Your browser doesn't support javascript.
loading
Energy Stress-Mediated Cytotoxicity in Tuberous Sclerosis Complex 2-Deficient Cells with Nelfinavir and Mefloquine Treatment.
McCann, Henry D; Johnson, Charlotte E; Errington, Rachel J; Davies, D Mark; Dunlop, Elaine A; Tee, Andrew R.
Afiliação
  • McCann HD; Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK. McCannHD@cardiff.ac.uk.
  • Johnson CE; Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK. johnson.ce2010@gmail.com.
  • Errington RJ; Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK. ErringtonRJ@cardiff.ac.uk.
  • Davies DM; Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK. DaviesDM@cardiff.ac.uk.
  • Dunlop EA; Department of Oncology, South West Wales Cancer Centre, Singleton Hospital, Swansea SA2 8QA, UK. DaviesDM@cardiff.ac.uk.
  • Tee AR; Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff CF14 4XN, UK. DunlopEA@cardiff.ac.uk.
Cancers (Basel) ; 10(10)2018 Oct 10.
Article em En | MEDLINE | ID: mdl-30308940
ABSTRACT
To find new anti-cancer drug therapies, we wanted to exploit homeostatic vulnerabilities within Tuberous Sclerosis Complex 2 (TSC2)-deficient cells with mechanistic target of rapamycin complex 1 (mTORC1) hyperactivity. We show that nelfinavir and mefloquine synergize to selectively evoke a cytotoxic response in TSC2-deficient cell lines with mTORC1 hyperactivity. We optimize the concentrations of nelfinavir and mefloquine to a clinically viable range that kill cells that lack TSC2, while wild-type cells tolerate treatment. This new clinically viable drug combination causes a significant level of cell death in TSC2-deficient tumor spheroids. Furthermore, no cell recovery was apparent after drug withdrawal, revealing potent cytotoxicity. Transcriptional profiling by RNA sequencing of drug treated TSC2-deficient cells compared to wild-type cells suggested the cytotoxic mechanism of action, involving initial ER stress and an imbalance in energy homeostatic pathways. Further characterization revealed that supplementation with methyl pyruvate alleviated energy stress and reduced the cytotoxic effect, implicating energy deprivation as the trigger of cell death. This work underpins a critical vulnerability with cancer cells with aberrant signaling through the TSC2-mTORC1 pathway that lack flexibility in homeostatic pathways, which could be exploited with combined nelfinavir and mefloquine treatment.
Palavras-chave

Texto completo: 1 Bases de dados: MEDLINE Idioma: En Revista: Cancers (Basel) Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Reino Unido

Texto completo: 1 Bases de dados: MEDLINE Idioma: En Revista: Cancers (Basel) Ano de publicação: 2018 Tipo de documento: Article País de afiliação: Reino Unido