment with Wnt-1 subsequent Erk2/CK2α

activation lead to decreased α-catenin/β-catenin association [144, 145]. Loss of α-

catenin/β-catenin association promoted the dissociation of β-catenin from E-Cadherin.

The reduced β-catenin/E-Cadherin association mediates the buildup of β-catenin in the

cytoplasm and this accumulation of stabilized β-catenin promotes its translocation into the nucleus [143]. The mechanism of β-catenin’s nuclear translocation is mediated by several mechanisms including direct interaction with nuclear pore proteins, Bcl-9-

dependent and/or TCF/LEF-1-dependent trafficking [146-148]. In the nucleus, β-catenin

can act as a transcriptional coactivator binding to the lymphoid enhancing factor/T-cell

factor (LEF/TCF) and eventually to TCF response elements in DNA. Later studies

reported that β-catenin/TCF response genes that were up-regulated were mostly pro-

proliferation (cyclin-D1), pro-survival (myc) and pro-migration (uPAR). It is also

important to note that β-catenin/TCF signaling plays an important role in mammary gland

development and maintenance [Reviewed in [140]].

The dissociation of β-catenin from adherens junctions results in down-regulation

of E-Cadherin and an up-regulation of neural-cadherin (N-cadherin). This cadherin

switch is in part a result of β-catenin-dependent transcriptional activation and SLUG1/2- 51

dependent transcriptional repression [149, 150]. In addition, the cadherin switch is a

common trait observed in metastatic cancers and is also utilized to determine the

occurrence of the epithelial to mesenchymal transition (EMT) [151]. Cancer cells that

have undergone EMT are often more migratory and invasive, which results in metastatic

tumors [Reviewed in [152]]. These secondary tumors will often result in organ failure

and are the main cause of cancer related deaths.

Peptide Therapy

The goal of understanding the basic science behind breast cancer development

and progression is to design less toxic and more effective targeted chemotherapies. For

example, the tyrosine kinase activity of erbB receptors has become a favored therapeutic

target (Refer to page 33). With respect to EGFR targeted small molecule tyrosine kinase

inhibitors (Iressa or Lapitinib) or antibody therapies (Cetuximab and Matuzimab), in vitro

and in vivo breast cancer demonstrated studies these drugs were highly effective.

However, in breast cancer clinical trials with these therapies have resulted in limited

efficacy and toxic side-effects (heart damage and skin damage). Recently, the use of cell-

penetrating peptide-based therapies has offered the benefit of targeted therapy with

minimal side-effects.

Cell penetrating peptides (CPPs) are 9-35mer cationic and/or amphipathic peptides that are rapidly endocytosed across cell membranes [153, 154]. Importantly, they can be linked to a variety of cargo, including anti-cancer therapeutics, making CPPs 52

an efficient, effective and non-toxic mechanism for drug delivery [123, 155-157]. CPP

conjugated therapies (CTTs) target a number of different processes specific to cancer

progression, including proliferation, survival and migration. In addition, many of these

CTTs also increase sensitivity to current anti-cancer therapy modalities, including

radiation and other DNA damaging chemotherapies, thereby decreasing the toxic dosage

required for effective treatment [158, 159]. Mechanistically, these CTTs function in a

dominant-negative manner by blocking tumor-specific protein-protein interactions with

the CPP-conjugated peptide or protein. Treatment of both cell lines and mouse models

demonstrate that this method of molecular targeting results in equal if not greater efficacy

then current standards of care, including DNA damaging agents and topoisomerase

inhibitors. For the treatment of invasive carcinoma, these CTTs have significant clinical

potential to deliver highly targeted therapies without sacrificing the patient’s quality of

life. The use of anti-cancer target CTTs is further discussed in chapter VI.

53

STATEMENT OF THE PROBLEM

Independently, β-catenin, EGFR, and MUC1 have all been shown to be

responsible for carcinogenesis. EGFR is overexpressed in ~ 60% and MUC1 is overexpressed in ~90% of basal-like breast cancers and β-catenin activity is aberrant in

~50% of breast cancer. In mouse models of breast cancer the co-expression of these

oncogenes promotes tumor onset and progression. In normal cells, these proteins are spatially separated, which prevents their interactions, but as cells transform and lose

polarity they are able to interact. The aberrant cellular localization of these proteins plays an important role in the development and progression of breast cancer. Specifically,

MUC1 has been shown to affect EGFR trafficking and β-catenin localization, which

further promotes their oncogenic function (Figure 1.10). For example, increased MUC1-

dependent EGFR recycling promotes aberrant tyrosine kinase activity, which can

contribute to the dissociation of β-catenin from adherens junctions and EMT.

Additionally the treatment of basal-like breast cancers with tyrosine kinase inhibitors

have had limited success indicating that kinase activity should not be the sole target in

future development of anti-EGFR therapeutics. The hypothesis is that MUC1/EGFR and

MUC1/β-catenin interactions promote the oncogenic activity of each protein and that targeting their interaction is a viable therapeutic option. To address this hypothesis I asked the following questions:

I) What is the role of MUC1 in EGFR or β-catenin trafficking to the nucleus?

a. Does MUC1 alter nuclear EGFR or β-catenin accumulation? 54

b. Does MUC1 alter nuclear EGFR function?

II) Evaluate the use of a CPP conjugated therapy (PMIP) to inhibit MUC1/EGFR

and MUC1/β-catenin interactions.

a. Utilize in vitro and in vivo models of breast cancers to determine

PMIP’s efficacy

III) Examine the role of EGFR activity in a Wnt-dependent mouse model of

tumorigenesis

Figure 1.10 Working Model. Epithelial cell polarization is lost during the progression of normal breast tissue (A) to a hyperplastic state (B) to breast cancer (C). This allows novel protein-protein interactions to occur, such as MUC1 (red)/EGFR (green) and MUC1/β-catenin (blue). MUC1 inhibits

degradation of EGFR and promotes recycling to the membrane. MUC1/EGFR promotes β-catenin dissociation from adherens junctions resulting in upregulation of oncogenic β-catenin transcriptional activation. Ultimately, these interactions promote breast cancer proliferation and metastases. It is important to note that epithelial cells are surrounded by an extracellular matrix (Background image: SEM of a collagen I matrix) that play an important mechanical and signaling role (Modified from [9]). 55

II. MATERIALS AND METHODS

Cell Lines

MCF10A, MDA-MB-468, BT20, and MDA-MB-231 cells were obtained from

American Tissue Culture Collection. MCF10A is an immortalized breast epithelial cell line, whereas MDA-MB-468, BT20 and MDA-MB-231 are transformed breast epithelial cell lines. MCF10A cells were maintained in RPMI with 10% FBS (Omega Scientific),

1% Penicillin-streptomycin (Cellgro), 10ng/ml cholera toxin (Sigma), 0.5 μg/ml

o hydrocortisone and 5ng/ml EGF at 37 C and 5% CO2. Cells were grown in RPMI

(MDA-MB-468 and MDA-MB-231, Cellgro) or EMEM (BT20, ATCC) with 10% FBS

o and 1% Penicillin-streptomycin at 37 C and 5% CO2.

Transfection with siRNA

Transfection with siRNA was performed as in [87]. The MUC1-1 siRNA targets

the extracellular domain (31045’-AAGACTGATGCCAGTA-GCACT-3’) [160] and the

siGENOME SMARTpool (M-004019-02-0020, Dharmacon/ThermoScientific) MUC1-2

siRNA targets the 3’UTR and open-reading frame (1-5’-

ACCAAGAGCUGCAGAGAGA-3’, 2-5’-GAUCGUAGCCCUAUGAGA-3’, 3-5’-

GCCGAAAGAACUACGGGCA-3’ AND 4-5’-CCACCAAUUUCUCGGA-CAC-3’).

Additionally, the control siRNA has no known mammalian gene targets (5’-

AATTCTCC-GAACGTGTCACGT -3’)(Qiagen). The transfections were performed with Lipofectamine 2000 (Invitrogen) as per the manufacturers’ specifications. Cells 56

were transfected, incubated for a specific time (MDA-MB-468, two days and MCF10A,

three days) and used for experiments.

Antibodies and Growth Factors

Table 2.1 PrimaryAntibodies Antibody Species Use Manufacturer MUC1 (CT-2) Hamster WB, IP, IF Neomarkers MUC1 (H295) Rabbit IF Santa Cruz EGFR (1005) Rabbit WB, IP, IF Santa Cruz EGFR (Ab-13) Mouse IP, ChIP Neomarkers EGFR (Ab-1) Mouse IF Neomarkers Phospho-EGFR (1173) Rabbit WB Cell Signaling ERK (9102) Mouse WB Sigma dpERK (M-8159) Mouse WB Sigma β-catenin (C-18) Goat WB Santa Cruz β-catenin (C-19220) Mouse IF BD Scientific β-actin (AC-15) Mouse WB Sigma Affinity BAP31 (MA3-002) Rat WB Bioreagents Histone H3 (C-16) Goat WB Santa Cruz Myosin IIa (D-16) Goat WB Santa Cruz IGF-R1β Rabbit WB Santa Cruz Phosphorylated Tryosine (PY99) Mouse WB Santa Cruz EEA1 (H-300) Rabbit WB Santa Cruz Phosphorylated Serine- 2 RNA polymerase II (H-5) Mouse ChIP Abcam

Secondary Antibodies Antibody Species Tag Manufacturer Anti-mouse Donkey 488, 584, HRP Invitrogen Anti-rabbit Donkey 488, 594, HRP Invitrogen Anti-goat Donkey 488, 594, HRP Invitrogen Anti-Armenian HRP, Texas Hamster Red, 488 Jackson Labs

57

Epidermal growth factor (EGF) was stored at -20oC at a concentration of 100ng/μl

(Invitrogen).

Immunoprecipitation and Immunoblotting

Protein was treated as in [87]. Nuclear protein was isolated via differential

detergent fractionation and the protein concentration was assessed by a Bicinchoninic

Acid assay. Protein (400 μg) was incubated in TNEN buffer (50mM Tris pH 7.5,

150mM NaCl, 1mM EDTA pH 8.0, 0.5% Igepal CA630, 2.0mM Sodium orthovanadate,

50 M ammonium molybdate, 10mM sodium fluoride, and Complete protease inhibitors)

and primary antibody (EGFR, Ab-13, Neomarkers, 2 μg) or mouse IgG for 1.5 hours at

4oC. The protein/TNEN/antibody mixture were tumbled at 4oC with agarose G beads

(Molecular Probes, Invitrogen) for 4 hours. The precipitations were washed three times

with TNEN buffer and then incubated with 2%SDS-PAGE buffer. Immunoprecipitations

were separated on an SDS-PAGE (Running buffer: 1X Tris-Glycine/0.1%SDS),

transferred (15% Methanol, 1X Tris-Glycine, 0.04% SDS) to PVDF (Millipore, Billerica,

MA). The membrane was blocked in either a 3% Milk/PBS/0.1% Tween solution or a

3% BSA/TBS/0.1% Tween and then used for immunoblot. Protiens on the membrane were treated with Super Signal Chemoilluminescent Substrate (Pierce), visualized on

Imagetech-B film (American X-ray, Louisville, TN) and developed with a Konica SRX-

101C. Densitometry of immuno-blots was performed with Image J software (NIH).

Immunofluorescence 58

Cells were grown on cover slips, treated with siRNA, and incubated with EGF as described above. Cells were fixed with a 4% Paraformaldehyde /PBS solution and permeabilized with 0.5% Triton X-100/0.02% NaN3/PBS. Following fixation, cells were

o blocked with 20% Fetal Bovine Serum (FBS)/ 0.02% NaN3/PBS and incubated at 4 C for

18 hours with primary antibody (1:100) in 10% Fetal Bovine Serum (FBS)/ 0.02%

NaN3/PBS (stock buffer). Secondary antibody (1:200) was incubated at room

temperature for 45 min in stock buffer. The slides were visualized with a Zeiss Axiovert

200 confocal microscope (Molecular and Cellular Biology Microscopy Facility,

University of Arizona), images were captured using a Zeiss AxioCam and analyzed with

the Zeiss Meta software.

Growth Factor Treatment

Prior to growth factor treatment, cells were serum-starved overnight. Cells that

were to be used to study the effect of EGF treatment were incubated with 10 ng/ml or

20ng/ml receptor grade EGF (Invitrogen) for 10 minutes on ice. Free EGF was then

washed twice with PBS and serum-free media was added. Cells were then incubated at

37ºC in serum-free media for the required time period before being lysed and

fractionated.

Differential Detergent Fractionation 59

The differential detergent fractionation protocol followed was modified from

[161]. Prior to lysis, cells were washed, collected, and resuspended in cold PBS. The

cell suspension was then centrifuged for 2 minutes at 200 x g, and the PBS was removed.

Cytoplasmic Extraction. Cells obtained from the previous step were resuspended in 300 uL of digitonin lysis buffer [0.01% digitonin, 100mM NaCl, 300mM sucrose, 10mM

PIPES (pH 6.8), 3 mM MgCl2, 5mM EDTA, 2.0 mM Sodium orthovanadate, 50.0 μM

ammonium molybdate and 10.0 mM sodium fluoride and Complete protease inhibitors

(Roche)] and incubated on ice for 5 minutes. The insoluble material was then pelleted for

10 min at 400 x g and the supernatant was stored as the cytosolic fraction.

Membrane Extraction. The pellet obtained from the cytoplasmic centrifugation was resuspended in 300uL of TritonX-100 lysis buffer [0.5% TritonX-100, 100Mm NaCl,

300mM sucrose, 10mM PIPES (pH 7.4), 3mM MgCl2, 3mM EDTA, 2.0 mM Sodium

orthovanadate, 50.0 μM ammonium molybdate and 10.0 mM sodium fluoride and

Complete protease inhibitors (Roche)] and incubated on ice for 5 minutes. Cells were

then subjected to centrifugation for 10 min at 5000 x g and the supernatant was stored as

the membrane fraction.

Nuclear Extraction. The pellet obtained from the membrane centrifugation was resuspended in 500uL of Tween 40/ DOC lysis buffer [1% Tween-40, 10mM NaCl, 0.5%

Deoxycholate, 10mM PIPES (pH 7.4), 1mM MgCl2, 2.0 mM Sodium orthovanadate, 50.0

μM ammonium molybdate and 10.0 mM sodium fluoride and Complete protease

inhibitors (Roche)]. The pellets were then dounced 20 times with a B-dounce and 60

sonicated 3 times for 10 seconds each. This was followed by centrifugation for 10 min at

7000 x g and the supernatant was removed as the nuclear fraction.

Chromatin-Bound Protein Isolation

Chromatin-bound nuclear protein fractions were extracted per the manufacturers’

protocol using the Subcellular Protein Fractionation Kit from Thermo Scientific. After,

the soluble nuclear fraction was isolated the chromatin-bound protein fraction was

isolated by treating the DNA/protein complexes with Micrococcal nuclease, which

digested the DNA thereby releasing the protein into the supernatant. Protein

concentrations for all fractions were determined by Bicinchoninic Acid assay (Thermo

Scientific) and all of the samples were stored at -80ºC until use.

High Salt Nuclear Extraction

Protein was extracted as in [13]. Cells were collected in non-nuclear extraction

buffer [20mM HEPES, pH 7.0, 10mM KCl, 2mM MgCl2, 0.5%NP40, 2.0 mM Sodium

orthovanadate, 50.0 μM ammonium molybdate and 10.0 mM sodium fluoride and

Complete protease inhibitors (Roche)] and dounced 30 times. Lysates were centrifuged

for 5min at 1,500g and supernatant was removed. Pellet (nuclei) was washed 3 times

with non-nuclear extraction buffer, resuspended in high salt nuclear extraction buffer

[500mM NaCl, 20mM HEPES, pH 7.0, 10mM KCl, 2mM MgCl2, 0.5%NP40, 2.0 mM

Sodium orthovanadate, 50.0 μM ammonium molybdate and 10.0 mM sodium fluoride

and Complete protease inhibitors (Roche)] and incubated at 4oC for 10min. Suspension

was centrifuged for 10min at 15000g and supernatant was collected as nuclear fraction. 61

Chromatin Immunoprecipitation

Epithelial cells were grown in 100mm dishes and treated with MUC1 or controls siRNA. Cells were exposed to EGF treatment and allowed to incubate for 120 minutes.

Cells were then incubated with 1% paraformaldehyde /PBS for 10 minutes at room temperature and then treated 0.125M glycine for 5 minutes at room temperature. Cells were washed with PBS, resuspended in PBS with 2.0mM Sodium orthovanadate, 50μM

ammonium molybdate, 10mM sodium fluoride, and Complete protease inhibitors and

counted. Cells were then snap frozen and kept at -80oC. These cells were subjected to

chromatin immunoprecipitation based on the Millipore EZ-ChIP protocol (17-371).

Briefly, cells were lysed and the protein/DNA lysates were sonicated (five-10sec bursts)

and pre-cleared with DNA blocked agarose G beads (16-201, Millipore) for 30 minutes at

4oC. The pre-cleared lysates were incubated overnight at 4oC with 5 μg of antibody for

EGFR (Santa Cruz, 1005 and Neomarkers, Ab-13), Mouse IgG, Rabbit IgG, and RNA

Pol II (Upstate, clone CTD4H8). The antibody/protein/DNA complexes were isolated by incubating agarose G beads for 60 minutes at 4oC. The agarose G

bead/antibody/protein/DNA complexes were washed several times and the crosslink was

reversed by incubating with NaCl overnight at 65oC. DNA was treated with RNase A,

proteinase K, and isolated with a Qiagen spin column. DNA was used for PCR to

amplify the ATRS region of cyclin D1 (F: 5’-TGGTCTTGTCCCAGGCAGAG-3’ and R:

5’-GATGGTGGCCAGCATTTC-3’) and GAPDH (F: 5'-

TACTAGCGGTTTTACGGGCG-3’ and R: 5'-

TCGAACAGGAGGAGCAGAGAGCGA-3). The cyclin D1 PCR was performed for 35 62

cycles at 98oC for 10sec, 65.3oC for 10sec, and 72oC for 25sec. The GAPDH PCR was

performed for 32 cycles at 98oC for 10sec, 62oC for 10sec, and 72oC for 25sec. The PCR

products were analyzed on a 2% agarose gel and visualized using a Kodak Gel Logic

2000.

Invasion Assay

Collagen matrix (0.9 mg/ml Type I Rat tail collagen (BD Bioscicences, Billerica,

MA), 83.0% (v/v) M-199 medium (Life Technologies), and 0.18% NaHCO3 (Fisher,

Hampton, NH)) was poured into a 96 well plate. Prior to collagen polymerization a

0.8μm pore transwell insert (Corning Inc., Corning, NY) was placed on top of the matrix.

The polymerized collagen was rehydrated using a chemoattractant (20% FBS/EMEM).

BT-20 cells were treated with 10µM of hPMIP, CP, PTD4 peptides or water in serum free EMEM media overnight. Prior to loading the cells onto the transwell inserts they

were fluorescently labeled with Calcein-AM (Invitrogen) for 30 minutes and then washed

with PBS. The cells were then resuspended in a 10µM peptide/EMEM serum free media and loaded onto the inserts in each well (15,000 cells/well). The cells were allowed to

o invade into the matrix for 10hrs at 37 C with 5% CO2 in a humidified incubator. After

10hrs, the collagen matrix was treated with a 0.25% Collagenase in 40% FBS/PBS and

the inserts were removed (Calbiochem, San Diego, CA). The number of fluorescently

labeled cells that had invaded into the collagen were measured using a Molecular Devices

spectrophotometer (Ex:485, Em:538, and Cutoff:530). For each treatment group, a set of 63

cells were placed directly into the bottom chamber and used as a reference for 100% of

cell invasion. Invasion is then indicated as percent invasion (invaded cells/total cells).

MTT Assay

BT20 cells were plated into a 96 well plate (5 X 103 cells per well). The cells

were allowed to adhere and then they were treated daily with 10μM of hPMIP, CP, PTD4

peptides or water (treatments diluted in 10% FBS/EMEM) for seven days. After treatment the Vybrant MTT Cell Proliferation Assay (Molecular Probes) was performed following the manufacturers’ protocol.

Peptide Synthesis

hPMIP, Biotin-hPMIP, msPMIP, FITC-msPMIP, CP and PTD4 polypeptides were synthesized by GenScript (Scotch Plains, NJ) and delivered lyophilized. The peptides were resuspended at a concentration of 500μM in water and stored at -80oC in single-use aliquots. Peptide sequences are shown in Figure 5.1.

Mice

Mouse Genotyping. The genotypes of the mice were determined by extracting DNA from a small portion of the tail. The tail was incubated in a lysis buffer (10mM Tris, pH 8.0,

50mM NaCl, 25mM EDTA, pH 8.0, 0.5% SDS, and 0.5mg/ml Proteinase K) overnight and the DNA was extracted. Subsequently the DNA was utilized for the MMTV-Wnt-1

PCR (IMR513 - 5’-GGACTTGCTTCTCTTCTCATAGCC-3’ and IMR514 - 5’-

CCACACAGGCATAGAGTGTCTGC-3’) and waved-2 PCR (Wa2 F – 5’- 64

CCCAGAAAGGGATATGCG-3’ and Wa2 R – 5’-GCAACCGTAGGGCATGAG-3’, 45

cycles of 30 seconds at 94oC, 30 seconds at 55 oC, and 30 seconds at 72 oC). Note the

product of the waved-2 PCR (170bp, Egfrwildtype) was treated with a restriction enzyme

(FokI) to confirm the presence of a 95bp and 75bp piece ( Egfrwa2). The digested PCR products were separated on a 2% agarose gel to confirm size.

Mouse Protein Lysates

Protein lysates from mouse tissues were generated as in [8]. Mammary glands were removed from the mouse and immediately homogenizing in a tissue lysis buffer

(20mM HEPES, pH 7.0, 150mM NaCl, 1% Triton X-100, 2mM EDTA, pH 8.0, 2mM

EGTA, pH 8.0, 2.0 mM Sodium orthovanadate, 50.0 μM ammonium molybdate and 10.0 mM sodium fluoride and Complete protease inhibitors). Lysates were stored at -80oC.

Whole Mounts

As performed in [8]. Mammary glands were removed from the mice and fixed in

1:3 glacial acetic acid:ethanol for one hour. Fixed glands were washed with 95% and

100% ethanol for 15 minutes and then defatted with acetone for three days. Following

the acetone treatment, the glands were subsequently washed with 100%, 95%, and 70% for 15 minutes and briefly with distilled water to rehydrate the gland. Rehydrated glands were stained with 0.2% carmine/0.5% aluminum potassium sulfate overnight and then destained with subsequent 70%, 95%, 100%, 95% and 70% ethanol washes for 15 minutes each. Stained mammary glands were stored in glycerol solution, imaged with a

Leica dissecting microscope and analyzed using the MagnaFire software. 65

Xenograft Tumor Studies (PMIP)

Immunocompromised (scid) mice (Taconic, Rockville, MD) were tested for the

presence of serum IgG and found to be <20 μg/ml IgG. Female mice (four to six weeks

old) were injected with 1x107 cells embedded in Matri-gel (BD Biosciences) into the

mammary fat pad and allowed to grow to either 100mm3 or 500mm3, based on the

2 formula a x b/2 where a is the smaller diameter and b is the larger diameter. Mice were

injected intraperitoneally (i.p.) with 50 μg/g body weight of either PMIP or PTD4 control

peptide for 21 days and measured with calipers every two days. Either at the end of 21

days, or after the tumor had reached 800mm3, tumors were resected by injecting the mice

with buprenorphine (2.5 mg/kg body weight, Infusion Solutions, Totowa, NJ) at least one

hour prior to resection and anesthetizing the mice with isoflurane (Abbott, Abbott Park,

Il). Following surgery, mice were treated with buprenorphine (2.5mg/kg body weight) 8

and 16hrs post-surgery. Animals were then followed for 10 days to examine regrowth at

the primary tumor site or at secondary mammary glands, then sacrificed.

Genetically-Driven Tumor Studies (PMIP)

MMTV- pyV mT mice on an FVB background (W. Muller [67], obtained from

Jackson Laboratories) entered into study upon the development of tumors that measured

> 0.5cm in at least one diameter. Only mice greater than seven weeks of age were included in the study and mice that developed fluid cysts were excluded. Animals were injected i.p. with 50μg/g body weight of msPMIP or PTD4 once daily for 21 days. Each of ten mammary glands were measured using calipers every two days, and measurements 66

2 were used to determine tumor volume based on the formula a x b/2, with a being the

smaller diameter and b being the larger diameter. After treatment for 21 days, mice were

sacrificed by CO2 inhalation and tissues resected. Several msPMIP treated mice were

injected with FITC-msPMIP (50μg/g body weight) one or four hours prior to sacrifice

and intact tissues were visualized using a fluorescence MZFLIII Leica dissection

microscope. Both the xenograft and transgenic PMIP mouse studies were performed by

the Experimental Mouse Shared Service (University of Arizona, Tucson).

MMTV-Wnt-1 Transgenic Mouse Study

MMTV-Wnt-1 mice were obtained from Jackson Laboratories and waved-2 mice

were provided by Dr. Noreen Luetteke from the University of South Florida, Tampa.

The mice were maintained at the Arizona Health Science Center Animal facility (Tucson,

AZ) under conditions specified by the Institutional Animal Care and Use Committee.

