Skip to main content

Advertisement

Log in

124I-PET Assessment of Human Sodium Iodide Symporter Reporter Gene Activity for Highly Sensitive In Vivo Monitoring of Teratoma Formation in Mice

  • Research Article
  • Published:
Molecular Imaging and Biology Aims and scope Submit manuscript

Abstract

Purpose

Pluripotent stem cell (PSC)-based therapies possess great potential to restore the function of irreversibly damaged organs. PSCs can be differentiated in vitro into any cell type. However, pluripotent potential bears the risk of teratoma formation. In vivo monitoring of teratoma formation is indispensable, as 100 % purity of the cell preparation cannot be achieved. We aimed at establishing the human sodium iodide symporter (hNIS) as reporter gene for PET monitoring of teratoma formation.

Procedures

Murine PSC stably expressing hNIS were injected into the hind limbs of SCID mice to induce teratoma formation. Positron emission tomography (PET) scans were acquired weekly between days 14 and 42 after transplantation. Two teratomas were excised at each time point for histology and size measurement. Tracer uptake was correlated with teratoma weight. Specificity of tumoural iodine uptake was assessed by blocking hNIS in vivo with perchlorate.

Results

Neither hNIS expression nor I-124 exposure adversely impacted viability or differentiation potential of PSCs. Iodine uptake was highly specific in teratomas, as in vivo blocking of hNIS with perchlorate led to uptake rates comparable to tracer uptake in non-transgene tumours. Tumour mass and tracer uptake showed a positive correlation.

Conclusions

This is the first study to generate stably hNIS-expressing murine PSCs. Since the differentiation potential was preserved, hNIS-expressing cells are suitable for PSC-based forward programming approaches. Teratoma formation from undifferentiated cells can be monitored in vivo by PET with high specificity on a quantitative level. Due to its anticipated lack of immunogenicity in humans, hNIS is a promising reporter gene for clinical translation.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1.
Fig. 2.
Fig. 3.
Fig. 4.
Fig. 5.
Fig. 6.
Fig. 7.

Similar content being viewed by others

Abbreviations

%ID:

Percentage of the injected dose

I-124:

Iodine-124

I-125:

Iodine-125

BMSC:

Bone marrow stem cells

DMEM:

Dulbecco’s modified eagle medium

EB:

Embryoid body

ESC:

Embryonic stem cells

[18F]FDG:

2-Deoxy-2-[18F]fluoro-d-glucose

hNIS:

Human sodium iodide symporter

iPS:

Induced pluripotent stem cells

LAD:

Left anterior descending artery

MAP:

Maximum a posteriori

MI:

Myocardial infarction

OSEM:

Ordered subset expectation maximization

PBS:

Phosphate buffered saline

PET:

Positron emission tomography

PFA:

Paraformaldehyde

PSC:

Pluripotent stem cells

SCID:

Severe combined immunodeficiency

VOI:

Volume of interest

[18F]FHBG:

9-(4-[18F]fluoro-3-[hydroxymethyl]butyl)guanine

References

  1. David R, Stieber J, Fischer E et al (2009) Forward programming of pluripotent stem cells towards distinct cardiovascular cell types. Cardiovasc Res 84:263–272

    Article  CAS  PubMed  Google Scholar 

  2. Yang D, Zhang Z-J, Oldenburg M et al (2008) Human embryonic stem cell-derived dopaminergic neurons reverse functional deficit in parkinsonian rats. Stem Cells 26:55–63

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  3. Craft AM, Ahmed N, Rockel JS et al (2013) Specification of chondrocytes and cartilage tissues from embryonic stem cells. Development 140:2597–2610

    Article  CAS  PubMed  Google Scholar 

  4. Mauritz C, Schwanke K, Reppel M et al (2008) Generation of functional murine cardiac myocytes from induced pluripotent stem cells. Circulation 118:507–517

    Article  PubMed  Google Scholar 

  5. Roessler R, Smallwood SA, Veenvliet JV et al (2014) Detailed analysis of the genetic and epigenetic signatures of iPSC-derived mesodiencephalic dopaminergic neurons. Stem Cell Rep 2:520–533

