The formulation for cancer prevention & therapy

Abstract

Our natural product research over nine years has been to discover effective over-the-counter-supplements, designed to facilitate classical treatments in fighting disease, specifically cancer. These supplements are important in antiangiogenic, immune and antioxidant defense systems. The components include Acetyl L-Carnitine (ALC), Alpha Lipoic Acid (ALA), Coenzyme Q10 (CoQ10), Curcumin with Piperine, Genistein, Lentinan, N-Acetylcysteine (NAC), Resveratrol, selenium, Vitamin B Complex, Vitamin C, Vitamin E and zinc. These supplement components are supported for human cancer by over 15,000 references in the scientific literature withover of these being published clinical trials, not including zinc and the vitamins. These chemical defined components from natural origins have demonstrated, either individually or collectively, to have antioxidant, anti-angiogenesis, and immune stimulation properties. Furthermore, the direct cancer cell cytotoxicity for Curcumin, Genistein and NAC have been shown. Some of the antiangiogenesis components that affect the majority if not all pathways of angiogenesis, such as Curcumin, Genistein and NAC, actually stimulate the in vivo production of natural antiangiogenic compounds that include Angiostatin, Endostatin and Thrombospotin 1. All of the components play a role in serving as either water or lipid soluble (able to cross the blood-brain barrier) antioxidants. Curcumin, Genistein, Resveratrol, Lentinan, NAC, zinc, selenium and vitamins B and C all stimulate the immune system. Except for ALC and CoQ10, the other components show anti-inflammatory activity. ALC, Resveratrol along with the B and C vitamins are helpful in treating fatigue. The components that help protect the brain and promote nerve regeneration include ALA, CoQ10, Resveratrol, NAC, selenium, zinc and the B and C vitamins. In conclusion, effective prevention and treatment for diseases such as cancer, heart disease and immune deficiency will require multiple compounds.

Share and Cite:

Thornthwaite, J. , Shah, H. , Shah, P. , Peeples, W. and Respess, H. (2013) The formulation for cancer prevention & therapy. Advances in Biological Chemistry, 3, 356-387. doi: 10.4236/abc.2013.33040.

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] Pietras, R.J. and Weinberg, O.K. (2005) Antiangiogenic steroids in human cancer therapy. Evidence-Based Complementary and Alternative Medicine, 2, 49-57. doi:10.1093/ecam/neh066
[2] Tascilar, M., et al. (2006) Complementary and alternative medicine during cancer treatment: Beyond innocence. The Oncologist, 11, 732-741. doi:10.1634/theoncologist.11-7-732
[3] Hagerty, R.G., et al. (2005) Communicating with realism and hope: Incurable cancer patients’ views on the disclosure of prognosis. Journal of Clinical Oncology, 23, 1278-1288.
[4] Surh, Y.J. and Chun, K.S. (2007) Cancer chemopreventive effects of curcumin. Advances in Experimental Medicine and Biology, 595, 149-172. doi:10.1007/978-0-387-46401-5_5
[5] Chainani-Wu, N. (2003) Safety and anti-inflammatory activity of curcumin: A component of tumeric (Curcuma longa). The Journal of Alternative and Complementary Medicine, 9, 161-168. doi:10.1089/107555303321223035
[6] Duvoix, A., et al. (2005) Chemopreventive and therapeutic effects of curcumin. Cancer Letters, 223, 181-190. doi:10.1016/j.canlet.2004.09.041
[7] Hsu, C.H. and Cheng, A.L. (2007) Clinical studies with curcumin. Advances in Experimental Medicine and Biology, 595, 471-480. doi:10.1007/978-0-387-46401-5_21
[8] Bisht, M., Bist, S.S. and Dhasmana, D.C. (2010) Biological response modifiers: Current use and future prospects in cancer therapy. Indian Journal of Cancer, 47, 443-451. doi:10.4103/0019-509X.73559
[9] Johnson, J.J. and Mukhtar, H. (2007) Curcumin for chemoprevention of colon cancer. Cancer Letters, 255, 170181. doi:10.1016/j.canlet.2007.03.005
[10] Ferguson, L.R. and Philpott, M. (2007) Cancer prevention by dietary bioactive components that target the immune response. Current Cancer Drug Targets, 7, 459-464.
[11] Singh, S. and Khar, A. (2006) Biological effects of curcumin and its role in cancer chemoprevention and therapy. Anti-Cancer Agents in Medicinal Chemistry, 6, 259-270.
[12] Jagetia, G.C. and Aggarwal, B.B. (2007) “Spicing up” of the immune system by curcumin. Journal of Clinical Immunology, 27, 19-35. doi:10.1007/s10875-006-9066-7
[13] Sharma, R.A., Gescher, A.J. and Steward, W.P. (2005) Curcumin: The story so far. European Journal of Cancer, 41, 1955-1968. doi:10.1016/j.ejca.2005.05.009
[14] John, V.D., Kuttan, G. and Krishnankutty, K. (2002) Antitumour studies of metal chelates of synthetic curcumanoids. Journal of Experimental & Clinical Cancer Research, 21, 219-224.
[15] Hadi, S.M., et al. (2000) Putative mechanism for anticancer and apoptosis-inducing properties of plant-derived polyphenolic compounds. IUBMB Life, 50, 167-171.
[16] Yoshino, M., et al. (2004) Prooxidant activity of curcumin: Copper-dependent formation of 8-hydroxy-2’-deoxyguanosine in DNA and induction of apoptotic cell death. Toxicology in Vitro, 18, 783-789. doi:10.1016/j.tiv.2004.03.009
[17] Thompson, K.H., et al. (2004) Complementary inhibition of synoviocyte, smooth muscle cell or mouse lymphoma cell proliferation by a vanadyl curcumin complex compared to curcumin alone. Journal of Inorganic Biochemistry, 98, 2063-2070. doi:10.1016/j.jinorgbio.2004.09.011
[18] Garcea, G., et al. (2005) Consumption of the putative chemopreventive agent curcumin by cancer patients: Assessment of curcumin levels in the colorectum and their pharmacodynamic consequences. Cancer Epidemiology Biomarkers & Prevention, 14,120-125.
[19] Adams, B.K., et al. (2004) Synthesis and biological evaluation of novel curcumin analogs as anti-cancer and anti-angiogenesis agents. Bioorganic & Medicinal Chemistry, 12, 3871-3883. doi:10.1016/j.bmc.2004.05.006
[20] Sun, C.Y., et al. (2004) Experimental study on anticancer effect of curcumin on Raji cells in Vitro. Zhongguo Zhong Xi Yi Jie He Za Zhi, 24, 1003-1006.
