Skip to main content
Top
Published in: EJNMMI Research 1/2020

Open Access 01-12-2020 | Positron Emission Tomography | Original research

Tracking a TGF-β activator in vivo: sensitive PET imaging of αvβ8-integrin with the Ga-68-labeled cyclic RGD octapeptide trimer Ga-68-Triveoctin

Authors: Neil Gerard Quigley, Katja Steiger, Frauke Richter, Wilko Weichert, Sebastian Hoberück, Jörg Kotzerke, Johannes Notni

Published in: EJNMMI Research | Issue 1/2020

Login to get access

Abstract

Purpose

As a major activator of transforming growth factor β (TGF-β), the RGD receptor αvβ8-integrin is involved in pathogenic processes related to TGF-β dysregulation, such as tumor growth, invasion, and radiochemoresistance, metastasis and tumor cell stemness, as well as epithelial-mesenchymal transition. The novel positron emission tomography (PET) radiopharmaceutical Ga-68-Triveoctin for in vivo mapping of αvβ8-integrin expression might enhance the prognosis of certain tumor entities, as well as support and augment TGF-β-targeted therapeutic approaches.

Methods

Monomeric and trimeric conjugates of cyclo(GLRGDLp(NMe)K(pent-4-ynoic amide)) were synthesized by click chemistry (CuAAC), labeled with Ga-68, and evaluated in MeWo (human melanoma) xenografted SCID mice by means of PET and ex-vivo biodistribution. αvβ8-integrin expression in murine tissues was determined by β8-IHC. A human subject received a single injection of 173 MBq of Ga-68-Triveoctin and underwent 3 subsequent PET/CT scans at 25, 45, and 90 min p.i..

Results

The trimer Ga-68-Triveoctin exhibits a 6.7-fold higher αvβ8-integrin affinity than the monomer (IC50 of 5.7 vs. 38 nM, respectively). Accordingly, biodistribution showed a higher tumor uptake (1.9 vs. 1.0%IA/g, respectively) but a similar baseline upon blockade (0.25%IA/g for both). IHC showed an intermediate β8-expression in the tumor while most organs and tissues were found β8-negative. Low non-target tissue uptakes (< 0.4%IA/g) confirmed a low degree of unspecific binding. Due to its hydrophilicity (log D = − 3.1), Ga-68-Triveoctin is excreted renally and shows favorable tumor/tissue ratios in mice (t/blood: 6.7; t/liver: 6.8; t/muscle: 29). A high kidney uptake in mice (kidney-to-blood and -to-muscle ratios of 126 and 505, respectively) is not reflected by human PET (corresponding values are 15 and 30, respectively), which furthermore showed notable uptakes in coeliac and choroid plexus (SUVmean 6.1 and 9.7, respectively, 90 min p.i.).

