Early detection of response to experimental chemotherapeutic Top216 with [18F]FLT and [18F]FDG PET in human ovary cancer xenografts in mice

Mette Munk Jensen, Kamille Dumong Erichsen, Fredrik Björkling, Jacob Madsen, Peter Buhl Jensen, Liselotte Højgaard, Maxwell Sehested, Andreas Kjær, Mette Munk Jensen, Kamille Dumong Erichsen, Fredrik Björkling, Jacob Madsen, Peter Buhl Jensen, Liselotte Højgaard, Maxwell Sehested, Andreas Kjær

Abstract

Background: 3'-Deoxy-3'-[(18)F]fluorothymidine ((18)F-FLT) is a tracer used to assess cell proliferation in vivo. The aim of the study was to use (18)F-FLT positron emission tomography (PET) to study treatment responses to a new anti-cancer compound. To do so, we studied early anti-proliferative effects of the experimental chemotherapy Top216 non-invasively by PET.

Methodology/principal findings: In vivo uptake of (18)F-FLT in human ovary cancer xenografts in mice (A2780) was studied at various time points after Top216 treatment (50 mg/kg i.v. at 0 and 48 hours) was initiated. Baseline (18)F-FLT scans were made before either Top216 (n = 7-10) or vehicle (n = 5-7) was injected and repeated after 2 and 6 hours and 1 and 5 days of treatment. A parallel study was made with 2'-deoxy-2'-[(18)F]fluoro-D-glucose ((18)F-FDG) (n = 8). Tracer uptake was quantified using small animal PET/CT. Imaging results were validated by tumor volume changes and gene-expression of Ki67 and TK1. Top216 (50 mg/kg 0 and 48 hours) inhibited the growth of the A2780 tumor compared to the control group (P<0.001). (18)F-FLT uptake decreased significantly at 2 hours (-52%; P<0.001), 6 hours (-49%; P = 0.002) and Day 1 (-47%; P<0.001) after Top216 treatment. At Day 5 (18)F-FLT uptake was comparable to uptake in the control group. Uptake of (18)F-FLT was unchanged in the control group during the experiment. In the treatment group, uptake of (18)F-FDG was significantly decreased at 6 hours (-21%; P = 0.003), Day 1 (-29%; P<0.001) and Day 5 (-19%; P = 0.05) compared to baseline.

Conclusions/significance: One injection with Top216 initiated a fast and significant decrease in cell-proliferation assessable by (18)F-FLT after 2 hours. The early reductions in tumor cell proliferation preceded changes in tumor size. Our data indicate that (18)F-FLT PET is promising for the early non-invasive assessment of chemotherapy effects in both drug development and for tailoring therapy in patients.

Conflict of interest statement

Competing Interests: The following co-authors have conflict of interests: Peter Buhl Jensen: Ownership Interests and Employment in Topotarget A/S. Maxwell Sehested: Ownership Interests and Employment in Topotarget A/S. Fredrik Björkling: Employment in Topotarget A/S. Kamille Dumong Erichsen: Employment in Topotarget A/S. All other authors have no conflict of interests. That some of the co-authors are employed by Topotarget A/S does not alter the authors' adherence to the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. Chemical structure of Top216.
Figure 1. Chemical structure of Top216.
Figure 2. Schematic view of the experimental…
Figure 2. Schematic view of the experimental design.
Figure 3. Tumor growth.
Figure 3. Tumor growth.
A) The effects of Top216 on the growth of A2780 tumor xenografts. Tumor volume was determined by microCT. Mice were treated with Top216 (50 mg/kg) or vehicle at 0 and 48 hours. *) P# # #) P<0.001 vs. control. n = 15 tumors per group. B) Changes in tumor volume assessed by ratio Day 5/baseline in the control group as a function of baseline FLT uptake. n = 7 tumors. R2 = 0.61, P = 0.04.
Figure 4. Fused PET/CT images.
Figure 4. Fused PET/CT images.
A) The eight images at left are representative coronal fused PET/CT images of two mice scanned with 18F-FLT at baseline and at 6 hours and 1 and 5 days after treatment start. The images at the top show one mouse treated with Top216 and the images at the bottom show one control mouse which received vehicle. The four images at right show fused PET/CT pictures of two representative mice treated with either Top216 or vehicle and scanned at baseline and 2 hours after treatment initiation. The arrows point towards the tumors. B) Representative coronal fused PET/CT images of two mice scanned with 18F-FDG at baseline and 6 hours and 1 and 5 days after treatment start. The images at the top show one mouse treated with Top216 and the images at the bottom show one control mouse which received vehicle. The arrows point towards the tumors.
Figure 5. 18 F-FLT and 18 F-FDG…
Figure 5. 18F-FLT and 18F-FDG uptake assessed by SUVmean and SUVmax at baseline and following treatment with Top216.
Top216/vehicle treatment was initiated at 0 hours and repeated at 48 hours after the first injection. N = 5–10 tumors per group. *) P18F-FLT experiments and the two graphs at right show data from the 18F-FDG experiments.
Figure 6. Expression of Ki67 and TK1…
Figure 6. Expression of Ki67 and TK1 normalized to expression of reference gene TBP.
Data are presented as fold changes following treatment with Top216/vehicle relative to baseline levels (n = 7 tumors per group). Top216 treatment was initiated at 0 hours and repeated at 48 hours after the first injection. *) P

