A polymeric nanoparticle encapsulated small-molecule inhibitor of Hedgehog signaling (NanoHHI) bypasses secondary mutational resistance to Smoothened antagonists.

TitleA polymeric nanoparticle encapsulated small-molecule inhibitor of Hedgehog signaling (NanoHHI) bypasses secondary mutational resistance to Smoothened antagonists.
Publication TypeJournal Article
Year of Publication2012
AuthorsChenna V, Hu C, Pramanik D, Aftab BT, Karikari C, Campbell NR, Hong S-M, Zhao M, Rudek MA, Khan SR, Rudin CM, Maitra A
JournalMol Cancer Ther
Volume11
Issue1
Pagination165-73
Date Published2012 Jan
ISSN1538-8514
KeywordsAnimals, Cell Line, Tumor, Cell Proliferation, Deoxycytidine, Hedgehog Proteins, Humans, Male, Medulloblastoma, Mice, Mice, Nude, Nanoparticles, Pancreatic Neoplasms, Patched Receptors, Patched-1 Receptor, Receptors, Cell Surface, Receptors, G-Protein-Coupled, Signal Transduction, Smoothened Receptor, Transcription Factors, Tumor Suppressor Protein p53, Xenograft Model Antitumor Assays, Zinc Finger Protein GLI1
Abstract

Aberrant activation of the hedgehog (Hh) signaling pathway is one of the most prevalent abnormalities in human cancer. Tumors with cell autonomous Hh activation (e.g., medulloblastomas) can acquire secondary mutations at the Smoothened (Smo) antagonist binding pocket, which render them refractory to conventional Hh inhibitors. A class of Hh pathway inhibitors (HPI) has been identified that block signaling downstream of Smo; one of these compounds, HPI-1, is a potent antagonist of the Hh transcription factor Gli1 and functions independent of upstream components in the pathway. Systemic administration of HPI-1 is challenging due to its minimal aqueous solubility and poor bioavailability. We engineered a polymeric nanoparticle from [poly(lactic-co-glycolic acid); (PLGA)] conjugated with polyethylene glycol (PEG), encapsulating HPI-1 (NanoHHI). NanoHHI particles have an average diameter of approximately 60 nm, forms uniform aqueous suspension, and improved systemic bioavailability compared with the parent compound. In contrast to the prototype targeted Smo antagonist, HhAntag (Genentech), NanoHHI markedly inhibits the growth of allografts derived from Ptch(-/+); Trp53(-/-) mouse medulloblastomas that harbor a Smo(D477G) binding site mutation (P < 0.001), which is accompanied by significant downregulation of mGli1 as well as bona fide Hh target genes (Akna, Cltb, and Olig2). Notably, NanoHHI combined with gemcitabine also significantly impedes the growth of orthotopic Pa03C pancreatic cancer xenografts that have a ligand-dependent, paracrine mechanism of Hh activation when compared with gemcitabine alone. No demonstrable hematologic or biochemical abnormalities were observed with NanoHHI administration. NanoHHI should be amenable to clinical translation in settings where tumors acquire mutational resistance to current Smo antagonists.

DOI10.1158/1535-7163.MCT-11-0341
Alternate JournalMol. Cancer Ther.
PubMed ID22027695
PubMed Central IDPMC3256300
Grant ListR01 CA113669 / CA / NCI NIH HHS / United States
R01 CA113669-08 / CA / NCI NIH HHS / United States
U54 CA151838 / CA / NCI NIH HHS / United States
U54 CA151838-03 / CA / NCI NIH HHS / United States