DOI QR코드

DOI QR Code

Psammaplin A-Modified Novel Radiosensitizers for Human Lung Cancer and Glioblastoma Cells

  • Wee, Chan Woo (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Kim, Jin Ho (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Kim, Hak Jae (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Kang, Hyun-Cheol (Department of Radiation Oncology, Seoul National University College of Medicine) ;
  • Suh, Soo Youn (Cancer Research Institute, Seoul National University College of Medicine) ;
  • Shin, Beom Soo (School of Pharmacy, Sungkyunkwan University) ;
  • Ma, Eunsook (College of Pharmacy, Daegu Catholic University) ;
  • Kim, Il Han (Department of Radiation Oncology, Seoul National University College of Medicine)
  • Received : 2018.11.30
  • Accepted : 2019.02.11
  • Published : 2019.03.31

Abstract

Background: Psammaplin A (PsA) is a radiosensitizer whereas its clinical application is hampered by poor bioavailability. This study aimed to synthesize novel radiosensitizers using PsA as the lead compound. Materials and Methods: Eight homodimeric disulfides were synthesized from corresponding acid and cystamine dihydrochloride in N-hydroxysuccinimide and dicyclohexylcarbodiimide coupling conditions. One monomeric thiol analog was obtained by reduction of homodimeric disulfide with dithiothreitol. Clonogenic assay was used to measure cell survival after irradiation and drug treatment in human lung cancer (A549) and glioblastoma (U373MG) cells. Results and Discussion: Using the PsA backbone, nine compounds were synthesized. Eight compounds showed variable cytotoxicity with 50% inhibitory concentrations ranging $16.14{\mu}M$ to $150.10{\mu}M$ (A549), and $13.25{\mu}M$ to $50.15{\mu}M$ (U373MG). Four and six compounds radiosensitized A549 and U373MG cells, respectively. Two compounds that radiosensitized both cell lines were tested for its inhibitory effects on DNMT1. One of them was shown to significantly inhibit DNMT1 activity. Conclusion: Novel compounds with radiosensitizing activity were synthesized. These compounds have a great potential to serve as a basis for the development of future radiosensitizers. Further investigation is warranted for their clinical application.

