Fluorodeoxyglucose Positron Emission Tomography Correlates With Akt Pathway Activity but Is Not Predictive of Clinical Outcome During mTOR Inhibitor Therapy

Purpose Positron emission tomography (PET) with [F]fluorodeoxyglucose (FDG-PET) has increasingly been used to evaluate the efficacy of anticancer agents. We investigated the role of FDG-PET as a predictive marker for response to mammalian target of rapamycin (mTOR) inhibition in advanced solid tumor patients and in murine xenograft models. Patients and Methods Thirty-four rapamycin-treated patients with assessable baseline and treatment FDG-PET and computed tomography scans were analyzed from two clinical trials. Clinical response was evaluated according to Response Evaluation Criteria in Solid Tumors, and FDG-PET response was evaluated by quantitative changes and European Organisation for Research and Treatment of Cancer (EORTC) criteria. Six murine xenograft tumor models were treated with temsirolimus. Small animal FDG-PET scans were performed at baseline and during treatment. The tumors were analyzed for the expression of pAkt and GLUT1. Results Fifty percent of patients with increased FDG-PET uptake and 46% with decreased uptake had progressive disease (PD). No objective response was observed. By EORTC criteria, the sensitivity of progressive metabolic disease on FDG-PET in predicting PD was 19%. Preclinical studies demonstrated similar findings, and FDG-PET response correlated with pAkt activation and plasma membrane GLUT1 expression. Conclusion FDG-PET is not predictive of proliferative response to mTOR inhibitor therapy in both clinical and preclinical studies. Our findings suggest that mTOR inhibitors suppress the formation of mTORC2 complex, resulting in the inhibition of Akt and glycolysis independent of proliferation in a subset of tumors. Changes in FDG-PET may be a pharmacodynamic marker for Akt activation during mTOR inhibitor therapy. FDG-PET may be used to identify patients with persistent Akt activation following mTOR inhibitor therapy. J Clin Oncol 27:2697-2704. © 2009 by American Society of Clinical Oncology

[1]  A. Jimeno,et al.  Pharmacodynamic-guided modified continuous reassessment method-based, dose-finding study of rapamycin in adult patients with solid tumors. , 2008, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[2]  David McDermott,et al.  Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. , 2007, The New England journal of medicine.

[3]  A. Marx,et al.  A recent application of fluoro-18-deoxyglucose positron emission tomography, treatment monitoring with a mammalian target of rapamycin inhibitor: an example of a patient with a desmoplastic small round cell tumor. , 2007, Hellenic journal of nuclear medicine.

[4]  D. Sabatini,et al.  Rapamycin derivatives reduce mTORC2 signaling and inhibit AKT activation in AML. , 2007, Blood.

[5]  D. Sabatini mTOR and cancer: insights into a complex relationship , 2006, Nature Reviews Cancer.

[6]  R. Hruban,et al.  An In vivo Platform for Translational Drug Development in Pancreatic Cancer , 2006, Clinical Cancer Research.

[7]  A. Godwin,et al.  Therapeutic effect of imatinib in gastrointestinal stromal tumors: AKT signaling dependent and independent mechanisms. , 2006, Cancer research.

[8]  D. Sabatini,et al.  Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. , 2006, Molecular cell.

[9]  R. Memmott,et al.  Handicapping the Race to Develop Inhibitors of the Phosphoinositide 3-Kinase/Akt/Mammalian Target of Rapamycin Pathway , 2006, Clinical Cancer Research.

[10]  C. Thompson,et al.  Akt-dependent transformation: there is more to growth than just surviving , 2005, Oncogene.

[11]  E. Wardelmann,et al.  Very early detection of response to imatinib mesylate therapy of gastrointestinal stromal tumours using 18fluoro-deoxyglucose-positron emission tomography. , 2005, Anticancer research.

[12]  Donghui Li,et al.  The rapamycin analog CCI-779 is a potent inhibitor of pancreatic cancer cell proliferation. , 2005, Biochemical and biophysical research communications.

[13]  E. Fiala,et al.  Induction of preneoplastic lung lesions in guinea pigs by cigarette smoke inhalation and their exacerbation by high dietary levels of vitamins C and E. , 2005, Carcinogenesis.

[14]  R. Loewith,et al.  Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive , 2004, Nature Cell Biology.

[15]  J. Testa,et al.  Frequent activation of AKT in non-small cell lung carcinomas and preneoplastic bronchial lesions. , 2004, Carcinogenesis.

[16]  A. Alavi,et al.  Akt Stimulates Aerobic Glycolysis in Cancer Cells , 2004, Cancer Research.

[17]  Jingxia Li,et al.  Nickel Compounds Act through Phosphatidylinositol-3-kinase/Akt-Dependent, p70S6k-Independent Pathway to Induce Hypoxia Inducible Factor Transactivation and Cap43 Expression in Mouse Epidermal Cl41 Cells , 2004, Cancer Research.

[18]  J Nuyts,et al.  18FDG-Positron emission tomography for the early prediction of response in advanced soft tissue sarcoma treated with imatinib mesylate (Glivec). , 2003, European journal of cancer.

[19]  A. D. Van den Abbeele,et al.  Use of positron emission tomography in oncology and its potential role to assess response to imatinib mesylate therapy in gastrointestinal stromal tumors (GISTs). , 2002, European journal of cancer.

[20]  A. D. Van den Abbeele,et al.  Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. , 2002, The New England journal of medicine.

[21]  R. Wahl,et al.  Expression of hexokinase II and Glut-1 in untreated human breast cancer. , 2002, Nuclear medicine and biology.

[22]  G. Koehl,et al.  Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor , 2002, Nature Medicine.

[23]  T. Slaga,et al.  Functional roles of Akt signaling in mouse skin tumorigenesis , 2002, Oncogene.

[24]  R. Valeri,et al.  Rapamycin impairs recovery from acute renal failure: role of cell-cycle arrest and apoptosis of tubular cells. , 2001, American journal of physiology. Renal physiology.

[25]  T. Suhara,et al.  Phosphatidylinositol 3-Kinase/Akt Signaling Controls Endothelial Cell Sensitivity to Fas-Mediated Apoptosis via Regulation of FLICE-Inhibitory Protein (FLIP) , 2001, Circulation research.

[26]  K. Herholz,et al.  Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: review and 1999 EORTC recommendations. European Organization for Research and Treatment of Cancer (EORTC) PET Study Group. , 1999, European journal of cancer.

[27]  I. Mellinghoff,et al.  Hypoxia-inducible factor determines sensitivity to inhibitors of mTOR in kidney cancer , 2006, Nature Medicine.

[28]  M Van Glabbeke,et al.  New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. , 2000, Journal of the National Cancer Institute.