Myeloma is the second most common hematologic cancer. It thrives in the bone marrow and aggressively disseminates throughout the skeleton causing osteolysis, debilitating pain and devastating side effects. The tumors eventually become chemoresistant leading to patient death. The Sanderson Lab is dedicated to understanding how heparanase promotes the aggressive behavior of myeloma and to using that knowledge to develop curative therapies for myeloma patients. During the previous funding period, we identified multiple mechanisms through which heparanase drives myeloma progression and metastasis and established heparanase as a viable target for myeloma therapy. Moreover, we catalyzed collaborative efforts aimed at developing and testing novel anti-heparanase drugs and demonstrated their efficacy against myeloma in vivo. Based on our work, one of these inhibitors recently entered clinical trials in myeloma patients. Thus, we accomplished the goals proposed in our previous application including translation of our work to the clinic. The challenge now is t maximize anti-heparanase therapy to enhance tumor killing; a goal that will be attained by a thorough understanding of heparanase mechanisms of action. To this end, we have made two discoveries that provide novel insight into how heparanase regulates myeloma behavior: (i) heparanase enhances the secretion and alters the composition and function of tumor cell-derived exosomes and (ii) heparanase facilitates development of myeloma chemoresistance. These discoveries lay the foundation for studies proposed in this competitive renewal. Our working hypothesis for Aim 1 is that heparanase regulation of exosomes facilitates robust tumor-host crosstalk that drives myeloma progression and metastasis and that these exosomes are targets for anti-heparanase therapy and sources of disease biomarkers. Exosomes will be tested for their impact on behavior of both host and tumor cells using in vitro and in vivo models and anti-heparanase therapy will be tested for its ability to diminish exosome secretion by tumors thereby inhibiting tumor progression. The protein composition of exosomes from myeloma patient sera will be assessed for levels of heparanase, syndecan-1 and other cargo to determine their value as biomarkers of disease. Our working hypothesis for Aim 2 is that heparanase promotes chemoresistance and that use of heparanase inhibitors in tandem with chemotherapeutic drugs will overcome chemoresistance and enhance tumor killing. Using in vitro and in vivo models we will identify which commonly used anti-myeloma drugs exhibit a diminished ability to kill when tumor cells are expressing high levels of heparanase. Once identified, those drugs will be tested in combination with anti-heparanase drugs to determine if this increases tumor killing efficiency in vivo. The proposed work is innovative and significant because it couples the discovery of new heparanase mechanisms of action with the objectives of identifying novel biomarkers, maximizing anti- heparanase therapy and improving patient outcome.

Public Health Relevance

Heparanase is a protein made by cancer cells that plays a major role in helping them grow and spread throughout the body. This project is designed to study novel aspects of heparanase function with the goal of maximizing the use of new anti-heparanase drugs to block cancer growth and spread.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA138340-10
Application #
9477435
Study Section
Tumor Progression and Metastasis Study Section (TPM)
Program Officer
Woodhouse, Elizabeth
Project Start
2009-07-03
Project End
2019-04-30
Budget Start
2018-05-01
Budget End
2019-04-30
Support Year
10
Fiscal Year
2018
Total Cost
Indirect Cost
Name
University of Alabama Birmingham
Department
Pathology
Type
Schools of Medicine
DUNS #
063690705
City
Birmingham
State
AL
Country
United States
Zip Code
35294
Vlodavsky, Israel; Gross-Cohen, Miriam; Weissmann, Marina et al. (2018) Opposing Functions of Heparanase-1 and Heparanase-2 in Cancer Progression. Trends Biochem Sci 43:18-31
Bandari, Shyam K; Purushothaman, Anurag; Ramani, Vishnu C et al. (2018) Chemotherapy induces secretion of exosomes loaded with heparanase that degrades extracellular matrix and impacts tumor and host cell behavior. Matrix Biol 65:104-118
Purushothaman, Anurag; Bandari, Shyam K; Chandrashekar, Darshan S et al. (2017) Chondroitin sulfate proteoglycan serglycin influences protein cargo loading and functions of tumor-derived exosomes. Oncotarget 8:73723-73732
Sanderson, Ralph D; Bandari, Shyam K; Vlodavsky, Israel (2017) Proteases and glycosidases on the surface of exosomes: Newly discovered mechanisms for extracellular remodeling. Matrix Biol :
Higgs, Jerome T; Lee, Joo Hyoung; Wang, Hong et al. (2017) Mesenchymal stem cells expressing osteoprotegerin variants inhibit osteolysis in a murine model of multiple myeloma. Blood Adv 1:2375-2385
Sanderson, Ralph D; Elkin, Michael; Rapraeger, Alan C et al. (2017) Heparanase regulation of cancer, autophagy and inflammation: new mechanisms and targets for therapy. FEBS J 284:42-55
Hao, M; Franqui-Machin, R; Xu, H et al. (2017) NEK2 induces osteoclast differentiation and bone destruction via heparanase in multiple myeloma. Leukemia 31:1648-1650
Purushothaman, Anurag; Bandari, Shyam Kumar; Liu, Jian et al. (2016) Fibronectin on the Surface of Myeloma Cell-derived Exosomes Mediates Exosome-Cell Interactions. J Biol Chem 291:1652-63
Beauvais, DeannaLee M; Jung, Oisun; Yang, Yang et al. (2016) Syndecan-1 (CD138) Suppresses Apoptosis in Multiple Myeloma by Activating IGF1 Receptor: Prevention by SynstatinIGF1R Inhibits Tumor Growth. Cancer Res 76:4981-93
Jung, O; Trapp-Stamborski, V; Purushothaman, A et al. (2016) Heparanase-induced shedding of syndecan-1/CD138 in myeloma and endothelial cells activates VEGFR2 and an invasive phenotype: prevention by novel synstatins. Oncogenesis 5:e202

Showing the most recent 10 out of 34 publications