Vitamin D as Radiosensitizer: A Review in Cell Line
PDF

Keywords

Vitamin D, cancer, radiosensitizer, radiotherapy, cell line, review.

How to Cite

Fahmi Radityamurti, Fauzan Herdian, Tiara Bunga Mayang Permata, Handoko Handoko, Henry Kodrat, Endang Nuryadi, Heri Wibowo, & Soehartati A Gondhowiardjo. (2020). Vitamin D as Radiosensitizer: A Review in Cell Line. Journal of Pharmacy and Nutrition Sciences, 10(6), 315–324. https://doi.org/10.29169/1927-5951.2020.10.06.1

Abstract

Introduction: Vitamin D has been shown to have anti-cancer properties such as antioxidants, anti-proliferative, and cell differentiation. The property of vitamin D as an anticancer agent triggers researchers to find out whether vitamin D is useful as a radiosensitizer. Multiple studies have been carried out on cell lines in various types of cancer, but the benefits of vitamin D as a radiosensitizer still controversial. This paperwork aims to investigate the utilization of Vitamin D3 (Calcitriol) as radiosensitizer in various cell line through literature review.
Methods: A systematic search of available medical literature databases was performed on in-vitro studies with Vitamin D as a radiosensitizer in all types of cell lines. A total of 11 in-vitro studies were evaluated.
Results: Nine studies in this review showed a significant effect of Vitamin D as a radiosensitizer agent by promoting cytotoxic autophagy, increasing apoptosis, inhibiting of cell survival and proliferation, promoting gene in ReIB inhibition, inducing senescene and necrosis. The two remaining studies showed no significant effect in the radiosensitizing mechanism of Vitamin D due to lack of evidence in-vitro settings.
Conclusion: Vitamin D have anticancer property and can be used as a radiosensitizer by imploring various mechanism pathways in various cell lines. Further research especially in-vivo settings need to be evaluated.

https://doi.org/10.29169/1927-5951.2020.10.06.1
PDF

References

International Agency for Research on Cancer. Vitamin D and cancer. International Agency for Research on Cancer 2008. 449.

Bouillon R, de Groot L, Jameson J. Vitamin D: From Photosynthesis, Metabolism, and Action to Clinical Applications. New South Wales, Australia: Saunders 2001. 1010-1028.

Feldman D, Leeuwen J, Munoz A, Driel M. Vitamin D. 4th ed. Vol. 2. Elsevier 2018.

Dunlap N, Schwartz GG, Eads D, Cramer SD, Sherk AB, John V, et al. 1?,25-Dihydroxyvitamin D3 (calcitriol) and its analogue, 19-nor-1?,25(OH)2D2, potentiate the effects of ionising radiation on human prostate cancer cells. British Journal of Cancer 2003; 89(4). https://doi.org/10.1038/sj.bjc.6601161

Polar MK, Gennings C, Park M, Gupta MS, Gewirtz DA. Effect of the vitamin D3 analog ILX 23-7553 on apoptosis and sensitivity to fractionated radiation in breast tumor cells and normal human fibroblasts. Cancer Chemotherapy and Pharmacology 2003; 51(5). https://doi.org/10.1007/s00280-003-0606-z

Xu Y, Fang F, st. Clair DK, Josson S, Sompol P, Spasojevic I, et al. Suppression of RelB-mediated manganese superoxide dismutase expression reveals a primary mechanism for radiosensitization effect of 1 ,25-dihydroxyvitamin D3 in prostate cancer cells. Molecular Cancer Therapeutics 2007; 6(7). https://doi.org/10.1158/1535-7163.MCT-06-0700

Chaudhry M, Sundaram S, Gennings C, Carter H, Gewirtz DA. The vitamin D 3 analog, ILX-23-7553, enhances the response to Adriamycin and irradiation in MCF-7 breast tumor cells. Cancer Chemotherapy and Pharmacology 2001; 47(5). https://doi.org/10.1007/s002800000251

Wilson EN, Bristol ML, Di X, Maltese WA, Koterba K, Beckman MJ, et al. A Switch Between Cytoprotective and Cytotoxic Autophagy in the Radiosensitization of Breast Tumor Cells by Chloroquine and Vitamin D. Hormones and Cancer 2011; 2(5). https://doi.org/10.1007/s12672-011-0081-7

