Epigenetic drugs in somatostatin type 2 receptor radionuclide theranostics and radiation transcriptomics in mouse pheochromocytoma models

Research output: Contribution to journalResearch articleContributedpeer-review

Contributors

Abstract

Pheochromocytomas and paragangliomas (PCCs/PGLs) are catecholamine-producing tumors. In inoperable and metastatic cases, somatostatin type 2 receptor (SSTR2) expression allows for peptide receptor radionuclide therapy with [177Lu]Lu-DOTA-TATE. Insufficient receptor levels, however, limit treatment efficacy. This study evaluates whether the epigenetic drugs valproic acid (VPA) and 5-Aza-2'-deoxycytidine (DAC) modulate SSTR2 levels and sensitivity to [177Lu]Lu-DOTA-TATE in two mouse PCC models (MPC and MTT). Methods: Drug-effects on Sstr2/SSTR2 were investigated in terms of promoter methylation, mRNA and protein levels, and radiotracer binding. Radiotracer uptake was measured in subcutaneous allografts in mice using PET and SPECT imaging. Tumor growth and gene expression (RNAseq) were characterized after drug treatments. Results: DAC alone and in combination with VPA increased SSTR2 levels along with radiotracer uptake in vitro in MPC (high-SSTR2) and MTT cells (low-SSTR2). MTT but not MPC allografts responded to DAC and VPA combination with significantly elevated radiotracer uptake, although activity concentrations remained far below those in MPC tumors. In both models, combination of DAC, VPA and [177Lu]Lu-DOTA-TATE was associated with additive effects on tumor growth delay and specific transcriptional responses in gene sets involved in cancer and treatment resistance. Effects of epigenetic drugs were unrelated to CpG island methylation of the Sstr2 promoter. Conclusion: This study demonstrates that SSTR2 induction in mouse pheochromocytoma models has some therapeutic benefit that occurs via yet unknown mechanisms. Transcriptional changes in tumor allografts associated with epigenetic treatment and [177Lu]Lu-DOTA-TATE provide first insights into genetic responses of PCCs/PGLs, potentially useful for developing additional strategies to prevent tumor recurrence.

Details

Original languageEnglish
Pages (from-to)278-294
Number of pages17
JournalTheranostics
Volume13
Issue number1
Publication statusPublished - 2023
Peer-reviewedYes

External IDs

PubMed 36593963
ORCID /0000-0002-3549-2477/work/142244878

Keywords

Research priority areas of TU Dresden

Sustainable Development Goals

Keywords

  • decitabine, neuroendocrine tumors, radiation biology, small animal multimodal imaging, valproic acid, Neuroendocrine Tumors/pathology, Epigenesis, Genetic, Transcriptome, Pheochromocytoma/drug therapy, Radioisotopes/metabolism, Receptors, Somatostatin/genetics, Animals, Neoplasm Recurrence, Local/drug therapy, Mice, Somatostatin, Adrenal Gland Neoplasms, Precision Medicine, Octreotide/therapeutic use