Synthesis and positron emission tomography evaluation of 18F-Glu-Urea-Lys, a prostate-specific membrane antigen-based imaging agent for prostate cancer

Corrigendum in: /10.3892/ol.2015.4039

  • Authors:
    • Weiwei Fan
    • Zhiwei Zhang
    • Zheng Zhu
    • Deyong Yang
    • Xiaochi Chen
    • Jianbo Wang
    • Feng Chen
    • Xishuang Song
  • View Affiliations

  • Published online on: August 19, 2015     https://doi.org/10.3892/ol.2015.3625
  • Pages: 2299-2302
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In recent years, single-photon emission computed tomography and positron emission tomography (PET) have also been used, in addition to computed tomography and magnetic resonance imaging, in targeting the diagnosis of prostate cancer. The aim of this study was to synthesize the prostate‑specific membrane antigen (PSMA)‑based imaging agent 2‑{3‑[1‑Carboxy‑5‑(4‑[18F] fluoro‑benzoylamino)‑pentyl]‑ureido}‑pentanedioic acid (18F‑Glu‑Urea‑Lys, [18F]3) and to detect its PET imaging efficiency for high PSMA expression in prostate cancer. In this study, 18F‑Glu‑Urea‑Lys was synthesized in two steps from the p‑methoxybenzyl‑protected Glu‑Urea‑Lys precursor using N‑Hydroxysuccinimidyl‑4‑[18F] fluorobenzoate ([18F]SFB). PET imaging evaluation was conducted in nude mice using LNCaP (PSMA+), and PC‑3, 231 and A549 (all PSMA‑) xenograft models. The results indicated that 18F‑Glu‑Urea‑Lys was produced in radiochemical yields of 28.7%. The radiochemical purity was 99.1% and the mean total synthesis time was 168 min. In nude mice models 18F‑Glu‑Urea‑Lys clearly delineated PSMA+ LNCaP prostate tumor xenografts on PET imaging. At 4 h post‑injection, the contrast agents were only observed in renal, liver, bladder and PSMA+ LNCaP tumors. The PSMA‑ tumor (PC‑3, 231 and A549) was clear. In conclusion, 18F‑Glu‑Urea‑Lys was found to be easily synthesized. This radiotracer demonstrated high tumor and low‑to‑normal tissue uptake, fast clearance from non-target tissues and retention in PSMA+ prostate tumor xenografts.

Introduction

Prostate cancer is one of the most common types of tumor and the second highest cause of cancer-related mortality in males (1). The majority of patients succumb to tumor recurrence and metastasis. Early diagnosis, targeted therapy and effective monitoring following radical prostatectomy may have a significant impact on the prognosis of patients. The location of the tumor determines the subsequent treatment. In recent years not only have computed tomography (CT) and magnetic resonance imaging been used in prostate cancer diagnosis, single-photon emission computed tomography (SPECT) and positron emission tomography (PET) also offer new ways of targeting diagnosis (2,3).

Prostate-specific membrane antigen (PSMA) is a type 2 transmembrane glycoprotein expressed in prostate epithelial cells. It is shown to be highly expressed in prostate cancer in a disease progression-dependent manner (4). This study introduces a means of synthesis of 2-{3-[1-Carboxy-5-(4-[18F] fluoro-benzoylamino)-pentyl]-ureido}-pentanedioic acid (18F-Glu-Urea-Lys, [18F]3). This low molecular weight agent is easily prepared and demonstrates a high uptake in PSMA+ tumors.

Materials and methods

General procedures

All reagents and solvents were purchased from Sigma-Aldrich (Milwaukee, WI, USA). 1H NMR spectra were obtained on an Avance 400 MHz spectrometer (Bruker Corporation, Ettlingen, Germany). Electrospray ionization (ESI) mass spectra were obtained on a Bruker Esquire 3000 plus system. High-performance liquid chromatography (HPLC) purification was performed on a Waters 2998 and Waters 2487 system (Waters Corp., Milford, MA, USA). [18F]-fluoride was obtained using the M-7 Cyclotron (Sumitomo Heavy Industries, Ltd., Tokyo, Japan). Solid-phase extraction cartridges (Sep-Pak C18 Plus) were purchased from Waters Corp. The precursor 2-[3-(5-amino-1-carboxy-pentyl)- ureido]-pentanedioic acid 1 was synthesized in Dalian Medical University, China (5).

