Broccoli eats cancer sm - Revitalize Health and Wellness

Oncology References

(2020) Gurdev Parmar, ND, FABNO. Textbook of Naturopathic Oncology

Dr. Neil McKinney, BSc, ND. Naturopathic Oncology 4: An Encyclopedic Guide for Patients & Physicians  2017.  

Lesley Braun, Marc Cohen. “Herbs and Natural Supplements, Volume 2: An Evidence-Based Guide, Volume 2.”

(2009) A.R. Amin, O. Kucuk, F.R. Khuri, D.M. Shin.  “Perspectives for cancer prevention with natural compounds.”  J. Clin. Oncol., 27 (2009), pp. 2712-2725

(2010) Henning SM, Zhang Y, Seeram NP, Lee RP, Wang P, Bowerman S, Heber D.  “Antioxidant capacity and phytochemical content of herbs and spices in dry, fresh and blended herb paste form.” Int J Food Sci Nutr. 2011 May;62(3):219-25. doi: 10.3109/09637486.2010.530595. Epub 2010 Dec 1. PMID: 21118053.

(2013) Nousheen Zaidi, et al.  “Lipogenesis and lipolysis: The pathways exploited by the cancer cells to acquire fatty acids.” Progress in Lipid Research. Volume 52, Issue 4, October 2013, Pages 585-589.

(2014) J.F. Lesgards, N. Baldovini, N. Vidal, S. Pietri.  “Anticancer activities of essential oils constituents and synergy with conventional therapies: a review.”  Phytother. Res., 28 (2014), pp. 1423-1446.

(2014) Saedi, Tayebeh Azam et al. “The effects of herbs and fruits on leukaemia.” Evidence-based complementary and alternative medicine: eCAM vol. 2014 (2014): 494136. doi:10.1155/2014/494136

(2015) Mulpur, Bhageeradh H et al. “Complementary therapy and survival in glioblastoma.” Neuro-oncology practice vol. 2,3 (2015): 122-126. doi:10.1093/nop/npv008

(2015) Greenwell, M, and P K S M Rahman. “Medicinal Plants: Their Use in Anticancer Treatment.” International journal of pharmaceutical sciences and research vol. 6,10 (2015): 4103-4112. doi:10.13040/IJPSR.0975-8232.6(10).4103-12

(2016) Fleischer T, Chang TT, Chiang JH, et al. “Improved Survival With Integration of Chinese Herbal Medicine Therapy in Patients With Acute Myeloid Leukemia: A Nationwide Population-Based Cohort Study.” Integr Cancer Ther. Aug 16 2016.

(2017) James A. McCubrey, etal. “Effects of resveratrol, curcumin, berberine and other nutraceuticals on aging, cancer development, cancer stem cells and microRNAs.” Aging 2017. Vol 9, No. 6. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5509453/pdf/aging-09-1477.pdf. 

(2017) Vilas Desai and Alok Bhushan.  “Natural Bioactive Compounds: Alternative Approach to the Treatment of Glioblastoma Multiforme.” Hindawi BioMed Research International. Volume 2017, Article ID 9363040, 10 pages.

(2017) Lin SR, Fu YS, Tsai MJ, Cheng H, Weng CF. “Natural Compounds from Herbs that can Potentially Execute as Autophagy Inducers for Cancer Therapy.”  Int J Mol Sci. 2017;18(7):1412. Published 2017 Jul 1. doi:10.3390/ijms18071412

(2018) Trogrlić, Ivo et al. “Treatment of glioblastoma with herbal medicines.” World journal of surgical oncology vol. 16,1 28. 13 Feb. 2018, doi:10.1186/s12957-018-1329-2

(2018) Mazumder A, et al.  “Natural scaffolds in anticancer therapy and precision medicine.” Biotechnology Advances. Volume 36, Issue 6, 1 November 2018, Pages 1563-1585

 

N-ACETYL-CYSTEINE (NAC) 

(2006) Cetinkaya A, Bulbuloglu E, Kurutas EB, Kantarceken B. “N-acetylcysteine ameliorates methotrexate-induced oxidative liver damage in rats.” Med Sci Monit. 2006;12:BR274–BR278.

(2006) Ciralik H, Bulbuloglu E, Cetinkaya A, Kurutas EB, Celik M, Polat A. “Effects of N-acetylcysteine on methotrexate-induced small intestinal damage in rats.” Mt Sinai J Med. 2006;73:1086–1092.

(2007)  Cetinkaya A, Kurutas EB, Bulbuloglu E, Kantarceken B. “The effects of N-acetylcysteine on methotrexate-induced oxidative renal damage in rats.” Nephrol Dial Transplant. 2007;22:284–285.

(2013) Caglar Y, Ozgur H, Matur I, et al. “Ultrastructural evaluation of the effect of N-

acetylcysteine on methotrexate nephrotoxicity in rats.” Histol Histopathol. 2013;28:865–874. 

(2014) Afshin Amini, et al. “Bromelain and N-acetylcysteine inhibit proliferation and survival of gastrointestinal cancer cells in vitro: significance of combination therapy.”  Journal of Experimental & Clinical Cancer Research. 2014, 33:9. https://doi.org/10.1186/s13046-014-0092-7.

(2014) Sayin V.I., Ibrahim M.X., Larsson E., Nilsson J.A., Lindahl P., Bergo M.O. “Antioxidants Accelerate Lung Cancer Progression in Mice. ” Sci. Transl. Med. 2014;6:221ra15. doi: 10.1126/scitranslmed.3007653.

(2017) Daniel Monti, et al. “Pilot study demonstrating metabolic and anti-proliferative effects of in vivo anti-oxidant supplementation with N-Acetylcysteine in Breast Cancer.” Seminars in Oncology, 2017; DOI: 10.1053/j.seminoncol.2017.10.001

(2018) Salvatore Grisanti, et al. “Hepatoprotective effect of N-acetylcysteine in trabectedin-induced liver toxicity in patients with advanced soft tissue sarcoma.”  Support Care Cancer (2018) 26: 2929. https://doi.org/10.1007/s00520-018-4129-x.

(2019) Deng J, Liu AD, Hou GQ, Zhang X, Ren K, Chen XZ, Li SSC, Wu YS, Cao X. “N-acetylcysteine decreases malignant characteristics of glioblastoma cells by inhibiting Notch2 signaling.” J Exp Clin Cancer Res. 2019;38:2.

(2019) Šalamon, Špela et al. “Medical and Dietary Uses of N-Acetylcysteine.” Antioxidants (Basel, Switzerland) vol. 8,5 111. 28 Apr. 2019, doi:10.3390/antiox8050111

ALPHA LIPOIC ACID

(2002) Jones, et al. Uptake, Recycling, and Antioxidant Actions of Alpha-lipoic acid in Endothelial Cells.” Free Radic. Biol. Med. 2002; 33 (1): 83-93.

(2002) Gedlicka, et al. Effective Treatment of Oxaliplatin-Induced Cumulative Poly-Neuropathy with Alpha-Lipoic Acid.”  J. Clin. Oncol. 2002; 20 (5): 3359-3361.

(2003) van de MK, Chen JS, Steliou K, Perrine SP, Faller DV. Alpha-lipoic acid induces p27Kip-dependent cell cycle arrest in non-transformed cell lines and apoptosis in tumor cell lines. J Cell Physiol2003;194:325-40.

(2005) Wenzel, et al.  Alpha Lipoic Acid Induces Apoptosis in Human Colon Cancer Cells by Increasing Mitochondrial Respiration with a Concomitant O2* Generation.” Apoptosis 2005; 10 (2): 359-368.

(2006) Mantovani G, Macció A, Madeddu C, et al. “A phase II study with antioxidants, both in the diet and supplemented, pharmaconutritional support, Progestagen, and anti-cyclooxygenase-2 showing efficacy and safety in patients with cancer-related anorexia/cachexia and oxidative stress.” Cancer Epidemiol Biomarkers Prev. 2006;15:1030-1034. doi:10.1158/1055-9965.EPI-05-0538

(2006) Moungiaroen, et al.  Reactive Oxygen Species Mediate Caspase Activation and Apoptosis Induced by Alpha Lipoic Acid in Human Lung Epithelial Cancer Cells Through Bcl-2 Down-regulation.  J. Pharmacolo. Exp. 2006; 319 (3): 1062-1069.

(2006) Berkson, et al.  The Long-Term Survival of a Patient with Pancreatic Cancer with Metastases to the Liver After Treatment with the Intravenous Alpha-Lipoic Acid/Low-Dose Naltrexone Protocol.Integr. Cancer Ther. 2006; 5 (1): 83-89.

(2007) Simbula G, et al. Increased ROS generation and p53 activation in alpha-lipoic acid-induced apoptosis of hepatoma cells. Apoptosis 2007 Jan;12(1):113-23.

(2008) Shi DY, Liu HL, Stern JS, et al. “Alpha-lipoic acid induces apoptosis in hepatoma cells via the PTEN/Akt pathway”. FEBS Lett. 2008 May 28;582(12):1667-71.

(2008) Park KG, Min AK, Koh EH, et al. “Alpha-lipoic acid decreases hepatic lipogenesis through adenosine monophosphate- activated protein kinase (AMPK)-dependent and AMPK-independent pathways.” Hepatology 2008 Nov;48(5):1477-86.

(2008) N.A. Mikirova, et al. “Differential Effect of Alpha-lipoic Acid on Healthy Peripheral Blood Lymphocytes and Leukemic Cells.” Journal of Orthomolecular Medicine Vol. 23, No. 2, 2008 (83-89).

(2009) Berkson BM, Rubin DM, Berkson AJ. “Revisiting the ALA/N (a-Lipoic Acid/Low-Dose Naltrexone) protocol for people with metastatic and nonmetastatic pancreatic cancer: a report of 3 new cases.” Integ Cancer Ther. 2009;8:416-422. doi: 10.1177/1534735409352082.

(2010) Dozio E, et al. “The natural antioxidant alpha-lipoic acid induces p27(Kip1)-dependent cell cycle arrest and apoptosis in MCF-7 human breast cancer cells.” Eur J Pharmacol. 2010 Sep 1;641(1):29-34.

(2010) Kisurina-Evgen’eva OP, Onishchenko GE. “Alpha-lipoic acid triggers elimination of cells with abnormal nuclei in human carcinoma epidermoid cell line.” Tsitologiia. 2010;52(3):225-34.

(2010) Srinivasan Krishna, et al. Differential Effects of Short‐Chain Fatty Acids on Head and Neck Squamous Carcinoma Cells.” The Laryngoscope. Volume 112, Issue 4. April 2002, Pages 645-650 https://cam.cancer.gov/news_and_events/newsletter/2012-spring/feature.htm

(2012) “Alpha-Lipoic Acid Plus Low-Dose Naltrexone Reviewed for Cancer Treatment.”  NIH, Division of Cancer Treatment and Diagnosis. Lymphomas and pancreatic cancers.

https://cam.cancer.gov/news_and_events/newsletter/2012-spring/feature.htm

(2013) Kapoor, Shailendra.  “The Anti-neoplastic Effects of Alpha-Lipoic Acid: Clinical Benefits in System Tumors besides Lung Carcinomas.” Korean Journal of Thoracic Cardiovascular Surgery. 2013; 46:162-163  (melanoma).

(2013) Hiratsuka, T., et al. “DHL-TauZnNa, a newly synthesized α-lipoic acid derivative, induces autophagy in human colorectal cancer cells”.  Oncology Reports 29.6 (2013): 2140-2146.

(2014) Moon.  “Chemopreventive Effects of Alpha Lipoic Acid on Obesity-Related Cancers.” Ann Nutr Metab 2016;68:137-144. (breast, thyroid, colon, pancreatic and liver).

(2014) Udensi, Udensi K, and Paul B Tchounwou. “Dual effect of oxidative stress on leukemia cancer induction and treatment.” Journal of experimental & clinical cancer research : CR vol. 33 106. 18 Dec. 2014, doi:10.1186/s13046-014-0106-5.

(2014) Guo Y, Jones D, Palmer JL, et al. “Oral alpha-lipoic acid to prevent chemotherapy-induced peripheral neuropathy: a randomized, double-blind, placebo-controlled trial.” Support Care Cancer. 2014;22:1223-1231. doi: 10.1007/s00520-013-2075-1.

(2015) Dörsam B, et al. “Lipoic acid induces p53-independent cell death in colorectal cancer cells and potentiates the cytotoxicity of 5-fluorouracil.” Arch Toxicol. 2015 Oct;89(10):1829-46.

(2015) Jeon MJ, Kim WG, Lim S, et al. “Alpha lipoic acid inhibits proliferation and epithelial mesenchymal transition of thyroid cancer cells.” Mol Cell Endocrinol. 2016;419:113-123. doi: 10.1016/j.mce.2015.10.005

(2015) Kafara P, Icard P, Guillamin M, et al. “Lipoic acid decreases Mcl-1, Bcl-xL and up regulates Bim on ovarian carcinoma cells leading to cell death.” J Ovarian Res. 2015;8:36-49. doi: 10.1186/s13048-015-0165-z.

(2016) Puchsaka P, Chaotham C, Chanvorachote P. “α-Lipoic acid sensitizes lung cancer cells to chemotherapeutic agents and anoikis via integrin β1/β3 downregulation.” Int J Oncol. 2016;49:1445-1456. doi: 10.3892/ijo.2016.3624

(2016) Alpay M, et al. “Antileukemic effects of piperlongumine and alpha lipoic acid combination on Jurkat, MEC1 and NB4 cells in vitro.” Journal of Cancer Research and Therapeutics. 2016;12:2:556-560

(2017) Kuban-Jankowska A, Gorska-Ponikowska M, Wozniak M. “Lipoic acid decreases the viability of breast cancer cells and activity of PTP1B and SHP2.” Anticancer Res. 2017;37:2893-2898.

(2017) Kismali G, Yurdakok-Dikmem B, Kuzukiran O, et al. “Phthalate induced toxicity in prostate cancer cell lines and effects of alpha lipoic acid.” Bratisl Med J. 2017;118:460-466. doi: 10.4149/BLL_2017_089.

(2017) Desideri I, Francolini G, Becherini C, et al. “Use of an alpha lipoic, methylsulfonylmethane and bromelain dietary supplement (Opera®) for chemotherapy-induced peripheral neuropathy management, a prospective study.” Med Oncol. 2017;34:46. doi: 10.1007/s12032-017-0907-4.

(2018) Berkson, Burton M, and Francisco Calvo Riera. “The Long-Term Survival of a Patient With Stage IV Renal Cell Carcinoma Following an Integrative Treatment Approach Including the Intravenous α-Lipoic Acid/Low-Dose Naltrexone Protocol.” Integrative cancer therapies vol. 17,3 (2018): 986-993. doi:10.1177/1534735417747984

(2018) Deveci HA, Akyuva Y, Nur G, Nazıroğlu M. “Alpha lipoic acid attenuates hypoxia-induced apoptosis, inflammation and mitochondrial oxidative stress via inhibition of TRPA1 channel in human glioblastoma cell line.” Biomed Pharmacother. 2019;111:292-304. doi:10.1016/j.biopha.2018.12.077

(2018) Dinicola S, et al.  “Natural products – alpha-lipoic acid and acetyl-L-carnitine – in the treatment of chemotherapy-induced peripheral neuropathy.” European Review for Medical and Pharmacological Sciences. 2018; 22: 4739-4754.

