Limits of [18F]-FLT PET as a biomarker of proliferation in oncology

Eliot T McKinley, Gregory D Ayers, R Adam Smith, Samir A Saleh, Ping Zhao, Mary Kay Washington, Robert J Coffey, H Charles Manning, Eliot T McKinley, Gregory D Ayers, R Adam Smith, Samir A Saleh, Ping Zhao, Mary Kay Washington, Robert J Coffey, H Charles Manning

Abstract

Background: Non-invasive imaging biomarkers of cellular proliferation hold great promise for quantifying response to personalized medicine in oncology. An emerging approach to assess tumor proliferation utilizes the positron emission tomography (PET) tracer 3'-deoxy-3'[(18)F]-fluorothymidine, [(18)F]-FLT. Though several studies have associated serial changes in [(18)F]-FLT-PET with elements of therapeutic response, the degree to which [(18)F]-FLT-PET quantitatively reflects proliferative index has been continuously debated for more that a decade. The goal of this study was to elucidate quantitative relationships between [(18)F]-FLT-PET and cellular metrics of proliferation in treatment naïve human cell line xenografts commonly employed in cancer research.

Methods and findings: [(18)F]-FLT-PET was conducted in human cancer xenograft-bearing mice. Quantitative relationships between PET, thymidine kinase 1 (TK1) protein levels and immunostaining for proliferation markers (Ki67, TK1, PCNA) were evaluated using imaging-matched tumor specimens. Overall, we determined that [(18)F]-FLT-PET reflects TK1 protein levels, yet the cell cycle specificity of TK1 expression and the extent to which tumors utilize thymidine salvage for DNA synthesis decouple [(18)F]-FLT-PET data from standard estimates of proliferative index.