Sharma K, Goehe RW, Di X, Hicks MA, Torti S v, Torti FM, et al. A novel cytostatic form of autophagy in sensitization of non-small cell lung cancer cells to radiation by vitamin D and the vitamin D analog, EB 1089. Autophagy 2014; 10(12). https://doi.org/10.4161/15548627.2014.993283

Mineva ND, Wang X, Yang S, Ying H, Xiao Z-XJ, Holick MF, et al. Inhibition of RelB by 1,25-dihydroxyvitamin D 3 promotes sensitivity of breast cancer cells to radiation. Journal of Cellular Physiology 2009; 220(3). https://doi.org/10.1002/jcp.21765

Podgorska E, Drzal A, Matuszak Z, Swakon J, Slominski A, Elas M, et al. Calcitriol and Calcidiol Can Sensitize Melanoma Cells to Low-LET Proton Beam Irradiation. International Journal of Molecular Sciences 2018; 19(8). https://doi.org/10.3390/ijms19082236

Bristol ML, Di X, Beckman MJ, Wilson EN, Henderson SC, Maiti A, et al. Dual functions of autophagy in the response of breast tumor cells to radiation. Autophagy 2012; 8(5). https://doi.org/10.4161/auto.19313

DeMasters G, Di X, Newsham I, Shiu R, Gewirtz DA. Potentiation of radiation sensitivity in breast tumor cells by the vitamin D3 analogue, EB 1089, through promotion of autophagy and interference with proliferative recovery. Molecular Cancer Therapeutics 2006; 5(11). https://doi.org/10.1158/1535-7163.MCT-06-0316

Gavrilov V, Leibovich Y, Ariad S, Lavrenkov K, Shany S. A combined pretreatment of 1,25-dihydroxyvitamin D3 and sodium valproate enhances the damaging effect of ionizing radiation on prostate cancer cells. The Journal of Steroid Biochemistry and Molecular Biology 2010; 121(1-2). https://doi.org/10.1016/j.jsbmb.2010.03.004

Chen N, Karantza-Wadsworth V. Role and regulation of autophagy in cancer. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 2009; 1793(9). https://doi.org/10.1016/j.bbamcr.2008.12.013

Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nature Reviews Molecular Cell Biology 2007; 8(9). https://doi.org/10.1038/nrm2239

Gorka M, Daniewski WM, Gajkowska B, Lusakowska E, Godlewski MM, Motyl T. Autophagy is the dominant type of programmed cell death in breast cancer MCF-7 cells exposed to AGS 115 and EFDAC, new sesquiterpene analogs of paclitaxel. Anti-Cancer Drugs 2005; 16(7). https://doi.org/10.1097/01.cad.0000171514.50310.85

Kirisako T, Ichimura Y, Okada H, Kabeya Y, Mizushima N, Yoshimori T, et al. The Reversible Modification Regulates the Membrane-Binding State of Apg8/Aut7 Essential for Autophagy and the Cytoplasm to Vacuole Targeting Pathway. Journal of Cell Biology 2000; 151(2). https://doi.org/10.1083/jcb.151.2.263

Nedelsky NB, Todd PK, Taylor JP. Autophagy and the ubiquitin-proteasome system: Collaborators in neuroprotection. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 2008; 1782(12). https://doi.org/10.1016/j.bbadis.2008.10.002

Ghosh S, May MJ, Kopp EB. NF-?B and rel proteins: Evolutionarily Conserved Mediators of Immune Responses. Annual Review of Immunology 1998; 16(1). https://doi.org/10.1146/annurev.immunol.16.1.225

Sonenshein GE. Rel/NF-?B transcription factors and the control of apoptosis. Seminars in Cancer Biology 1997; 8(2). https://doi.org/10.1006/scbi.1997.0062

Karin M. Nuclear factor-?B in cancer development and progression. Nature 2006; 441(7092). https://doi.org/10.1038/nature04870

Magné N, Toillon R-A, Bottero V, Didelot C, Houtte P van, Gérard J-P, et al. NF-?B modulation and ionizing radiation: mechanisms and future directions for cancer treatment. Cancer Letters 2006; 231(2). https://doi.org/10.1016/j.canlet.2005.01.022