(2018) Arpag H, Gül M, Aydemir Y, et al. “Protective Effects of Alpha-Lipoic Acid on Methotrexate-Induced Oxidative Lung Injury in Rats.”  J Invest Surg. 2018;31(2):107-113. doi:10.1080/08941939.2017.1296513

(2019) Bahadur S, Sahu AK, Baghel P, Saha S. “Current promising treatment strategy for glioblastoma multiform: A review.” Oncology Reviews. 2019 Jul;13(2):417. DOI: 10.4081/oncol.2019.417.

ARTESUNATE/ARTEMISININ

(2014) Odaka, et al. Dihydroartemisinin inhibits the mammalian target of rapamycin-mediated signaling pathways in tumor cells.” Carcinogenesis. 2014 Jan;35(1):192-200. doi: 10.1093/carcin/bgt277. Rhabdomyosarcoma.

(2015) Hargraves, et al. Phytochemical regulation of the tumor suppressive microRNA, miR-34a, by p53-dependent and independent responses in human breast cancer cells.” Mol Carcinog. 2016 May;55(5):486-98. doi: 10.1002/mc.22296. Epub 2015 Mar 19.

(2015) . Zhang ZS, et al. “Dihydroartemisinin increases temozolomide efficacy in glioma cells by inducing autophagy.” Oncol Lett 2015;10:379-83

(2016) Drenberg, Christina D et al. “Evaluation of artemisinins for the treatment of acute myeloid leukemia.” Cancer chemotherapy and pharmacology vol. 77,6 (2016): 1231-43. doi:10.1007/s00280-016-3038-2

(2017) Slezakova, et alAnticancer Activity of Artemisinin and its Derivatives.” Anticancer Research November 2017 vol. 37 no. 11 5995-6003.

(2017) Kumar B. et al“Antileukemic activity and cellular effects of the antimalarial agent artesunate in acute myeloid leukemia.” Leuk Res. 2017 May. DOI: 10.1016/j.leukres.2017.05.007.

(2018) Dou, et al. Cytotoxic effect of Artesunate on myeloid leukemia cell lines through up-regulating miR-29c expression.” Translational Cancer Research. Vol 7, No 6 (December 2018)

(2019) Lam, et al. “Artemisinin and its derivatives: A potential treatment for leukemia.” (AML) Anti-Cancer Drugs 2019 Jan;30(1):1-18:1-18. 

ASTRAGALUS
(2006) McCulloch M, See C, Shu XJ, et al. “Astragalus-based Chinese herbs and platinum-based chemotherapy for advanced non-small-cell lung cancer: meta-analysis of randomized trials.” J Clin Oncol. 2006 Jan 20;24(3):419-30.

(2012) Auyeung KK, Woo PK, Law PC, Ko JK. “Astragalus saponins modulate cell invasiveness and angiogenesis in human gastric adenocarcinoma cells.”  JEthnopharmacol. 2012;141(2):635-41.

(2012) Guo L, Bai SP, Zhao L, Wang XH. “Astragalus polysaccharide injection integrated with vinorelbine and cisplatin for patients with advanced non-small cell lung cancer: effects on quality of life and survival.”  Med Oncol. 2012;29(3):1656-62.

(2012) Law PC, Auyeung KK, Chan LY, et al. “Astragalus saponins downregulate vascular endothelial growth factor under cobalt chloride-stimulated hypoxia in colon cancer cells.” BMC Complement Altern Med. 2012;12:160.

(2012) Huang, et al.  Astragalus membranaceus lectin (AML) induces caspase-dependent apoptosis in human leukemia cells (CML).” Cell Prolif. 2012 Feb;45(1):15-21.

(2014) Auyeung KK, Law PC, Ko JK. “Combined therapeutic effects of vinblastine and Astragalus saponins in human colon cancer cells and tumor xenograft via inhibition of tumor growth and proangiogenic factors.” Nutr Cancer. 2014;66(4):662-674.

(2014) Wang Y, Auyeung KK, Zhang X, et al. “Astragalus saponins modulates colon cancer development by regulating calpain-mediated glucose-regulated protein expression.” BMC Complement Altern Med. 2014;14:401.

(2016) Wang, S F et al. “Astragalus-containing Traditional Chinese Medicine, with and without prescription based on syndrome differentiation, combined with chemotherapy for advanced non-small-cell lung cancer: a systemic review and meta-analysis.” Current oncology (Toronto, Ont.) vol. 23,3 (2016): e188-95.

(2016) Huang WH, Liao WR, Sun RX. “Astragalus polysaccharide induces the apoptosis of human hepatocellular carcinoma cells by decreasing the expression of Notch1.” Int J Mol Med. Aug 2016;38(2):551-557.

(2016) Auyeung KK, Han QB, Ko JK. Astragalus membranaceus: A Review of its Protection Against Inflammation and Gastrointestinal Cancers.” Am J Chin Med. 2016;44(1):1-22.

(2016) Tseng A, Yang CH, Chen CH, et al. “An in vivo molecular response analysis of colorectal cancer treated with Astragalus membranaceus extract.” Oncol Rep. Feb 2016;35(2):659-668.

(2018) Zhang J, Liu L, Wang J, Ren B, Zhang L, Li W. “Formononetin, an isoflavone from Astragalus membranaceus inhibits proliferation and metastasis of ovarian cancer cells.”  J Ethnopharmacol. 2018 Jul 15;221:91-99.

BERBERINE

(2006) Mantena SK, Sharma SD, Katiyar SK. “Berberine inhibits growth, induces G1 arrest and apoptosis in human epidermoid carcinoma A431 cells by regulating Cdki-Cdk-cyclin cascade, disruption of mitochondrial membrane potential and cleavage of caspase 3 and PARP.” Carcinogenesis 27: 2018–2027.

(2007) Yu, et al.  “Berberine inhibits WEHI-3 leukemia cells in vivo.” In vivo (Athens, Greece) 21(2):407-12 · March 2007

(2008) LI, et al.  Berberine inhibits SDF-1-induced AML cells and leukemic stem cells migration via regulation of SDF-1 level in bone marrow stromal cells.” Biomed Pharmacother. 2008 Nov;62(9):573-8. doi: 10.1016/j.biopha.2008.08.003. Epub 2008 Sep 4.

(2010) Li GH, et al.  “Berberine inhibits acute radiation intestinal syndrome in human with abdomen radiotherapy.” Medical Oncology. 2010 Sep;27(3):919-25. doi: 10.1007/s12032-009-9307-8. Epub 2009 Sep 16.

(2014) Parham Jabbarzadeh Kaboli, et al. “Targets and mechanisms of berberine, a natural drug with potential to treat cancer with special focus on breast cancer.” European Journal of Pharmacology 740:584-595 · June 2014. DOI: 10.1016/j.ejphar.2014.06.025.

(2014) Luis Miguel Guamán Ortiz, Micol Tillhon, Michael Parks, et al. “Multiple Effects of Berberine Derivatives on Colon Cancer Cells.” BioMed Research International, vol. 2014, Article ID 924585, 12 pages, 2014. https://doi.org/10.1155/2014/924585.

(2014) Zhu, Yu et al. “Berberine induces apoptosis and DNA damage in MG-63 human osteosarcoma cells.” Molecular medicine reports vol. 10,4 (2014): 1734-8.

(2014), Zhang, et al. “Berberine inhibits the expression of hypoxia induction factor-1alpha and increases the radiosensitivity of prostate cancer.” Diagn Pathol (2014) 9: 98. https://doi.org/10.1186/1746-1596-9-98

(2015) Pandey A.  Berberine and Curcumin Target Survivin and STAT3 in Gastric Cancer Cells and Synergize Actions of Standard Chemotherapeutic 5-Fluorouracil.”  Nutrition and Cancer. 2015;67(8):1293-304. doi: 10.1080/01635581.2015.1085581. Epub 2015 Oct 22.

(2015) Liu, et al. “Berberine Inhibits Invasion and Metastasis of Colorectal Cancer Cells via COX-2/PGE2 Mediated JAK2/STAT3 Signaling Pathway.” PLoS One. 2015; 10(5): e0123478.

(2015) Zhu T, Li LL, Xiao GF, Luo QZ, Liu QZ, Yao KT, et al. “Berberine increases doxorubicin sensitivity by suppressing STAT3 in lung cancer.” Am J Chin Med 2015; 43:1487-1502.

(2016) Wang, Jiwei et al. “Berberine induces autophagy in glioblastoma by targeting the AMPK/mTOR/ULK1-pathway.” Oncotarget vol. 7,41 (2016): 66944-66958.

(2016) Jin, Hao et al. “Berberine affects osteosarcoma via downregulating the caspase-1/IL-1β signaling axis.” Oncology reports vol. 37,2 (2017): 729-736. doi:10.3892/or.2016.5327.

(2017) Pan, Yue et al. “Berberine Enhances Chemosensitivity and Induces Apoptosis Through Dose-orchestrated AMPK Signaling in Breast Cancer.” Journal of Cancer. vol. 8,9 1679-1689. 5 Jun. 2017, doi:10.7150/jca.19106.

(2017) Mohammadi S, Seyedhoseini FS, Asadi J, et al. “Effects of berberine on the secretion of cytokines and expression of genes involved in cell cycle regulation in THP-1 monocytic cell line.” (AML) Iran J Basic Med Sci. 2017;20:530–537.

(2017) Mehrzadi S, Fatemi I, Esmaeilizadeh M, Ghaznavi H, Kalantar H, Goudarzi M. “Hepatoprotective effect of berberine against methotrexate induced liver toxicity in rats.” Biomed Pharmacother. 2018;97:233-239. doi:10.1016/j.biopha.2017.10.113

(2018) Jin, Hao et al. “Berberine affects osteosarcoma via downregulating the caspase-1/IL-1β signaling axis.” Oncology reports vol. 37,2 (2016): 729-736.

(2018) Li, J., Yang, L., Shen, R. et al. “Self-nanoemulsifying system improves oral absorption and enhances anti-acute myeloid leukemia activity of berberine.”  J Nanobiotechnol 16, 76 (2018) doi:10.1186/s12951-018-0402-x

(2019) Shinji, et al. Berberine and palmatine inhibit the growth of human rhabdomyosarcoma cells.” Biosci Biotechnol Biochem. 2019 Aug 29:1-13. doi: 10.1080/09168451.2019.1659714.

(2019) Dai, et al. “Berberine Promotes Apoptosis of Colorectal Cancer via Regulation of the Long Non-Coding RNA (lncRNA) Cancer Susceptibility Candidate 2 (CASC2)/AU-Binding Factor 1 (AUF1)/B-Cell CLL/Lymphoma 2 (Bcl-2) Axis.” Medical Science Monitor. 2019 Jan 25;25:730-738. doi: 10.12659/MSM.912082.

BICARBONATE

(2009) Robey, et al. Bicarbonate increases tumor pH and inhibits spontaneous metastases.” Cancer Res. 2009 Mar 15;69(6):2260-8. doi: 10.1158/0008-5472.CAN-07-5575. Epub 2009 Mar 10.

BLACK CUMIN/ BLACK SEED OIL/ NIGELLA SATIVA

(2007) Mbarek L, et al.  “Anti-tumor effect of blackseed (Nigella sativa L) extracts.” Brazilian Journal of Medical and Biological Research (2007) 40: 839-847 ISSN 0100-879X

(2011) Khan, Md Asaduzzaman et al. “Anticancer activities of Nigella sativa (black cumin).” African journal of traditional, complementary, and alternative medicines.” AJTCAM vol. 8,5 Suppl (2011): 226-32. doi:10.4314/ajtcam.v8i5S.10

(2014) Arshad H. Rahmani, et al.  “Therapeutic Implications of Black Seed and Its Constituent Thymoquinone in the Prevention of Cancer through Inactivation and Activation of Molecular Pathways.” Evidence-Based Complementary and Alternative Medicine. Volume 2014 |Article ID 724658 | https://doi.org/10.1155/2014/724658

(2016) Amin F.Majdalawieh and Muneera W.Fayyad.  “Recent advances on the anti-cancer properties of Nigella sativa, a widely used food additive.” Journal of Ayurveda and Integrative Medicine. Volume 7, Issue 3, July–September 2016, Pages 173-180

(2019) Ebrahim M. Yimer, et al.  “Nigella sativa L. (Black Cumin): A Promising Natural Remedy for Wide Range of Illnesses.” Evidence-Based Complementary and Alternative Medicine. Volume 2019 |Article ID 1528635 | https://doi.org/10.1155/2019/1528635

BOSWELLIA

(1999) Jing, Y., S. Nakajo, L. Xia, K. Nakaya, Q. Fang, S. Waxman and R. Han, 1999. “Boswellic acid acetate induces differentiation and apoptosis in leukemia cell lines.” Leukemia Res., 23: 43-50.

(1999) Glaser T, Winter S, Groscurth P et al.  “Boswellic acids and malignant glioma: Induction of apoptosis but no modulation of drug sensitivity.” Br J Cancer. 80:756-765, 1999.

(1999) Hoernlein, R.F., T. Orlikowsky, C. Zehrer, D. Niethammer and E.R. Sailer et al., 1999. “Acetyl-11-keto-beta-boswellic acid induces apoptosis in HL-60 and CCRF-CEM cells and inhibits topoisomerase I.” J. Pharmacol. Exp. Ther., 288: 613-619.

(2000) Huang MT, Badmaev V, Ding Y, Liu Y, Xie JG, Ho CT. “Anti-tumor and anti-carcinogenic activities of triterpenoid, β-boswellic acid.” Biofactors. 2000;13:225-230.

(2000) Winking M, Sarikaya S, Rahmanian A, Jodicke A, Boker DK. “Boswellic acids inhibit glioma growth: a new treatment option?” J Neurooncol. 2000;46:97-103. 

(2002) Hostanska K, Daum G, Saller R. “Cytostatic and apoptosisinducing activity of boswellic acids toward malignant cell lines in vitro.” Anticancer Res. 2002;22:2853-2862. 

(2005) Syrovets, T.; Gschwend, J.E.; Buchele, B.; Laumonnier, Y.; Zugmaier, W.; Genze, F.; Simmet, T. “Inhibition of IkappaB kinase activity by acetyl-boswellic acids promotes apoptosis in androgen-independent PC-3 prostate cancer cells in vitro and in vivo.”  J. Biol. Chem. 2005, 280, 6170–6180. 

(2005) Xia L, Chen D, Han R, Fang Q, Waxman S, Jing Y. “Boswellic acid acetate induces apoptosis through caspase-mediated pathways in myeloid leukemia cells.” Mol Cancer Ther. 2005;4(3):381-388. doi:10.1158/1535-7163.MCT-03-0266.

