CONTRACTING ORGANIZATION: Rush University Medical Center Chicago, IL 60612

Similar documents
CONTRACTING ORGANIZATION: University of California Lawrence Berkeley National Laboratory Berkeley, CA 94720

Fort Detrick, Maryland

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

CONTRACTING ORGANIZATION: Western Institute For Biomedical Research Salt Lake City, UT

TITLE: Targeting the Immune System s Natural Response to Cell Death to Improve Therapeutic Response in Breast Cancers

TITLE: Neural Protein Synuclein (SNCG) in Breast Cancer Progression

TITLE: Dietary Genistein and Prostate Cancer Chemoprevention

CONTRACTING ORGANIZATION: Mount Sinai School of Medicine New York, New York

TITLE: 64Cu-DOTA-trastuzumab PET imaging in women with HER2 overexpressing breast cancer

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

La Jolla, CA Approved for Public Release; Distribution Unlimited

CONTRACTING ORGANIZATION: North Eastern Ohio Universities Rootstown OH 44202

Award Number: W81XWH TITLE: Global Positioning Systems (GPS) Technology to Study Vector-Pathogen-Host Interactions

TITLE: A PSCA Promoter Based Avian Retroviral Transgene Model of Normal and Malignant Prostate

CONTRACTING ORGANIZATION: Vanderbilt University Medical Center Nashville, TN

TITLE: Evaluation of DNA Methylation as a Target for Intraductal Therapy for Ductal Carcinoma in Situ of the Breast

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Award Number: W81XWH

TITLE: New Advanced Technology to Improve Prediction and Prevention of Type 1 Diabetes

Award Number: W81XWH TITLE: Characterizing an EMT Signature in Breast Cancer. PRINCIPAL INVESTIGATOR: Melanie C.

TITLE: Selective Oncolytic Therapy for Hypoxic Breast Cancer Cells. CONTRACTING ORGANIZATION: Ordway Research Institute Albany, New York 12208

TITLE: Genes Associated with Food Allergy and Eosinophilic Esophagitis

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Role of ADAM15 in Tumor/Endothelial Interactions Prostate Cancer Regression

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: A Tissue Engineering Approach to Study the Progression of Breast Tumor Metastasis in Bone

TITLE: The Role of hcdc4 as a Tumor Suppressor Gene in Genomic Instability Underlying Prostate Cancer

TITLE: Enhancing the Anti-Tumor Activity of ErbB Blockers with Histone Deaccetylase (HDAC) Inhibition in Prostate Cancer Cell Lines

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Investigation of the Akt/PKB Kinase in the Development of Hormone-Independent Prostate Cancer

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Effect of MUC1 Expression on EGFR Endocytosis and Degradation in Human Breast Cancer Cell Lines

CONTRACTING ORGANIZATION: Johns Hopkins Bloomberg School of Public Health

CONTRACTING ORGANIZATION: Cold Spring Harbor Laboratory Cold Spring Harbor, NY 11724

Award Number: W81XWH TITLE: A Novel Membrane-Permeable, Breast-Targeting, Pro-Apoptotic Peptide for Treatment of Breast Cancer

TITLE: Investigating the Role of TBX2 in the Inhibition of Senescence in Prostate Cancer

TITLE: Induction of Ephs/Ephrins-Mediated Tumor Cells-Endothelial Cells Repulsion as an Anti-Cancer Therapeutic Approach

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Award Number: W81XWH TITLE: Dietary Fish Oil in Reducing Bone Metastasis of Breast Cancer

Approved for public release; distribution unlimited

TITLE: Oxidative Stress, DNA Repair and Prostate Cancer Risk. PRINCIPAL INVESTIGATOR: Hua Zhao, Ph.D.

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

CONTRACTING ORGANIZATION: Regents of the University of Michigan Ann Arbor, MI 48109

CONTRACTING ORGANIZATION: Sloan-Kettering Institute for Cancer Research New York, NY 10021

TITLE: Identification of New Serum Biomarkers for Early Breast Cancer Diagnosis and Prognosis Using Lipid Microarrays.

