• No results found

Development and preclinical comparison of two non- non-peptidomimetic MMP/ADAM inhibitors for PET

Methyl 1-((4-nitrophenyl)sulfonyl)piperazine-2-carboxylate hydrochloride (14) Thionyl chloride (7.00 mL, 96.5 mmol) was added dropwise to a solution of

4.10 Preclinical studies

Animals were injected with [18F]-1A or [18F]-2 when the tumors had reached an adequate size (0.3-0.6 mL), after 14 to 21 days of inoculation. The mice were randomly divided into two groups: tumor-bearing mice injected with [18F]-1A (or [18F]-2) and with or without coinjection of 2.5 mg/kg of 1A (or 2). [18F]-1A (1.55 ± 1.38 MBq, 0.031 ± 0.028 nmol) (or [18F]-2 (2.12 ± 3.28 MBq, 0.043 ± 0.080 nmol)), dissolved in saline (maximum volume 200 μL per injection), was intravenously injected into the penile vein of mice anesthetized with 5% isoflurane (Pharmacie BV, The Netherlands) that was mixed with medical air at a flow rate of 2 mL.min-1, after which anesthesia was maintained with 2% isoflurane. For each tracer, 12 mice were used. In each group (either baseline or block), one animal was scanned using PET. Following induction of anesthesia, the mice were then positioned in the bed of the microPET camera (Focus 220, Siemens Medical Solutions USA, Inc.) in transaxial position. The body temperature of mice was maintained by electronically regulated heating pads. Data acquisition of the microPET camera was initiated and continued for a period of 90 min. After the completion of the dynamic emission scan, a 515 sec transmission scan with a Co-57 point source was made for correc-tion of attenuacorrec-tion of 511 keV photons by tissue. After microPET scanning, the mice were terminated by administering a high dose of isoflurane (5%) for about 20 min.

CHAPTER

5

Ex vivo biodistribution

Ex vivo biodistribution studies were performed on the sacrificed mice 90 min p.i of [18F]-1A (± 1A) or [18F]-2 (± 2). The following organs were taken: bladder, bone, brain, heart, kidney, large intestine, liver, lungs, muscle, pancreas, small intestine, spleen, stomach, tumor and urine. A small drop of infusate was collected for cal-culation of SUVmean. The blood was centrifuged in order to collect plasma and red blood cells. All samples were weighed and levels of radioactivity were determined using a gamma counter. Tracer uptake was expressed as the standardized uptake value (SUVmean), defined as [tissue activity concentration (MBq/g)/(injected activity (MBq)/mouse body weight (g))].

MicroPET image analysis

Emission sinograms were iteratively reconstructed (OSEM2d) after being nor-malized, corrected for attenuation, and corrected for decay of radioactivity. The list-mode data of the emission scans were separated into 22 frame sinograms (15 frames of 2 minutes, 3 frames of 5 minutes, 2 frames of 7.5 minutes, 2 frames of 15 minutes; zoom factor, 4). PET image analysis was performed using Inveon Research Workplace (Siemens) software. Regions of interest were drawn around the tumor.

The uptake of the tracer in the region of interest was determined in Bq.cm-3, which was converted to PET-SUVmean using the following formula: [tissue activity concen-tration (MBq/cc)/(injected activity (MBq)/mouse body weight (g))].

Acknowledgements

The authors wish to thank the Dutch Technology Foundation (STW) for financial support (project 08008).

References

1. Southan C. A genomic perspective on human proteases as drug targets. Drug Discov Today. 2001;

6(13):681-8.

2. Flipo M, Beghyn T, Charton J, Leroux VA, Deprez BP, Deprez-Poulain RF. A library of novel hydroxamic acids targeting the metallo-protease family: design, parallel synthesis and screening. Bioorg Med Chem. 2007;15(1):63-76.

