DOI QR코드

DOI QR Code

Natural radioprotectors and their impact on cancer drug discovery

  • Kuruba, Vinutha (Department of Biotechnology, NMAM Institute of Technology) ;
  • Gollapalli, Pavan (Department of Biotechnology, Vignan's Foundation for Science, Technology and Research (Deemed to be University))
  • Received : 2018.08.14
  • Accepted : 2018.11.08
  • Published : 2018.12.31

Abstract

Cancer is a complex multifaceted illness that affects different patients in discrete ways. For a number of cancers the use of chemotherapy has become standard practice. Chemotherapy is a use of cytostatic drugs to cure cancer. Cytostatic agents not only affect cancer cells but also affect the growth of normal cells; leading to side effects. Because of this, radiotherapy gained importance in treating cancer. Slaughtering of cancerous cells by radiotherapy depends on the radiosensitivity of the tumor cells. Efforts to improve the therapeutic ratio have resulted in the development of compounds that increase the radiosensitivity of tumor cells or protect the normal cells from the effects of radiation. Amifostine is the only chemical radioprotector approved by the US Food and Drug Administration (FDA), but due to its side effect and toxicity, use of this compound was also failed. Hence the use of herbal radioprotectors bearing pharmacological properties is concentrated due to their low toxicity and efficacy. Notably, in silico methods can expedite drug discovery process, to lessen the compounds with unfavorable pharmacological properties at an early stage of drug development. Hence a detailed perspective of these properties, in accordance with their prediction and measurement, are pivotal for a successful identification of radioprotectors by drug discovery process.