TITLE: Prevention and Treatment of Neurofibromatosis Type 1-Associated Malignant Peripheral Nerve Sheath Tumors

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: TREATMENT OF ENDOCRINE-RESISTANT BREAST CANCER WITH A SMALL MOLECULE C-MYC INHIBITOR

CONTRACTING ORGANIZATION: University of Illinois Chicago, IL

Award Number: W81XWH TITLE: Regenerative Stem Cell Therapy for Breast Cancer Bone Metastasis

CONTRACTING ORGANIZATION: University of Mississippi Medical Center Jackson, MS 39216

CONTRACTING ORGANIZATION: Children s Hospital Los Angeles Los Angeles, CA 90027

TITLE: Adipose Estrogen and Increased Breast Cancer Risk in Obesity: Regulation by Leptin and Insulin

TITLE: Effect of COX-2 (PGE2) and IL-6 on Prostate Cancer Bone Mets

TITLE: Hyaluronic acid is overexpressed in fibrotic lung tissue and promotes collagen expression

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: HER2/neu Antisense Therapeutics in Human Breast Cancer. CONTRACTING ORGANIZATION: Washington University School of Medicine St.

Detection of Prostate Cancer Progression by Serum DNA Integrity

Award Number: W81XWH TITLE: Synthetic Lethal Gene for PTEN as a Therapeutic Target. PRINCIPAL INVESTIGATOR: Kounosuke Watabe, Ph.D.

TITLE: Breast Tumor-Generated Type 1 Collagen Breakdown Fragments Act as Matrikines to Drive Osteolysis

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

AWARD NUMBER: W81XWH TITLE: Treatment of Pain and Autonomic Dysreflexia in Spinal Cord Injury with Deep Brain Stimulation

CONTRACTING ORGANIZATION: University of Texas M.D. Anderson Cancer Center Houston, TX 77030

AD (Leave blank) PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: The Importance of Autophagy in Breast Cancer Development and Treatment

AD (Leave blank) Award Number: W81XWH TITLE: Targeting Autophagy for the Treatment of TSC and LAM. PRINCIPAL INVESTIGATOR: Elizabeth Henske

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

Sphingosine-1-Phosphate Receptor Subtype 3: A Novel Therapeutic Target of K-Ras Mutant Driven Non-Small Cell Lung Carcinoma

TITLE: Properties of Leukemia Stem Cells in a Novel Model of CML Progression to Lymphoid Blast Crisis

TITLE: The Role of HOX Proteins in Androgen-Independent Prostate Cancer

U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Systemic Oncolytic Cytokine HSV Therapy of Prostate Cancer. CONTRACTING ORGANIZATION: Massachusetts General Hospital Boston, MA

TITLE: Overcoming Resistance to Inhibitors of the Akt Protein Kinase by Modulation of the Pim Kinase Pathway

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: "Impact of Obesity on Tamoxifen Chemoprevention in a Model of Ductal Carcinoma in Situ"

TITLE: Harnessing GPR17 Biology for Treating Demyelinating Disease

AD (Leave blank) TITLE: Proteomic analyses of nipple fluid for early detection of breast cancer

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

TITLE: Maximizing Energy After Traumatic Brain Injury: A Novel Intervention

AWARD NUMBER: W81XWH TITLE: Androgen Deprivation Therapy and Cognitive Impairment. PRINCIPAL INVESTIGATOR: Robert N. Pechnick, Ph.D.

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, MD

TITLE: Inhibitors of Histone Deacetylases for Radiosensitization of Prostate Cancer

TITLE: Prostate Expression Databases: Gene Expression Resources for Comparative Studies of Prostate Carcinogenesis

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Post-Traumatic Headache and Psychological Health: Mindfulness Training for Mild Traumatic Brain Injury

CONTRACTING ORGANIZATION: Oregon Health & Science University Portland OR 97239

TITLE: Functional Genetics for Predisposition to Development of Type 2 Diabetes in Obese Individuals. PRINCIPAL INVESTIGATOR: Assia Shisheva

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Targeted Eradication of Prostate Cancer Mediated by Engineered Mesenchymal Stem Cells

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Short-Term Exercise and Prostate Cancer Prevention in African American Men. CONTRACTING ORGANIZATION: Howard University Washington DC 20059

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Role of CDK5 as a Tumor Suppressor Gene in Non-Small Cell Lung Cancer