[21] Shao, Z.M., et al. (2002) Curcumin exerts multiple suppressive effects on human breast carcinoma cells. International Journal of Cancer, 98, 234-240. doi:10.1002/ijc.10183
[22] Kim, J.H., et al. (2002) Microarray-based analysis of anti-angiogenic activity of demethoxycurcumin on human umbilical vein endothelial cells: Crucial involvement of the down-regulation of matrix metalloproteinase. Cancer Science, 93, 1378-1385. doi:10.1111/j.1349-7006.2002.tb01247.x
[23] Bemis, D.L., Katz, A.E. and Buttyan, R. (2006) Clinical trials of natural products as chemopreventive agents for prostate cancer. Expert Opinion on Investigational Drugs, 15, 1191-1200. doi:10.1517/13543784.15.10.1191
[24] Campbell, F.C. and Collett, G.P. (2005) Chemopreventive properties of curcumin. Future Oncology, 1, 405-414. doi:10.1517/14796694.1.3.405
[25] Shanmugam, M.K., Kannaiyan, R. and Sethi, G. (2011) Targeting cell signaling and apoptotic pathways by dietary agents: Role in the prevention and treatment of cancer. Nutrition and Cancer, 63, 161-173. doi:10.1080/01635581.2011.523502
[26] Su, S.J., et al., The novel targets for anti-angiogenesis of genistein on human cancer cells. Biochemical Pharmacology, 69, 307-318. doi:10.1016/j.bcp.2004.09.025
[27] Ravindranath, M.H., et al. (2004) Anticancer therapeutic potential of soy isoflavone, genistein. Advances in Experimental Medicine and Biology, 546, 121-165.
[28] Kyle, E., et al. (1997) Genistein-induced apoptosis of prostate cancer cells is preceded by a specific decrease in focal adhesion kinase activity. Molecular Pharmacology, 51, 193-200.
[29] Lazarevic, B., et al. (2012) The effects of short-term genistein intervention on prostate biomarker expression in patients with localised prostate cancer before radical prostatectomy.The British Journal of Nutrition, 108, 1-10.
[30] Lazarevic, B., et al. (2011) Efficacy and safety of shortterm genistein intervention in patients with localized prostate cancer prior to radical prostatectomy: A randomized, placebo-controlled, double-blind phase 2 clinical trial. Nutrition and Cancer, 63, 889-898. doi:10.1080/01635581.2011.582221
[31] Sasamura, H., et al. (2004) Antiproliferative and antiangiogenic activities of genistein in human renal cell carcinoma. Urology, 64, 389-393. doi:10.1016/j.urology.2004.03.045
[32] Hillman, G.G., et al. (2001) Genistein potentiates the radiation effect on prostate carcinoma cells. Clinical Cancer Research, 7, 382-390.
[33] de la Taille, A., et al. (2001) Cancer of the prostate: Influence of nutritional factors. A new nutritional approach. La Presse Médicale, 30, 561-564.
[34] Ghafar, M.A., et al. (2002) Regression of prostate cancer following administration of genistein combined polysaccharide (GCP), a nutritional supplement: A case report. The Journal of Alternative and Complementary Medicine, 8, 493-497. doi:10.1089/107555302760253694
[35] Davis, J.N., et al. (2000) Inhibition of prostate specific antigen expression by genistein in prostate cancer cells. International Journal of Oncology, 16, 1091-1097.
[36] Davis, J.N., Kucuk, O. and Sarkar, F.H. (2002) Expression of prostate-specific antigen is transcriptionally regulated by genistein in prostate cancer cells. Molecular Carcinogenesis, 34, 91-101. doi:10.1002/mc.10053
[37] Shen, J.C., et al. (2000) Low-dose genistein induces cyclin-dependent kinase inhibitors and G1 cell-cycle arrest in human prostate cancer cells. Molecular Carcinogenesis, 29, 92-102. doi:10.1002/1098-2744(200010)29:2<92::AID-MC6>3.0.CO;2-Q
[38] Piao, M., et al. (2006) Inhibition of endothelial cell proliferation, in Vitro angiogenesis, and the down-regulation of cell adhesion-related genes by genistein. Combined with a cDNA microarray analysis. Endothelium, 13, 249266. doi:10.1080/10623320600903940
[39] Shichinohe, T., et al. (2001) Development of lentiviral vectors for antiangiogenic gene delivery. Cancer Gene Therapy, 8, 879-889. doi:10.1038/sj.cgt.7700388
[40] Shao, Z.M., et al. (1998) Genistein exerts multiple suppressive effects on human breast carcinoma cells. Cancer Research, 58, 4851-4857.
[41] Li, Y. and Sarkar, F.H. (2002) Gene expression profiles of genistein-treated PC3 prostate cancer cells. The Journal of Nutrition, 132, 3623-3631.
[42] Li, Y. and Sarkar, F.H. (2002) Down-regulation of invasion and angiogenesis-related genes identified by cDNA microarray analysis of PC3 prostate cancer cells treated with genistein. Cancer Letters, 186, 157-164. doi:10.1016/S0304-3835(02)00349-X
[43] Sarkar, F.H. and Li, Y. (2002) Mechanisms of cancer chemoprevention by soy isoflavone genistein. Cancer and Metastasis Reviews, 21, 265-280. doi:10.1023/A:1021210910821
[44] Konstantakopoulos, N., et al. (2006) Changes in gene expressions elicited by physiological concentrations of genistein on human endometrial cancer cells. Molecular Carcinogenesis, 45, 752-763. doi:10.1002/mc.20187
[45] Khoshyomn, S., et al. (2000) Synergistic action of genistein and cisplatin on growth inhibition and cytotoxicity of human medulloblastoma cells. Pediatric Neurosurgery, 33, 123-131. doi:10.1159/000028993
[46] Raynal, N.J., et al. (2008) Antileukemic activity of genistein, a major isoflavone present in soy products. Journal of Natural Products, 71, 3-7. doi:10.1021/np070230s
[47] Banerjee, S., et al. (2007) In Vitro and in Vivo molecular evidence of genistein action in augmenting the efficacy of cisplatin in pancreatic cancer. International Journal of Cancer, 120, 906-917. doi:10.1002/ijc.22332
[48] Dijkstra, S.C., et al. (2010) Biomarkers of dietary exposure are associated with lower risk of breast fibroadenomas in Chinese women. The Journal of Nutrition, 140, 1302-1310. doi:10.3945/jn.109.119727
[49] Vauzour, D., et al. (2007) Inhibition of cellular proliferation by the genistein metabolite 5,7,3',4'-tetrahydroxylisoflavone is mediated by DNA damage and activation of the ATR signalling pathway. Archives of Biochemistry and Biophysics, 468, 159-166. doi:10.1016/j.abb.2007.09.021
[50] Meeran, S.M. and Katiyar, S.K. (2008) Cell cycle control as a basis for cancer chemopre-vention through dietary agents. Frontiers in Bioscience, 13, 2191-2202. doi:10.2741/2834
[51] Ohigashi, H. and Murakami, A. (2004) Cancer prevention with food factors: Alone and in combination. Biofactors, 22, 49-55. doi:10.1002/biof.5520220109
[52] Ramos, S. (2007) Effects of dietary flavonoids on apoptotic pathways related to cancer chemoprevention. The Journal of Nutritional Biochemistry, 18, 427-442. doi:10.1016/j.jnutbio.2006.11.004
[53] Sarkar, F.H., et al. (2006) The role of genistein and synthetic derivatives of isoflavone in cancer prevention and therapy. Mini Reviews in Medicinal Chemistry, 6, 401407.