Conclusion

Ga-68-Triveoctin enables sensitive in-vivo imaging αvβ8-integrin expression in murine tumor xenografts. PET in a human subject confirmed a favorable biodistribution, underscoring the potential of Ga-68-Triveoctin for mapping of αvβ8-integrin expression in a clinical setting.
Appendix
Available only for authorised users
Literature
1.
go back to reference Moyle M, Napier MA, McLean JW. Cloning and expression of a divergent integrin subunit β8. J Biol Chem. 1991;266:19650–8.PubMed Moyle M, Napier MA, McLean JW. Cloning and expression of a divergent integrin subunit β8. J Biol Chem. 1991;266:19650–8.PubMed
2.
go back to reference Nishimura SL, Sheppard D, Pytela R. Integrin αvβ8. J Biol Chem. 1994;269:28708–15.PubMed Nishimura SL, Sheppard D, Pytela R. Integrin αvβ8. J Biol Chem. 1994;269:28708–15.PubMed
3.
go back to reference Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor β in human disease. N Engl J Med. 2000;34:1350–8.CrossRef Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor β in human disease. N Engl J Med. 2000;34:1350–8.CrossRef
4.
go back to reference Worthington JJ, Klementowicz JE, Travis MA. TGF-β: a sleeping giant awoken by integrins. Trends Biochem Sci. 2011;36:47–54.CrossRef Worthington JJ, Klementowicz JE, Travis MA. TGF-β: a sleeping giant awoken by integrins. Trends Biochem Sci. 2011;36:47–54.CrossRef
5.
go back to reference Khan Z, Marshall JF. Thr role of integrins in TGF-β activation in the tumour stroma. Cell Tissue Res. 2016;365:657–73.CrossRef Khan Z, Marshall JF. Thr role of integrins in TGF-β activation in the tumour stroma. Cell Tissue Res. 2016;365:657–73.CrossRef
6.
7.
go back to reference Dong X, Zhao B, Iacob RE, Zhu J, Koksal AC, Lu C, Engen JR, Springer TA. Force interacts with macromolecular structure in activation of TGF-β. Nature. 2017;542:55–9.CrossRef Dong X, Zhao B, Iacob RE, Zhu J, Koksal AC, Lu C, Engen JR, Springer TA. Force interacts with macromolecular structure in activation of TGF-β. Nature. 2017;542:55–9.CrossRef
8.
go back to reference Mu D, Cambier S, Fjellbirkeland L, Baron JL, Munger JS, Kawakatsu H, Sheppard D, Broaddus VC, Nishimura SL. The integrin αvβ8 mediates epithelial homeostasis through MT1-MMP-dependent activation of TGF-β. J Cell Biol. 2002;157:493–507.CrossRef Mu D, Cambier S, Fjellbirkeland L, Baron JL, Munger JS, Kawakatsu H, Sheppard D, Broaddus VC, Nishimura SL. The integrin αvβ8 mediates epithelial homeostasis through MT1-MMP-dependent activation of TGF-β. J Cell Biol. 2002;157:493–507.CrossRef
9.
go back to reference Brown NF, Marshall JF. Integrin-mediated TGF-β activation modulates the tumour microenvironment. Cancers. 2019;11:1221.CrossRef Brown NF, Marshall JF. Integrin-mediated TGF-β activation modulates the tumour microenvironment. Cancers. 2019;11:1221.CrossRef
10.
go back to reference Inman GJ. Switching TGF-β from a tumor suppressor to a tumor promoter. Curr Opin Genet Dev. 2011;21:93–9.CrossRef Inman GJ. Switching TGF-β from a tumor suppressor to a tumor promoter. Curr Opin Genet Dev. 2011;21:93–9.CrossRef
11.
go back to reference Cambier S, Mu DZ, O’Connell D, Boylen K, Travis W, Liu WH, Broaddus VC, Nishimura SL. A role for the integrin αvβ8 in the negative regulation of epithelial cell growth. Cancer Res. 2000;60:7084–93.PubMed Cambier S, Mu DZ, O’Connell D, Boylen K, Travis W, Liu WH, Broaddus VC, Nishimura SL. A role for the integrin αvβ8 in the negative regulation of epithelial cell growth. Cancer Res. 2000;60:7084–93.PubMed