MMTV-Wnt-1 mice were outbred (C57Bl6/FVB) onto the Egfrwa-2/wa2 background and at

least 14 virgin female mice from each genotype were generated (n=16, MMTV-Wnt-1

Egfr+/+ ; n=14, MMTV-Wnt-1 Egfrwa2/+ ; n=15, MMTV-Wnt-1 Egfrwa-2/wa2). After eight

weeks of age mice were palpated once a week until formation of a mammary gland tumor

and then the tumor volume was measured three times a week based on the formula a2 x b/2 where a is the smaller diameter and b is the larger diameter. Once the tumors grew to

larger than one centimeter the mouse was sacrificed and the tumors, contralateral

mammary, and lungs were collected. Protein lysates were generated for the tumors and

contralateral mammary glands. A small portion of the tumor and another contralateral 67

mammary gland were fixed in 10% buffered formalin and embedded in paraffin. Finally,

another contralateral mammary gland was used for whole mount.

All studies were performed under protocols approved by the Institutional Animal Care

and Use Committee of the University of Arizona.

Statistical Analysis

All statistics were performed in Excel (Microsoft) or STATA 10.0 (StataCorp). Test

implemented were two-tailed student t-test and one way analysis of varience (ANOVA).

68

III. MUC1 REGULATES NUCLEAR LOCALIZATION AND FUNCTION OF THE EPIDERMAL GROWTH FACTOR RECEPTOR

Note: The work presented in this chapter is published in Journal of Cell Science (ADD FINAL CITATION). Except Figure 3.3

INTRODUCTION

Subcellular protein trafficking is a highly regulated process that is essential for

maintaining normal cell function and viability. In fact, the mislocalization of proteins

plays a fundamental role in promoting diseases such as cystic fibrosis, diabetes, and

several types of cancer [Reviewed in [162]]. In cancer specifically, aberrant nuclear

localization of receptor tyrosine kinases, such as, c-MET, ErbB1/EGFR, ErbB2, ErbB3,

ErbB4, FGF, and VEGF, results in increased transcription of genes that promote

chemoresistance, proliferation and disease progression [52-55, 163-165]. Notably, these

effects are not due to increased signal transduction, but are instead due to the ability of

these proteins to act as transcriptional cofactors [55]. A number of these proteins contain

nuclear localization signals (NLS), including the erbB family of receptors (ErbB1-4),

which share a highly conserved tri-partite NLS (Consensus Sequence: [RK](3)-x{2,3}-

[RK]{3,4}-x{2,3}-RR) [58]. While this NLS can drive nuclear translocation, the

receptors typically remain localized to the plasma membrane in polarized epithelium.

The Epidermal Growth Factor Receptor (ErbB1/EGFR) is an example of a protein

that has altered subcellular localization in cancer. In normal polarized epithelium, EGFR

is targeted to the basolateral membrane by well-conserved localization sequences 69

(KRTLRRLLQERELVEPLTPSGEAP) [29]. Under these circumstances, ligand stimulation induces receptor dimerization, ubquitination, endocytosis and degradation in the lysosome, thereby limiting EGFR expression and signaling [Reviewed in [166]].

However, in cancer cells it has recently been discovered that EGFR undergoes retrograde trafficking through the endoplasmic reticulum Sec61 translocon, which is followed by release into the cytosol [56]. Following this release from the membrane, EGFR’s tripartite nuclear NLS interacts with Importinβ1 allowing it to be shuttled through the nuclear pore complex into the nucleus [5]. Once inside the nucleus, it has been proposed that EGFR directly interacts with the endogenous promoter and initiates transcription of the cell cycle regulator, cyclin D1 [52]. Finally, the CRM1 exportin is capable of facilitating the nuclear export of EGFR [5]. Of note, the presence of EGFR in the nucleus of tumor cells correlates with a poor clinical outcome in breast, ovarian, oropharyngeal, esophageal squamous cell, and epithelial carcinomas [47].

The heterodimeric transmembrane mucin MUC1 is also highly expressed in epithelial carcinomas and inhibits the ubquitination of EGFR, altering normal EGFR trafficking [87]. In polarized ductal epithelium, MUC1 is constitutively internalized and recycled between the apical membrane and the golgi, which enables it to maintain a high level of O-glycosylation [119]. The apical localization of MUC1 is regulated by a well- conserved targeting sequence in the juxtamembrane domain (CQC) [113]. In non- polarized breast epithelium, MUC1 and EGFR interact at the plasma membrane resulting in increased EGFR internalization, the loss of lysosomal degradation and an increase in

EGFR recycling [87]. In addition to membrane localization, MUC1 is overexpressed and 70

mislocalized to the cytosol and nucleus in about 90% of breast , correlating with poor prognosis [89, 98, 110, 122]. Notably, the loss of Muc1 significantly delays tumor onset in an EGFR-dependent (WAP-TGFα) transgenic breast cancer model [8, 167]. In this study, loss of Muc1 within EGFR-dependent mammary gland tumors is also associated with a reduction of cyclin D1 expression [8].

MUC1 trafficking to the nucleus is due to the presence of a non-classical NLS

(RRK) that interacts with Nucleoporin 62 and Importinβ1, allowing for the nuclear co- translocation of numerous proteins, including γ-catenin, β-catenin and estrogen receptor

[97, 112, 121, 168]. In fact, the transcriptional activity of β-catenin is dependent on

MUC1’s nuclear localization, indicating that MUC1 is acting as co-factor [111, 112]. In addition, transcriptional targets of these proteins are activated in a MUC1-dependent manner. Given MUC1’s influence on EGFR trafficking following receptor activation and

EGFR’s newly described nuclear trafficking pathway, our current study was designed to determine if MUC1 functions as a regulator of EGFR nuclear translocation and transcriptional activation of the gene, cyclin-D1.

In this report, I demonstrate a novel role for MUC1 in EGFR regulation. Through biochemical and genetic analysis, I show that MUC1 alters the nuclear localization and function of EGFR in a manner that may have important consequences for EGFR- dependent tumors.

RESULTS

71

MUC1 promotes accumulation of nuclear EGFR

The loss of Muc1 expression significantly suppresses EGFR-dependent cancer

progression and cyclin D1 protein expression in the WAP-TGFα model [8]. In the

current study, I set out to determine if the mechanism by which MUC1 alters EGFR-

dependent cyclin D1 expression is via a novel regulation of nuclear trafficking and direct

transcriptional function of EGFR.

To examine the effect of MUC1 on EGFR cellular localization, I utilized two cell

lines expressing MUC1 (MCF10A and MDA-MB-468). Cells were transfected with either MUC1-specific siRNA to knock down MUC1 protein expression or a control

siRNA, and changes in EGFR localization were evaluated. Optimal knockdown of

MUC1 was observed after treatment with either MUC1-1 or MUC1-2 specific siRNAs after three days (MCF10A), as previously demonstrated with these MUC1 target sequences (Figure 3.1) [87]. For all fractionation studies, cells were serum-starved for

18 hours, treated with 10ng/mL EGF on ice for 10 minutes (to saturate membrane-bound receptors), and then excess ligand was removed. Cells were then incubated at 37oC for

120 minutes to induce receptor endocytosis and trafficking. Cells were then fractionated by differential detergent fractionation which takes advantage of the ionic properties of different detergents to isolate the membrane (Triton X100), cytosolic (Digitonin), and nuclear (Tween40) protein fractions [161]. Note that the membrane fractions include plasma membrane, mitochondria, golgi, endosomes, and the endoplasmic reticulum.

Proteins isolated from the membrane, cytosolic, and nuclear compartments were then separated by SDS-PAGE and examined by subsequent immunoblotting to determine the 72

effects of MUC1 expression on EGFR localization. To confirm fraction purity and equal

loading, expression of proteins known to localize in each cellular compartment were

analyzed (Plasma Membrane, IGF-1Rβ; Endoplasmic Reticulum Membrane, BAP31;

Endosomes, EEA1; Cytosolic, Myosin IIa; Nucleus, Histone H3) [161].

Analysis of protein lysates from MDA-MB-468 cells revealed that MUC1 and

EGFR are present in both the membrane and nuclear fractions under conditions of serum

starvation and EGF treatment (Figure 3.1A, top 2 panels). MUC1 expression did not

affect EGFR localization to the membrane fraction in the absence of EGF, although loss

of MUC1 resulted in decreased levels of EGFR in the membrane fraction in the presence

of EGF. This recapitulates the result of a previous study that described increased EGFR

membrane recycling in the presence of MUC1 (Figure 3.1A, top panel) [87]. The

maximal nuclear EGFR accumulation was observed after 120 minutes of EGF

stimulation, a finding similar to what was previously observed by another group [169].

Interestingly, EGFR was observed in the nucleus in the presence or absence of EGF,

indicating that nuclear EGFR trafficking can occur in the absence of serum in these cell

lines (Figure 3.1A, top panel). Importantly, in both serum starved and EGF-treated cell lines, I observed that the absence of MUC1 inhibited the accumulation of EGFR in the

nucleus (Figure 3.1A, top panel). The effect of MUC1 on EGFR localization was found

to be similar in MCF10a cells with both MUC1-1 and MUC1-2 siRNAs (Figure 3.1B).

To determine if this result was an artifact of endoplasmic reticulum or post-lysis

contamination I performed a high-salt nuclear protein extraction on cells treated with

control or MUC1-1 siRNA. Cells were serum starved or stimulated with EGF for 0min 73

or 120min, subjected to high-salt protein extraction and proteins were analyzed by

immunoblotting. I discovered that after serum starvation and 0 min of EGF treatment there was a minimal difference in total nuclear EGFR with respect to MUC1 expression.

However, after 120 minutes of EGF treatment there was a significant MUC1-dependent accumulation of total EGFR in the nucleus (Figure 3.1C). This demonstrates that MUC1 promotes the accumulation of non-membrane bound nuclear EGFR.

74

Figure 3.1 MUC1 promotes nuclear EGFR Localization. A and C) MDA-MB-468 breast cancer cells or B) MCF10a immortalized breast epithelial cells were treated for three days with control (Ctrl) or MUC1 (MUC1-1 or MUC1-2) siRNA, were either serum starved (-Serum) for 18hrs or serum starved and then treated with EGF (10ng/ml, 120’ EGF). Cells were fractionated and protein (6μg) was separated by SDS-PAGE. After transfer to PVDF, proteins were immunoblotted (IB) for EGFR (Santa Cruz, 1005) and MUC1 (Neomarkers, CT2). Fraction purity and loading were determined by immunoblotting for BAP31(Affinity Bio-Reagents, MA3-002; Endoplasmic Reticulum Membrane, Mem), IGF-1Rβ (Santa Cruz, SC-713; Plasma Membrane), EEA1 (Santa Cruz, SC-33585; Endosomes), Myosin IIa (Santa Cruz, D16; Cytosolic, Cyto), Sp1 (Santa Cruz, 1C6), and Histone H3 (Santa Cruz, C-16; Nuclear, Nuc). White lines through blots indicate same samples and exposure but were non-contiguous. The molecular weights (kDa) are indicated on the right. Note that the cytoplasmic domain of MUC1 runs as multiple species 14-28 kDa, because of different glycosylation states [110].

75

To visualize the subcellular localization of EGFR and MUC1, control and MUC1 siRNA treated cells were analyzed by laser-scanning confocal microscopy. Cells were serum-starved and either left untreated (Figure 3.2, A-H’) or treated with 20ng/mL of

EGF for 120 minutes (Figure 3.2, I-P’), then immunolocalized for MUC1 (Figure 3.2, A,

E, I, M) and EGFR (Figure 3.2, B, F, J, N). I observed that in the absence of EGF, EGFR was mainly localized to the plasma membrane with limited cytosolic and nuclear localization, regardless of MUC1 expression (Figure 3.2, B and F). Under similar conditions, MUC1 localization was largely nuclear and cytosolic (Figure 3.2, A).

Following EGF treatment, punctate staining of EGFR was observed within the cytosol of cells from both treatment conditions (Figure 3.2, J and N, outlined arrows), but in MUC1 siRNA treated cells the cytosolic EGFR mainly localized to perinuclear regions (Figure

3.2, N and P’, outlined arrows) whereas in control siRNA treated cells that maintained

MUC1 expression, significant punctate EGFR staining was observed in the nucleus

(Figure 3.2, J and L’, arrowheads). These data corroborate our observation that MUC1 expression promotes EGFR nuclear accumulation.

76

Figure 3.2 MUC1 promotes nuclear EGFR accumulation. Immortalized breast epithelial cells (MCF10A) were treated with MUC1-1 (E-H’, M-P’) or Control (Ctrl) siRNA (A-D’, I-L’) and stimulated with EGF (20ng/mL) for 120 minutes. (I-P’) Cells were fixed, permeabilized, and used for immunostaining against EGFR (Neomarkers, Ab-1) (green, B, F, J, N) and MUC1 (Neomarkers, CT2) (red, A, E, I, M). (A-H’) Under serum starvation conditions (- serum) EGFR localized to the plasma membrane (white arrows), nucleus and cytoplasm. (I-L) In presence of EGF and MUC1, EGFR is localized within the nucleus (white arrowheads, L’). (M-P’) EGF treatment in the absence of MUC1 resulted in EGFR localization to perinuclear regions (white open arrows, P’). DAPI= Blue nuclei. A-P magnifications are 630X. Scale Bars=20μm

77

Inhibiting MUC1/EGFR interaction decreases nuclear EGFR

Knocking down MUC1 expression resulted in a significant decrease in nuclear

EGFR localization. To confirm that the interaction of MUC1 and EGFR promoted nuclear accumulation of EGFR I utilized the MUC1 mimetic peptide, PMIP, to inhibit the interaction. PMIP was observed to efficiently inhibit EGFR and MUC1 interactions in the BT20 breast cancer cell line resulting in a significant decrease in the proliferative and migratory potential (Figure 5.2 and 5.3) [123].

I next examined the effect of PMIP of the localization of EGFR. MDA-MB-468 cells were treated for 18 hrs with 20μM PMIP in serum-free media, stimulated with EGF for 120 minutes and the protein from cells was fractionated. Protein fractions were analyzed by immunoblotting as in previous experiment. I discovered that PMIP was able to inhibit nuclear EGFR accumulation compared to non-treated and control peptide treated cells (Figure 3.3). Lysate purity was confirmed by immunoblotting for endoplasmic reticulum membrane (BAP31) and nuclear (Histone H3). 78

Figure 3.3 Inhibiting MUC1/EGFR interaction inhibits nuclear EGFR accumulation. MDA-MB-468 cells were treated with PMIP (20uM) for 18hrs and then stimulated with EGF (10ng/mL) for 10 minutes at 4oC. Cells were then incubated for 120 minutes and subjected to differential detergent fractionation. Protein fractions were separated on an SDS-PAGE and used for immunoblotting against EGFR (Santa Cruz, 1005), BAP31 Affinity Bio-Reagents, MA3-002; Endoplasmic Reticulum Membrane, Mem) and Histone H3 (Santa Cruz, C-16; Nuclear, Nuc).

79

MUC1 and EGFR Interact in the Nucleus

The loss of MUC1 results in a significant decrease in nuclear EGFR. I next

wanted to determine if MUC1 and EGFR were interacting within the nuclear

compartment. MDA-MB-468 and MCF10A cells were treated as described in the

previous section and subjected to differential detergent fractionation. To determine if

MUC1 and EGFR were interacting within the nucleus of these cells, I isolated the nuclear

protein (400μg) and performed immunoprecipitation directed against EGFR and mouse

IgG, followed by immunoblotting against EGFR and MUC1. Following 120 minutes of

EGF treatment at 37oC, MUC1 and EGFR form a complex in the nucleus (Figure 3.4A,

top panel). Additionally, nuclear protein lysates were analyzed to confirm MUC1

expression and the resulting EGFR nuclear accumulation (Figure 3.1B).

To determine if interacting MUC1 and EGFR in the nucleus were also binding

nuclear DNA, I performed a fractionation that would allow us to distinguish between

soluble and chromatin-bound nuclear proteins. After EGF treatment, cells were

subjected to subcellular fractionation and the nuclear protein bound to chromatin was

isolated as a separate fraction from the soluble nuclear protein. This was accomplished

by first gently collecting protein from the nucleus and then collecting the

protein/chromatin complexes. These complexes were then treated with Micrococcal

nuclease, which released chromatin-associated protein into the supernatant. I found that

while EGFR was present at the same levels in the soluble nuclear fraction regardless of

MUC1 expression, EGFR was only found bound to chromatin in the presence of MUC1 80

(Figure 3.4B, Nuc CB). This data demonstrates that EGFR’s interaction with chromatin

is MUC1-dependent. Moreover, this data addresses the question of whether MUC1-

dependent changes in EGFR levels in the nucleus are a result of altered EGFR

degradation, as soluble nuclear EGFR was equivalent regardless of MUC1 expression

(Figure 3.4B, Nuc S). Therefore, the presence of MUC1 appears not to alter

accumulation of soluble EGFR in the nucleus, but appears to be vital in EGFR’s ability to

interact with the DNA.

81

Figure 3.4 MUC1/EGFR interaction in the nucleus promotes EGFR/chromatin interaction. A) MCF10A immortalized breast epithelial cells were treated with control or MUC1-1 siRNA. Cells were serum-starved and treated with EGF (10ng/ml, 120 minutes at 37oC). The nuclear protein fraction (400μg) was used for immunoprecipitation (IP) with antibodies against EGFR (Neomarkers, Ab-13) and mouse IgG, and the precipitated protein was separated on an SDS-PAGE and immunoblotted (IB) for EGFR (Santa Cruz, 1005) and MUC1 (Neomarker, CT2). SL= straight lysate (non-IP protein, 12μg). Fraction purity was confirmed by immunoblotting for Histone H3 (Santa Cruz, C-16) and Myosin IIa (Santa Cruz, D16). Note: Straight lysates for this experiment are shown in Figure 3.1B. B) MDA-MB-468 breast cancer cells were was fractionated into membrane (Mem), cytosolic (Cyto), nuclear soluble (Nuc S), and chromatin bound (Nuc CB) fractions and separated (15μg) on an SDS-PAGE and immunoblotted (IB) for EGFR (Santa Cruz, 1005) and MUC1 (Neomarker, CT2). Fraction purity and loading was determined by immunoblotting for BAP31 (American Bio-Reagents, MA3-002), Myosin (Santa Cruz, D16), Sp1 (Santa Cruz, 1C6) and Histone H3 (Santa Cruz, C-16). The molecular weights (kDa) are indicated on the side.

82

EGFR Colocalizes with Phosphorylated-RNA Polymerase II

Considering the ability of EGFR to interact with chromatin and ability to directly induce transcription, I next wanted to determine if MUC1 could affect EGFR-mediated transcriptional activation [52]. A classical marker for transcription initiation is phosphorylated serine-2 RNA polymerase II (pS2-CTD-RNA Pol II) [170]. Therefore, to visualize EGFR and pS2-CTD-RNA Pol II localization, MCF10A cells were transfected

with control or MUC1-1 siRNA and stimulated with 20ng/mL EGF for 120 minutes.

Following EGF treatment, cells were fixed, permeabilized, and pS2-CTD-RNA Pol II and

EGFR were immunolocalized. Under serum starvation conditions, EGFR was mainly

observed at the plasma membrane and there was no positive pS2-CTD-RNA Pol II

staining regardless of MUC1 status (Data not shown). Alternatively, in the presence of

EGF, pS2-CTD-RNA Pol II was readily observed in the nucleus in the presence of

MUC1 (Figure 3.5B, outlined arrows). Additionally, following 120 minutes of EGF

treatment in MUC1-expressing cells, there were a significant number of EGFR-positive

punctate spots within the nucleus (Figure, 3.5A arrows) as observed in figure 3.2J. This

nuclear EGFR was found colocalized with pS2-CTD-RNA Pol II in the presence, but not

in the absence of MUC1 (Figure 3.5, D’ compared to H’, white arrowheads). Note that

the colocalization indicates that EGFR is localizing to transcriptional start sites [170]. To

confirm that the activity of pS2-CTD-RNA Pol II localization was not MUC1-dependent,

I also treated cells with 50ng/mL of . I found that insulin treatment for 120

minutes resulted in punctate pS2-CTD-RNA Pol II nuclear localization (Figure 3.5J,

outlined arrows) and did not affect membrane localization of EGFR (Figure 3.5I, white 83

arrowhead). These data suggest that MUC1 plays a regulatory role in both EGFR

trafficking to the nucleus and its interaction with active transcriptional elements.

84

Figure 3.5 EGFR colocalizes with phosphorylated RNA polymerase II. MCF10A cells were treated with MUC1-1 (E-H’) or control (Ctrl) siRNA (A-D’, I-L’) and stimulated with 20ng/mL of EGF for 120 minutes. Following treatment the cells were fixed, permeabilized and incubated with antibodies against EGFR (Santa Cruz, 1005) (green, A, E, I) and phosphorylated serine-2 RNA polymerase II (pS2-CTD-RNA Pol II)(Abcam, H5) (red, B, F, J). Cells were treated with DAPI to visualize nuclei (C, G, K) and composite colocalization of all three images are shown in D, H, and L. (A-D): Colocalization (yellow) is seen at the white arrowheads (D’). (E-H): Cytoplasmic EGFR staining is shown by a white arrow, E. (I-L): Cells were also treated with insulin (50ng/mL) to illustrate membrane EGFR localization (large white arrowhead, I) and pS2- CTD-RNA Pol II without EGF treatment (white outline arrow, L’). DAPI= Blue Nuclei. A-L’ magnifications are 630X. Scale Bars=10μm

85

MUC1 promotes EGFR-mediated cyclin D1 expression

EGFR’s ability to activate transcription of cyclin D1 and the importance of cyclin

D1 in breast cancer progression has previously been described [52]. EGFR has been

shown to interact with the Adenine/Thymine Rich Sequence (ATRS) promoter region of

the cyclin D1 gene on promoter constructs, although EGFR has not yet been reported as

actively engaging the endogenous cyclin D1 promoter. Additionally, in an EGFR-

dependent tumorigenesis mouse model, the protein expression of cyclin D1 was found to

be MUC1-dependent. Therefore, to determine MUC1’s role in EGFR induced

transcriptional activation of cyclin D1, I utilized chromatin immunoprecipitation (ChIP)

directed against EGFR.

MCF10A cells were treated with MUC1-specific siRNA and subsequently

stimulated with EGF to promote endocytosis and trafficking of EGFR. The EGFR/DNA

complexes were isolated and the ATRS region of the cyclin D1 promoter was amplified

by PCR to determine if EGFR association was altered by MUC1. Additionally, I

amplified the promoter of the housekeeping gene, GAPDH, to confirm RNA Pol II

interaction and to determine the purity of the EGFR ChIP. I discovered EGFR efficiently

interacts with the ATRS region of the cyclin D1 promoter and this interaction is enhanced

in the presence of MUC1 (Figure 3.6). This ChIP analysis was performed using

extracellular EGFR (Neomarkers, Ab-13) antibody, indicating that the EGFR’s

ectodomain is also translocating to the nucleus and engaging the promoter. Additionally,

in cells that express MUC1, a ChIP performed against RNA Pol II revealed that RNA Pol

II was interacting with the cyclin D1 promoter more frequently compared to cells that 86

lacked MUC1 (Figure 3.6). With respect to the GAPDH, I found that RNA Pol II was

associated with its promoter.

Previously, I demonstrated that the loss of MUC1 expression corresponded to a

significant decrease of the interaction of EGFR with the cyclin D1 promoter. To

determine if this increased interaction with the cyclin D1 promoter translated into

increased cyclin D1 protein, I extracted protein lysates from cells treated with control or

MUC1 siRNA. The loss of MUC1 resulted in significant loss of cyclin D1 protein

expression (Figure 3.7A). In addition, the increase was EGF dependent, indicating that

although EGFR can be found in the nucleus of serum starved cells, EGF treatment is

necessary to promote cyclin D1 expression. Densitometry analysis of three separate

experiments indicated there was a significant 1.4 fold increase in the expression of cyclin

D1 (Figure 3.7B). These observations demonstrate that EGFR in the nucleus interacts

with MUC1 to promote the transcription of cyclin D1.

87

Figure 3.6 EGFR interaction with the cyclin D1 promoter is MUC1-dependent. MCF10A cells were transfected with either MUC1-1 or control (Ctrl) siRNA, serum starved, treated with 10ng/mL EGF, and incubated for 120 min. Protein/DNA complexes were collected and chromatin immunoprecipitations were performed against EGFR (Neomarker, AB-13), Mouse IgG, and RNA Pol II (Millipore, clone CTD4H8). DNA was isolated from the immunoprecipitations and used for PCR against the ATRS region of the cyclin D1 promoter and the GAPDH promoter.

Figure 3.7 MUC1 promotes Cyclin D1 Expression. A) MCF10A cells were treated with control (Ctrl) or MUC1-1 siRNA and stimulated with 20ng/mL of EGF for 120 minutes. Cytosolic protein was isolated, analyzed on an SDS-PAGE and immunoblotted for cyclin D1 (Cell Signaling, 2922), MUC1 (Neomarker, CT2), and β-actin (Sigma, AC- 15). The molecular weights (kDa) are indicated on the right. B) Relative Cyclin D1 expression levels were determined by densitometry and two-tailed student t-test was performed based on three experiments.

