Award Number: W81XWH TITLE: A Novel Association and Therapeutic Targeting of Neuropilin-1 and MUC1 in Pancreatic Cancer

Similar documents
TITLE: A Novel Association and Therapeutic Targeting of Neuropilin-1 and MUC1 in Pancreatic Cancer

CONTRACTING ORGANIZATION: University of North Carolina at Charlotte Charlotte, NC

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

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

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

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

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

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

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

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

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

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

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

Fort Detrick, Maryland

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

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

CONTRACTING ORGANIZATION: Vanderbilt University Medical Center Nashville, TN

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

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

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

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

CONTRACTING ORGANIZATION: North Eastern Ohio Universities Rootstown OH 44202

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

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: Rush University Medical Center Chicago, IL 60612

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

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

CONTRACTING ORGANIZATION: Johns Hopkins Bloomberg School of Public Health

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

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

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

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

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

La Jolla, CA Approved for Public Release; Distribution Unlimited

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

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

TITLE: Inhibitors of Histone Deacetylases for Radiosensitization of Prostate Cancer

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

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

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

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

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: Dietary Genistein and Prostate Cancer Chemoprevention

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

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

TITLE: Notch in Pathological Angiogenesis and Lymphangiogenesis

TITLE: Genes Associated with Food Allergy and Eosinophilic Esophagitis

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

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

TITLE: The Role of Constitutively Active Prolactin Receptors in the Natural History of Breast Cancer

Targeting of the MUC1-C Oncoprotein in Colitis-Associated Colorectal Cancer

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

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

CONTRACTING ORGANIZATION: Massachusetts General Hospital Boston, MA

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

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

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

CONTRACTING ORGANIZATION: University of Southern California Los Angeles, CA 90033

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

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

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

Award Number: W81XWH

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

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

TITLE: Harnessing GPR17 Biology for Treating Demyelinating Disease

TITLE: Uncarboxylated Osteocalcin and Gprc6a Axis Produce Intratumoral Androgens in Castration- Resistant Prostate Cancer

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

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

RECIPIENT: Lankenau Institute for Medical Research, Wynnewood, PA 19096

Award Number: W81XWH TITLE: Dependency on SRC-Family Kinases for Recurrence of Androgen- Independent Prostate Cancer

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

TITLE: Uncarboxylated Osteocalcin and Gprc6a Axis Produce Intratumoral Androgens in Castration-Resistant Prostate Cancer

TITLE: Harnessing GPR17 Biology for Treating Demyelinating Disease

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

TITLE: Genetic and epigenetic biomarkers for recurrent prostate cancer after radiotherapy

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

TITLE: MicroRNA in Prostate Cancer Racial Disparities and Aggressiveness

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

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: University of Mississippi Medical Center Jackson, MS 39216

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

CONTRACTING ORGANIZATION: Fred Hutchinson Cancer Research Center Seattle, WA 98109

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

Award Number: W81XWH

Roles of microrna-mediated Drug Resistance in Tumor Stem Cells of Small Cell Lung Carcinoma

AD (Leave blank) TITLE: Trial of Naltrexone and Dextromethorphan for Gulf War Veterans Illnesses

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

TITLE: Central Tolerance Blockade to Augment Checkpoint Immunotherapy in Melanoma

Co-PI: Amy Trentham-Dietz, PhD

Page 1 AWARD NUMBER: W81XWH TITLE: A Novel Pleiotropic Anti-Inflammatory Drug to Reduce ARDS Incidence. PRINCIPAL INVESTIGATOR: Gary Nieman

TITLE: A Novel Immune-Intact Mouse Model of Prostate Cancer Bone Metastasis: Mechanisms of Chemotaxis and Bone Colonization

Transcription:

AD Award Number: W81XWH-12-1-0220 TITLE: A Novel Association and Therapeutic Targeting of Neuropilin-1 and MUC1 in Pancreatic Cancer PRINCIPAL INVESTIGATOR: Pinku Mukherjee CONTRACTING ORGANIZATION: University of North Carolina at Charlotte Charlotte, NC 28223-0001 REPORT DATE: October 2013 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. 1

