FDG-PET as a predictive biomarker for therapy with everolimus in metastatic renal cell cancer

James L Chen, Daniel E Appelbaum, Masha Kocherginsky, Charles L Cowey, Wendy Kimryn Rathmell, David F McDermott, Walter M Stadler, James L Chen, Daniel E Appelbaum, Masha Kocherginsky, Charles L Cowey, Wendy Kimryn Rathmell, David F McDermott, Walter M Stadler

Abstract

The mTOR (mammalian target of rapamycin) inhibitor, everolimus, affects tumor growth by targeting cellular metabolic proliferation pathways and delays renal cell carcinoma (RCC) progression. Preclinical evidence suggests that baseline elevated tumor glucose metabolism as quantified by FDG-PET ([(18)F] fluorodeoxy-glucose positron emission tomography) may predict antitumor activity. Metastatic RCC (mRCC) patients refractory to vascular endothelial growth factor (VEGF) pathway inhibition were treated with standard dose everolimus. FDG-PET scans were obtained at baseline and 2 weeks; serial computed tomography (CT) scans were obtained at baseline and every 8 weeks. Maximum standardized uptake value (SUVmax) of the most FDG avid lesion, average SUVmax of all measured lesions and their corresponding 2-week relative changes were examined for association with 8-week change in tumor size. A total of 63 patients were enrolled; 50 were evaluable for the primary endpoint of which 48 had both PET scans. Patient characteristics included the following: 36 (72%) clear cell histology and median age 59 (range: 37-80). Median pre- and 2-week treatment average SUVmax were 6.6 (1-17.9) and 4.2 (1-13.9), respectively. Response evaluation criteria in solid tumors (RECIST)-based measurements demonstrated an average change in tumor burden of 0.2% (-32.7% to 35.9%) at 8 weeks. Relative change in average SUVmax was the best predictor of change in tumor burden (all evaluable P = 0.01; clear cell subtype P = 0.02), with modest correlation. Baseline average SUVmax was correlated with overall survival and progression-free survival (PFS) (P = 0.023; 0.020), but not with change in tumor burden. Everolimus therapy decreased SUVs on follow-up PET scans in mRCC patients, but changes were only modestly correlated with changes in tumor size. Thus, clinical use of FDG-PET-based biomarkers is challenged by high variability.

Keywords: Kidney carcinoma; mTOR protein; pharmacological biomarkers; positron emission tomography.

Figures

Figure 1
Figure 1
Patient disposition of this clinical trial exploring FDG-PET as a predictive biomarker for therapy with everolimus in metastatic renal cell carcinoma.
Figure 2
Figure 2
(A) RECIST-based changes in tumor size at 8 weeks post treatment with everolimus are heterogeneous as seen in the waterfall plot. (B) FDG-PET uptake decreased after 2 weeks of treatment for most renal cancer patients.
Figure 3
Figure 3
Kaplan–Meier estimates of overall survival among patients with high (>4) and low (≤4) baseline avgSUVmax. (A) Significant difference in overall survival among all patients (n = 50). (B) The overall survival difference is no longer statistically significant among the clear cell subtype (n = 36).

