Anticancer effects of niclosamide in human glioblastoma

A Wieland, D Trageser, S Gogolok, R Reinartz… - Clinical Cancer …, 2013 - AACR
A Wieland, D Trageser, S Gogolok, R Reinartz, H Höfer, M Keller, A Leinhaas, R Schelle…
Clinical Cancer Research, 2013AACR
Purpose: Glioblastoma is a highly malignant, invariably fatal brain tumor for which effective
pharmacotherapy remains an unmet medical need. Experimental Design: Screening of a
compound library of 160 synthetic and natural toxic substances identified the antihelmintic
niclosamide as a previously unrecognized candidate for clinical development. Considering
the cellular and interindividual heterogeneity of glioblastoma, a portfolio of short-term
expanded primary human glioblastoma cells (pGBM; n= 21), common glioma lines (n= 5) …
Abstract
Purpose: Glioblastoma is a highly malignant, invariably fatal brain tumor for which effective pharmacotherapy remains an unmet medical need.
Experimental Design: Screening of a compound library of 160 synthetic and natural toxic substances identified the antihelmintic niclosamide as a previously unrecognized candidate for clinical development. Considering the cellular and interindividual heterogeneity of glioblastoma, a portfolio of short-term expanded primary human glioblastoma cells (pGBM; n = 21), common glioma lines (n = 5), and noncancer human control cells (n = 3) was applied as a discovery platform and for preclinical validation. Pharmacodynamic analysis, study of cell-cycle progression, apoptosis, cell migration, proliferation, and on the frequency of multipotent/self-renewing pGBM cells were conducted in vitro, and orthotopic xenotransplantation was used to confirm anticancer effects in vivo.
Results: Niclosamide led to cytostatic, cytotoxic, and antimigratory effects, strongly reduced the frequencies of multipotent/self-renewing cells in vitro, and after exposure significantly diminished the pGBMs' malignant potential in vivo. Mechanism of action analysis revealed that niclosamide simultaneously inhibited intracellular WNT/CTNNB1-, NOTCH-, mTOR-, and NF-κB signaling cascades. Furthermore, combinatorial drug testing established that a heterozygous deletion of the NFKBIA locus in glioblastoma samples could serve as a genomic biomarker for predicting a synergistic activity of niclosamide with temozolomide, the current standard in glioblastoma therapy.
Conclusions: Together, our data advocate the use of pGBMs for exploration of compound libraries to reveal unexpected leads, for example, niclosamide that might be suited for further development toward personalized clinical application. Clin Cancer Res; 19(15); 4124–36. ©2013 AACR.
AACR