12.
go back to reference Adorno M, Cordenonsi M, Montagner M, Dupont S, Wong C, Hann B, Solari A, Bobisse S, Rondina MB, Guzzardo V, et al. A Mutant-p53/Smad complex opposes p63 to empower TGF-β-induced metastasis. Cell. 2009;137:87–98.CrossRef Adorno M, Cordenonsi M, Montagner M, Dupont S, Wong C, Hann B, Solari A, Bobisse S, Rondina MB, Guzzardo V, et al. A Mutant-p53/Smad complex opposes p63 to empower TGF-β-induced metastasis. Cell. 2009;137:87–98.CrossRef
13.
go back to reference Zhang B, Halder SK, Kashikar ND, Cho YJ, Datta A, Gorden DL, Datta PK. Antimetastatic role of Smad4 signaling in colorectal cancer. Gastroenterology. 2010;138:969–80.CrossRef Zhang B, Halder SK, Kashikar ND, Cho YJ, Datta A, Gorden DL, Datta PK. Antimetastatic role of Smad4 signaling in colorectal cancer. Gastroenterology. 2010;138:969–80.CrossRef
14.
go back to reference Villalba M, Evans SR, Vidal-Vanaclocha F, Calvo A. Role of TGF-β in metastatic colon cancer: it is finally time for targeted therapy. Cell Tissue Res. 2017;320:29–39.CrossRef Villalba M, Evans SR, Vidal-Vanaclocha F, Calvo A. Role of TGF-β in metastatic colon cancer: it is finally time for targeted therapy. Cell Tissue Res. 2017;320:29–39.CrossRef
15.
go back to reference Guerrero PA, Tchaicha JH, Chen Z, Morales JE, McCarty N, Wang Q, Sulman EP, Fuller G, Lang FF, Rao G, McCarty JH. Glioblastoma stem cells exploit the αvβ8 integrin-TGFβ1 signaling axis to drive tumor initiation and progression. Oncogene. 2017;47:6568–80.CrossRef Guerrero PA, Tchaicha JH, Chen Z, Morales JE, McCarty N, Wang Q, Sulman EP, Fuller G, Lang FF, Rao G, McCarty JH. Glioblastoma stem cells exploit the αvβ8 integrin-TGFβ1 signaling axis to drive tumor initiation and progression. Oncogene. 2017;47:6568–80.CrossRef
16.
go back to reference Mertens-Walker I, Fernandini BC, Maharaj MSN, Rockstroh A, Nelson CC, Herington AC, Stephenson SA. The tumour-promoting receptor tyrosine kinase, EphB4, regulates expression of Integrin-β8 in prostate cancer cells. BMC Cancer. 2015;15:164.CrossRef Mertens-Walker I, Fernandini BC, Maharaj MSN, Rockstroh A, Nelson CC, Herington AC, Stephenson SA. The tumour-promoting receptor tyrosine kinase, EphB4, regulates expression of Integrin-β8 in prostate cancer cells. BMC Cancer. 2015;15:164.CrossRef
17.
go back to reference Cambier S, Gline S, Mu D, Collins R, Araya J, Dolganov G, Einheber S, Boudreau N, Nishimura SL. Integrin αvβ8-mediated activation of transforming growth factor-β by perivascular astrocytes. Am J Pathol. 2005;166:1883–94.CrossRef Cambier S, Gline S, Mu D, Collins R, Araya J, Dolganov G, Einheber S, Boudreau N, Nishimura SL. Integrin αvβ8-mediated activation of transforming growth factor-β by perivascular astrocytes. Am J Pathol. 2005;166:1883–94.CrossRef
18.
go back to reference Reyes SB, Narayanan AS, Lee HS, Tchaicha JH, Aldape KD, Lang FF, Tolias KF, McCarty JH. αvβ8 integrin interacts with RhoGDI1 to regulate Rac1 and Cdc42 activation and drive glioblastoma cell invasion. Mol Biol Cell. 2013;24:474–82.CrossRef Reyes SB, Narayanan AS, Lee HS, Tchaicha JH, Aldape KD, Lang FF, Tolias KF, McCarty JH. αvβ8 integrin interacts with RhoGDI1 to regulate Rac1 and Cdc42 activation and drive glioblastoma cell invasion. Mol Biol Cell. 2013;24:474–82.CrossRef
