Your browser doesn't support javascript.
loading
Etomoxir, a carnitine palmitoyltransferase 1 inhibitor, combined with temozolomide reduces stemness and invasiveness in patient-derived glioblastoma tumorspheres.
Shim, Jin-Kyoung; Choi, Seonah; Yoon, Seon-Jin; Choi, Ran Joo; Park, Junseong; Lee, Eun Hee; Cho, Hye Joung; Lee, Suji; Teo, Wan-Yee; Moon, Ju Hyung; Kim, Hyun Sil; Kim, Eui Hyun; Cheong, Jae-Ho; Chang, Jong Hee; Yook, Jong In; Kang, Seok-Gu.
Afiliación
  • Shim JK; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Choi S; Brain Tumor Translational Research Laboratory, Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Yoon SJ; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Choi RJ; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Park J; Brain Tumor Translational Research Laboratory, Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Lee EH; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Cho HJ; Brain Tumor Translational Research Laboratory, Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Lee S; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Teo WY; Brain Tumor Translational Research Laboratory, Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Moon JH; Precision Medicine Research Center, College of Medicine, The Catholic University of Korea, Seoul, 03722, Republic of Korea.
  • Kim HS; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Kim EH; Brain Tumor Translational Research Laboratory, Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Cheong JH; Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Chang JH; Brain Tumor Translational Research Laboratory, Severance Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Yook JI; Department of Medical Science, BK21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
  • Kang SG; Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, 169857, Singapore.
Cancer Cell Int ; 22(1): 309, 2022 Oct 11.
Article en En | MEDLINE | ID: mdl-36221088
ABSTRACT

INTRODUCTION:

The importance of fatty acid oxidation (FAO) in the bioenergetics of glioblastoma (GBM) is being realized. Etomoxir (ETO), a carnitine palmitoyltransferase 1 (CPT1) inhibitor exerts cytotoxic effects in GBM, which involve interrupting the FAO pathway. We hypothesized that FAO inhibition could affect the outcomes of current standard temozolomide (TMZ) chemotherapy against GBM.

METHODS:

The FAO-related gene expression was compared between GBM and the tumor-free cortex. Using four different GBM tumorspheres (TSs), the effects of ETO and/or TMZ was analyzed on cell viability, tricarboxylate (TCA) cycle intermediates and adenosine triphosphate (ATP) production to assess metabolic changes. Alterations in tumor stemness, invasiveness, and associated transcriptional changes were also measured. Mouse orthotopic xenograft model was used to elucidate the combinatory effect of TMZ and ETO.

RESULTS:

GBM tissues exhibited overexpression of FAO-related genes, especially CPT1A, compared to the tumor-free cortex. The combined use of ETO and TMZ further inhibited TCA cycle and ATP production than single uses. This combination treatment showed superior suppression effects compared to treatment with individual agents on the viability, stemness, and invasiveness of GBM TSs, as well as better downregulation of FAO-related gene expression. The results of in vivo study showed prolonged survival outcomes in the combination treatment group.

CONCLUSION:

ETO, an FAO inhibitor, causes a lethal energy reduction in the GBM TSs. When used in combination with TMZ, ETO effectively reduces GBM cell stemness and invasiveness and further improves survival. These results suggest a potential novel treatment option for GBM.
Palabras clave

Texto completo: 1 Bases de datos: MEDLINE Idioma: En Revista: Cancer Cell Int Año: 2022 Tipo del documento: Article

Texto completo: 1 Bases de datos: MEDLINE Idioma: En Revista: Cancer Cell Int Año: 2022 Tipo del documento: Article