(2006) Liu, J.J., B. Huang and S.C. Hooi, 2006. “Acetyl-keto-β-boswellic acid inhibits cellular proliferation through a p21-dependent pathway in colon cancer cells.” Br. J. Pharmacol., 148: 1099-1107.

(2007) Bhushan, S., A. Kumar, F. Malik, S.S. Andotra and V.K. Sethi et al., 2007. “A triterpenediol from Boswellia serrata induces apoptosis through both the intrinsic and extrinsic apoptotic pathways in human leukemia HL-60 cells.” Apoptosis, 12: 1911-1926.

(2008) Lu, M.; Xia, L.; Hua, H.; Jing, Y. “Acetyl-keto-beta-boswellic acid induces apoptosis through a death receptor 5-mediated pathway in prostate cancer cells.” Cancer Res. 2008, 68, 1180–1186.

(2008) Yuan, H.Q.; Kong, F.; Wang, X.L.; Young, C.Y.; Hu, X.Y.; Lou, H.X. “Inhibitory effect of acetyl-11-ketobeta-boswellic acid on androgen receptor by interference of Sp1 binding activity in prostate cancer cells.” Biochem. Pharmacol. 2008, 75, 2112–2121.

(2008) Kim HR, Kim, MS, Kwon DY, et al. “Boswellia serrata-induced apoptosis in related with ER stress and calcium release.”  Genes Nutr.  2:371-374, 2008.

(2009) Bhushan S, Malik F, Kumar A, et al. “Activation of p53/p21/PUMA alliance and disruption of PI-3/Akt multimodal targeting of apoptotic signaling cascades in cervical cancer cells by pentacyclic triterpenediol from Boswellia serrata.”  Mol Carcinog. 48:1093-1108, 2009.

(2009) Liu, J.J. and R.D. Duan, 2009. “LY294002 enhances boswellic acid-induced apoptosis in colon cancer cells.” Anticancer Res., 29: 2987-2991.

(2009) Frank MB, Yang Q, Osban J, et al. “Frankincense oil derived from Boswellia carteri induces tumor cell specific cytotoxicity”. BMC Complement Altern Med. 2009;9:6. 

(2009) Pang, X.; Yi, Z.; Zhang, X.; Sung, B.; Qu, W.; Lian, X.; Aggarwal, B.B.; Liu, M. “Acetyl-11-keto-beta-boswellic acid inhibits prostate tumor growth by suppressing vascular endothelial growth factor receptor 2-mediated angiogenesis.” Cancer Res. 2009, 69, 5893–5900.

(2010) Qurishi, Y., A. Hamid, M.A. Zargar, S.K. Singh and A.K. Saxena, 2010. “Potential role of natural molecules in health and disease: Importance of boswellic acid.” J. Med. Plants Res., 4: 2778-2785.

(2011) Suhail MM, Wu W, Cao A, et al. “Boswellia sacra essential oil induces tumor cell-specific apoptosis and suppresses tumor aggressiveness in cultured human breast cancer cells.” BMC Complement Altern Med. 2011;11:129.

(2011) Park, B., B. Sung, V.R. Yadav, S.G. Cho, M. Liu and B.B. Aggarwal, 2011. “Acetyl-11-keto-β-boswellic acid suppresses invasion of pancreatic cancer cells through the downregulation of CXCR4 chemokine receptor expression.” Int. J. Cancer, 129: 23-33.

(2011) Eichhorn, Tolga et al. “Molecular Determinants of the Response of Tumor Cells to Boswellic Acids.” Pharmaceuticals vol. 4,8 1171–1182. 19 Aug. 2011, doi:10.3390/ph4081171

(2011) Kirste S, Treier M, Wehrle SJ, et al. “Boswellia extract acts on cerebral edema in patients irradiated for brain tumors: A prospective, randomized, placebo-controlled, double-blind pilot trial.”  Cancer. 117:3788-3795, 2011.

(2012) Paul, M., 2012. “Chemotaxonomic investigation on resins of the frankincense species Boswellia papyriferaBoswellia serrataBoswellia sacra respectively, Boswellia carterii.” Ph.D. Thesis, Saarland University, Saarbrucken, Saarland, Germany.

(2012) Ni X, Suhail MM, Yang Q, et al. “Frankincense essential oil prepared from hydrodistillation of Boswellia sacra gum resins induces human pancreatic cancer cell death in cultures and in a xenograft murine model.” BMC Complement Altern Med. 2012;12:253. 

(2013) Fung KM, Suhail MM, McClendon B, Woolley CL, Young DG, Lin HK. “Management of basal cell carcinoma of the skin using frankincense (Boswellia sacra) essential oil: a case report.” OA Altern Med. 2013;1:14.

(2013) Qurishi Y, Hamid A, Sharma PR, et al. “NF-ƙB down-regulation and PARP cleavage by novel 3-α-butyryloxy-β-boswellic acid results in cancer cell specific apoptosis and in vivo tumor regression.”  Anticancer Agents Med Chem.  13:777-790, 2013.

(2014) Dozmorov MG, Yang Q, Wu W, et al. “Differential effects of selective frankincense (Ru Xiang) essential oil versus nonselective sandalwood (Tan Xiang) essential oil on cultured bladder cancer cells: a microarray and bioinformatics study.” Chin Med. 2014;9:18.

(2014) Neeta and Harish Dureja, “Role of Boswellic Acids in Cancer Treatment.” Journal of Medical Sciences, 2014:14: 261-269.

(2014) Wang r, Wany Y, Gao Z, et al. “The comparative study of acetyl-11-keto-beta-boswellic acid (AKBA) and aspirin in the prevention of intestinal adenomatous polyposis in APC(Min/+) mice.” Drug Discov Ther. 8:25-32, 2014.

(2015) Togni S, Maramaldi G, Bonetta A, Giacomelli L, Di Pierro F. “Clinical evaluation of safety and efficacy of Boswellia based cream for prevention of adjuvant radiotherapy skin damage in mammary carcinoma: a randomized placebo controlled trial.” Eur Rev Med Pharmacol Sci. 2015;19: 1338-1344.

(2015) Pasta V, Gullo G, Giuliani A, et al. “An association of Boswellia, betaine and myo-inositol (EUmastos) in the treatment of mammographic breast density: A randomized, double-blind study.”  Eur Rev med Pharmacol Sci. 19:4419-4426, 2015.

(2016) Hamidpour R, Hamidpour S, Hamidpour M et al. “Frankincense (Boswellia Species): The Novel Phytotherapy for Drug Targeting in Cancer.” Arch Cancer Res. 2016, 4:1.

(2017) Ranjbarnejad T, Saidijam M, Moradkhani S, et al. “Methanolic extract of Boswellia serrata exhibits anti-cancer activities by targeting microsomal prostaglandin E synthase-1 in human colon cells.” Prostaglandins Other Lipid Mediat. 131:1-8, 2017.

(2018) Huang, M.; Li, A.; Zhao, F.; Xie, X.; Li, K.; Jing, Y.; Liu, D.; Zhao, L. “Design, synthesis and biological evaluation of ring A modified 11-keto-boswellic acid derivatives as Pin1 inhibitors with remarkable anti-prostate cancer activity.” Bioorganic Med. Chem. Lett. 2018, 28, 3187–3193.

(2018) Conti S, Vexler A, Edry-Botzer L, et al. “Combined acetyl-11-ketoboswellic acid and radiation treatment inhibited glioblastoma tumor cells.” PLoS One 13:e0198627, 2018.

(2019) Liu, YQ, et al. Acetyl-11-keto-β-boswellic acid suppresses docetaxel-resistant prostate cancer cells in vitro and in vivo by blocking Akt and Stat3 signaling, thus suppressing chemoresistant stem cell-like properties.”  Acta Pharmacol Sin. 2019 May;40(5):689-698. doi: 10.1038/s41401-018-0157-9. Epub 2018 Aug 31. (colon cancer, malignant glioma, prostate cancer, and leukemia)

CAM MEDICINE

(2011) Michael McCulloch, Michael Broffman, Mark van der Laan, Alan Hubbard, Lawrence Kushi, Alan Kramer, Jin Gao and John M. Colford, Jr.  “Follow-up Data Analyzed With Marginal Structural Models and Propensity Score Methods Lung Cancer Survival With Herbal Medicine and Vitamins in a Whole-Systems Approach : Ten-Year”.  Integrative Cancer Therapies.  2011 10: 260 originally published online 8 August 2011.

L-CARNITINE

(2018) Dinicola S, et al.  “Natural products – alpha-lipoic acid and acetyl-L-carnitine – in the treatment of chemotherapy-induced peripheral neuropathy.” European Review for Medical and Pharmacological Sciences. 2018; 22: 4739-4754.

COQ10

(1994) Lockwood, et al. Partial and complete regression of breast cancer in patients in relation to dosage of coenzyme Q10.” Biochem Biophys Res Commun. 1994 Mar 30;199(3):1504-8. “PMID:7908519 DOI:10.1006/bbrc.1994.1401

(2005) Malik, et al. “Coenzyme Q10 inhibits proliferation of breast cancer cells while stabilizing growth in primary cells in vitro.”  Cancer Res May 1 2005 (65) (9 Supplement) 1384-1385;

(2009) Hertz, et al. “Improved Survival in Patients with Endstage Cancer Treated with Coenzyme Q10 and Other Antioxidants: a Pilot Study.” The Journal of International Medical Research. 2009; 37: 1961 – 1971

(2014) Mathews, et al.  “The effects of coenzyme Q10 on women with breast cancer: a systematic review protocol.” JBI Database of Systematic Reviews & Implementation Reports 2014;12(8) 127 – 144. 

(2016) Iwase, S, et al. “Efficacy and safety of an amino acid jelly containing coenzyme Q10 and L-carnitine in controlling fatigue in breast cancer patients receiving chemotherapy: a multi-institutional, randomized, exploratory trial (JORTC-CAM01).” Support Care Cancer. 2016 Feb;24(2):637-646. doi: 10.1007/s00520-015-2824-4. Epub 2015 Jun 24.

(2017) Tafazoli, A. “Coenzyme Q10 in breast cancer care.” Future Oncol. 2017 May;13(11):1035-1041. doi: 10.2217/fon-2016-0547.

(2017) Jang, et al. Effect of coenzyme Q10 via nitric oxide production and growth arrest of human colon cancer HCT116 cells.” J. Prev. Vet. Med 2017; 41(2): 59-65.

(2018) Coenzyme Q10 PDQ. https://www.ncbi.nlm.nih.gov/books/NBK65890/

(2019) Aksoy, et al.  “An investigation of oxidative stress and coenzyme Q10 levels in patients with head and neck squamous cell carcinomas.” Eur Arch Otorhinolaryngol. 2019 Apr;276(4):1197-1204. doi: 10.1007/s00405-019-05328-5. Epub 2019 Feb 7.

 

CURCUMIN

(2004) Myung-Ju Ahn, et al. “Mechanisms of proapoptotic properties of curcumin in chronic and acute myeloid leukemia.” AACR. April 2004: Vol 64:7 

(2009) Ravindran, Jayaraj et al. “Curcumin and cancer cells: how many ways can curry kill tumor cells selectively?” AAPS journal vol. 11,3 (2009): 495-510. 

(2013) Yu, Jianhua et al. “Curcumin down-regulates DNA methyltransferase 1 and plays an anti-leukemic role in acute myeloid leukemia.” PloS one vol. 8,2 (2013): e55934. doi:10.1371/journal.pone.0055934

(2014) Troselj KG.  “Curcumin in combined cancer therapy.” Current Pharmaceutical Design. 2014;20(42):6682-96.

(2015) Zeng, Yingjian et al. “Curcumin reduces the expression of survivin, leading to enhancement of arsenic trioxide-induced apoptosis in myelodysplastic syndrome and leukemia stem-like cells.” Oncology reports vol. 36,3 (2016): 1233-42.

(2016) Yi Deng, et al. “Molecular Mechanisms of Anti-metastatic Activity of Curcumin.” Anticancer Research. 36: 5639-5648 (2016) doi:10.21873/anticanres.11147.

(2016) Gardane, et al. “Curcumin sensitizes quiescent leukemic cells to antimitotic drug 5-fluorouracil by inducing proliferative responses in them.” J Cancer Metastasis Treat 2016;2:245-52.

(2017) Animesh Kumar, et al.  “Enhanced apoptosis, survivin down-regulation and assisted immunochemotherapy by curcumin loaded amphiphilic mixed micelles for subjugating endometrial cancer.” Nanomedicine: Nanotechnology, Biology and Medicine. Volume 13, Issue 6, August 2017, Pages 1953-1963.

(2017) Zaidi, Abbas et al. “Long-term stabilisation of myeloma with curcumin.” BMJ case reports vol. 2017 bcr2016218148. 16 Apr. 2017, doi:10.1136/bcr-2016-218148

(2018) Sourav Banerjee, et al.  “Ancient drug curcumin impedes 26S proteasome activity by direct inhibition of dual-specificity tyrosine-regulated kinase 2.” Proceedings of the National Academy of Sciences, 2018; 201806797 DOI: 10.1073/pnas.1806797115. Anti-cancer therapy.

(2018) Li, et al. “Evaluation of curcumin, a natural product in turmeric, on Burkitt lymphoma and acute myeloid leukemia cancer stem cell markers.” Future Oncol. 2018 Oct;14(23):2353-2360.

FATS/LIPIDS

(2017) Pabst, Thomas et al. “The plasma lipidome in acute myeloid leukemia at diagnosis in relation to clinical disease features.” BBA clinical vol. 7 105-114. 8 Mar. 2017, doi:10.1016/j.bbacli.2017.03.002

FEBENDAZOLE/MEBENDAZOLE

(2008) Gao, Ping et al. “Unexpected antitumorigenic effect of fenbendazole when combined with supplementary vitamins.” Journal of the American Association for Laboratory Animal Science : JAALAS vol. 47,6 (2008): 37-40.

(2014) Pantziarka Pan, at al. “Repurposing Drugs in Oncology (ReDO)—mebendazole as an anti-cancer agent” ecancer 8 443

(2018) Dogra, Nilambra et al. “Fenbendazole acts as a moderate microtubule destabilizing agent and causes cancer cell death by modulating multiple cellular pathways.” Scientific reports vol. 8,1 11926. 9 Aug. 2018, doi:10.1038/s41598-018-30158-6

(2019) Guerini, et al. Mebendazole as a Candidate for Drug Repurposing in Oncology: An Extensive Review of Current Literature.” Cancers 201911(9), 1284; https://doi.org/10.3390/cancers11091284

(2019) Rushworth, L.K., Hewit, K., Munnings-Tomes, S. et al. “Repurposing screen identifies mebendazole as a clinical candidate to synergise with docetaxel for prostate cancer treatment.” Br J Cancer (2019). https://doi.org/10.1038/s41416-019-0681-5

FLAVONOIDS

(2007) J. J. Raffoul, F. H. Sarkar, and G. G. Hillman. “Radiosensitization of prostate cancer by soy isoflavones.” Current Cancer Drug Targets, vol. 7, no. 8, pp. 759–765, 2007.