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 October 2013 2. REPORT TYPE Annual 3. DATES COVERED 30September2012 29September2013 4. TITLE AND SUBTITLE 5a. CONTRACT NUMBER W81XWH-12-1-0220 A Novel Association and Therapeutic Targeting of Neuropilin-1 and MUC1 in Pancreatic Cancer 5b. GRANT NUMBER W81XWH-12-1-0220 5c. PROGRAM ELEMENT NUMBER 6. AUTHOR(S) 5d. PROJECT NUMBER Jennifer Curry, Ru Zhou, Pinku Mukherjee E-Mail: pmukherj@uncc.edu 7. PERFORMING ORGANIZATION NAME(S) AND ADDRESS(ES) University of North Carolina at Charlotte Charlotte, NC 28223-0001 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 We hypothesized that MUC1, a transmembrane glycoprotein that is overexpressed in >80% of pancreatic ductal adenocarcinoma induces a pro-angiogenic tumor microenvironment by increasing the level of NRP-1 and VEGF thereby enhancing angiogenesis and metastasis. We report that MUC1 hi PC cells and tumors in vitro and in vivo not only express higher levels of NRP1 but also express higher levels of VEGFR2, pvegfr2 1175, pvegfr2 996, and VEGFR3 as well as secrete higher levels of VEGF than MUC1 low PC cells. This enables the MUC1 hi /NRP hi cells to generate long ectopic blood vessels and enhanced distant metastasis. In the proposal, we also hypothesized that blocking the interaction between VEGF 165 and NRP-1 within the tumor microenvironment will lead to therapeutic benefit. Indeed, in vivo blocking NRP1 significantly reduces tumor burden in the MUC1 hi tumors. In fact 4 out of 7 mice had a complete response, 2 had small tumors and 1 mouse did not respond and developed large tumor. This treatment had no significant effect on the MUC1 low tumors. This is not surprising since MUC1 low tumors have very low levels of NRP1 and VEGFR2. It should be noted that these MUC1 low tumors do however grow but may eventually form stable disease. Thus, we conclude that NRP1 may be a promising target for MUC1 hi PC and in the future conjugating to the MUC1 antibody for targeted delivery may enhance its potency. 15. SUBJECT TERMS Neuropilin-1, MUC1, Angiogenesis, Pancreatic Cancer 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) 2

Table of Contents Page Introduction.....4 Body..4-16 Key Research Accomplishments...17 Reportable Outcomes 17 Conclusion 18 References.18 3

Introduction: Pancreatic cancer is a lethal disease. We hypothesize that MUC1 induces a pro-angiogenic tumor microenvironment by increasing the level of NRP-1, thus enhancing angiogenesis, disease progression and metastases. Second, blocking the interaction between VEGF and NRP-1 within the tumor 165 microenvironment will lead to therapeutic benefit. We report progress for all three tasks that we had proposed. I have thus broken the report down by tasks. The objective and specific aims are stated below. This is followed by the individual tasks with the progress made for that task. We show 11 figures in total. Figures 1-3 are for Task 1, Figures 4-7 are for Task 2, and Figures 8-11 are for Task 3. Objectives/Specific Aims: Task 1: To test the effects of MUC1+ve PC cells on in vitro endothelial cell function in an NPR-1-dependent fashion. To determine if MUC1 up-regulates NRP-1 and creates a pro-angiogenic niche in vivo. To directly target angiogenesis within the tumor microenvironment by using TAB 004 MUC1 antibody conjugated to a peptide blocking VEGF-NPR-1 binding. 1) To test the effects of MUC1 + PC cells on in vitro endothelial cell function in an NRP-1- dependent fashion (0-9 months). For this task we perform in vitro angiogenesis assays to assess the ability of MUC1-expressing pancreatic cells to enhance endothelial cell proliferation, invasion and tube formation via NRP-1 Thus, we first determined if a correlation exists between in MUC1 and NRP1 in a panel of human PDA cell lines that endogenously express high or low MUC1. Next we determined if MUC1 regulates NRP1 expression by conducting gain of function (stably transfected full length MUC1 in MUC1-low cells) and loss of function studies (by knocking down MUC1 using MUC1 specific sirna). Figure 1A clearly shows that cells expressing high endogenous MUC1 also have high NRP1 while cells with low endogenous MUC1 has low NRP with a few exceptions (Su86.86 and Panc1). We show that overexpressing full length MUC1 in two separate MUC1low cell lines (BxPC3 and Panc) can induce higher expression of NRP1 (Figure 1B) while knocking down MUC1 from two other MUC1hi cell lines (CFPAC and HPAFII) can reduce NRP1 expression. Whether this regulation is direct or indirect is not yet delineated. Next we studied whether the MUC1hi/NRP1hi cells affects formation of ectopic blood vessel (number and length) in a zebra fish embryo model of angiogenesis [1] [2]. We show for the first time that human PDA cell line BxPC3.MUC1 induces significantly more and longer vessels in vivo in a zebra fish embryo model than its counterpart BxPC3.Neo cells (Figure 2A-C). Furthermore, we show that BxPC3.MUC1 tumors are highly invasive and form significantly more metastasis compared to BxPC3.Neo tumors (Figure 3A and B) clearly suggesting that the tumors with high MUC1 can enhance proliferation by affecting vessel formation. We will confirm this result in an in vitro tube formation assay. However we believe that the in vivo assay is more relevant and does not depend on the matrigel and the type of endothelial cells used. 4