Conclusions: Our findings illustrate that [(18)F]-FLT-PET reflects tumor proliferation as a function of thymidine salvage pathway utilization. Unlike more general proliferation markers, such as Ki67, [(18)F]-FLT PET reflects proliferative indices to variable and potentially unreliable extents. [(18)F]-FLT-PET cannot discriminate moderately proliferative, thymidine salvage-driven tumors from those of high proliferative index that rely primarily upon de novo thymidine synthesis. Accordingly, the magnitude of [(18)F]-FLT uptake should not be considered a surrogate of proliferative index. These data rationalize the diversity of [(18)F]-FLT-PET correlative results previously reported and suggest future best-practices when [(18)F]-FLT-PET is employed in oncology.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Thymidine salvage and de novo…
Figure 1. Thymidine salvage and de novo synthesis pathways.
In thymidine salvage, thymidine is transported across the cell membrane and phosphorylated by TK1 into thymidine monophosphate (TMP). The thymidine is further phosphorylated into thymidine diphosphate (TDP) and thymidine triphosphate (TTP) and then incorporated into DNA. Alternatively, using the de novo synthesis pathway, deoxyuridine monophosphate (dUMP) is converted to TMP by TS which can then be further phosphorylated and incorporated into DNA. Similarly to thymidine, [18F]-FLT is transported into the cell and phosphorylated into [18F]-FLT monophosphate ([18F]-FLTMP) and trapped by TK1. [18F]-FLTMP can be further phosphorylated into [18F]-FLT diphosphate ([18F]-FLTDP) and [18F]-FLT triphosphate ([18F]-FLTTP), however, due to the substitution of OH with 18F in the 5-prime position, [18F]-FLTTP is not incorporated into the DNA.
Figure 2. IHC markers of proliferation in…
Figure 2. IHC markers of proliferation in HCT-116 and DiFi human CRC xenografts.
Representative high power microscopic images (40x) of HCT-116 and DiFi xenograft tissue stained for Ki67, PCNA, and TK1 are shown along with quantification of the percentage of positive cells per field for each marker. HCT-116 and DiFi xenograft tissues exhibit a similar proportion of proliferating cells as measured by Ki67 indices (68.50 ± 5.52 % vs. 72.37 ± 12.64%; p  =  0.0651). The proportion of PCNA positive cells, representing S-phase, was significantly increased in HCT-116 xenografts (53.91 ± 3.18 %) compared to DiFi xenografts (13.59 ±1.48%; p  =  0.0007). Similar to PCNA, the percentage of TK1-positive cells was significantly higher in HCT-116 xenografts (38.32 ± 1.90 %) than DiFi cells (17.95 ± 3.95 %; p  =  0.0007). TK1 indices for both cell lines were reduced compared to Ki67 indices.
Figure 3. HCT-116 xenografts exhibit higher […
Figure 3. HCT-116 xenografts exhibit higher [18F]-FLT flux than DiFi xenografts.
Representative time activity curves for HCT-116 (A) and DiFi (D) xenografts and left ventricle estimates of blood pool activity are shown. In both models, a sharp rise in activity in the blood pool is observed following tracer injection, followed by rapid clearance. Fit lines derived from compartmental modeling closely matched the measured data. Parametric maps reveal heterogeneous intratumoral delivery of [18F]-FLT in both HCT-116 (B) and DiFi (E) xenografts (tumor localized by arrowhead). Increased [18F]-FLT delivery was observed in the urinary bladder (denoted by UB) in both cell line xenografts. The tumor regions exhibiting the greatest [18F]-FLT flux in HCT-116 tumors (C) also exhibited the highest tracer delivery. Only modest [18F]-FLT flux was observed in DiFi xenografts (F), despite having similar delivery as HCT-116 xenografts (B).
Figure 4. Static [ 18 F]-FLT PET…
Figure 4. Static [18F]-FLT PET uptake is 2-fold greater in HCT-116 xenografts than DiFi xenografts.
Representative 20 minute static PET scans reflect the relative 60 minute accumulation of [18F]-FLT (%ID/g) qualitatively and show increased tracer uptake in HCT-116 xenografts (A) compared to DiFi (B) xenografts (tumor localized by arrowhead). Spatially, %ID/g maps (A,B) are similar to KFLT maps derived from parametric modeling (Fig. 3C,F). Static PET imaging reveals approximately a 2-fold increase in [18F]-FLT accumulation in HCT-116 xenografts (8.56 ± 1.17 %ID/g) compared to DiFi xenografts (3.92 ± 1.08 %ID/g; p < 0.0001) (C) and was similar to [18F]-FLT flux (KFLT) means derived from compartmental modeling (Table 1).
Figure 5. Isogenically matched cell line xenografts…
Figure 5. Isogenically matched cell line xenografts illustrate that de novo pathway utilization results in decreased [18F]-FLT uptake.
Western blot analysis illustrated reduced TK1 levels and elevated TS levels in HCT-116p21-/- xenografts compared to wild type HCT-116 xenografts (A). Densitometry of the western blot illustrated that HCT-116p21-/- tumors exhibited one-third less TK1 protein and double the TS protein compared to the parental line (B). Illustrating the sensitivity of [18F]-FLT PET to de novo pathway utilization, PET imaging of HCT-116 (C) and HCT-116p21-/- (D) xenografts closely reflected the relative tumor cell TK1 levels inherent to each model (tumor localized by arrowhead). HCT-116 xenografts (8.56 ± 1.17 %ID/g) exhibited approximately 1/3 greater uptake than analogous HCT-116p21-/- xenografts (6.91 ± 1.07 %ID/g; p  =  0.005) (E).