88

DISCUSSION

This report identifies a novel regulatory function of MUC1 on EGFR’s cellular

localization and nuclear activity. In breast cancer cells, I observed that both MUC1 and

EGFR localize to the nucleus and that MUC1 expression promotes the nuclear

accumulation of EGFR regardless of exogenous ligand. Focusing on the nuclear proteins,

I were able to efficiently immunoprecipitate EGFR/MUC1 complexes and found that the

presence of MUC1 regulated the interaction of EGFR and chromatin. After EGF

stimulation in MUC1-expressing cells, I also detected significant colocalization of

nuclear EGFR and phosphorylated RNA Pol II. Subsequent examination of chromatin-

bound EGFR with an extracellular EGFR antibody revealed that MUC1 expression

promoted the interaction of EGFR with the ATRS region of the cyclin D1 promoter. The

loss of the cyclin D1 promoter/EGFR interaction directly translated to a MUC1-

dependent decrease in the expression of cyclin D1.

Previously, MUC1 and EGFR have been reported to interact in a cancer-

dependent fashion at the plasma membrane and this interaction inhibited the lysosomal

degradation of EGFR [8, 87]. In addition, overexpression of EGFR leads to altered

trafficking of internalized receptor [56]. Altered trafficking can include the loss of

lysosomal targeting, increase recycling to the plasma membrane, and retrograde

trafficking. Retrograde trafficking can occur when transmembrane receptors are

endocytosed and returned to the endoplasmic reticulum (ER). Once in the ER, EGFR

associates with the Sec61 translocon and is released into the cytosol. EGFR then

associates with Importinβ1 via its tri-partite NLS and is imported into the nucleus [5]. In 89

the current report I demonstrate that MUC1 plays a novel regulatory role in EGFR

trafficking to the nucleus, by promoting accumulation of nuclear localized chromatin-

bound EGFR. This implies that both EGFR and MUC1 undergo retrograde trafficking

and that MUC1 promotes EGFR interaction with chromatin. Additionally, a recent study has demonstrated that MUC1 directly modulates the interaction of a transcription factor with DNA, which results in MUC1-dependent transcriptional activation [100]. Therefore it is possible that the protein-protein interaction between MUC1 and EGFR is altering

EGFR’s conformation in a way that allows EGFR to interact with DNA. In addition,

MUC1 and EGFR could be serving as transcriptional cofactors, and this will be the subject of future studies.

Interestingly, I found that the nuclear translocation of EGFR was not dependent upon the addition of exogenous ligand. Alternatively, I found that the ability of MUC1 to influence EGFR/chromatin interaction did require exogenous EGF. In addition, I observed that EGFR’s ability to colocalize with pS2-CTD-RNA Pol II and interact with the cyclin D1 promoter was both MUC1 and EGF-dependent. Although some reports have described that EGFR’s nuclear localization is dependent on EGF stimulation, others have determined that overexpression of EGFR or autocrine ligand production can drive nuclear accumulation in the absence of exogenous ligand [52, 56]. Our data suggest that both EGF-dependent and independent mechanisms may be utilized during nuclear translocation.

Recently, the importance of EGFR’s kinase activity was examined by utilizing a kinase-dead mutant of EGFR. The authors demonstrated that this kinase-dead EGFR was 90

able to inhibit autophagic cell death in human cancer cells [79]. This study demonstrated

a mechanism of EGFR oncogenic action that is independent of EGFR’s kinase activity.

It also suggests that targeting the kinase domain should not be the only focus of EGFR

therapeutics. The widely-held concept that the tyrosine activity of EGFR confers its role

in tumor formation has resulted in several therapeutic approaches that specifically target

EGFR’s kinase domain. For example, gefitinib was designed to inhibit EGFR

dimerization and the resulting receptor activation. These types of therapies were shown

to be effective in vitro and in mouse models of tumorigenesis; however the response rates

were not recapitulated in human studies [Reviewed in [21]]. Interestingly, one of the

cell lines used in this study (MDA-MB-468) is gefitinib-resistant [65]. The authors

discovered that in MDA-MB-468 cells treated with gefitinib, EGFR’s nuclear

accumulation was unchanged. I have demonstrated that in MDA-MB-468 cells, MUC1

alters EGFR’s ability to interact with chromatin and it is possible that this interaction is

promoting gefitinib resistance.

Signal transduction cascades starting at the plasma membrane and utilizing

intracellular signaling proteins such as MAPK are considered to be the main regulatory

mechanism of cyclin D1 expression. Recently, it has been proposed that the expression

of cyclin D1 can be regulated through the direct interaction of EGFR and cyclin D1’s promoter. Understanding the regulation of cyclin D1 expression is essential to clarifying its role in cancer progression. Aberrant expression of cyclin D1 has been observed in

30% of breast cancer, which in both in vivo and in vitro models of breast cancer leads to increased proliferation and genetic instability [171, 172]. Loss of cyclin D1 expression in 91

transformed human cells results in decreased tumorigenicity in nude mice [173]. In

addition, the overexpression of cyclin D1 correlates with gefitinib resistance in head and

neck squamous carcinoma cells [174]. In breast cancer, the overexpression of EGFR and

cyclin D1 is not associated with gene amplification, but postulated to be a consequence of

an unknown stimulatory mechanism [171]. Our data suggest that expression of MUC1

allows EGFR to bind the cyclin D1 promoter, driving tumor progression (Figure 3.8).

In conclusion, this report offers a mechanism of transformation as a result of aberrant trafficking and mislocalization of a receptor tyrosine kinase. This novel regulatory function of MUC1 to mediate EGFR’s role as a transcriptional cofactor sheds important light on tumor-dependent alterations in trafficking.

92

Figure 3.8 Model A) Absence of EGF – 1. MUC1’s constitutive internalization to maintain glycosylation also results in EGFR being endocytosed. 2. EGFR and MUC1 both undergo retrograde trafficking through the endoplasmic reticulum and are released into the cytosol by the Sec61 translocon. 3. Both proteins are able to interact with Importin β1 which mediates entry in the nucleus. 4. MUC1 and EGFR’s interaction in the nucleus promotes nuclear EGFR accumulation. B) EGF Stimulation in the presence of MUC1 1. Following EGF treatment, EGFR is rapidly endocytosed in a clathrin-coated vesicle 2. EGFR and MUC1 undergo retrograde trafficking 3. Both proteins are imported into the nucleus and form a complex (4). 5. EGFR colocalizes with pRNA Pol II and associates with the cyclin D1 promoter to activate transcription. 6. Expression of MUC1 promotes the recycling of EGFR back to the plasma membrane. C) EGF stimulation in the absence of MUC1 1. EGF treatment results in EGFR endocytosis 2. EGFR is either retrograde trafficked or is degraded in the lysosome (5). 3. Cytosolic EGFR is imported in the nucleus and the absence of MUC1 inhibits EGFR nuclear accumulation and EGFR is exported out of the nucleus (4).

93

IV. EGFR ACTIVITY PROMOTES TUMORIGENESIS IN MMTV-WNT-1 MOUSE MODEL OF BREAST CANCER

INTRODUCTION

Breast cancer is the 2nd most common cancer among American women and the

role of oncogenes in the transformation of normal to cancerous tissue is still being

investigated. In mice the role of oncogenes to promote mammary gland transformation

has been well studied mainly through the use of the promoter isolated from the mouse

mammary tumor virus (MMTV). Utilization of the MMTV promoter allows for the

overexpression of the protein of interest specifically in the mammary gland with limited

expression in the salivary gland tissue, male reproductive organs, and lymphoid tissue

[175]. One of the first MMTV proto-oncogenes to be inserted into the mouse genome

was int-1 or Wnt-1 [176]. MMTV-Wnt-1 mammary gland tumors develop unifocally and

will infrequently metastasize in the presence of the primary tumor. Approximately 50% of female mice (SJL strain) develop within 6 months of being born and the remaining will develop tumors by one year [177]. Wnt-1 has been found to be associated with breast cancer, but is not normally expressed in normal breast tissue [178].

In addition, the expression of other Wnt ligands (e.g. Wnt -2, 3, 4 and 7b) has been demonstrated to be expressed in normal human breast tissue [179]. Overexpression of

Wnt-1 in the mammary gland of mice promotes transformation through the overactivation of the canonical Wnt/β-catenin pathway and its resulting targets genes. 94

The canonical Wnt/β-catenin signaling pathway is mediated by the binding of a

Wnt ligand (eg. Wnt-1) to the seven-pass transmembrane frizzled (Frz) receptor and this results in the inhibition of GSK3β by the Frz activated dishevelled (Dsh) protein

[Reviewed in [180]]. Normally, the Wnt signaling pathway is important in development and is reported to induce differentiation and cell survival [Reviewed in [140]]. However, in most transformed tissues β-catenin is not degraded, because of specific mutations

within β-catenin, upregulation of Wnt ligands, mutations in the GSK3β/Axin/ APC

degradation pathway and/or deregulation of critical signaling pathways. Although these

events are not common in breast cancer, the negative regulator of Wnt signaling secreted

Frizzled-related protein 1 (sFRP1) is often lost as a result of promoter methylation [181].

Importantly, the Wnt/β-catenin signaling has been reported to be upregulated in greater than 50% of all breast tumors [Reviewed in [130]]. Sabilization and transcription factor function of β-catenin has been proposed to be the primary mechanism of Wnt- dependent transformation.

β-catenin is a 92kDa protein that has been shown to be conserved among several different organisms and interacts with E-Cadherin and α-catenin to maintain cell-cell interactions [133]. β-catenin has three highly conserved tyrosines that are critical in maintaining cell-cell adhesion function, which upon phosphorylation will lead to reduced

β-catenin/E-Cadherin association. The reduced β-catenin/E-Cadherin association mediates the buildup of β-catenin in the cytoplasm [143]. In the presence of Wnt-1 signaling the accumulation of stabilized β-catenin in the cytoplasm promotes its translocation into the nucleus. In the nucleus, β-catenin can act as a transcription factor 95

binding to the lymphoid enhancing factor/T-cell factor (LEF/TCF) and eventually to TCF

response elements in DNA. Later studies reported that the genes that were up-regulated

in response to the β-catenin/TCF complex were mostly pro-proliferation (cyclin D1), pro-

survival and pro-migration (MMPs and uPAR) [182, 183].

A prior study has examined the effect of mammary gland tumorigeneis on increased β-catenin/Wnt signaling in the presence of epidermal growth factor receptor hyperactivation. The authors discovered that overexpression of Wnt ligands (Wnt-1 and

Wnt-3) in an epidermal growth factor receptor (EGFR)-dependent mouse model of

tumorigenesis (WAP-TGFα) enhanced mammary gland tumorigenesis compared to the

transgenic WAP-TGFα alone [184]. EGFR has been demonstrated to phosphorylate β-

catenin on three highly conserved tyrosines that are critical in maintaining cell-cell

adhesion function, which subsequent phosphorylation promotes adherens junction

dissociation [Reviewed in [135]]. In breast cancer EGFR is often over-activated and

over-expressed which leads to tyrosine phosphorylation of β-catenin, cytoplasmic

accumulation, and increased transcriptional activity. Schroeder et al. demonstrated in

MMTV-Wnt-1 mice that β-catenin tyrosine phosphorylation was tumor-dependent and

treatment with an EGFR specific ligand (TGFα and AR) resulted in the tumor-specific

phosphorylation of β-catenin [185]. The authors established that EGFR and β-catenin

interacted in both MMTV-Wnt-1 and MMTV-Neu derived tumors and they found

significant colocalization in both human primary and metastatic breast cancers [185].

Another report discovered that Wnt-1 treatment of breast cancer cells resulted in the

activation of ERK and ultimately led to the EGFR-dependent increase of cyclin D1 [144]. 96

ERK activation was a result of Wnt-1 induced transactivation of EGFR, which

demonstrates the potential oncogenic signaling crosstalk between Wnts and ErbBs.

The role of EGFR activity in these studies is somewhat unclear, because it is

unknown if overactivation of EGFR and Wnt ligands are acting independent of each

other. However, examining the role of EGFR activity in mouse models is difficult

because total EGFR knockdown is embryonic lethal [186]. Therefore to clarify the role

of EGFR activity versus Wnt signaling in the transgenic mouse models I utilized the

waved-2 EGFR mutant. These waved-2 mutant mice have a single nucleotide mutation in

the kinase domain of EGFR results in the same levels of total EGFR but an 80% loss of

activity [187]. In a mouse model of colon cancer (Apcmin) Roberts et al. examined the effect of the waved-2 EGFR mutation and they discovered that the loss of EGFR activity significantly reduced the number of pre-cancerous polyps [188].

These studies demonstrate the possible cooperation between EGFR and β-catenin signaling mechanisms in the promotion of tumorigenesis. Again, to more closely examine the effects of EGFR activity on this cooperation I utilized the β-catenin- dependent mouse model of tumorigenesis (MMTV-Wnt-1) and crossed it onto the EGFR tyrosine kinase activity deficient waved-2 background. I discovered that when MMTV-

Wnt-1 mice were either hetero or homozygous for the waved-2 genotype there was a significant delay in tumor onset and slower tumor growth alteration. Additionally, reduced EGFR activity in the MMTV-Wnt-1 mouse resulted in altered mammary gland development. Interestingly, total levels and cellular localization of β-catenin in the 97

tumors were unchanged compared across the different genotypes. This study

demonstrates that EGFR activity is important in the Wnt-1-dependent transformation of

mouse mammary glands.

RESULTS

EGFR kinase activity promotes Wnt-1 dependent tumorigenesis. MMTV-Wnt-1

transgenic mice were outbred (C57Bl6/FVB) onto an EGFR kinase deficient waved-2

background. Female mice were collected and after 8 weeks of age the mice were

examined weekly until mammary gland tumors were detected. After a tumor had become

established (>50mm3) the tumor volume was measured every other day and when one

edge exceeded one centimeter the mouse was sacrificed and the tissues were collected.

The loss of EGFR kinase activity resulted in the significant delay of mammary

gland tumor onset. Half of the MMTV-Wnt-1 mice with wildtype Egfr developed

mammary gland tumors in 22 weeks, similarly to the 24 weeks first described for the

Wnt-1 transgenic model (Figure 4.1). In contrast, Wnt-1 mice with either one or two

copies of the Egfr waved-2 gene displayed a significant delay in tumor onset. Tumors

that developed from the waved-2 mice grew at a similar rate until day 10 and then the

waved-2 grew a significantly slower rate compared to Wnt-1 Egfr wildtype derived

tumors (9.65mm3/day compared 41.1mm3/day)(Figure 4.2). 98

β-catenin and E-Cadherin protein expression is EGFR kinase independent. Wnt-1-

dependent downstream signaling results in the inhibition of β-catenin degradation

following dissociation from E-Cadherin and promotes β-catenin cytosolic accumulation.

Cytosolic β-catenin is then trafficked into the nucleus where it interacts with TCF/LEF to

activate transcription. To determine any alterations in total E-Cadherin and β-catenin

levels I examined proteins isolated from both MMTV-Wnt-1 Egfr wildtype and waved-2

tumors. Immunoblotting against both E-Cadherin and β-catenin I observed that there was

not a significant difference in expression levels (Figure 4.3).

β-catenin localization in MMTV-Wnt-1 tumors is EGFR kinase independent. In

breast cancer EGFR has been shown to tyrosine phosphorylate adherens junction

associated β-catenin, which will mediate the dissociation of E-Cadherin and cytosolic

release of β-catenin. I next examined if there were any differences cytosolic and nuclear

localization of β-catenin in tumors derived from EGFR wildtype and waved-2 mice. A

portion of each tumor was fixed, embedded in paraffin, and sectioned (5μM). Tumor

sections from each genotype were used to analyze changes in β-catenin localization and I

found that most of the β-catenin was associated with the plasma membrane independent

of EGFR’s kinase activity (Figure 4.4). 99

Figure 4.1 Loss of EGFR kinase activity significantly delays Wnt-1-dependent tumorigenesis. Outbred (C57Bl6/FVB) MMTV-Wnt-1 mice were crossed on to an EGFR tyrosine kinase mutant, waved-2 (wa2), and tumor onset was examined. (MMTV- Wnt-1 Egfr+/+, n=16; MMTV-Wnt-1 Egfrwa2/+, n=14; MMTV-Wnt-1 Egfrwa2/wa2, n=15).

The image cannot be displayed. Your computer may not have enough memory to open the image, or the image may have been corrupted. Restart your computer, and then open the file again. If the red x still appears, you may have to delete the image and then insert it again.

Figure 4.2 MMTV-Wnt-1 Egfr waved-2 tumors grow significantly slower. Outbred (C57Bl6/FVB) MMTV-Wnt-1 mice were crossed on to an EGFR tyrosine kinase mutant, waved-2 (wa2), and following tumor development (>50mm3) the growth rate was examined. n ≥ 3 for all genotypes and lines indicate moving average. (MMTV-Wnt-1 Egfrwa2/wa2, 9.65mm3/day compared to MMTV-Wnt-1 Egfr+/+, 41.1mm3/day) 100

Figure 4.3 Total β-catenin proteins is independent of EGFR tyrosine kinase activity. A) Tumors from MMTV-Wnt-1 Egfr wildtype and waved-2 were collected and protein from the tissue was isolated. Protein lysates were separated on an SDS-PAGE and transferred to a PVDF membrane. Proteins were analyzed by immunoblotting against β- catenin (SC-1496-R, Santa Cruz), E-Cadherin (610182, BD), and β-actin (Sigma) B) Densitometry performed of β-catenin relative to β-actin found no significant difference between genotypes.

Figure 4.4 β-catenin localization is independent of EGFR tyrosine kinase activity. Parrafin embedded tumors from each genotype were sectioned and used for immunofluorescence against β-catenin (green) and nuclei (blue). 101

EGFR kinase activity alters mammary gland development. The role of EGFR in the

developing mouse mammary gland has been described and its function in stromal associated cells (fibroblast) has been determined to be necessary for proper gland development [189]. At the time of sacrifice a normal contralateral mammary gland used

utilized to determine any developmental abnormalities. Upon staining the ductal

epithelium of the mammary glands, I examined gross differences of the epithelial

branching between genotypes (Figure 4.5). In the MMTV-Wnt-1 mouse that lacked

EGFR kinase activity secondary and tertiary branching was less distinguished compared

to wildtype mice. Again, this demonstrates that EGFR kinase activity is important in the

normal development of the mammary gland.

Figure 4.5 EGFR kinase activity is required for normal branching morphogenesis. Mammary glands were removed, fixed, defatted, and stained with carmine. Carmine stains the epithelium (black). Branching epithelia is clearly observed in the wildtype mice (left) compared to the less distinguishable epithelia observed in the waved-2 mice (right). 102

DISCUSSION

Wnt signaling cooperates with EGFR to promote β-catenin-dependent

transcriptional activation of TCF/LEF response genes, such as cyclin D1, VEGF, myc,

uPAR, and MMPs. All of these genes play a role in breast cancer progression to a

metastatic condition. In this current study, I utilized the MMTV-Wnt-1 transgenic mouse

model of breast cancer to determine the effect of EGFR tyrosine kinase activity in β- catenin-dependent tumorigenesis. Reduction of EGFR tyrosine kinase activity in the

Wnt-1 transgenic model resulted in a significant delay in tumor onset and tumor growth

rate.

EGFR is commonly overexpressed in the poorly differentiated triple-negative

breast cancer, which is the most chemoresistance and metastatic subtype. EGFR activity

and aberrant cellular localization in this subtype of breast cancer has been described as a

poor prognostic indicator [44, 46]. The loss of EGFR kinase activity in the Wnt-1 model

resulted in a slower growing tumor, which could possibly be explained through reduced

VEGF transcription and a lack of angiogenesis. Transgenic expression of Wnt-1 could

drive tumor formation and initial growth, but lack of nutrients would ultimately slow

tumor cell proliferation. This type of growth curve was observed in the waved-2 mice,

where after early growth the tumor became more cytostatic and took longer to reach at

least one centimeter in size.

Tumorigenesis in the MMTV-Wnt-1 model is heavily dependent on the

transcriptional activity of β-catenin and previous study has reported that Wnt ligand 103

expression can modulate EGFR-dependent tumorigenesis [14]. However, in MMTV-

Wnt-1 derived tumors I observed no difference in β-catenin localization and expression

with respect to reduced EGFR activity. Although EGFR kinase activity is reduced

compensatory activity and function of the other erbB receptors (erbB2 and erbB3) could

possibly explain the minimal b-catenin differences observed in the tumors. To address

this possible issue it will be important to examine the contralateral “normal” mammary

glands to see any alterations in β-catenin localization and expression. In addition,

determine if there are any preferential erbB dimers in the wildtype versus the waved-2

tumors. However, an obvious explanation could be that the delay in tumor onset

observed could be a result of other downstream EGFR signaling effectors, such as dual

phosphorylated ERK (ERK1/2).

A recent report discovered that EGF stimulation resulted in the activation of the

serine/theronine kinase CK2a by an ERK-2 dependent mechanism [145]. Following CK2

activation, it can serine phosphorylate α-catenin to promote adherens junctions disassembly and β-catenin stabilization. These events promoted the β-catenin dependent

transcriptional activation, increased cellular invasion and EMT. This reiterates the role of

EGFR activity and β-catenin stabilization but importantly provides a possible mechanism

of the delay of tumor onset observed in the EGFR activity deficient MMTV-Wnt-1 mice.

Although differences in β-catenin localization were not observed in the tumors between

genotypes examination of its localization in the contralateral normal mammary gland

could provide insight into possible early events driving tumorigenesis. 104

Although it is important to note that the mechanism driving the different tumor

onset and tumor growth rate is still unknown, the study does reveal that in an in vivo

model that EGFR tyrosine kinase activity and Wnt-1 signaling cooperate to drive tumorigenesis. These findings indicate that in triple negative breast cancer where EGFR is highly expressed the cotreatment of a Wnt and EGFR inhibitor could have additive or synergistic anti-cancer effects.

105

V. INTRACELLULAR MUC1 PEPTIDES INHIBIT CANCER

PROGRESSION

Note: The experiments described in this chapter have been published in [123].

Additionally, the experiment and resulting figure shown as Figure 5.2a was performed by

Dr. Ina Menzl. Xenograft and genetically-driven mouse studies were performed by

Experimental Mouse Shared Service (Tucson, AZ).

INTRODUCTION

MUC1 (DF3, CD227, episialin, PEM) is a heavily O-glycosylated heterodimeric

protein of >300 kDa, normally expressed abundantly on the apical surface of glandular

epithelia. In greater than 90% of human breast carcinomas and metastases, apical

localization is lost and MUC1 is overexpressed (by greater than 10 fold) and

underglycosylated [95, 190]. Deregulated expression of MUC1 is found in many other

types of adenocarcinomas as well, including cancers of the lung, pancreas, ovary and

prostate, in addition to being highly expressed in leukemias, myelomas and lymphomas

[191-193]. Studies in both genetic mouse models and cell line models have demonstrated

that MUC1 is an oncogene. A transgenic mouse model driving MUC1 (human)

overexpression to the mouse mammary gland (MMTV-MUC1) results in the

development of breast cancer and is accompanied by a failure of the mammary gland to

undergo complete postlactational regression via apoptosis [98]. Transfection of MUC1

into 3Y1 fibroblasts induces their transformation, and transfection of MUC1 into colon 106

cancer cells demonstrates that MUC1 overexpression inhibits drug-induced apoptosis

[120].

The cytoplasmic domain of MUC1 contains sites for multiple protein interactions, although these interactions go largely unformed in the polarized epithelium of the normal

breast, as the binding partners of MUC1 are typically found on the basolateral membrane

(Reviewed in [103] and [8, 87]. During cancer progression, when there is a loss of

cellular polarization, MUC1 is overexpressed and functionally interacts with src, GSK3β,

epidermal growth factor receptor (EGFR) and β-catenin, among others [87, 124, 126].

The sites for interaction between MUC1 and these proteins have been mapped to distinct

domains within the 72-amino acid cytoplasmic tail of MUC1 [126, 194, 195]. Both

EGFR and src can phosphorylate MUC1 on a YEKV motif, and this phosphorylation

results in increased binding of MUC1 to β-catenin through an SAGNGGSSLS domain

[126]. Recent evidence demonstrates that the interaction between MUC1 and EGFR can

significantly modulate EGFR biology and effect EGFR-dependent transformation [8, 87].

It has been established in both human breast cancer cell lines and transgenic mice

overexpressing MUC1 (MMTV-MUC1) that MUC1 and EGFR biochemically interact,

resulting in the potentiation of EGF-dependent p42/44 ERK activation during lactation

[110, 126]. Recently, our laboratory has demonstrated that MUC1 expression inhibits the

ligand-mediated ubiquitination and degradation of EGFR while enhancing its

internalization and recycling [87]. To evaluate the role of Muc1 in transformation,

Pochampalli et al generated WAP-TGFα mice on a Muc1 null background which 107

revealed that Muc1 expression has a dominant effect on TGFα-dependent transformation

of the breast, promoting both onset and progression [8].

While it is now established that EGFR is potently regulated by MUC1 expression,

transgenic mouse models have also implicated the cell adhesion protein, β-catenin, in

EGFR and MUC1 signaling. In a study of the WAP-TGFα transgenic model, Wnt1 and

Wnt3 were found to be selectively activated in the most aggressive breast tumors [184].

The Wnts are secreted glycoproteins that bind the transmembrane frizzled receptor,

resulting in a signaling cascade that inactivates the mechanism for β-catenin degradation

and results in transformation [183, 196-200]. Additionally, in MMTV-Wnt1 transgenic

mice, EGFR was found to interact with and phosphorylate β-catenin in a tumor-specific

manner [185]. These studies demonstrate that β-catenin and EGFR can affect their respective pathways to promote transformation. Finally, MUC1 is also implicated in β- catenin dependent transformation, indicating that these three proteins have the ability to cooperatively promote cancer progression. In MMTV-Wnt-1 transgenic mouse models crossed onto a Muc1-null background, loss of Muc1 corresponds to a significant reduction in tumor progression [124]. In a subsequent study, interactions between MUC1 and β-catenin were found to be highly increased in samples from human metastatic breast tumors, indicating that these interactions are clinically relevant [124].