(2010) I. U. Ahmad, J. D. Forman, F. H. Sarkar et al. “Soy isoflavones in conjunction with radiation therapy in patients with prostate cancer.” Nutrition and Cancer, vol. 62, no. 7, pp. 996–1000, 2010.

(2011) G. G. Hillman and V. Singh-Gupta. “Soy isoflavones sensitize cancer cells to radiotherapy.” Free Radical Biology & Medicine, vol. 51, no. 2, pp. 289–298, 2011.

(2011) V. Singh-Gupta, M. C. Joiner, L. Runyan et al. “Soy isoflavones augment radiation effect by inhibiting APE1/ref-1 DNA repair activity in non-small cell lung cancer.” Journal of Thoracic Oncology, vol. 6, no. 4, pp. 688–698, 2011.

FLAXSEEDS

(1998) Lin Yan, et al.  “Dietary flaxseed supplementation and experimental metastasis of melanoma cells in mice.” Cancer Letters. Volume 124, Issue 2, 27 February 1998, Pages 181-186

(2005) Dwivedi C, Natarajan K, Matthees DP.  “Chemopreventive effects of dietary flaxseed oil on colon tumor development.” Nutr Cancer. 2005;51(1):52-8.

(2015) Han, J, et al.  “Flax seed oil inhibits metastatic melanoma and reduces lung tumor formation in mice.” Journal BUON. 2015 Nov-Dec;20(6):1546-52.

(2017) Kim, Dong Joon et al. “Herbacetin suppresses cutaneous squamous cell carcinoma and melanoma cell growth by targeting AKT and ODC.” Carcinogenesis vol. 38,11 (2017): 1136-1146.

(2018) Calado, Ana et al. “The Effect of Flaxseed in Breast Cancer: A Literature Review.” Frontiers in nutrition vol. 5 4. 7 Feb. 2018, doi:10.3389/fnut.2018.00004

(2018) Shahira M. Ezzat, et al. “Anticancer potentiality of lignan rich fraction of six Flaxseed cultivars.” Nature Scientific Reports. January 11, 2018.  https://www.nature.com/articles/s41598-017-18944-0 .pdf

FOLATE

(2018) Michelle Claire Rudd, Prof Robert Lea, Dr Jane Alder; University of Central Lancashire. “Folate: Friend or Foe? An Investigation Into the Opposing Roles of Folate in Glioma.”  Neuro-oncology.  Jan 2018.

(2018) Pieroth, Renee et al. “Folate and Its Impact on Cancer Risk.” Current nutrition reports vol. 7,3 (2018): 70-84.

FUCOIDAN (Brown Seaweed)

(2006) Moreau, D., et al. “An extract from the brown alga bifurcaria bifurcata induces irreversible arrest of cell proliferation in a non-small-cell bronchopulmonary carcinoma line.”  J. Appl. Phycol. 2006, 18, 87–93.

(2010) Kim, E.J., et al.  “Fucoidan present in brown algae induces apoptosis of human colon cancer cells.”  BMC Gastroenterol. 2010, 10, 96, doi:10.1186/1471-230X-10-96.

(2011) Boo, H.J., et al. “Fucoidan from undaria pinnatifida induces apoptosis in a549 human lung carcinoma cells.”  Phytother. Res. 2011, 25, 1082–1086.

(2011) Ikeguchi, M., et al.  “Fucoidan reduces the toxicities of chemotherapy for patients with unresectable advanced or recurrent colorectal cancer.” Oncol. Lett. 2011, 2, 319–322. 62. 

(2012) Lv, Y., et al. “Comparison of the effects of marchantin c and fucoidan on sflt-1 and angiogenesis in glioma microenvironment.”  J. Pharm. Pharmacol. 2012, 64, 604–609.

(2013) Kimura, Ryuichiro et al. “Cytotoxic effects of fucoidan nanoparticles against osteosarcoma.” Marine drugs vol. 11,11 4267-78. 30 Oct. 2013, doi:10.3390/md11114267.

(2013) Boo, H.J, et al. “The anticancer effect of fucoidan in pc-3 prostate cancer cells.” Mar. Drugs 2013, 11, 2982–2999.

(2013) Banafa, A.M., et al. “Fucoidan induces g1 phase arrest and apoptosis through caspases-dependent pathway and ros induction in human breast cancer mcf-7 cells.”  J. Huazhong Univ. Sci. Technol. Med. Sci. 2013, 33, 717–724.

(2015) Atashrazm, Farzaneh et al. “Fucoidan and cancer: a multifunctional molecule with anti-tumor potential.” Marine drugs vol. 13,4 2327-46. 14 Apr. 2015, doi:10.3390/md13042327.

(2015) Kalimuthu, S.; Kim, S. “Fucoidan, a sulfated polysaccharides from brown algae as therapeutic target for cancer.” In Handbook of Anticancer Drugs from Marine Origin; Kim, S., Ed.; Springer International Publishing: Cham, Switzerland, 2015; p. 147.

(2015) Wei, et al. “Fucoidan inhibits proliferation of the SKM-1 acute myeloid leukaemia cell line via the activation of apoptotic pathways and production of reactive oxygen species.” Mol Med Rep. 2015 Nov;12(5):6649-55. 

(2016) Atashrazm, et al.  “Fucoidan Suppresses the Growth of Human Acute Promyelocytic Leukemia Cells In Vitro and In Vivo.” J Cell Physiol. 2016 Mar;231(3):688-97.

(2016) Liu, et al.  “Fucoidan inhibits angiogenesis induced by multiple myeloma cells.” Spandidos Pub, Oncology Reports. October 2016. Volume 36 Issue 4 (1963-1972).

(2019) Hsu, Hsien-Yeh, and Pai-An Hwang. “Clinical applications of fucoidan in translational medicine for adjuvant cancer therapy.” Clinical and translational medicine vol. 8,1 15. 1 May. 2019, doi:10.1186/s40169-019-0234-9

GLUCOSE

Rath,Linda. Cancer and Sugar: Is There a Link?” https://www.webmd.com/cancer/features/cancer-sugar-link#1

Heather Christofk, PhD, co-leader of the UCLA Metabolomics Center. “Most Cancers Use Glucose.” https://medschool.ucla.edu/metabolism/cancer-glucose-metabolism

(2015) Granja, et al. “Glucose Addiction in Cancer Therapy: Advances and Drawbacks.”

Curr Drug Metab. 2015;16(3):221-42.

(2016) Yu, et al.  “The sweet trap in tumors: aerobic glycolysis and potential targets for therapy.” Oncotarget. 2016 Jun 21;7(25):38908-38926. doi: 10.18632/oncotarget.7676.

(2017) Santye, Lauren. “Squamous Cell Carcinoma Significantly More Dependent on Sugar Than Other Cancer Types.” https://www.pharmacytimes.com/news/squamous-cell-carcinoma-significantly-more-dependent-on-sugar-than-other-cancer-types. May 31,2017.

(2018) A Breakthrough in the Connection Between Sugar and Lung Cancer.” American Lung Association. https://www.lung.org/about-us/blog/2018/02/sugar-and-lung-cancer.html.  February 12, 2018.

(2018) Bise, et al, “Glucose Metabolism in Cancer.” Adv Exp Med Biol. 2018;1063:3-12. doi: 10.1007/978-3-319-77736-8_1.

(2019) Arthur Dyer, et al.  “Antagonism of Glycolysis and Reductive Carboxylation of Glutamine Potentiates Activity of Oncolytic Adenoviruses in Cancer Cells.” Cancer Res January 15 2019 (79) (2) 331-345; DOI: 10.1158/0008-5472.CAN-18-1326 Published January 2019

GLUTAMINE

(2016) Bajpai, R et al. “Targeting glutamine metabolism in multiple myeloma enhances BIM binding to BCL-2 eliciting synthetic lethality to venetoclax.” Oncogene vol. 35,30 (2016): 3955-64. doi:10.1038/onc.2015.464

(2016) Bolzoni, et al.  “Dependence on glutamine uptake and glutamine addiction characterize myeloma cells: A new attractive target.” Blood 128(5) · June 2016 

(2018) Choi, Yeon-Kyung, and Keun-Gyu Park. “Targeting Glutamine Metabolism for Cancer Treatment.” Biomolecules & therapeutics vol. 26,1 (2018): 19-28. doi:10.4062/biomolther.2017.178

(2019) Jiang, Jie et al. “Starve Cancer Cells of Glutamine: Break the Spell or Make a Hungry Monster?.” Cancers vol. 11,6 804. 11 Jun. 2019, doi:10.3390/cancers11060804

(2019) Grinde, M.T., Hilmarsdottir, B., Tunset, H.M. et al. “Glutamine to proline conversion is associated with response to glutaminase inhibition in breast cancer.” Breast Cancer Res 21, 61 (2019) doi:10.1186/s13058-019-1141-0

GLUTATHIONE

(1986) Midander J, Deschavanne PJ, Debieu D, Malaise EP, Revesz L. “Reduced repair of potentially lethal radiation damage in glutathione synthetase-deficient human fibroblasts after X-irradiation.” Int J Radiat Biol Relat Stud Phys Chem Med. 1986 Mar;49(3):403-13. PMID: 3485589.

(1995) Cascinu S, Cordella L, Del Ferro E, Fronzoni M, Catalano G. “Neuroprotective effect of reduced glutathione on cisplatin-based chemotherapy in advanced gastric cancer: a randomized double-blind placebo-controlled trial.” J Clin Oncol. 1995 Jan;13(1):26-32. PubMed PMID: 7799029.

(1997) Smyth JF, et al. “Glutathione reduces the toxicity and improves quality of life of women diagnosed with ovarian cancer treated with cisplatin: results of a double-blind, randomized trial.” Ann Oncol. 1997 Jun;8(6):569-73. PubMed PMID: 9261526.

(2002) Cascinu S, et al. “Neuroprotective effect of reduced glutathione on oxaliplatin-based chemotherapy in advanced colorectal cancer: a randomized, double-blind, placebo-controlled trial.” J Clin Oncol. 2002 Aug 15;20(16):3478-83. PubMed PMID: 12177109.

(2007) Franco R, Schoneveld OJ, Pappa A, Panayiotidis MI. “The central role of glutathione in the pathophysiology of human diseases.” Arch Physiol Biochem. 2007 Oct-Dec;113(4-5):234-58. Review. PubMed PMID: 18158646.

(2007) King-TehLee, et al. “Glutathione status in the blood and tissues of patients with virus-originated hepatocellular carcinoma.” Clinical Biochemstry. Volume 40, Issue 15, October 2007, Pages 1157-1162.

(2008) Sreeja L, Syamala V, Hariharan S, Syamala VS, Raveendran PB, Sivanandan CD, Madhavan J, Ankathil R. “Glutathione S-transferase M1, T1 and P1 polymorphisms: susceptibility and outcome in lung cancer patients.” J Exp Ther Oncol. 2008;7(1):73-85. PubMed PMID: 18472644.

(2009) Pujari G, Berni A, Palitti F, Chatterjee A. “Influence of glutathione levels on radiation-induced chromosomal DNA damage and repair in human peripheral lymphocytes.” Mutat Res. 2009 JunJul;677(1-2):109-10. PMID: 19386243

(2009) Milla P, et al. “Administration of reduced glutathione in FOLFOX4 adjuvant treatment for colorectal cancer: effect on oxaliplatin pharmacokinetics, Pt-DNA adduct formation, and neurotoxicity.” Anticancer Drugs. 2009 Jun;20(5):396-402. PubMed PMID: 19287306.

(2010) Kilburn L, et al.  “Glutathione S-transferase polymorphisms are associated with survival in anaplastic glioma patients.” Cancer. 2010 Feb 24. [Epub ahead of print] PubMed PMID: 20187096.

(2011) Angel L. Ortega, at al. “Glutathione in Cancer Cell Death.”  Cancers 2011, 3, 1285-1310; doi:10.3390/cancers3011285.

(2013) Nicola Traverso, et al., “Role of Glutathione in Cancer Progression and Chemoresistance (AML).” Oxidative Medicine and Cellular Longevity, vol. 2013, Article ID 972913, 10 pages, 2013. doi:10.1155/2013/972913.

(2014) Mitchell, Gaynor.  Complete Remission of Widely Metastatic Melanoma: A Case Report.” Cancer Strategies Journal.  Spring 2014. 

(2017) Zhou, et al. “Detection of Glutathione in Oral Squamous Cell Carcinoma Cells With a Fluorescent Probe During the Course of Oxidative Stress and Apoptosis.” J Oral Maxillofac Surg. 2017 Jan;75(1):223.e1-223.e10. doi: 10.1016/j.joms.2016.08.010. Epub 2016 Aug 24.

(2018) Bansal, Ankita and Simon, Celeste.  “Glutathione metabolism in cancer progression and treatment resistance.”  J Cell Biol (2018) 217 (7): 2291–2298.

GREEN TEA EXTRACT

(2002) Sartippour, Shao, Heber, et al. “Green Tea Inhibits VEGF Induction in Human Breast Cancer Cells.”  J. Nutr. 2002; 132: 2307-2311.

(2004) Kay. Green Tea Component Destroys Leukemia Cells.”  Blood Mar.2, 2004 On-line edition.

(2004) Yokoyama, Noguchi, Nakao, et al. “The Tea Polyphenol, (-)-Epigallocatechin Gallate Effects on Growth, Apoptosis, and Telomerase Activity in Cervical Cell Lines.” Gynecol. Oncol. 2004; 92 (1): 197-204.

(2004) Mittal, Pate, Wylie, et al.  “EGCG Down-Regulates Telomerase in Human Breast Carcinoma MCF-7 Cells, Leading to Suppression of Cell Viability and Induction of Apoptosis.” Int. J. Oncol. 2004; 24 (3): 703-710.

(2004) Pezzato, Sartor, Dell’Aica, et al. Prostate Carcinoma and Green Tea: PSA-Triggered Basement Membrane Degradation and MMP-2 Activation are Inhibited by (-) Epigallocatechin-3-Gallate.”  Int. J. Cancer 2004; 112 (5): 787-792.

 

(2005) Shimizu, Deguchi , Joe, et al. “EGCG Inhibits Activation of HER3 and Expression of Cyclooxygenase-2 in Human Colon Cancer Cells.”  J. Exp. Ther. Oncol. 2005; 5 (1): 69-78.

(2006) Shanafelt, Lee, Call, et al. Clinical Effects of Oral Green Tea Extracts in Four Patients with Low Grade B-Cell Malignancies.”  Leuk. Res2006; 30: 707-712.

(2008) Ergruder, Namusui, Sozener, et al. Effects of Aqueous Green Tea Extract on Activities of DNA Turn-over Enzymes in Cancerous and Non-Cancerous Human Gastric and Colon Tissues.”  Altern. Ther. Health Med.  2008; 14 (3): 30-33. 

(2008) Shankar, Ganapathy, Hingorani & Srivastava. EGCG Inhibits Growth, Invasion, Angiogenesis and Metastasis of Pancreatic Cancer.”  Front. Biosci. 2008; 13: 440-452.

