Despite recent advances in both targeted therapy and immunotherapy, the overall survival for metastatic melanoma remains low. This renewal application is to continue our productive efforts to develop novel tubulin inhibitors and their targeted deliveries for more effective treatment of metastatic melanoma. In the first funding period, we identified several potent scaffolds and developed two nanoparticle formulations that significantly improved in vivo efficacy against melanoma metastasis. Our objectives in this renewal are to perform focused lead optimization using integrated medicinal chemistry, structural biology and molecular biology approaches, and to optimize nanoparticle formulations for high drug loading and nanoparticle kinetic stability.
Aim 1 : Perform focused lead optimization using the two best scaffolds and iteratively optimize their activities.
Aim 1. 1. Using the comprehensive SAR we have accumulated, we will perform focused lead optimization with the two best scaffolds to further improve their interactions with the colchicine binding site.
Aim 1. 2. We will screen new analogs using a panel of melanoma cell lines, including multidrug resistant phenotypes and melanoma colony formation. Melanocytes will serve as a model of normal cells to assess the selectivity. We will select up to the 20 best analogs for Aim 2.
Aim 2 : Confirm the modes of action, determine molecular interactions with tubulin, and assess drug- like properties.
Aim 2. 1. We will determine their ability to inhibit tubulin polymerization, quantitatively assess their binding to the colchicine site in tubulin, investigate their effects on apoptosis, and evaluate their anti- angiogenesis potential.
Aim 2. 2. We will determine molecular interactions between the best analogs and tubulin by solving the X-ray crystal structures of tubulin in complex with a diverse set of potent analogs. These results will feed back to Aim 1 to guide more efficient structural optimization.
Aim 2. 3. We will assess drug-like properties and off-target effects for the selected new analogs to select up to 6 of the best analogs for Aim 3.
Aim 3 : Optimize nanoparticle formulations, determine in vivo efficacy against melanoma tumor growth and metastasis using both free drugs and their optimal nanoparticle formulations.
Aim 3. 1. We will use hydrolyzable bonds to conjugate new analogs to polymer backbones before forming nanoparticles for higher drug loading capacity and improved kinetic stability of nanoparticles. We will incorporate melanoma tumor- specific peptides to the drug loaded nanoparticles for active targeting.
Aim 3. 2 We will evaluate in vivo efficacy against melanoma growth, metastasis, tumor angiogenesis, and potential toxicities. We will compare the efficacy of free drugs and their optimal nanoparticle formulations, using Abraxane (nanoparticle formulated paclitaxel) as the reference control.
Aim 3. 3. For the three most efficacious tubulin inhibitors an their nanoparticle formulations, we will determine their bio-distribution and pharmacokinetic profiles in mice after systemic drug administration.

Public Health Relevance

Currently there is no curative therapy for malignant melanoma. Tubulin inhibitors are successful chemotherapeutic agents but prolonged use of existing anti-tubulin drugs often results in drug resistance and toxicity due to their non-selectiv tissue distributions. We proposed in this grant to continue our productive efforts to develop a new generations of tubulin inhibitors along with their targeted nanoparticle deliveries that can overcome clinically important multidrug resistance problems and reduce systemic toxicity. Completion of the proposed studies will provide several drug-like candidates for future clinical evaluation of use of these compounds as a more efficacious therapy to treat malignant melanoma and other types of cancers.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA148706-07
Application #
9206458
Study Section
Drug Discovery and Molecular Pharmacology Study Section (DMP)
Program Officer
Fu, Yali
Project Start
2011-01-01
Project End
2020-12-31
Budget Start
2017-01-01
Budget End
2017-12-31
Support Year
7
Fiscal Year
2017
Total Cost
Indirect Cost
Name
University of Tennessee Health Science Center
Department
Pharmacology
Type
Schools of Pharmacy
DUNS #
941884009
City
Memphis
State
TN
Country
United States
Zip Code
38103
Chen, Guan; Li, Wei (2018) Editorial Preface for Targeted Cancer Therapy. Acta Pharm Sin B 8:501-502
Bumbaca, Brandon; Li, Wei (2018) Taxane resistance in castration-resistant prostate cancer: mechanisms and therapeutic strategies. Acta Pharm Sin B 8:518-529
Banerjee, Souvik; Arnst, Kinsie E; Wang, Yuxi et al. (2018) Heterocyclic-Fused Pyrimidines as Novel Tubulin Polymerization Inhibitors Targeting the Colchicine Binding Site: Structural Basis and Antitumor Efficacy. J Med Chem 61:1704-1718
Wang, Qinghui; Arnst, Kinsie E; Wang, Yuxi et al. (2018) Structural Modification of the 3,4,5-Trimethoxyphenyl Moiety in the Tubulin Inhibitor VERU-111 Leads to Improved Antiproliferative Activities. J Med Chem 61:7877-7891
Arnst, Kinsie E; Wang, Yuxi; Hwang, Dong-Jin et al. (2018) A Potent, Metabolically Stable Tubulin Inhibitor Targets the Colchicine Binding Site and Overcomes Taxane Resistance. Cancer Res 78:265-277
Yang, Ruinan; Mondal, Goutam; Ness, Rachel A et al. (2017) Polymer conjugate of a microtubule destabilizer inhibits lung metastatic melanoma. J Control Release 249:32-41
Chen, Hao; Lin, Zongtao; Arnst, Kinsie E et al. (2017) Tubulin Inhibitor-Based Antibody-Drug Conjugates for Cancer Therapy. Molecules 22:
Wang, B; Liu, Y; Huang, L et al. (2017) A CNS-permeable Hsp90 inhibitor rescues synaptic dysfunction and memory loss in APP-overexpressing Alzheimer's mouse model via an HSF1-mediated mechanism. Mol Psychiatry 22:990-1001
Patil, Shivaputra A; Addo, James K; Deokar, Hemantkumar et al. (2017) Synthesis, Biological Evaluation and Modeling Studies of New Pyrido[3,4-b]indole Derivatives as Broad-Spectrum Potent Anticancer Agents. Drug Des 6:
Wu, Xiaoxin; Wang, Qinghui; Li, Wei (2016) Recent Advances in Heterocyclic Tubulin Inhibitors Targeting the Colchicine Binding Site. Anticancer Agents Med Chem 16:1325-38

Showing the most recent 10 out of 36 publications