This study was approved by the ethics committee of the First Affiliated Hospital of Dalian Medical University (Dalian, China).

Cell lines

LNCaP, PC-3, 231 and A549 cells were obtained from WUXI Molecular Imaging CRO (Wuxi, China). Nude mice were purchased from JiangNan University, China. Cells (5×106) were implanted subcutaneously into the right flank of models. Mice were imaged when the tumor xenografts reached 5–8 mm in diameter.

2-{3-[1-tert-Butoxycarbonyl-5-(4-fluoro-benzoylamino)-pent yl]-ureido}-pentanedioic acid di-tert-butyl ester 2

N-Hydroxysuccinimidyl-4-[18F] fluorobenzoate (SFB, 145 mg), hydroxybenzotriazole (HOBt, 165 mg) and 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDCI, 235 mg) were added to a solution of CH2Cl2 (50 ml), then mixed with Triethylamine (208 mg) and stirred for 1 h. Precursor 1 (500 mg) was added and stirred at room temperature overnight. The crude material was purified on a silica column to obtain 550 mg compound 2.

2-{3-[1-Carboxy-5-(4-fluoro-benzoylamino)-pentyl]-ureido}-pe ntanedioic acid 3

Compound 2 (110 mg) was added to HCl/diethyl ether solution (20 ml), and stirred overnight at room temperature. The crude material was purified using HPLC to obtain 24 mg compound 3.

2-[3-[1-Carboxy-5-(4-[18F]fluoro-benzoylamino)-pentyl]-ure ido]-pentanedioic acid [18F]3

Compound 1 (1 mg) was added to phosphate-buffered saline (PBS) solution (100 µl). Then [18F]SFB (100 µl) and Na2CO3 (40 µl) was added, and the mixture was regulated to pH 7.6, stirred and reacted in an oil bath at 50°C for 30 min. When the reaction cooled down, trifluoroacetic acid (100 µl) and benzaldehyde (3 µl) were added, and reacted in an oil bath at 50°C for 30 min. Water (5 ml) was added, then the mixture was purified on a silica column, and washed with acetonitrile (0.5 ml). Finally, it was purified using HPLC to obtain [18F]3.

PET imaging

Small animal PET was used to image the nude mice implanted with PSMA+ (LNCaP) and PSMA- (PC-3, 231 and A549) xenografts. The nude mice were anesthetized with diethyl ether and injected intravenously with 0.2 mCi 18F-Glu-Urea-Lys in 200 µl PBS. The images were obtained at post-injection times of 1, 2 and 4 h.

Results

Synthesis of the compounds 2, 3 and [18F]3

The final quantity of 2-{3-[1-tert-Butoxycarbonyl-5-(4-flu oro-benzoylamino)-pentyl]-ureido}-pentanedioic acid di-tert-butyl ester 2 obtained was 550 mg, with a produce yield of 88%. The associated parameters are listed as the followings: 1H NMR (400 MHz, CDCl3) δ7.91–7.96 (m, 2H), 7.26–7.45 (m, 1H), 7.05–7.11 (m, 2H), 5.70–5.72 (m, 1H), 5.40–5.43 (m, 1H), 4.20–4.23 (m, 2H), 3.34–3.51 (m, 2H), 2.24–2.29 (m, 2H), 2.16 (m, 1H), 1.99–2.04 (m, 2H), 1.64–1.77 (m, 32H). The [M+H]+ ESI mass calculated for C31H48FN3O8 was 609.7.