Together, these studies demonstrate the strong potential for MUC1, EGFR and β-catenin to affect each other during transformation, including their striking co-upregulation during transformation and metastasis. MUC1 can inhibit the downregulation of EGFR and 108

promote the transforming ability of both EGFR and β-catenin. Additionally, genetically

derived mouse models implicate MUC1 in both EGFR- and β-catenin-dependent

transformation and metastasis. Interestingly, the interaction sites on MUC1 for both

EGFR and β-catenin lie in tandem on the MUC1 cytoplasmic domain. This study

demonstrates that targeting the interaction domain of MUC1 for both EGFR and β-

catenin through the utilization of MUC1 dominant negative peptides can significantly

affect breast cancer progression. Importantly, this dominant negative peptide can

significantly inhibit tumor progression in a genetically-driven mouse model of breast

cancer over a relatively short treatment time and may have important clinical

applications.

RESULTS

PMIP efficiently enters the cells, reduces EGFR protein levels and inhibits MUC1

colocalization with β-catenin.

The MUC1 cytoplasmic domain is composed of 72 amino acids, within which lies

a 15 amino acid domain containing sites of EGFR phosphorylation (human=YEKV and

mouse=YEEV) and β-catenin binding (human=SAGNGGSSLS and

mouse=SAGNGSSSLS,Figure 5.1a) [126, 201]. A 15 amino acid peptide was

synthesized to determine if it could act in a dominant negative fashion to block

interactions between endogenous MUC1 and EGFR/β-catenin. To allow this peptide to

gain entrance to the cell, a protein transduction domain was synthesized in tandem

[PTD4, Figure 5.1a [202], Reviewed in [203]]. To demonstrate that PMIP was entering 109

the cells, a biotin-labeled hPMIP (10μM) peptide (biotin-hPMIP) was pulsed onto cells in vitro. We found that the peptide was taken up and is retained in BT20 breast cancer cells

(Figure 5.1b).

To determine if the hPMIP peptide was able to block the interaction between

EGFR and MUC1, BT20 cells were treated overnight with either 10 μM hPMIP, 10 μM

control peptide (CP) or peptide vehicle (V), immunoprecipitated EGFR and

immunoblotted for MUC1 (Figure 5.2a). Coimmunoprecipitation between MUC1 and

EGFR in control treated cells was detected; however interaction was significantly

reduced in hPMIP treated cells (Figure 5.2a, top panel). In addition to protein interaction,

Pochampalli et al previously demonstrated that MUC1 expression inhibits the EGF- dependent degradation of EGFR [87]. To determine if PMIP was able to block the

MUC1-mediated effects on EGFR degradation, BT20 and MDA-MB-231 cells were treated with hPMIP or PTD4 control, and induced EGFR endocytosis by treatment with

EGF for 30 minutes. Examination of EGFR protein levels by immunoblotting demonstrated that hPMIP treatment resulted in a reduction of EGFR levels following

EGF treatment (Figure 5.2b and data not shown). Note that in the absence of EGF treatment, hPMIP treatment does not affect EGFR levels.

To determine if PMIP could block the interactions between MUC1 and β-catenin that occur during transformation, the effects of hPMIP treatment on protein colocalization in the BT-20 breast cancer cell line was evaluated [124, 185]. Cells were serum starved and treated with either 10 μM hPMIP, 10 μM control peptide (CP), 10 μM PTD4, or 110

vehicle control (V) overnight, fixed and immunofluorescence was performed. While β-

catenin and MUC1 colocalization at the cell cortex could be identified in control

conditions (Figure 5.2 c-f, arrows), their interactions were significantly inhibited by

hPMIP treatment (Figure 2c).

These results demonstrate that PMIP treatment inhibits both MUC1/β-catenin and

MUC1/EGFR interactions. In addition, PMIP inhibits MUC1-mediated inhibition of

EGF-dependent degradation of EGFR.

111

Figure 5.1 a) MUC1 peptide (PMIP) can efficiently enter cells. a) MUC1’s cytoplasmic amino acid sequence (MUC1CT), which includes an EGFR phosphorylation site (underline) and β-catenin binding site (double underline). Human and mouse sequence for the PMIP peptide (human PMIP, hPMIP and mouse PMIP, msPMIP), the PTD4 protein transduction domain and the control peptide (CP) containing the last 12 amino acids of the human MUC1 cytoplasmic domain are shown. b) BT-20 cells were treated with 10μM Biotin-hPMIP for 4hrs (Green=Biotin-PMIP and Blue=DAPI (nucleus), 400x and 630x)

112

Figure 5.2 PMIP blocks MUC1/β-catenin and MUC1/EGFR interactions. a) BT-20 cells were treated overnight (-serum) with either 10μM hPMIP, 10μM control peptide (CP) or vehicle control (water; V), and protein lysates generated. Lysates (500μg) were immunoprecipitated (IP) with either anti-EGFR (Ab-13) (lanes 1-6, top and middle panel) or IgG (lane 7, top and middle panel) and immunoblotted (IB) with anti-MUC1 (top panel; CT2) or anti-EGFR (middle panel; 1005). Total levels of MUC1 (35 μg) are shown in the bottom panel and in right column of top panel (straight lysate, SL). White lines through blots indicate same gel and exposure but were non-contiguous. b) BT-20 cells were treated for 18hrs (-S) with hPMIP (10μM) or PTD4 (10μM). The cells were treated with EGF (30’ EGF, 10ng/ml) to induce endocytosis or left serum free (-S) and protein lysates were generated. The lysates were immunoblotted (IB) for EGFR (1005) and β-actin (AC-15) c-f) BT-20 cells were treated (-serum) overnight with (c) hPMIP (10μM), (d) control peptide (10μM), (e) PTD4 (10μM) or (f) water (V) and pulsed with the same peptides for an additional 30’ prior to fixation. The cells were probed for MUC1 (primary; H295: red) and β-catenin (primary; C-14: green). Co-localization (arrows) is designated with white pixels. Magnification=400x. 113

PMIP inhibits cellular invasion and proliferation in vitro

As hPMIP treatment demonstrated a strong biochemical inhibition of MUC1 interactions with both β-catenin and EGFR, potential effects of PMIP treatment on cell

growth and invasion were evaluated. To determine the effects of PMIP treatment on cell

proliferation, BT20 cells were treated with either 10 μM hPMIP, 10 μM control peptide

(CP), 10 μM PTD4 or peptide vehicle (V) for 7 days continually, then the number of cells was evaluated by quantifying the differences in formazan conversion through a colormetric MTT assay [204]. Following seven days of hPMIP treatment the growth of

BT20 cells was significantly inhibited compared to the control peptide (CP), PTD4 and water (V) (*, p<0.001, **, p<0.0002, ***, p<0.002,Figure 5.3a).

The effect of PMIP on cellular invasion was next evaluated. BT20 or MDA-MB-

231 breast cancer cells were treated overnight with either hPMIP or PTD4 control peptide

(10μM), then induced to invade through an 8.0μM filter into Type I collagen using 20% fetal bovine serum (FBS) as a chemoattractant. Cells that were able to migrate through the filter and invade into the gels were fluorescently labeled and quantified. PMIP treatment significantly inhibited the ability of cells to invade compared to cells treated with controls in both BT-20 cells and MDA-MB-231 (#p<0.0001 and data not

shown,Figure 5.3b).

114

Figure 5.3 PMIP inhibits cell proliferation and invasion of breast cancer cells in vitro. a) BT20 cells were cultured in a 96-well dish (5 x 103 cells/well) and treated daily for 7 days with either hPMIP (10μM), CP (10μM), PTD4 (10μM) or water (V) in EMEM 10% FBS. An MTT assay was performed to quantify cell number at the start of treatment (Day 0, DO) and after treatment was complete (Day 7, D7) (*, p<0.001, **, p<0.0002, ***, p<0.002, ANOVA). Error bars represent standard error. b) BT-20 cell lines were treated with either hPMIP (10μM), CP (10μM), PTD4 (10μM) or water (V) overnight, labeled with Calcein AM, allowed to invade through a transwell (8.0μM) insert into a Type I collagen gel, and the invaded cells were fluorescently measured. (#p<0.0001, ANOVA). The data from the invasion and proliferation assays represents four independent experiments with at least seven replicates per experiment. Error bars represent standard deviation.

115

PMIP inhibits tumor growth and recurrence in a xenograft breast cancer model

As hPMIP strongly inhibited cellular growth and invasion in vitro, the ability of

hPMIP to suppress tumor growth and metastasis in vivo was evaluated. In these

experiments, highly metastatic MDA-MB-231 breast cancer cells were implanted into the

mammary fat pad of severe combined immune deficiency (scid) mice and tumor growth

was evaluated.

To determine potential effects of hPMIP on primary tumor growth, scid mice

bearing MDA-MB-231 tumors (100mm3) were treated for 21 days with either hPMIP or

PTD4 control peptide (i.p. injections) (Figure 5.4a-c). Upon removal of drug, tumors were allowed to continue to grow until they reached a volume of 1000mm3, then surgical

resection of primary tumors was performed (Figure 5.4b). Treatment with hPMIP

resulted in a significant decrease in tumor size compared to control treated animals at 21

days when drug administration was stopped (*p=0.028, Figure 5.4a). Additionally, this

corresponded to a significant increase in the length of time required for hPMIP treated

mice to reach resection size of 1000mm3 (**p=0.03,Figure 5.4b). Although treatment

ended approximately 20 days prior to resection, hPMIP treatment substantially decreased the amount of tumor regrowth and spread 10 days after resection (Figure 5.4c). Also, a

decrease in the size of metastatic tumors in the hPMIP-treated animals was observed

compared to control, and next designed an experiment that would allow us to determine if

hPMIP was affecting tumor spread following tumor resection. 116

To evaluate the effects of PMIP on tumor spread (Figure 5.4a and b), MDA-MB-

231 breast cancer cells were allowed to establish a large tumor mass (500mm3) and mice

were injected (i.p.) for 21 days with hPMIP or control peptide (PTD4) (Figure 4.4d).

Immediately after the end of treatment, primary breast tumors were resected, and animals

were followed to examine rates of tumor regrowth and/or metastasis to secondary

mammary glands. While the presence of secondary mammary gland tumors were found

in equal number for both treatment groups, the tumor volume for the control treated

animals averaged 760mm3 while the PMIP treated averaged only 73mm3 (Figure 5.4e).

Note that mice were not treated with drug during the 10 days in which regrowth was

followed. These xenograft models revealed that PMIP treatment could have long lasting

anti-growth effect, which was observed in tumor regrowth following resection.

117

Figure 5.4 PMIP significantly inhibits tumor growth and recurrence in vivo. a) MDA-MB-231 cells in Matrigel were injected into the mammary fat pad of scid mice, and daily peptide treatment (50μg/g body weight of hPMIP or PTD4) began when tumors reached 100mm3 (hPMIP and PTD4 n=8 mice) and primary tumor growth was assessed 2 (*, p=0.028, ANOVA). Tumor burden represents the average volume (v= a x b/2) of all tumors at the stage indicated. b) After the end of treatment, the amount of time the tumors took to progress to 1000mm3 was measured (**, p=0.03, ANOVA). c) After resection, mice were observed for tumor regrowth at the primary site or secondary mammary glands (hPMIP n=7 mice and PTD4 n=8 mice). d) MDA-MB-231 cells in Matrigel were injected into the mammary fat pad of scid mice, and daily peptide treatment (50μg/g of hPMIP or PTD4) began when tumors reached 500mm3 (hPMIP n=6 mice and PTD4 n=4 mice) and primary tumor growth was assessed. e) Following 21 days of treatment the tumors were resected immediately and tumor regrowth at the primary site and spread to secondary mammary glands was monitored (hPMIP n=4 mice and PTD4 n=4 mice). Error bars represent standard error.

118

PMIP inhibits tumor growth and induces regression in genetically-driven breast

cancer

While the xenograft model demonstrated the effect of hPMIP treatment on growth

and progression of established cell lines, we wanted to determine how msPMIP would

affect tumor initiation and progression in a mouse model which better recapitulates

human breast cancer. The MMTV-pyV mT transgenic mouse model of breast cancer

strongly resembles human breast cancer by activating multiple signaling pathways,

including AKT, src and shc [67, 205]. Studies have demonstrated that the resulting breast

cancer pathologically and molecularly mimics the full progression of hyperplasia, ductal carcinoma in situ and adenocarcinomas observed in human disease [206, 207]. To first determine if peptide could be delivered to the mammary glands and tumors of these animals, FITC-labeled msPMIP was injected and analyzed peptide retention (Figure

5.5a). At two hours post-injection, FITC was detected throughout the animal’s body cavity, including all organs (data not shown). After four hours, FITC-msPMIP was found to be retained selectively in the mammary gland, mammary gland tumors and in the colon and skin (Figure 5.5a and data not shown).

To determine the effects of msPMIP on genetically-driven breast cancer progression, MMTV-pyV mT mice bearing at least one mammary gland tumor of >0.5cm

in diameter (in MMTV-pyV mT mice there are ten potential tumor sites) were treated for

21 days with either msPMIP or PTD4 control peptide. Treatment had a dramatic effect

on tumor growth, as msPMIP significantly slowed the total tumor growth from ~590% to 119

~194% over the 21 days of treatment (*p=0.039;Figure 5.5b). Additionally, msPMIP

treatment significantly decreased the tumor growth rate compared to control (PTD4)

treated tumors (**p=0.007;Figure 5.5c). Note that treatment of MMTV-pyV mT mice with hPMIP (as opposed to msPMIP) had no effect on tumor growth, emphasizing the amino acid specificity of PMIP.

Overall size of tumors that arose throughout the study were next analyzed. This analysis demonstrates that while 13% of the tumors in the control (PTD4) group grew larger than 500mm3 by the end of the study, only 1% of the tumors in the msPMIP treated

group reached that size (Figure 5.5d). As this transgenic model had continual expression of the polyoma middle T transgene driving tumorigenesis throughout the study, the

effects of drug treatment on the formation of new tumors were examined. Although both

msPMIP and control (PTD4) groups had a similar number of tumors sized 100-300mm3 at the beginning of treatment, this number nearly doubled by the end of treatment in the control group, but remained the similar in the msPMIP group (Figure 5.5d). These data indicated that msPMIP treatment inhibits tumor initiation in this model (Initiation equals percent of tumor transitions from 0mm3 to 100mm3). To analyze tumor initiation further,

the percent of tumors that were initiated during drug treatment were determined. This

analysis demonstrates that in the msPMIP group there was a significant decrease (12 vs.

20 initiated tumors, data not shown, p=0.0045) of tumor initiation during the study.

Although highly significant decreases in tumor formation and growth were

observed from treatment of tumor-bearing MMTV-pyV mT mice with msPMIP, not all 120

tumors in the study responded to treatment (Figure 5.6a). Analysis of each mammary

gland demonstrates that while most tumor growth rates slowed substantially in response

to msPMIP (white bars), a number of tumors continued to grow, indicative of the stochastic pathway activation in this model. Importantly, a subset of established tumors treated with msPMIP (four tumors) regressed completely under treatment, although none of the control treated tumors did so (Figure 5.6a, *). In addition, there was no detectable toxicity (no weight loss, organ failure or disorientation) following treatment with msPMIP or PTD4. Further investigation of tumor tissue sections by cleaved caspase-3 immuno-histochemistry and hemotoxylin-eosin staining of tumors that regressed or responded to msPMIP treatment did not reveal a difference in apoptotic cells compared to

PTD4 control peptide treated tumors (Figure 5.6b). 121

Figure 5.5 PMIP significantly slows progression of MMTV-pyV mT induced mammary gland tumors. a) MMTV-pyV mT transgenic mice were injected with FITC-msPMIP (50μg/g body weight), sacrificed four hours later and various tissues visualized using fluorescence microscopy. FITC-msPMIP localization in the mouse mammary gland tumors is shown (2.5x and 8x). b) Mammary gland tumors (>0.5 cm in diameter) were allowed to develop and mice were injected daily (50μg/g body weight, 21 day treatment, i.p. injection, 1X per day) with either msPMIP (7 mice) or PTD4 (6 mice). At the end of treatment, animals were sacrificed and proteins lysates made of the tumors for later analysis. In the course of treatment, total tumor growth for all tumor sites (msPMIP n=70, PTD4 n=60) was significantly lower in the msPMIP treated mice than in the PTD4 mice (193.8% ± 77.7% vs. 589.5% ± 283.6%, *, p=0.039, ANOVA). c) Mammary gland tumors of msPMIP treated mice grew at a significantly slower rate than PTD4 treated tumors (p=0.0076, ANOVA). d) Tumor size distribution for the msPMIP or PTD4 treated transgenic mice revealed that 47% (28 out of 60 possible tumor sites) of the PTD4 treated tumors were larger than 100mm3 compared to 27% (19 out of 70 possible tumor sites) of the msPMIP treated tumors. Numbers above data are numbers of tumors that meet the size criteria over the total potential tumor sites. 122

Figure 5.6 MMTV-pyV mT tumors have differential response to msPMIP. a) Individual growth of each tumor site (each bar is an individual tumor site) from animals described in figure 3 were treated daily with msPMIP (open bars) or PTD4 (grey bars) at 50μg/g body weight for 21 days. Tumor progression was observed every three days during the 21 day treatment (PTD4, n=60 tumor sites and PMIP, n=70 tumor sites). In four instances (*), msPMIP treated tumors completely regressed, however none of the control (PTD4) treated tumors regressed. Individual tumor sites are grouped by mammary fat pads (1-10) to allow for the comparison of similar anatomical sites. MFP=mammary fat pad b) Tumors from msPMIP (1, left 2 panels) and PTD4 (2, right 2 panels) treated mice were sectioned (3μm) and subsequently used for hemotoxylin-eosin staining and cleaved caspase-3 immunohistochemistry (200x). 123

msPMIP treatment results in a reduction of EGFR and Muc1 levels

To determine if msPMIP was affecting EGF-dependent degradation of EGFR in

vivo, protein lysates from tumors of MMTV- pyV mT animals injected with EGF and

peptide 30 minutes prior to animal sacrifice (after a standard 21 day drug treatment). A

striking reduction in the expression of EGFR and corresponding phosphotyrosine in the

msPMIP treated mouse compared to the control treated animal (Figure 5.7a). Note that

in this mouse, there were no frank tumors remaining after the 21 day msPMIP treatment

(remaining mammary fat pads were analyzed), while we obtained six tumors of greater

than 400mm3 from control treated animals. Analysis of EGFR expression and activation of the MDA-MB-231 xenograft tumors was restricted to tumors that were not treated with EGF. Nonetheless, a slight reduction in the levels of phospho-EGFR (Y1173) in hPMIP treated compared to control treated tumors was observed (Figure 5.8, top panel).

Tyrosine 1173 phosphorylation can result in the recruitment of shc to EGFR, resulting in activation of the MAP Kinase ERK. By examination of activated ERK, found that hPMIP treatment resulted in a strong downregulation of ERK activity compared to controls (Figure 5.8).

To determine if msPMIP blocked interaction between Muc1 and β-catenin, we began by establishing levels of Muc1 protein expression in the tumor lysates.

Interestingly, msPMIP treatment induced a loss of Muc1 protein in both the MMTV- pyV mT model (Figure 5.7a and b) and in the MDA-MB-231 xenograft model (Figure 5.7c and d). Total levels of β-catenin and its target genes cyclin D1 and Twist are unchanged

between PMIP and control (Data not shown). 124

Figure 5.7 PMIP is associated with reduced Muc1 expression. a) A representative MMTV- pyV mT msPMIP (P) treated mouse and a PTD4 mouse (C) were each injected 30 minutes prior to sacrifice with epidermal growth factor (1μg/g body weight) and with peptide. Following sacrifice the tumors were collected and protein lysates were generated. Protein (50μg) was separated by SDS-PAGE, transferred and immunoblotted for expression of phosphotyrosine (PY99), EGFR (1005), Muc1 (CT2), and β-actin (AC- 15). b) Lysates from MDA-MD-231 xenograft tumors (not treated with EGF; described in Figure 2) were similarly analyzed to determine levels of EGFR and MUC1 protein expression. Relative protein levels of Muc1 were measured by densitometry and graphed (Muc1/β-actin, *, p=0.014, ANOVA). Molecular weights are shown on the right. IB = immunoblot.

125

Figure 5.8 hPMIP treatment results in reduced levels of phosphorylated EGFR and dpERK. Protein lysates were prepared from the tumors described in Figure 5.4d. Protein (50μg) was separated by SDS-PAGE, transferred and immunoblotted for expression of 1173pEGFR (1173), EGFR (1005), dpERK (M-8159), ERK (9102), and β- actin (AC-15). White lines through blots indicate same gel and exposure but were non- contiguous.

PMIP Downregulates Met Expression

To better understand any possible alterations in downstream signaling and

transcriptional activation a whole genome cDNA microarray was utilized to determine

changes in transcripts following PMIP treatment. After extracting total RNA from BT20

cells treated with PMIP (10μM), PTD4 (10μM) or vehicle a cDNA reaction was

performed with fluorescently labeled nucleotides. The cDNA was then hybridized to a

Codelink Whole Genome microarray and the intensities were normalized with internal

controls. Several genes were differentially regulated after PMIP treatment compared to

controls (Table 5.1). Of interest, RT-PCR confirmed that the oncogenic hepatocyte

growth factor receptor (c-Met) c-Met was significantly downregulated greater than 3 fold

following PMIP treatment (Figure 5.9). 126

Table 5.1 Genes that are differentially regulated following PMIP treatment. BT20 cells were treated with PMIP (10μM) for 18hours and total RNA was extracted. RNA was processed into cDNA and was hybridized onto a Codelink Whole Genome microarray. These arrays were analyzed using Genespring. Genes downregulated (a) and upregulated (b) are listed in the table, with function and fold change compared to PTD4 and vehicle controls. 127

Figure 5.9 MET is significantly downregulated in response to PMIP. a) Microarray intensities from BT20 breast cancer cells treated with PMIP (10μM), PTD4 (10μM), or vehicle. b) RNA from PMIP, PTD4, and vehicle treated cells was extracted and used for RT-PCR against MET. RT-PCR was performed by Derrick Broka.

128

Discussion A MUC1 mimetic peptide (MIP) linked to a protein transduction domain (PTD4)

can freely enter transformed cells and inhibit their invasion in vitro. This same peptide

can inhibit primary tumor growth, tumor spread, and recurrence of tumors after resection

in an orthotopically implanted breast cancer model. PMIP can survive in circulation with

tissue specific retention and no detectable toxicity. Importantly, PMIP can

significantly inhibit tumor growth in a genetically-driven mouse model that mimics

human breast cancer. Mechanistically, this tumor-inhibitory effect is closely associated

with a decrease in EGFR and MUC1 expression. Together these data demonstrate that

this peptide-based intracellular drug shows strong efficacy as a non-toxic treatment for

breast cancer.

The over-expression of human MUC1 in mouse mammary glands promotes

transformation and the loss of MUC1 in several transgenic models can significantly delay

tumor onset [8, 98, 124]. This may be due to the number of oncogenic partners MUC1

has been demonstrated to interact with, namely β-catenin, src, and EGFR [Reviewed in

[103]]. This mimetic peptide is designed to block interactions between MUC1, β-catenin

and EGFR, and the study demonstrated that PMIP treatment does block MUC1

interactions with these proteins. In addition, a loss of MUC1 expression in response to

PMIP treatment under certain conditions, which may be the result of downregulation of

EGFR. While the mechanism of MUC1 loss is unknown, it would certainly result in a

loss of Muc1-dependent oncogenic signaling. It is tempting to speculate that in the

presence of PMIP, MUC1 and EGFR are alternatively trafficked and enter the lysosomal 129

degradation pathway, leading to their enhanced degradation. PMIP treatment of MDA-

MB-231 cells overexpressing MUC1 under a CMV promoter for only 3 hours in culture

induces a loss of MUC1 overexpression, indicating that the effect is not transcriptionally

regulated (data not shown). Loss of MUC1 in another study demonstrated a significant

decrease in activated ERK, which is a similar observation in PMIP treated tumors [99].

Importantly, PMIP treatment resulted in significant downregulation of the growth factor

receptor, MET. In non-small EGFR and Met can act synergistically to

promote tumorigenesis, proliferation and invasion [208]. Therefore a loss of Met and

EGFR could be another mechanism of PMIP’s anti-tumor effect. However, future studies

will focus on determining the precise mechanism by which PMIP suppresses protein

expression and tumor progression.

One important observation is the lack of toxicity associated with PMIP treatment

(no weight loss, signs of distress or organ failure). While this is not an unexpected result with the use of an endogenous peptide, it points to a potentially low level of toxicity in patients. We have also examined the possibility that PMIP has activated an immune response in the immune-intact MMTV- pyV mT transgenic animals. Examining leukocytic infiltrates using protein levels of CD45 as a marker, there was increase in those tumors treated with PMIP versus control ([209]; data not shown). Additionally, studies by groups investigating the potential adjuvant activity of the protein transduction domain have found that the TAT protein transduction domain is not immunogenic [210].

One potential reason for PMIP’s lack of toxicity may be due to the relatively fast clearance of small peptides from the body [211]. Importantly, while PMIP-FITC 130

appeared to be cleared overall from the body 4 hours after injection, it was retained in

specific organ sites, including the mammary gland and colon. This site specific retention

may be due to an increase in peptide binding partners in these particular tissues.