19.
go back to reference Reichart F, Maltsev OV, Kapp TG, Räder AFB, Weinmüller M, Marelli UK, Notni J, Wurzer A, Beck R, Wester HJ, Steiger K, Di Maro S, Di Leva FS, Marinelli L, Nieberler M, Reuning U, Schwaiger M, Kessler H. Selective targeting of integrin αvβ8 by a highly active cyclic peptide. J Med Chem. 2019;62:2024–37.CrossRef Reichart F, Maltsev OV, Kapp TG, Räder AFB, Weinmüller M, Marelli UK, Notni J, Wurzer A, Beck R, Wester HJ, Steiger K, Di Maro S, Di Leva FS, Marinelli L, Nieberler M, Reuning U, Schwaiger M, Kessler H. Selective targeting of integrin αvβ8 by a highly active cyclic peptide. J Med Chem. 2019;62:2024–37.CrossRef
20.
go back to reference Kapp TG, Rechenmacher F, Neubauer S, Maltsev O, Cavalcanti-Adam AE, Zarka R, Reuning U, Notni J, Wester HJ, Mas-Moruno C, Spatz J, Geiger B, Kessler H. A comprehensive evaluation of the activity and selectivity profile of ligands for RGD-binding integrins. Sci Rep. 2017;7:39805.CrossRef Kapp TG, Rechenmacher F, Neubauer S, Maltsev O, Cavalcanti-Adam AE, Zarka R, Reuning U, Notni J, Wester HJ, Mas-Moruno C, Spatz J, Geiger B, Kessler H. A comprehensive evaluation of the activity and selectivity profile of ligands for RGD-binding integrins. Sci Rep. 2017;7:39805.CrossRef
21.
go back to reference Notni J, Šimeček J, Hermann P, Wester HJ. TRAP, a powerful and versatile framework for gallium-68 radiopharmaceuticals. Chemistry. 2011;17:14718–22.CrossRef Notni J, Šimeček J, Hermann P, Wester HJ. TRAP, a powerful and versatile framework for gallium-68 radiopharmaceuticals. Chemistry. 2011;17:14718–22.CrossRef
22.
go back to reference Färber SF, Wurzer A, Reichart F, Beck R, Kessler H, Wester HJ, Notni J. Therapeutic radiopharmaceuticals targeting integrin αvβ6. ACS Omega. 2018;3:2428–36.CrossRef Färber SF, Wurzer A, Reichart F, Beck R, Kessler H, Wester HJ, Notni J. Therapeutic radiopharmaceuticals targeting integrin αvβ6. ACS Omega. 2018;3:2428–36.CrossRef
23.
go back to reference Pohle K, Notni J, Bussemer J, Kessler H, Schwaiger M, Beer AJ. 68Ga-NODAGA-RGD is a suitable substitute for 18F-Galacto-RGD and can be produced with high specific activity in a cGMP/GRP compliant automated process. Nucl Med Biol. 2012;39:777–84.CrossRef Pohle K, Notni J, Bussemer J, Kessler H, Schwaiger M, Beer AJ. 68Ga-NODAGA-RGD is a suitable substitute for 18F-Galacto-RGD and can be produced with high specific activity in a cGMP/GRP compliant automated process. Nucl Med Biol. 2012;39:777–84.CrossRef
24.
go back to reference Stabin MG, Sparks RB, Crowe E. OLINDA/EXM: the second-generation personal computer software for internal dose assessment in nuclear medicine. J Nucl Med. 2005;46:1023–7.PubMed Stabin MG, Sparks RB, Crowe E. OLINDA/EXM: the second-generation personal computer software for internal dose assessment in nuclear medicine. J Nucl Med. 2005;46:1023–7.PubMed
25.
go back to reference Baranyai Z, Reich D, Vágner A, et al. A shortcut to high-affinity Ga-68 and Cu-64 radiopharmaceuticals: one-pot click chemistry trimerisation on the TRAP platform. Dalton Trans. 2015;44:11137–46.CrossRef Baranyai Z, Reich D, Vágner A, et al. A shortcut to high-affinity Ga-68 and Cu-64 radiopharmaceuticals: one-pot click chemistry trimerisation on the TRAP platform. Dalton Trans. 2015;44:11137–46.CrossRef
