With the traditional therapies for ovarian cancer (OC) that include surgery followed by cytotoxic chemotherapy patient survival is still extremely poor and 5-year survival rate is less than 30%. Therefore, new treatment strategies are needed to cure or improve dismal patient survival. Recent clinical and experimental evidence including ours indicate that the monocyte-macrophage axis (MMA) and tumor-associated macrophages (TAMs) play a significant role in tumor growth and progression by contributing to angiogenesis, invasion/metastasis, and drug resistance. We found OC patients with increased numbers of circulating monocytes and intratumoral TAMs have poor clinical outcomes and significantly shorter patient survival. Our studies also indicate that the tumor microenvironment and TAMs are attractive therapeutic targets in ovarian and other cancers and reduction of TAMs inhibits angiogenesis, invasion and metastasis, and drug resistance in ovarian cancer models. Overall data indicate that in contrast to the conventional drugs that target only tumor cells, targeting tumor microenvironment is required to achieve maximal anti-tumor efficacy. Also, lack of critical molecular targets for this purpose and together with a lack of effective delivery systems, biological barriers for drugs, including poor tissue-specific delivery, toxicity, and the inability to deliver high concentrations of therapeutics into tumor microenvironment, therapeutic strategies fail to cure OC. The use of siRNA bearing- nanoparticles targeted to the tumor and the microenvironment is a particularly attractive approach for targeting molecular targets, reprogramming the tumor microenvironment and development of the most effective therapeutic strategies for OC. To directly address these unmet needs we recently developed various nanoparticle (NP) platforms including, (1) long-acting, slow-release dual assembly NPs (DANPs); (2) serum resistant AXL-receptor binding-aptamers for development of targeted therapies in OC and validated each NP- based approach in multiple OC tumor models with robust and sustained target silencing and significant antitumor efficacy. In this study we will test the hypothesis that the blockade of the tumor and tumor microenvironment interactions by highly versatile NPs that encase therapeutic cargos will provide significant antitumor activity and enhance the efficacy of current regimens in OC. Thus we will determine mechanism of TAM regulation and determine the biological and therapeutic efficacy of targeting MMA/TAMs using highly targeted dual effect NPs in OC models as well as by applying mathematical models.

Public Health Relevance

Development of innovative and most effective targeted therapies for ovarian cancer is an urgent need due to the fact that this cancer has the poorest patient survival rate <30% for more than 75% of the cases due to lack of appropriate targeted therapies and insensitivity to current regimens. We recently demonstrated that tumor microenvironment especially monocytes and tumor associated macrophages (TAMs) are associated with poor patient survival and developed long-acting slow release dual-assembly nanoparticles (DANP) for siRNA based therapies and highly specific serum-stable AXL-targeted aptamers with significant in vivo efficacy in ovarian cancer models, suggesting that these strategies could be used to develop therapeutic approaches that can target both tumor cells and TAMs and multiple oncogenic targets. We will determine the mechanisms by which TAMs accumulate and polarized to protumorigenic phenotype and validate this highly novel tumor-targeting therapeutic strategies in metastatic ovarian cancer models, and evaluate antitumor efficacy, survival and toxicity in preclinical models alone and in combination with currently approved first-line chemotherapies and eradicate the disease or have significantly improve patient survival.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project--Cooperative Agreements (U01)
Project #
5U01CA213759-02
Application #
9476217
Study Section
Special Emphasis Panel (ZCA1)
Program Officer
Hartshorn, Christopher
Project Start
2017-06-01
Project End
2022-05-31
Budget Start
2018-06-01
Budget End
2019-05-31
Support Year
2
Fiscal Year
2018
Total Cost
Indirect Cost
Name
University of Texas MD Anderson Cancer Center
Department
Internal Medicine/Medicine
Type
Hospitals
DUNS #
800772139
City
Houston
State
TX
Country
United States
Zip Code
77030
Brocato, Terisse A; Coker, Eric N; Durfee, Paul N et al. (2018) Understanding the Connection between Nanoparticle Uptake and Cancer Treatment Efficacy using Mathematical Modeling. Sci Rep 8:7538
Gharpure, Kshipra M; Pradeep, Sunila; Sans, Marta et al. (2018) FABP4 as a key determinant of metastatic potential of ovarian cancer. Nat Commun 9:2923
Koay, Eugene J; Lee, Yeonju; Cristini, Vittorio et al. (2018) A Visually Apparent and Quantifiable CT Imaging Feature Identifies Biophysical Subtypes of Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 24:5883-5894
Chen, Xiuhui; Mangala, Lingegowda S; Rodriguez-Aguayo, Cristian et al. (2018) RNA interference-based therapy and its delivery systems. Cancer Metastasis Rev 37:107-124
Kanlikilicer, Pinar; Ozpolat, Bulent; Aslan, Burcu et al. (2017) Therapeutic Targeting of AXL Receptor Tyrosine Kinase Inhibits Tumor Growth and Intraperitoneal Metastasis in Ovarian Cancer Models. Mol Ther Nucleic Acids 9:251-262