[54] Perabo, F.G., et al. (2008) Soy isoflavone genistein in prevention and treatment of prostate cancer. Prostate Cancer and Prostatic Diseases, 11, 6-12. doi:10.1038/sj.pcan.4501000
[55] Nagata, Y., et al. (2007) Dietary isoflavones may protect against prostate cancer in Japanese men. The Journal of Nutrition, 137, 1974-1979.
[56] Heald, C.L., et al. (2006) Phyto-oestrogen intake in Scottish men: Use of serum to validate a self-administered food-frequency questionnaire in older men. European Journal of Clinical Nutrition, 60, 129-135. doi:10.1038/sj.ejcn.1602277
[57] Kurahashi, N., et al. (2007) Soy product and isoflavone consumption in relation to prostate cancer in Japanese men. Cancer Epidemiology, Biomarkers & Prevention, 16, 538-545. doi:10.1158/1055-9965.EPI-06-0517
[58] Ko, K.P., et al. (2010) Isoflavones from phytoestrogens and gastric cancer risk: A nested case-control study within the Korean Multicenter Cancer Cohort. Cancer Epidemiology, Biomarkers & Preven-tion, 19, 1292-1300. doi:10.1158/1055-9965.EPI-09-1004
[59] Cho, J. and Kim, Y. (2002) Sharks: A potential source of antiangiogenic factors and tumor treatments. Marine Biotechnology, 4, 521-525. doi:10.1007/s10126-002-0064-3
[60] Williams, J.I., et al. (2001) Squalamine treatment of human tumors in nu/nu mice enhances platinum-based chemotherapies. Clinical Cancer Research, 7, 724-733.
[61] Bhargava, P., et al. (2001) A phase I and pharmacokinetic study of squalamine, a novel antiangiogenic agent, in patients with advanced cancers. Clinical Cancer Research, 7, 3912-3919.
[62] Herbst, R.S., et al. (2003) A phase I/IIA trial of continuous five-day infusion of squalamine lactate (MSI-1256F) plus carboplatin and paclitaxel in patients with advanced non-small cell lung cancer. Clinical Cancer Research, 9, 4108-4115.
[63] Hao, D., et al. (2003) A Phase I and pharmacokinetic study of squalamine, an aminosterol angiogenesis inhibittor. Clinical Cancer Research, 9, 2465-2471.
[64] Bayes, M., Rabasseda, X. and Prous, J.R. (2005) Gateways to clinical trials. Methods and Findings in Experimental and Clinical Pharmacology, 27, 331-372.
[65] Emerson, M.V. and Lauer, A.K. (2007) Emerging therapies for the treatment of neovascular age-related macular degeneration and diabetic macular edema. BioDrugs, 21, 245-257. doi:10.2165/00063030-200721040-00005
[66] Connolly, B., et al. (2006) Squalamine lactate for exudative age-related macular degeneration. Ophthalmology Clinics of North America, 19, 381-391.
[67] Michels, S., Schmidt-Erfurth, U. and Rosenfeld, P.J. (2006) Promising new treatments for neovascular age-related macular degeneration. Expert Opinion on Investigational Drugs, 15, 779-793. doi:10.1517/13543784.15.7.779
[68] Shepherd, F.A. and Sridhar, S.S. (2003) Angiogenesis inhibitors under study for the treatment of lung cancer. Lung Cancer, 41, 63-72. doi:10.1016/S0169-5002(03)00144-2
[69] Li, D., Williams, J.I. and Pietras, R.J. (2002) Squalamine and cisplatin block angiogenesis and growth of human ovarian cancer cells with or without HER-2 gene overexpression. Oncogene, 21, 2805-2814. doi:10.1038/sj.onc.1205410
[70] Akhter, S., et al. (1999) Squalamine, a novel cationic steroid, specifically inhibits the brush-border Na+/H+ exchanger isoform NHE3. American Journal of Physiology -Cell Physiology, 276, C136-C144.
[71] Teicher, B.A., et al. (1998) Potential of the aminosterol, squalamine in combination therapy in the rat 13,762 mammary carcinoma and the murine Lewis lung carcinoma. Anticancer Research, 18, 2567-2573.
[72] Paley, P.J., et al. (1997) Vascular endothelial growth factor expression in early stage ovarian carcinoma. Cancer, 80, 98-106. doi:10.1002/(SICI)1097-0142(19970701)80:1<98::AID-CNCR13>3.0.CO;2-A
[73] Yamamoto, S., et al. (1997) Expression of vascular endothelial growth factor (VEGF) in epithelial ovarian neoplasms: Correlation with clinicopathology and patient survival, and analysis of serum VEGF levels. British Journal of Cancer, 76, 1221-1227. doi:10.1038/bjc.1997.537
[74] Alvarez, A.A., et al. (1999) The prognostic significance of angiogenesis in epithelial ovarian carcinoma. Clinical Cancer Research, 5, 587-591.
[75] Brunel, J.M., et al. (2005) Squalamine: A polyvalent drug of the future? Current Cancer Drug Targets, 5, 267-272.
[76] Havre, P.A., et al. (2002) Transformed and tumor-derived human cells exhibit preferential sensitivity to the thiol antioxidants, N-acetyl cysteine and penicillamine. Cancer Research, 62, 1443-1449.
[77] Alonso, A., et al. (2004) Prevention of radiocontrast nephropathy with N-acetylcysteine in patients with chronic kidney disease: A meta-analysis of randomized, controlled trials. American Journal of Kidney Diseases, 43, 1-9.
[78] Agarwal, A., et al. (2004) N-acetyl-cysteine promotes angiostatin production and vascular collapse in an orthotopic model of breast cancer. The American Journal of Pathology, 164, 1683-1696. doi:10.1016/S0002-9440(10)63727-3
[79] Albini, A., et al. (2001) Inhibition of angiogenesis-driven Kaposi’s sarcoma tumor growth in nude mice by oral N-acetylcysteine. Cancer Research, 61, 8171-8178.
[80] Choi, C.H., et al. (2007) Phase II study of neoadjuvant chemotherapy with mitomycin-c, vincristine and cisplatin (MVC) in patients with stages IB2-IIB cervical carcinoma. Gynecologic Oncology, 104, 64-69. doi:10.1016/j.ygyno.2006.07.006
[81] Betten, D.P., et al. (2007) A prospective evaluation of shortened course oral N-acetylcysteine for the treatment of acute acetaminophen poisoning. Annals of Emergency Medicine, 50, 272-279. doi:10.1016/j.annemergmed.2006.11.010
[82] Paterson, R.L., Galley, H.F. and Webster, N.R. (2003) The effect of N-acetylcysteine on nuclear factor-kappa B activation, interleukin-6, interleukin-8, and intercellular adhesion molecule-1 expression in patients with sepsis. Critical Care Medicine, 31, 2574-2578. doi:10.1097/01.CCM.0000089945.69588.18
[83] Uwe, S. (2008) Antiinflammatory interventions of NFkappaB signaling: Potential applications and risks. Biochemical Pharmacology, 75, 1567-1579. doi:10.1016/j.bcp.2007.10.027
[84] Vecchiarelli, A., et al. (1994) Macrophage activation by N-acetyl-cysteine in COPD patients. Chest, 105, 806-811.