AWARD NUMBER: W81XWH TITLE: Gulf War Illness as a Brain Autoimmune Disorder. PRINCIPAL INVESTIGATOR: Apostolos Georgopoulos, MD, PhD

CONTRACTING ORGANIZATION: UNIVERSITY OF ILLINOIS Chicago, IL 60612

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

TITLE: Crosstalk Between Cancer Cells and Bones Via the Hedgehog Pathway Determines Bone Metastasis of Breast Cancer

Transcription:

AD Award Number: W81XWH-11-1-0302 TITLE: Yin and Yang of Heparanase in breast Tumor Initiation PRINCIPAL INVESTIGATOR: Xiulong Xu, Ph.D. CONTRACTING ORGANIZATION: Rush University Medical Center Chicago, IL 60612 REPORT DATE: April 2014 TYPE OF REPORT: Annual PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland 21702-5012 DISTRIBUTION STATEMENT: Approved for Public Release; Distribution Unlimited The views, opinions and/or findings contained in this report are those of the author(s) and should not be construed as an official Department of the Army position, policy or decision unless so designated by other documentation.

REPORT DOCUMENTATION PAGE Form Approved OMB No. 0704-0188 Public reporting burden for this collection of information is estimated to average 1 hour per response, including the time for reviewing instructions, searching existing data sources, gathering and maintaining the data needed, and completing and reviewing this collection of information. Send comments regarding this burden estimate or any other aspect of this collection of information, including suggestions for reducing this burden to Department of Defense, Washington Headquarters Services, Directorate for Information Operations and Reports (0704-0188), 1215 Jefferson Davis Highway, Suite 1204, Arlington, VA 22202-4302. Respondents should be aware that notwithstanding any other provision of law, no person shall be subject to any penalty for failing to comply with a collection of information if it does not display a currently valid OMB control number. PLEASE DO NOT RETURN YOUR FORM TO THE ABOVE ADDRESS. 1. REPORT DATE 2. REPORT TYPE: Annual 3. DATES COVERED April 2014 4. TITLE AND SUBTITLE Yin and Yang of Heparanase in Breast Tumor Initiation 1 April 2013-31 March 2014 5a. CONTRACT NUMBER 5b. GRANT NUMBER W81XWH-11-1-0302 5c. PROGRAM ELEMENT NUMBER 6. AUTHOR(S) Xiulong Xu, Ph.D. E-Mail: xxu@rush.edu 7. PERFORMING ORGANIZATION NAME(S) AND ADDRESS(ES) Rush University Medical Center, Chicago, IL 60612 5d. PROJECT NUMBER 5e. TASK NUMBER 5f. WORK UNIT NUMBER 8. PERFORMING ORGANIZATION REPORT NUMBER 9. SPONSORING / MONITORING AGENCY NAME(S) AND ADDRESS(ES) 10. SPONSOR/MONITOR S ACRONYM(S) U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland 21702-5012 11. SPONSOR/MONITOR S REPORT NUMBER(S) 12. DISTRIBUTION / AVAILABILITY STATEMENT Approved for Public Release; Distribution Unlimited 13. SUPPLEMENTARY NOTES 14. ABSTRACT Heparanase (HPR1) is an endoglycosidase that specifically degrades heparan sulfate proteoglycans, a main constituent on the cell surface and in the extracellular matrix and basement membrane. The role of heparanase in breast cancer tumorigenesis remains unclear. In particular, whether HPR1 enzymatic activity is required for its stimulatory effect on tumor growth and initiation are not fully understood. Our earlier study showed that the C-terminus of HPR1, which lacks the enzymatic activity, was able to accelerate breast cancer formation in a somatic breast cancer mouse model since mice infected with RCAS-Neu virus plus RCAS-8C (a vector encoding the C terminus of HPR1), developed breast cancer significantly faster than that infected with RCAS-Neu plus RCAS-GFP (Green fluorescence protein) control virus. Here we report that the similar observations were made with PyMT oncogene-induced breast cancer, e.g. co-infection with RCAS-PyMT virus plus RCAS-8C virus induced breast cancer significantly faster than RCAS-PyMT virus plus RCAS-GFP. Enzymatically dead HPR1 had similar effect in stimulating breast cancer formation in the C-terminus of HPR1. In contrast, co-infection of TVA transgenic mice with RCAS-PyMT virus plus RCAS-HPR1 virus significantly delayed breast cancer formation than those infected with RCAS-PyMT virus plus RCAS-GFP control virus. Our results strongly suggest that HPR1 promotes tumor growth independent of its enzymatic activity, and that the enzymatic activity of HPR1 antagonizes the tumor initiating activity of HPR1. 15. SUBJECT TERMS 16. SECURITY CLASSIFICATION OF: 17. LIMITATION OF ABSTRACT a. REPORT U b. ABSTRACT U c. THIS PAGE U UU 18. NUMBER OF PAGES 19a. NAME OF RESPONSIBLE PERSON USAMRMC 19b. TELEPHONE NUMBER (include area code)