References

    1. Shields AF, Mankoff DA, Link JM, Graham MM, Eary JF, et al. (1998) Carbon-11-thymidine and FDG to measure therapy response. J Nucl Med 39: 1757–1762.
    1. Shields AF, Mankoff D, Graham MM, Zheng M, Kozawa SM, et al. (1996) Analysis of 2-carbon-11-thymidine blood metabolites in PET imaging. J Nucl Med 37: 290–296.
    1. Reinhardt MJ, Kubota K, Yamada S, Iwata R, Yaegashi H (1997) Assessment of cancer recurrence in residual tumors after fractionated radiotherapy: a comparison of fluorodeoxyglucose, L-methionine and thymidine. J Nucl Med 38: 280–287.
    1. Barthel H, Cleij MC, Collingridge DR, Hutchinson OC, Osman S, et al. (2003) 3'-deoxy-3'-[18F]fluorothymidine as a new marker for monitoring tumor response to antiproliferative therapy in vivo with positron emission tomography. Cancer Res 63: 3791–3798.
    1. Chen W, Cloughesy T, Kamdar N, Satyamurthy N, Bergsneider M, et al. (2005) Imaging proliferation in brain tumors with F-18-FLT PET: Comparison with F-18-FDG. Journal of Nuclear Medicine 46: 945–952.
    1. Choi SJ, Kim JS, Kim JH, Oh SJ, Lee JG, et al. (2005) [F-18]3 '-deoxy-3 '-fluorothymidine PET for the diagnosis and grading of brain tumors. European Journal of Nuclear Medicine and Molecular Imaging 32: 653–659.
    1. Cobben DCP, Elsinga PH, van Waarde A, Jager PL (2003) Correspondence re: H. Barthel et al., 3 '-deoxy-3 '-[F-18]fluorothymidine as a new marker for monitoring tumor response to antiproliferative therapy in vivo with positron emission tomography. Cancer Res., 63: 3791-3798,2003. Cancer Research 63: 8558–8559.
    1. Shields AF, Grierson JR, Dohmen BM, Machulla HJ, Stayanoff JC, et al. (1998) Imaging proliferation in vivo with [F-18]FLT and positron emission tomography. Nat Med 4: 1334–1336.
    1. Arner ES, Eriksson S (1995) Mammalian deoxyribonucleoside kinases. Pharmacol Ther 67: 155–186.
    1. Mankoff DA, Eary JF, Link JM, Muzi M, Rajendran JG, et al. (2007) Tumor-specific positron emission tomography imaging in patients: [18F] fluorodeoxyglucose and beyond. Clin Cancer Res 13: 3460–3469.
    1. Grierson JR, Shields AF (2000) Radiosynthesis of 3'-deoxy-3'-[(18)F]fluorothymidine: [(18)F]FLT for imaging of cellular proliferation in vivo. Nucl Med Biol 27: 143–156.
    1. Grierson JR, Schwartz JL, Muzi M, Jordan R, Krohn KA (2004) Metabolism of 3 '-deoxy-3 '-[F-18]fluorothymidine in proliferating A549 cells: validations for positron emission tomography. Nuclear Medicine and Biology 31: 829–837.
    1. Muzi M, Spence AM, O'Sullivan F, Mankoff DA, Wells JM, et al. (2006) Kinetic analysis of 3'-deoxy-3'-18F-fluorothymidine in patients with gliomas. J Nucl Med 47: 1612–1621.
    1. Schwartz JL, Tamura Y, Jordan R, Grierson JR, Krohn KA (2003) Monitoring tumor cell proliferation by targeting DNA synthetic processes with thymidine and thymidine analogs. J Nucl Med 44: 2027–2032.
    1. Wagner M, Seitz U, Buck A, Neumaier B, Schultheiss S, et al. (2003) 3 '-[F-18]fluoro-3 '-deoxythymidine ([F-18]-FLT) as positron emission tomography tracer for imaging proliferation in a murine B-cell lymphoma model and in the human disease. Cancer Research 63: 2681–2687.
    1. Brockenbrough JS, Souquet T, Morihara JK, Stern JE, Hawes SE, et al. Tumor 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) uptake by PET correlates with thymidine kinase 1 expression: static and kinetic analysis of (18)F-FLT PET studies in lung tumors. J Nucl Med 52: 1181–1188.
    1. Olive M, Untawale S, Coffey RJ, Siciliano MJ, Wildrick DM, et al. (1993) Characterization of the DiFi rectal carcinoma cell line derived from a familial adenomatous polyposis patient. In Vitro Cell Dev Biol 29A: 239–248.
    1. Waldman T, Kinzler KW, Vogelstein B (1995) p21 is necessary for the p53-mediated G1 arrest in human cancer cells. Cancer Res 55: 5187–5190.
    1. Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, et al. (1998) Requirement for p53 and p21 to sustain G2 arrest after DNA damage. Science 282: 1497–1501.
    1. Dy DY, Whitehead RH, Morris DL (1992) SMS 201.995 inhibits in vitro and in vivo growth of human colon cancer. Cancer Res 52: 917–923.
    1. Manning HC, Merchant NB, Foutch AC, Virostko JM, Wyatt SK, et al. (2008) Molecular imaging of therapeutic response to epidermal growth factor receptor blockade in colorectal cancer. Clin Cancer Res 14: 7413–7422.