[85] Mantovani, G., et al. (2003) Subcutaneous interleukin-2 in combination with medroxyprogesterone acetate and antioxidants in advanced cancer responders to previous chemotherapy: Phase II study evaluating clinical, quality of life, and laboratory parameters. Journal of Experimental Therapeutics and Oncology, 3, 205-219. doi:10.1046/j.1359-4117.2003.01096.x
[86] Pendyala, L. and Creaven, P.J. (1995) Pharmacokinetic and pharmacodynamic studies of N-acetylcysteine, a potential chemopreventive agent during a phase I trial. Cancer Epidemiology, Biomarkers & Prevention, 4, 245251.
[87] Wolchok, J.D., et al. (2003) Phase I trial of high dose paracetamol and carmustine in patients with metastatic melanoma. Melanoma Research, 13, 189-196. doi:10.1097/00008390-200304000-00013
[88] Haase, M., et al. (2007) Phase II, randomized, controlled trial of high-dose N-acetylcysteine in high-risk cardiac surgery patients. Critical Care Medicine, 35, 1324-1331.
[89] Mantovani, G., et al. (2002) Phase II study of subcutaneously administered interleukin-2 in combination with medroxyprogesterone acetate and antioxidant agents as maintenance treatment in advanced cancer responders to previous chemotherapy. Oncology Reports, 9, 887-896.
[90] Zoran, D.L., et al. (1997) Wheat bran diet reduces tumor incidence in a rat model of colon cancer independent of effects on distal luminal butyrate concentrations. The Journal of Nutrition, 127, 2217-2225.
[91] Reddy, B.S., et al. (2000) Preventive potential of wheat bran fractions against experimental colon carcinogenesis: Implications for human colon cancer prevention. Cancer Research, 60, 4792-4797.
[92] Martinez, M.E., et al. (1998) Design and baseline characteristics of study participants in the wheat bran fiber trial. Cancer Epidemiology, Biomarkers & Prevention, 7, 813-816.
[93] Compher, C.W., et al. (1999) Wheat bran decreases aberrant crypt foci, preserves normal proliferation, and increases intraluminal butyrate levels in experimental colon cancer. Journal of Parenteral Enteral Nutrition, 23, 269277.
[94] Folino, M., McIntyre, A. and Young, G.P. (1995) Dietary fibers differ in their effects on large bowel epithelial proliferation and fecal fermentation-dependent events in rats. The Journal of Nutrition, 125, 1521-1528.
[95] Lupton, J.R. and Kurtz, P.P. (1993) Relationship of colonic luminal short-chain fatty acids and pH to in Vivo cell proliferation in rats. The Journal of Nutrition, 123, 15221530.
[96] Zhang, J. and Lupton, J.R. (1994) Dietary fibers stimulate colonic cell proliferation by different mechanisms at different sites. Nutrition and Cancer, 22, 267-276. doi:10.1080/01635589409514352
[97] McIntyre, A., et al. (1991) Different fibers have different regional effects on luminal contents of rat colon. Gastroenterology, 101, 1274-1281.
[98] McIntyre, A., Gibson, P.R. and Young, G.P. (1993) Butyrate production from dietary fibre and protection against large bowel cancer in a rat model. Gut, 34, 386391. doi:10.1136/gut.34.3.386
[99] Newmark, H.L. and Lupton, J.R. (1990) Determinants and consequences of colonic luminal pH: Implications for colon cancer. Nutrition and Cancer, 14, 161-173. doi:10.1080/01635589009514091
[100] Kestell, P., et al. (1999) Studies on the mechanism of cancer protection by wheat bran: Effects on the absorption, metabolism and excretion of the food carcinogen 2-amino-3-methylimidazo[4,5-f]quinoline (IQ). Carcinogenesis, 20, 2253-2260. doi:10.1093/carcin/20.12.2253
[101] Ghoneum, M. and Matsuura, M. (2004) Augmentation of macrophage phagocytosis by modified arabinoxylan rice bran (MGN-3/biobran). International Journal of Immunopathology and Pharmacology, 17, 283-292.
[102] Chang, H., et al. (2004) Prognostic relevance of immunophenotyping in 379 patients with acute myeloid leukemia. Leukemia Research, 28, 43-48. doi:10.1016/S0145-2126(03)00180-2
[103] Lee, C.Y. and Wan, J.M. (2002) Immunoregulatory and antioxidant performance of alpha-tocopherol and selenium on human lymphocytes. Biological Trace Element Research, 86, 123-136. doi:10.1385/BTER:86:2:123
[104] Muralikrishna, G. and Rao, M.V. (2007) Cereal noncellulosic polysaccharides: Structure and function relationship—An overview. Critical Reviews in Food Science and Nutrition, 47, 599-610. doi:10.1080/10408390600919056
[105] Glei, M., et al. (2006) Both wheat (Triticum aestivum) bran arabinoxylans and gut flora-mediated fermentation products protect human colon cells from genotoxic activities of 4-hydroxynonenal and hydrogen peroxide. Journal of Agricultural and Food Chemistry, 54, 20882095. doi:10.1021/jf052768e
[106] Ferreira, I.C., et al. (2010) Compounds from wild mushrooms with antitumor potential. Anti-Cancer Agents in Medicinal Chemistry, 10, 424-436.
[107] Wasser, S.P. and Weis, A.L. (1999) Therapeutic effects of substances occurring in higher basidiomycetes mushrooms: A modern perspective. Critical Reviews in Immunology, 19, 65-96.
[108] Chihara, G. (1992) Recent progress in immunopharmacology and therapeutic effects of polysaccha-rides. Developments in Biological Standardization, 77, 191-197.
[109] Ooi, V.E. and Liu, F. (2000) Immunomodulation and anti-cancer activity of polysaccharide-protein complexes. Current Medicinal Chemistry, 7, 715-729.
[110] Borchers, A.T., et al. (1999) Mushrooms, tumors, and immunity. Proceedings of the Society for Experimental Biology and Medicine, 221, 281-293. doi:10.1046/j.1525-1373.1999.d01-86.x
[111] Jong, S.C., Birmingham, J.M. and Pai, S.H. (1991) Immunomodulatory substances of fungal origin. Journal of Immunology and Immunopharmacology, 9, 115-122.
[112] Hobbs, C.R. (2000) Medicinal value of Lentinus edodes (Berk.) Sing. (Agaricomy-cetideae). A literature review. International Journal of Medicinal Mushrooms, 2, 287302.
[113] Taguchi, T., et al. (1985) Results of phase III study of lentinan. Gan to Kagaku Ryoho, 12, 366-378.
