Back to Search Start Over

The human somatostatin receptor type 2 as an imaging and suicide reporter gene for pluripotent stem cell-derived therapy of myocardial infarction.

Authors :
Neyrinck K
Breuls N
Holvoet B
Oosterlinck W
Wolfs E
Vanbilloen H
Gheysens O
Duelen R
Gsell W
Lambrichts I
Himmelreich U
Verfaillie CM
Sampaolesi M
Deroose CM
Source :
Theranostics [Theranostics] 2018 Apr 14; Vol. 8 (10), pp. 2799-2813. Date of Electronic Publication: 2018 Apr 14 (Print Publication: 2018).
Publication Year :
2018

Abstract

Rationale: Pluripotent stem cells (PSCs) are being investigated as a cell source for regenerative medicine since they provide an infinitive pool of cells that are able to differentiate towards every cell type of the body. One possible therapeutic application involves the use of these cells to treat myocardial infarction (MI), a condition where billions of cardiomyocytes (CMs) are lost. Although several protocols have been developed to differentiate PSCs towards CMs, none of these provide a completely pure population, thereby still posing a risk for neoplastic teratoma formation. Therefore, we developed a strategy to (i) monitor cell behavior noninvasively via site-specific integration of firefly luciferase (Fluc) and the human positron emission tomography (PET) imaging reporter genes, sodium iodide symporter (hNIS) and somatostatin receptor type 2 (hSSTr2), and (ii) perform hSSTr2-mediated suicide gene therapy via the clinically used radiopharmacon <superscript>177</superscript> Lu-DOTATATE. Methods: Human embryonic stem cells (ESCs) were gene-edited via zinc finger nucleases to express Fluc and either hNIS or hSSTr2 in the safe harbor locus, adeno-associated virus integration site 1. Firstly, these cells were exposed to 4.8 MBq <superscript>177</superscript> Lu-DOTATATE in vitro and cell survival was monitored via bioluminescence imaging (BLI). Afterwards, hNIS <superscript>+</superscript> and hSSTr2 <superscript>+</superscript> ESCs were transplanted subcutaneously and teratomas were allowed to form. At day 59, baseline <superscript>124</superscript> I and <superscript>68</superscript> Ga-DOTATATE PET and BLI scans were performed. The day after, animals received either saline or 55 MBq <superscript>177</superscript> Lu-DOTATATE. Weekly BLI scans were performed, accompanied by <superscript>124</superscript> I and <superscript>68</superscript> Ga-DOTATATE PET scans at days 87 and 88, respectively. Finally, hSSTr2 <superscript>+</superscript> ESCs were differentiated towards CMs and transplanted intramyocardially in the border zone of an infarct that was induced by left anterior descending coronary artery ligation. After transplantation, the animals were monitored via BLI and PET, while global cardiac function was evaluated using cardiac magnetic resonance imaging. Results: Teratoma growth of both hNIS <superscript>+</superscript> and hSSTr2 <superscript>+</superscript> ESCs could be followed noninvasively over time by both PET and BLI. After <superscript>177</superscript> Lu-DOTATATE administration, successful cell killing of the hSSTr2 <superscript>+</superscript> ESCs was achieved both in vitro and in vivo , indicated by reductions in total tracer lesion uptake, BLI signal and teratoma volume. As undifferentiated hSSTr2 <superscript>+</superscript> ESCs are not therapeutically relevant, they were differentiated towards CMs and injected in immune-deficient mice with a MI. Long-term cell survival could be monitored without uncontrolled cell proliferation. However, no improvement in the left ventricular ejection fraction was observed. Conclusion: We developed isogenic hSSTr2-expressing ESCs that allow noninvasive cell monitoring in the context of PSC-derived regenerative therapy. Furthermore, we are the first to use the hSSTr2 not only as an imaging reporter gene, but also as a suicide mechanism for radionuclide therapy in the setting of PSC-derived cell treatment.<br />Competing Interests: Competing interests: Katrien Neyrinck works as an aspirant for the FWO, while both Natacha Breuls and Bryan Holvoet work under an SB-grant of the FWO. Christophe M. Deroose is a consultant for Advanced Accelerator Applications (AAA) and Novartis, who hold rights to commercialize 177Lu-DOTATATE. He is also a consultant for Sirtex, Bayer, Ipsen and Terumo. He received travel grants from GE Healthcare. No other potential conflict of interest relevant to this article was reported.

Details

Language :
English
ISSN :
1838-7640
Volume :
8
Issue :
10
Database :
MEDLINE
Journal :
Theranostics
Publication Type :
Academic Journal
Accession number :
29774076
Full Text :
https://doi.org/10.7150/thno.22980