    Article  CAS  Google Scholar 

  6. Wei Y, Zeng W, Wan R et al (2012) Chondrogenic differentiation of induced pluripotent stem cells from osteoarthritic chondrocytes in alginate matrix. Eur Cell Mater 23:1–12

    CAS  PubMed  Google Scholar 

  7. Ieda M, Fu J-D, Delgado-Olguin P et al (2010) Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142:375–386

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  8. Caiazzo M, Dell’Anno MT, Dvoretskova E et al (2011) Direct generation of functional dopaminergic neurons from mouse and human fibroblasts. Nature 476:224–227

    Article  CAS  PubMed  Google Scholar 

  9. Outani H, Okada M, Yamashita A et al (2013) Direct induction of chondrogenic cells from human dermal fibroblast culture by defined factors. PLoS ONE 8, e77365

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  10. Kolossov E, Bostani T, Roell W et al (2006) Engraftment of engineered ES cell-derived cardiomyocytes but not BM cells restores contractile function to the infarcted myocardium. J Exp Med 203:2315–2327

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  11. Moon J, Lee H-S, Kang JM et al (2013) Stem cell grafting improves both motor and cognitive impairments in a genetic model of Parkinson’s disease, the aphakia (ak) mouse. Cell Transplant 22:1263–1279

    Article  PubMed Central  PubMed  Google Scholar 

  12. Marini JC, Forlino A (2012) Replenishing cartilage from endogenous stem cells. N Engl J Med 366:2522–2524

    Article  CAS  PubMed  Google Scholar 

  13. Atala A (2012) Human embryonic stem cells: early hints on safety and efficacy. Lancet 379:689–690

    Article  PubMed  Google Scholar 

  14. Cyranoski D (2013) Stem cells cruise to clinic. Nature 494:413

    Article  CAS  PubMed  Google Scholar 

  15. David R, Schwarz F, Rimmbach C et al (2013) Selection of a common multipotent cardiovascular stem cell using the 3.4-kb MesP1 promoter fragment. Basic Res Cardiol 108:312

    Article  PubMed  Google Scholar 

  16. Bondue A, Tännler S, Chiapparo G et al (2011) Defining the earliest step of cardiovascular progenitor specification during embryonic stem cell differentiation. J Cell Biol 192:751–765

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  17. Su W, Zhou M, Zheng Y et al (2011) Bioluminescence reporter gene imaging characterize human embryonic stem cell-derived teratoma formation. J Cell Biochem 112:840–848

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  18. Cao F, Li Z, Lee A et al (2009) Noninvasive de novo imaging of human embryonic stem cell-derived teratoma formation. Cancer Res 69:2709–2713

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  19. Cao F, Lin S, Xie X et al (2006) In vivo visualization of embryonic stem cell survival, proliferation, and migration after cardiac delivery. Circulation 113:1005–1014

    Article  PubMed  Google Scholar 

  20. Pomper MG, Hammond H, Yu X et al (2009) Serial imaging of human embryonic stem-cell engraftment and teratoma formation in live mouse models. Cell Res 19:370–379

    Article  CAS  PubMed  Google Scholar 

  21. Dohán O, la Vieja DA, Paroder V et al (2003) The sodium/iodide Symporter (NIS): characterization, regulation, and medical significance. Endocr Rev 24:48–77

    Article  PubMed  Google Scholar 

  22. Terrovitis J, Kwok KF, Lautamäki R et al (2008) Ectopic expression of the sodium-iodide symporter enables imaging of transplanted cardiac stem cells in vivo by single-photon emission computed tomography or positron emission tomography. J Am Coll Cardiol 52:1652–1660

    Article  PubMed  Google Scholar 

  23. Miyagawa M, Anton M, Wagner B et al (2005) Non-invasive imaging of cardiac transgene expression with PET: comparison of the human sodium/iodide symporter gene and HSV1-tk as the reporter gene. Eur J Nucl Med Mol Imaging 32:1108–1114