[114] Oba, K., et al. (2009) Individual patient based metaanalysis of lentinan for unresectable/recurrent gastric cancer. Anticancer Research, 29, 2739-2745.
[115] Hazama, S., et al. (2009) Efficacy of orally administered superfine dispersed lentinan (beta-1,3-glucan) for the treatment of advanced colorectal cancer. Anticancer Research, 29, 2611-2617.
[116] Ross, G.D., et al. (1999) Therapeutic intervention with complement and beta-glucan in cancer. Immunopharmacology, 42, 61-74. doi:10.1016/S0162-3109(99)00013-2
[117] Kerekgyarto, C., et al. (1996) Strain differences in the cytotoxic activity and TNF production of murine macrophages stimulated by lentinan. International Journal of Immunopharmacology, 18, 347-353. doi:10.1016/S0192-0561(96)00038-0
[118] Yoshino, S., et al. (2010) Improvement of QOL and prognosis by treatment of superfine dispersed lentinan in patients with advanced gastric cancer. Hepatogastroenterology, 57, 172-177.
[119] Kataoka, H., et al. (2009) Lentinan with S-1 and paclitaxel for gastric cancer chemotherapy improve patient quality of life. Hepatogastroenterology, 56, 547-550.
[120] Wang, J.L., et al. (2012) Combination therapy with lentinan improves outcomes in patients with esophageal carcinoma. Molecular Medicine Report, 5, 745-748.
[121] Vanotti, A., et al. (2007) Overview on pathophysiology and newer approaches to treatment of peripheral neuropathies. CNS Drugs, 21, 3-12. doi:10.2165/00023210-200721001-00002
[122] D. De Grandis, (2007) Acetyl-l-carnitine for the treatment of chemotherapy-induced peripheral neuropathy: A short review. CNS Drugs, 21, 39-43. doi:10.2165/00023210-200721001-00006
[123] Youle, M. (2007) Acetyl-l-carnitine in HIV-associated antiretroviral toxic neuropathy. CNS Drugs, 21, 25-30. doi:10.2165/00023210-200721001-00004
[124] Sima, A.A. (2007) Acetyl-l-carnitine in diabetic polyneuropathy: Experimental and clinical data. CNS Drugs, 21, 13-23. doi:10.2165/00023210-200721001-00003
[125] Youle, M. and Osio, M. (2007) A double-blind, parallel-group, placebo-controlled, multicentre study of acetyll-carnitine in the symptomatic treatment of antiretroviral toxic neuropathy in patients with HIV-1 infection. HIV Medicine, 8, 241-250. doi:10.1111/j.1468-1293.2007.00467.x
[126] Murosaki, S., et al. (2007) A combination of caffeine, arginine, soy isoflavones, and L-carnitine enhances both lipolysis and fatty acid oxidation in 3T3-L1 and HepG2 cells in Vitro and in KK mice in Vivo. The Journal of Nutrition, 137, 2252-2257.
[127] Carroll, J.K., et al. (2007) Pharmacologic treatment of cancer-related fatigue. The Oncologist, 12, 43-51. doi:10.1634/theoncologist.12-S1-43
[128] Gramignano, G., et al. (2006) Efficacy of l-carnitine administration on fatigue, nutritional status, oxidative stress, and related quality of life in 12 advanced cancer patients undergoing anticancer therapy. Nutrition, 22, 136-145. doi:10.1016/j.nut.2005.06.003
[129] Cruciani, R.A., et al. (2006) Safety, tolerability and symptom outcomes associated with L-carnitine supplementation in patients with cancer, fatigue, and carnitine deficiency: A phase I/II study. Journal of Pain and Symptom Management, 32, 551-559. doi:10.1016/j.jpainsymman.2006.09.001
[130] Sood, A., et al. (2007) A critical review of complementtary therapies for cancer-related fatigue. Integrative Cancer Therapies, 6, 8-13. doi:10.1177/1534735406298143
[131] Delaney, C.E., Hopkins, S.P. and Addison, C.L. (2007) Supplementation with l-carnitine does not reduce the efficacy of epirubicin treatment in breast cancer cells. Cancer Letters, 252, 195-207. doi:10.1016/j.canlet.2006.12.027
[132] Dong, M.H. and Kaunitz, J.D. (2006) Gastroduodenal mucosal defense. Current Opinion in Gastroenterology, 22, 599-606. doi:10.1097/01.mog.0000245540.87784.75
[133] Lebrun, C., et al. (2006) Levocarnitine administration in multiple sclerosis patients with immunosuppressive therapy-induced fatigue. Multiple Sclerosis Journals, 12, 321-324. doi:10.1191/135248506ms1275oa
[134] Mantovani, G., et al. (2003) Antioxidant agents are effective in inducing lymphocyte progression through cell cycle in advanced cancer patients: Assessment of the most important laboratory indexes of cachexia and oxidative stress. Journal of Molecular Medicine, 81, 664-673. doi:10.1007/s00109-003-0476-1
[135] Weitsman, G.E., et al. (2003) Vitamin D enhances caspase-dependent and independent TNF-induced breast cancer cell death: The role of reactive oxygen species. Annals of New York Academy of Sciences, 1010, 437-440.
[136] Mantovani, G., et al. (2003) The impact of different antioxidant agents alone or in combination on reactive oxygen species, antioxidant enzymes and cytokines in a series of advanced cancer patients at different sites: Correlation with disease progression. Free Radical Research, 37, 213-223. doi:10.1080/10715760303849
[137] Zahid, M., et al. (2007) Inhibition of depurinating estrogen-DNA adduct formation by natural compounds. Chemical Research in Toxicology, 20, 1947-1953. doi:10.1021/tx700269s
[138] Novotny, L., Rauko, P. and Cojocel, C. (2008) Alphalipoic acid: The potential for use in cancer therapy. Neoplasma, 55, 81-86.
[139] Rock, E. and DeMichele, A. (2003) Nutritional approaches to late toxicities of adjuvant chemotherapy in breast cancer survivors. The Journal of Nutrition, 133, 3785S-3793S.
[140] Schwartz, L., et al. (2012) Tumor regression with a combination of drugs interfering with the tumor metabolism: Efficacy of hydroxycitrate, lipoic acid and capsaicin. Investigational New Drugs, 31, 256-264.
[141] Moungjaroen, J., et al. (2006) Reactive oxygen species mediate caspase activation and apoptosis induced by lipoic acid in human lung epithelial cancer cells through Bcl-2 down-regulation. The Journal of Pharmacology and Experimental Therapeutics, 319, 1062-1069. doi:10.1124/jpet.106.110965
[142] Miquel, J., et al. (2006) Menopause: A review on the role of oxygen stress and favorable effects of dietary antioxidants. Archives of Gerontology and Geriatrics, 42, 289306. doi:10.1016/j.archger.2005.08.005
[143] Valko, M., et al. (2006) Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chemico-Biological Interactions, 160, 1-40. doi:10.1016/j.cbi.2005.12.009
[144] Ratnam, D.V., et al. (2006) Role of antioxidants in prophylaxis and therapy: A pharmaceutical perspective. Journal of Controlled Release, 113, 189-207. doi:10.1016/j.jconrel.2006.04.015
[145] Matkovics, A. (2006) Recent changes in concepts of antioxidant treatment. Orvosi Hetilap, 147, 747-752.
