@article{oai:repo.qst.go.jp:00048558, author = {Yoshii, Yukie and Hiroki Matsumoto and Yoshimoto, Mitsuyoshi and Zhang, Ming-Rong and Ooe, Yoko and Kurihara, Hiroaki and Narita, Yoshitaka and Jin, Zhao-Hui and Tsuji, Atsushi and Yoshinaga, Keiichiro and Fujibayashi, Yasuhisa and Higashi, Tatsuya and 吉井 幸恵 and 張 明栄 and 大江 洋子 and 金 朝暉 and 辻 厚至 and 吉永 恵一郎 and 藤林 康久 and 東 達也}, issue = {1}, journal = {Translational Oncology}, month = {Nov}, note = {Glioblastoma is the most aggressive malignant brain tumor in humans and is difficult to cure using current treatment options. Hypoxic regions are frequently found in glioblastoma, and increased levels of hypoxia are associated with poor clinical outcomes of glioblastoma patients. Hypoxia plays important roles in the progression and recurrence of glioblastoma because of drug delivery deficiencies and induction of hypoxia-inducible factor-1α in tumor cells, which lead to poor prognosis. We focused on a promising hypoxia-targeted internal radiotherapy agent, 64Cu-diacetyl-bis (N4-methylthiosemicarbazone) (64Cu-ATSM), to address the need for additional treatment for glioblastoma. This compound can target the overreduced state under hypoxic conditions within tumors. Clinical positron emission tomography studies using radiolabeled Cu-ATSM have shown that Cu-ATSM accumulates in glioblastoma and its uptake is associated with high hypoxia-inducible factor-1α expression. To evaluate the therapeutic potential of this agent for glioblastoma, we examined the efficacy of 64Cu-ATSM in mice bearing U87MG glioblastoma tumors. Administration of single dosage (18.5, 37, 74, 111, and 148 MBq) and multiple dosages (37 MBq × 4) of 64Cu-ATSM was investigated. Single administration of 64Cu-ATSM in high-dose groups dose-dependently inhibited tumor growth and prolonged survival, with slight and reverse signs of adverse events. Multiple dosages of 64Cu-ATSM remarkably inhibited tumor growth and prolonged survival. By splitting the dose of 64Cu-ATSM, no adverse effects were observed. Our findings indicate that multiple administrations of 64Cu-ATSM have effective antitumor effects in glioblastoma without side effects, indicating its potential for treating this fatal disease.}, pages = {24--30}, title = {Multiple Administrations of 64Cu-ATSM as a Novel Therapeutic Option for Glioblastoma: a Translational Study Using Mice with Xenografts}, volume = {11}, year = {2017} }