Figure 1: High MUC1 expressing PC cells have high expression of NRP1 A) T3M4, BxPC3.MUC1, Capan2, CFPAC, HPAFII, and HPAC express moderate/high levels of MUC1. These MUC1-hi cells all express moderate to high levels of NRP1. Capan1, MiaPaca2, BxPC3, Su86.86, HS766T, Panc1, and Panc2 cells have low/intermediate levels of MUC1. Other than Su86.96 and Panc1, these low MUC1 cells also express low levels of NRP1. B) When BxPC3 and Panc02 cells were stably transfected with full length MUC1, NRP 1 expression was substantially increased. C) Similarly, when MUC1 was down regulated in HPAC and CFPAC cells using sirna, NRP1 levels were significantly downregulated. B-actin served as control for equal loading of protein. A. B. C MUC1 BxPC3.Neo BxPC3.WT BxPC3.MUC1 Pan02.MUC1 Panc02.Neo Untreated HPAC Control sirna MUC1siRNA 48h MUC1siRNA Untreated CFPAC Control sirna MUC1siRNA 48h MUC1siRNA MUC1 NRP1 NRP1 β-actin β-actin 5

Figure 2: MUC1 expressing BxPC3 induced tumor angiogenesis in zebrafish tumor xenograft model. A) Representative image of zebrafish embryo with formation of ectopic blood vessels. B. Quantitative measurement of number of ectopic vessels formed in the embryo C. Quantitative measurement of the length of the ectopic vessels formed in the embryo (n=100 embryos each). t test*p=0.005 between BxPC3.Neo and BxPC3.MUC1. A. Ctrl-No cells BXPC3-Neo BXPC3-MUC1 B. C. # of ectopic vessels in embryo 7 6 5 4 3 2 1 0 No cells BxPC3.Neo BxPC3.MUC1 * Length of ectopic vessels in embryo 600 500 400 300 200 100 0 * No cells BxPC3.Neo BxPC3.MUC1 6

Figure 3: MUC1 overexpression in BxPC3 promotes tumor metastasis in zebrafish/tumor xenograft. A. Represenetative image of zebra fish embryo with metastatic lesions. B. Quantitative measurement of % metastasis in xenografted tumor cells. Table shows number of embryos and the significance. A. No Metastasis with xenograft Metastasis with BxPC3-Neo xenograft Metastasis with BxPC3-MUC1 xenograft B. % metastasis in xenograft embryos 40.0% 35.0% 30.0% 25.0% 20.0% 15.0% 10.0% 5.0% 0.0% BXPC3-Neo * BXPC3-Muc1 Cell Line %Metastasis SEM BxPC3-Neo 5.71% 2.97% 88 BxPC3-Muc1 31.28% 4.36% 104 3-group total embryos t-test (Neo vs. Muc1) 0.0042 p 0.01 7

