Drug therapy has thus far attained little success in managing cancers, primarily due to a lack clinical efficacy and presence of drug-induced toxicity. In the previous project, we instituted principles to overcome such obstacles in achieving an ideal drug therapy of maximum therapeutic efficacy with minimal drug-induced toxicity. This drug delivery system (DDS) is comprised of two components: [1] a targeting part composed of heparin linked to a mAb, and [2] a drug part, comprised of a drug coupled with a cell penetrating peptide (CPP). These two components can self-assemble into a tight complex via charge interaction between heparin and CPP. Binding CPP with heparin would mask its cell-penetrating function due to inhibition of its adsorption to the cell surfaces. The drug complex provides a prodrug behavior and prevents CPP-mediated uptake by normal tissues, alleviating drug-induced side effects. Following tumor targeting via the mAb, protamine will be administered to unmask heparin inhibition and restore the activity of the released CPP-Drug, inducing apoptosis to only tumor cells. Exceptional progress was made during the past grant period, with publication of 36 papers and graduation of 12 PhD/Postdoctoral trainees.
All aims were accomplished, and utility/applicability of this DDS was confirmed. Despite promise, two crucial factors - efficacy of antibody targeting and extent of prodrug protection - were found in need of improvement in order to realize a clinically enabled drug therapy. Recent advancement in colorectal cancer research seems to offer a solution to these two issues. First, carcinoembryonic antigen (CEA), a surface antigen known to associate with tumor growth/metastasis, was shown to be overly expressed in colonic cancers but limited in normal tissues. Secondly, we discovered that >85% of the injected T84.66, a mouse mAb against human CEA, accumulated at the colorectal tumor target via an antigen-mediated elimination pathway. Thirdly, the protease legumain was recently reported to express profoundly on the surface of colorectal tumors but is almost undetectable in normal tissues. Using this information, we further engineered a highly improved DDS to cross the afore-cited barriers. Briefly, a potent toxin (gelonin) or siRNA drug will be covalently linked with LMWP, a proven but non-toxic CPP, via S-S bond. This LMWP-Drug will be further linked to T84.66 through a peptide linker containing the legumain-degradable specific AANL sequence. The drug complex would then exhibit a prodrug behavior during tumor targeting, since LMWP is embedded between the antibody and drug molecule, thus unable to transverse the drug through tumor cells in exerting cytotoxic effects. Following tumor accumulation via T84.66 targeting, release of LMWP-Drug from T84.66 will take place on tumor surface via cleavage of the AANL linker by surface-expressed legumain. LMWP-Drug will then internalize into tumor cells via LMWP-mediated cell entry action, initiating tumor apoptosis.
The specific aims are to validate the pre-clinical capability of this system in managing specifically colorectal cancers, through extensive in vitro and in vivo studies using a well-established LS174T xenograft mouse model; thus paving the road for future clinical translation.

Public Health Relevance

Drug therapy has thus far attained little success in managing cancers, primarily due to a lack of clinical efficacy and presence of drug-induced toxicity. In this project, we propose to develop a drug delivery system, which would take advantage of the tumor-specific expression of legumain as the activator of a prodrug that consists of T84.66 antibody, a protein (or siRNA) toxin, and a cell-penetrating peptide. The construction of the DDS is designed to improve treatment efficacy against colorectal cancers, while also reducing systemic toxicities.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA114612-10
Application #
8842594
Study Section
Bioengineering, Technology and Surgical Sciences Study Section (BTSS)
Program Officer
Fu, Yali
Project Start
2005-05-04
Project End
2016-04-30
Budget Start
2015-05-01
Budget End
2016-04-30
Support Year
10
Fiscal Year
2015
Total Cost
Indirect Cost
Name
University of Michigan Ann Arbor
Department
Pharmacology
Type
Schools of Pharmacy
DUNS #
073133571
City
Ann Arbor
State
MI
Country
United States
Zip Code
48109
Shin, Meong Cheol; Min, Kyoung Ah; Cheong, Heesun et al. (2016) Preparation and Characterization of Gelonin-Melittin Fusion Biotoxin for Synergistically Enhanced Anti-Tumor Activity. Pharm Res 33:2218-2228
Shin, Meong Cheol; Zhang, Jian; Min, Kyoung Ah et al. (2015) PTD-Modified ATTEMPTS for Enhanced Toxin-based Cancer Therapy: An In Vivo Proof-of-Concept Study. Pharm Res 32:2690-703
Shin, Meong Cheol; Zhao, Jingwen; Zhang, Jian et al. (2015) Recombinant TAT-gelonin fusion toxin: synthesis and characterization of heparin/protamine-regulated cell transduction. J Biomed Mater Res A 103:409-419
He, Huining; Ye, Junxiao; Wang, Yinsong et al. (2014) Cell-penetrating peptides meditated encapsulation of protein therapeutics into intact red blood cells and its application. J Control Release 176:123-132
Zhang, Jian; Shin, Meong Cheol; Yang, Victor C (2014) Magnetic targeting of novel heparinized iron oxide nanoparticles evaluated in a 9L-glioma mouse model. Pharm Res 31:579-92
Shin, Meong Cheol; Zhang, Jian; Ah Min, Kyoung et al. (2014) Combination of antibody targeting and PTD-mediated intracellular toxin delivery for colorectal cancer therapy. J Control Release 194:197-210
Shin, Meong Cheol; Zhang, Jian; Min, Kyoung Ah et al. (2014) Cell-penetrating peptides: achievements and challenges in application for cancer treatment. J Biomed Mater Res A 102:575-87
He, Huining; Sheng, Jianyong; David, Allan E et al. (2013) The use of low molecular weight protamine chemical chimera to enhance monomeric insulin intestinal absorption. Biomaterials 34:7733-43
Zhang, Jian; Shin, Meong Cheol; David, Allan E et al. (2013) Long-circulating heparin-functionalized magnetic nanoparticles for potential application as a protein drug delivery platform. Mol Pharm 10:3892-902
Min, Kyoung Ah; Shin, Meong Cheol; Yu, Faquan et al. (2013) Pulsed magnetic field improves the transport of iron oxide nanoparticles through cell barriers. ACS Nano 7:2161-71

Showing the most recent 10 out of 57 publications