References

    1. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70.
    1. Shields AF, Grierson JR, Dohmen BM, Machulla HJ, Stayanoff JC, et al. Imaging proliferation in vivo with [F-18]FLT and positron emission tomography. Nat Med. 1998;4:1334–1336.
    1. Kong XB, Zhu QY, Vidal PM, Watanabe KA, Polsky B, et al. Comparisons of anti-human immunodeficiency virus activities, cellular transport, and plasma and intracellular pharmacokinetics of 3′-fluoro-3′-deoxythymidine and 3′-azido-3′-deoxythymidine. Antimicrob Agents Chemother. 1992;36:808–818.
    1. Arner ES, Eriksson S. Mammalian deoxyribonucleoside kinases. Pharmacol Ther. 1995;67:155–186.
    1. Sherley JL, Kelly TJ. Regulation of human thymidine kinase during the cell cycle. J Biol Chem. 1988;263:8350–8358.
    1. Rasey JS, Grierson JR, Wiens LW, Kolb PD, Schwartz JL. Validation of FLT uptake as a measure of thymidine kinase-1 activity in A549 carcinoma cells. J Nucl Med. 2002;43:1210–1217.
    1. Barthel H, Perumal M, Latigo J, He Q, Brady F, et al. The uptake of 3′-deoxy-3′-[18F]fluorothymidine into L5178Y tumours in vivo is dependent on thymidine kinase 1 protein levels. Eur J Nucl Med Mol Imaging. 2005;32:257–263.
    1. Barthel H, Cleij MC, Collingridge DR, Hutchinson OC, Osman S, et al. 3′-deoxy-3′-[18F]fluorothymidine as a new marker for monitoring tumor response to antiproliferative therapy in vivo with positron emission tomography. Cancer Res. 2003;63:3791–3798.
    1. Leyton J, Latigo JR, Perumal M, Dhaliwal H, He Q, et al. Early detection of tumor response to chemotherapy by 3′-deoxy-3′-[18F]fluorothymidine positron emission tomography: the effect of cisplatin on a fibrosarcoma tumor model in vivo. Cancer Res. 2005;65:4202–4210.
    1. Leyton J, Alao JP, Da CM, Stavropoulou AV, Latigo JR, et al. In vivo biological activity of the histone deacetylase inhibitor LAQ824 is detectable with 3′-deoxy-3′-[18F]fluorothymidine positron emission tomography. Cancer Res. 2006;66:7621–7629.
    1. Waldherr C, Mellinghoff IK, Tran C, Halpern BS, Rozengurt N, et al. Monitoring antiproliferative responses to kinase inhibitor therapy in mice with 3′-deoxy-3′-18F-fluorothymidine PET. J Nucl Med. 2005;46:114–120.
    1. Buck AK, Halter G, Schirrmeister H, Kotzerke J, Wurziger I, et al. Imaging proliferation in lung tumors with PET: 18F-FLT versus 18F-FDG. J Nucl Med. 2003;44:1426–1431.
    1. Francis DL, Freeman A, Visvikis D, Costa DC, Luthra SK, et al. In vivo imaging of cellular proliferation in colorectal cancer using positron emission tomography. Gut. 2003;52:1602–1606.
    1. Vesselle H, Grierson J, Muzi M, Pugsley JM, Schmidt RA, et al. In vivo validation of 3′deoxy-3′-[(18)F]fluorothymidine ([(18)F]FLT) as a proliferation imaging tracer in humans: correlation of [(18)F]FLT uptake by positron emission tomography with Ki-67 immunohistochemistry and flow cytometry in human lung tumors. Clin Cancer Res. 2002;8:3315–3323.
    1. Apisarnthanarax S, Alauddin MM, Mourtada F, Ariga H, Raju U, et al. Early detection of chemoradioresponse in esophageal carcinoma by 3′-deoxy-3′-3H-fluorothymidine using preclinical tumor models. Clin Cancer Res. 2006;12:4590–4597.
    1. Brepoels L, Stroobants S, Verhoef G, De GT, Mortelmans L, et al. 18F-FDG and 18F-FLT Uptake Early After Cyclophosphamide and mTOR Inhibition in an Experimental Lymphoma Model. J Nucl Med. 2009;50:1102–1109.