Previously Pochampalli et al demonstrated that MUC1 inhibits the ligand-

dependent degradation of EGFR, resulting in enhanced receptor stability [87].

Furthermore, Pochampalli et al have demonstrated that this interaction promotes the

oncogenic properties of EGFR [8]. Neither of the mouse models used in these

experiments were previously shown to be dependent upon EGFR for progression, and

yet, PMIP is significantly effective in each model. This indicates that PMIP may have

broad applications against tumors overexpressing MUC1, which encompasses most

epithelial neoplasias [212]. In addition, PMIP may serve as an important adjuvant

therapy with anti-EGFR treatments [Reviewed in [213]]. Our data indicate that MUC1

induces the internalization, altered trafficking and enhanced signaling of EGFR [87].

Therefore, if PMIP blocks these interactions, anti-EGFR therapy that relies on surface-

localization of EGFR could by enhanced by PMIP co-delivery.

This study demonstrates the efficacy of PTD4-linked peptide-based drugs and the

value of MUC1-directed targets in breast cancer. Importantly, these data indicate that

PMIP is a potent drug that is active at all stages of tumor progression; inhibiting growth,

inducing regression and inhibiting metastatic spread.

131

VI. ANTI-CANCER THERAPIES THAT UTILIZE CELL PENETRATING PEPTIDES

Note: The review presented in this chapter has been published in [214].

INTRODUCTION

Significant progress has been made in the identification of tumor specific cellular pathways which can be successfully blocked in vitro. Unfortunately, the safe, systemic delivery of these blocking agents to patients has been difficult and sometimes impossible due to difficulty with tissue penetration, toxicity and functional activity in vivo. The recent development of cell penetrating peptides (CPPs) has overcome these barriers, leading to the development of novel tumor-specific molecular therapeutics. Also known as protein transduction domains (PTD), CPPs consist of short amino acid sequences that are able to mediate the translocation of a conjugated cargo across the plasma membrane.

Once inside the cell, CPP-associated cargos are then able to interact with their intracellular targets, and block their tumorigenic properties. Importantly, the non-toxic mechanism of cell penetration allows for the safe and effective systemic delivery of cancer therapeutics.

Over the last decade, several laboratories have utilized naturally occurring and synthetic CPPs to deliver several types of therapeutic cargo including proteins, antibodies, peptides, and nucleic acids directly into the cell’s cytoplasm [123, 155-157].

CPPs are easily manipulated and optimized, and can be used to target cargo to a specific 132

cell-type or tissue [215]. For example, one sub-group of CPPs can cross the blood-brain

barrier, allowing for the development of therapies that target brain cancers which are

normally difficult to treat with conventional chemotherapies [154, 216]. Importantly,

CPP bound therapies offer a novel approach to treating cancer in a specific and non-toxic

manner, as damage to the cell membrane, receptors or transporters are not required for

entry into the cell. This chapter will discuss naturally occurring and optimized protein

transduction domains and their utilization in the delivery of anti-cancer therapies into

intracellular compartments of diseased cells. Additionally, this chapter will focus on

studies with pending or recently awarded patents whose functionality has been verified

and published, and discuss the very promising future of these therapeutics for the

treatment of cancer.

Cell Penetrating Peptides

CPPs were first discovered through the study of model organisms such as

Drosophila melanogaster (Antennapedia, Antp) and the Human Immunodeficiency Virus

(HIV Tat) retrovirus, and there are now two fundamental types of CPPs that have been

utilized for intracellular delivery of therapeutic molecules [153, 154]. The first and most

common are the cationic CPPs, which are short amino acid sequences that are mainly

composed of arginine, lysine, and histidine. These amino acids are responsible for the

cationic charge of the peptide that will mediate the interaction of the peptide with anionic

motifs on the plasma membrane. Both naturally occurring and synthetic cationic CPPs 133

have been described and are shown in Table 1. A second class of CPPs is the

amphipathic peptides, which have lipophilic and hydrophilic tails that are responsible for

mediating the peptide translocation across the plasma membrane Table 1. Although these

classes of CPPs are composed of different amino acids, the mechanism of intracellular

delivery is similar.

134

Type of Name Amino Acid Sequence CPP Ref

1 HIV Tat (47-58) YGRKKRRQRRR Cationic [154] 1a PTD-3 YARKARRQARR Cationic [202] 1b PTD-4 YARAAARQARA Cationic [202] 1c PTD-5 YARAARRAARR Cationic [202] 1d PTD-6 YARAARRAARA Cationic [202] 1e P3 YGRKKRRQR Cationic [217]

1f Polyarginine Rn Cationic [218]

Antennapedia 2 (Antp) RQIKIWFQNRRMKWKK Cationic [153]

DAATATRGRSAASRPTERPRAPAR- VP-22 3 SASRPRRPVD Cationic [219]

4 PTD-SN1 RKMLKSTRRQRR Cationic [220]

5 Bac RRIRPRPPRLPRPRPRLPFPRPG Cationic [221]

6 Transportan GWTLNSAGYLLGKINLKALAALAKKIL Amphipathic [222]

7 Pep-1 KETWWETWWTEWSQPKKKRKV Amphipathic [223]

Table 6.1. Commonly Utilized Cationic and Amphipathic Cell Penetrating Peptides. More specifically, this chapter will discuss recent patents that utilize CPPs 1, 2, 4 and 6. However, the remaining CPPs are also relevant to the field of peptide-based therapies. 135

Mechanism of Cell Penetrating Peptides The detailed mechanism of action by which CPPs transverse the plasma

membrane and interact with cytosolic proteins was recently reviewed, and readers are

encouraged to consult those publications for the details summarized here [224, 225].

Briefly, CPPs are composed of several cationic amino acids such as lysine or arginine,

and a group of these amino acids result in a highly positively charged peptide sequence.

These positively charged sequences are able to interact with acidic motifs on the plasma

membrane in a receptor-independent fashion. Acidic motifs can be several different

substrates found on the plasma membrane including proteoglycans and glycolipids [226,

227]. A recent patent has introduced the use of synthetic CPP, PTD-SN1, to improve the

efficiency of this interaction and ultimately the peptide’s cytosolic translocation [220].

Following interaction with these substrates, the peptides undergo cell-type independent

internalization [228]. The exact mechanism of this internalization process is yet to be

elucidated but it is dependent on several peptide attributes including type of CPP and the

size and charge of the cargo. A recent study has indicated that Antp and Tat CPP

internalization can be mediated by clathrin-dependent endocytosis, caveolin-dependent

endocytosis, macropinocytosis, or direct intracellular translocation [226] (Figure 6.1).

Another report used therapeutic cargo to alter the rate of endocytosis, which allowed the

therapy to act in an extracellular fashion [229]. Following endosomal internalization, the

CPP-tagged therapy (CTT) escapes lysosomal degradation by a process that is poorly

understood. It is known however, that endosomal escape is the rate-limiting step in

delivering therapeutic doses of CTTs to the cytosol [224]. Once the CTT is released into 136

the cytosol, it is able to directly interact with its target. In vitro, a CPP (Tat) linked

therapy has been demonstrated to enter the cell and start affecting their target within five

minutes of treatment [230]. In a mouse tumor model, a GFP-CTT (Tat) was observed to

be localized at the tumor site within 30 minutes of treatment, thus illustrating CPPs’ uptake speed and efficiency [123].

137

Figure 6.1 Mechanism of Peptide Membrane Transduction. 1) CPP-Cargo peptide is delivered to site of disease. 2) The CPP’s highly cationic amino acids interact with proteoglycans (left) or glycolipids (right) on the cell surface. Note: There is evidence that the CPP-cargo directly enters the cell (center arrow) 3) The peptide is endocytosed via clathrin or caveoloae or it is macropinocytosed. 4) Endosomal escape of peptide. 5) Interaction with target protein.

138

Non-Immunogenic

One of the benefits of using CPPs for therapeutic delivery is the lack of toxicity

compared to other cytoplasmic delivery devices, such as liposomes, etc. One potential

drawback to using exogenous peptides, though, is the ability to induce a humoral immune

response against the therapy, which can be highly detrimental for subsequent treatments.

A commonly used CPP derived from HIV’s Transcriptional Transactivator (Tat) was

investigated for immunogenicity, and the portions of the Tat protein involved in eliciting

a humoral immune response were determined. The authors discovered that the portion of

Tat that conveys protein transduction, PTD, failed to induce an immune response [210].

In addition, our group has evaluated the induction of an immune response following

repeated PTD-4 injection in immune intact mice, and observed no significant change in

CD45-positive lymphocyte infiltration into tumors [123]. These studies indicate that the

use of Tat’s PTD to deliver anti-cancer therapy does not induce a measurable immune

response, although long-term toxicity studies in patients have not yet been performed.

With respect to another PTD, Antp, several studies have determined that Antp efficiently

elicits an immune response to the peptide cargo following processing by an antigen presenting cell [231, 232]. These examples demonstrate the possibility of modulating the immune response to the CTT based on the PTD used to deliver the therapeutic peptide.

Anti-Cancer Cell Penetrating Peptides

Since 1988, when the first protein transduction domains were described, the use of CPPs to deliver anti-cancer therapies (CTTs) to the cytosol and nuclear compartments has become more common. Over the last decade numerous reports and patents have 139

been published demonstrating the potential of CTTs for the treatment of leukemia and

breast, lung, pancreatic, ovarian, colon cancers [123, 155, 159, 217, 229, 233-235]. With respect to anti-cancer therapies, CTTs have been used to inhibit oncoprotein nuclear translocation, modulate oncoprotein signaling, and block metastases-promoting cell-ECM interactions (Figure 6.2) [123, 217, 229, 233]. The mechanism of inhibition is similar in most cases: a target interaction is determined and a portion or the whole interacting protein is attached to a CPP. The CTT is then transduced across the plasma membrane and delivered into the cytosol or nucleus to block the endogenous pro-cancerous interactions from occurring.

140

Figure 6.2 Mechanism of CTT Anti-Cancer Therapy. 1) The CPP portion of the CTT attaches to the plasma membrane and directly inhibits ECM Receptor/ECM interaction. This inhibits the ability of metastatic cancer cells to establish secondary tumor sites. 2) CTTs block cancer dependent protein-protein interactions, resulting in the inhibition of proliferation, angiogenesis, and chemoresistance. 3) Additionally, inhibition of protein interactions can modulate nuclear translocation of oncogenic transcription factors. 141

Nuclear Translocation

Protein trafficking is a common cellular mechanism that regulates signal

transduction and transcriptional activation. Following oncoprotein signaling, the

translocation of activated proteins into the nucleus often results in the transcription of

genes that are responsible for conveying many of the hallmark of cancers [66]. Increases in oncoprotein nuclear translocation are often observed in cancer cells and can further promote transformation and metastases.

Following DNA damage, there are a number of cell-cycle inhibitor proteins that become activated and are translocated to the nucleus. For example, in normal cells, DNA damage induces p21 nuclear translocation and cell cycle arrest, however, the extent of

DNA damage determines whether the cell undergoes caspase-dependent apoptosis or is able to repair its DNA [236, 237]. Current anti-cancer modalities such as radiation and

most chemotherapies cause irreparable DNA aberrations, which induces the cell to

undergo apoptosis. However, these treatments can also result in the generation of a sub-

population of cancer cells that aberrantly upregulate p21, which results in the inhibition

of caspase 3-dependent apoptosis, thereby allowing the cells to survive the treatment

[158, 159]. Blocking p21 nuclear translocation could inhibit the ability of p21 to convey

chemoresistance, resulting in sensitizing cancer cells and allowing for a reduction in

chemotherapy dosages. In an effort to sensitize cancer cells to the DNA damaging

effects of radiation and chemotherapy, a recent study utilized a CPP conjugated to an

anti-p21 antibody. The CPP-anti-p21 effectively blocked p21 translocation, allowing 142

cells to progress through the cell cycle following chemotherapeutic exposure and

replicate severe DNA adducts, inducing apoptosis [233]. These authors demonstrated

that an IgG region could be conjugated to Tat’s PTD, (immunoconjugates; ICs) and the

tat-anti-p21 IC was shown to inhibit the translocation of p21 into the nucleus [233].

Inhibition of nuclear trafficking of p21 resulted in the loss of the p21 dependent anti- apoptotic effect, and sensitized breast cancer cells to both γ-radiation and Camptothecin, a Topoisomerase inhibitor [233]. This study offers an example of how the tat-anti-p21

IC, in combination with chemotherapies, could result in lower and more effective dosages as well as less discomfort for the patient. The use of CPPs to deliver antibodies directly into the cytosol and nucleus of cancer cells has tremendous potential, allowing for the intracellular targeting of cancer-specific protein mutations.

Aberrant nuclear translocation of proteins is also observed when cells are exposed to pro-inflammatory cytokines, an event frequently observed in epithelial cancers

[Reviewed in [238]]. A protein commonly activated and translocated to the nucleus in the presence of these cytokines is the transcription factor, NF-κB /p65. NF-κB is normally found in the cytosol as a complex of several different proteins (p50, p65, and

IκBα) and following an inflammatory signal, the inhibitor IκBα dissociates from the complex. Following dissociation, p65 is serine phosphorylated and translocated to the nucleus where it activates transcription of several pro-cancerous genes such as, cyclin-

D1, MMP-9 and COX-2 [239]. To inhibit the nuclear translocation of p65, a peptide was designed to block its serine phosphorylation sites. A recently approved patent utilized a p65 derived peptide conjugated with Antp (CPP) to inhibit p65 from activating 143

transcription [240]. The peptide, PTD-p65-1, mimics the amino acid sequence of the p65 protein that spans two serine residues essential to nuclear translocation. In KBM-5 leukemia cells, treatment with PTD-p65-1 was shown to inhibit p65 phosphorylation, p65 nuclear translocation and p50-p65 DNA binding. These PTD-p65-1 treated leukemia cells also demonstrated an increase in the cytotoxic sensitivity following exposure to a chemotherapeutic agent, doxorubicin. Additionally, PTD-p65-1 was able to block the p65-dependent transcription of cyclin-D1, MMP-9, and COX-2 [159]. This indicates that peptides like the PTD-p65-1 can be utilized to sensitize chemotherapy resistant cancer cells to apoptosis or be used to treat chronic inflammation.

Cancer cells often have deregulation of protein expression due to a variety of reasons, including gene amplification, epigenetic alterations, aberrant signaling or atypical protein translation. The altered protein expression of the eukaryotic initiation factor 4E (eIF4E) has been described to increase the translation of mRNAs with a long 5’ untranslated regions [241]. Increased eIF4E has been shown to be directly responsible for the altered protein expression of several oncogenes such as cyclin D1, VEGF, and

FGF2. Additionally, the overexpression of eIF4E has recently been described as an indicator of breast cancer recurrence [242].

eIF4E functions by entering the nucleus, interacting with mRNA, and assist in presenting the mRNA to the ribosome for translation. The nuclear translocation of eIF4E is vital for its function and blocking nuclear entry can promote cytoplasmic accumulation and apoptosis. A recent patent discusses the use of a CPP attached to a portion of the 144

potato virus Y VPg protein to inhibit eIF4E nuclear translocation [243]. In Drosophila

the authors have previously demonstrated that the VPg protein directly binds to eIF4e and

inhibits cell proliferation. The conserved nature of imitation factors allowed a similar

approach in mammalian cells. The authors demonstrated that treatment with CPP-VPg

inhibited eIF4E nuclear translocation, loss of cell proliferation and cell death [243].

Nuclear translocation also plays an important role in signal transduction,

transmitting signals from membrane-bound receptors to the nucleus. The erbB growth

factor receptor family (erbB1/EGFR, erbB2/Her2, erbB3, and erbB4) has been implicated

in the progression of several cancer types including breast, lung, gastrointestinal stromal,

and prostate [Reviewed in [244]]. For example, erbB2 is a common therapeutic target

that is amplified and overexpressed in approximately 30% of all breast cancers [245].

One of the downstream signaling events following erbB2 activation is the tyrosine

phosphorylation, dimerization, and nuclear trafficking of the signal transducers and

activators of transcription 3 (STAT3) transcription factor. STAT3 is been found

deregulated in most solid tumors (breast, ovarian, pancreatic, prostate, and )

and hematological diseases (lymphoma and leukemia) [246-251]. Following STAT3

nuclear translocation, it activates the transcription of several oncogenes that play a role in

conveying a chemoresistant phenotype, such as VEGF, Bcl-2, CyclinD1, and MMP9

[252]. Therefore, nuclear translocation of STAT3 mediated by ErbB2 is an attractive

therapeutic target. 145

In 2006, a report was published that demonstrated that the CPP Tat could be

manipulated to specifically target breast cancer cells that overexpress erbB2/Her2, by

targeting cells with an erbB2 binding peptide conjugated to Tat (P3-AHNP). By

concentrating the P3-AHNP peptide to the cells that were erbB2-positive, the authors

reported that cells overexpressing erbB2 preferentially absorbed the peptide [217]. P3-

AHNP was then additionally conjugated to a STAT3 binding peptide, STAT3BP,

resulting in a CTT that could specifically localize to erbB2 overexpressing breast cancer

cells and block STAT3 nuclear translocation, by blocking STAT3 dimerization [217].

Importantly, P3-AHNP-STAT3BP inhibited STAT3-DNA interactions and its resulting

oncogenic transcriptional activity. A significant decrease in proliferation and an increase

in apoptosis were observed in P3-AHNP-STAT3BP treated breast cancer cell lines and

xenograft tumors [217]. This is a novel example of how the protein expression profile of certain cancers can lead to the development of CTTs that specifically target certain cell- types.

Oncogenic Signaling Modulation

Oncogenic signaling as a result of cancer-dependent protein-protein interactions can promote many aspects of cancer progression, including cell cycle progression, inhibition of apoptosis and insensitivity to anti-growth signals. In addition, changes in

cellular polarity can result in novel protein-protein interactions that are present in cancer

cells, but do not occur in polarized epithelium due to junctional separations. In some

instances these cancer-dependent protein-protein interactions result in aberrant signaling 146

and trafficking. These interactions and signaling events thus provide potential targets for a cancer-specific therapy.

Our laboratory has focused on targeting those interactions that occur in tumor cells, but not in normal polarized epithelium. Basolateral proteins, such as the Epidermal

Growth Factor Receptor (erbB1/EGFR), and apical proteins, such as MUC1 are separated when epithelial cells maintain junctional formations. Loss of polarization in tumor cells, and in metastatic cancer specifically, results in novel protein-protein interactions that promote neoplasia. In the case of MUC1 and EGFR, interactions result in altered protein trafficking and oncogenic signal transduction events that promote breast cancer progression [87]. EGFR is a potent driver of tumor growth and when it interacts with

MUC1, EGFR signaling is amplified [87]. Targeting the interaction domains between these proteins, demonstrated that a CTT, PMIP, blocks MUC1 and EGFR interactions resulting in significantly slower tumor growth [253]. PMIP was designed from a small portion of the MUC1 cytoplasmic tail and conjugated with the CPP, PTD4 [202]. In in vitro studies with breast cancer cell lines, PMIP’s inhibitory effect corresponded to a significant decrease in invasion and proliferation. In the xenograft mouse model, PMIP treatment of the aggressive MDA-MB-231 breast cancer cell line resulted in slower growth and a reduction in distant metastases. Additionally, PMIP dramatically slowed tumor growth and in several instances caused regression of solids tumors in the genetically-driven mouse mammary tumor model (MMTV- py MT). Again, as others have observed, PMIP treated mice showed no weight loss or gross tissue damage, indicating a lack of toxic side-effects [123]. 147

Immune response modulation against cancer has been the main focus of many

cancer researchers because the concept that the body’s natural defense could target and

eliminate tumor cells is an ideal therapeutic approach. Though there are many drawbacks

to the immune modulation approach mainly autoimmunity. However, a recent patent has

utilized a peptide to promote potential tumors antigens to the cell surface of dying tumor

cells which elicits an anti-tumor response. The underlying concept of this patent is based

on the finding that following anthracyclin (common type of chemotherapy, a DNA

intercalating agent) the proteins calreticulin (CRT), KDEL receptor, and ERp57 are

found at the plasma membrane. The membrane localization of these protein results in an

immune response against these tumor antigens. However, this membrane localization is

inefficient in dying cancer cells so a peptide was designed to promote membrane

localization of these proteins. The authors found that inhibition of a protein phosphatase,

GADD34, with a CTT (DPT-PP1 inhibitor) in colon and cervical cancer cells lines

resulted in increased membrane localization of CRT, KDEL receptor, and ERp57.

Additionally, this elicited a greater immune response compared to peptide controls or no

treatment [254]. This a novel use of a CTT to alter intracellular pathways that play an

important role in extracellular processes, such as an immune response.

Most anti-cancer therapies used in the clinic take advantage of the replicative

nature of cancer cells by disrupting DNA replication and synthesis. Radiotherapy (IR) is

often used to cause double-strand breaks in proliferating tumor cells, thereby promoting

apoptosis. However, following radiation exposure the cell has an innate signaling

mechanism to arrest the cell cycle to allow for DNA repair mechanism. For example, 148

upon IR exposure the ataxia telangiectasia mutant (ATM) is activated through the

interaction with another protein, Nijmegen breakage syndrome 1 (NSB-1), and this

activation leads to downstream signaling events which result in cell cycle arrest [255].

The cell cycle arrest allows for DNA repair, which can promote the viability of the cell

and inhibition of apoptosis. Although in normal cells this mechanism is important in

preventing genomic instability however in cancer cells this pathway assists tumor cells in

becoming radioinsensitive. A recent patent describes the use of a peptide based approach

to inhibit the NSB-1/ATM interaction to sensitize tumor cells to IR [256]. The peptide

designed utilized the polyarginine (R9) PTD attached to a NSB-1 mimetic peptide

(wtNIP). A study found that treatment of cervical and prostate cancer cells with wtNIP

resulted in efficient cellular uptake and limited cytotoxicity. However, the authors found

that wtNIP treatment following radiation was able to sensitize these cells and increase

cell death compared to peptide controls [257].

The drawback to the previous approach would be the non-specific nature of the

CTT. In contrast, another patent has utilized a peptide-based therapy to specifically

target the increased availability of single-stranded DNA (ssDNA) in cancer stem cells. In

2006, a report was published demonstrating the presence of “bell-shaped” nuclei in

colorectal tumors and upon closer investigation the authors found this population of cells

contained stem cell like properties [258]. The authors reported that “bell-shaped” nuclei

undergo symmetrical and asymmetrical nuclear fission and this phenomenon results in

increased availability of ssDNA. This led to the design of a peptide that could enter cells,

bind ssDNA, and promote apoptosis of the cancer stem cells. This patent however, 149

mentions the use of CPP to deliver the peptide but does not provide a specific CPP [259].

This a novel therapeutic approach which takes advantage of increased ssDNA to

specifically target cancer stem cells for apoptosis.

Other laboratories have targeted protein-protein interactions that are important in

anti-apoptotic signaling. An important step that leads to the inhibition of apoptosis in

cancer cells is the increased signaling and stabilization of inhibitors of apoptosis proteins

(IAP). One protein that has been found to stabilize IAPs is heat-shock protein 90

(Hsp90). Hsp90 is commonly deregulated in cancer and has been shown to assist the cell

in adapting to environmental stresses [260]. Importantly, Hsp90 is able to directly

interact with an IAP, survivin, and inhibit its proteosomal mediated degradation.

Recently approved patents propose the use of CTTs that specifically targets the

interaction between Hsp90 and survivin [261, 262]. The peptide, shepherdin, was designed to mimic a small domain of the survivin protein that interacts with the ATPase domain of Hsp90 and was then conjugated with the CPPs Antp or Tat. The authors demonstrated that in vitro treatment of several cancer cell lines (breast, prostate, colon, and cervical) with shepherdin caused caspase-dependent apoptosis and loss of membrane integrity [263]. In addition, shepherdin treated breast and prostate tumors grown superficially in immunocompromised mice showed slower tumor growth versus controls and no toxic side-effects were observed. Notably, this report revealed that shepherdin effects were cancer cell specific and did not have any effect on normal fibroblast [263].

Therefore, shepherdin demonstrates the inhibition of protein-protein interactions that 150

results in the non-toxic targeting of tumors via the sensitization of cells to apoptotic

signals.

The use of Hsp90 as a therapeutic peptide target has not been limited to IAPs.

Another recent patent application describes the use of an Hsp90 peptide to promote the

degradation of eNOS [264]. Activation of endothelial nitric-oxide synthase (eNOS) by

Hsp90 results in the increased generation of nitric oxide (NO) which allows the cancer cell to become insensitive to hypoxic and stress conditions. In this manner, activation of eNOS by Hsp90 is able to convey an anti-apoptotic response in several cancers, including oral squamous cell, colon, breast and prostate [265-268]. Therefore the interaction of

Hsp90 and eNOS provides a potential anti-cancer target. An Hsp90 directed CTT was generated, and treatment of endothelial cells resulted in a significant decrease in nitric-

oxide production [269]. The loss of nitric-oxide production has been observed to slow

the proliferation of a malignant melanoma cell line [270]. Thus, the targeting of Hsp90

may be useful in the treatment of multiple cancer types and further demonstrates that the

blocking of cancer-specific protein-protein interactions may be a viable therapeutic

approach.