Task 2: To determine if MUC1 up-regulates NRP-1 and creates a pro-angiogenic niche in vivo (0-18 months). For Task 2, we will use our mouse models of pancreatic cancer to determine if MUC1- expressing pancreatic tumors have enhanced intra-tumoral levels of NRP-1 and angiogenesis. We first demonstrate that expression of NRP1 is significantly higher in the spontaneously arising PDA tumors in WT (Muc1 intact) versus PDA in Muc1 null mice (Figure 4A) [3]. Cell lines from these tumors were developed and NRP1 and Muc1 levels tested by western blotting and flow cytometry (Figure 4B and C) and confirm the data in Figure 4A. We then determined that knocking down NRP1 with specific sirna significantly reduces proliferation only in Muc1 expressing KC cells but not in KCKO cells suggesting for the first time that Muc1-ve tumors are not dependent on NRP1 for their growth but Muc1+ve tumors are (Figure 4D and E). To further elucidate the mechanism, we determined the other VEGF receptor levels as well as secreted VEGF levels in these cells. This is especially important as NRP1 is only a co-receptor of VEGF and signaling through VEGFR is critical for the angiogenic signaling to occur [4]. We show that KCKO (Muc1 null) cells have moderately lower levels of VEGF receptor 1, 2, and 3 than its counterpart KC cells which have an intact Muc1 (Figure 5A). Further, KC cells secrete significantly higher levels of VEGF than KCKO cells (Figure 5B). It must be noted that in human cell line BxPC3.MUC1, there was only an increase in VEGFR 2 from its counterpart BxPC3.Neo. There was no effect on levels of VEGFR1 and VEGFR3 was absent in these cells (Figure 5C). We have already shown previously that BxPC3.MUC1 cells express high levels of several pro-angiogenic factors compared to BxPC3.Neo cells [5]. Why this difference between mouse and human cells in VEGFR is not clear at this time. NRP1 levels are always lower in Muc1/MUC1 low cells compared to Muc1/MUC1hi cells. Next we studied if supernatant from these KC and KCKO cells can differentially regulate the NRP1 levels of murine endothelial cells, 2H11. As shown in Figure 6A, 2H11 cells endogenously express high levels of NRP1 and VEGFR1 but low levels of VEGFR2 and 3 (Figure 6A). However, stimulating with supernatant had no effect on the NRP1 levels as tested by flow cytometry (Figure 6B and C). We did not test the other VEGF receptors and are planning to conduct those analyses soon. Other endothelial cell lines will also be tested similarly. Even though it might not affect NRP1 levels, it is possible that there will be differential angiogenic/oncogenic signaling. VEGFR-2, the major positive signal transducer for both physiological and pathological angiogenesis is selectively expressed on vascular endothelial cells. The binding of VEGF to its receptors induces dimerization and subsequent receptor phosphorylation, which then leads to the activation of several intracellular downstream signaling pathways promoting angiogenesis [6]. Thus we looked at the levels of VEGFR2 phosphorylation in vivo in KC and KCKO tumors. It was of great interest to demonstrate that not only are the levels of NRP1 and VEGFR2 significantly lower in the KCKO tumors, the phosphotylation at tyrosines 1175, and 996 are significantly low (Figure 7). This is of course because the receptor level itself is low but phosphorylation of tyrosine 996 is completely absent in KCKO tumors as compared to the KC tumors suggesting that KCKO tumors that lack MUC1 regulate NRP1 and VEGFR2 and thereby downregulate the angiogenic signaling. The question still remains whether the low receptors are due to low endothelial cells and low vessels or due to the tumor cells themselves as NRP1 and VEGFR are expressed on both the epithelial and the endothelial cells. This will be studied in great detail in the coming year. This also explains why KC cells were the only ones that responded to NRP sirna and showed significantly lower proliferation in vitro (Figure 4E). This is significant since NRP1 binds VEGF-A 165 and only when co-expressed with VEGFR2, it enhances the binding of VEGF-A 165 to VEGFR-2 by 4- to 6-fold and results in sustained vascular permeability, inflammation, and endothelial cell migration. 8

