Developing a clinically relevant radiosensitizer for temozolomide-resistant gliomas

PLoS One. 2020 Sep 3;15(9):e0238238. doi: 10.1371/journal.pone.0238238. eCollection 2020.

Abstract

The prognosis for patients with glioblastoma (GB) remains grim. Concurrent temozolomide (TMZ) radiation-the cornerstone of glioma control-extends the overall median survival of GB patients by only a few months over radiotherapy alone. While these survival gains could be partly attributed to radiosensitization, this benefit is greatly minimized in tumors expressing O6-methylguanine DNA methyltransferase (MGMT), which specifically reverses O6-methylguanine lesions. Theoretically, non-O6-methylguanine lesions (i.e., the N-methylpurine adducts), which represent up to 90% of TMZ-generated DNA adducts, could also contribute to radiosensitization. Unfortunately, at concentrations attainable in clinical practice, the alkylation capacity of TMZ cannot overwhelm the repair of N-methylpurine adducts to efficiently exploit these lesions. The current therapeutic application of TMZ therefore faces two main obstacles: (i) the stochastic presence of MGMT and (ii) a blunted radiosensitization potential at physiologic concentrations. To circumvent these limitations, we are developing a novel molecule called NEO212-a derivatization of TMZ generated by coupling TMZ to perillyl alcohol. Based on gas chromatography/mass spectrometry and high-performance liquid chromatography analyses, we determined that NEO212 had greater tumor cell uptake than TMZ. In mouse models, NEO212 was more efficient than TMZ at crossing the blood-brain barrier, preferentially accumulating in tumoral over normal brain tissue. Moreover, in vitro analyses with GB cell lines, including TMZ-resistant isogenic variants, revealed more potent cytotoxic and radiosensitizing activities for NEO212 at physiologic concentrations. Mechanistically, these advantages of NEO212 over TMZ could be attributed to its enhanced tumor uptake presumably leading to more extensive DNA alkylation at equivalent dosages which, ultimately, allows for N-methylpurine lesions to be better exploited for radiosensitization. This effect cannot be achieved with TMZ at clinically relevant concentrations and is independent of MGMT. Our findings establish NEO212 as a superior radiosensitizer and a potentially better alternative to TMZ for newly diagnosed GB patients, irrespective of their MGMT status.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Blood-Brain Barrier / drug effects
  • Blood-Brain Barrier / metabolism
  • Brain / metabolism
  • Brain Neoplasms / drug therapy
  • Brain Neoplasms / pathology
  • Cell Line, Tumor
  • Cell Survival / drug effects
  • DNA Damage / drug effects
  • Dacarbazine / analogs & derivatives*
  • Dacarbazine / analysis
  • Dacarbazine / metabolism
  • Dacarbazine / pharmacology
  • Dacarbazine / therapeutic use
  • Drug Resistance, Neoplasm* / genetics
  • Gas Chromatography-Mass Spectrometry
  • Glioma / drug therapy*
  • Glioma / pathology
  • Humans
  • Mice
  • Mice, Inbred C57BL
  • O(6)-Methylguanine-DNA Methyltransferase / metabolism
  • Radiation-Sensitizing Agents / analysis
  • Radiation-Sensitizing Agents / metabolism
  • Radiation-Sensitizing Agents / pharmacology
  • Radiation-Sensitizing Agents / therapeutic use*
  • Temozolomide / analysis
  • Temozolomide / metabolism
  • Temozolomide / pharmacology
  • Temozolomide / therapeutic use*
  • Xenograft Model Antitumor Assays

Substances

  • NEO212
  • Radiation-Sensitizing Agents
  • Dacarbazine
  • O(6)-Methylguanine-DNA Methyltransferase
  • Temozolomide

Grants and funding

This study was funded in part by the Hale Family Research Fund and Sounder Foundation (provided to T.C.C), and by generous support from NeOnc Technologies, Inc. (Los Angeles, CA). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.