26.
go back to reference Notni J, Reich D, Maltsev OV, Kapp TG, Steiger K, Hoffmann F, Esposito I, Weichert W, Kessler H, Wester HJ. In vivo PET imaging of the cancer integrin αvβ6 using 68Ga-labeled cyclic RGD nonapeptides. J Nucl Med. 2017;58:671–7.CrossRef Notni J, Reich D, Maltsev OV, Kapp TG, Steiger K, Hoffmann F, Esposito I, Weichert W, Kessler H, Wester HJ. In vivo PET imaging of the cancer integrin αvβ6 using 68Ga-labeled cyclic RGD nonapeptides. J Nucl Med. 2017;58:671–7.CrossRef
27.
go back to reference Wurzer A, Pollmann J, Schmidt A, Reich D, Wester HJ, Notni J. Molar activity of Ga-68 labeled PSMA inhibitor conjugates determines PET imaging results. Mol Pharmaceutics. 2018;15:4296–302.CrossRef Wurzer A, Pollmann J, Schmidt A, Reich D, Wester HJ, Notni J. Molar activity of Ga-68 labeled PSMA inhibitor conjugates determines PET imaging results. Mol Pharmaceutics. 2018;15:4296–302.CrossRef
28.
go back to reference Notni J, Šimeček J, Wester HJ. Phosphinic acid functionalized polyazacycloalkane chelators for radiodiagnostics and radiotherapeutics: unique characteristics and applications. ChemMedChem. 2014;9:1107–15.CrossRef Notni J, Šimeček J, Wester HJ. Phosphinic acid functionalized polyazacycloalkane chelators for radiodiagnostics and radiotherapeutics: unique characteristics and applications. ChemMedChem. 2014;9:1107–15.CrossRef
29.
go back to reference Notni J, Hermann P, Havlíčková J, et al. A triazacyclononane-based bifunctional phosphinate ligand for the preparation of multimeric 68Ga tracers for positron emission tomography. Chem Eur J. 2010;16:7174–85.CrossRef Notni J, Hermann P, Havlíčková J, et al. A triazacyclononane-based bifunctional phosphinate ligand for the preparation of multimeric 68Ga tracers for positron emission tomography. Chem Eur J. 2010;16:7174–85.CrossRef
30.
go back to reference Notni J, Wester HJ. A practical guide on the synthesis of metal chelates for molecular imaging and therapy by means of click chemistry. Chem Eur J. 2016;22:11500–8.CrossRef Notni J, Wester HJ. A practical guide on the synthesis of metal chelates for molecular imaging and therapy by means of click chemistry. Chem Eur J. 2016;22:11500–8.CrossRef
31.
go back to reference Hirota S, Liu Q, Lee HS, Hossain MG, Lacy-Hulbert A, McCarty JH. The astrocyte-expressed integrin αvβ8 governs blood vessel sprouting in the developing retina. Development. 2011;138:5157–66.CrossRef Hirota S, Liu Q, Lee HS, Hossain MG, Lacy-Hulbert A, McCarty JH. The astrocyte-expressed integrin αvβ8 governs blood vessel sprouting in the developing retina. Development. 2011;138:5157–66.CrossRef
32.
go back to reference Khan S, Lakhe-Reddy S, McCarty JH, Sorenson CM, Sheibani N, Reichardt LF, Kim JH, Wang B, Sedor JR, Schelling JR. Mesangial cell integrin αvβ8 provides glomerular endothelial cell cytoprotection by sequestering TGF-β and regulating PECAM-1. Am J Pathol. 2011;178:609–20.CrossRef Khan S, Lakhe-Reddy S, McCarty JH, Sorenson CM, Sheibani N, Reichardt LF, Kim JH, Wang B, Sedor JR, Schelling JR. Mesangial cell integrin αvβ8 provides glomerular endothelial cell cytoprotection by sequestering TGF-β and regulating PECAM-1. Am J Pathol. 2011;178:609–20.CrossRef
33.