The final quantity of 2-{3-[1-Carboxy-5-(4-fluoro-benzoylamino)-pentyl]ureido}-pentanedioic acid 3 obtained was 24 mg, with a produce yield of ~30%. The associated parameters are listed as the followings: 1H NMR (400 MHz, CDCl3) δ8.51 (s, 1H), 7.89–7.92 (m, 2H), 7.27–7.31 (m, 2H), 6.34 (m, 2H), 4.06–4.08 (m, 2H), 3.23–3.55 (m, 3H), 2.25–2.51 (m, 2H), 1.50–1.60 (m, 7H), 1.06–1.35 (m, 3H). The [M+H]+ ESI mass calculated for C19H24FN3O8 was 441.4.

The radiochemical yield of [18F]3 achieved was 28.7%. The radiochemical purity was 99.1% and the mean synthesis time was 168 min (Fig. 1).

PET imaging

Following the injection, 18F-Glu-Urea-Lys rapidly and notably delineated PSMA+ LNCaP prostate tumor xenografts on the PET imaging. At 4 h post-injection, the contrast was only observed in renal, liver, bladder (the intense renal uptake was partially due to the specific binding of 18F-Glu-Urea-Lys to proximal renal tubules (6) as well as to the excretion of this hydrophilic compound) and PSMA+ LNCaP tumors. PSMA tumors (PC-3, 231 and A549) were clear according to the radiotracer (Fig. 2).

Discussion

Due to the relatively low metabolic rate of prostate cancer, PET with [18F] fluorodeoxy glucose (FDG PET) has proven ineffective. Other agents for imaging prostate cancer include the choline series (7), radiolabeled acetates (8), [18F] F-FACBC (9), [18F] FMAU (10) and [18F] FDHT (11). However, each has disadvantages, including cost, difficulty to synthesize or low specificity to prostate cancer.

Overexpressed in prostate cancer, PSMA is becoming an attractive target for cancer imaging and therapy (12). PSMA has an internalization signal that allows internalization of the protein on the cell surface into an endosomal compartment (13). Previous studies reveal that a type of monoclonal antibody against PSMA is available for imaging diagnosis and therapy of prostate cancer (14,15). These agents have long circulation times, low specificity to target tissue and were expensive to synthesize, limiting their clinical use in the diagnosis of prostate cancer.

Maresca et al (16) designed and synthesized a type of Glu-Urea-R compound which could be marked by 123I and 131I. This R-group and the substrate coupling with it may notably affect the affinity of the compounds to PSMA. To improve the diagnosis and therapy of prostate cancer, in recent years researchers have developed a series of PSMA-based small molecular agents. This type of agent was based on various R-groups, including [11C] DCMC (17), [125I] DCIT (18) and [18F] DCFBC (19), each having its own benefits.

The use of these compounds is not limited to the area of diagnosis of prostate cancer. Kularatne et al (20) coupled the chelate 99mTc-Dap-Asp-Cys with Glu-Urea-R for use in SPECT as an imaging agent. In combination with the chemotherapy drug TubH, this compound was capable of killing PSMA+ LNCaP cells in vitro. Zhang et al (21) coupled dinitrophenyl (DNP) with Glu-Urea-R to target prostate cancer. The DNP-end increased the immune antibodies and killed the cancer cells.

These small molecular agents demonstrate high specificity and affinity with PSMA (22). The use of 18F-Glu-Urea-Lys provides a new strategy in diagnosis, preoperative or tumor recurrence staging, and also could be extended from molecular imaging to the gene target therapy area.

In conclusion, 18F-Glu-Urea-Lys demonstrated high PSMA+ tumor uptake and low-to-normal tissue uptake. This radiotracer could be quickly cleared from non-target tissues and retention may occur in PSMA+ prostate tumor. With its relatively simple and convenient method of synthesis, this type of PSMA-based small molecular imaging agent may have a variety of clinical uses to help localize prostate cancer.

Acknowledgements

The authors are grateful for the financial support from grants NSFC30670544 and NSFC81271603 from the National Natural Science Foundation of China. They also thank WUXI Molecular Imaging CRO for performing the imaging studies and providing excellent technical support.