    1. Shah C, Miller TW, Wyatt SK, McKinley ET, Olivares MG, et al. (2009) Imaging biomarkers predict response to anti-HER2 (ErbB2) therapy in preclinical models of breast cancer. Clin Cancer Res 15: 4712–4721.
    1. Muzi M, Mankoff DA, Grierson JR, Wells JM, Vesselle H, et al. (2005) Kinetic modeling of 3'-deoxy-3'-fluorothymidine in somatic tumors: mathematical studies. J Nucl Med 46: 371–380.
    1. Muzic RF Jr, Cornelius S (2001) COMKAT: compartment model kinetic analysis tool. J Nucl Med 42: 636–645.
    1. Li W, Araya M, Elliott M, Kang X, Gerk PM, et al. Monitoring cellular accumulation of 3'-deoxy-3'-fluorothymidine (FLT) and its monophosphate metabolite (FLT-MP) by LC-MS/MS as a measure of cell proliferation in vitro. J Chromatogr B Analyt Technol Biomed Life Sci 879: 2963–2970.
    1. Efron B, Tibshirani R (1993) An Introduction to the Bootstrap. Washington, D.C.: CRC Press.
    1. Zhang CC, Yan Z, Li W, Kuszpit K, Painter CL, et al. [18F]FLT-PET imaging does not always "light up" proliferating tumor cells. Clin Cancer Res.
    1. Kim SJ, Lee JS, Im KC, Kim SY, Park SA, et al. (2008) Kinetic modeling of 3'-deoxy-3'-18F-fluorothymidine for quantitative cell proliferation imaging in subcutaneous tumor models in mice. J Nucl Med 49: 2057–2066.
    1. Wang H, Zhang J, Tian J, Qu B, Li T, et al. (2009) Using dual-tracer PET to predict the biologic behavior of human colorectal cancer. J Nucl Med 50: 1857–1864.
    1. Krieger-Hinck N, Gustke H, Valentiner U, Mikecz P, Buchert R, et al. (2006) Visualisation of neuroblastoma growth in a Scid mouse model using [18F]FDG and [18F]FLT-PET. Anticancer Res 26: 3467–3472.
    1. Buck AK, Bommer M, Stilgenbauer S, Juweid M, Glatting G, et al. (2006) Molecular imaging of proliferation in malignant lymphoma. Cancer Res 66: 11055–11061.
    1. Yap CS, Czernin J, Fishbein MC, Cameron RB, Schiepers C, et al. (2006) Evaluation of thoracic tumors with 18F-fluorothymidine and 18F-fluorodeoxyglucose-positron emission tomography. Chest 129: 393–401.
    1. Dittmann H, Dohmen BM, Paulsen F, Eichhorn K, Eschmann SM, et al. (2003) [18F]FLT PET for diagnosis and staging of thoracic tumours. Eur J Nucl Med Mol Imaging 30: 1407–1412.
    1. van Westreenen HL, Cobben DC, Jager PL, van Dullemen HM, Wesseling J, et al. (2005) Comparison of 18F-FLT PET and 18F-FDG PET in esophageal cancer. J Nucl Med 46: 400–404.
    1. Buck AK, Schirrmeister H, Hetzel M, Von Der Heide M, Halter G, et al. (2002) 3-deoxy-3-[(18)F]fluorothymidine-positron emission tomography for noninvasive assessment of proliferation in pulmonary nodules. Cancer Res 62: 3331–3334.
    1. Yamamoto Y, Nishiyama Y, Ishikawa S, Nakano J, Chang SS, et al. (2007) Correlation of 18F-FLT and 18F-FDG uptake on PET with Ki-67 immunohistochemistry in non-small cell lung cancer. Eur J Nucl Med Mol Imaging 34: 1610–1616.
    1. Kenny LM, Vigushin DM, Al-Nahhas A, Osman S, Luthra SK, et al. (2005) Quantification of cellular proliferation in tumor and normal tissues of patients with breast cancer by [18F]fluorothymidine-positron emission tomography imaging: evaluation of analytical methods. Cancer Res 65: 10104–10112.
    1. Smyczek-Gargya B, Fersis N, Dittmann H, Vogel U, Reischl G, et al. (2004) PET with [18F]fluorothymidine for imaging of primary breast cancer: a pilot study. Eur J Nucl Med Mol Imaging 31: 720–724.
    1. Yamamoto Y, Kameyama R, Izuishi K, Takebayashi R, Hagiike M, et al. (2009) Detection of colorectal cancer using (1)F-FLT PET: comparison with (1)F-FDG PET. Nucl Med Commun 30: 841–845.
    1. Francis DL, Freeman A, Visvikis D, Costa DC, Luthra SK, et al. (2003) In vivo imaging of cellular proliferation in colorectal cancer using positron emission tomography. Gut 52: 1602–1606.
    1. McKinley ET, Smith RA, Tanksley JP, Washington MK, Walker R, et al. [(18)F]FLT-PET to predict pharmacodynamic and clinical response to cetuximab therapy in Menetrier's disease. Ann Nucl Med.
    1. Moroz MA, Kochetkov T, Cai S, Wu J, Shamis M, et al. Imaging colon cancer response following treatment with AZD1152: a preclinical analysis of [18F]fluoro-2-deoxyglucose and 3'-deoxy-3'-[18F]fluorothymidine imaging. Clin Cancer Res 17: 1099–1110.

Source: PubMed

3
Subscribe