Modulation of ECM Interactions

Aberrant interactions between tumor cells and the extracellular matrix is an

important component of tumor progression. The ability of tumor cells to form productive

interactions with components of the ECM is critical for movement from the primary 151

organ to distant sites. For example, cells from the primary tumor of the breast

disassociate from the tumor mass and utilize the surrounding ECM to migrate towards

blood and lymph vessels where they intravasate and are transported throughout the body,

eventually forming secondary tumor sites in many tissues, including liver, bone, and

brain [Reviewed in [271]]. This metastatic process is similar in most solid tumors and

demonstrates the critical role of the ECM interactions with the tumor in metastatic

progression [Reviewed in [272]]. A recent study has utilized the Tat derived CPP linked

with an elastin-like polypeptide (Tat-ELP) to inhibit ovarian cancer invasion, migration,

and metastases [229]. Elastin is an extracellular matrix protein commonly found in

connective tissue throughout the body that conveys elasticity to tissues such as blood

vessels, lungs, skin, and the bladder. This CTT was designed to block the interaction

between elastin and circulating tumor cells, resulting in the loss of secondary tumor site

establishment. The concept presented in this report is unique in the sense that the authors

attached a CPP to a large elastin-like polypeptide (800aa), which resulted in Tat-ELP

binding to the cell surface followed by a more gradual endocytosis [229]. In this way,

Tat-ELP is attached to the cell membrane and is functioning to inhibit ECM interactions

prior to internalization. Treatment of ovarian cancer cells with Tat-ELP resulted in an

inhibition of the circulating tumor cells from attaching and establishing secondary tumor

sites [229]. The use of Tat to bind a therapy to the cell surface is a novel approach to

inhibiting ECM Receptor/ECM interactions that could have potential in treating pre-

metastatic and metastatic disease.

152

Novel Therapeutic Targets

The complexity of cancer biology and our understanding of mechanisms

underlying cancer development are ever growing. As new areas of cancer research are

being explored the discovery of new therapeutic targets has been uncovered. Recently,

several patents applications have been submitted which plan on utilizing CPPs to deliver

anti-cancer therapies directed against these novel targets. For example, the role of

microRNA (miRNA) in cancer formation and progression is a relatively new field in

cancer biology so a recent patent describes a miRNA processing protein as a potential

therapeutic target [273]. Another patent plans to utilize CPPs to target a new area of cancer biology, epigenetics. Specifically, this patent describes the use of a CPP to deliver a antisense nucleic acid to block the translation of a splice variant of DNA methyltransferase 3B, which has been shown to promote hypermethylation and gene silencing in cancer [274-276].

In this chapter, I have examined the use of CTTs to target cancer in a manner that is non-toxic to normal cells. In vitro and in vivo CTTs have been shown to translocate across the plasma membrane, directly interact with their target in the cytosol or nucleus, and promote cancer cell death. Additionally, CTTs have also been used to induce sensitivities to chemotherapies and radiation. CTTs have shown to be effective in many cancer types, and while the shift from the laboratory to the clinic has been slow, recent progress in should promote this translation. 153

Endosomal Escape

As more CTTs are being developed and tested in the clinic setting, there is a great

need for a better understanding of how these peptides escape the endosomal

compartments, which limit their therapeutic potential. The ability to optimize endosomal escape would allow for higher therapeutic concentrations in the cytoplasm, resulting in an increase in the CTT/target interaction. However, several of the studies examined in this reviewed show a significant amount of CTT in the tumor several hours after injection.

One possible explanation for this phenomenon is the increased bio-availability of the target found within tumor cells. The CTT fails to be degraded because it is interacting with its target [277].

CPP Stability and Modifications

One concern with using peptides as therapeutics is the potential effects of serum proteases on peptide degradation prior to cellular internalization. A potential modification to CTTs could involve incubation with SDS or polysialic acid, which has been shown to increase peptide stability and diffusion throughout the body [231].

Inhibiting serum protease activity in the blood would also assist in maintaining a higher concentration of therapeutic peptide. Although few studies have attempted this, Yi et al. discovered that treatment of human serum samples with heparin or citrate could stabilize the serum peptides [278]. Another approach to stabilize peptides in the circulatory system is the manipulation of the amino-acid isomerization, which could have a dramatic effect on peptide bioavailability and potency. For example, the isomerization of an L- 154

isoleucine to a D-isoleucine in an anti-angiogenic thrombospondin-1 peptide (Mal II D-

Ile) resulted in better pharmocodynamic properties [279]. The proposed mechanism of

this result was the limited protease activity towards the D-amino acid, thereby increasing

the circulatory concentration of the peptide. Another biological characteristic of D-amino

acids is that processing and presentation to the immune system of D-amino acid peptide

is inefficient thereby limiting an immune response [280]. A recent patent describes the

use of a CPP containing D-amino acid to deliver a targeted anti-cancer therapy to

leukocytes and endothelial cells [281]. In addition to amino acid isomerization, cyclic

peptides that contain a disulfide or amide bonds show an increased therapeutic advantage

versus linear peptides [282]. These studies indicate that the amino acids of CTTs can be

manipulated to optimize peptide bioavailability and efficacy.

Another potential drawback of CTTs is that most of the time the CTT is a low

molecular weight peptide, which results in efficient renal clearance, and poor tumor

penetration. To address this issue one study utilized a hydrophilic polyethylene glycol

coat to act as an efficient “macromolecular drug carrier” [283]. More recently the use of

nanoparticles to delivery macromolecules drugs such as, peptides and genes, offers a potential solution to the drawbacks of just treating with a CTT alone [284]. Theoretically

these carriers would function by forming a “capsule” around the therapy, which would

able to deliver relatively larger doses of CTT through mucosal barriers to target tissues.

In conclusion, the use of CTTs to treat cancer is a relatively new field and there are still many areas that need to be explored. However CTTs show great promise in the 155

field of anti-cancer therapy because of their many benefits including low toxicity,

minimal modulation of immune system, tissue specificity, ability to target most pro- cancerous interactions and regulation of oncogenic signal transduction. The therapeutic

approaches/patents discussed in this chapter are only a snapshot of all new CTTs that have been reported, but it provides a glimpse of the potential these peptides have in the fight against cancer.

156

VII. CONCLUDING STATEMENTS

There are approximately 200,000 new patients diagnosed with breast cancer each

year and over 40,000 will succumb to the disease. Stage and subtype are major determinants of patient prognosis and survival. Breast cancers that are classified into the

basal-like subtype tend to more aggressive and metastatic, which results in decreased

patient survival. Basal-like breast cancers are often described based on their genetic and

proteomic profile. Commonly, these types of breast cancer are estrogen receptor,

progesterone receptor, and erbB2 negative, which means that therapies designed to target

these proteins are ineffective. However, there are several proteins that are highly

upregulated in basal-like breast cancer and these include EGFR and basal epithelium

cytokeratins.

Normally, EGFR expression and signaling is regulated by endocytosis and trafficking mechanisms, but in cancer, mutations in these pathways can result in aberrant

EGFR activity and localization. Loss of polarity in epithelial cells will promote the

mislocalization of EGFR and will result in novel EGFR-protein interactions. For

example, EGFR interacts with another oncogene, MUC1, in a cancer dependent fashion and this interaction is facilitated by the loss of spatial organization. Additionally,

EGFR’s interaction with an adheren junction protein, β-catenin, will promote dissociation

of cell-cell junctions and ultimately loss of cell polarity. 157

The role of MUC1/EGFR and MUC1/β-catenin cancer-dependent protein-protein

interactions in tumorigenesis was established by examining the loss of Muc1 in an EGFR

and an β-catenin-dependent mouse models of breast cancer. In both of these models, the

loss of MUC1 resulted in a significant delay in tumor onset. Mechanistically, in the

EGFR-dependent tumor model cyclin D1 protein expression with directly correlated with

MUC1’s expression status. Previously, our laboratory has observed that the presence of

MUC1 promotes EGFR internalization and recycling. The body of work presented here

demonstrates that the presence of MUC1 promotes EGFR nuclear localization and

mediates EGFR-dependent transcriptional activity. Specifically, we discovered that

MUC1’s interaction with EGFR within the nucleus mediated the EGFR’s interaction with

chromatin and the cyclin-D1 promoter. Upon the loss of MUC1, we found a significant

decrease in cyclin-D1 protein expression. Therefore to inhibit MUC1/EGFR and

MUC1/β-catenin interactions we developed a CPP-mimetic peptide, which mimics a

portion of the MUC1 CT (PMIP). We discovered that the CPP (PTD4) promoted the

efficient uptake of PMIP into breast cancer cells and blocked the target interactions. In

addition, blocking these interactions resulted in a significant decrease of EGFR levels

following EGF treatment. We also observed that by blocking EGFR/MUC1 interactions

in in vitro models of breast cancer we could inhibit EGFR nuclear translocation,

proliferation, and invasion. Importantly, the treatment of PMIP in a xenograft and a

transgenic model of breast cancer significantly inhibited primary tumor growth and the

establishment of secondary tumors. In some cases PMIP was observed to promote the

full regression of mammary gland tumors. This result indicates that although EGFR’s 158

signaling pathways are important in tumorigenesis, that MUC1 and EGFR’s oncogenic

activity could be dependent on each other (Figure 7.3).

MUC1 and EGFR’S Role In Autophagy

The role of autophagy of cancer is a relatively new field and the process of

autophagy can promote cancer cell survival or death. The autophagic outcome is highly

dependent on the extent of cell stress and nutrient bioavailability. Autophagy occurs

when starving cells redistribute nutrients to vital cellular machinery thereby promoting

cell survival in a nutrient depleted environment. In 2009, a new MUC1-dependent cell

survival role was described in which the overexpression of MUC1 promoted ATP

production and cell survival in a low glucose microenvironment. The mechanism

driving this phenotype was determined to be MUC1’s promotion of autophagy in starving

cancer cells [285]. In contrast, the overexpression of a kinase dead EGFR mutant

inhibited autophagy and promoted cell survival by stabilizing a sodium/glucose

transporter (SGLT1)[79]. Stabilization of SGLT1 promoted high levels of intracellular

glucose which prevented the cell from undergoing autophagic death. These studies

demonstrate that both MUC1 and EGFR could be utilized as an autophagic target to

promote cytotoxicity in starving cancer cells.

159

Targeting MUC1/EGFR Interaction and EGFR Activity In Combination

Recently, aberrant EGFR localization within the cell has been associated with

increased chemotherapeutic resistance. Additionally, overexpression of a kinase-dead

mutant of EGFR inhibited autophagic cancer cell death following treatment with a

cytotoxic agent [79]. However, EGFR tyrosine inhibitors do effectively block

downstream signaling and by blocking EGFR/MUC1 interactions we observed a

significant decrease in total EGFR levels. PMIP treatment of breast cancer cells also resulted in the loss of nuclear EGFR localization (Figure 3.3). Therefore we

hypothesized that co-treatment of breast cancer cells with a tyrosine kinase inhibitor and

PMIP could have synergistic affects. The basis of this hypothesis is that by inhibiting

MUC1/EGFR interactions with PMIP we are promoting EGFR degradation, inhibiting

nuclear EGFR function and that by inhibiting the kinase activity of remaining EGFR we

could further exacerbate the anti-tumor effects. Initial experiments using an EGFR

specific kinase-inhibitor (AG1478) and PMIP have resulted in a significant inhibition of

proliferation compared to the single agents (Figure 7.1). This demonstrates that targeting

EGFR kinase activity should not be the sole focus of future anti-EGFR therapeutic. In

the future, targeting EGFR’s aberrant localization in conjunction with EGFR-dependent

downstream signaling pathways (eg. PI3K/Akt) could result in greater anti-tumor

response in basal-like breast cancers.

160

Figure 7.1. EGFR Tyrosine kinase inhibitor (TKI) and PMIP combination significantly inhibit proliferation. An MTT assay using BT20 breast cancer cells treated with a combination of 100nM EGFR TKI (AG1478) and 20μM PMIP for 7 days demonstrated a significant inhibition of proliferation (61% compared to single agent alone (PMIP, 31% and AG1478, 40%)). Student t-test was performed by comparing treatments versus control peptide (CP). MTT assay was performed by Aarthi Goverdhan. 161

EGFR As A Biomarker

The use of EGFR as anti-cancer target is widely accepted, but the use of EGFR

expression in breast cancer diagnosis is still unclear. Overexpression of EGFR in the

basal-like subtype is a poor predictor of clinical outcome, but detection of EGFR expression is not standardize so its use as biomarkers has been limited. However, the

work presented here indicates that although EGFR expression is important the cellular localization of EGFR is also important. The presence of nuclear EGFR has ready been determined to be a poor prognostic indicator in ovarian and breast cancer [44, 46, 47].

Additionally, the mislocalization of MUC1 also is indicative of clinical outcome, the

more cytosolic MUC1 directly correlates with a poorer prognosis [122]. In the future of

breast cancer diagnosis examining the expression and localization of MUC1 with respect to EGFR could be a powerful biomarker.

Preventing Loss of Cell Polarity

This work again re-establishes the importance of MUC1/EGFR and MUC1/β-

catenin interactions, but the underlying fact that is often overlooked is that these

interactions are highly dependent on the loss of cellular polarity. Maintenance of cellular

polarity is an important “tumor suppressor” and breakdown of this cellular organization

promotes novel oncogenic protein-protein interactions. Although this step is critical in

tissue transformation, the mechanism of the loss of cell polarity is poorly understood. In

addition, there has been evidence that EGFR and β-catenin are playing roles in the loss of 162

cell polarity [85, 86]. Given MUC1’s ability to promote EGFR and β-catenin

tumorigenesis, MUC1 overexpression could also be playing a significant role in driving

the loss of polarity. To determine MUC1’s role in the loss of polarity ongoing studies in

our laboratory are examining the overexpression of an apical versus a basolateral

targeting MUC1. These studies will yield important insight into the role of MUC1 in the

early transformation events in polarized epithelial cells.

Role of Heat Shock Proteins In EGFR And MUC1-Dependent Tumorigenesis

Although MUC1’s role in early cancer progression has yet to be established,

MUC1’s interactions with other proteins in cancer cells is now known to promote

proliferation, invasion and chemoresistance. Hsp70/90 have both been determined to

independently interact with EGFR and MUC1. Heat shock proteins 70 and 90

(Hsp70/90) assist in the refolding of proteins, prevent protein aggregation, and are vital in

antigen presentation. Hsp70 interaction with EGFR prevents its aggregation in the

cytosol following Sec61-dependent retrotranslocation from the endoplasmic reticulum

[56]. Hsp70/90 interact with a phosphorylated tyrosine on MUC1’s CT, which promotes

MUC1 mitochondrial localization resulting in the inhibition of the intrinsic apoptosis

pathway [129]. Recently in our laboratory, the interaction of Hsp70 and EGFR has been

determined to be MUC1-dependent [286]. Additionally, the deacetylation of the N-

terminus of proteins has recently been described to inhibit lysosomal degradation [287].

Aberrant endosomal trafficking of EGFR has been observed to be perturbed in the 163

presence of the interacting heat shock protein, Histone Deacetylase-6 (HDAC-6). EGFR

interaction with heat shock proteins and HDAC6 resulted in a significant reduction of

acetylated EGFR and its lysosomal trafficking [288]. HDAC6 regulates this process by

binding to heat shock proteins associated with the aggresomes and will aid in the

chaperoning process [288]. Therefore in the presence of MUC1, Hsp70/90 could be

recruited to EGFR cytoplasmic tail, which increases HDAC6 interaction thereby reducing

EGFR degradation. This could be a possible mechanism of how MUC1 is promoting

EGFR recycling and signaling, but also a mechanism of increased nuclear EGFR in the

presence of MUC1. Importantly, the interactions of MUC1 and EGFR with HSPs are

novel therapeutic targets that could be targeted with a peptide-based therapy.

MUC1’s Role In EGFR Cytosolic Release

The mechanisms of MUC1 and EGFR nuclear translocation are still being

investigated and the work presented invokes the questions do MUC1 and EGFR traffic

together or are their paths to the nucleus independent? I hypothesize that MUC1 and

EGFR are being trafficked together and that the presence of MUC1 in this trafficking

plays an important role in the cytosolic release of EGFR. In the endoplasmic reticulum,

misfolded proteins are presented to Sec61 and this interaction will mediate the

ubquitination of misfolded proteins and subsequent proteosomal degradation [289].

However, if EGFR and MUC1 are being trafficked together, then MUC1 could inhibit

EGFR’s ubquitination as observed in an earlier study, which examined MUC1’s role in 164

aberrant EGFR trafficking [87]. Although this study examined the effect of MUC1 on

ubquitination of membrane associated-EGFR following EGF stimulation the principal

that the presence of MUC1 inhibits EGFR ubquitination could still be a valid mechanism

for increased EGFR cytosolic release and decreased proteosomal degradation.

Nuclear EGFR Trafficking: Anti-Cancer Target

Overexpression of EGFR is commonly seen in the basal-like subtype of breast

cancer, but therapeutic responses to anti-EGFR tyrosine kinase inhibitors has been

limited. The work presented here demonstrates the EGFR oncogenic function can be

multi-faceted, because although EGFR signaling is important, cellular localization in

cancer cells is starting to be appreciated as a mechanism of increased proliferation and

radioresistance. Trafficking of EGFR to the nucleus involves the interaction with several

other proteins including heat shock proteins and importins, therefore to inhibit nuclear

EGFR accumulation and function we have developed an EGFR/Importinβ1 inhibitory

peptide. We utilized the protein transduction domain (PTD4) and mimicked EGFR’s

NLS to create a peptide (eNLS) which would compete for importin interaction thereby

inhibit EGFR nuclear translocation. Preliminary results indicated that targeting EGFR

nuclear trafficking inhibits proliferation in cancer cells (Figure 7.2). Novel anti-cancer

therapies that target aberrant localization of EGFR and other proteins offers a new field

of cancer therapeutics.

165

0.2 Day 0 Day 3 0.15

0.1

0.05 Relative Absorbance Relative 0 50uM eNLS 50uM CP 50uM Water NT

Figure 7.2. Blocking nuclear EGFR translocation inhibits cell proliferation. BT20 breast cancer cells were treated with eNLS, control peptide (CP), vehicle (water), or non- treated for 3 days. Cells were then counted using an MTT assay. Student t-test was performed to determine that eNLS significantly inhibited proliferation (p=0.0001). Assay was performed by Aarthi Goverdhan. 166

The work presented in this dissertation offers a new insight into a novel

mechanism of EGFR activity in breast cancer development, progression, and chemo-

resistance. Additionally, a 'first-in-class' peptide-based therapy was developed that

demonstrates the oncogenic importance of MUC1/EGFR and MUC1/beta-catenin

interactions. In the future, peptide-based drugs may be viable options in the pursuit of

non-toxic cancer-specific therapies. For example, a peptide that could inhibit nuclear

translocation of EGFR could result increased chemotherapy sensitivity and cancer-

specific cytotoxicity. Overall, this body of works demonstrates the importance of

EGFR's activity and localization within cells and the use of mimetic peptides as potential

anti-breast cancer therapeutics.

167

Figure 7.3. Working Model with PMIP Treatment A) In a normal polarized epithelial cell

MUC1 (red, 1) is constitutively recycled, after EGF stimulation EGFR (green, 2) is degraded in the lysosome with minimal recycling (3), and b-catenin (blue) is associated with adherens REFERENCESjunctions. B) As cell undergoes transformation MUC1 expression and EGFR activity are increased, which promotes the loss of polarization by dissociation of β-catenin from adherens junctions (1). Loss of polarity promotes the mislocalization of proteins resulting in novel protein-protein interactions. C) These interactions promote EGFR recycling (1) and β- catenin-dependent transcriptional activation, and trafficking of EGFR into the nucleus (2). These cancer-dependent events result in increase metastases, proliferation and chemoresistance. D) Treatment with PMIP inhibits MUC1/EGFR (1) and MUC1/ β-catenin interactions (2), which results in decreased invasion, proliferation, cell growth, and chemoresistance. Additionally, PMIP inhibits EGFR nuclear accumulation, which downregulates the cell-cycle regulator, cyclin D1.

168

IIX. REFERENCES

1. Eikuch. Epidermal growth factor receptor (EGFR) signaling pathway. 2007 [cited February 14th, 2010]; Available from: http://commons.wikimedia.org/wiki/File:EGFR_signaling_pathway.png. 2. Brayman, M., A. Thathiah, and D.D. Carson, MUC1: a multifunctional cell surface component of reproductive tissue epithelia. Reprod Biol Endocrinol, 2004. 2: p. 4. 3. Kufe, D.W., Mucins in cancer: function, prognosis and therapy. Nat Rev Cancer, 2009. 9(12): p. 874-85. 4. Carson, D.D., The cytoplasmic tail of MUC1: a very busy place. Sci Signal, 2008. 1(27): p. pe35. 5. Lo, H.W. and M.C. Hung, Nuclear EGFR signalling network in cancers: linking EGFR pathway to cell cycle progression, nitric oxide pathway and patient survival. Br J Cancer, 2006. 94(2): p. 184-8. 6. Hynes, N.E. and H.A. Lane, ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer, 2005. 5(5): p. 341-54. 7. Visvader, J.E., Keeping abreast of the mammary epithelial hierarchy and breast tumorigenesis. Genes Dev, 2009. 23(22): p. 2563-77. 8. Pochampalli, M.R., B.G. Bitler, and J.A. Schroeder, Transforming growth factor alpha dependent cancer progression is modulated by Muc1. Cancer Res, 2007. 67(14): p. 6591-8. 9. Tamoxifen for early breast cancer: an overview of the randomised trials. Early Breast Cancer Trialists' Collaborative Group. Lancet, 1998. 351(9114): p. 1451-67. 10. Perou, C.M., et al., Molecular portraits of human breast tumours. Nature, 2000. 406(6797): p. 747-52. 11. Nielsen, T.O., et al., Immunohistochemical and clinical characterization of the basal- like subtype of invasive breast carcinoma. Clin Cancer Res, 2004. 10(16): p. 5367-74. 12. Thike, A.A., et al., Triple-negative breast cancer: clinicopathological characteristics and relationship with basal-like breast cancer. Mod Pathol. 23(1): p. 123-33. 13. Livasy, C.A., et al., Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol, 2006. 19(2): p. 264-71. 14. DiRenzo, J., et al., Growth factor requirements and basal phenotype of an immortalized mammary epithelial cell line. Cancer Res, 2002. 62(1): p. 89-98. 15. Foulkes, W.D., et al., Germline BRCA1 mutations and a basal epithelial phenotype in breast cancer. J Natl Cancer Inst, 2003. 95(19): p. 1482-5. 16. Chen, J.J., et al., BRCA1, BRCA2, and Rad51 operate in a common DNA damage response pathway. Cancer Res, 1999. 59(7 Suppl): p. 1752s-1756s. 17. Futreal, P.A., et al., BRCA1 mutations in primary breast and ovarian carcinomas. Science, 1994. 266(5182): p. 120-2. 18. Miki, Y., et al., A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science, 1994. 266(5182): p. 66-71. 19. Narod, S., et al., Increasing incidence of breast cancer in family with BRCA1 mutation. Lancet, 1993. 341(8852): p. 1101-2. 20. Burness, M.L., T.A. Grushko, and O.I. Olopade, Epidermal growth factor receptor in triple-negative and Basal-like breast cancer: promising clinical target or only a marker? Cancer J, 2010. 16(1): p. 23-32. 169

21. Anders, C. and L.A. Carey, Understanding and treating triple-negative breast cancer. Oncology (Williston Park), 2008. 22(11): p. 1233-9; discussion 1239-40, 1243. 22. Strunk, K.E., V. Amann, and D.W. Threadgill, Phenotypic variation resulting from a deficiency of epidermal growth factor receptor in mice is caused by extensive genetic heterogeneity that can be genetically and molecularly partitioned. Genetics, 2004. 167(4): p. 1821-32. 23. Ullrich, A., et al., Human epidermal growth factor receptor cDNA sequence and aberrant expression of the amplified gene in A431 epidermoid carcinoma cells. Nature, 1984. 309(5967): p. 418-25. 24. Carpenter, G., L. King, Jr., and S. Cohen, Epidermal growth factor stimulates phosphorylation in membrane preparations in vitro. Nature, 1978. 276(5686): p. 409- 10. 25. Davies, R.L., et al., Genetic analysis of epidermal growth factor action: assignment of human epidermal growth factor receptor gene to chromosome 7. Proc Natl Acad Sci U S A, 1980. 77(7): p. 4188-92. 26. Ishii, S., et al., Characterization and sequence of the promoter region of the human epidermal growth factor receptor gene. Proc Natl Acad Sci U S A, 1985. 82(15): p. 4920-4. 27. Kageyama, R., G.T. Merlino, and I. Pastan, Epidermal growth factor (EGF) receptor gene transcription. Requirement for Sp1 and an EGF receptor-specific factor. J Biol Chem, 1988. 263(13): p. 6329-36. 28. Mialon, A., et al., DNA topoisomerase I is a cofactor for c-Jun in the regulation of epidermal growth factor receptor expression and cancer cell proliferation. Mol Cell Biol, 2005. 25(12): p. 5040-51. 29. He, C., et al., The epidermal growth factor receptor juxtamembrane domain has multiple basolateral plasma membrane localization determinants, including a dominant signal with a polyproline core. J Biol Chem, 2002. 277(41): p. 38284-93. 30. Goldman, R., et al., Heterodimerization of the erbB-1 and erbB-2 receptors in human breast carcinoma cells: a mechanism for receptor transregulation. Biochemistry, 1990. 29(50): p. 11024-8. 31. Yarden, Y. and J. Schlessinger, Epidermal growth factor induces rapid, reversible aggregation of the purified epidermal growth factor receptor. Biochemistry, 1987. 26(5): p. 1443-51. 32. Samanta, A., et al., Ligand and p185c-neu density govern receptor interactions and tyrosine kinase activation. Proc Natl Acad Sci U S A, 1994. 91(5): p. 1711-5. 33. Guy, P.M., et al., Insect cell-expressed p180erbB3 possesses an impaired tyrosine kinase activity. Proc Natl Acad Sci U S A, 1994. 91(17): p. 8132-6. 34. Zhang, W. and H.T. Liu, MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res, 2002. 12(1): p. 9-18. 35. Kennedy, S.G., et al., Akt/Protein kinase B inhibits cell death by preventing the release of cytochrome c from mitochondria. Mol Cell Biol, 1999. 19(8): p. 5800-10. 36. Hellyer, N.J., K. Cheng, and J.G. Koland, ErbB3 (HER3) interaction with the p85 regulatory subunit of phosphoinositide 3-kinase. Biochem J, 1998. 333 ( Pt 3): p. 757- 63. 37. Sithanandam, G., et al., Alternate paths from epidermal growth factor receptor to Akt in malignant versus nontransformed lung epithelial cells: ErbB3 versus Gab1. Am J Respir Cell Mol Biol, 2005. 33(5): p. 490-9. 170