(2009) Angelo, Laura S, and Razelle Kurzrock. “Turmeric and green tea: a recipe for the treatment of B-chronic lymphocytic leukemia.” Clinical cancer research : an official journal of the American Association for Cancer Research vol. 15,4 (2009): 1123-5. doi:10.1158/1078-0432.CCR-08-2791.

(2009) Park, Lee, Hwang, et al. Green Tea Catechin Controls Apoptosis in Colon Cancer Cells by Attenuation of H2O2-Stimulated COX-2 Expression via the AMPK Signaling Pathway at Low-Dose H2O2.Ann. N.Y. Acad. Sci. 2009; 1171: 538-544.  [re: NAC blocks EGCG effects].

(2010) Shanafelt, Call, Zent, et al. Phase II Trial of Daily, Oral Green Tea Extract in Patients with Asymptomatic, Rai Stage 0-II Chronic Lymphocytic Leukemia (CLL).”  J. Clin. Oncol. 2010; 28 (Suppl.): S7. Abstract 6522 ASCO 2010.

(2017) Y. Zhang, S.-X. Wang, J.-W. Ma et al. “EGCG inhibits properties of glioma stem-like cells and synergizes with temozolomide through downregulation of P-glycoprotein inhibition.” Journal of Neuro-Oncology, vol. 121, no. 1, pp. 41–52, 2015.

(2018) Ugai T, Matsuo K, Sawada N, et al. “Coffee and green tea consumption and subsequent risk of acute myeloid leukemia and myelodysplastic syndromes in Japan.” Int J Cancer. 2018;142(6):1130-1138. doi:10.1002/ijc.31135

(2020) Filippini T, Malavolti M, Borrelli F, Izzo AA, Fairweather-Tait SJ, Horneber M, Vinceti M. “Green tea (Camellia sinensis) for the prevention of cancer. Cochrane Database Syst Rev. 2020 Mar 2;3(3):CD005004. doi: 10.1002/14651858.CD005004.pub3.PMID: 32118296

(2020) Almatroodi SA, Almatroudi A, Khan AA, Alhumaydhi FA, Alsahli MA, Rahmani AH. “Potential Therapeutic Targets of Epigallocatechin Gallate (EGCG), the Most Abundant Catechin in Green Tea, and Its Role in the Therapy of Various Types of Cancer.” Molecules. 2020;25(14):3146. Published 2020 Jul 9. doi:10.3390/molecules25143146

HYPERBARIC OXYGEN THERAPY (HBOT)

(1966) Van den Brenak, et al.RESULTS FROM TOURNIQUET ANOXIA AND HYPERBARIC OXYGEN TECHNIQUES COMBINED WITH MEGAVOLTAGE TREATMENT OF SARCOMAS Of bone and soft tissue.” From the Radiobiological and Hyperbaric Oxygen Research Units, Cancer Institute Board, Melbourne, Australia. March 1966, Pgs 760-766.

(2003) Perte, et al.  Hyperbaric oxygen as a chemotherapy adjuvant in the treatment of metastatic lung tumors in a rat model.”  The Journal of Thoracic and Cardiovascular Surgery

Volume 125, Issue 1, January 2003, Pages 85-95

(2009) Tie Fu Liu, Jiaozhong Cai, Denise M. Gibo and Waldemar Debinski.  Reoxygenation of Hypoxic Glioblastoma Multiforme Cells Potentiates the Killing Effect of an Interleukin-13-Based Cytotoxin.” Clinical Cancer Research. DOI: 10.1158/1078-0432.CCR-08-2151 Published January 2009.

(2012) Moen, Ingrid, and Linda E B Stuhr. “Hyperbaric oxygen therapy and cancer–a review.” Targeted oncology vol. 7,4 (2012): 233-42. doi:10.1007/s11523-012-0233-x

(2013) Ogawa K, et al.  “Old but new methods in radiation oncology: hyperbaric oxygen therapy.” Int J Clin Oncol. 2013 Jun;18(3):364-70. doi: 10.1007/s10147-013-0537-6. Epub 2013 Mar 5.  https://www.ncbi.nlm.nih.gov/pubmed/23463521

(2014) Peng, Hai-Shan, et al. “Synergistic inhibitory effect of hyperbaric oxygen combined with sorafenib on hepatoma cells.”  PloS one vol. 9,6 e100814. 23 Jun. 2014, doi:10.1371/journal.pone.0100814.  https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4067386/

(2015) Bennett MH, Feldmeier J, Hampson NB, Smee R, Milross C. “Hyperbaric oxygen therapy for late radiation tissue injury.”  Cochrane Database of Systematic Reviews 2016, Issue 4. Art. No.: CD005005. DOI: 10.1002/14651858.CD005005.pub4

(2016) Stępień, Katarzyna et al. “Hyperbaric oxygen as an adjunctive therapy in treatment of malignancies, including brain tumours.” Medical oncology (Northwood, London, England) vol. 33,9 (2016): 101. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4971045/

(2016) Yahara, Katsuya et al. “Radiotherapy using IMRT boosts after hyperbaric oxygen therapy with chemotherapy for glioblastoma.” Journal of radiation research vol. 58,3 (2016): 351-356.https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5440883/

(2017) Singh, et al. “Hyperbaric Oxygen Therapy for Radiation-Related CNS Necrosis in Children with Brain Tumors: A Single Institution Experience.” International Journal of Radiation Oncology  Biology  Physics, 3360.

(2018) Huang, Lei et al. “Hyperbaric oxygen therapy as adjunctive strategy in treatment of glioblastoma multiforme.” Medical gas research vol. 8,1 24-28. 18 Apr. 2018, doi:10.4103/2045-9912.229600https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5937300/

(2018) “NCI Awards Omar Aljitawi, M.D., $1M to Study Hyperbaric Oxygen Therapy for Cancer.” University of Rochester. Multiple Myeloma. https://www.urmc.rochester.edu/news/story/5334/nci-awards-omar-aljitawi-m.d.-1m-to-study-hyperbaric-oxygen-therapy-for-cancer.aspx

(2018) Bennett MH, Feldmeier J, Smee R, Milross C. “Hyperbaric oxygenation for tumour sensitisation to radiotherapy.” Cochrane Database of Systematic Reviews 2018, Issue 4. Art. No.: CD005007. DOI: 10.1002/14651858.CD005007.pub4

HYPERTHERMIA

Hyperthermia Cancer Institute– Santa Monica, CA.  (888)580-5900.  

Texas Oncology– multiple locations.

UCSF Health- Dr. I-Chow (Joe) Hsu, Dr Sue Yom. San Francisco. (415) 353-7175.

Integrated Health Clinic– Fort Langley, British Columbia, Sarah Soles, MSc, ND. (604) 888-8325

Hufeland Klinik- Bad Mergentheim, Germany.

Medical Center Germany– Frankfurt.  Holistic Care.

Sanoviv Medical Institute– Baja California, Mexico

The Grace Gawler Institute– Australia, New Zealand

(2002) Ohno S, et al.  “Hyperthermia for rectal cancer.” Surgery. 2002 Jan;131(1 Suppl):S121-7.

(2004) Hildebrandt B, et al. “Treatment of locally recurrent rectal cancer with special focus on regional pelvic hyperthermia.” Onkologie. 2004 Oct;27(5):506-11.

(2012) Lin CY, et al. “Ultrasound sonication with microbubbles disrupts blood vessels and enhances tumor treatments of anticancer nanodrug.” Int J Nanomedicine. 2012;7:2143-52. doi: 10.2147/IJN.S29514. Epub 2012 Apr 24.

(2012) Lee, Doo Yun, et al.  “Complete Remission of SCLC with Chemotherapy and Oncothermia (Case Report)”. Oncothermia Journal 5:43-51 (2012)

(2013) Gao, et al.  “Water bath hyperthermia reduces stemness of colon cancer cells.” Clinical Biochemistry. Volume 46, Issues 16–17, November 2013, Pages 1747-1750

(2014) Gianfranco Baronzio, et al. “A brief overview of hyperthermia in cancer treatment.” J Integr Oncol 2014, 3:1.  

(2014) Wu, Sheng-Kai et al. “Short-time focused ultrasound hyperthermia enhances liposomal doxorubicin delivery and antitumor efficacy for brain metastasis of breast cancer.” International journal of nanomedicine vol. 9 4485-94. 19 Sep. 2014, doi:10.2147/IJN.S68347

(2015) Aryal M, et al. “Enhancement in blood-tumor barrier permeability and delivery of liposomal doxorubicin using focused ultrasound and microbubbles: evaluation during tumor progression in a rat glioma model.” Phys Med Biol. 2015 Mar 21;60(6):2511-27. doi: 10.1088/0031-9155/60/6/2511. Epub 2015 Mar 6.

(2015) PLOS ONE Staff. “Correction: First clinical experience of intra-operative high intensity focused ultrasound in patients with colorectal liver metastases: a phase I-IIa study.” PLoS One. 2015 Mar 30;10(3):e0123751. doi: 10.1371/journal.pone.0123751. eCollection 2015.

(2016) Chu W, Staruch RM, Pichardo S, Tillander M, Köhler MO, Huang Y, Ylihautala M, McGuffin M, Czarnota G, Hynynen K. “Magnetic Resonance-Guided High-Intensity Focused Ultrasound Hyperthermia for Recurrent Rectal Cancer: MR Thermometry Evaluation and Preclinical Validation.” Int J Radiat Oncol Biol Phys. 2016 Jul 15;95(4):1259-67. doi: 10.1016/j.ijrobp.2016.03.019. Epub 2016 Mar 24.

(2016) VanOsdol J, Ektate K, Ramasamy S, Maple D, Collins W, Malayer J, Ranjan A. “Sequential HIFU heating and nanobubble encapsulation provide efficient drug penetration from stealth and temperature sensitive liposomes in colon cancer.” J Control Release. 2017 Feb 10;247:55-63. doi: 10.1016/j.jconrel.2016.12.033. Epub 2016 Dec 30.

(2017) Wu sk, et al. “Pulsed-wave low-dose ultrasound hyperthermia selectively enhances nanodrug delivery and improves antitumor efficacy for brain metastasis of breast cancer.” Ultrason Sonochem. 2017 May;36:198-205. doi: 10.1016/j.ultsonch.2016.11.033. Epub 2016 Nov 28.

(2017) Fahimeh Faghihi Moghadam, et al. “A BRIEF REVIEW OF HYPERTHERMIA AS A NEOADJUVANT THERAPY METHOD RELATED TO CANCER TREATMENT”.  Journal of Cellular Immunotherapy 3 (2017) 2-30. Department of Medical Physics, Faculty of Medical Sciences, Shahid Beheshti University of Medical Science, Tehran, Iran; Department of Radiology, Faculty of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran;  Institute for Hyperthermia Research, Partner of the Marien Hospital Herne, Hospital of the Ruhr-University, Bochum, Germany

(2017) VanOsdol J, Ektate K, Ramasamy S, Maples D, Collins W, Malayer J, Ranjan A. “Sequential HIFU heating and nanobubble encapsulation provide efficient drug penetration from stealth and temperature sensitive liposomes in colon cancer.” J Control Release. 2017 Feb 10;247:55-63. doi: 10.1016/j.jconrel.2016.12.033. Epub 2016 Dec 30.

(2017) Boissenot T, Bordat A, Larrat B, Varna M, Chacun H, Paci A, Poinsignon V, Fattal E, Tsapis N. “Ultrasound-induced mild hyperthermia improves the anticancer efficacy of both Taxol® and paclitaxel-loaded nanocapsules.” J Control Release. 2017 Oct 28;264:219-227. doi: 10.1016/j.jconrel.2017.08.041. Epub 2017 Sep 1.

(2017) Yagawa Y, et al. “Cancer immunity and therapy using hyperthermia with immunotherapy, radiotherapy, chemotherapy, and surgery.” J Cancer Metastasis Treat 2017;3:218-230

(2017) Maeda M, Muragaki Y, Okamoto J, Yoshizawa S, Abe N, Nakamoto H, Ishii H, Kawabata K, Umemura S, Nishiyama N, Kataoka K, Iseki H. “Sonodynamic Therapy Based on Combined Use of Low Dose Administration of Epirubicin-Incorporating Drug Delivery System and Focused Ultrasound.” Ultrasound Med Biol. 2017 Oct;43(10):2295-2301. doi: 10.1016/j.ultrasmedbio.2017.06.003. Epub 2017 Jul 10.

(2018) Ektate K, Munteanu MC, Ashar H, Malayer J, Ranjan A. “Chemo-immunotherapy of colon cancer with focused ultrasound and Salmonella-laden temperature sensitive liposomes (thermobots).” Sci Rep. 2018 Aug 30;8(1):13062. doi: 10.1038/s41598-018-30106-4.

(2018) Byun YH, et al. “Local recurrence of brain metastasis reduced by intra-operative hyperthermia treatment.” Int J Hyperthermia. 2019 Jan 1;35(1):168-175. doi: 10.1080/02656736.2018.1488004. Epub 2018 Oct 8. 

(2018) Vancsik T, et a;. “Modulated electro-hyperhtermia induced loco-regional and systemic tumor destruction in colorectal cancer allografts.”  J Cancer 2018; 9(1):41-53. Doi:10.7150/jca.21520

(2019) Chiang CF, et al. Pulsed-wave Ultrasound Hyperthermia Enhanced Nanodrug Delivery Combined with Chloroquine Exerts Effective Antitumor Response and Postpones Recurrence.” Sci Rep. 2019 Aug 28;9(1):12448. doi: 10.1038/s41598-019-47345-8.

(2019) Liu, Zichuan. “Clinical effects of high frequency hyperthermia-assisted irinotecan chemotherapy on patients with middle and advanced colorectal cancer and its safety assessment.” Oncology letters vol. 17,1 (2019): 215-220. doi:10.3892/ol.2018.9574.

(2019) Mantso T, et al. Hyperthermia Suppresses Post – In Vitro Proliferation and Tumor Growth in Murine Malignant Melanoma and Colon Carcinoma.” ANTICANCER RESEARCH 39: 2307-2315 (2019) doi:10.21873/anticanres.13347

(2019) Li M, Wan G, Yu H, Xiong W. “High-intensity focused ultrasound inhibits invasion and metastasis of colon cancer cells by enhancing microRNA-124-mediated suppression of STAT3.” FEBS Open Bio. 2019 Jun;9(6):1128-1136. doi: 10.1002/2211-5463.12642. Epub 2019 May 4.

(2019) Ma B, Liu X, Yu Z. “The effect of high intensity focused ultrasound on the treatment of liver cancer and patients’ immunity.” Cancer Biomark. 2019;24(1):85-90. doi: 10.3233/CBM-181822.

ICD- 10 codes: DDY58ZZ Hyperthermia of colon, DDY78ZZ Hyperthermia of rectum

INDOLE-3-CARBINOL

(1999) Cover, Hsieh, Cram, et al. Indole-3-Carbinol and Tamoxifen Cooperate to Arrest the Cell Cycle of MCF-7 Human Breast Cancer Cells.”  Cancer Res. 1999; 59 (6):1244-1251.