Table of Contents Page Introduction..... 3 Body 3-4 Key Research Accomplishments... 5 Reportable Outcomes 5 Conclusion 5 References.6 Appendices

Heparanase-1 (HPR1) is an endoglycosidase overexpressed in many malignancies including breast cancer (1; 2). Previous studies suggest that the enzymatic activity of HPR1 can promote tumor angiogenesis and growth by degrading extra cellular matrix and releasing the growth factors. Using a xenograft mouse model, Flux et al. (3) reported that the full-length of the HPR1 gene stimulates tumor growth less effectively than the enzymatically inactive HPR1 gene, suggesting that HRP1 enzymatic activity may be disposable for its effect on stimulating tumor growth. Since the C-terminus of HPR1 can activate the PI-3 kinase pathway and induce endothelial and tumor cell migration independent of its enzymatic activity, it is not clear whether its enzymatic activity or C-terminus or both contribute to breast tumor initiation and growth. The goal of this project is to dissect the effect of HPR1 enzymatic activity and HPR1 C-terminus epitope on breast tumor initiation in a clinically relevant mouse breast cancer model. We proposed to determine if HPR1 knockdownn will suppress or accelerate breast tumor initiation mediated by three oncogenes, PyMT, Neu and Wnt, and whether HPR1 C-terminus or an enzymatically deadd HPR1 can stimulates breast tumor initiation, whereas full- length HPR1 has no effect or is less effective in stimulating breast tumor initiation and progression. The effect of HPR1 on the PI-3 pathway activation. Three RCAS vectors containing a full-length HPR1 gene, an kinase and MAP kinase enzymatic activity-dead HPR1 gene (RCAS-DM-HPR1, double mutations at amino acid residues 225 & 343), and a C-terminus gene fragment (RCAS-8C, with a fusion of 8-kDa and the C- terminus of HPR1 including amino acid residues from 415-543). Alll inserts were tagged with a Myc epitope. This allowed us to titrate virus concentrations and monitor the expression levels in vivo. Western blot analysis with an anti- Myc tag antibody revealed that HPR1 was detected as an 18-kDa protein in DF-1 cells transfected with RCAS-8CC vector, whereas the full-length HPR1 was detected as 50-kDa protein. Immunofluorescence staining revealed that RCAS-HPR1 virus-infected DF-1 heparan sulfate levels, compared to RCAS-8C- infected DF-1 cells. These results confirmed that the C terminus of HPR1 did not have HPR1 enzymatic activity. cells had lower cell surface We next determinedd if the full- length HPR1 and enzymatically inactive HPR1 were functional in stimulating the PI-3 kinase pathway. KAT-18 cells (a thyroid tumor cell line), NIH 3T3 murine Introduction Experim mental procedures and results Fig. 1. HPR1 expression and cell surface HS levels. DF-1 cells were infected with RCAS-HPR1 (left), RCAS-DM-HPR1 (middle) or RCAS- and HPR1-8C. After incubation for 48 hr, the cells were harvested analyzed for HPR1 expression by Western blot with an anti-myc epitope antibody or for cell surface HS levels by staining with an anti- control; HS IgM mab followed by FACS analysis. Green line, isotype Black area, anti-hs IgM. A pakt AKT perk ERK pakt AKT None RCAS 8CC RCAS dhpr1 RCAS HPR1 perk ERK KAT 18 DF 1 NIH 3T3 Fig. 2. The effect of HPR1 on AKT and ERK phosphorylation. KAT-18, DF-1, and NIH-3T3 cells were starved in the medium containing 0.5% FBS overnight. The cells were then left untreated or treated for 30 min with HPR1 or enzymatically dead HPR1 (dhpr1) purified via heparin-agarose beads from the conditioned media of DF1 cells infected with RCAS retroviral vector. AKT and ERK phosphorylation was detected by their specific antibodies. Antibodies against total proteins were used as control. 3