3 Figure 4: A) In vivo expression of NRP1 and Muc1 from KC and KCKO tumors (spontaneously arising PDA developing in WT (intact Muc1) or Muc1 null mice respectively. KC tumors have higher NRP1 compared to KCKO tumors. B) In vitro expression of NRP1 in KC and KCKO cell lines. KC cells have higher expression of NRP1 than KCKO cells and expression increases with increasing confluency. C) In vitro expression of Muc1 by flow cytometry using CT2 antibody. KCKO is null for Muc1 and KC has high levels of Muc1. D) KC and KCKO cells were subjected to NRP1 specific sirna and NRP1 levels determined at 48 and 72h post treatment by western blotting. E) KC cells treated with NRP-siRNA had significantly lower proliferation than control sirna treated KC cells as determined by 3H-thymidine assay (*p<0.01). KCKO cells had no response to the peptide. A B C +ve KCKO KC NRP1 MUC1 KCKO KC 0.5M 1M 2M 0.5M 1M 2M NRP1 % of Max 100 80 60 40 20 unstain KC KCKO b-actin b-actin 0 0 10 2 10 3 10 4 10 5 Muc1 (CT2) D E Unt Ctl KC NRP1 sirna 48h 72h Unt KCKO NRP1 sirna Ctl 48h 72h 3H-Thymidine Uptake (CPM) 90000 80000 70000 60000 50000 40000 30000 20000 10000 Ctl sirna NRP1 sirna * 0 KCKO KC 9

Figure 5: A) Along with NRP1, KC cells express higher levels of VEGF receptors 1, 2, and 3 and B) KC cells secrete higher levels of VEGF than KCKO cells (p<0.0001). C) BxPC3.MUC1 cells also express higher levels of VEGFR2 and NRP1. TAB 004 and CT2 are two MUC1 monoclonal antibodies that detect the extracellular tandem repeat of MUC1 and the cytoplasmic tail respectively. TAB 004 does not recognize mouse Muc1 and therefore not used for murine cell lines. KC and KCKO cells were plated in medium with 10% FBS in 6-well plates overnight. Media was replaced with fresh medium without FBS. After 24hr culture, supernatants were collected and VEGF concentrations determined by ELISA. A B C BxPC3 Neo MUC1 VEGFR1 KC KCKO VEGFR2 VEGFR1 VEGFR3 VEGFR2 NRP-1 VEGFR3 * TAB 004 NRP-1 * CT-2 * CT-2 b-actin b-actin Figure 6: A) Western blot analysis of murine endothelial cell line 2H11. These cells express high levels of NRP1 and VEGFR1 B) Expression of NRP1 is confirmed by flow cytometry. C) Tumor derived conditioned medium from KC or KCKO cells did not alter levels of NRP1 in 2H11 cells as determined by flow cytometry. A B C VEGFR1 VEGFR2 VEGFR3 % of Max 100 80 60 40 20 unstain 2H11 % of Max Isotype KC-5x10 5 cells sup KCKO-5x10 5 cells sup NRP-1 b-actin 0 0 10 2 10 3 10 4 10 5 NRP-1 NRP-1 10

Figure 7: KCKO tumors in vivo display lower level of NRP1 and VEGFR2 and its phosphorylations in vivo. Western blotting with the indicated antibodies: N=6 KC mice and 8 KCKO mice tumors were evaluated. KC #1 KC #2 KC #3 KC #4 KC #5 KC #6 KCKO #1 KCKO #2 KCKO #3 KCKO #4 KCKO #5 KCKO #6 KCKO #7 KCKO #8 VEGFR2 (Y1175) VEGFR2 (Y996) VEGFR2 (Y951) VEGFR2 NRP-1 b-actin 11

Task 3: To directly target angiogenesis within the tumor microenvironment by using TAB 004 conjugated to an inhibitory peptide that blocks VEGF-NPR-1 interaction (12-24 months). In this aim, we will attempt targeted drug delivery using an antibody specific for tumor-associated MUC1 (TAB 004) conjugated to a peptide inhibitor of VEGF-NRP-1 binding. Before we injected the cells in vivo, we first confirmed that BxPC3.MUC1 tumor cells proliferate at a significantly higher rate than BxPC3.Neo (Figure 8A). Next we show that the tumors in vivo recapitulate the in vitro finding that MUC1-expressing cells express more NRP1 and VEGF (Figure 8B). Finally tumor bearing mice were treated with the NRP1 blocking peptide. Fourteen nude mice injected with 3 x10 6 BxPC3 Neo or MUC1 cells. After 2 weeks, we randomized groups and started therapy with A7R - NRP1 blocking peptide - 20mg/kg injected subcutaneously 3 times a week for 5 weeks, PBS served as the vehicle control. Mice bearing BxPC3.MUC1 tumors responded well to the mono-therapy with significantly lower tumor burden between treated and untreated mice and with 4 out of 7 mice showing a complete response. In contrast the BxPC3.Neo tumors did not respond as well to the therapy with no significant change in the tumor burden between treated and vehicle treated mice (Figure 8). 12

