Despite unprecedented gains in the basic understanding of cancer genes, only a few molecular therapies have become standard of care in thirty years. This may reflect the extraordinary molecular heterogeneity of human tumors, but also a relatively narrow path to cancer drug discovery, which typically focuses on individual """"""""drugable"""""""" targets in isolation. Conversely, agents that disable multiple essential networks of tumor maintenance, i.e. pathway inhibitors, may offer broader therapeutic opportunities. One attractive candidate for this approach is the molecular chaperone Heat Shock Protein-90 (Hsp90), which, together with its related molecules, orchestrates pivotal cancer networks of cell proliferation, survival and adaptation. Recently, we identified Shepherdin, a novel peptidomimetic inhibitor of Hsp90. Compared to other Hsp90 antagonists currently in the clinic, Shepherdin exhibits superior anticancer activity in vitro and in vivo, efficacy against heterogeneous tumor cell types regardless of their genetic makeup, and no toxicity for normal tissues. New experimental evidence has now uncovered a complex Shepherdin pathway, underlying this unique anticancer activity. We found that Shepherdin acts on mitochondria, inducing sudden organelle collapse and cell death. At a molecular level, this is due to the inhibition of a pool of Hsp90 chaperones present in mitochondria of tumor cells, and orchestrating a novel pathway of cell survival. In contrast, none of the current Hsp90 antagonists targets the mitochondrial chaperones. Therefore, the hypothesis that Shepherdin is a novel """"""""pathway inhibitor"""""""" with a unique mechanism of anticancer activity compared to all other previously known Hsp90 antagonists can be formulated, and will constitute the focus of the present application. Experiments in the first specific aim will characterize the dynamics of Shepherdin trafficking and import to tumor mitochondria, and dissect the molecular requirements for activation of organelle dysfunction. The second specific aim will elucidate the survival functions of a mitochondrial Hsp90 network in tumor cells, and map their interactions with the molecular machinery of organelle homeostasis. The third specific aim will test the efficacy of Shepherdin in xenograft and transgenic models of drug-resistant disease, metastasis, and angiogenesis, in vivo. The approach combines mechanistic and preclinical studies to elucidate the Shepherdin pathway and its anticancer activity. The results will complement the clinical development of Shepherdin now underway at the National Cancer Institute (NCI), Rapid Access to Intervention Development (RAID) program, and provide a requisite molecular foundation to direct the upcoming testing of this agent in humans.

Public Health Relevance

New anticancer drugs that target fundamental mechanisms of tumor cell maintenance are urgently needed. The present application will characterize the anticancer activity of Shepherdin: a novel and potent inhibitor of the Heat Shock Protein-90 (Hsp90) chaperone network with a unique mechanism of action.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
1R01CA118005-01A2
Application #
7729315
Study Section
Developmental Therapeutics Study Section (DT)
Program Officer
Forry, Suzanne L
Project Start
2009-07-01
Project End
2014-05-31
Budget Start
2009-07-01
Budget End
2010-05-31
Support Year
1
Fiscal Year
2009
Total Cost
$340,386
Indirect Cost
Name
University of Massachusetts Medical School Worcester
Department
Biology
Type
Schools of Medicine
DUNS #
603847393
City
Worcester
State
MA
Country
United States
Zip Code
01655
Chae, Young Chan; Angelin, Alessia; Lisanti, Sofia et al. (2015) Corrigendum: Landscape of the mitochondrial Hsp90 metabolome in tumours. Nat Commun 6:7605
Chae, Young Chan; Angelin, Alessia; Lisanti, Sofia et al. (2013) Landscape of the mitochondrial Hsp90 metabolome in tumours. Nat Commun 4:2139
Vaira, Valentina; Faversani, Alice; Martin, Nina M et al. (2013) Regulation of lung cancer metastasis by Klf4-Numb-like signaling. Cancer Res 73:2695-705
Altieri, Dario C (2013) Hsp90 regulation of mitochondrial protein folding: from organelle integrity to cellular homeostasis. Cell Mol Life Sci 70:2463-72
Altieri, Dario C (2013) Targeting survivin in cancer. Cancer Lett 332:225-8
Tavecchio, Michele; Lisanti, Sofia; Lam, Aaron et al. (2013) Cyclophilin D extramitochondrial signaling controls cell cycle progression and chemokine-directed cell motility. J Biol Chem 288:5553-61
Caino, M Cecilia; Chae, Young Chan; Vaira, Valentina et al. (2013) Metabolic stress regulates cytoskeletal dynamics and metastasis of cancer cells. J Clin Invest 123:2907-20
Altieri, Dario C; Stein, Gary S; Lian, Jane B et al. (2012) TRAP-1, the mitochondrial Hsp90. Biochim Biophys Acta 1823:767-73
Chae, Young Chan; Caino, M Cecilia; Lisanti, Sofia et al. (2012) Control of tumor bioenergetics and survival stress signaling by mitochondrial HSP90s. Cancer Cell 22:331-44
Vaira, V; Faversani, A; Dohi, T et al. (2012) miR-296 regulation of a cell polarity-cell plasticity module controls tumor progression. Oncogene 31:27-38

Showing the most recent 10 out of 29 publications