(2001) Chinni, Li, Upadhyay, et al.  “Indole-3-Carbinol (I3C) Induced Cell Growth Inhibition, G1 Cell Cycle Arrest and Apoptosis in Prostate Cancer Cells.”  Oncogene 2001.

(2003) Zhang, Hsu, Kinseth, et al.  Indole-3-Carbinol Induces a G1 Cell Cycle Arrest and Inhibits Prostate-Specific Antigen Production in Human LNCaP Prostate Carcinoma Cells.” Cancer 2003; 98 (11): 2511-2520.

 

(2004) Sarkar. Indole-3-Carbinol and Prostate Cancer.”  J. Nutr. 2004; 134 (12 Suppl): 3493S-3498S.

(2005) Takada, Yasunari et al. “Indole-3-carbinol suppresses NF-kappaB and IkappaBalpha kinase activation, causing inhibition of expression of NF-kappaB-regulated antiapoptotic and metastatic gene products and enhancement of apoptosis in myeloid and leukemia cells.” Blood vol. 106,2 (2005): 641-9. doi:10.1182/blood-2004-12-4589

(2005) Hsu, Zhang, Dev, et al. “Indole-3-Carbinol Inhibition of Androgen Receptor Expression and Down-regulation of Androgen Responsiveness in Human Prostate Cancer Cells.”  Carcinogenesis 2005.

(2005) Garikpaty, et al.  Anti-Carcinogenic and Anti-Metastatic Properties of Indole-3-Carbinol in Prostate Cancer.”  Oncol. Rep.  2005; 13 (1): 89-93.

(2005) Li, Chinni & Sarkhar.  Selective Growth Regulatory and Pro-apoptotic Effects of DIM is Mediated by AKT and NF-kappaB Pathways in Prostate Cancer Cells.”  Front. Biosci. 2005; 10: 236-243.

(2006) Kim, Jeong, Moon, et al.  Indole-3-Carbinol Enhances Ultraviolet-B-induced Apoptosis by Sensitizing Human Melanoma Cells.” Cell. Mol. Life Sci. 2006; 63 (22): 2661-2668.

(2010) Nguyen, et al. “1-Benzyl-indole-3-carbinol is a novel indole-3-carbinol derivative with significantly enhanced potency of anti-proliferative and anti-estrogenic properties in human breast cancer cells.” Chemico-biological interactions 186(3):255-66 · August 2010.

(2013) Bai LY, Weng JR, Chiu CF, et al. “OSU-A9, an indole-3-carbinol derivative, induces cytotoxicity in acute myeloid leukemia through reactive oxygen species-mediated apoptosis.” Biochem Pharmacol. 2013;86:1430–1440.

(2014), Tin, et al.  “Essential role of the cancer stem/progenitor cell marker nucleostemin for indole-3-carbinol anti-proliferative responsiveness in human breast cancer cells.” BMC Biology volume 12, Article number: 72 (2014).

(2014) Tavasoli B, Ebrahimi S, Manafi R, et al.  “Indole-3-carbinol induces G1 cell cycle arrest in pre-B acute lymphoblastic leukemia cell line.”  J Arak Univ Med Sci. 2014;17:30–40.

(2016) Perez-Chacon G, Martinez-Laperche C, Rebolleda N, et al. “Indole-3-carbinol synergizes with and restores fludarabine sensitivity in chronic lymphocytic leukemia cells irrespective of p53 activity and treatment resistances.” Clin Cancer Res. 2016;22:134–145.

(2017) Mohammadi, et al. Indole-3-carbinol induces G1 cell cycle arrest and apoptosis through aryl hydrocarbon receptor in THP-1 monocytic cell line.” (AML) Journal of Receptors and Signal Transduction.  Volume 37, 2017 – Issue 5.

  • (2017) Safa M, et al. “Indole-3-carbinol induces apoptosis of chronic myelogenous leukemia cells through suppression of STAT5 and Akt signaling pathways.” Tumor Biology 39(6):101042831770576. June 2017.

LOW DOSE NALTREXONE

See Alpha-lipoic section for further combination treatment.

(2007) Berkson, B, et al.  “Reversal of Signs and Symptoms of a B-Cell Lymphoma in a Patient Using Only Low-Dose Naltrexone.” INTEGRATIVE CANCER THERAPIES 6(3); 2007 pp. 293-296

(2011) “Low-dose naltrexone (LDN): Tricking the body to heal itself.” Science Daily. September 2, 2011. https://www.sciencedaily.com/releases/2011/09/110902133047.htm

(2018) Miskoff, Jeffrey A, and Moiuz Chaudhri. “Low Dose Naltrexone and Lung Cancer: A Case Report and Discussion.” Cureus vol. 10,7 e2924. 5 Jul. 2018, doi:10.7759/cureus.2924

(2018) Li Z, You Y, Griffin N, Feng J, Shan F. “Low-dose naltrexone (LDN): a promising treatment in immune-related diseases and cancer therapy.” Int Immunopharmacol. 2018 Aug;61:178-184.

LYCOPENE

(2007) Keren Hirsch, et al.” Lycopene and other carotenoids inhibit estrogenic activity of 17-estradiol and genistein in cancer cells.” Breast Cancer Research and Treatment. September 2007: 104(2):221-30.

(2008) van Breemen, Richard B, and Natasa Pajkovic. “Multitargeted therapy of cancer by lycopene.” Cancer letters vol. 269,2 (2008): 339-51. doi:10.1016/j.canlet.2008.05.016

(2011) Chih-Min Yang. “Lycopene inhibits the proliferation of androgen-dependent human prostate tumor cells through activation of PPARγ-LXRα-ABCA1 pathway.” The Journal of Nutritional Biochemistry. February 2011: 23(1):8-17. 

(2017) Nina Pauline Holzapfel. “Investigation of the Potential Anticancer Effects of Lycopene in Tissue Engineered in Vitro and in Vivo Models.” Doctoral research project. Queensland University of Technology, School of Biomedical Sciences, Faculty of Health. 2016/2017. https://eprints.qut.edu.au/107571/1/Nina%20Pauline_Holzapfel_Thesis.pdf

(2019) Jiang LN, et al. “Lycopene exerts anti-inflammatory effect to inhibit prostate cancer progression.” Asian J Androl 2019;21: 80-5 

MELATONIN

(2006) Büyükavci M, et al. “Melatonin cytotoxicity in human leukemia cells: relation with its pro-oxidant effect.” Fundam Clin Pharmacol. 2006 Feb;20(1):73-9.

(2011) Büyükavci M, et al.  “Effect of melatonin on the cytotoxicity of chemotherapeutic drugs in human leukemia cells.” In Vivo. 2011 May-Jun;25(3):405-9.

(2013) Martin V, et al. “Melatonin-induced methylation of the ABCG2/BCRP promoter as a novel mechanism to overcome multidrug resistance in brain tumour stem cells.” Br J Cancer 2013;108:2005-12

(2014) Mihandoost E, Shirazi A, Mahdavi SR, Aliasgharzadeh A. Can melatonin help us in radiation oncology treatments?Biomed Res Int. 2014;2014:578137. doi:10.1155/2014/578137.

(2017) Najafi, M et al. “The melatonin immunomodulatory actions in radiotherapy.”  Biophysical reviews vol. 9,2 (2017): 139-148. doi:10.1007/s12551-017-0256-8.

(2017) Li, Ya et al. “Melatonin for the prevention and treatment of cancer.” Oncotarget vol. 8,24 (2017): 39896-39921. doi:10.18632/oncotarget.16379.

(2018) Favero, et al.  “Promising Antineoplastic Actions of Melatonin.” Front. Pharmacol., 16 October 2018.

(2018) Farhood, et al. “Melatonin as an adjuvant in radiotherapy for radioprotection and radiosensitization.”  Clin Transl Oncol. 2019 Mar;21(3):268-279. doi: 10.1007/s12094-018-1934-0.

METHYLSULFONYLMETHAN (MSM)

(2010) Caron, Joan McIntyre et al. “Methyl sulfone induces loss of metastatic properties and reemergence of normal phenotypes in a metastatic cloudman S-91 (M3) murine melanoma cell line.” PloS one vol. 5,8 e11788. 4 Aug. 2010, doi:10.1371/journal.pone.0011788

(2012) Jafari N, et al.  “Cytotoxicity of methylsulfonylmethane on gastrointestinal (AGS, HepG2, and KEYSE-30) cancer cell lines.”  J Gastrointestinal Cancer. 2012 Sep;43(3):420-5. doi: 10.1007/s12029-011-9291-z.

(2012) Lim, Eun Joung et al. “Methylsulfonylmethane suppresses breast cancer growth by down-regulating STAT3 and STAT5b pathways.” PloS one vol. 7,4 (2012): e33361.

(2014) Joung YH, et al.  “Combination of AG490, a Jak2 inhibitor, and methylsulfonylmethane synergistically suppresses bladder tumor growth via the Jak2/STAT3 pathway.”   Int J Oncol. 2014 Mar;44(3):883-95. doi: 10.3892/ijo.2014.2250. Epub 2014 Jan 8.

(2014) Kim JH, et al.  “Methylsulfonylmethane suppresses hepatic tumor development through activation of apoptosis.”  World J Hepatol. 2014 Feb 27;6(2):98-106. doi: 10.4254/wjh.v6.i2.98.

(2015) SPN, et al. “The combination of methylsulfonylmethane and tamoxifen inhibits the Jak2/STAT5b pathway and synergistically inhibits tumor growth and metastasis in ER-positive breast cancer xenografts.”  BMC Cancer. 2015 Jun 19;15:474. doi: 10.1186/s12885-015-1445-0.

(2016) Kang DY, etal.  “Methylsulfonylmethane inhibits HER2 expression through STAT5b in breast cancer cells.”  Int J Oncol. 2016 Feb;48(2):836-42. doi: 10.3892/ijo.2015.3277. Epub 2015 Dec 7.

(2016) Karahay AZ, et al.  “Methylsulfonylmethane Induces p53 Independent Apoptosis in HCT-116 Colon Cancer Cells.”  Int J Mol Sci. 2016 Jul 15;17(7). pii: E1123. doi: 10.3390/ijms17071123.

MISTLETOE

(2009) Ostermann, Thomas et al. “Survival of cancer patients treated with mistletoe extract (Iscador): a systematic literature review.” BMC cancer vol. 9 451. 18 Dec. 2009, doi:10.1186/1471-2407-9-451

MODIFIED CITRUS PECTIN

(2011) Ramachandran, Cheppail et al. “Activation of human T-helper/inducer cell, T-cytotoxic cell, B-cell, and natural killer (NK)-cells and induction of natural killer cell activity against K562 chronic myeloid leukemia cells with modified citrus pectin.” BMC complementary and alternative medicine vol. 11 59. 4 Aug. 2011, doi:10.1186/1472-6882-11-59

(2013) Leclere Lionel, Van Cutsem Pierre, Michiels Carine. “Anti-cancer activities of pH- or heat-modified pectin.”  Frontiers in Pharmacology. 4:128, 2013. URL=https://www.frontiersin.org/article/10.3389/fphar.2013.00128

(2018) Contj S, et al. Modified Citrus Pectin as a Potential Sensitizer for Radiotherapy in Prostate Cancer.”  Integrative Cancer Therapies. Volume: 17 issue: 4, page(s): 1225-1234

(2019) Eliaz, Isaac, and Avraham Raz. “Pleiotropic Effects of Modified Citrus Pectin.” Nutrients vol. 11,11 2619. 1 Nov. 2019, doi:10.3390/nu11112619

MUSHROOMS

(2006) Muller, et al. “Ganoderma lucidum causes apoptosis in leukemia, lymphoma and multiple myeloma cells.” (Reishi) Leuk Res. 2006 Jul;30(7):841-8. Epub 2006 Jan 19. (Reishi)

(2011) Hetland, Geir et al. “The Mushroom Agaricus blazei Murill Elicits Medicinal Effects on Tumor, Infection, Allergy, and Inflammation through Its Modulation of Innate Immunity and Amelioration of Th1/Th2 Imbalance and Inflammation.” Advances in pharmacological sciences vol. 2011 (2011): 157015. doi:10.1155/2011/157015. (Brazilian rain forest)

(2014) Patel, Seema, and Arun Goyal. “Recent developments in mushrooms as anti-cancer therapeutics: a review.” 3 Biotech vol. 2,1 (2012): 1-15. doi:10.1007/s13205-011-0036-2.

(2017) Tangen, et al. “Cytotoxic Effect on Human Myeloma Cells and Leukemic Cells by the Agaricus blazei Murill Based Mushroom Extract, Andosan.” BioMed Research International
Volume 2017, Article ID 2059825, 7 pages.

NICLOSAMIDE ETHANOLAMINE

 

 

NITRIC OXIDE

(2015) Sudjit Luanpitpong and Pithi Chanvorachote.  “Nitric Oxide and Aggressive Behavior of Lung Cancer Cells “. Anticancer Research. September 2015 vol. 35no. 9 4585-4592

(2016) Vahora, Huzefa et al. “The Potential Role of Nitric Oxide in Halting Cancer Progression Through Chemoprevention” Journal of cancer prevention vol. 21,1 (2016): 1-12.

OREGANO 

(2009) Savini I, Arnone R, Catani MV, Avigliano L. “Origanum vulgare induces apoptosis in human colon cancer caco2 cells.” Nutr Cancer. 24 Volume 3, Number 3 • May 2014 • www.gahmj.com GLOBAL ADVANCES IN HEALTH AND MEDICINE Original Research 2009;61(3):381-9

(2015) H.S. Elshafie, M.F. Armentano, M. Carmosino, S.A. Bufo, V. De Feo, I. Camele.  “Cytotoxic activity of Origanum vulgare L. on hepatocellular carcinoma cell line HepG2 and evaluation of its biological activity.”  Molecules, 22 (9) (2015), p. E1435

(2017) K.R. Begnini, et al.  “Composition and antiproliferative effect of essential oil of Origanum vulgare against tumor cell lines.” J. Med. Food., 17(10)(2017), pp. 1-5

(2018) Sri Renukadevi Balusamy, Haribalan Perumalsamy, Md. Amdad Huq, Balamuralikrishnan Balasubramanian.  “Anti-proliferative activity of Origanum vulgare inhibited lipogenesis and induced mitochondrial mediated apoptosis in human stomach cancer cell lines.”  Biomedicine & Pharmacotherapy, Volume 108, 2018, Pages 1835-1844.

OZONE THERAPY

(1980) Sweet F, Kao MS, Lee SC, Hagar WL, Sweet WE. “Ozone selectively inhibits growth of human cancer cells.” Science. 1980 Aug 22;209(4459):931-3. doi: 10.1126/science.7403859. PMID: 7403859.

(2004) Clavo B, Ruiz A, Lloret M, López L, Suárez G, Macías D, Rodríguez V, Hernández MA, Martín-Oliva R, Quintero S, Cuyás JM, Robaina F.  “Adjuvant Ozonetherapy in Advanced Head and Neck Tumors: A Comparative Study.” Evid Based Complement Alternat Med. 2004 Dec;1(3):321-325. doi: 10.1093/ecam/neh038. Epub 2004 Oct 16. PMID: 15841266; PMCID: PMC538509.