Figure 8: Blocking with NRP blocker significantly reduced tumor growth in BxPC3.MUC1 tumors but had no effect on BxPC3.Neo tumors. A) 3H-Thymidine assay showing higher proliferation of BxPC3.MUC1 cells (t-test: * P=0.06, **p<0.05, ***p<0.01) B) Western blotting and densitometric analysis of NRP1 levels and VEGF levels in vivo in the BxPC3 tumor xenografts in nude mice. BxPC3.MUC1 tumors have higher levels of NRP1 and VEGF than BxPC3.Neo tumors. C) NRP-1 blocking peptide causes significant reduction in BxPC3.MUC1 tumor burden (p=0.07). Four out of seven mice showed a complete response. A B C * P=0.07 13

To determine what downstream factors may be altered due to the ineffective NRP1/VEGF signaling axis and contribute to the difference observed in angiogenesis between MUC1hi and low cells, we conducted a multiplex bioassay on tumor derived conditioned media. The most change was observed in Leukocyte Inhibitory Factor (LIF) (Figure 9). LIF inhibits angiogenesis in vitro, an effect that can be correlated with a LIF-mediated decrease in endothelial cell proliferation, migration and extracellular proteolysis [7]. LIF was lower both in the supernatant of BxPC3.MUC1 and HPAF cells compared to BxPC3.Neo and HPAF cells treated with MUC1siRNA. In contrast, MMP2 was higher in the BxPC3.MUC1 cell supernatant. This data is significant as it is the first indication of LIF being involved in tumor progression and angiogenesis in pancreatic cancer. Figure 9: Multiplex analysis of the supernatants showed decreased LIF and increased MMP2 in BxPC3.MUC1 versus BxPC3.Neo cells. Similar trend was seen in HPAF cells treated with control and MUC1 sirna. Average of two separate experiments are shown. Only statistical significant differences (p<0.05) are reported in the multiplex analysis. Thus the difference is significant in the data shown in the figure. LIF (ngs/ml) 70 60 50 40 30 20 10 0 BxPC3.Neo BxPC3.MUC1 6h 12h 48h LIF ( ngs/ml) 180 160 140 120 100 80 60 40 20 0 HPAF MUC1siRNA HPAF CTl sirna 6h 12h 24h MMP2 (ngs/ml) 25 20 15 10 5 0 BxPC3.Neo BxPC3.MUC1 6h 12h 24h 48h Finally, with regard to targeted drug delivery using the TAB 004 MUC1 monoclonal antibody, we first show that TAB 004 when injected intra peritoneal and intravenously specifically reaches the tumor within 24hours (Figure 10). With iv injection, we observe some reaching the spleen. Therefore, for our future experiments, we plan to inject the antibody intraperitoneal. Second, we show by in vitro confocal analysis that TAB 004 is internalized into KCM cells through endocytosis (Figure 11). Thus, we have shown that the blocking peptide is effective in vivo and that targeting antibody internalizes in the cells and reaches the tremor bed in vivo. Now we are ready to conjugate the blocking peptide to the antibody and start the treatment in the following year. 14

Figure 10: Localization of TAB004 in mice bearing the MUC1+ve tumor. Mice were injected with biotin-tab004 (50ugs/mouse) via the ip, it, and iv route. After 24hrs, mice were euthanized and TAB004 binding detected by IHC probing with streptavidin-hrp. Brown staining indicates antibody binding. 15

Figure 11: TAB004 internalizes in KCM cells. KCM cells were plated on chamber tissue culture slides. Cell surface was labeled green with Cell Light Plasma Membrane-GFP. BacMam 2.0. Lysozyme organelles were labeled red with Lysotracker Deep Red. TAB004 labeled with yellow was incubated with the cells at 1ugs/ml concentration for 2h. Slides were fixed with ProLong Gold Antifade with DAPI and imaged with confocal microscopy. DAPI Plasma Membrane MUC1 Endosome Merged 16