Keywords

References

  1. Mustonen T, Alitalo K. Endothelial receptor tyrosine kinases involved in angiogenesis. J Cell Biol 1995;129:895-8.
  2. Alison MR. Cancer [Internet]. Hoboken, NJ: John Wiley & Sons Inc.; 2001 [cited 2018 Nov 10]. Available from: https://onlinelibrary.wiley.com/doi/full/10.1038/npg.els.0001471.
  3. Chabner BA, Boral AL, Multani P. Translational research: walking the bridge between idea and cure: seventeenth Bruce F. Cain Memorial Award lecture. Cancer Res 1998;58:4211-6.
  4. International Agency for Research on Cancer. GLOBOCAN 2008: cancer incidence and mortality worldwide [Internet]. Lyon, France: International Agency for Research on Cancer; 2010 [cited 2017 Nov 10]. Available from: https://www.iarc.fr/en/media-centre/iarcnews/2010/globocan2008.php.
  5. Lawrence TS, Ten Haken RK, Giaccia A. Principles of radiation oncology. In: DeVita VT, Lawrence TS, Rosenberg SA, editors. DeVita, Hellman, and Rosenberg's cancer: principles and practice of oncology. 8th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2008. p. 307-36.
  6. Nair CK, Parida DK, Nomura T. Radioprotectors in radiotherapy. J Radiat Res 2001;42:21-37.
  7. Schulz-Ertner D, Jakel O, Schlegel W. Radiation therapy with charged particles. Semin Radiat Oncol 2006;16:249-59.
  8. Barnett GC, West CM, Dunning AM, et al. Normal tissue reactions to radiotherapy: towards tailoring treatment dose by genotype. Nat Rev Cancer 2009;9:134-42.
  9. Wasserman TH, Chapman JD. Radiation response modulation. Part A. Chemical sensitizers and protectors. In: Perez & Brady's principles and practice of radiation oncology. 4th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2004.
  10. Ross GM. Induction of cell death by radiotherapy. Endocr Relat Cancer 1999;6:41-4.
  11. Citrin D, Cotrim AP, Hyodo F, Baum BJ, Krishna MC, Mitchell JB. Radioprotectors and mitigators of radiation-induced normal tissue injury. Oncologist 2010;15:360-71.
  12. Raviraj J, Bokkasam VK, Kumar VS, Reddy US, Suman V. Radiosensitizers, radioprotectors, and radiation mitigators. Indian J Dent Res 2014;25:83-90.
  13. Arora R, Gupta D, Chawla R, et al. Radioprotection by plant products: present status and future prospects. Phytother Res 2005;19:1-22.
  14. Monig H, Messerschmidt O, Streffer C. Chemical radioprotection in mammals and in man. In: Schere E, Streffer C, Trott KR, editors. Radiation exposure and occupational risks. Heidelberg: Springer; 1990. p. 97-143.
  15. Bourhis J, Rosine D. Radioprotective effect of amifostine in patients with head and neck squamous cell carcinoma. Semin Oncol 2002;29(6 Suppl 19):61-2.
  16. Hazra B, Ghosh S, Kumar A, Pandey BN. The prospective role of plant products in radiotherapy of cancer: a current overview. Front Pharmacol 2012;2:94.
  17. Tannehill SP, Mehta MP. Amifostine and radiation therapy: past, present, and future. Semin Oncol 1996;23(4 Suppl 8):69-77.
  18. Lans C. Ethnomedicines used in Trinidad and Tobago for reproductive problems. J Ethnobiol Ethnomed 2007;3:13.
  19. Mantena SK, Unnikrishnan MK, Uma Devi P. Radioprotective effect of sulfasalazine on mouse bone marrow chromosomes. Mutagenesis 2008;23:285-92.
  20. Sweeney TR. A survey of compounds from the antiradiation drug development program of the US army medical research and development command. Washington, DC: Walter Reed Army Institute of Research; 1979.
  21. Brown JM, Diehn M, Loo BW Jr. Stereotactic ablative radiotherapy should be combined with a hypoxic cell radiosensitizer. Int J Radiat Oncol Biol Phys 2010;78:323-7.
  22. Hicks KO, Siim BG, Jaiswal JK, et al. Pharmacokinetic/pharmacodynamic modeling identifies SN30000 and SN29751 as tirapazamine analogues with improved tissue penetration and hypoxic cell killing in tumors. Clin Cancer Res 2010;16:4946-57.
  23. Skellett A, Swift L, Tan E, Garioch J. A randomized, doubleblind, negatively controlled pilot study to determine whether the use of emollients or calcipotriol alters the sensitivity of the skin to ultraviolet radiation during phototherapy with narrowband ultraviolet B. Br J Dermatol 2011;164:402-6.
  24. Zenk JL. New therapy for the prevention and prophylactic treatment of acute radiation syndrome. Expert Opin Investig Drugs 2007;16:767-70.
  25. Ghosh SP, Perkins MW, Hieber K, et al. Radiation protection by a new chemical entity, Ex-Rad: efficacy and mechanisms. Radiat Res 2009;171:173-9.
  26. Fukuzawa N, Petro M, Baldwin WM 3rd, Gudkov AV, Fairchild RL. A TLR5 agonist inhibits acute renal ischemic failure. J Immunol 2011;187:3831-9.
  27. Stickney DR, Dowding C, Authier S, et al. 5-androstenediol improves survival in clinically unsupported rhesus monkeys with radiation-induced myelosuppression. Int Immunopharmacol 2007;7:500-5.
  28. Srinivasan V, Doctrow S, Singh VK, Whitnall MH. Evaluation of EUK-189, a synthetic superoxide dismutase/catalase mimetic as a radiation countermeasure. Immunopharmacol Immunotoxicol 2008;30:271-90.
  29. Goff JP, Epperly MW, Dixon T, et al. Radiobiologic effects of GS-nitroxide (JP4-039) on the hematopoietic syndrome. In Vivo 2011;25:315-23.
  30. Thotala DK, Geng L, Dickey AK, Hallahan DE, Yazlovitskaya EM. A new class of molecular targeted radioprotectors: GSK-3beta inhibitors. Int J Radiat Oncol Biol Phys 2010;76:557-65.