References

1 

Chen W, Mao K, Liu Z and Dinh-Xuan AT: The role of the RhoA/Rho kinase pathway in angiogenesis and its potential value in prostate cancer (Review). Oncol Lett. 8:1907–1911. 2014.(Review). PubMed/NCBI

2 

Geus-Oei LF and Oyen WJ: Predictive and prognostic value of FDG-PET. Cancer Imaging. 8:70–80. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Liu Y: Diagnostic role of fluorodeoxyglucose positron emission tomography-computed tomography in prostate cancer. Oncol Lett. 7:2013–2018. 2014.PubMed/NCBI

4 

Risk MC, Knudsen BS, Coleman I, Dumpit RF, Kristal AR, LeMeur N, Gentleman RC, True LD, Nelson PS and Lin DW: Differential gene expression in benign prostate epithelium of men with and without prostate cancer: evidence for a prostate cancer field effect. Clin Cancer Res. 16:5414–5423. 2010. View Article : Google Scholar : PubMed/NCBI

5 

Chen XC, Yang DY and Che XY: Synthesis of PSMA-targeted small molecule Glu-urea-Lys analogue. J Dalian Med Univer. 34:13–17. 2012.

6 

Silver DA, Pellicer I, Fair WR, Heston WD and Cordon-Cardo C: Prostate-specific membrane antigen expression in normal and malignant human tissues. Clin Cancer Res. 3:81–85. 1997.PubMed/NCBI

7 

Rinnab L, Mottaghy FM, Blumstein NM, Reske SN, Hautmann RE, Hohl K, Möller P, Wiegel T, Kuefer R and Gschwend JE: Evaluation of [11C]-choline positron-emission/computed tomography in patients with increasing prostate-specific antigen levels after primary treatment for prostate cancer. BJU Int. 100:786–793. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Ponde DE, Dence CS, Oyama N, Kim J, Tai YC, Laforest R, Siegel BA and Welch MJ: 18F-fluoroacetate: A potential acetate analog for prostate tumor imaging - in vivo evaluation of 18F-fluoroacetate versus 11C-acetate. J Nucl Med. 48:420–428. 2007.PubMed/NCBI

9 

Oka S, Hattori R, Kurosaki F, Toyama M, Williams LA, Yu W, Votaw JR, Yoshida Y, Goodman MM and Ito O: A preliminary study of anti-1-amino-3-18F-fluorocyclobutyl-1-carboxylic acid for the detection of prostate cancer. J Nucl Med. 48:46–55. 2007.PubMed/NCBI

10 

Tehrani OS, Muzik O, Heilbrun LK, Douglas KA, Lawhorn-Crews JM, Sun H, Mangner TJ and Shields AF: Tumor imaging using 1-(2′-deoxy-2′-18F-fluoro-beta-D-arabinofuranosyl)thymine and PET. J Nucl Med. 48:1436–1441. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Larson SM, Morris M, Gunther I, Beattie B, Humm JL, Akhurst TA, Finn RD, Erdi Y, Pentlow K, Dyke J, et al: Tumor localization of 16beta-18F-fluoro-5alpha-dihydrotestosterone versus 18F-FDG in patients with progressive, metastatic prostate cancer. J Nucl Med. 45:366–373. 2004.PubMed/NCBI

12 

Wang W and Mo ZN: Advances in prostate-specific membrane antigen targeted therapies for prostate cancer. Zhonghua Nan Ke Xue. 16:547–551. 2010.(In Chinese). PubMed/NCBI

13 

Rajasekaran SA, Anilkumar G, Oshima E, Bowie JU, Liu H, Heston W, Bander NH and Rajasekaran AK: A novel cytoplasmic tail MXXXL motif mediates the internalization of prostate-specific membrane antigen. Mol Biol Cell. 14:4835–4845. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Kim H, Shoji S, Tomonaga T, Shima M, Terachi T and Uchida T: Prostate cancer with cyst formation detected by whole body positron emission tomography/computed tomography: A case report. Oncol Lett. 8:2037–2039. 2014.PubMed/NCBI