fibroblast cells, and DF-1 chicken fibroblast cells were pre-starved in serum-free medium overnight and then stimulated with purified HPR1 and dhpr1. As expected, both forms of HPR1 induced AKT phosphorylation in KAT-18 cells but were slightly less effective in inducing AKT phosphorylation in two fibroblast cells (Fig. 2). Interestingly, both forms of HPR1 stimulated ERK phospshorylation in KAT-18 cells. Of note, only p42 ERK was detected in chicken fibroblast cells with the anti-erk antibodies. The effect of HPR1 on tumor latency. We originally proposed to use TVA transgenic mice intercrossed with MMTV-PyMT, Neu, or Wnt transgenic mice. We have changed these experimental procedures by co-infecting TVA transgenic mice with RCAS vectors encoding the oncogene PyMT, Neu or Wnt plus the control RCAS vector or the vector encoding RCAS-8C. As shown in Fig. 3, mice infected with RCAS-PyMT plus RCAS-8C vectors developed breast cancer significantly faster than mice infected with RCAS-PyMT plus RCAS-GFP vector. The mean tumor latency in TVA transgenic mice infected with RCAS- Neu plus RCAS-GFP were 86±6 days versus 52±4 days in TVA transgenic mice infected with RCAS-PyMT plus RCAS-8C. Statistical analysis revealed that the C terminus of HPR1 significantly accelerated breast formation induced by PyMT. Survival 1.0 0.8 0.6 0.4 0.2 RCAS-PyMT + RCAS=GFP RCAS-PyMT + RCAS-8C 0.0 0 20 40 60 80 100 120 140 160 Time (days) Fig. 3. HPR1 C-terminus accelerates PyMTinduced breast cancer formation. Female TVA transgenic mice (8-12 weeks old) were infected with RCAS-PyMT plus RCAS-dHPR1- (Black line) or RCAS-Neu plus RCAS-GFP virus (red line) by intraductal injection of 1x10 7 virions each. Mice were monitored for tumor formation by palpation. Percent of tumor-free glands were plotted and statistically analyzed by using Log- Rank test (p=0.002) We next determined if an enzymatically inactive dead HPR1 was able to accelerate breast tumor initiation. As shown in Fig. 4, TVA transgenic mice infected with RCAS-PyMT or RCAS-Neu plus RCASdHPR1 vectors developed breast cancer much faster than mice infected with RCAS-PyMT or RCAS-Neu plus a control RCAS-vector, respectively. The mean tumor latency in TVA transgenic mice infected with RCAS-PyMT plus RCAS-dHPR1 were 89±6 versus 63±5 days in TVA 0.0 0 100 200 300 400 Time (days) transgenic mice infected with RCAS-PyMT plus RCAS-GFP. The mean tumor latency in TVA transgenic mice infected with RCAS-Neu plus RCAS-dHPR1 were 95±11 versus 228±21 days in TVA transgenic mice infected with RCAS-Neu plus RCAS-GFP. These observations strongly suggested that that HPR1 enzymatic activity is disposable for its tumor promoting effect. Survival 1.0 0.8 0.6 0.4 0.2 RCAS-Neu + RCAS-dHPR1 RCAS-Neu + RCAS-GFP Fig. 4. Enzymatically inactive dead HPR1 (dhpr1) accelerates PyMT- and Neuinduced breast cancer formation. (A) Female TVA transgenic mice (8-12 weeks old) were infected with RCAS-PyMT plus RCAS-dHPR1- (Black line) or RCAS-Neu plus RCAS-GFP virus (red line) by intraductal injection of 1x10 7 virions each. (p<0.001) (B) Female TVA transgenic mice (8-12 weeks old) were infected with RCAS-Neu plus RCAS-dHPR1or RCAS-Neu plus RCAS-GFP virus (red line) by intraductal injection of 1x10 7 virions each. Mice were monitored for tumor formation by palpation. Percent of tumor-free glands were plotted and statistically analyzed by using Log-Rank test (p<0.001) 4

