Skip to main content

Advertisement

Log in

2-Deoxy-2-[18F]fluoro-d-glucose Positron Emission Tomography Demonstrates Target Inhibition with the Potential to Predict Anti-Tumour Activity Following Treatment with the AKT Inhibitor AZD5363

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

Abstract

Purpose

The phosphatidyl inositol 3 kinase, AKT and mammalian target of rapamycin are frequently deregulated in human cancer and are among one of the most promising targets for cancer therapy. AZD5363 (AstraZeneca) is an AKT inhibitor in phase 1 clinical trials. Given its utility in assessing glucose metabolism, we investigated the role of 2-Deoxy-2-[18F]fluoro-d-glucose (18F-FDG) positron emission tomography (PET) as a biomarker to demonstrate target inhibition and its potential to predict and demonstrate the anti-tumour activity of AZD5363.

Methods

18F-FDG PETscans were performed in nude mice in a number of xenograft models (U87-MG glioblastoma, BT474C breast carcinoma and Calu-6 lung). Mice were fasted prior to imaging, and either static or dynamic 18F-FDG PET imaging was performed.

Results

We have shown that 18F-FDG uptake in tumour xenografts was reduced by 39 % reduction compared to vehicle after a single dose of AZD5363, demonstrating activation of the AKT pathway after only 4 h of dosing. Multiple doses of AZD5363 showed an anti-tumour volume effect and a reduction in 18F-FDG uptake (28 % reduction compared to vehicle), highlighting the potential of 18F-FDG PET as an efficacy biomarker. Furthermore, the degree of inhibition of 18F-FDG uptake corresponded with the sensitivity of the tumour model to AZD5363. The use of dynamic 18F-FDG PET and a two-compartmental analysis identified the mechanism of this change to be due to a change in cellular uptake of 18F-FDG following administration of AZD5363.

Conclusions

We conclude that 18F-FDG PET is a promising pharmacodynamic biomarker of AKT pathway inhibition, with potential to predict and demonstrate anti-tumour activity. It is a biomarker that may stop ineffective drug schedules, helping to make early stop decisions and identify responding subsets of patients, resulting in improved clinical decision making both during drug development and patient management.

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
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Luo J, Manning BD, Cantley LC (2003) Targeting the PI3K-Akt pathway in human cancer: rationale and promise. Cancer Cell 4:257–262

    Article  PubMed  CAS  Google Scholar 

  2. Marone R, Cmiljanovic V, Giese B, Wymann MP (2008) Targeting phosphoinositide 3-kinase: moving towards therapy. Biochim Biophys Acta 1784:159–185

    Article  PubMed  CAS  Google Scholar 

  3. Yuan TL, Cantley LC (2008) PI3K pathway alterations in cancer: variations on a theme. Oncogene 27:5497–5510

    Article  PubMed  CAS  Google Scholar 

  4. Markman B, Dienstmann R, Tabernero J (2010) Targeting the PI3K/AKT/mTOR pathway beyond rapalogs. Oncotarget 1:530–543

    PubMed  Google Scholar 

  5. Hirai H, Sootome H, Nakatsuru Y, Miyama K et al (2010) MK-2206, an allosteric AKT inhibitor, enhances tumour efficacy by standard chemotherapeutic agents or molecular targeted drugs in-vitro and in-vivo. Mol Cancer Ther 9:1956–1967

    Google Scholar 

  6. Davies BR, Greenwood H, Dudley P, Crafter C et al (2012) Preclinical Pharmacology of AZD5363: an inhibitor of AKT: pharmacodynamics, antitumour activity, and correlation of monotherapy activity with genetic background. Mol Cancer Ther 11:873–887

    Article  PubMed  CAS  Google Scholar 

  7. Wahl RL, Zasadny K, Helvie M, Hutchins GD et al (1993) Metabolic monitoring of breast cancer chemohormonotherapy using positron emission tomography: initial evaluation. J Clin Oncol 11:2101–2111

    PubMed  CAS  Google Scholar 

  8. Kidd EA, Siegel BA, Dehdashti F, Grigsby PW (2007) The standardised uptake value for F-18 fluorodeoxyglucose is a sensitive predictive biomarker for cervical cancer treatment response and survival. Cancer 15:1738–1744