  31. C Jagetia G. Radioprotective potential of plants and herbs against the effects of ionizing radiation. J Clin Biochem Nutr 2007;40:74-81.
  32. Hosseinimehr SJ. Trends in the development of radioprotective agents. Drug Discov Today 2007;12:794-805.
  33. Weiss JF, Landauer MR. Protection against ionizing radiation by antioxidant nutrients and phytochemicals. Toxicology 2003;189:1-20.
  34. Sandhya T, Mishra KP. Cytotoxic response of breast cancer cell lines, MCF 7 and T 47 D to triphala and its modification by antioxidants. Cancer Lett 2006;238:304-13.
  35. Veeraraghavan J, Aravindan S, Natarajan M, Awasthi V, Herman TS, Aravindan N. Neem leaf extract induces radiosensitization in human neuroblastoma xenograft through modulation of apoptotic pathway. Anticancer Res 2011;31:161-70.
  36. Bache M, Zschornak MP, Passin S, et al. Increased betulinic acid induced cytotoxicity and radiosensitivity in glioma cells under hypoxic conditions. Radiat Oncol 2011;6:111.
  37. Bansal P, Paul P, Nayak PG, et al. Phenolic compounds isolated from Pilea microphylla prevent radiation-induced cellular DNA damage. Acta Pharm Sin B 2011;1:226-35.
  38. Manach C, Williamson G, Morand C, Scalbert A, Remesy C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr 2005;81(1 Suppl):230S-242S.
  39. Nijveldt RJ, van Nood E, van Hoorn DE, Boelens PG, van Norren K, van Leeuwen PA. Flavonoids: a review of probable mechanisms of action and potential applications. Am J Clin Nutr 2001;74:418-25.
  40. Conney AH, Lysz T, Ferraro T, et al. Inhibitory effect of curcumin and some related dietary compounds on tumor promotion and arachidonic acid metabolism in mouse skin. Adv Enzyme Regul 1991;31:385-96.
  41. Wang S, Sim TB, Kim YS, Chang YT. Tools for target identification and validation. Curr Opin Chem Biol 2004;8:371-7.
  42. Gorbounova V, Khokhlova S, Orel N. Docetaxel and cisplatin as firstline chemotherapy in patients with advanced ovarian cancer. In: 36th Annual American Society of Clinical Oncology Meeting; 2000 May 20-23; New Orleans, LA. Abstract 1536.
  43. Zhou J, Liu M, Aneja R, Chandra R, Joshi HC. Enhancement of paclitaxel-induced microtubule stabilization, mitotic arrest, and apoptosis by the microtubule-targeting agent EM012. Biochem Pharmacol 2004;68:2435-41.
  44. Cragg GM, Newman DJ, Weiss RB. Coral reefs, forests, and thermal vents: the worldwide exploration of nature for novel antitumor agents. Semin Oncol 1997;24:156-63.
  45. Newman DJ, Cragg GM. Natural products as sources of new drugs over the last 25 years. J Nat Prod 2007;70:461-77.
  46. Searls DB. Data integration: challenges for drug discovery. Nat Rev Drug Discov 2005;4:45-58.
  47. Apic G, Ignjatovic T, Boyer S, Russell RB. Illuminating drug discovery with biological pathways. FEBS Lett 2005;579:1872-7.
  48. Claus BL, Underwood DJ. Discovery informatics: its evolving role in drug discovery. Drug Discov Today 2002;7:957-66.
  49. Imming P, Sinning C, Meyer A. Drugs, their targets and the nature and number of drug targets. Nat Rev Drug Discov 2006;5:821-34.
  50. Huang CM, Elmets CA, Tang DC, Li F, Yusuf N. Proteomics reveals that proteins expressed during the early stage of Bacillus anthracis infection are potential targets for the development of vaccines and drugs. Genomics Proteomics Bioinformatics 2004;2:143-51.
  51. Jackson RC. Update on computer-aided drug design. Curr Opin Biotechnol 1995;6:646-51.
  52. Stears RL, Martinsky T, Schena M. Trends in microarray analysis. Nat Med 2003;9:140-5.
  53. Hormozdiari F, Salari R, Bafna V, Sahinalp SC. Protein-protein interaction network evaluation for identifying potential drug targets. J Comput Biol 2010;17:669-84.
  54. Shoichet BK. Virtual screening of chemical libraries. Nature 2004;432:862-5.
  55. Dennis M, Sammes PG, Taylor JB. Absorption processes. In: Hansch C, editor. Comprehensive medicinal chemistry. Oxford, UK: Pergamon Press; 1990. p. 1-44.
  56. Faller B, Wohnsland F. Physiochemical parameters as tools. In: Testa B, van der Waterbeemd H, Folkers G, Guy R, editors. Pharmacokinetic optimization in drug research. Zurich, Switzerland: Wiley-VCH; 2001. p. 257-73.
  57. Good AC, Oprea TI. Optimization of CAMD techniques 3. Virtual screening enrichment studies: a help or hindrance in tool selection? J Comput Aided Mol Des 2008;22:169-78.
  58. Maurya DK, Salvi VP, Krishnan Nair CK. Radioprotection of normal tissues in tumor-bearing mice by troxerutin. J Radiat Res 2004;45:221-8.
  59. Hollstein M, Rice K, Greenblatt MS, et al. Database of p53 gene somatic mutations in human tumors and cell lines. Nucleic Acids Res 1994;22:3551-5.
  60. Ren L, Xie D, Li P, et al. Radiation protective effects of baclofen predicted by a computational drug repurposing strategy. Pharmacol Res 2016;113(Pt A):475-83.
  61. Horst JA, Laurenzi A, Bernard B, Samudrala R. Computational multitarget drug discovery. In: Peters JU, editor. Polypharmacology in drug discovery. Hoboken, NJ: John Wiley & Sons; 2012. p. 263-302.
  62. Jin G, Wong ST. Toward better drug repositioning: prioritizing and integrating existing methods into efficient pipelines. Drug Discov Today 2014;19:637-44.