go back to reference Lakhe-Reddy S, Li V, Arnold TD, Khan S, Schelling JR. Mesangial cell αvβ8 -integrin regulates glomerular capillary integrity and repair. Am J Physiol Renal Physiol. 2014;306:F1400–9.CrossRef Lakhe-Reddy S, Li V, Arnold TD, Khan S, Schelling JR. Mesangial cell αvβ8 -integrin regulates glomerular capillary integrity and repair. Am J Physiol Renal Physiol. 2014;306:F1400–9.CrossRef
34.
go back to reference Šimeček J, Hermann P, Havlíčková J, et al. A cyclen-based tetraphosphinate chelator for preparation of radiolabeled tetrameric bioconjugates. Chemistry. 2013;19:7748–57.CrossRef Šimeček J, Hermann P, Havlíčková J, et al. A cyclen-based tetraphosphinate chelator for preparation of radiolabeled tetrameric bioconjugates. Chemistry. 2013;19:7748–57.CrossRef
35.
go back to reference Thumshirn G, Hersel U, Goodman SL, Kessler H. Multimeric cyclic RGD peptides as potential tools for tumor targeting: solid-phase peptide synthesis and chemoselective oxime ligation. Chemistry. 2003;9:2717–25.CrossRef Thumshirn G, Hersel U, Goodman SL, Kessler H. Multimeric cyclic RGD peptides as potential tools for tumor targeting: solid-phase peptide synthesis and chemoselective oxime ligation. Chemistry. 2003;9:2717–25.CrossRef
36.
go back to reference Dijkgraaf I, Kruijtzer JAW, Liu S, et al. Improved targeting of the αvβ3 integrin by multimerisation of RGD peptides. Eur J Nucl Med Mol Imaging. 2007;34:267–73.CrossRef Dijkgraaf I, Kruijtzer JAW, Liu S, et al. Improved targeting of the αvβ3 integrin by multimerisation of RGD peptides. Eur J Nucl Med Mol Imaging. 2007;34:267–73.CrossRef
37.
go back to reference Wängler C, Maschauer S, Prante O, et al. Multimerization of cRGD peptides by click chemistry: synthetic strategies, chemical limitations, and influence on biological properties. ChemBioChem. 2010;11:1–15.CrossRef Wängler C, Maschauer S, Prante O, et al. Multimerization of cRGD peptides by click chemistry: synthetic strategies, chemical limitations, and influence on biological properties. ChemBioChem. 2010;11:1–15.CrossRef
38.
go back to reference Kaeopookum P, Petrik M, Summer D, Klinger M, Zhai C, Rangger C, Haubner R, Haas H, Hajduch M, Decristoforo C. Comparison of 68Ga-labeled RGD mono- and multimers based on a clickable siderophore-based scaffold. Nucl Med Biol. 2019;78–79:1–10.CrossRef Kaeopookum P, Petrik M, Summer D, Klinger M, Zhai C, Rangger C, Haubner R, Haas H, Hajduch M, Decristoforo C. Comparison of 68Ga-labeled RGD mono- and multimers based on a clickable siderophore-based scaffold. Nucl Med Biol. 2019;78–79:1–10.CrossRef
39.
go back to reference Quigley NG, Tomassi S, Di Leva FS, Di Maro S, Richter F, Steiger K, Kossatz S, Marinelli L, Notni J. Click-chemistry (CuAAC) trimerization of an αvβ6-integrin targeting Ga-68-peptide: enhanced contrast for in-vivo PET imaging of human lung adenocarcinoma xenografts. ChemBioChem. 2020. https://doi.org/10.1002/cbic.202000200.CrossRefPubMed Quigley NG, Tomassi S, Di Leva FS, Di Maro S, Richter F, Steiger K, Kossatz S, Marinelli L, Notni J. Click-chemistry (CuAAC) trimerization of an αvβ6-integrin targeting Ga-68-peptide: enhanced contrast for in-vivo PET imaging of human lung adenocarcinoma xenografts. ChemBioChem. 2020. https://​doi.​org/​10.​1002/​cbic.​202000200.CrossRefPubMed
40.