[146] Schmelzer, C., et al. (2007) Functional connections and pathways of coenzyme Q10-inducible genes: An in-silico study. IUBMB Life, 59, 628-633. doi:10.1080/15216540701545991
[147] Pepe, S., et al. (2007) Coenzyme Q10 in cardiovascular disease. Mitochondrion, 7, S154-S167. doi:10.1016/j.mito.2007.02.005
[148] Rosenfeldt, F., et al. (2005) Coenzyme Q10 therapy before cardiac surgery improves mitochondrial function and in Vitro contractility of myocardial tissue. The Journal of Thoracic and Cardiovascular Surgery, 129, 25-32. doi:10.1016/j.jtcvs.2004.03.034
[149] Bailey, D.M., et al. (2007) Electron paramagnetic spectroscopic evidence of exercise-induced free radical accumulation in human skeletal muscle. Free Radical Research, 41, 182-190. doi:10.1080/10715760601028867
[150] Thomas, J.E., Lee, N. and Thompson, P.D. (2007) Statins provoking MELAS syndrome. A case report. European Neurology, 57, 232-235. doi:10.1159/000101287
[151] Langsjoen, P.H., et al. (2005) Treatment of statin adverse effects with supplemental Coenzyme Q10 and statin drug discontinuation. Biofactors, 25, 147-152. doi:10.1002/biof.5520250116
[152] Lamperti, C., et al. (2005) Muscle coenzyme Q10 level in statin-related myopathy. Archives of Neurology, 62, 17091712. doi:10.1001/archneur.62.11.1709
[153] Stocker, R., et al. (2006) Neither plasma coenzyme Q10 concentration, nor its decline during pravastatin therapy, is linked to recurrent cardiovascular disease events: A prospective case-control study from the LIPID study. Atherosclerosis, 187, 198-204. doi:10.1016/j.atherosclerosis.2005.09.004
[154] Siemieniuk, E. and Skrzydlewska, E. (2005) Coenzyme Q10: Its biosynthesis and biological significance in animal organisms and in humans. Postepy Higieny I Medycyny Doswiadczalnej (Online), 59, 150-159.
[155] Tavintharan, S., et al. (2007) Reduced mitochondrial coenzyme Q10 levels in HepG2 cells treated with highdose simvastatin: A possible role in statin-induced hepatotoxicity? Toxicology and Applied Pharmacology, 223, 173-179. doi:10.1016/j.taap.2007.05.013
[156] McCarthy, S., et al. (2004) Paraquat induces oxidative stress and neuronal cell death; neuroprotection by water-soluble Coenzyme Q10. Toxicology and Applied Pharmacology, 201, 21-31. doi:10.1016/j.taap.2004.04.019
[157] Korkina, L., et al. (2003) Coenzyme Q10-containing composition (Immugen) protects against occupational and environmental stress in workers of the gas and oil Industry. Biofactors, 18, 245-254. doi:10.1002/biof.5520180227
[158] Bruge, F., et al. (2003) Effect of UV-C mediated oxidative stress in leukemia cell lines and its relation to ubiquinone content. Biofactors, 18, 51-63. doi:10.1002/biof.5520180207
[159] Ebadi, M., et al. (2004) Coenzyme Q10 inhibits mitochondrial complex-1 down-regulation and nuclear factor-kappa B activation. Journal of Cellular and Molecular Medicine, 8, 213-222. doi:10.1111/j.1582-4934.2004.tb00276.x
[160] Hyun, D.H., et al. (2006) Calorie restriction up-regulates the plasma membrane redox system in brain cells and suppresses oxidative stress during aging. Proceedings of the National Academy of Sciences of the United States of America, 103, 19908-19912. doi:10.1073/pnas.0608008103
[161] Bhagavan, H.N., et al. (2007) Assessment of coenzyme Q10 absorption using an in Vitro digestion-Caco-2 cell model. International Journal of Pharmaceutics, 333, 112-117. doi:10.1016/j.ijpharm.2006.10.007
[162] Niklowitz, P., et al. (2007) Coenzyme Q10 concentration in the plasma of children suffering from acute lymphoastic leukaemia before and during induction treatment. Biofactors, 29, 83-89. doi:10.1002/biof.552029208
[163] van Dalen, E.C., Caron, H.N. and Kremer, L.C. (2007) Prevention of anthracycline-induced cardiotoxicity in children: The evidence. European Journal of Cancer, 43, 1134-1140. doi:10.1016/j.ejca.2007.01.040
[164] Conklin, K.A. (2005) Coenzyme Q10 for prevention of anthracycline-induced cardiotoxicity. Integrative Cancer Therapies, 4, 110-130. doi:10.1177/1534735405276191
[165] Bryant, J., et al. (2007) Cardioprotection against the toxic effects of anthracyclines given to children with cancer: A systematic review. Health Technology Assessment, 11, iii, ix-x, 1-84.
[166] Premkumar, V.G., et al. (2007) Effect of coenzyme Q10, riboflavin and niacin on serum CEA and CA 15-3 levels in breast cancer patients undergoing tamoxifen therapy. Biological & Pharmaceutical Bulletin, 30, 367-370. doi:10.1248/bpb.30.367
[167] Yuvaraj, S., et al. (2007) Ameliorating effect of coenzyme Q10, riboflavin and niacin in tamoxifen-treated postmenopausal breast cancer patients with special reference to lipids and lipoproteins. Clinical Biochemistry, 40, 623628. doi:10.1016/j.clinbiochem.2007.02.003
[168] Palan, P.R., et al. (2005) Effects of menopause and hormone replacement therapy on serum levels of coenzyme Q10 and other lipid-soluble antioxidants. Biofactors, 25, 61-66. doi:10.1002/biof.5520250107
[169] Rusciani, L., et al. (2007) Recombinant interferon alpha-2b and coenzyme Q10 as a postsurgical adjuvant therapy for melanoma: A 3-year trial with recombinant interferon-alpha and 5-year follow-up. Melanoma Research, 17, 177-183. doi:10.1097/CMR.0b013e32818867a0
[170] Rusciani, L., et al. (2006) Low plasma coenzyme Q10 levels as an independent prognostic factor for melanoma progression. Journal of the American Academy of Dermatology, 54, 234-241. doi:10.1016/j.jaad.2005.08.031
[171] Brea-Calvo, G., et al. (2006) Chemotherapy induces an increase in coenzyme Q10 levels in cancer cell lines. Free Radical Biology and Medicine, 40, 1293-1302. doi:10.1016/j.freeradbiomed.2005.11.014
[172] Roffe, L., Schmidt, K. and Ernst, E. (2004) Efficacy of coenzyme Q10 for improved tolerability of cancer treatments: A systematic review. Journal of Clinical Oncology, 22, 4418-4424.