3. Hu J, Van den Steen PE, Sang Q-XA, Opdenakker G. Matrix metalloproteinase inhibitors as therapy for inflammatory and vascular diseases. Nat Rev Drug Discov. 2007;6(6):480-98.

4. Galis ZS, Sukhova GK, Lark MW, Libby P. Increased expression of matrix metalloproteinase and matrix degrading activity in vulnerable regions of human atherosclerotic plaques. J Clin Invest.

1994;94:2493–503.

5. Moss ML, White JM, Lambert MH, Andrews RC. TACE and other ADAM proteases as targets for drug discovery. Drug Discov Today. 2001;6(8):417–26.

6. Whittaker M, Floyd CD, Brown P, Gearing AJH. Design and therapeutic application of matrix metal-loproteinase inhibitors. Chem Rev. 1999;99(9):2735-76.

7. Matusiak N, van Waarde A, Bischoff R, et al. Probes for non-invasive matrix metalloproteinase-targeted imaging with PET and SPECT. Curr Pharm Des. 2013;19:4647-72.

8. Konstantinopoulos PA, Karamouzis MV, Papatsoris AG, Papavassiliou AG. Matrix metalloproteinase inhibitors as anticancer agents. Int J Biochem Cell Biol. 2008;40(6-7):1156-68.

9. Cheng M, De B, Pikul S, Almstead NG, et al. Design and synthesis of piperazine-based matrix metal-loproteinase inhibitors. J Med Chem. 2000;43:369–80.

10. Achilefu S. Introduction to concepts and strategies for molecular imaging. Chem Rev.

2010;110(5):2575-8.

11. Fei X, Zheng Q-H, Liu X, et al. Synthesis of MMP inhibitor radiotracer [11C]CGS 25966, a new poten-tial PET tumor imaging agent. J Labelled Comp Radiopharm. 2003;46(4):343-51.

12. Bremer C, Bredow S, Mahmood U, et al. Optical imaging of matrix metalloproteinase – 2 activity in tumors: feasibility study in a mouse model. Radiology. 2001;221(2):523-9.

13. Zhao T, Harada H, Teramura Y, et al. A novel strategy to tag matrix metalloproteinases-positive cells for in vivo imaging of invasive and metastatic activity of tumor cells. J Control Release. 2010;144:

109–14.

14. Olson ES, Jiang T, Aguilera TA, et al. Activatable cell penetrating peptides linked to nanoparticles as dual probes for in vivo fluorescence and MR imaging of proteases. Proc Natl Acad Sci USA. 2010;107:

4311–16.

15. Cheng T-L, Chuang C-H, Chuang K-H, et al. In vivo positron emission tomography imaging of protease activity by generation of a hydrophobic product from a non-inhibitory protease substrate. Clin Cancer Res. 2012;18(1):238-47.

16. Bellac CL, Li Y, Lou Y, et al. Novel MMP inhibitor [18F]-Marimastat-aryltrifluoroborate as a probe for in vivo PET imaging in cancer. Cancer Res. 2010;70(19):7562-9.

17. Matusiak N, Castelli R, Tuin AW et al. A dual inhibitor of matrix metalloproteinases and a disintegrin and metalloproteinases, [18F]FB-ML5, as a molecular probe for non-invasive MMP/ADAM-targeted imaging. Bioorg Med Chem. 2015;23(1):192-202.

18. Murphy, G. Tissue inhibitors of metalloproteinases. Genome Biol. 2011;12(233):1-7.

19. Shipley JM, Doyle GA, Fliszar CJ, et al. The structural basis for the elastolytic activity of the 92-kDa and 72-kDa gelatinases. Role of the fibronectin type II-like repeats. J Biol Chem. 1996;271:4335-41.

CHAPTER

5

20. Parkar AA, Stow MD, Smith K, et al. Large-scale expression, refolding, and purification of the catalytic domain of human macrophage metalloelastase (MMP-12) in Escherichia coli. Protein Expr Purif. 2000;20:152-61.

6 |