38. Panasyuk, G., et al., mTORbeta splicing isoform promotes cell proliferation and tumorigenesis. J Biol Chem, 2009. 284(45): p. 30807-14. 39. Rawlings, J.S., K.M. Rosler, and D.A. Harrison, The JAK/STAT signaling pathway. J Cell Sci, 2004. 117(Pt 8): p. 1281-3. 40. Li, W.X., Canonical and non-canonical JAK-STAT signaling. Trends Cell Biol, 2008. 18(11): p. 545-51. 41. Nyati, M.K., et al., Integration of EGFR inhibitors with radiochemotherapy. Nat Rev Cancer, 2006. 6(11): p. 876-85. 42. Slamon, D.J., et al., Human breast cancer: correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science, 1987. 235(4785): p. 177-82. 43. Alimandi, M., et al., Cooperative signaling of ErbB3 and ErbB2 in neoplastic transformation and human mammary carcinomas. Oncogene, 1995. 10(9): p. 1813-21. 44. Hadzisejdic, I., et al., Nuclear EGFR in ductal invasive breast cancer: correlation with cyclin-D1 and prognosis. Mod Pathol, 2010. 45. Hsu, S.C., et al., Nuclear EGFR is required for cisplatin resistance and DNA repair. Am J Transl Res, 2009. 1(3): p. 249-58. 46. Lo, H.W., et al., Novel prognostic value of nuclear epidermal growth factor receptor in breast cancer. Cancer Res, 2005. 65(1): p. 338-48. 47. Xia, W., et al., Nuclear expression of epidermal growth factor receptor is a novel prognostic value in patients with ovarian cancer. Mol Carcinog, 2008. 48. Carpenter, G., The EGF receptor: a nexus for trafficking and signaling. Bioessays, 2000. 22(8): p. 697-707. 49. de Melker, A.A., G. van der Horst, and J. Borst, c-Cbl directs EGF receptors into an endocytic pathway that involves the ubiquitin-interacting motif of Eps15. J Cell Sci, 2004. 117(Pt 21): p. 5001-12. 50. Smit, G.D., et al., Cbl controls EGFR fate by regulating early endosome fusion. Sci Signal, 2009. 2(102): p. ra86. 51. Vieira, A.V., C. Lamaze, and S.L. Schmid, Control of EGF receptor signaling by clathrin-mediated endocytosis. Science, 1996. 274(5295): p. 2086-9. 52. Lin, S.Y., et al., Nuclear localization of EGF receptor and its potential new role as a transcription factor. Nat Cell Biol, 2001. 3(9): p. 802-8. 53. Ni, C.Y., et al., gamma -Secretase cleavage and nuclear localization of ErbB-4 receptor tyrosine kinase. Science, 2001. 294(5549): p. 2179-81. 54. Offterdinger, M., et al., c-erbB-3: a nuclear protein in mammary epithelial cells. J Cell Biol, 2002. 157(6): p. 929-39. 55. Wang, S.C., et al., Binding at and transactivation of the COX-2 promoter by nuclear tyrosine kinase receptor ErbB-2. Cancer Cell, 2004. 6(3): p. 251-61. 56. Liao, H.J. and G. Carpenter, Role of the Sec61 translocon in EGF receptor trafficking to the nucleus and gene expression. Mol Biol Cell, 2007. 18(3): p. 1064-72. 57. Johnson, A.E. and M.A. van Waes, The translocon: a dynamic gateway at the ER membrane. Annu Rev Cell Dev Biol, 1999. 15: p. 799-842. 58. Hsu, S.C. and M.C. Hung, Characterization of a novel tripartite nuclear localization sequence in the EGFR family. J Biol Chem, 2007. 282(14): p. 10432-40. 59. Lo, H.W., et al., Nuclear-cytoplasmic transport of EGFR involves receptor endocytosis, importin beta1 and CRM1. J Cell Biochem, 2006. 98(6): p. 1570-83. 60. Giri, D.K., et al., Endosomal transport of ErbB-2: mechanism for nuclear entry of the cell surface receptor. Mol Cell Biol, 2005. 25(24): p. 11005-18. 171

61. Hanada, N., et al., Co-regulation of B-Myb expression by E2F1 and EGF receptor. Mol Carcinog, 2006. 45(1): p. 10-7. 62. Lo, H.W., et al., Nuclear interaction of EGFR and STAT3 in the activation of the iNOS/NO pathway. Cancer Cell, 2005. 7(6): p. 575-89. 63. Hung, L.Y., et al., Nuclear epidermal growth factor receptor (EGFR) interacts with signal transducer and activator of transcription 5 (STAT5) in activating Aurora-A gene expression. Nucleic Acids Res, 2008. 36(13): p. 4337-51. 64. Lo, H.W., et al., Cyclooxygenase-2 is a novel transcriptional target of the nuclear EGFR-STAT3 and EGFRvIII-STAT3 signaling axes. Mol Cancer Res, 2010. 8(2): p. 232-45. 65. Oliveras-Ferraros, C., et al., Growth and molecular interactions of the anti-EGFR antibody cetuximab and the DNA cross-linking agent cisplatin in gefitinib-resistant MDA-MB-468 cells: new prospects in the treatment of triple-negative/basal-like breast cancer. Int J Oncol, 2008. 33(6): p. 1165-76. 66. Hanahan, D. and R.A. Weinberg, The hallmarks of cancer. Cell, 2000. 100(1): p. 57- 70. 67. Guy, C.T., et al., Expression of the neu protooncogene in the mammary epithelium of transgenic mice induces metastatic disease. Proc Natl Acad Sci U S A, 1992. 89(22): p. 10578-82. 68. Rose-Hellekant, T.A. and E.P. Sandgren, Transforming growth factor alpha- and c- myc-induced mammary carcinogenesis in transgenic mice. Oncogene, 2000. 19(8): p. 1092-6. 69. Moulder, S.L. and C.L. Arteaga, A Phase I/II Trial of trastuzumab and gefitinib in patients with Metastatic Breast Cancer that overexpresses HER2/neu (ErbB-2). Clin Breast Cancer, 2003. 4(2): p. 142-5. 70. Bhargava, R., et al., EGFR gene amplification in breast cancer: correlation with epidermal growth factor receptor mRNA and protein expression and HER-2 status and absence of EGFR-activating mutations. Mod Pathol, 2005. 18(8): p. 1027-33. 71. Wikstrand, C.J., et al., Monoclonal antibodies against EGFRvIII are tumor specific and react with breast and lung carcinomas and malignant gliomas. Cancer Res, 1995. 55(14): p. 3140-8. 72. Casci, T., J. Vinos, and M. Freeman, Sprouty, an intracellular inhibitor of Ras signaling. Cell, 1999. 96(5): p. 655-65. 73. Sirotnak, F.M., et al., Efficacy of cytotoxic agents against human tumor xenografts is markedly enhanced by coadministration of ZD1839 (Iressa), an inhibitor of EGFR tyrosine kinase. Clin Cancer Res, 2000. 6(12): p. 4885-92. 74. Kokubo, Y., et al., Reduction of PTEN protein and loss of epidermal growth factor receptor gene mutation in lung cancer with natural resistance to gefitinib (IRESSA). Br J Cancer, 2005. 92(9): p. 1711-9. 75. Engelman, J.A. and P.A. Janne, Mechanisms of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small cell lung cancer. Clin Cancer Res, 2008. 14(10): p. 2895-9. 76. Baselga, J., Why the epidermal growth factor receptor? The rationale for cancer therapy. Oncologist, 2002. 7 Suppl 4: p. 2-8. 77. Elie, C., et al., Lack of relationship between EGFR-1 immunohistochemical expression and prognosis in a multicentre clinical trial of 93 patients with advanced primary ovarian epithelial cancer (GINECO group). Br J Cancer, 2004. 91(3): p. 470-5. 172

78. Saltz, L.B., et al., Phase II trial of cetuximab in patients with refractory that expresses the epidermal growth factor receptor. J Clin Oncol, 2004. 22(7): p. 1201-8. 79. Weihua, Z., et al., Survival of cancer cells is maintained by EGFR independent of its kinase activity. Cancer Cell, 2008. 13(5): p. 385-93. 80. Li, C., et al., Nuclear EGFR contributes to acquired resistance to cetuximab. Oncogene, 2009. 28(43): p. 3801-13. 81. Lee, M. and V. Vasioukhin, Cell polarity and cancer--cell and tissue polarity as a non- canonical tumor suppressor. J Cell Sci, 2008. 121(Pt 8): p. 1141-50. 82. Betschinger, J., K. Mechtler, and J.A. Knoblich, The Par complex directs asymmetric cell division by phosphorylating the cytoskeletal protein Lgl. Nature, 2003. 422(6929): p. 326-30. 83. Kuwada, S.K., et al., Differential signaling and regulation of apical vs. basolateral EGFR in polarized epithelial cells. Am J Physiol, 1998. 275(6 Pt 1): p. C1419-28. 84. Aranda, V., et al., Par6-aPKC uncouples ErbB2 induced disruption of polarized epithelial organization from proliferation control. Nat Cell Biol, 2006. 8(11): p. 1235- 45. 85. Wang, Y., et al., Tyrosine phosphorylated Par3 regulates epithelial tight junction assembly promoted by EGFR signaling. EMBO J, 2006. 25(21): p. 5058-70. 86. Lu, Z., et al., Downregulation of caveolin-1 function by EGF leads to the loss of E- cadherin, increased transcriptional activity of beta-catenin, and enhanced tumor cell invasion. Cancer Cell, 2003. 4(6): p. 499-515. 87. Pochampalli, M.R., R.M. el Bejjani, and J.A. Schroeder, MUC1 is a novel regulator of ErbB1 receptor trafficking. Oncogene, 2007. 26(12): p. 1693-701. 88. Park, S.Y., et al., Expression of MUC1, MUC2, MUC5AC and MUC6 in cholangiocarcinoma: prognostic impact. Oncol Rep, 2009. 22(3): p. 649-57. 89. Zotter, S., et al., Monoclonal antibodies to epithelial sialomucins recognize epitopes at different cellular sites in adenolymphomas of the parotid gland. Int J Cancer Suppl, 1988. 3: p. 38-44. 90. Ajioka, Y., L.J. Allison, and J.R. Jass, Significance of MUC1 and MUC2 mucin expression in colorectal cancer. J Clin Pathol, 1996. 49(7): p. 560-4. 91. Ando, H., et al., Prognostic significance of the expression of MUC1 and collagen type IV in advanced gastric carcinoma. Br J Surg, 2009. 96(8): p. 901-9. 92. Gao, J., et al., MUC1 is a downstream target of STAT3 and regulates lung cancer cell survival and invasion. Int J Oncol, 2009. 35(2): p. 337-45. 93. Aubert, S., et al., MUC1, a new hypoxia inducible factor target gene, is an actor in clear renal cell carcinoma tumor progression. Cancer Res, 2009. 69(14): p. 5707-15. 94. Hilkens, J., et al., Monoclonal antibodies against human milk-fat globule membranes detecting differentiation antigens of the mammary gland and its tumors. Int J Cancer, 1984. 34(2): p. 197-206. 95. Zotter, S., et al., Tissue and tumor distribution of human polymorphic epithelial mucin. Cancer Reviews, 1988. 11-12: p. 55-101. 96. Rakha, E.A., et al., Expression of mucins (MUC1, MUC2, MUC3, MUC4, MUC5AC and MUC6) and their prognostic significance in human breast cancer. Mod Pathol, 2005. 18(10): p. 1295-304. 97. Li, Y., et al., Human DF3/MUC1 carcinoma-associated protein functions as an oncogene. Oncogene, 2003. 22(38): p. 6107-10. 173

98. Schroeder, J.A., et al., MUC1 overexpression results in mammary gland tumorigenesis and prolonged alveolar differentiation. Oncogene, 2004. 23(34): p. 5739-47. 99. Hattrup, C.L. and S.J. Gendler, MUC1 alters oncogenic events and transcription in human breast cancer cells. Breast Cancer Res, 2006. 8(4): p. R37. 100. Ahmad, R., et al., MUC1-C Oncoprotein Functions as a Direct Activator of the Nuclear Factor-{kappa}B p65 Transcription Factor. Cancer Res, 2009. 101. Wreschner, D.H., et al., Generation of ligand-receptor alliances by "SEA" module- mediated cleavage of membrane-associated mucin proteins. Protein Sci, 2002. 11(3): p. 698-706. 102. Strous, G.J. and J. Dekker, Mucin-type glycoproteins. Crit Rev Biochem Mol Biol, 1992. 27(1-2): p. 57-92. 103. Hollingsworth, M.A. and B.J. Swanson, Mucins in cancer: protection and control of the cell surface. Nat Rev Cancer, 2004. 4(1): p. 45-60. 104. McGuckin, M.A., et al., Muc1 mucin limits both Helicobacter pylori colonization of the murine gastric mucosa and associated gastritis. Gastroenterology, 2007. 133(4): p. 1210-8. 105. Yu, L.G., et al., Galectin-3 interaction with Thomsen-Friedenreich disaccharide on cancer-associated MUC1 causes increased cancer cell endothelial adhesion. J Biol Chem, 2007. 282(1): p. 773-81. 106. Hayashi, T., et al., MUC1 mucin core protein binds to the domain 1 of ICAM-1. Digestion, 2001. 63 Suppl 1: p. 87-92. 107. Zhang, K., et al., Secreted MUC1 mucins lacking their cytoplasmic part and carrying sialyl-Lewis a and x epitopes from a tumor cell line and sera of colon carcinoma patients can inhibit HL-60 leukocyte adhesion to E-selectin-expressing endothelial cells. J Cell Biochem, 1996. 60(4): p. 538-49. 108. Ramasamy, S., et al., The MUC1 and galectin-3 oncoproteins function in a microRNA-dependent regulatory loop. Mol Cell, 2007. 27(6): p. 992-1004. 109. Zhao, Q., et al., Circulating galectin-3 promotes metastasis by modifying MUC1 localization on cancer cell surface. Cancer Res, 2009. 69(17): p. 6799-806. 110. Schroeder, J.A., et al., Transgenic MUC1 interacts with epidermal growth factor receptor and correlates with mitogen-activated protein kinase activation in the mouse mammary gland. J Biol Chem, 2001. 276(16): p. 13057-64. 111. Huang, L., et al., MUC1 cytoplasmic domain coactivates Wnt target gene transcription and confers transformation. Cancer Biol Ther, 2003. 2(6): p. 702-6. 112. Leng, Y., et al., Nuclear import of the MUC1-C oncoprotein is mediated by nucleoporin Nup62. J Biol Chem, 2007. 282(27): p. 19321-30. 113. Pemberton, L.F., et al., The epithelial mucin MUC1 contains at least two discrete signals specifying membrane localization in cells. J Biol Chem, 1996. 271(4): p. 2332- 40. 114. Zaretsky, J.Z., et al., MUC1 gene overexpressed in breast cancer: structure and transcriptional activity of the MUC1 promoter and role of estrogen receptor alpha (ERalpha) in regulation of the MUC1 gene expression. Mol Cancer, 2006. 5: p. 57. 115. Gaemers, I.C., et al., A stat-responsive element in the promoter of the episialin/MUC1 gene is involved in its overexpression in carcinoma cells. J Biol Chem, 2001. 276(9): p. 6191-9. 116. Abba, M.C., et al., GATA3 protein as a MUC1 transcriptional regulator in breast cancer cells. Breast Cancer Res, 2006. 8(6): p. R64. 174

117. Ligtenberg, M.J., et al., Cell-associated episialin is a complex containing two proteins derived from a common precursor. J Biol Chem, 1992. 267(9): p. 6171-7. 118. Carson, D.D., M.M. DeSouza, and E.G. Regisford, Mucin and proteoglycan functions in embryo implantation. Bioessays, 1998. 20(7): p. 577-83. 119. Litvinov, S.V. and J. Hilkens, The epithelial sialomucin, episialin, is sialylated during recycling. J Biol Chem, 1993. 268(28): p. 21364-71. 120. Ren, J., et al., Human MUC1 carcinoma-associated protein confers resistance to genotoxic anticancer agents. Cancer Cell, 2004. 5(2): p. 163-75. 121. Wen, Y., et al., Nuclear association of the cytoplasmic tail of MUC1 and beta-catenin. J Biol Chem, 2003. 278(39): p. 38029-39. 122. Rahn, J.J., et al., The importance of MUC1 cellular localization in patients with breast carcinoma: an immunohistologic study of 71 patients and review of the literature. Cancer, 2001. 91(11): p. 1973-82. 123. Bitler, B.G., et al., Intracellular MUC1 peptides inhibit cancer progression. Clin Cancer Res, 2009. 15(1): p. 100-9. 124. Schroeder, J.A., et al., MUC1 alters beta-catenin-dependent tumor formation and promotes cellular invasion. Oncogene, 2003. 22(9): p. 1324-32. 125. Huang, L., et al., MUC1 oncoprotein blocks glycogen synthase kinase 3beta-mediated phosphorylation and degradation of beta-catenin. Cancer Res, 2005. 65(22): p. 10413- 22. 126. Li, Y., et al., The epidermal growth factor receptor regulates interaction of the human DF3/MUC1 carcinoma antigen with c-Src and beta-catenin. J Biol Chem, 2001. 276(38): p. 35239-42. 127. Al Masri, A. and S.J. Gendler, Muc1 affects c-Src signaling in PyV MT-induced mammary tumorigenesis. Oncogene, 2005. 24(38): p. 5799-808. 128. Raina, D., S. Kharbanda, and D. Kufe, The MUC1 oncoprotein activates the anti- apoptotic phosphoinositide 3-kinase/Akt and Bcl-xL pathways in rat 3Y1 fibroblasts. J Biol Chem, 2004. 279(20): p. 20607-12. 129. Ren, J., et al., MUC1 oncoprotein is targeted to mitochondria by heregulin-induced activation of c-Src and the molecular chaperone HSP90. Oncogene, 2006. 25(1): p. 20- 31. 130. Cowin, P., T.M. Rowlands, and S.J. Hatsell, Cadherins and catenins in breast cancer. Curr Opin Cell Biol, 2005. 17(5): p. 499-508. 131. Orsulic, S. and M. Peifer, An in vivo structure-function study of armadillo, the beta- catenin homologue, reveals both separate and overlapping regions of the protein required for cell adhesion and for wingless signaling. J Cell Biol, 1996. 134(5): p. 1283-300. 132. Lickert, H., et al., Casein kinase II phosphorylation of E-cadherin increases E- cadherin/beta-catenin interaction and strengthens cell-cell adhesion. J Biol Chem, 2000. 275(7): p. 5090-5. 133. Pokutta, S., et al., Biochemical and structural definition of the l-afadin- and actin- binding sites of alpha-catenin. J Biol Chem, 2002. 277(21): p. 18868-74. 134. Qian, X., et al., E-cadherin-mediated adhesion inhibits ligand-dependent activation of diverse receptor tyrosine kinases. EMBO J, 2004. 23(8): p. 1739-48. 135. Lilien, J. and J. Balsamo, The regulation of cadherin-mediated adhesion by tyrosine phosphorylation/dephosphorylation of beta-catenin. Curr Opin Cell Biol, 2005. 17(5): p. 459-65. 175

136. Davis, M.A., R.C. Ireton, and A.B. Reynolds, A core function for p120-catenin in cadherin turnover. J Cell Biol, 2003. 163(3): p. 525-34. 137. Hart, M., et al., The F-box protein beta-TrCP associates with phosphorylated beta- catenin and regulates its activity in the cell. Curr Biol, 1999. 9(4): p. 207-10. 138. Amit, S., et al., Axin-mediated CKI phosphorylation of beta-catenin at Ser 45: a molecular switch for the Wnt pathway. Genes Dev, 2002. 16(9): p. 1066-76. 139. Noordermeer, J., et al., dishevelled and armadillo act in the wingless signalling pathway in Drosophila. Nature, 1994. 367(6458): p. 80-3. 140. Knudsen, K.A. and M.J. Wheelock, Cadherins and the mammary gland. J Cell Biochem, 2005. 95(3): p. 488-96. 141. Chen, G., et al., Up-regulation of Wnt-1 and beta-catenin production in patients with advanced metastatic prostate carcinoma: potential pathogenetic and prognostic implications. Cancer, 2004. 101(6): p. 1345-56. 142. Morin, P.J., et al., Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science, 1997. 275(5307): p. 1787-90. 143. Orsulic, S., et al., E-cadherin binding prevents beta-catenin nuclear localization and beta-catenin/LEF-1-mediated transactivation. J Cell Sci, 1999. 112 ( Pt 8): p. 1237-45. 144. Civenni, G., T. Holbro, and N.E. Hynes, Wnt1 and Wnt5a induce cyclin D1 expression through ErbB1 transactivation in HC11 mammary epithelial cells. EMBO Rep, 2003. 4(2): p. 166-71. 145. Ji, H., et al., EGF-induced ERK activation promotes CK2-mediated disassociation of alpha-Catenin from beta-Catenin and transactivation of beta-Catenin. Mol Cell, 2009. 36(4): p. 547-59. 146. Fagotto, F., U. Gluck, and B.M. Gumbiner, Nuclear localization signal-independent and importin/karyopherin-independent nuclear import of beta-catenin. Curr Biol, 1998. 8(4): p. 181-90. 147. Townsley, F.M., A. Cliffe, and M. Bienz, Pygopus and Legless target Armadillo/beta- catenin to the nucleus to enable its transcriptional co-activator function. Nat Cell Biol, 2004. 6(7): p. 626-33. 148. Huber, O., et al., Nuclear localization of beta-catenin by interaction with transcription factor LEF-1. Mech Dev, 1996. 59(1): p. 3-10. 149. Kim, K., Z. Lu, and E.D. Hay, Direct evidence for a role of beta-catenin/LEF-1 signaling pathway in induction of EMT. Cell Biol Int, 2002. 26(5): p. 463-76. 150. Cano, A., et al., The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol, 2000. 2(2): p. 76-83. 151. Tomita, K., et al., Cadherin switching in human prostate cancer progression. Cancer Res, 2000. 60(13): p. 3650-4. 152. Lee, J.M., et al., The epithelial-mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol, 2006. 172(7): p. 973-81. 153. Derossi, D., et al., The third helix of the Antennapedia homeodomain translocates through biological membranes. J Biol Chem, 1994. 269(14): p. 10444-50. 154. Schwarze, S.R., et al., In vivo protein transduction: delivery of a biologically active protein into the mouse. Science, 1999. 285(5433): p. 1569-72. 155. Yu, Z., et al., A recombinant cell-permeable p53 is selectively stabilized under hypoxia and inhibits tumor cell growth. Cancer Lett, 2009. 279(1): p. 101-7. 156. Hu, M., et al., Site-specific conjugation of HIV-1 tat peptides to IgG: a potential route to construct radioimmunoconjugates for targeting intracellular and nuclear epitopes in cancer. Eur J Nucl Med Mol Imaging, 2006. 33(3): p. 301-10. 176

