Skip to main content

Advertisement

Log in

MRI with Magnetic Nanoparticles Monitors Downstream Anti-Angiogenic Effects of mTOR Inhibition

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

Abstract

Purpose

To study the effect of mammalian target of rapamycin (mTOR) inhibition on angiogenesis with magnetic resonance imaging (MRI) using magnetic iron oxide nanoparticles (MNP).

Procedures

One million CAK-1 renal cell carcinoma cells were subcutaneously implanted into each of 20 nude mice. When tumors reached ∼750 μl, four daily treatment arms began and continued for 4 weeks: rapamycin (mTOR inhibitor) 10 mg/kg/day; sorafenib (VEGF inhibitor) high dose (80 mg/kg/day) and low dose (30 mg/kg/day); and saline control. Weekly MRI (4.7 T Bruker Pharmascan) was performed before and after IV MION-48, a prototype MNP similar to MNP in clinical trials. Vascular volume fraction (VVF) was quantified as ΔR2 (from multi-contrast T2 sequences) and normalized to assumed muscle VVF of 3%. Linear regression compared VVF to microvascular density (MVD) as determined by histology.

Results

VVF correlated with MVD (R2 = 0.95). VVF in all treatment arms differed from control (p < 0.05) and declined weekly with treatment. VVF changes with rapamycin were similar to high-dose sorafenib.

Conclusion

This study demonstrates noninvasive, in vivo anti-angiogenic monitoring using MRI of mTOR inhibition.

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.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Escudier B et al (2007) Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 356:125–134

    Article  PubMed  CAS  Google Scholar 

  2. Hudes G et al (2007) Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 356:2271–2281

    Article  PubMed  CAS  Google Scholar 

  3. Motzer RJ et al (2007) Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 356:115–124

    Article  PubMed  CAS  Google Scholar 

  4. Jac J et al (2007) A phase II trial of RAD001 in patients with metastatic renal cell carcinoma (MRCC). Proc Am Soc Clin Oncol 25 (18S): 5107

  5. Lamuraglia M et al (2006) To predict progression-free survival and overall survival in metastatic renal cancer treated with sorafenib: pilot study using dynamic contrast-enhanced Doppler ultrasound. Eur J Cancer 42:2472–2479

    Article  PubMed  CAS  Google Scholar 

  6. Marzola P et al (2004) In vivo assessment of antiangiogenic activity of SU6668 in an experimental colon carcinoma model. Clin Cancer Res 10:739–750

    Article  PubMed  CAS  Google Scholar 

  7. Marzola P et al (2005) Early antiangiogenic activity of SU11248 evaluated in vivo by dynamic contrast-enhanced magnetic resonance imaging in an experimental model of colon carcinoma. Clin Cancer Res 11:5827–5832

    Article  PubMed  CAS  Google Scholar 

  8. Del Bufalo D et al (2006) Antiangiogenic potential of the mammalian target of rapamycin inhibitor temsirolimus. Cancer Res 66:5549–5554

    Article  PubMed  Google Scholar 

  9. Thomas GV et al (2006) Hypoxia-inducible factor determines sensitivity to inhibitors of mTOR in kidney cancer. Nat Med 12:122–127

    Article  PubMed  CAS  Google Scholar 

  10. Heng DY, Bukowski RM (2008) Anti-angiogenic targets in the treatment of advanced renal cell carcinoma. Curr Cancer Drug Targets 8:676–682

    Article  PubMed  CAS  Google Scholar 

  11. Lainakis G, Bamias A (2008) Targeting angiogenesis in renal cell carcinoma. Curr Cancer Drug Targets 8:349–358

    Article  PubMed  CAS  Google Scholar 

  12. Lane HA et al (2009) mTOR inhibitor RAD001 (everolimus) has antiangiogenic/vascular properties distinct from a VEGFR tyrosine kinase inhibitor. Clin Cancer Res 15:1612–1622

    Article  PubMed  CAS  Google Scholar 

  13. Lee DF, Hung MC (2007) All roads lead to mTOR: integrating inflammation and tumor angiogenesis. Cell Cycle 6:3011–3014

    Article  PubMed  CAS  Google Scholar 

  14. Mabuchi S et al (2007) RAD001 (Everolimus) delays tumor onset and progression in a transgenic mouse model of ovarian cancer. Cancer Res 67:2408–2413

    Article  PubMed  CAS  Google Scholar 

  15. Szczylik C, Demkow T, Staehler M (2007) Final results of the randomized phase III trial of sorafenib in advanced renal cell carcinoma: survival and biomarker analysis. Proc Am Soc Clin Oncol Meet

  16. Flaherty KT et al (2008) Pilot study of DCE-MRI to predict progression-free survival with sorafenib therapy in renal cell carcinoma. Cancer Biol Ther 7:496–501

    Article  PubMed  CAS  Google Scholar 

  17. Rosen MA, Schnall MD (2007) Dynamic contrast-enhanced magnetic resonance imaging for assessing tumor vascularity and vascular effects of targeted therapies in renal cell carcinoma. Clin Cancer Res 13:770s–776s

    Article  PubMed  CAS  Google Scholar 

  18. Schnell CR et al (2008) Effects of the dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 on the tumor vasculature: implications for clinical imaging. Cancer Res 68:6598–6607

    Article  PubMed  CAS  Google Scholar 

  19. Bremer C et al (2003) Steady-state blood volume measurements in experimental tumors with different angiogenic burdens a study in mice. Radiology 226:214–220