[173] Fulda, S. and Debatin, K.M. (2006) Resveratrol modulation of signal transduction in apoptosis and cell survival: A mini-review. Cancer Detection and Prevention, 30, 217-223. doi:10.1016/j.cdp.2006.03.007
[174] Vinod, B.S., Maliekal, T.T. and Anto, R.J. (2012) Phytochemicals as chemosensitizers: From molecular mechanism to clinical significance. Antioxidants & Redox Signaling, 18, 1307-1348.
[175] Hull, M.A. (2012) Nutritional agents with anti-lnflammatory properties in chemoprevention of colorectal neoplasia. Recent Results in Cancer Research, 191, 143-156. doi:10.1007/978-3-642-30331-9_8
[176] Athar, M., et al. (2007) Resveratrol: A review of preclinical studies for human cancer prevention. Toxicology and Applied Pharmacology, 224, 274-283. doi:10.1016/j.taap.2006.12.025
[177] Leon-Galicia, I., et al. (2012) Resveratrol induces downregulation of DNA repair genes in MCF-7 human breast cancer cells. European Journal Cancer Prevention, 22, 11-20.
[178] Kim, Y.S., Sull, J.W. and Sung, H.J. (2012) Suppressing effect of resveratrol on the migration and invasion of human metastatic lung and cervical cancer cells. Molecular Biology Reports, 39, 8709-8716. doi:10.1007/s11033-012-1728-3
[179] Scoditti, E., et al. (2012) Mediterranean diet polyphenols reduce inflammatory angiogenesis through MMP-9 and COX-2 inhibition in human vascular endothelial cells: A potentially protective mechanism in atherosclerotic vascular disease and cancer. Archives of Biochemistry and Biophysics, 527, 81-89.doi:10.1016/j.abb.2012.05.003
[180] Liu, M.M., Huang, Y. and Wang, J. (2012) Developing phytoestrogens for breast cancer prevention. Anti-Cancer Agents in Medicinal Chemistry, 12, 1306-1313.
[181] Shankar, S., Singh, G. and Srivastava, R.K. (2007) Chemoprevention by resveratrol: Molecular mechanisms and therapeutic potential. Frontiers in Bioscience, 12, 48394854. doi:10.2741/2432
[182] Boocock, D.J., et al. (2007) Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiology, Biomarkers & Prevention, 16, 1246-1252. doi:10.1158/1055-9965.EPI-07-0022
[183] Delmas, D., et al. (2006) Resveratrol as a chemopreventive agent: A promising molecule for fighting cancer. Current Drug Targets, 7, 423-442.
[184] Aziz, M.H., et al. (2005) Chemoprevention of skin cancer by grape constituent resveratrol: Relevance to human disease? FASEB Journal, 19, 1193-1195.
[185] Stojanovic, S., Sprinz, H. and Brede, O. (2001) Efficiency and mechanism of the antioxidant action of trans-resveratrol and its analogues in the radical liposome oxidation. Archives of Biochemistry and Biophysics, 391, 79-89. doi:10.1006/abbi.2001.2388
[186] Brito, P., Almeida, L.M. and Dinis, T.C. (2002) The interaction of resveratrol with ferrylmyoglobin and peroxynitrite; Protection against LDL oxidation. Free Radical Research, 36, 621-631. doi:10.1080/10715760290029083
[187] Bradamante, S., Barenghi, L. and Villa, A. (2004) Cardiovascular protective effects of resveratrol. Cardiovascular Drug Reviews, 22, 169-188. doi:10.1111/j.1527-3466.2004.tb00139.x
[188] Das, S. and Das, D.K. (2007) Anti-inflammatory responses of resveratrol. Inflammation & Allergy Drug Targets, 6, 168-173. doi:10.2174/187152807781696464
[189] Goswami, S.K. and Das, D.K. (2009) Resveratrol and chemoprevention. Cancer Letters, 284, 1-6. doi:10.1016/j.canlet.2009.01.041
[190] Aggarwal, B.B., et al. (2004) Role of resveratrol in prevention and therapy of cancer: Preclinical and clinical studies. Anticancer Research, 24, 2783-2840.
[191] Gescher, A.J. and Steward, W.P. (2003) Relationship between mechanisms, bioavailibility, and preclinical chemopreventive efficacy of resveratrol: A conundrum. Cancer Epidemiology, Biomarkers & Prevention, 12, 953957.
[192] Bass, T.M., et al. (2007) Effects of resveratrol on lifespan in Drosophila melanogaster and Caenorhabditis elegans. Mechanisms of Ageing and Development, 128, 546-552. doi:10.1016/j.mad.2007.07.007
[193] Bartel, J., et al. (2007) Activity of the glutathione peroxidase-2. Differences in the selenium-dependent expression between colon and small intestine. Cancer Genomics & Proteomics, 4, 369-372.
[194] Steinbrenner, H., et al. (2007) Post-translational processing of selenoprotein P: Implications of glycosylation for its utilisation by target cells. Biological Chemistry, 388, 1043-1051. doi:10.1515/BC.2007.136
[195] Philchenkov, A., et al. (2007) Comparative analysis of apoptosis induction by selenium compounds in human lymphoblastic leukemia MT-4 cells. Experimental Oncology, 29, 257-261.
[196] Meplan, C. and Hesketh, J. (2012) The influence of selenium and selenoprotein gene variants on colorectal cancer risk. Mutagenesis, 27, 177-186. doi:10.1093/mutage/ger058
[197] Bardia, A., et al. (2008) Efficacy of antioxidant supplementation in reducing primary cancer incidence and mortality: Systematic review and meta-analysis. Mayo Clinic Proceedings, 83, 23-34. doi:10.4065/83.1.23
[198] Li, S., et al. (2007) Doxorubicin and selenium cooperatively induce fas signaling in the absence of Fas/Fas ligand interaction. Anticancer Research, 27, 3075-3082.
[199] Santos, R.A. and Takahashi, C.S. (2008) Anticlastogenic and antigenotoxic effects of selenomethionine on doxorubicin-induced damage in Vitro in human lymphocytes. Food and Chemical Toxicology, 46, 671-677. doi:10.1016/j.fct.2007.09.090
[200] Cheung, E., et al. (2008) Diet and prostate cancer risk reduction. Expert Review of Anticancer Therapy, 8, 43-50. doi:10.1586/14737140.8.1.43
[201] Juliger, S., et al. (2007) Chemosensitization of B-cell lymphomas by methylseleninic acid involves nuclear factor-kappaB inhibition and the rapid generation of other selenium species. Cancer Research, 67, 10984-10992. doi:10.1158/0008-5472.CAN-07-0519
[202] Murawaki, Y., et al. (2008) Aberrant expression of selenoproteins in the progression of colorectal cancer. Cancer Letters, 259, 218-230. doi:10.1016/j.canlet.2007.10.019
[203] Ma, J.L., et al. (2012) Fifteen-year effects of Helicobacter pylori, garlic, and vitamin treatments on gastric cancer incidence and mortality. Journal of the National Cancer Institiute, 104, 488-492. doi:10.1093/jnci/djs003
[204] Chen, K.M., et al. (2007) Inhibition of nuclear factorkappaB DNA binding by organoselenocyanates through covalent modification of the p50 subunit. Cancer Research, 67, 10475-10483. doi:10.1158/0008-5472.CAN-07-2510
[205] Lee, E.H., et al. (2011) Effects of selenium supplements on cancer prevention: Meta-analysis of randomized controlled trials. Nutrition and Cancer, 63, 1185-1195. doi:10.1080/01635581.2011.607544
[206] Sanmartin, C., et al. (2011) Selenium and clinical trials: New therapeutic evidence for multiple diseases. Current Medicinal Chemistry, 18, 4635-4650.