go back to reference Maschauer S, Einsiedel J, Reich D, Hübner H, Gmeiner P, Wester HJ, Prante O, Notni J. Theranostic value of multimers: lessons learned from trimerization of neurotensin receptor ligands and other targeting vectors. Pharmaceuticals. 2017;10:29.CrossRef Maschauer S, Einsiedel J, Reich D, Hübner H, Gmeiner P, Wester HJ, Prante O, Notni J. Theranostic value of multimers: lessons learned from trimerization of neurotensin receptor ligands and other targeting vectors. Pharmaceuticals. 2017;10:29.CrossRef
41.
go back to reference Šimeček J, Notni J, Kapp TG, Kessler H, Wester HJ. Benefits of NOPO as chelator in gallium-68 peptides, exemplified by preclinical characterization of 68Ga-NOPO-c(RGDfK). Mol Pharm. 2014;11:1687–95.CrossRef Šimeček J, Notni J, Kapp TG, Kessler H, Wester HJ. Benefits of NOPO as chelator in gallium-68 peptides, exemplified by preclinical characterization of 68Ga-NOPO-c(RGDfK). Mol Pharm. 2014;11:1687–95.CrossRef
42.
go back to reference Vágner A, Forgács A, Brücher E, Tóth I, Maiocchi A, Wurzer A, Wester HJ, Notni J, Baranyai Z. Equilibrium thermodynamics, formation, and dissociation kinetics of trivalent iron and gallium complexes of triazacyclononane-triphosphinate (TRAP) chelators: unraveling the foundations of highly selective Ga-68 labeling. Front Chem. 2018;6:170.CrossRef Vágner A, Forgács A, Brücher E, Tóth I, Maiocchi A, Wurzer A, Wester HJ, Notni J, Baranyai Z. Equilibrium thermodynamics, formation, and dissociation kinetics of trivalent iron and gallium complexes of triazacyclononane-triphosphinate (TRAP) chelators: unraveling the foundations of highly selective Ga-68 labeling. Front Chem. 2018;6:170.CrossRef
43.
go back to reference Šimeček J, Hermann P, Wester HJ, Notni J. How is 68Ga-labeling of macrocyclic chelators influenced by metal ion contaminants in 68Ge/68Ga generator eluates? ChemMedChem. 2013;8:95–103.CrossRef Šimeček J, Hermann P, Wester HJ, Notni J. How is 68Ga-labeling of macrocyclic chelators influenced by metal ion contaminants in 68Ge/68Ga generator eluates? ChemMedChem. 2013;8:95–103.CrossRef
44.
go back to reference Notni J, Pohle K, Wester HJ. Comparative gallium-68 labeling of TRAP-, NOTA-, and DOTA-peptides: practical consequences for the future of gallium-68-PET. EJNMMI Res. 2012;2:28.CrossRef Notni J, Pohle K, Wester HJ. Comparative gallium-68 labeling of TRAP-, NOTA-, and DOTA-peptides: practical consequences for the future of gallium-68-PET. EJNMMI Res. 2012;2:28.CrossRef
45.
go back to reference Notni J, Wester HJ. Re-thinking the role of radiometal isotopes: Towards a future concept for theranostic radiopharmaceuticals. J Label Compd Radiopharm. 2018;61:141–53.CrossRef Notni J, Wester HJ. Re-thinking the role of radiometal isotopes: Towards a future concept for theranostic radiopharmaceuticals. J Label Compd Radiopharm. 2018;61:141–53.CrossRef
46.
go back to reference Takasaka N, Seed RI, Cormier A, Bondesson AJ, Lou J, Elattma A, Ito S, Yanagisawa H, Hashimoto M, Ma R, Levine MD, Publicover J, Potts R, Jespersen JM, Campbell MG, Conrad F, Marks JD, Cheng Y, Baron JL, Nishimura SL. Integrin αvβ8-expressing tumor cells evade host immunity by regulating TGF-β activation in immune cells. JCI Insight. 2018;3:e122591.CrossRef Takasaka N, Seed RI, Cormier A, Bondesson AJ, Lou J, Elattma A, Ito S, Yanagisawa H, Hashimoto M, Ma R, Levine MD, Publicover J, Potts R, Jespersen JM, Campbell MG, Conrad F, Marks JD, Cheng Y, Baron JL, Nishimura SL. Integrin αvβ8-expressing tumor cells evade host immunity by regulating TGF-β activation in immune cells. JCI Insight. 2018;3:e122591.CrossRef