Xu Y, Kiningham KK, Devalaraja MN, Yeh C-C, Majima H, Kasarskis EJ, et al. An Intronic NF-kappaB Element Is Essential for Induction of the Human Manganese Superoxide Dismutase Gene by Tumor Necrosis Factor-alpha and Interleukin-1beta. DNA and Cell Biology 1999; 18(9). https://doi.org/10.1089/104454999314999

Dong X, Craig T, Xing N, Bachman LA, Paya C v., Weih F, et al. Direct Transcriptional Regulation of RelB by 1?,25-Dihydroxyvitamin D3 and Its Analogs. Journal of Biological Chemistry 2003; 278(49). https://doi.org/10.1074/jbc.M308448200

Dong X, Lutz W, Schroeder TM, Bachman LA, Westendorf JJ, Kumar R, et al. Regulation of relB in dendritic cells by means of modulated association of vitamin D receptor and histone deacetylase 3 with the promoter. Proceedings of the National Academy of Sciences 2005; 102(44). https://doi.org/10.1073/pnas.0506516102

Falck J, Mailand N, Syljuåsen RG, Bartek J, Lukas J. The ATM-Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature 2001; 410(6830). https://doi.org/10.1038/35071124

Debacq-Chainiaux F, Erusalimsky JD, Campisi J, Toussaint O. Protocols to detect senescence-associated beta-galactosidase (SA-?gal) activity, a biomarker of senescent cells in culture and in vivo. Nature Protocols 2009; 4(12). https://doi.org/10.1038/nprot.2009.191

Lambert JR, Kelly JA, Shim M, Huffer WE, Nordeen SK, Baek SJ, et al. Prostate derived factor in human prostate cancer cells: Gene induction by vitamin D via a p53-dependent mechanism and inhibition of prostate cancer cell growth. Journal of Cellular Physiology 2006; 208(3). https://doi.org/10.1002/jcp.20692

Kovalenko PL, Zhang Z, Cui M, Clinton SK, Fleet JC. 1,25 dihydroxyvitamin D-mediated orchestration of anticancer, transcript-level effects in the immortalized, non-transformed prostate epithelial cell line, RWPE1. BMC Genomics 2010; 11(1). https://doi.org/10.1186/1471-2164-11-26

Bao B-Y, Ting H-J, Hsu J-W, Lee Y-F. Protective role of 1?, 25-dihydroxyvitamin D3 against oxidative stress in nonmalignant human prostate epithelial cells. International Journal of Cancer 2008; 122(12). https://doi.org/10.1002/ijc.23460

Chandel NS, Tuveson DA. The Promise and Perils of Antioxidants for Cancer Patients. New England Journal of Medicine 2014; 371(2). https://doi.org/10.1056/NEJMcibr1405701

Yasueda A, Urushima H, Ito T. Efficacy and Interaction of Antioxidant Supplements as Adjuvant Therapy in Cancer Treatment. Integrative Cancer Therapies 2016; 15(1). https://doi.org/10.1177/1534735415610427

Meyer F, Bairati I, Fortin A, Gélinas M, Nabid A, Brochet F, et al. Interaction between antioxidant vitamin supplementation and cigarette smoking during radiation therapy in relation to long-term effects on recurrence and mortality: A randomized trial among head and neck cancer patients. International Journal of Cancer 2007; 122(7). https://doi.org/10.1002/ijc.23200

Moss RW. Do Antioxidants Interfere With Radiation Therapy for Cancer? Integrative Cancer Therapies 2007; 6(3). https://doi.org/10.1177/1534735407305655

Wilson MK, Baguley BC, Wall C, Jameson MB, Findlay MP. Review of high-dose intravenous vitamin C as an anticancer agent. Asia-Pacific Journal of Clinical Oncology 2014; 10(1). https://doi.org/10.1111/ajco.12173

Cain L, Flynn J, Kelly D. Effect of complementary alternative medical (CAM) therapy on tumor response, control and recurrence in prostate cancer patients (PCpts) treated with radiation therapy (RT). Journal of Clinical Oncology 2007; 25: 15585-undefined. https://doi.org/10.1200/jco.2007.25.18_suppl.15585

Jaakkola K, Lähteenmäki P, Laakso J, Harju E, Tykkä H, Mahlberg K. Treatment with antioxidant and other nutrients in combination with chemotherapy and irradiation in patients with small-cell lung cancer. Anticancer Research 1992; 12: 599-606.

Creative Commons License

This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

Copyright (c) 2020 Journal of Pharmacy and Nutrition Sciences