References

    1. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N. Engl. J. Med. 2007;356:2271–2281.
    1. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet. 2008;372:449–456.
    1. Khandani AH, Rathmell WK. Positron emission tomography in renal cell carcinoma: an imaging biomarker in development. Semin. Nucl. Med. 2012;42:221–230.
    1. Brugarolas J. Renal-cell carcinoma – molecular pathways and therapies. N. Engl. J. Med. 2007;356:185–187.
    1. Hager M, Haufe H, Lusuardi L, Schmeller N, Kolbitsch C. PTEN, pAKT, and pmTOR expression and subcellular distribution in primary renal cell carcinomas and their metastases. Cancer Invest. 2011;29:427–438.
    1. Chan DA, Sutphin PD, Nguyen P, Turcotte S, Lai EW, Banh A, et al. Targeting GLUT1 and the Warburg effect in renal cell carcinoma by chemical synthetic lethality. Sci. Transl. Med. 2011;3:94ra70.
    1. Hu CJ, Wang LY, Chodosh LA, Keith B, Simon MC. Differential roles of hypoxia-inducible factor 1alpha (HIF-1alpha) and HIF-2alpha in hypoxic gene regulation. Mol. Cell. Biol. 2003;23:9361–9374.
    1. Kim JW, Tchernyshyov I, Semenza GL, Dang CV. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006;3:177–185.
    1. Thomas GV, Tran C, Mellinghoff IK, Welsbie DS, Chan E, Fueger B, et al. Hypoxia-inducible factor determines sensitivity to inhibitors of mTOR in kidney cancer. Nat. Med. 2006;12:122–127.
    1. Posadas EM, Figlin RA. Systemic therapy in renal cell carcinoma: advancing paradigms. Oncology (Williston Park) 2012;26:290–301.
    1. Kayani I, Avril N, Bomanji J, Chowdhury S, Rockall A, Sahdev A, et al. Sequential FDG-PET/CT as a biomarker of response to sunitinib in metastatic clear cell renal cancer. Clin. Cancer Res. 2011;17:6021–6028.
    1. Khandani AH, Cowey CL, Moore DT, Gohil H, Rathmell WK. Primary renal cell carcinoma: relationship between FDG uptake and response to neoadjuvant sorafenib. Nucl. Med. Commun. 2012;33:967–973.
    1. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, Rubinstein L, 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. J. Natl. Cancer Inst. 2000;92:205–216.
    1. Patard JJ, Leray E, Rioux-Leclercq N, Cindolo L, Ficarra V, Zisman A, et al. Prognostic value of histologic subtypes in renal cell carcinoma: a multicenter experience. J. Clin. Oncol. 2005;23:2763–2771.
    1. Sharma MR, Maitland ML, Ratain MJ. Why RECIST works and why it should stay – reply to counterpoint. Cancer Res. 2012;72:5158.
    1. Cho D, Signoretti S, Regan M, Mier JW, Atkins MB. The role of mammalian target of rapamycin inhibitors in the treatment of advanced renal cancer. Clin Cancer Res. 2007;13:758s–763s.
    1. Brouwers AH, Dorr U, Lang O, Boerman OC, Oyen WJ, Steffens MG, et al. 131 I-cG250 monoclonal antibody immunoscintigraphy versus [18F]FDG-PET imaging in patients with metastatic renal cell carcinoma: a comparative study. Nucl. Med. Commun. 2002;23:229–236.
    1. Majhail NS, Urbain JL, Albani JM, Kanvinde MH, Rice TW, Novick AC, et al. F-18 fluorodeoxyglucose positron emission tomography in the evaluation of distant metastases from renal cell carcinoma. J. Clin. Oncol. 2003;21:3995–4000.
    1. Lavin PT. An alternative model for the evaluation of antitumor activity. Cancer Clin. Trials. 1981;4:451–457.
    1. Ratain MJ, Eisen T, Stadler WM, Flaherty KT, Kaye SB, Rosner GL, et al. Phase II placebo-controlled randomized discontinuation trial of sorafenib in patients with metastatic renal cell carcinoma. J. Clin. Oncol. 2006;24:2505–2512.
    1. Patard JJ. New treatment options for renal cell cancer – critical evaluation. Eur. Urol. Suppl. 2008;7:443–446.
    1. Ho CL, Chen S, Ho KM, Chan WK, Leung YL, Cheng KC, et al. Dual-tracer PET/CT in renal angiomyolipoma and subtypes of renal cell carcinoma. Clin. Nucl. Med. 2012;37:1075–1082.
    1. Ma, W. W., H. Jacene, D. Song, F. Vilardell, W. A. Messersmith, D. Laheru, et al. [18F]fluorodeoxyglucose positron emission tomography correlates with Akt pathway activity but is not predictive of clinical outcome during mTOR inhibitor therapy. J. Clin. Oncol. 2009;27:2697–2704.
    1. Demichelis F, Setlur SR, Beroukhim R, Perner S, Korbel JO, Lafargue CJ, et al. Distinct genomic aberrations associated with ERG rearranged prostate cancer. Genes Chromosomes Cancer. 2009;48:366–380.

Source: PubMed

3
Suscribir