47.
go back to reference Rotteveel L, Poot AJ, Bogaard HJ, ten Dijke P, Lammertsma AA, Windhorst AD. In vivo imaging of TGF signalling components using positron emission tomography. Drug Discov Today. 2019;24:2258–72.CrossRef Rotteveel L, Poot AJ, Bogaard HJ, ten Dijke P, Lammertsma AA, Windhorst AD. In vivo imaging of TGF signalling components using positron emission tomography. Drug Discov Today. 2019;24:2258–72.CrossRef
48.
go back to reference Korpal M, Kang Y. Targeting the transforming growth factor β signalling pathway in metastatic cancer. Eur J Cancer. 2010;46:1232–40.CrossRef Korpal M, Kang Y. Targeting the transforming growth factor β signalling pathway in metastatic cancer. Eur J Cancer. 2010;46:1232–40.CrossRef
49.
go back to reference Anderton MJ, Mellor HR, Bell A, Sadler C, Pass M, Powell S, Steele SJ, Roberts RR, Heier A. Induction of heart valve lesions by small-molecule ALK5 inhibitors. Toxicol Pathol. 2011;39:916–24.CrossRef Anderton MJ, Mellor HR, Bell A, Sadler C, Pass M, Powell S, Steele SJ, Roberts RR, Heier A. Induction of heart valve lesions by small-molecule ALK5 inhibitors. Toxicol Pathol. 2011;39:916–24.CrossRef
50.
go back to reference Vitsky A, Waire J, Pawliuk R, Bond A, Matthews D, Lacasse E, Hawes ML, Nelson C, Richards S, Piepenhagen PA, Garman RD, Andrews L, Thurberg BL, Lonning S, Ledbetter S, Ruzek MC. Homeostatic role of transforming growth factor-beta in the oral cavity and esophagus of mice and its expression by mast cells in these tissues. Am J Pathol. 2009;174:2137–49.CrossRef Vitsky A, Waire J, Pawliuk R, Bond A, Matthews D, Lacasse E, Hawes ML, Nelson C, Richards S, Piepenhagen PA, Garman RD, Andrews L, Thurberg BL, Lonning S, Ledbetter S, Ruzek MC. Homeostatic role of transforming growth factor-beta in the oral cavity and esophagus of mice and its expression by mast cells in these tissues. Am J Pathol. 2009;174:2137–49.CrossRef
51.
go back to reference Wang WW, Wang YB, Wang DQ, Lin Z, Sun RJ. Integrin β-8 (ITGB8) silencing reverses gefitinib resistance of human hepatic cancer HepG2/G cell line. Int J Clin Exp Med. 2015;8:3063–71.PubMed Wang WW, Wang YB, Wang DQ, Lin Z, Sun RJ. Integrin β-8 (ITGB8) silencing reverses gefitinib resistance of human hepatic cancer HepG2/G cell line. Int J Clin Exp Med. 2015;8:3063–71.PubMed
52.
go back to reference Jin S, Lee WC, Aust D, Pilarsky C, Cordes N. β8 integrin mediates pancreatic cancer cell radiochemoresistance. Mol Cancer Res. 2019;17:2126–38.CrossRef Jin S, Lee WC, Aust D, Pilarsky C, Cordes N. β8 integrin mediates pancreatic cancer cell radiochemoresistance. Mol Cancer Res. 2019;17:2126–38.CrossRef
Metadata
Title
Tracking a TGF-β activator in vivo: sensitive PET imaging of αvβ8-integrin with the Ga-68-labeled cyclic RGD octapeptide trimer Ga-68-Triveoctin
Authors
Neil Gerard Quigley
Katja Steiger
Frauke Richter
Wilko Weichert
Sebastian Hoberück
Jörg Kotzerke
Johannes Notni
Publication date
01-12-2020
Publisher
Springer Berlin Heidelberg
Published in
EJNMMI Research / Issue 1/2020
Electronic ISSN: 2191-219X
DOI
https://doi.org/10.1186/s13550-020-00706-1

Other articles of this Issue 1/2020

EJNMMI Research 1/2020 Go to the issue