    Article  Google Scholar 

  9. Larson SM, Schwartz LH (2006) 18F-FDG PET as a candidate for “Qualified Biomarker”: functional assessment of treatment response in oncology. J Nucl Med 47:901–903

    PubMed  CAS  Google Scholar 

  10. Stroobants S, Goeminne J, Seegers M, Dimitrijevic S et al (2003) 18FDG Positron emission tomography for the early prediction of response in advanced soft tissue sarcoma treated with imatinib mesylate (Glivec). Eur J Cancer 39:2012–2020

    Article  PubMed  CAS  Google Scholar 

  11. Sunaga N, Oricuhi N, Kaira K, Yanagitani N et al (2008) Usefulness of FDG-PET for early prediction of the response to Gefitinib in non small cell lung cancer. Lung Cancer 59:203–210

    Article  PubMed  Google Scholar 

  12. Bendell JC, Rodon J, Burris HA, de Jonge M et al (2012) Phase 1, dose-escalation study of BKM120, an oral pan-class I PI3K inhibitor, in patients with advanced solid tumours. J Clin Oncol 30:282–290

    Article  PubMed  CAS  Google Scholar 

  13. Young H, Baum R, Cremerius U, Herholz K et al (1999) Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: review and 1999 EORTC recommendations. European Organisation for Research and Treatment of Cancer (EORTC) PET Study Group. Eur J Cancer 35:1773–1782

    Article  PubMed  CAS  Google Scholar 

  14. Roe K, Aleksandersen TB, Kristian A, Nilsen LB et al (2010) Pre-clinical dynamic 18F-FDG PET—tumour characterization and radiotherapy response assessment by kinetic compartment analysis. Acta Oncol 49:914–921

    Article  PubMed  Google Scholar 

  15. Manning BD, Cantley LC (2007) AKT/PKB signaling: navigating downstream. Cell 29:1261–1274

    Article  Google Scholar 

  16. Elstrom RL, Bauer DE, Buzzai M, Karnauskas R et al (2004) AKT stimulates aerobic glycolysis in cancer cells. Cancer Res 64:3892–3899

    Article  PubMed  CAS  Google Scholar 

  17. Ma WW, Jacene H, Song D, Vilardell F et al (2009) 18F Fluorodeoxyglucose positron emission tomography correlates with AKT pathway activity but is not predictive of clinical outcome during mTOR inhibitor therapy. J Clin Oncol 27:2697–2704

    Article  PubMed  CAS  Google Scholar 

  18. Nogova L, Boellaard R, Kobe C, Hoetjes N et al (2009) Downregulation of 18F-FDG uptake in PET as an early pharmacodynamic effect in treatment of non-small cell lung cancer with the mTOR inhibitor everolimus. J Nucl Med 50:1815–1819

    Article  PubMed  Google Scholar 

  19. Bao Q, Newport D, Chen M, Stout D et al (2009) Performance evaluation of the inveon dedicated PET preclinical tomograph based on the NEMA NU-4 standards. J Nucl Med 50:401–408

    Article  PubMed  Google Scholar 

  20. Gambhir S (2004) Quantitative assay development for PET. In: Phelps ME (ed) PET: molecular imaging and its biological applications. Springer-Verlag, Berlin Germany, pp 125–216

    Google Scholar 

  21. Laforest R, Sharp TL, Engelbach JA, Fettia NM et al (2005) Measurement of input functions in rodents: challenges and solutions. Nucl Med Biol 32:679–685

    Article  PubMed  CAS  Google Scholar 

  22. Nguyen QD, Perumal M, Waldman TA, Abogaye EO (2011) Glucose metabolism measured by [18F] fluorodeoxyglucose positron emission tomography is independent of PTEN/AKT status in human colon carcinoma cells. Transl Oncol 4:241–248

    PubMed  Google Scholar 

  23. Keen H, Ricketts SA, Bales J, Shannon A et al (2009) The mTOR kinase inhibitor AZD8055 modulates 18 F-FDG uptake in vivo in the human glioma xenograft model U87-MG. Mol Cancer Ther 8(supplement 1):A225

    Article  Google Scholar 

  24. Wei LH, Su H, Hildebrandt IJ, Phelps ME et al (2008) Changes in tumour metabolism as readout for mammalian target of rapamycin kinase inhibition by rapamycin in glioblastoma. Clin Cancer Res 14:3416–3426