15 

Tagawa ST, Beltran H, Vallabhajosula S, Goldsmith SJ, Osborne J, Matulich D, Petrillo K, Parmar S, Nanus DM and Bander NH: Anti-prostate-specific membrane antigen-based radioimmunotherapy for prostate cancer. Cancer. 116:(Suppl). 1075–1083. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Maresca KP, Hillier SM, Femia FJ, Keith D, Barone C, Joyal JL, Zimmerman CN, Kozikowski AP, Barrett JA, Eckelman WC, et al: A series of halogenated heterodimeric inhibitors of prostate specific membrane antigen (PSMA) as radiolabeled probes for targeting prostate cancer. J Med Chem. 52:347–357. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Pomper MG, Musachio JL, Zhang J, Scheffel U, Zhou Y, Hilton J, Maini A, Dannals RF, Wong DF and Kozikowski AP: 11C-MCG: Synthesis, uptake selectivity, and primate PET of a probe for glutamate carboxypeptidase II (NAALADase). Mol Imaging. 1:96–101. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Foss CA, Mease RC, Fan H, Wang Y, Ravert HT, Dannals RF, Olszewski RT, Heston WD, Kozikowski AP and Pomper MG: Radiolabeled small-molecule ligands for prostate-specific membrane antigen: In vivo imaging in experimental models of prostate cancer. Clin Cancer Res. 11:4022–4028. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Mease RC, Dusich CL, Foss CA, Ravert HT, Dannals RF, Seidel J, Prideaux A, Fox JJ, Sgouros G, Kozikowski AP, et al: N-[N-[(S)-1,3-Dicarboxypropyl]carbamoyl]-4-[18F]fluorobenzyl-L-cysteine, [18F]DCFBC: a new imaging probe for prostate cancer. Clin Cancer Res. 14:3036–3043. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Kularatne SA, Wang K, Santhapuram HK and Low PS: Prostate-specific membrane antigen targeted imaging and therapy of prostate cancer using a PSMA inhibitor as a homing ligand. Mol Pharm. 6:780–789. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Zhang AX, Murelli RP, Barinka C, Michel J, Cocleaza A, Jorgensen WL, Lubkowski J and Spiegel DA: A remote arene-binding site on prostate specific membrane antigen revealed by antibody-recruiting small molecules. J Am Chem Soc. 132:12711–12716. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Qi Y, Zhang Q, Huang Y and Wang D: Manifestations and pathological features of solitary thin-walled cavity lung cancer observed by CT and PET/CT imaging. Oncol Lett. 8:285–290. 2014.PubMed/NCBI

Related Articles

Journal Cover

October-2015
Volume 10 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fan W, Zhang Z, Zhu Z, Yang D, Chen X, Wang J, Chen F and Song X: Synthesis and positron emission tomography evaluation of 18F-Glu-Urea-Lys, a prostate-specific membrane antigen-based imaging agent for prostate cancer Corrigendum in /10.3892/ol.2015.4039. Oncol Lett 10: 2299-2302, 2015
APA
Fan, W., Zhang, Z., Zhu, Z., Yang, D., Chen, X., Wang, J. ... Song, X. (2015). Synthesis and positron emission tomography evaluation of 18F-Glu-Urea-Lys, a prostate-specific membrane antigen-based imaging agent for prostate cancer Corrigendum in /10.3892/ol.2015.4039. Oncology Letters, 10, 2299-2302. https://doi.org/10.3892/ol.2015.3625
MLA
Fan, W., Zhang, Z., Zhu, Z., Yang, D., Chen, X., Wang, J., Chen, F., Song, X."Synthesis and positron emission tomography evaluation of 18F-Glu-Urea-Lys, a prostate-specific membrane antigen-based imaging agent for prostate cancer Corrigendum in /10.3892/ol.2015.4039". Oncology Letters 10.4 (2015): 2299-2302.
Chicago
Fan, W., Zhang, Z., Zhu, Z., Yang, D., Chen, X., Wang, J., Chen, F., Song, X."Synthesis and positron emission tomography evaluation of 18F-Glu-Urea-Lys, a prostate-specific membrane antigen-based imaging agent for prostate cancer Corrigendum in /10.3892/ol.2015.4039". Oncology Letters 10, no. 4 (2015): 2299-2302. https://doi.org/10.3892/ol.2015.3625