Key Research Accomplishments Establishment of the zebra fish angiogenic and metastatic assay MUC1 in pancreatic cancer cells regulate levels of NRP1, VEGFR and angiogenic signaling. VEGFR, NRP and phosphorylation of VEGFR2 tyrosines is reduced in MUC1-ve tumors LIF may be one of the underlying factors contributing to inhibition of angiogenesis in MUC1-ve cells MUC1+ve tumors respond to NRP1 blocking peptide but MUC1-ve cells do not. This suggests that that MUC1 could be a companion diagnostic for anti-angiogenic therapy targeting NRP1. MUC1 antibody (TAB 004) internalizes in the MUC1+ve tumor cells through endocytosis suggesting that the drug will be internalized when conjugated to TAB 004. Established the four color confocal images for plasma membrane, lysosomes, MUC1, and DAPI TAB 004 localizes to the tumor when injected intraperitoneal and intravenous within 24hours. Reportable Outcomes Two manuscripts are in preparation, 1) The effect of NRP1-VEGF signaling axis on MUC1 +ve epithelial tumor cells and 2) The effect of NRP1-VEGF signaling axis in endothelial cells within MUC1+ve tumors. Abstracts are being prepared for submission to the AACR 2014 meeting next month. It will be presented in April of 2014. Postdoctoral fellow hired for the project, Graduate students are working on parts of the project and in the future will get publications and degrees out of this work. Patient pancreatic cancer serum and tissue collected for future analysis. KC, KCM, and KCKO cells are further characterized due to this project We plan to submit an RO1 in February of 2014 based on data from this project, especially to understand how LIF plays a role in tumor progression and metastasis Development of a novel antibody-anti-angiogenic drug conjugate that can be used in immune competent mouse model of spontaneous PC and can be potentially translated to human clinical trials. 17

Conclusion In conclusion, we show that NRP1 may be an excellent target for treating MUC1+ve PDA but not MUC1-ve PDA. The mechanistic role of LIF needs to be further studied. If LIF indeed is the underlying anti-angiogenic signal for MUC1-ve pancreatic cancer, we can envision the potential therapeutic application of LIF1 therapy. It is doubtful that a monotherapy with NRP1 blockade will work and therefore using the TAB 004-conjugated to the drug may show promise due to ADCC caused by the antibody and thereby making the tumors more vulnerable to NRP1 blockade. In the long term, combination therapy with standard of care drug and NRP1 blockade conjugated to TAB 004 or two drugs packaged in nanoparticles and conjugated to the antibody might be highly efficacious for patients with pancreatic cancer. References 1. Serbedzija GN, Flynn E, Willett CE: Zebrafish angiogenesis: a new model for drug screening. Angiogenesis 1999, 3(4):353-359. 2. Norrby K: In vivo models of angiogenesis. J Cell Mol Med 2006, 10(3):588-612. 3. Besmer DM, Curry JM, Roy LD, Tinder TL, Sahraei M, Schettini J, Hwang SI, Lee YY, Gendler SJ, Mukherjee P: Pancreatic ductal adenocarcinoma mice lacking mucin 1 have a profound defect in tumor growth and metastasis. Cancer Res 2011, 71(13):4432-4442. 4. Ferrara N, Gerber HP, LeCouter J: The biology of VEGF and its receptors. Nat Med 2003, 9(6):669-676. 5. Roy LD, Sahraei M, Subramani DB, Besmer D, Nath S, Tinder TL, Bajaj E, Shanmugam K, Lee YY, Hwang SI et al: MUC1 enhances invasiveness of pancreatic cancer cells by inducing epithelial to mesenchymal transition. Oncogene 2011, 30(12):1449-1459. 6. Sinha S, Vohra PK, Bhattacharya R, Dutta S, Mukhopadhyay D: Dopamine regulates phosphorylation of VEGF receptor 2 by engaging Src-homology-2-domain-containing protein tyrosine phosphatase 2. J Cell Sci 2009, 122(Pt 18):3385-3392. 7. Pepper MS, Ferrara N, Orci L, Montesano R: Leukemia inhibitory factor (LIF) inhibits angiogenesis in vitro. J Cell Sci 1995, 108 ( Pt 1):73-83. 18