(2005) Bocci V, Larini A, Micheli V. “Restoration of normoxia by ozone therapy may control neoplastic growth: a review and a working hypothesis.”  J Altern Complement Med. 2005 Apr;11(2):257-65. doi: 10.1089/acm.2005.11.257. PMID: 15865491.

(2014) Li, X., Wang, X., Zhang, M. et al. “Quercetin Potentiates the Antitumor Activity of Rituximab in Diffuse Large B-Cell Lymphoma by Inhibiting STAT3 Pathway.” Cell Biochem Biophys 70, 1357–1362 (2014). https://doi.org/10.1007/s12013-014-0064-8

(2015) Kızıltan HŞ, Bayir AG, Yucesan G, Eris AH, İdin K, Karatoprak C, Aydin T, Akcakaya A, Mayadagli A. “Medical ozone and radiotherapy in a peritoneal, Erlich-ascites, tumor-cell model.” Altern Ther Health Med. 2015 Mar-Apr;21(2):24-9. PMID: 25830277.

(2017) Luongo, et al. “Review: Possible Therapeutic Effects of Ozone Mixture on Hypoxia in Tumor Development.” Anticancer Research, February 2017 37 (2) 425-435.

(2018) Dogan R, Hafız AM, Kiziltan HS, Yenigun A, Buyukpinarbaslili N, Eris AH, Ozturan O. “Effectiveness of radiotherapy+ozone on tumoral tissue and survival in tongue cancer rat model.” Auris Nasus Larynx. 2018 Feb;45(1):128-134. doi: 10.1016/j.anl.2017.03.017. Epub 2017 Apr 6. PMID: 28390748.

(2018) Clavo, Bernardino et al. “Ozone Therapy as Adjuvant for Cancer Treatment: Is Further Research Warranted?” Evidence-based complementary and alternative medicine : eCAM vol. 2018 7931849. 9 Sep. 2018, doi:10.1155/2018/7931849

(2019) Bernardino Clavo, et al. “Modulation of Oxidative Stress by Ozone Therapy in the Prevention and Treatment of Chemotherapy-Induced Toxicity: Review and Prospects.” Antioxidants 2019, 8, 588; doi:10.3390/antiox8120588

QUERCETIN

(2011) Wang K, et al. “Quercetin induces protective autophagy in gastric cancer cells: involvement of Akt-mTOR- and hypoxia-induced factor 1α-mediated signaling”. Autophagy. 2011;7(9):966–978. doi: 10.4161/auto.7.9.15863.

(2012) Jacquemin, Guillaume et al. “Quercetin-mediated Mcl-1 and survivin downregulation restores TRAIL-induced apoptosis in non-Hodgkin’s lymphoma B cells.” Haematologica vol. 97,1 (2012): 38-46. doi:10.3324/haematol.2011.046466

(2013) Dneg, et al. “Effects of quercetin on the proliferation of breast cancer cells and expression of survivin in vitro.” EXPERIMENTAL AND THERAPEUTIC MEDICINE  6:  1155-1158,  2013.

(2014) Maso V, et al. “Multitarget effects of quercetin in leukemia.” Cancer Prev Res (Phila) 2014;7(12):1240–1250. doi: 10.1158/1940-6207.CAPR-13-0383

(2017) Granato, Marisa, et al. Quercetin induces apoptosis and autophagy in primary effusion lymphoma cells by inhibiting PI3K/AKT/mTOR and STAT3 signaling pathways.” J Nutr Biochem. 2017 Mar;41:124-136. doi: 10.1016/j.jnutbio.2016.12.011.

(2018) Wang, Rong et al. “Quercetin Inhibits Breast Cancer Stem Cells via Downregulation of Aldehyde Dehydrogenase 1A1 (ALDH1A1), Chemokine Receptor Type 4 (CXCR4), Mucin 1 (MUC1), and Epithelial Cell Adhesion Molecule (EpCAM).” Medical science monitor : international medical journal of experimental and clinical research vol. 24 412-420. 21 Jan. 2018, doi:10.12659/msm.908022.

(2018) Nezami MA, et al. “Proof of concept in a case study of glioblastoma multiforme successfully treated with IV Quercetin in combination with leading edge gamma knife and standard treatments.”  J Cancer Ther 2018;9:522-8.

(2018) Alvarez, Marisa Claudia et al. “The polyphenol quercetin induces cell death in leukemia by targeting epigenetic regulators of pro-apoptotic genes.” Clinical epigenetics vol. 10,1 139. 8 Nov. 2018, doi:10.1186/s13148-018-0563-3

(2018) Calgarotto, A.K., Maso, V., Junior, G.C.F. et al.  “Antitumor activities of Quercetin and Green Tea in xenografts of human leukemia HL60 cells.” Sci Rep 8, 3459 (2018). https://doi.org/10.1038/s41598-018-21516-5

(2019) Naimi A, Entezari A, Hagh MF, Hassanzadeh A, Saraei R, Solali S. “Quercetin sensitizes human myeloid leukemia KG-1 cells against TRAIL-induced apoptosis.” J Cell Physiol. 2019;234(8):13233-13241. doi:10.1002/jcp.27995

(2020) Vafadar, A., Shabaninejad, Z., Movahedpour, A. et al. “Quercetin and cancer: new insights into its therapeutic effects on ovarian cancer cells.” Cell Biosci 10, 32 (2020). https://doi.org/10.1186/s13578-020-00397-0

RESVERATROL

(2004) Simone Fulda and Klaus-Michael Debati. “Sensitization for anticancer drug-induced apoptosis by the chemopreventive agent resveratrol.” Oncogene (2004) 23, 6702–6711.

(2007) C. Gill, S. E. Walsh, C. Morrissey, J. M. Fitzpatrick, and R. W. G. Watson. “Resveratrol sensitizes androgen independent prostate cancer cells to death-receptor mediated apoptosis through multiple mechanisms.” The Prostate, vol. 67, no. 15, pp. 1641– 1653, 2007.

(2009) Nguyen, et al. “Results of a phase I pilot clinical trial examining the effect of plant-derived resveratrol and grape powder on Wnt pathway target gene expression in colonic mucosa and colon cancer.” Cancer Management and Research 2009:1 25–37.

(2010) K. R. Patel, V. A. Brown, D. J. L. Jones et al. “Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients.” Cancer Research, vol. 70, no. 19, pp. 7392–7399, 2010.

(2011) ] J. Ryu, B. M. Ku, Y. K. Lee et al. “Resveratrol reduces tnf-alphainduced u373mg human glioma cell invasion through regulating nf-kappab activation and upa/upar expression.” Anticancer Research, vol. 31, pp. 4223–4230, 2011.

(2012) Meher U. Nessa, et al. “Combinations of Resveratrol, Cisplatin and Oxaliplatin Applied to Human Ovarian Cancer Cells.” Anticancer Research. 32: 53-60 (2012)

(2012) (2012) Vetvicka V, Vetvickova J. “Combination of glucan, resveratrol and vitamin C demonstrates strong anti-tumor potential.” Anticancer Res. 2012;32:81–87.

(2014) Chottanapund S, et al.  “Anti-aromatase effect of resveratrol and melatonin on hormonal positive breast cancer cells co-cultured with breast adipose fibroblasts.”  Toxicol In Vitro. 2014 Oct;28(7):1215-21. doi: 10.1016/j.tiv.2014.05.015. Epub 2014 Jun 12.

(2016) Zhong, L-X et al. “Resveratrol and STAT inhibitor enhance autophagy in ovarian cancer cells.” Cell death discovery vol. 2 15071. 25 Jan. 2016, doi:10.1038/cddiscovery.2015.71

(2017) Andreani, et al. “Resveratrol fuels HER2 and ERα-positive breast cancer behaving as proteasome inhibitor.” AGING 2017, Vol 9.

(2017) C. Cilibrasi, G. Riva, G. Romano et al. “Resveratrol impairs glioma stem cells proliferation and motility by modulating the wnt signaling pathway.” PLoS ONE, vol. 12, no. 1, Article ID e0169854, 2017.

(2018) Liu Y, et al. “Resveratrol inhibits the proliferation and induces the apoptosis in ovarian cancer cells via inhibiting glycolysis and targeting AMPK/mTOR signaling pathway.” J Cell Biochem. 2018 Jul;119(7):6162-6172. doi: 10.1002/jcb.26822. Epub 2018 Apr 16.

 (2018) Stefan Poschner, et al.  “Resveratrol Inhibits Key Steps of Steroid Metabolism in a Human Estrogen-Receptor Positive Breast Cancer Model: Impact on Cellular Proliferation.” Frontiers in Pharmacology. 10 July 2018. https://doi.org/10.3389/fphar.2018.00742

SELENIUM

(2010) Muecke, et al.  “Multicenter, phase 3 trial comparing selenium supplementation with observation in gynecologic radiation oncology.” Int J Radiat Oncol Biol Phys. 2010 Nov 1;78(3):828-35. doi: 10.1016/j.ijrobp.2009.08.013. Epub 2010 Feb 3.

(2016) Cai, Xianlei et al. “Selenium Exposure and Cancer Risk: an Updated Meta-analysis and Meta-regression.” Scientific reports vol. 6 19213. 20 Jan. 2016, doi:10.1038/srep19213.

SULFORPHANE

(2012) Abbaoui, Besma et al. “Inhibition of bladder cancer by broccoli isothiocyanates sulforaphane and erucin: characterization, metabolism, and interconversion.” Molecular nutrition & food research vol. 56,11 (2012): 1675-87.

(2014) Bergantin, Elisa et al. “Sulforaphane induces apoptosis in rhabdomyosarcoma and restores TRAIL-sensitivity in the aggressive alveolar subtype leading to tumor elimination in mice.” Cancer biology & therapy vol. 15,9 (2014): 1219-25.

(2014) Jo, G. H., et al. “Sulforaphane induces apoptosis in T24 human urinary bladder cancer cells through a reactive oxygen species-mediated mitochondrial pathway: The involvement of endoplasmic reticulum stress and the Nrf2 signaling pathway.” International Journal of Oncology 45.4 (2014): 1497-1506.

(2017) Leone, Andrew et al. “Sulforaphane for the chemoprevention of bladder cancer: molecular mechanism targeted approach.” Oncotarget vol. 8,21 (2017): 35412-35424.

(2018) Lei Huang, et al. “Sulforaphane inhibits human bladder cancer cell invasion by reversing epithelial-to-mesenchymal transition via directly targeting microRNA-200c/ZEB1 axis.”  Journal of Functional Foods. Volume 41, February 2018, Pages 118-126.

(2018) Sato S, et al. “Sulforaphane Inhibits Liver Cancer Cell Growth and Angiogenesis.” iMed Pub Journals. 2018. Vol 6. No 4:23. DOI: 10.21767/2254-6081.100189

VITAMIN B12

(2018) Fanidi A, Et al. “Is high vitamin B12 status a cause of lung cancer?” Int J Cancer. 2018 Nov 29. doi: 10.1002/ijc.32033

VITAMIN C

Linus Pauling Institute.  Oregon State University.

https://www.cancer.gov/about-cancer/treatment/cam/hp/vitamin-c-pdq#_16

https://www.medicalnewstoday.com/articles/319017.php#1

https://now.uiowa.edu/2017/01/why-high-dose-vitamin-c-kills-cancer-cells#.WJo_O1qD2kY.twitter

https://www.cell.com/cancer-cell/fulltext/S1535-6108(17)30062-4

University of Kansas Medical Center Integrative Medicine.  Clinical Trials high dose vitamin C.

University of Wisconsin, School of Medicine and Public Health.  High-Dose Vitamin C (PDQ®): Integrative, alternative, and complementary therapies – Health Professional Information [NCI]

(1975) Cameron E, et al. The orthomolecular treatment of cancer. III. Reticulum cell sarcoma: double complete regression induced by high-dose ascorbic acid therapy.” Chemico-biological Interactions. 1975 Nov;11(5):387-93.

(1978) Cameron E, Pauling L. Supplemental ascorbate in the supportive treatment of cancer: reevaluation of prolongation of survival times in terminal human cancer.” Proceedings of the National academy of Science of the USA. 1978 Sep;75(9):4538-42.

(1996) Kurbacher, et al. “Ascorbic acid (vitamin C) improves the antineoplastic activity of doxorubicin, cisplatin, and paclitaxel in human breast carcinoma cells in vitro.” Cancer letters. 103. 183-9. 10.1016/0304-3835(96)04212-7.

(2000) Andrew G, Bowie2 , O’Neill LAJ. “Vitamin C Inhibits NF-kB Activation by TNF Via the Activation of p38 Mitogen-Activated Protein Kinase.” The J Immunol. 2000, 165: 7180-7188.

(2001) Casciari JJ, Riordan NH. “Cytotoxicity of ascorbate, lipoic acid, and other antioxidants in hollow fiber in vitro tumors.” Br J Cancer. 2001, 84: 1544-1550. 10.1054/bjoc.2001.1814.

(2001) Khaw KT, et al.  “Relation between plasma ascorbic acid and mortality in men and women in EPIC-Norfolk prospective study: a prospective population study. European Prospective Investigation into Cancer and Nutrition.”  Lancet. 2001 Mar 3;357(9257):657-63.

(2002) Bowie AG, Carcamo JM, Pedraza A, Borquez-Qjeda O, Golde DW. “Vitamin C suppresses TNFa-induced NFkB activation by inhibiting IkαB phosphorilation.” Biochemistry. 2002, 41: 12995-13002. 10.1021/bi0263210.

(2004) Padayatty SJ, Sun H, Wang Y, et al. “Vitamin C pharmacokinetics: implications for oral and intravenous use.” Ann Intern Med 140 (7): 533-7, 2004.

(2004) Park, et al. “L-ascorbic acid induces apoptosis in acute myeloid leukemia cells via hydrogen peroxide-mediated mechanisms.” The International Journal of Biochemisby & Cell Biology 36 (2004) 2180-2195.

(2004) Park, et al. “L-Ascorbic Acid Represses Constitutive Activation of NF-KB and COX-2 Expression in Human Acute Myeloid Leukemia, HL-60.” Journal of Cellular Biochemistry, 2004, 93(2):257-270.

(2005) Levine M, Espey MG, Chen Q.  “Losing and finding a way at C: new promise for pharmacologic ascorbate in cancer treatment.”  Free Radic Biol Med. 2009 Jul 1;47(1):27-9. doi: 10.1016/j.freeradbiomed.2009.04.001. Epub 2009 Apr 8.

(2005) Chen Q, Espey MG, Krishna MC. “Pharmacologic ascorbic acid concentrations selectively kill cancer cells: action as a pro-drug to deliver hydrogen peroxide to tissues.” Proc Natl Acad Sci U S A. 2005, 102: 13604-13609. 10.1073/pnas.0506390102.