Keywords

References

  1. Begg AC, Stewart FA, Conchita V. Strategies to improve radiotherapy with targeted drugs. Nat. Rev. Cancer. 2011;11(4):239-253. https://doi.org/10.1038/nrc3007
  2. Esteller M. Relevance of DNA methylation in the management of cancer. Lancet. Oncol. 2003;4(6):351-358. https://doi.org/10.1016/S1470-2045(03)01115-X
  3. Lyko F, Brown R. DNA Methyltransferase inhibitors and the development of epigenetic cancer therapies. J. Natl. Cancer. Inst. 2005;97(20):1498-1506. https://doi.org/10.1093/jnci/dji311
  4. Manoharan M, Ramachandran K, Soloway MS, Singal R. Epigenetic targets in the diagnosis and treatment of prostate cancer. Int. Braz. J. Urol. 2007;33(1):11-18. https://doi.org/10.1590/S1677-55382007000100003
  5. Lin RK, Hsu HS, Chang JW, Chen CY, Chen JT, Wang YC. Alteration of DNA methyltransferases contributes to 5'CpG methylation and poor prognosis in lung cancer. Lung. Cancer. 2007; 55(2):205-213. https://doi.org/10.1016/j.lungcan.2006.10.022
  6. Eads CA, Danenberg KD, Kawakami K, Saltz LB, Danenberg PV, Laird PW. CpG island hypermethylation in human colorectal tumors is not associated with DNA methyltransferase overexpression. Cancer. Res. 1999;59(10):2302-2306.
  7. Silverman LR, Demakos EP, Peterson BL. Randomized controlled trial of azacitidine in patients with the myelodysplastic syndrome: a study of the cancer and leukemia group B. J. Clin. Oncol. 2002;20(10):2429-2440. https://doi.org/10.1200/JCO.2002.04.117
  8. Pechalrieu D, Etievant C, Arimondo PB. DNA methyltransferase inhibitors in cancer: From pharmacology to translational studies. Biochem. Pharmacol. 2017;129;1-13. https://doi.org/10.1016/j.bcp.2016.12.004
  9. Kristensen LS, Nielsen HM, Hansen LL. Epigenetic and cancer treatment. Eur. J. Pharmacol. 2009;625(1-3):131-142. https://doi.org/10.1016/j.ejphar.2009.10.011
  10. Oki Y, Aoki E, Issa JP. Decitabine-bedside to bench. Crit. Rev. Oncol. 2007;61(2):140-152. https://doi.org/10.1016/j.critrevonc.2006.07.010
  11. Dote H, Cerna D, Burgan WE, Carter DJ, Cerra MA, Hollingshead MG, Camphausen K, Tofilon PJ. Enhancement of in vitro and in vivo tumor cell radiosensitivity by the DNA methylation inhibitor zebularine. Clin. Cancer. Res. 2005;11(12):4571-4579. https://doi.org/10.1158/1078-0432.CCR-05-0050
  12. Cho HJ, Kim SY, Kim KH, Kang WK, Kim JI, Oh ST, Kim JS, An CH. The combination effect of sodium butyrate and 5-Aza-2'- deoxycytidine on radiosensitivity in RKO colorectal cancer and MCF-7 breast cancer cell lines. World. J. Surg. Oncol. 2009;7:49. https://doi.org/10.1186/1477-7819-7-49
  13. Qiu H, Yashiro M, Shinto O, Matsuzaki T, Hirakawa K. DNA methyltransferase inhibitor 5-aza-CdR enhances the radiosensitivity of gastric cancer cells. Cancer. Sci. 2009;100(1):181-188. https://doi.org/10.1111/j.1349-7006.2008.01004.x
  14. De Schutter H, Kimpe M, Isebaert S, Nuyts S. A systematic assessment of radiation dose enhancement by 5-aza-2'-deoxycytidine and histone deacetylase inhibitors in head-and-neck squamous cell carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 2009;73(3):904-912. https://doi.org/10.1016/j.ijrobp.2008.10.032
  15. Lübbert M. DNA methylation inhibitors in the treatment of leukemias, myelodysplastic syndromes and hemoglobinopathies: clinical results and possible mechanisms of action. Curr. Top. Microbiol. Immunol. 2000;249:135-164.
  16. Aparicio A, Weber JS. Review of the clinical experience with 5-azacytidine and 5-aza-2'-deoxycytidine in solid tumors. Curr. Opin. Investig. Drugs. 2002;3(4):627-633.
  17. Arabshahi L, Schmitz FJ. Brominated tyrosine metabolites from an unidentified sponge. J. Org. Chem. 1987;52(16):3584-3586. https://doi.org/10.1021/jo00392a016
  18. Pina IC, et al. Psammaplins from the sponge Pseudoceratina purpurea: inhibition of both histone deacetylase and DNA methyltransferase. J. Org. Chem. 2003;68:3866-3873. https://doi.org/10.1021/jo034248t
  19. Baud MG, et al. Defining the mechanism of action and enzymatic selectivity of psammaplin A against its epigenetic targets. J. Med. Chem. 2012;55(4):1731-1750. https://doi.org/10.1021/jm2016182
  20. Kim JH, Kim IH, Shin JH, Kim HJ, Kim IA. Sequence-dependent radiosensitization of histone deacetylase inhibitors trichostatin A and SK-7041. Cancer. Res. Treat. 2013;45(4):334-342. https://doi.org/10.4143/crt.2013.45.4.334
  21. Kim HJ, Kim JH, Chie EK, Park DY, Kim IA, Kim IH. DNMT (DNA methyltransferase) inhibitors radiosensitize human cancer cells by suppressing DNA repair activity. Radiat. Oncol. 2012;7:39. https://doi.org/10.1186/1748-717X-7-39
  22. Kim HJ, Kim TH, Seo WS, Yoo SD, Kim IH, Joo SH, Shin S, Park ES, Ma ES, Shin BS. Pharmacokinetics and tissue distribution of psammaplin A, a novel anticancer agent, in mice. Arch. Pharm. Res. 2012;35(10):1849-1854. https://doi.org/10.1007/s12272-012-1019-5
  23. Löffelhardt W. The biosynthesis of phenylacetic acids in the blue-green alga Anacytis nidulans: Evidence for the involvement of a thylakoid-bound L-amino acid oxidase. Z. Naturforsch. 1977;32(5-6):345-350. https://doi.org/10.1515/znc-1977-5-606
  24. Nicolaou KC, Hughes R, Pfefferkorn JA, Barluenga S, Roecker AJ. Combinational synthesis through disulfide exchange: Discovery of potent psammaplin A type antibacterial agent active against methicillin-resistant Staphylococcus aureus (MRSA). Chem. Eur. J. 2001;7(19):4280-4295. https://doi.org/10.1002/1521-3765(20011001)7:19<4280::AID-CHEM4280>3.0.CO;2-3
  25. Mautner HG, Chu SH, Gunther WHH. The aminolysis of thioacyl and selenoacyl analogs. J. Am. Chem. Soc. 1963;85(21):3458-3462. https://doi.org/10.1021/ja00904a034
  26. Godert AM, Angelino N, Woloszynska-Read A, Morey SR, James SR, Karpf AR, Sufrin JR. An improved synthesis of psammaplin A. Bioorg. Med. Chem. Lett. 2006;16(12):3330-3333. https://doi.org/10.1016/j.bmcl.2006.03.008
  27. Kim DK, et al. Synthesis and biological evaluation of 3-(4-substituted- phenyl)-N-hydroxy-2-propenamides, a new class of histone deacetylase inhibitors. J. Med. Chem. 2003;46(26):5745-5751. https://doi.org/10.1021/jm030377q
  28. Chie EK, Shin JH, Kim IA, Kim IH. In vivo radiosensitization effect of HDAC inhibitor, SK-7041 on RIF-1 cell line. J. Korean. Soc. Ther. Radiol. Oncol. 2010;28(4):219-223. https://doi.org/10.3857/jkstro.2010.28.4.219
  29. Lee JY, Lee MY, Ha MW, Won TH, Cho HJ, Shin J, Park HG, Kim DD. Determination and validation of psammaplin A and its derivatives in rat plasma by liquid chromatography-tandem mass spectrometry and its application in pharmacokinetic study. J. Chromatogr. B. 2015;1000:155-162. https://doi.org/10.1016/j.jchromb.2015.07.014
  30. Gillis EP, Eastman KJ, Hill MD, Donnelly DJ, Meanwell NA. Applications of fluorine in medicinal chemistry. J. Med. Chem. 2015; 58(21):8315-8359. https://doi.org/10.1021/acs.jmedchem.5b00258
  31. Wasserman TH, Brizel DM. The role of amifostine as a radioprotector. Oncology. 2001;15(10):1349-1360.
  32. Hosseinimehr SJ. Trends in the development of radioprotective agents. Drug. Discovery. Today. 2007;12(19-20):794-805. https://doi.org/10.1016/j.drudis.2007.07.017