    Article  PubMed  Google Scholar 

  20. Guimaraes AR et al (2008) Magnetic resonance imaging monitors physiological changes with antihedgehog therapy in pancreatic adenocarcinoma xenograft model. Pancreas 37:440–444

    Article  PubMed  CAS  Google Scholar 

  21. Persigehl T et al (2007) Antiangiogenic tumor treatment: early noninvasive monitoring with USPIO-enhanced MR imaging in mice. Radiology 244:449–456

    Article  PubMed  Google Scholar 

  22. Boxerman J et al (1995) MR contrast due to intravascular magnetic susceptibility perturbations. Magn Reson Med 34:555–566

    Article  PubMed  CAS  Google Scholar 

  23. Dennie J et al (1998) NMR imaging of changes in vascular morphology due to tumor angiogenesis. Magn Reson Med 40:793–799

    Article  PubMed  CAS  Google Scholar 

  24. Tang Y et al (2005) In vivo assessment of RAS-dependent maintenance of tumor angiogenesis by real-time magnetic resonance imaging. Cancer Res 65:8324–8330

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge grant support from the Dana Farber Renal Spore where Dr. Ross, received funding to support this work from a career development award. In addition, the authors would like to acknowledge funding support from the AstraZeneca Pharmaceuticals, Inc. where Dr. Guimaraes received funding to support part of this work. Lastly, the authors would like to acknowledge Claire Kaufman and Carlos Rangel for technological support in imaging and data analysis

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alexander R. Guimaraes.

Additional information

Alexander R. Guimaraes and Robert Ross contributed equally as first authors to this article.

Funding

Funding provided by the Renal Spore-Dana Farber Cancer Institute & MGH-AstraZeneca Strategic Alliance

Brief Article

Significance—showing the application of steady state MRI with magnetic nanoparticles to monitor the anti-angiogenic effect of rapamycin on xenograft model in vivo.

Appendix

Appendix

The theory behind the use of superparamagnetic contrast agents with long vascular half lives to determine blood volumes by MRI, which is supported by detailed numerical simulations, is that the change in gradient-echo transverse relaxation rate (ΔR 2*) relative to the pre-injection baseline is proportional to the local blood volume times some function of the plasma concentration of the agent or \( \Delta {R_2} {*} = k \times f(P) \times V \). If a steady state concentration of the contrast agent is assumed in the blood plasma then there is a simple linear relationship between ΔR 2* and blood volume at any time (t) and the formula reduces to \( \Delta {R_2} {*} (t) = K \times V(t) \). Stated another way, \( V(t) = {{{\left[ {{{\Delta}}{{\hbox{R}}_2} {*} (t)} \right]}} \left/ {K} \right.} \) where V(t) is the blood volume, ΔR 2* (t) is the change in the transverse relaxation rate of the region, and the constant K includes the contrast agent blood pool concentration and is therefore dose dependent. While this technique allows for easy measurement of total blood volume in a given voxel, tissue slice, or entire organs, additional methods more sensitive to microvessels have also been developed.

These methods are based on the property that compartmentalization of these magnetic nanoparticles also induces long-range magnetic field perturbations that extend over many microns and increase both transverse relaxation rates, R 2 and R 2*, of the tissues. The enhancement of R 2 and R 2* caused by these agents can be expressed as follows: \( \Delta {R_2} = { }{{{1}} \left/ {{T{{2}_{\rm{post}}}}} \right.}-{{{1}} \left/ {{T{{2}_{\rm{pre}}}}} \right.} \approx - {{{1}} \left/ {\hbox{TE}} \right.}{\, \ln }\left( {{{{{S_{\rm{post}}}}} \left/ {{{S_{\rm{pre}}}}} \right.}} \right) \) and \( \Delta {R_2} {*} { } = {{{1}} \left/ {{{\hbox{T2}}{{*}_{\hbox{post}}}}} \right.}-{{{1}} \left/ {{{\hbox{T2}}{{*}_{\hbox{pre}}}}} \right.} \approx {{{ - {1}}} \left/ {\hbox{TE}} \right.}{ \ln }\left( {{{{{S_{\hbox{post}}}}} \left/ {{{S_{\hbox{pre}}}}} \right.}} \right) \) where S is the signal intensity, TE the echo time, and T2 is the transverse relaxation time. It has previously been shown that there exists a unique relationship between ∆R 2 (spin echo), ∆R 2* (gradient echo) as a function of vessel diameter and contrast agent concentration. The ∆R 2 peaks for vessels 1-2 μm in diameter whereas ∆R 2* is fairly independent of vessel size beyond 3-4 μm. These studies thus provide the rationale for spin-echo imaging being more sensitive to microvasculature, while gradient-echo imaging is more sensitive to total vasculature. Ultimately, gradient echo/spin ratio imaging could theoretically be utilized to measure vessel size noninvasively, as the ΔR 2*/ΔR 2 ratio increases nearly linearly with vessel size. Sequestered magnetic nanoparticles, as occurs with uptake by macrophages, may also be detected based on long-range magnetic field perturbations utilizing a similar approach.

Therefore, if the R 2 is calculated prior to and following administration of MNP, then the ∆R 2 can be determined accurately for both the tumor, over multiple slices, and the musculature. Following this conversion, a straightforward proportionality, utilizing a well-accepted muscular VVF of 3%, can be performed.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Guimaraes, A.R., Ross, R., Figuereido, J.L. et al. MRI with Magnetic Nanoparticles Monitors Downstream Anti-Angiogenic Effects of mTOR Inhibition. Mol Imaging Biol 13, 314–320 (2011). https://doi.org/10.1007/s11307-010-0357-2

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11307-010-0357-2

Key words

Navigation