Cited by

  1. (−)-Epicatechin mitigates radiation-induced intestinal injury and promotes intestinal regeneration via suppressing oxidative stress vol.53, pp.8, 2019, https://doi.org/10.1080/10715762.2019.1635692
  2. Assessment of monoamine neurotransmitters in the cortex and cerebellum of gamma-irradiated mice: A neuromodulatory role of Cynodon dactylon vol.19, pp.1, 2018, https://doi.org/10.4103/jcar.jcar_13_19
  3. Reflectance structured illumination imaging of internalized cerium oxide nanoparticles modulating dose-dependent reactive oxygen species in breast cancer cells vol.22, pp.None, 2018, https://doi.org/10.1016/j.bbrep.2020.100745
  4. Roles of Therapeutic Bioactive Compounds in Hepatocellular Carcinoma vol.2021, pp.None, 2018, https://doi.org/10.1155/2021/9068850
  5. Radioprotective potential of Costus afer against the radiation-induced hematological and histopathological damage in mice vol.39, pp.1, 2018, https://doi.org/10.3857/roj.2021.00017
  6. Cytotoxicity and radiosensitizing potency of Moscatilin in cancer cells at low radiation doses of X-ray and UV-C vol.11, pp.6, 2021, https://doi.org/10.1007/s13205-021-02827-3
  7. Effects of radiation and role of plants in radioprotection: A critical review vol.779, pp.None, 2021, https://doi.org/10.1016/j.scitotenv.2021.146431
  8. Protective effects of Brownea grandiceps (Jacq.) against ϒ-radiation-induced enteritis in rats in relation to its secondary metabolome fingerprint vol.146, pp.None, 2022, https://doi.org/10.1016/j.biopha.2021.112603
  9. Bamboo leaf extract ameliorates radiation induced genotoxicity: An in vitro study of chromosome aberration assay vol.31, pp.None, 2018, https://doi.org/10.1016/j.hermed.2021.100528