    Article  PubMed  CAS  Google Scholar 

  25. Nogova L, Gross SH, Dimitrijevic et al (2008). Pharmacodynamics of RAD001 measured by early FDG-PET in patients with recurrent NSCLC (abstract). J Clin Oncol 26(May 20 suppl): Abstract 14616

  26. Contractor KN, Aboagye EO (2009) Monitoring predominantly cytostatic treatment response with 18F-FDG PET. J Nucl Med 50:97S–105S

    Article  PubMed  CAS  Google Scholar 

  27. Benz MR, Czernin J, Allen Auebrach MS, Itap UD et al (2009) FDG-PET/CT imaging predicts histopathological treatment responses after the initial cycle of neoadjuvant therapy in high grade soft-tissue sarcomas. Clin Cancer Res 15:2856–2863

    Article  PubMed  CAS  Google Scholar 

  28. De Geus Oei LF, Vriens D, Van Laarloven HW, Van der Graaf WT et al (2009) Monitoring and predicting response to therapy with 18F-FDG PET in colorectal cancer: a systematic review. J Nucl Med 50:543–554

    Google Scholar 

  29. Cheebsumon P, Velasquez LM, Hoekstra CJ, Hayes W et al (2011) Measuring response to therapy using 18F-FDG PET. Semi-quantitative and full kinetic analysis. Eur J Nucl Med Mol Imaging 38:832–842

    Article  PubMed  CAS  Google Scholar 

  30. Minn H, Leskinen-Kallio S, Lindholm P, Bergmani J et al (1993) 18F Flourodeoxyglucose uptake in tumours. Kinetic vs steady state methods with reference to plasma insulin. J Comput Assist Tomgr 17:115–123

    Article  CAS  Google Scholar 

  31. Minn H, Zasadny KR, Quint LE, Wahl RL (1995) Lung cancer: reproducibility of quantitative measurements for evaluating 2[F18]-fluorodeoxyglucose uptake of PET. Radiology 196:167–173

    PubMed  CAS  Google Scholar 

  32. Plathow C, Weber WA (2008) Tumour cell metabolism imaging. J Nucl Med 49(Suppl2):43S–63S

    Article  PubMed  CAS  Google Scholar 

  33. Zhao S, Kuge Y, Mochizuki T, Takahashi T et al (2005) Biologic correlates of intra-tumoural heterogeneity in 18F-FDG distribution with regional expression of glucose transporters and hexokinase II in experimental tumour. J Nucl Med 46:675–682

    PubMed  CAS  Google Scholar 

  34. Pugachev A, Ruan S, Carlin S, Larson SM et al (2005) Dependence of FDG uptake on tumour micro-environment. Int J Radiat Oncol Biol Phys 62:545–553

    Article  PubMed  CAS  Google Scholar 

  35. Tseng J, Dunnwald LK, Schubert EK, Link JM et al (2004) 18F-FDG kinetics in locally advanced breast cancer: correlation with tumour blood flow and changes in response to neoadjuvant chemotherapy. J Nucl Med 45:1829–1837

    PubMed  CAS  Google Scholar 

  36. Phelps ME, Huang SC, Hoffman EJ, Selin MS et al (1999) Tomographic measurement of local cerebral glucose metabolic rate in humans with (F18) 2-flourodeoxyglucose: validation of method. Ann Neurol 6:371–388

    Article  Google Scholar 

Download references

Acknowledgments

AZD5363 was discovered by AstraZeneca subsequent to a collaboration with Astex Therapeutics. The authors thank Neill Gingles, Leigh Williams, Gareth Parker and Heather Keen for their contribution to the in vivo imaging procedures.

Conflict of Interest

The authors declare that they have no conflicts of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Juliana Maynard.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Maynard, J., Ricketts, SA., Gendrin, C. et al. 2-Deoxy-2-[18F]fluoro-d-glucose Positron Emission Tomography Demonstrates Target Inhibition with the Potential to Predict Anti-Tumour Activity Following Treatment with the AKT Inhibitor AZD5363. Mol Imaging Biol 15, 476–485 (2013). https://doi.org/10.1007/s11307-013-0613-3

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11307-013-0613-3

Key words

Navigation