[207] Cuello, S., et al. (2007) Selenium methylselenocysteine protects human hepatoma HepG2 cells against oxidative stress induced by tert-butyl hydroperoxide. Analytical and Bioanalytical Chemistry, 389, 2167-2178. doi:10.1007/s00216-007-1626-z
[208] Schmutzler, C., et al. (2007) Selenoproteins of the thyroid gland: Expression, localization and possible function of glutathione peroxidase 3. Biological Chemistry, 388, 1053-1059. doi:10.1515/BC.2007.122
[209] Yanagisawa, H. (2008) Zinc deficiency and clinical practice—Validity of zinc preparations. Yakugaku Zasshi, 128, 333-339. doi:10.1248/yakushi.128.333
[210] Yan, M., et al. (2008) Zinc deficiency alters DNA damage response genes in normal human prostate epithelial cells. The Journal of Nutrition, 138, 667-673.
[211] Lonn, E.M. and Yusuf, S. (1997) Is there a role for antioxidant vitamins in the prevention of cardiovascular diseases? An update on epidemiological and clinical trials data. The Canadian Journal of Cardiology, 13, 957-965.
[212] Bansal, S.S., et al. (2011) Advanced drug delivery systems of curcumin for cancer chemoprevention. Cancer Prevention Research, 4, 1158-1171. doi:10.1158/1940-6207.CAPR-10-0006
[213] Kaefer, C.M. and Milner, J.A. (2011) Herbs and spices in cancer prevention and treatment, in herbal medicine, In: I.F.F. Benzie and S. Wachtel-Galor, Eds., Biomolecular and Clinical Aspects, Llc, Boca Raton.
[214] Gullett, N.P., et al. (2010) Cancer prevention with natural compounds. Seminars in Oncology, 37, 258-281. doi:10.1053/j.seminoncol.2010.06.014
[215] Taylor, C.K., et al. (2009) The effect of genistein aglycone on cancer and cancer risk: A review of in Vitro, preclinical, and clinical studies. Nutrition Reviews, 67, 398415. doi:10.1111/j.1753-4887.2009.00213.x
[216] Gerhauser, C. (2013) Cancer chemoprevention and nutriepigenetics: State of the art and future challenges. Topics in Current Chemistry, 329, 73-132. doi:10.1007/128_2012_360
[217] Li, Y., et al. (2011) Implications of cancer stem cell theory for cancer chemoprevention by natural dietary compounds. The Journal of Nutritional Biochemistry, 22, 799-806. doi:10.1016/j.jnutbio.2010.11.001
[218] Kim, Y.S., et al. (2012) Cancer stem cells: Potential target for bioactive food components. The Journal of Nutritional Biochemistry, 23, 691-698. doi:10.1016/j.jnutbio.2012.03.002
[219] Ullah, M.F., et al. (2012) Ascorbic acid in cancer chemoprevention: Translational perspectives and efficacy. Current Drug Targets, 13, 1757-1771. doi:10.2174/138945012804545669
[220] Khan, S.I., et al. (2012) Epigenetic events associated with breast cancer and their prevention by dietary components targeting the epigenome. Chemical Research in Toxicology, 25, 61-73. doi:10.1021/tx200378c
[221] Ozten-Kandas, N. and Bosland, M.C. (2011) Chemoprevention of prostate cancer: Natural compounds, antiandrogens, and antioxidants-In Vivo evidence. Journal of Carcinogenesis, 10, 27. doi:10.4103/1477-3163.90438
[222] Khan, N., Adhami, V.M. and Mukhtar, H. (2010) Apoptosis by dietary agents for prevention and treatment of prostate cancer. Endocrine-Related Cancer, 17, R39-R52. doi:10.1677/ERC-09-0262
[223] Howells, L.M., et al. (2011) Phase I randomized, double-blind pilot study of micronized resveratrol (SRT501) in patients with hepatic metastases—Safety, pharmacokinetics, and pharmacodynamics. Cancer Prevention Research, 4, 1419-1425. doi:10.1158/1940-6207.CAPR-11-0148
[224] Ye, F., et al. (2012) Suppression of esophageal cancer cell growth using curcumin, (-)-epigallocatechin-3-gallate and lovastatin. World Journal of Gastroenterology, 18, 126135. doi:10.3748/wjg.v18.i2.126
[225] Hossain, D.M., et al. (2012) Curcumin: The multi-targeted therapy for cancer regression. Front in Bioscience, 4, 335-355.
[226] Basnet, P. and Skalko-Basnet, N. (2011) Curcumin: An anti-inflammatory molecule from a curry spice on the path to cancer treatment. Molecules, 16, 4567-4598. doi:10.3390/molecules16064567
[227] Darvesh, A.S., Aggarwal, B.B. and Bishayee, A. (2012) Curcumin and liver cancer: A review. Current Pharmaceutical Biotechnology, 13, 218-228.
[228] Shureiqi, I. and Baron, J.A. (2011) Curcumin chemoprevention: The long road to clinical translation. Cancer Prevention Research, 4, 296-298. doi:10.1158/1940-6207.CAPR-11-0060
[229] Rahman, M.A., Amin, A.R. and Shin, D.M. (2010) Chemopreventive potential of natural compounds in head and neck cancer. Nutrition and Cancer, 62, 973-987. doi:10.1080/01635581.2010.509538
[230] Shehzad, A., Wahid, F. and Lee, Y.S. (2010) Curcumin in cancer chemoprevention: Molecular targets, pharmacokinetics, bioavailability, and clinical trials. Archiv der Pharmazie, 343, 489-499. doi:10.1002/ardp.200900319
[231] Juan, M.E., Alfaras, I. and Planas, J.M. (2012) Colorectal cancer chemoprevention by trans-resveratrol. Pharmacological Research, 65, 584-591. doi:10.1016/j.phrs.2012.03.010
[232] Scott, E., et al. (2012) Resveratrol in human cancer chemoprevention—Choosing the “right” dose. Molecular Nutrition & Food Research, 56, 7-13. doi:10.1002/mnfr.201100400
[233] Vang, O., et al. (2011) What is new for an old molecule? Systematic review and recommendations on the use of resveratrol. PLoS One, 6, e19881. doi:10.1371/journal.pone.0019881

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.