Finally, we determine the effect of full-length HPR1 in breast cancer initiation. TVA transgenic mice infected with RCAS-PyMT plus RCAS-HPR1 vectors developed breast cancer significantly slower than mice infected with RCAS-PyMT plus a RCAS-GFP vector. As shown in Fig. 5, the mean tumor latency in TVA transgenic mice infected with RCAS-PyMT plus RCAS- HPR1 were 119±8 days. 1.0 whereas the mean tumor 0.8 latency in TVA transgenic 0.6 mice infected with RCAS- PyMT plus RCAS-GFP were 0.4 86±6. TVA transgenic mice 0.2 co-infected with RCAS-PyMT 0.0 and RCAS-HPR1 significantly prolonged breast cancer Time (days) formation. Studies are ongoing to determine if HPR1 also accelerates Neu-induced breast formation. Survival 0 20 40 60 80 100 120 140 160 180 KEY RESEARCH ACCOMPLISHMENTS: Task 2. To determine whether HPR1 enzymatic activity suppresses breast tumor initiation (Year 2) 1. Demonstrated that HPR1 C-terminus was able to promote breast cancer formation induced by PyMT oncogene 2. Demonstrated that the enzymatic activity of HPR1 had a negative effect on breast cancer formation induced by Neu and PyMT oncogene 3. Demonstrated that the enzymatically inactive HPR1 accelerated breast cancer formation induced by both RCAS-Neu and PyMT oncogenes. REPORTABLE OUTCOMES Manuscript: Domain-specific tumor-promoting activity of heparanase. Manuscript in preparation CONCLUSION RCAS-PyMT+ RCAS-GFP RCAS-PyMT + RCAS-HPR1 (N=32) Fig. 5. HPR1 accelerates breast cancer formation. Female TVA transgenic mice (8-12 weeks old) were infected with RCAS-PyMT plus RCAS-HPR1 (Red line) or RCAS- PyMT plus RCAS-GFP virus (Blue line) by intraductal injection of 1x10 7 virions each. Mice were monitored for tumor formation by palpation. Percent of tumor-free glands were plotted and statistically analyzed by using Log-Rank test (p=0.015) We proposed to determine if HPR1 enzymatic activity can antagonize the tumor promoting effect of the C terminus of HPR1. Our studies using a syngeneic breast cancer model and somatic mouse model suggest that knockdown of HPR1 expression causes the delay of tumorigenesis and the inhibition of tumor growth, suggesting the overall role of HPR1 is to promote tumor growth and formation, and that the C terminus of HPR1 alone was able to promote tumor initiation and growth induced by Neu ongocone in a somatic breast cancer model. Studies in the past year demonstrated that the C-terminus of HPR1 accelerated breast cancer formation induced by PyMT. In contrast, the full-length HRP1 not only delayed breast cancer initiation. In contrast, enzymatically dead HPR1 accelerated breast cancer formation. These results are consistent with our earlier observations that sulodexide, an HPR1 inhibitor, accelerated breast cancer formation. Our results collectively suggest that inhibition of HPR1 activity not only does not suppress but rather accelerate breast cancer initiation, and that the strategy using a specifically inhibitor of HPR1 enzyme could have an adverse effect on cancer relapse. 5

REFERENCES 1. Ilan N, Elkin M, Vlodavsky I: Regulation, function and clinical significance of heparanase in cancer metastasis and angiogenesis. Int J Biochem Cell Biol 38:2018-2039, 2006 2. Gotte M, Yip GW: Heparanase, hyaluronan, and CD44 in cancers: a breast carcinoma perspective. Cancer Res 66:10233-10237, 2006 3. Fux L, Feibish N, Cohen-Kaplan V, Gingis-Velitski S, Feld S, Geffen C, Vlodavsky I, Ilan N: Structurefunction approach identifies a COOH-terminal domain that mediates heparanase signaling. Cancer Res 69:1758-1767, 2009 6