Subcellular Location of Tirapazamine Reduction Dramatically Affects Aerobic but Not Anoxic Cytotoxicity

Molecules. 2020 Oct 22;25(21):4888. doi: 10.3390/molecules25214888.

Abstract

Hypoxia is an adverse prognostic feature of solid cancers that may be overcome with hypoxia-activated prodrugs (HAPs). Tirapazamine (TPZ) is a HAP which has undergone extensive clinical evaluation in this context and stimulated development of optimized analogues. However the subcellular localization of the oxidoreductases responsible for mediating TPZ-dependent DNA damage remains unclear. Some studies conclude only nuclear-localized oxidoreductases can give rise to radical-mediated DNA damage and thus cytotoxicity, whereas others identify a broader role for endoplasmic reticulum and cytosolic oxidoreductases, indicating the subcellular location of TPZ radical formation is not a critical requirement for DNA damage. To explore this question in intact cells we engineered MDA-231 breast cancer cells to express the TPZ reductase human NADPH: cytochrome P450 oxidoreductase (POR) harboring various subcellular localization sequences to guide this flavoenzyme to the nucleus, endoplasmic reticulum, cytosol or inner surface of the plasma membrane. We show that all POR variants are functional, with differences in rates of metabolism reflecting enzyme expression levels rather than intracellular TPZ concentration gradients. Under anoxic conditions, POR expression in all subcellular compartments increased the sensitivity of the cells to TPZ, but with a fall in cytotoxicity per unit of metabolism (termed 'metabolic efficiency') when POR is expressed further from the nucleus. However, under aerobic conditions a much larger increase in cytotoxicity was observed when POR was directed to the nucleus, indicating very high metabolic efficiency. Consequently, nuclear metabolism results in collapse of hypoxic selectivity of TPZ, which was further magnified to the point of reversing O2 dependence (oxic > hypoxic sensitivity) by employing a DNA-affinic TPZ analogue. This aerobic hypersensitivity phenotype was partially rescued by cellular copper depletion, suggesting the possible involvement of Fenton-like chemistry in generating short-range effects mediated by the hydroxyl radical. In addition, the data suggest that under aerobic conditions reoxidation strictly limits the TPZ radical diffusion range resulting in site-specific cytotoxicity. Collectively these novel findings challenge the purported role of intra-nuclear reductases in orchestrating the hypoxia selectivity of TPZ.

Keywords: DNA damage-response; DNA-targeted cytotoxin; cell membrane; cell nucleus; cytochrome P450 oxidoreductase; endoplasmic reticulum; hypoxia-activated prodrug; tirapazamine.

MeSH terms

  • Antineoplastic Agents / chemistry*
  • Antineoplastic Agents / pharmacology
  • Cell Engineering
  • Cell Hypoxia / drug effects
  • Cell Line, Tumor
  • Cell Membrane / drug effects
  • Cell Nucleus / drug effects
  • Cell Survival / drug effects
  • Copper / metabolism
  • DNA Damage / drug effects
  • DNA Damage / genetics
  • Humans
  • Hypoxia / drug therapy*
  • Models, Biological
  • NADPH-Ferrihemoprotein Reductase / genetics*
  • NADPH-Ferrihemoprotein Reductase / metabolism
  • NADPH-Ferrihemoprotein Reductase / ultrastructure
  • Oxygen / metabolism
  • Prodrugs / chemistry*
  • Prodrugs / metabolism
  • Tirapazamine / chemistry*
  • Tirapazamine / metabolism

Substances

  • Antineoplastic Agents
  • Prodrugs
  • Tirapazamine
  • Copper
  • NADPH-Ferrihemoprotein Reductase
  • Oxygen