1. Academic Validation
  2. Actionable cancer vulnerability due to translational arrest, p53 aggregation and ribosome biogenesis stress evoked by the disulfiram metabolite CuET

Actionable cancer vulnerability due to translational arrest, p53 aggregation and ribosome biogenesis stress evoked by the disulfiram metabolite CuET

  • Cell Death Differ. 2023 May 4. doi: 10.1038/s41418-023-01167-4.
Dimitris C Kanellis 1 Asimina Zisi 2 Zdenek Skrott 3 Bennie Lemmens 2 Jaime A Espinoza 2 Martin Kosar 2 Andrea Björkman 2 Xuexin Li 2 Stefanos Arampatzis 4 Jirina Bartkova 2 4 Miguel Andújar-Sánchez 5 Oscar Fernandez-Capetillo 2 6 Martin Mistrik 3 Mikael S Lindström 2 Jiri Bartek 7 8
Affiliations

Affiliations

  • 1 Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21, Stockholm, Sweden. dimitris.kanellis@ki.se.
  • 2 Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21, Stockholm, Sweden.
  • 3 Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic.
  • 4 Danish Cancer Society Research Center, DK-2100, Copenhagen, Denmark.
  • 5 Pathology Department, Complejo Hospitalario Universitario Insular, Las Palmas, Gran Canaria, Spain.
  • 6 Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain.
  • 7 Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, S-171 21, Stockholm, Sweden. jb@cancer.dk.
  • 8 Danish Cancer Society Research Center, DK-2100, Copenhagen, Denmark. jb@cancer.dk.
Abstract

Drug repurposing is a versatile strategy to improve current therapies. Disulfiram has long been used in the treatment of alcohol dependency and multiple clinical trials to evaluate its clinical value in oncology are ongoing. We have recently reported that the disulfiram metabolite diethyldithiocarbamate, when combined with copper (CuET), targets the NPL4 adapter of the p97VCP segregase to suppress the growth of a spectrum of Cancer cell lines and xenograft models in vivo. CuET induces proteotoxic stress and genotoxic effects, however important issues concerning the full range of the CuET-evoked tumor cell phenotypes, their temporal order, and mechanistic basis have remained largely unexplored. Here, we have addressed these outstanding questions and show that in diverse human Cancer cell models, CuET causes a very early translational arrest through the integrated stress response (ISR), later followed by features of nucleolar stress. Furthermore, we report that CuET entraps p53 in NPL4-rich aggregates leading to elevated p53 protein and its functional inhibition, consistent with the possibility of CuET-triggered cell death being p53-independent. Our transcriptomics profiling revealed activation of pro-survival adaptive pathways of ribosomal biogenesis (RiBi) and Autophagy upon prolonged exposure to CuET, indicating potential feedback responses to CuET treatment. The latter concept was validated here by simultaneous pharmacological inhibition of RiBi and/or Autophagy that further enhanced CuET's tumor cytotoxicity, using both Cell Culture and zebrafish in vivo preclinical models. Overall, these findings expand the mechanistic repertoire of CuET's anti-cancer activity, inform about the temporal order of responses and identify an unorthodox new mechanism of targeting p53. Our results are discussed in LIGHT of cancer-associated endogenous stresses as exploitable tumor vulnerabilities and may inspire future clinical applications of CuET in oncology, including combinatorial treatments and focus on potential advantages of using certain validated drug metabolites, rather than old, approved drugs with their, often complex, metabolic profiles.

Figures
Products