CUDC-907, a dual PI3K/histone deacetylase inhibitor, increases meta-iodobenzylguanidine uptake (123/131I-mIBG) in vitro and in vivo: a promising candidate for advancing theranostics in neuroendocrine tumors.

Details

Serval ID
serval:BIB_9B802C61B336
Type
Article: article from journal or magazin.
Collection
Publications
Institution
Title
CUDC-907, a dual PI3K/histone deacetylase inhibitor, increases meta-iodobenzylguanidine uptake (123/131I-mIBG) in vitro and in vivo: a promising candidate for advancing theranostics in neuroendocrine tumors.
Journal
Journal of translational medicine
Author(s)
Grand-Guillaume J., Mansi R., Gaonkar R.H., Zanger S., Fani M., Eugster P.J., Beck Popovic M., Grouzmann E., Abid K.
ISSN
1479-5876 (Electronic)
ISSN-L
1479-5876
Publication state
Published
Issued date
07/09/2023
Peer-reviewed
Oui
Volume
21
Number
1
Pages
604
Language
english
Notes
Publication types: Journal Article ; Research Support, Non-U.S. Gov't
Publication Status: epublish
Abstract
Neuroblastoma (NB) and pheochromocytoma/paraganglioma (PHEO/PGL) are neuroendocrine tumors. Imaging of these neoplasms is performed by scintigraphy after injection of radiolabeled meta-iodobenzylguanidine (mIBG), a norepinephrine analog taken up by tumoral cells through monoamine transporters. The pharmacological induction of these transporters is a promising approach to improve the imaging and therapy (theranostics) of these tumors.
Transporters involved in mIBG internalization were identified by using transfected Human Embryonic Kidney (HEK) cells. Histone deacetylase inhibitors (HDACi) and inhibitors of the PI3K/AKT/mTOR pathway were tested in cell lines to study their effect on mIBG internalization. Studies in xenografted mice were performed to assess the effect of the most promising HDACi on <sup>123</sup> I-mIBG uptake.
Transfected HEK cells demonstrated that the norepinephrine and dopamine transporter (NET and DAT) avidly internalizes mIBG. Sodium-4-phenylbutyrate (an HDACi), CUDC-907 (a dual HDACi and PI3K inhibitor), BGT226 (a PI3K inhibitor) and VS-5584 and rapamycin (two inhibitors of mTOR) increased mIBG internalization in a neuroblastoma cell line (IGR-NB8) by 2.9-, 2.1-, 2.5-, 1.5- and 1.3-fold, respectively, compared with untreated cells. CUDC-907 also increased mIBG internalization in two other NB cell lines and in one PHEO cell line. We demonstrated that mIBG internalization occurs primarily through the NET. In xenografted mice with IGR-NB8 cells, oral treatment with 5 mg/kg of CUDC-907 increased the tumor uptake of <sup>123</sup> I-mIBG by 2.3- and 1.9-fold at 4 and 24 h post-injection, respectively, compared to the untreated group.
Upregulation of the NET by CUDC-907 lead to a better internalization of mIBG in vitro and in vivo.
Keywords
Humans, Animals, Mice, Neuroendocrine Tumors, Histone Deacetylase Inhibitors/pharmacology, Histone Deacetylase Inhibitors/therapeutic use, 3-Iodobenzylguanidine/pharmacology, 3-Iodobenzylguanidine/therapeutic use, Phosphatidylinositol 3-Kinases, Precision Medicine, Neuroblastoma/diagnostic imaging, Neuroblastoma/drug therapy, Histone deacetylase inhibitor (HDACi), Meta-iodobenzylguanidine (mIBG), Neuroendocrine tumors, PI3K/AKT/mTOR inhibitor, Theranostics, Xenograft
Pubmed
Web of science
Open Access
Yes
Create date
25/09/2023 15:57
Last modification date
16/01/2024 8:12
Usage data