01.01 Mimetic sHDL Nanoparticles: a Novel Drug-Delivery Strategy to Target Triple-Negative Breast Cancer

P. T. White1, C. Subramanian1, R. Kuai2, J. Moon2,5, B. M. Timmermann3, A. Schwendeman2, M. S. Cohen1,4 5University Of Michigan,Department Of Biomedical Engineering,Ann Arbor, MI, USA 1University Of Michigan,Department Of Surgery,Ann Arbor, MI, USA 2University Of Michigan,College Of Pharmacy,Ann Arbor, MI, USA 3University Of Kansas,Department Of Medicinal Chemistry,Lawrence, KS, USA 4University Of Michigan,Department Of Pharmacology,Ann Arbor, MI, USA

Introduction: Triple negative breast cancer (TNBC) remains a therapeutic challenge today, highlighting the critical need for discovery and development of safer novel therapies. Withanolides are a unique class of naturally-derived Hsp90 inhibitors that are highly efficacious in preclinical TNBC models, but fairly hydrophobic in vivo. Synthetic high-density lipoprotein (sHDL) nanoparticles have been safely used in clinical cardiology trials and recently shown by our group to conjugate to withanolides to improve drug delivery, solubility, and efficacy in aggressive adrenal cancers in vivo. sHDL provides targeted drug delivery as a ligand to the SR-B1 surface receptor leading to cholesterol uptake and efflux. TNBCs highly overexpress SR-B1 and we hypothesize that combining withanolides with sHDL will enhance their delivery to TNBCs through improved targeting via SR-B1.

Methods: Validated human TNBC cell lines (MD-MBA468LN,MDA-MB231,SUM159) were evaluated for SR-B1 mRNA expression levels by qPCR, and protein levels confirmed by Western blot. Fluorescent labeled sHDL was used to evaluate SR-B1 mediated drug uptake in vitro under fluorescence microscopy, and used for tumor targeting in vivo with whole body IVIS spectrum imaging of mouse xenograft tumors (MD-MBA468LN). Withanolide (WGA-TA) IC50 values were obtained using 72 h CellTiter-Glo (CTG) assays.

Results: All TNBC cell lines had significantly (p<0.01) higher SRB1 expression by qPCR and Western blot compared to fibroblast or Jurkat cells (SUM159 4-5 fold, MDA-MB468LN 8-10 fold, MDA-MB231 2-4 fold). Fluorescent labeled sHDL uptake in vitro demonstrated cytosolic uptake of nanoparticles at significantly (p<0.05) higher levels within the high SR-B1 expressing cell line (MDA-MB468LN) compared to the lower SR-B1 expressing cell line (MDA-MB231). Fluorescent sHDL uptake was almost completely inhibited by receptor saturation through pre-treatment with 10-fold excess sHDL (p<0.05). In vivo tumor uptake of fluorescent labeled sHDL using IVIS spectrum imaging in the MDA-MB469LN mouse xenograft model demonstrated highest radiant efficiency in the tumor, with uptake in the liver where it is metabolized and significantly (p<0.05) lower radiant efficiency in other organs. Using CTG assays, treatment with sHDL up to 20 μM showed no changes to cell viability. In TNBC, IC50 values of WGA-TA vs. sHDL-WGA-TA were not statistically different with IC50 concentrations ranging from 18-125 nM. Both formulations had significantly lower IC50 values when compared to MCR5 control cells (6-41 fold lower; p<0.05).

Conclusion: Conjugation of WGA-TA with sHDL nanoparticles confers improved SR-B1 receptor-mediated targeting both in vitro and in vivo in TNBC without inhibiting the potency of withanolides in these tumor cells. This nanocarrier delivery system warrants further translational evaluation in vivo in patient-derived xenograft of TNBC to determine if improved targeting will confer enhanced treatment benefit and survival.