157. Chiu, Y.L., et al., Visualizing a correlation between siRNA localization, cellular uptake, and RNAi in living cells. Chem Biol, 2004. 11(8): p. 1165-75. 158. Waldman, T., et al., Cell-cycle arrest versus cell death in cancer therapy. Nat Med, 1997. 3(9): p. 1034-6. 159. Takada, Y., S. Singh, and B.B. Aggarwal, Identification of a p65 peptide that selectively inhibits NF-kappa B activation induced by various inflammatory stimuli and its role in down-regulation of NF-kappaB-mediated gene expression and up- regulation of apoptosis. J Biol Chem, 2004. 279(15): p. 15096-104. 160. Lan, M.S., et al., Cloning and sequencing of a human pancreatic tumor mucin cDNA. J Biol Chem, 1990. 265(25): p. 15294-9. 161. Ramsby, M.L., G.S. Makowski, and E.A. Khairallah, Differential detergent fractionation of isolated hepatocytes: biochemical, immunochemical and two- dimensional gel electrophoresis characterization of cytoskeletal and noncytoskeletal compartments. Electrophoresis, 1994. 15(2): p. 265-77. 162. Davis, J.R., M. Kakar, and C.S. Lim, Controlling protein compartmentalization to overcome disease. Pharm Res, 2007. 24(1): p. 17-27. 163. Pozner-Moulis, S., D.J. Pappas, and D.L. Rimm, Met, the receptor, localizes to the nucleus in cells at low density. Cancer Res, 2006. 66(16): p. 7976-82. 164. Maher, P.A., Nuclear Translocation of (FGF) receptors in response to FGF-2. J Cell Biol, 1996. 134(2): p. 529-36. 165. Feng, Y., et al., VEGF induces nuclear translocation of Flk-1/KDR, endothelial nitric oxide synthase, and caveolin-1 in vascular endothelial cells. Biochem Biophys Res Commun, 1999. 256(1): p. 192-7. 166. Le Roy, C. and J.L. Wrana, Clathrin- and non-clathrin-mediated endocytic regulation of cell signalling. Nat Rev Mol Cell Biol, 2005. 6(2): p. 112-26. 167. Sandgren, E.P., et al., Inhibition of mammary gland involution is associated with transforming growth factor alpha but not c-myc-induced tumorigenesis in transgenic mice. Cancer Res, 1995. 55(17): p. 3915-27. 168. Wei, X., H. Xu, and D. Kufe, MUC1 oncoprotein stabilizes and activates estrogen receptor alpha. Mol Cell, 2006. 21(2): p. 295-305. 169. Liao, H.J. and G. Carpenter, Cetuximab/C225-induced intracellular trafficking of epidermal growth factor receptor. Cancer Res, 2009. 69(15): p. 6179-83. 170. Zhang, H.M., et al., Mitogen-induced recruitment of ERK and MSK to SRE promoter complexes by ternary complex factor Elk-1. Nucleic Acids Res, 2008. 36(8): p. 2594- 607. 171. Gillett, C., et al., Amplification and overexpression of cyclin D1 in breast cancer detected by immunohistochemical staining. Cancer Res, 1994. 54(7): p. 1812-7. 172. Aggarwal, P., et al., Nuclear accumulation of cyclin D1 during S phase inhibits Cul4- dependent Cdt1 proteolysis and triggers p53-dependent DNA rereplication. Genes Dev, 2007. 21(22): p. 2908-22. 173. Kornmann, M., N. Arber, and M. Korc, Inhibition of basal and mitogen-stimulated pancreatic cancer cell growth by cyclin D1 antisense is associated with loss of tumorigenicity and potentiation of cytotoxicity to cisplatinum. J Clin Invest, 1998. 101(2): p. 344-52. 174. Kalish, L.H., et al., Deregulated cyclin D1 expression is associated with decreased efficacy of the selective epidermal growth factor receptor tyrosine kinase inhibitor 177

gefitinib in head and neck squamous cell carcinoma cell lines. Clin Cancer Res, 2004. 10(22): p. 7764-74. 175. Rollini, P., et al., Expression pattern of mouse mammary tumor virus in transgenic mice carrying exogenous proviruses of different origins. J Virol, 1992. 66(7): p. 4580- 6. 176. Nusse, R. and H.E. Varmus, Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome. Cell, 1982. 31(1): p. 99-109. 177. Li, Y., W.P. Hively, and H.E. Varmus, Use of MMTV-Wnt-1 transgenic mice for studying the genetic basis of breast cancer. Oncogene, 2000. 19(8): p. 1002-9. 178. Wong, S.C., et al., Expression of frizzled-related protein and Wnt-signalling molecules in invasive human breast tumours. J Pathol, 2002. 196(2): p. 145-53. 179. Huguet, E.L., et al., Differential expression of human Wnt genes 2, 3, 4, and 7B in human breast cell lines and normal and disease states of human breast tissue. Cancer Res, 1994. 54(10): p. 2615-21. 180. MacDonald, B.T., K. Tamai, and X. He, Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell, 2009. 17(1): p. 9-26. 181. Veeck, J., et al., Aberrant methylation of the Wnt antagonist SFRP1 in breast cancer is associated with unfavourable prognosis. Oncogene, 2006. 25(24): p. 3479-88. 182. Brabletz, T., et al., beta-catenin regulates the expression of the matrix metalloproteinase-7 in human colorectal cancer. Am J Pathol, 1999. 155(4): p. 1033-8. 183. Tetsu, O. and F. McCormick, Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature, 1999. 398(6726): p. 422-6. 184. Schroeder, J.A., K.L. Troyer, and D.C. Lee, Cooperative induction of mammary tumorigenesis by TGFalpha and Wnts. Oncogene, 2000. 19(28): p. 3193-9. 185. Schroeder, J.A., et al., ErbB-beta-catenin complexes are associated with human infiltrating ductal breast and murine mammary tumor virus (MMTV)-Wnt-1 and MMTV-c-Neu transgenic carcinomas. J Biol Chem, 2002. 277(25): p. 22692-8. 186. Sibilia, M. and E.F. Wagner, Strain-dependent epithelial defects in mice lacking the EGF receptor. Science, 1995. 269(5221): p. 234-8. 187. Luetteke, N.C., et al., The mouse waved-2 phenotype results from a point mutation in the EGF receptor tyrosine kinase. Genes Dev, 1994. 8(4): p. 399-413. 188. Roberts, R.B., et al., Importance of epidermal growth factor receptor signaling in establishment of adenomas and maintenance of carcinomas during intestinal tumorigenesis. Proc Natl Acad Sci U S A, 2002. 99(3): p. 1521-6. 189. Wiesen, J.F., et al., Signaling through the stromal epidermal growth factor receptor is necessary for mammary ductal development. Development, 1999. 126(2): p. 335-44. 190. Hilkens, J., et al., Is episialin/MUC1 involved in breast cancer progression? Cancer Lett, 1995. 90(1): p. 27-33. 191. Brossart, P., et al., The epithelial tumor antigen MUC1 is expressed in hematological malignancies and is recognized by MUC1-specific cytotoxic T-lymphocytes. Cancer Res, 2001. 61(18): p. 6846-50. 192. Takahashi, T., et al., Expression of MUC1 on myeloma cells and induction of HLA- unrestricted CTL against MUC1 from a multiple myeloma patient. J Immunol, 1994. 153(5): p. 2102-9. 193. Teruya-Feldstein, J., et al., MUC-1 mucin protein expression in B-cell lymphomas. Appl Immunohistochem Mol Morphol, 2003. 11(1): p. 28-32. 178

194. Yamamoto, M., et al., Interaction of the DF3/MUC1 breast carcinoma-associated antigen and beta-catenin in cell adhesion. J Biol Chem, 1997. 272(19): p. 12492-4. 195. Li, Y., et al., Interaction of glycogen synthase kinase 3beta with the DF3/MUC1 carcinoma-associated antigen and beta-catenin. Mol Cell Biol, 1998. 18(12): p. 7216- 24. 196. Polakis, P., Wnt signaling and cancer. Genes Dev, 2000. 14(15): p. 1837-51. 197. He, T.C., et al., Identification of c-MYC as a target of the APC pathway. Science, 1998. 281(5382): p. 1509-12. 198. Shtutman, M., et al., The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc Natl Acad Sci U S A, 1999. 96(10): p. 5522-7. 199. Tsukamoto, A.S., et al., Expression of the int-1 gene in transgenic mice is associated with mammary gland hyperplasia and adenocarcinomas in male and female mice. Cell, 1988. 55(4): p. 619-25. 200. Michaelson, J.S. and P. Leder, beta-catenin is a downstream effector of Wnt-mediated tumorigenesis in the mammary gland. Oncogene, 2001. 20(37): p. 5093-9. 201. Spicer, A.P., et al., Delayed mammary tumor progression in Muc-1 null mice. J Biol Chem, 1995. 270(50): p. 30093-101. 202. Ho, A., et al., Synthetic protein transduction domains: enhanced transduction potential in vitro and in vivo. Cancer Res, 2001. 61(2): p. 474-7. 203. Wadia, J.S. and S.F. Dowdy, Transmembrane delivery of protein and peptide drugs by TAT-mediated transduction in the treatment of cancer. Adv Drug Deliv Rev, 2005. 57(4): p. 579-96. 204. Mosmann, T., Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods, 1983. 65(1-2): p. 55-63. 205. Webster, M.A., et al., Requirement for both Shc and phosphatidylinositol 3' kinase signaling pathways in polyomavirus middle T-mediated mammary tumorigenesis. Mol Cell Biol, 1998. 18(4): p. 2344-59. 206. Maglione, J.E., et al., Transgenic Polyoma middle-T mice model premalignant mammary disease. Cancer Res, 2001. 61(22): p. 8298-305. 207. Lin, E.Y., et al., Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am J Pathol, 2003. 163(5): p. 2113-26. 208. Puri, N. and R. Salgia, Synergism of EGFR and c-Met pathways, cross-talk and inhibition, in non-small cell lung cancer. J Carcinog, 2008. 7: p. 9. 209. Trivedi, P., et al., Augmentation of leukocyte infiltration in murine tumors expressing B-cell derived but not nasopharyngeal carcinoma derived EBV membrane protein LMP1. J Med Virol, 2000. 60(4): p. 417-24. 210. Kittiworakarn, J., et al., HIV-1 Tat raises an adjuvant-free humoral immune response controlled by its core region and its ability to form cysteine-mediated oligomers. J Biol Chem, 2006. 281(6): p. 3105-15. 211. Torchilin, V.P. and A.N. Lukyanov, Peptide and protein drug delivery to and into tumors: challenges and solutions. Drug Discov Today, 2003. 8(6): p. 259-66. 212. Packer, L.M., et al., Expression of the cell surface mucin gene family in adenocarcinomas. Int J Oncol, 2004. 25(4): p. 1119-26. 213. Dassonville, O., et al., EGFR targeting therapies: monoclonal antibodies versus tyrosine kinase inhibitors. Similarities and differences. Crit Rev Oncol Hematol, 2007. 62(1): p. 53-61. 179

214. Bitler, B.G. and J.A. Schroeder, Anti-Cancer Therapies that Utilize Cell Penetrating Peptides. Recent Pat Anticancer Drug Discov, 2009. 215. Hong, F.D. and G.L. Clayman, Isolation of a peptide for targeted drug delivery into human head and neck solid tumors. Cancer Res, 2000. 60(23): p. 6551-6. 216. Popiel, H.A., et al., Delivery of the aggregate inhibitor peptide QBP1 into the mouse brain using PTDs and its therapeutic effect on polyglutamine disease mice. Neurosci Lett, 2009. 449(2): p. 87-92. 217. Tan, M., et al., Selective inhibition of ErbB2-overexpressing breast cancer in vivo by a novel TAT-based ErbB2-targeting signal transducers and activators of transcription 3- blocking peptide. Cancer Res, 2006. 66(7): p. 3764-72. 218. Elferink, J.G., Changes of plasma membrane permeability in neutrophils treated with polycations. Inflammation, 1991. 15(2): p. 103-15. 219. Elliott, G. and P. O'Hare, Intercellular trafficking and protein delivery by a herpesvirus structural protein. Cell, 1997. 88(2): p. 223-33. 220. Karas, M., Intracellular delivery of small molecules, proteins, and nucleic acids, U.P. Office, Editor. 2007, Canbrex Bio Science Walkersville, Inc.: USA. 221. Sadler, K., et al., Translocating proline-rich peptides from the antimicrobial peptide bactenecin 7. Biochemistry, 2002. 41(48): p. 14150-7. 222. Pooga, M., et al., Cell penetration by transportan. FASEB J, 1998. 12(1): p. 67-77. 223. Morris, M.C., et al., A peptide carrier for the delivery of biologically active proteins into mammalian cells. Nat Biotechnol, 2001. 19(12): p. 1173-6. 224. Edenhofer, F., Protein transduction revisited: novel insights into the mechanism underlying intracellular delivery of proteins. Curr Pharm Des, 2008. 14(34): p. 3628- 36. 225. Heitz, F., M.C. Morris, and G. Divita, Twenty years of cell-penetrating peptides: from molecular mechanisms to therapeutics. Br J Pharmacol, 2009. 157(2): p. 195-206. 226. Duchardt, F., et al., A comprehensive model for the cellular uptake of cationic cell- penetrating peptides. Traffic, 2007. 8(7): p. 848-66. 227. Gump, J.M. and S.F. Dowdy, TAT transduction: the molecular mechanism and therapeutic prospects. Trends Mol Med, 2007. 13(10): p. 443-8. 228. Fawell, S., et al., Tat-mediated delivery of heterologous proteins into cells. Proc Natl Acad Sci U S A, 1994. 91(2): p. 664-8. 229. Massodi, I., et al., Inhibition of ovarian cancer cell metastasis by a fusion polypeptide Tat-ELP. Clin Exp Metastasis, 2009. 26(3): p. 251-60. 230. Wadia, J.S., R.V. Stan, and S.F. Dowdy, Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nat Med, 2004. 10(3): p. 310-5. 231. Schutze-Redelmeier, M.P., et al., Introduction of exogenous antigens into the MHC class I processing and presentation pathway by Drosophila antennapedia homeodomain primes cytotoxic T cells in vivo. J Immunol, 1996. 157(2): p. 650-5. 232. Schutze-Redelmeier, M.P., et al., Antennapedia transduction sequence promotes anti tumour immunity to epicutaneously administered CTL epitopes. Vaccine, 2004. 22(15- 16): p. 1985-91. 233. Hu, M., et al., HIV-1 Tat peptide immunoconjugates differentially sensitize breast cancer cells to selected antiproliferative agents that induce the cyclin-dependent kinase inhibitor p21WAF-1/CIP-1. Bioconjug Chem, 2006. 17(5): p. 1280-7. 180

234. Bidwell, G.L., 3rd, A.N. Davis, and D. Raucher, Targeting a c-Myc inhibitory polypeptide to specific intracellular compartments using cell penetrating peptides. J Control Release, 2009. 135(1): p. 2-10. 235. Harada, H., M. Hiraoka, and S. Kizaka-Kondoh, Antitumor effect of TAT-oxygen- dependent degradation-caspase-3 fusion protein specifically stabilized and activated in hypoxic tumor cells. Cancer Res, 2002. 62(7): p. 2013-8. 236. Huo, J.X., S.A. Metz, and G.D. Li, p53-independent induction of p21(waf1/cip1) contributes to the activation of caspases in GTP-depletion-induced apoptosis of insulin-secreting cells. Cell Death Differ, 2004. 11(1): p. 99-109. 237. Gartel, A.L. and A.L. Tyner, The role of the cyclin-dependent kinase inhibitor p21 in apoptosis. Mol Cancer Ther, 2002. 1(8): p. 639-49. 238. Shan, W., G. Yang, and J. Liu, The inflammatory network: bridging senescent stroma and epithelial tumorigenesis. Front Biosci, 2009. 14: p. 4044-57. 239. Shishodia, S., et al., Ursolic acid inhibits nuclear factor-kappaB activation induced by carcinogenic agents through suppression of IkappaBalpha kinase and p65 phosphorylation: correlation with down-regulation of cyclooxygenase 2, matrix metalloproteinase 9, and cyclin D1. Cancer Res, 2003. 63(15): p. 4375-83. 240. Aggarwal, B.B. and S. Singh, Selective inhibitors of nuclear factor-κB activation and uses thereof. 2008, Research Development Foundation. 241. Svitkin, Y.V., et al., The requirement for eukaryotic initiation factor 4A (elF4A) in translation is in direct proportion to the degree of mRNA 5' secondary structure. RNA, 2001. 7(3): p. 382-94. 242. McClusky, D.R., et al., A prospective trial on initiation factor 4E (eIF4E) overexpression and cancer recurrence in node-positive breast cancer. Ann Surg, 2005. 242(4): p. 584-90; discussion 590-2. 243. Chroboczek, J., Zagorski, W., Grezla, R. 2008. 244. Holbro, T., G. Civenni, and N.E. Hynes, The ErbB receptors and their role in cancer progression. Exp Cell Res, 2003. 284(1): p. 99-110. 245. Kallioniemi, O.P., et al., Association of c-erbB-2 protein over-expression with high rate of cell proliferation, increased risk of visceral metastasis and poor long-term survival in breast cancer. Int J Cancer, 1991. 49(5): p. 650-5. 246. Burke, W.M., et al., Inhibition of constitutively active Stat3 suppresses growth of human ovarian and breast cancer cells. Oncogene, 2001. 20(55): p. 7925-34. 247. Scholz, A., et al., Activated signal transducer and activator of transcription 3 (STAT3) supports the malignant phenotype of human pancreatic cancer. Gastroenterology, 2003. 125(3): p. 891-905. 248. Mora, L.B., et al., Constitutive activation of Stat3 in human prostate tumors and cell lines: direct inhibition of Stat3 signaling induces apoptosis of prostate cancer cells. Cancer Res, 2002. 62(22): p. 6659-66. 249. Niu, G., et al., Roles of activated Src and Stat3 signaling in melanoma tumor cell growth. Oncogene, 2002. 21(46): p. 7001-10. 250. Aoki, Y., G.M. Feldman, and G. Tosato, Inhibition of STAT3 signaling induces apoptosis and decreases survivin expression in primary effusion lymphoma. Blood, 2003. 101(4): p. 1535-42. 251. Nakajima, K., et al., A central role for Stat3 in IL-6-induced regulation of growth and differentiation in M1 leukemia cells. EMBO J, 1996. 15(14): p. 3651-8. 252. Hsieh, F.C., G. Cheng, and J. Lin, Evaluation of potential Stat3-regulated genes in human breast cancer. Biochem Biophys Res Commun, 2005. 335(2): p. 292-9. 181

253. Schroeder, J.A., Therapeutic peptides for the treatment of metastatic cancer. 2006. 254. Kroemer, G., Zitvogel, L. 2006. 255. Cerosaletti, K. and P. Concannon, Independent roles for nibrin and Mre11-Rad50 in the activation and function of Atm. J Biol Chem, 2004. 279(37): p. 38813-9. 256. Cariveau, M.J., Xu, B. 2006. 257. Cariveau, M.J., et al., Characterization of an NBS1 C-terminal peptide that can inhibit ataxia telangiectasia mutated (ATM)-mediated DNA damage responses and enhance radiosensitivity. Mol Pharmacol, 2007. 72(2): p. 320-6. 258. Gostjeva, E.V., et al., Bell-shaped nuclei dividing by symmetrical and asymmetrical nuclear fission have qualities of stem cells in human colonic embryogenesis and carcinogenesis. Cancer Genet Cytogenet, 2006. 164(1): p. 16-24. 259. Gostjeva, E.V., Thilly, W.G. 2007. 260. Isaacs, J.S., W. Xu, and L. Neckers, Heat shock protein 90 as a molecular target for cancer therapeutics. Cancer Cell, 2003. 3(3): p. 213-7. 261. Altieri, D.C., J. Plescia, and S. Whitney, Compounds that inhibit Hsp90 protein- protein interactions with IAP proteins, U.P. Office, Editor. 2008, University of Massachusetts. 262. Altieri, D.C., Kang, B.H. 2007. 263. Plescia, J., et al., Rational design of shepherdin, a novel anticancer agent. Cancer Cell, 2005. 7(5): p. 457-68. 264. Pritchard, K., H. Xu, and Y. Shi, HSP90 Decoy Peptides and Uses Thereof, U.P. Office, Editor. 2006: USA. 265. Shang, Z.J., Z.B. Li, and J.R. Li, In vitro effects of nitric oxide synthase inhibitor L- NAME on oral squamous cell carcinoma: a preliminary study. Int J Oral Maxillofac Surg, 2006. 35(6): p. 539-43. 266. Wang, L., et al., Expression of endothelial nitric oxide synthase correlates with the angiogenic phenotype of and predicts poor prognosis in human gastric cancer. Gastric Cancer, 2005. 8(1): p. 18-28. 267. Vakkala, M., P. Paakko, and Y. Soini, eNOS expression is associated with the estrogen and progesterone receptor status in invasive breast carcinoma. Int J Oncol, 2000. 17(4): p. 667-71. 268. Tong, X. and H. Li, eNOS protects prostate cancer cells from TRAIL-induced apoptosis. Cancer Lett, 2004. 210(1): p. 63-71. 269. Xu, H., et al., A heat shock protein 90 binding domain in endothelial nitric-oxide synthase influences enzyme function. J Biol Chem, 2007. 282(52): p. 37567-74. 270. Tao, J., et al., Endogenous production of nitric oxide contributes to proliferation effect of vascular endothelial growth factor-induced malignant melanoma cell. Clin Exp Dermatol, 2006. 31(1): p. 94-9. 271. Liotta, L.A. and E.C. Kohn, The microenvironment of the tumour-host interface. Nature, 2001. 411(6835): p. 375-9. 272. O'Hayre, M., et al., Chemokines and cancer: migration, intracellular signalling and intercellular communication in the microenvironment. Biochem J, 2008. 409(3): p. 635-49. 273. Gregory, R.I., Daley, G.Q., Viswanathan, S.R. 2007. 274. Godley, L.A., Ostler, K.R. 2008. 275. Rhee, I., et al., DNMT1 and DNMT3b cooperate to silence genes in human cancer cells. Nature, 2002. 416(6880): p. 552-6. 182

276. Ostler, K.R., et al., Cancer cells express aberrant DNMT3B transcripts encoding truncated proteins. Oncogene, 2007. 26(38): p. 5553-63. 277. Maeda, H., et al., Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Release, 2000. 65(1-2): p. 271-84. 278. Yi, J., et al., Intrinsic Peptidase Activity Causes a Sequential Multi-Step Reaction (SMSR) in Digestion of Human Plasma Peptides. J Proteome Res, 2008. 279. Dawson, D.W., et al., Three distinct D-amino acid substitutions confer potent antiangiogenic activity on an inactive peptide derived from a thrombospondin-1 type 1 repeat. Mol Pharmacol, 1999. 55(2): p. 332-8. 280. Uebel, S., et al., Recognition principle of the TAP transporter disclosed by combinatorial peptide libraries. Proc Natl Acad Sci U S A, 1997. 94(17): p. 8976-81. 281. Peer, D., Shimaoka, M. 2009. 282. Leduc, A.M., et al., Helix-stabilized cyclic peptides as selective inhibitors of steroid receptor-coactivator interactions. Proc Natl Acad Sci U S A, 2003. 100(20): p. 11273- 8. 283. Prego, C., et al., Transmucosal macromolecular drug delivery. J Control Release, 2005. 101(1-3): p. 151-62. 284. de la Fuente, M., et al., Nanoparticles as protein and gene carriers to mucosal surfaces. Nanomed, 2008. 3(6): p. 845-57. 285. Yin, L., S. Kharbanda, and D. Kufe, MUC1 oncoprotein promotes autophagy in a survival response to glucose deprivation. Int J Oncol, 2009. 34(6): p. 1691-9. 286. Hart, M., EGFR proteomics 2010. 287. Hwang, C.S., A. Shemorry, and A. Varshavsky, N-terminal acetylation of cellular proteins creates specific degradation signals. Science, 2010. 327(5968): p. 973-7. 288. Deribe, Y.L., et al., Regulation of epidermal growth factor receptor trafficking by lysine deacetylase HDAC6. Sci Signal, 2009. 2(102): p. ra84. 289. Deak, P.M. and D.H. Wolf, Membrane topology and function of Der3/Hrd1p as a ubiquitin-protein ligase (E3) involved in endoplasmic reticulum degradation. J Biol Chem, 2001. 276(14): p. 10663-9.

1. Eikuch. Epidermal growth factor receptor (EGFR) signaling pathway. 2007 [cited February 14th, 2010]; Available from: http://commons.wikimedia.org/wiki/File:EGFR_signaling_pathway.png. 2. Brayman, M., A. Thathiah, and D.D. Carson, MUC1: a multifunctional cell surface component of reproductive tissue epithelia. Reprod Biol Endocrinol, 2004. 2: p. 4. 3. Kufe, D.W., Mucins in cancer: function, prognosis and therapy. Nat Rev Cancer, 2009. 9(12): p. 874‐85. 4. Lee, M. and V. Vasioukhin, Cell polarity and cancer‐‐cell and tissue polarity as a non‐ canonical tumor suppressor. J Cell Sci, 2008. 121(Pt 8): p. 1141‐50. 5. Carson, D.D., The cytoplasmic tail of MUC1: a very busy place. Sci Signal, 2008. 1(27): p. pe35. 183

6. Lo, H.W. and M.C. Hung, Nuclear EGFR signalling network in cancers: linking EGFR pathway to cell cycle progression, nitric oxide pathway and patient survival. Br J Cancer, 2006. 94(2): p. 184‐8. 7. Hynes, N.E. and H.A. Lane, ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer, 2005. 5(5): p. 341‐54. 8. Visvader, J.E., Keeping abreast of the mammary epithelial hierarchy and breast tumorigenesis. Genes Dev, 2009. 23(22): p. 2563‐77. 9. Pochampalli, M.R., B.G. Bitler, and J.A. Schroeder, Transforming growth factor alpha dependent cancer progression is modulated by Muc1. Cancer Res, 2007. 67(14): p. 6591‐8. 10. Cheang, M.C., et al., Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst, 2009. 101(10): p. 736‐50. 11. Millikan, R.C., et al., Epidemiology of basal‐like breast cancer. Breast Cancer Res Treat, 2008. 109(1): p. 123‐39. 12. Yang, X.R., et al., Differences in risk factors for breast cancer molecular subtypes in a population‐based study. Cancer Epidemiol Biomarkers Prev, 2007. 16(3): p. 439‐43. 13. Lin, S.Y., et al., Nuclear localization of EGF receptor and its potential new role as a transcription factor. Nat Cell Biol, 2001. 3(9): p. 802‐8. 14. Schroeder, J.A., K.L. Troyer, and D.C. Lee, Cooperative induction of mammary tumorigenesis by TGFalpha and Wnts. Oncogene, 2000. 19(28): p. 3193‐9.