Your browser doesn't support javascript.
loading
18 kDa translocator protein positron emission tomography facilitates early and robust tumor detection in the immunocompetent SB28 glioblastoma mouse model.
Bartos, Laura M; Kirchleitner, Sabrina V; Blobner, Jens; Wind, Karin; Kunze, Lea H; Holzgreve, Adrien; Gold, Lukas; Zatcepin, Artem; Kolabas, Zeynep Ilgin; Ulukaya, Selin; Weidner, Lorraine; Quach, Stefanie; Messerer, Denise; Bartenstein, Peter; Tonn, Joerg C; Riemenschneider, Markus J; Ziegler, Sibylle; von Baumgarten, Louisa; Albert, Nathalie L; Brendel, Matthias.
Afiliação
  • Bartos LM; Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Kirchleitner SV; Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Blobner J; Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Wind K; Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Kunze LH; Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Holzgreve A; Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Gold L; Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Zatcepin A; Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Kolabas ZI; Helmholtz Center, Institute for Tissue Engineering and Regenerative Medicine (iTERM), Munich, Germany.
  • Ulukaya S; Institute for Stroke and Dementia Research, University Hospital of Munich, Ludwig- Maximilians University Munich, Munich, Germany.
  • Weidner L; Graduate School of Systemic Neurosciences (GSN), Munich, Germany.
  • Quach S; Helmholtz Center, Institute for Tissue Engineering and Regenerative Medicine (iTERM), Munich, Germany.
  • Messerer D; Faculty of Biology, Master of Science Program in Molecular and Cellular Biology, Ludwig-Maximilians-Universität München, Planegg, Germany.
  • Bartenstein P; Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany.
  • Tonn JC; Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Riemenschneider MJ; Department of Cardiology, University Hospital of Munich, LMU Munich, Munich, Germany.
  • Ziegler S; Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany.
  • von Baumgarten L; SyNergy, University of Munich, Munich, Germany.
  • Albert NL; German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany.
  • Brendel M; Department of Neurosurgery, University Hospital of Munich, LMU Munich, Munich, Germany.
Front Med (Lausanne) ; 9: 992993, 2022.
Article em En | MEDLINE | ID: mdl-36325388
ABSTRACT

Introduction:

The 18 kDa translocator protein (TSPO) receives growing interest as a biomarker in glioblastoma. Mouse models can serve as an important tool for the investigation of biomarkers in glioblastoma, but several glioblastoma models indicated only low TSPO-PET signals in contrast to high TSPO-PET signals of human glioblastoma. Thus, we aimed to investigate TSPO-PET imaging in the syngeneic immunocompetent SB28 mouse model, which is thought to closely represent the tumor microenvironment (TME) of human glioblastoma.

Methods:

Dynamic TSPO-PET/CT imaging was performed for 60 min after injection of 13.6 ± 4.2 MBq [18F]GE-180. Contrast enhanced CT (ceCT) was acquired prior to PET and served for assessment of tumor volumes and attenuation correction. SB28 and sham mice were imaged at an early (week-1; n = 6 SB28, n = 6 sham) and a late time-point (week-3; n = 8 SB28, n = 9 sham) after inoculation. Standard of truth ex vivo tumor volumes were obtained for SB28 mice at the late time-point. Tracer kinetics were analyzed for the lesion site and the carotid arteries to establish an image derived input function (IDIF). TSPO-PET and ceCT lesion volumes were compared with ex vivo volumes by calculation of root-mean-square-errors (RMSE). Volumes of distribution (VTmax/mean) in the lesion were calculated using carotid IDIF and standardized uptake values (SUVmax/mean) were obtained for a 40-60 min time frame.

Results:

Higher uptake rate constants (K1) were observed for week-1 SB28 tumor lesions when compared to week-3 SB28 tumor lesions. Highest agreement between TSPO-PET lesion volumes and ex vivo tumor volumes was achieved with a 50% maximum threshold (RMSE-VT 39.7%; RMSE-SUV 34.4%), similar to the agreement of ceCT tumor volumes (RMSE 30.1%). Lesions of SB28 mice had higher PET signal when compared to sham mice at week-1 (VTmax 6.6 ± 2.9 vs. 3.9 ± 0.8, p = 0.035; SUVmax 2.3 ± 0.5 vs. 1.2 ± 0.1, p < 0.001) and PET signals remained at a similar level at week-3 (VTmax 5.0 ± 1.6 vs. 2.7 ± 0.8, p = 0.029; SUVmax 1.9 ± 0.5 vs. 1.2 ± 0.2, p = 0.0012). VTmax correlated with SUVmax (R 2 = 0.532, p < 0.001).

Conclusion:

TSPO-PET imaging of immunocompetent SB28 mice facilitates early detection of tumor signals over sham lesions. SB28 tumors mirror high TSPO-PET signals of human glioblastoma and could serve as a valuable translational model to study TSPO as an imaging biomarker.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Tipo de estudo: Diagnostic_studies / Screening_studies Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Tipo de estudo: Diagnostic_studies / Screening_studies Idioma: En Ano de publicação: 2022 Tipo de documento: Article