    1. Molthoff CF, Klabbers BM, Berkhof J, Felten JT, van GM, et al. Monitoring response to radiotherapy in human squamous cell cancer bearing nude mice: comparison of 2′-deoxy-2′-[18F]fluoro-D-glucose (FDG) and 3′-[18F]fluoro-3′-deoxythymidine (FLT). Mol Imaging Biol. 2007;9:340–347.
    1. Oyama N, Ponde DE, Dence C, Kim J, Tai YC, et al. Monitoring of therapy in androgen-dependent prostate tumor model by measuring tumor proliferation. J Nucl Med. 2004;45:519–525.
    1. Yang YJ, Ryu JS, Kim SY, Oh SJ, Im KC, et al. Use of 3′-deoxy-3′-[18F]fluorothymidine PET to monitor early responses to radiation therapy in murine SCCVII tumors. Eur J Nucl Med Mol Imaging. 2006;33:412–419.
    1. Manning HC, Merchant NB, Foutch AC, Virostko JM, Wyatt SK, et al. Molecular imaging of therapeutic response to epidermal growth factor receptor blockade in colorectal cancer. Clin Cancer Res. 2008;14:7413–7422.
    1. Shah C, Miller TW, Wyatt SK, McKinley ET, Olivares MG, et al. Imaging biomarkers predict response to anti-HER2 (ErbB2) therapy in preclinical models of breast cancer. Clin Cancer Res. 2009;15:4712–4721.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–247.
    1. Therasse P, Arbuck SG, Eisenhauer EA, Wanders J, Kaplan RS, 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. Wahl RL, Jacene H, Kasamon Y, Lodge MA. From RECIST to PERCIST: Evolving Considerations for PET response criteria in solid tumors. J Nucl Med. 2009;50(Suppl 1):122S–150S.
    1. Weber WA, Wieder H. Monitoring chemotherapy and radiotherapy of solid tumors. Eur J Nucl Med Mol Imaging. 2006;33(Suppl 1):27–37.
    1. Kubota R, Yamada S, Kubota K, Ishiwata K, Tamahashi N, et al. Intratumoral distribution of fluorine-18-fluorodeoxyglucose in vivo: high accumulation in macrophages and granulation tissues studied by microautoradiography. J Nucl Med. 1992;33:1972–1980.
    1. van WA, Cobben DC, Suurmeijer AJ, Maas B, Waalburg W, et al. Selectivity of 18F-FLT and 18F-FDG for differentiating tumor from inflammation in a rodent model. J Nucl Med. 2004;45:695–700.
    1. Uddin MK, Reignier SG, Coulter T, Montalbetti C, Granas C, et al. Syntheses and antiproliferative evaluation of oxyphenisatin derivatives. Bioorg Med Chem Lett. 2007;17:2854–2857.
    1. Jensen MM, Jorgensen JT, Binderup T, Kjaer A. Tumor volume in subcutaneous mouse xenografts measured by microCT is more accurate and reproducible than determined by 18F-FDG-microPET or external caliper. BMC Med Imaging. 2008;8:16.
    1. Fueger BJ, Czernin J, Hildebrandt I, Tran C, Halpern BS, et al. Impact of animal handling on the results of 18F-FDG PET studies in mice. J Nucl Med. 2006;47:999–1006.
    1. Fleige S, Pfaffl MW. RNA integrity and the effect on the real-time qRT-PCR performance. Mol Aspects Med. 2006;27:126–139.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402–408.
    1. Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 2001;29:e45.
    1. McDermott GM, Welch A, Staff RT, Gilbert FJ, Schweiger L, et al. Monitoring primary breast cancer throughout chemotherapy using FDG-PET. Breast Cancer Res Treat. 2007;102:75–84.
    1. Chang ZF, Huang DY, Hsue NC. Differential phosphorylation of human thymidine kinase in proliferating and M phase-arrested human cells. J Biol Chem. 1994;269:21249–21254.
    1. Munch-Petersen B, Cloos L, Jensen HK, Tyrsted G. Human thymidine kinase 1. Regulation in normal and malignant cells. Adv Enzyme Regul. 1995;35:69–89.

Source: PubMed

3
Subscribe