    Article  PubMed  Google Scholar 

  24. Müller M, Fleischmann BK, Selbert S et al (2000) Selection of ventricular-like cardiomyocytes from ES cells in vitro. FASEB J 14:2540–2548

    Article  PubMed  Google Scholar 

  25. David R, Jarsch VB, Schwarz F et al (2011) Induction of MesP1 by Brachyury(T) generates the common multipotent cardiovascular stem cell. Cardiovasc Res 92:115–122

    Article  CAS  PubMed  Google Scholar 

  26. Spitzweg C, Baker CH, Bergert ER et al (2007) Image-guided radioiodide therapy of medullary thyroid cancer after carcinoembryonic antigen promoter-targeted sodium iodide symporter gene expression. Hum Gene Ther 18:916–924

    Article  CAS  PubMed  Google Scholar 

  27. Scholz IV, Cengic N, Baker CH et al (2005) Radioiodine therapy of colon cancer following tissue-specific sodium iodide symporter gene transfer. Gene Ther 12:272–280

    Article  CAS  PubMed  Google Scholar 

  28. Klutz K, Russ V, Willhauck MJ et al (2009) Targeted radioiodine therapy of neuroblastoma tumors following systemic nonviral delivery of the sodium iodide symporter gene. Clin Cancer Res 15:6079–6086

    Article  CAS  PubMed  Google Scholar 

  29. Thomson JA, Itskovitz-Eldor J, Shapiro SS et al (1998) Embryonic stem cell lines derived from human blastocysts. Science 282:1145–1147

    Article  CAS  PubMed  Google Scholar 

  30. Frantz S (2012) Embryonic stem cell pioneer Geron exits field, cuts losses. Nat Biotechnol 30:12–13

    Article  CAS  PubMed  Google Scholar 

  31. Amariglio N, Hirshberg A, Scheithauer BW et al (2009) Donor-derived brain tumor following neural stem cell transplantation in an ataxia telangiectasia patient. PLoS Med 6, e1000029

    Article  PubMed Central  PubMed  Google Scholar 

  32. Schwartz SD, Hubschman J-P, Heilwell G et al (2012) Embryonic stem cell trials for macular degeneration: a preliminary report. Lancet 379:713–720

    Article  CAS  PubMed  Google Scholar 

  33. Lukovic D, Stojkovic M, Moreno-Manzano V et al (2014) Perspectives and future directions of human pluripotent stem cell-based therapies: lessons from Geron’s clinical trial for spinal cord injury. Stem Cells Dev 23:1–4

    Article  PubMed  Google Scholar 

  34. Lee AS, Tang C, Cao F et al (2009) Effects of cell number on teratoma formation by human embryonic stem cells. Cell Cycle 8:2608–2612

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  35. Knoepfler PS (2009) Deconstructing stem cell tumorigenicity: a roadmap to safe regenerative medicine. Stem Cells 27:1050–1056

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  36. Prokhorova TA, Harkness LM, Frandsen U et al (2009) Teratoma formation by human embryonic stem cells is site dependent and enhanced by the presence of Matrigel. Stem Cells Dev 18:47–54

    Article  CAS  PubMed  Google Scholar 

  37. Castro CA, Ben-Yehudah A, Ozolek JA et al (2010) Semiquantitative histopathology and 3D magnetic resonance microscopy as collaborative platforms for tissue identification and comparison within teratomas derived from pedigreed primate embryonic stem cells. Stem Cell Res 5:201–211

    Article  PubMed Central  PubMed  Google Scholar 

  38. Lee AS, Tang C, Rao MS et al (2013) Tumorigenicity as a clinical hurdle for pluripotent stem cell therapies. Nat Med 19:998–1004

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  39. Baril P, Martin-Duque P, Vassaux G (2010) Visualization of gene expression in the live subject using the Na/I symporter as a reporter gene: applications in biotherapy. Br J Pharmacol 159:761–771

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  40. Yaghoubi SS, Campbell DO, Radu CG, Czernin J (2012) Positron emission tomography reporter genes and reporter probes: gene and cell therapy applications. Theranostics 2:374–391

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  41. Chen EQ, MacIntyre WJ, Go RT et al (1997) Myocardial viability studies using fluorine-18-FDG SPECT: a comparison with fluorine-18-FDG PET. J Nucl Med 38:582–586

    CAS  PubMed  Google Scholar 

  42. Knesaurek K, Machac J (2006) Comparison of 18F SPECT with PET in myocardial imaging: a realistic thorax-cardiac phantom study. BMC Nucl Med 6:5

    Article  PubMed Central  PubMed  Google Scholar 

  43. Valentin J (2003) Relative biological effectiveness (RBE), quality factor (Q), and radiation weighting factor (wR). Ann ICRP 33:1–121

    Google Scholar 

  44. Protection R (2007) ICRP Publication 103. Ann ICRP

  45. Freudenberg LS, Jentzen W, Stahl A et al (2011) Clinical applications of 124I-PET/CT in patients with differentiated thyroid cancer. Eur J Nucl Med Mol Imaging 38(Suppl 1):S48–S56

    Article  PubMed  Google Scholar 

  46. Templin C, Zweigerdt R, Schwanke K et al (2012) Transplantation and tracking of human-induced pluripotent stem cells in a pig model of myocardial infarction: assessment of cell survival, engraftment, and distribution by hybrid single photon emission computed tomography/computed tomography of sodium iodide symporter transgene expression. Circulation 126:430–439

    Article  CAS  PubMed  Google Scholar 

  47. Rodriguez-Porcel M (2010) In vivo imaging and monitoring of transplanted stem cells: clinical applications. Curr Cardiol Rep 12:51–58

    Article  PubMed Central  PubMed  Google Scholar 

  48. Wu JC, Spin JM, Cao F et al (2006) Transcriptional profiling of reporter genes used for molecular imaging of embryonic stem cell transplantation. Physiol Genomics 25:29–38

    Article  PubMed  Google Scholar 

  49. Wu JC, Cao F, Dutta S et al (2006) Proteomic analysis of reporter genes for molecular imaging of transplanted embryonic stem cells. Proteomics 6:6234–6249

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Lubberink M, Herzog H (2011) Quantitative imaging of 124I and 86Y with PET. Eur J Nucl Med Mol Imaging 38(Suppl 1):S10–S18

    Article  PubMed  Google Scholar 

Download references

Acknowledgments

We thank Christiane Groß, Barbara Markieton and Judith Arcifa for the expert technical assistance. This work was supported by the FöFoLe Program of the LMU Munich [C.L. and W.M.F.], the BMBF [01GN0960 to R.D. and W.M.F.] and the Deutsche Forschungsgemeinschaft [DA 1296/2-1 to R.D. and FR 705/14-2 to W.M.F.]. This work has been funded by the Federal Ministry of Education and Research Germany (FKZ 0312138A and FKZ 316159) and the State Mecklenburg-Western Pomerania with EU Structural Funds (ESF/IV-WM-B34-0030/10 and ESF/IV-BM-B35-0010/12), by the DFG (DA 1296-1) and the German Heart Foundation (F/01/12). A substantial part of this work originated from the doctoral thesis of Georgios Kaissis.

Conflict of Interest

The authors declare that they have no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Robert David.

Additional information

Robert David and Marcus Hacker share senior authorships.

Sebastian Lehner and Cajetan Lang contributed equally to this work.

Electronic supplementary material

Below is the link to the electronic supplementary material.

EB’s from transgenic ESC’s retained their cardiogenic potential in vitro, as evident from spontaneously contracting foci that occurred on day 12 of differentiation (A,B). (M4V 5188 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lehner, S., Lang, C., Kaissis, G. et al. 124I-PET Assessment of Human Sodium Iodide Symporter Reporter Gene Activity for Highly Sensitive In Vivo Monitoring of Teratoma Formation in Mice. Mol Imaging Biol 17, 874–883 (2015). https://doi.org/10.1007/s11307-015-0857-1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11307-015-0857-1

Key words

Navigation