(2006) Sebastian J Paddayatty, Hugh D Riordan, Stephen M Hewitt, Arie Katz, L John Hoffer, Mark Levine. “Intravenously Administered Vitamin C as Cancer Therapy: Three Cases.”  CMAJ: March 28, 2006: 174(7) 937 (B-cell lymphoma, renal cell carcinoma, bladder cancer)

(2007) Yeom CH, Jung GC, Song KJ. “Changes of terminal cancer patients’ health-related quality of life after high dose vitamin C administration.” J Korean Med Sci 22 (1): 7-11, 2007. 

(2007) Duconge J, Miranda-Massari JR, González MJ, Taylor PR, Riordan HD, Riordan NH, Casciari JJ, Alliston K.  “Vitamin C pharmacokinetics after continuous infusion in a patient with prostate cancer.” Ann Pharmacother. 2007, 41 (6): 1082-1083. 10.1345/aph.1H654.

(2008) Duconge J, Miranda-Massari JR, Gonzalez MJ, Jackson JA, Warnock W, Riordan NH. Pharmacokinetics of vitamin C: insights into the oral and intravenous administration of ascorbate.” P R Health Sci J. 2008 Mar;27(1):7-19.

(2008) Mikirova NA, Ichim TE, Riordan NH. “Anti-angiogenic effect of high doses of ascorbic acid.” J Transl Med. 2008 Sep 12;6:50. doi: 10.1186/1479-5876-6-50.

(2008) Cameron E, Pauling L. “Supplemental ascorbate in the supportive treatment of cancer: Prolongation of survival times in terminal human cancer.” Proc. Natl. Acad. Sci. USA. 1976, 73: 3685-3689. 10.1073/pnas.73.10.3685.corbate. P R Health Sci J. 2008, 27 (1): 7-19. Review

(2008) Hofer, et al. “Phase I clinical trial of i.v. ascorbic acid in advanced malignancy.” Annals of Oncology. 2008 Nov;19(11):1969-74. doi: 10.1093/annonc/mdn377. Epub 2008 Jun 9.

(2009) Levine M, Espey MG, Chen Q. “Losing and finding a way at C: new promise for pharmacologic ascorbate in cancer treatment.” Free Radic Biol Med. 2009 Jul 1;47(1):27-9. doi: 10.1016/j.freeradbiomed.2009.04.001. Epub 2009 Apr 8.

(2012) Monti DA, Mitchell E, Bazzan AJ, et al. “Phase I evaluation of intravenous ascorbic acid in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer.” PLoS One 7 (1): e29794, 2012.

(2009) Ohno, et al.  “High-dose Vitamin C (Ascorbic Acid) Therapy in the Treatment of Patients with Advanced Cancer.” Anticancer Research March 2009 vol. 29 no. 3 809-815.

(2010) Li W, Wu JX, Tu YY. “Synergistic effects of tea polyphenols and ascorbic acid on human lung adenocarcinoma SPC-A-1 cells.” J Zhejiang Univ Sci B. 2010;11:458–464.

(2011) Vollbracht C, Schneider B, Leendert V, et al. “Intravenous vitamin C administration improves quality of life in breast cancer patients during chemo-/radiotherapy and aftercare: results of a retrospective, multicentre, epidemiological cohort study in Germany.” In Vivo 25 (6): 983-90, 2011 Nov-Dec.

(2011) Nakano, et al. “Delayed treatment with vitamin C and N-acetyl-L-cysteine protects Schwann cells without compromising the anti-myeloma activity of bortezomib.” Int J Hematol. 2011 Jun;93(6):727-735. doi: 10.1007/s12185-011-0850-7. Epub 2011 Apr 28.

(2012) Klingelhoeffer C, Kammerer U, Koospal M, et al. “Natural resistance to ascorbic acid induced oxidative stress is mainly mediated by catalase activity in human cancer cells and catalase-silencing sensitizes to oxidative stress.” BMC Complement Altern Med. 2012;12:61.

(2012) Monit, et al. “Phase I evaluation of intravenous ascorbic acid in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer.” PLoS One. 2012;7(1):e29794. doi: 10.1371/journal.pone.0029794. Epub 2012 Jan 17.

(2012) Mikirova, Nina et al. “Effect of high-dose intravenous vitamin C on inflammation in cancer patients.” Journal of translational medicine vol. 10 189. 11 Sep. 2012, doi:10.1186/1479-5876-10-189

(2012) Chen P, et al. Pharmacological ascorbate induces cytotoxicity in prostate cancer cells through ATP depletion and induction of autophagy.” Anticancer Drugs. 2012 Apr;23(4):437-44. 

(2012) Vetvicka V, Vetvickova J. “Combination of glucan, resveratrol and vitamin C demonstrates strong anti-tumor potential.” Anticancer Res. 2012;32:81–87.

(2013) Riordan Clinic Research Institute. “The Riordan IVC Protocol for Adjunctive Cancer Care Intravenous Ascorbate as a Chemotherapeutic and Biological Response Modifying Agent.”  February 2013. file:///C:/Users/krisr06/Documents/Clinic%20Business/IV.IMJ%20Therapy/IV%20HDVC%20protocol%20cancer%202013.pdf

(2013) Stephenson CM, Levin RD, Spector T, Lis CG. “Phase I clinical trial to evaluate the safety, tolerability, and pharmacokinetics of high-dose intravenous ascorbic acid in patients with advanced cancer.” Cancer Chemother Pharmacol. 2013;72(1):139-146.

(2013) Welsh JL, Wagner BA, van’t Erve TJ, et al. “Pharmacological ascorbate with gemcitabine for the control of metastatic and node-positive pancreatic cancer (PACMAN): results from a phase I clinical trial.” Cancer Chemother Pharmacol 71 (3): 765-75, 2013.

(2013) Lam, Tram Kim et al. “Prediagnostic plasma vitamin C and risk of gastric adenocarcinoma and esophageal squamous cell carcinoma in a Chinese population” American journal of clinical nutrition vol. 98,5 (2013): 1289-97.

(2013) Viviana Ulloa, etal. “Human choroid plexus papilloma cells efficiently transport glucose and vitamin C.” Journal of Neurochemistry. 127: 403-414, 2013.

(2013) Alexander B, Fishman AI, Eshghi M, Choudhury M, Konno S. “Induction of cell death in renal cell carcinoma with combination of D-fraction and vitamin C.” Integr Cancer Ther. 2013;12:442–448.

(2014) Ma Y, Chapman J, Levine M, et al. “High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy.” Sci Transl Med 6 (222): 222ra18, 2014.

(2014) Riordan H, Riordan N, et al. “The Riordan intravenous vitamin C (IVC) protocol for adjunctive cancer care: IVC as a chemotherapeutic and biological response modifying agent.” In: “The orthomolecular treatment of chronic disease”, edited by AW Saul, Basic Health Publications, Inc. 2014, 750-766.

(2014) Carr, Anitra C et al. “The effect of intravenous vitamin C on cancer- and chemotherapy-related fatigue and quality of life.” Frontiers in oncology vol. 4 283. 16 Oct. 2014, doi:10.3389/fonc.2014.00283

(2104) Fritz, H., Flower, G., Weeks, L., Cooley, K., Callachan, M., McGowan, J. Seely, D. “Intravenous Vitamin C and Cancer: A Systematic Review.” Integrative Cancer Therapies13(4), 280–300.

(2015) Jocelyn Kaisar.  “Vitamin C kills tumor cells with hard-to-treat mutation.” Science.  November 5, 2015. doi:10.1126/science.aad7397

(2015) Michael J. Gonzalez NMD, DSc, PhD, FANMA, FACN,  Jorge R. Miranda-Massari PharmD, Jorge Duconge PhD,  Miguel J. Berdiel MD.  Increasing the Effectiveness of Intravenous Vitamin C as an Anticancer Agent.  Journal of Orthomolecular Medicine Volume 32, Number 1, 2017

(2015) Hoffer, L John et al. “High-dose intravenous vitamin C combined with cytotoxic chemotherapy in patients with advanced cancer: a phase I-II clinical trial.” PloS one vol. 10,4 e0120228. 7 Apr. 2015, doi:10.1371/journal.pone.0120228

(2015) Yun, J. Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH.” Science 2015, 350, 1391–1396.

(2016) Nina Mikirova, Neil Riordan, Joseph Casciari.  “Modulation of Cytokines in Cancer Patients by Intravenous Ascorbate Therapy.” Medical Science Monitor. 2016; 22:14-25

(2016) Lauren Rouleau, Anil Noronha Antony, Sara Bisetto, Andrew Newberg, Cataldo Doria, Mark Levine, Daniel A. Monti, and Jan B. Hoek  “Synergistic effects of ascorbate and sorafenib in hepatocellular carcinoma: New insights into ascorbate cytotoxicity.” Free Radic Biol Med . 2016 June ; 95: 308–322. doi:10.1016/j.freeradbiomed.2016.03.031.

(2017) University of Iowa Health Care. “Why High Dose Vitamin C Kills Cancer Cells.”  Science Daily.  January 9, 2017. file:///C:/Users/krisr06/Documents/Clinic%20Business/IV.IMJ%20Therapy/Why%20high-dose%20vitamin%20C%20kills%20cancer%20cells_%20Science%20Daily%202017.html

(2017) Schoenfeld, Joshua D. et al. “O2 and H2O2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate.”  Cancer Cell, Volume 31 , Issue 4 , 487 – 500.e8 

(2017) Jennifer Brown. High-dose vitamin C proves safe and well-tolerated in brain and lung cancer trials.  Study identifies flaws in cancer cell metabolism that make high-dose vitamin C toxic to tumor cells.  University of Iowa.

(2017) “High doses of vitamin C to improve cancer treatment passes human safety trial.”  Science Daily, Cell Press. March 30, 2017.

(2017) Xia, Jiliang et al.  “Multiple Myeloma Tumor Cells are Selectively Killed by Pharmacologically-dosed Ascorbic Acid.” EBioMedicine, Volume 18 , 41 – 49.

(2017) Brickner, et al.  “Abstract CT061: Novel safety of high dose ascorbic acid and induction chemotherapy for high risk pancreatic cancer.” AACR Publications. 10.1158/1538-7445.AM2017-CT061 Published July 2017

(2017) Cimmino, Luisa, et al. “Restoration of TET2 Function Blocks Aberrant Self-Renewal and Leukemia Progression.” Cell. Volume 170, Issue 6, P1079-1095.E20, September 07, 2017. (High dose Vitamin C.  

(2017) Gonzalez, et al. “High Dose IV Vitamin C and Metastatic Breast Cancer: A Case Report.” J Orthmol Med. 32(6)

(2017) Polireddy, et al. High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study.” Sci Rep. 2017 Dec 7;7(1):17188. doi: 10.1038/s41598-017-17568-8.

(2017) Agathocleous, et al. Ascorbate regulates haematopoietic stem cell function and leukaemogenesis.” Nature. 2017 Sep 28;549(7673):476-481. doi: 10.1038/nature23876. Epub 2017 Aug 21.

(2017) Violet, et al. “Pharmacologic Ascorbate in Myeloma Treatment: Doses Matter.” EBioMedicine 2017, 18, 9–10

(2017) Shenoy, et al. “Upregulation of TET activity with ascorbic acid induces epigenetic modulation of lymphoma cells.”  Blood Cancer J. 2017, 7, e587. 

(2017) Roa, Francisco J et al. “Therapeutic Use of Vitamin C in Cancer: Physiological Considerations.” Frontiers in pharmacology vol. 11 211. 3 Mar. 2020, doi:10.3389/fphar.2020.00211

(2017) Mastrangelo D, Massai L, Fioritoni G, et al. “High Doses of Vitamin C and Leukaemia: In Vitro Update.” Advanced Leuk Res Treat. 1(1):5-19, 2017. 

(2017) Clinical Trial Phase 3. “IV ascorbic acid in advanced gastric cancer.”  https://clinicaltrials.gov/ct2/show/NCT03015675.

(2017) University of Iowa Health Care. “Why high-dose vitamin C kills cancer cells: Low levels of catalase enzyme make cancer cells vulnerable to high-dose vitamin C.”  Science Daily.  January 9, 2017. https://www.sciencedaily.com/releases/2017/01/170109134014.htm.

(2017) University of Iowa Health Care. “High-dose vitamin C proves safe and well-tolerated in brain and lung cancer trials.” March 30, 2017. https://now.uiowa.edu/2017/03/high-dose-vitamin-c-proves-safe-and-well-tolerated-brain-and-lung-cancer-trials

(2017) Schoenfeld et al.  “O2 ,– and H2O2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate.” Cancer Cell 31, 487–500 April 10, 2017

(2017) Magdelana Kegel.  “High-dose vitamin C may be effective against lymphoma, other blood cancers, say researchers.”  Lymphoma News Today. August 21, 2017 https://lymphomanewstoday.com/2017/08/21/high-dose-vitamin-c-seen-to-fight-lymphoma-other-blood-cancers/

(2018) Michael Greger. “The vitamin C study that started it all.” Nutritionfacts.org. https://nutritionfacts.org/2018/06/14/the-vitamin-c-cancer-study-that-started-it-all/.  Non-Hodgkins Lymphoma. 

(2018) Jeanne A. Drisko, Oscar K. Serrano, Lisa R. Spruce, Qi Chen and Mark Levine.  Treatment of pancreatic cancer with intravenous vitamin C: a case report.  Anti-Cancer Drugs 2018, 29:373–379 

(2018) Klimant, E., Wright, H., Rubin, D., Seely, D., & Markman, M. (2018). Intravenous vitamin C in the supportive care of cancer patients: a review and rational approach. Current Oncology25(2), 139–148. http://doi.org/10.3747/co.25.3790

(2018) Sant DW, Mustafi S, Gustafson CB, Chen J, Slingerland JM, Wang G. Vitamin C promotes apoptosis in breast cancer cells by increasing TRAIL expression. Scientific Reports. 2018;8:5306. doi:10.1038/s41598-018-23714-7.

(2018) El Halabi, Ibrahim et al. “Ascorbic Acid in Colon Cancer: From the Basic to the Clinical Applications” International journal of molecular sciences vol. 19,9 2752. 13 Sep. 2018, doi:10.3390/ijms19092752

(2018) Baillie, Nicola et al. “The Use of Intravenous Vitamin C as a Supportive Therapy for a Patient with Glioblastoma Multiforme.” Antioxidants (Basel, Switzerland) vol. 7,9 115. 30 Aug. 2018, doi:10.3390/antiox7090115

(2018) Yuen Chuen Fong Raymond, et al.  “High-Dose Vitamin C Helps Prevent Recurrence of Stage IV Ovarian Cancer: A Case Report.” International Society for Orthomolecular Medicine. Vol. 33, No. 4.

(2018) Foster, et al. “Intravenous Vitamin C Administration Improved Blood Cell Counts and Health-Related Quality of Life of Patient with History of Relapsed Acute Myeloid Leukaemia.” Antioxidants 2018, 7, 92; doi:10.3390/antiox7070092.

(2018) Carr, Cook.  “Intravenous Vitamin C for Cancer Therapy – Identifying the Current Gaps in Our Knowledge.” Front. Physiol., 23 August 2018 | https://doi.org/10.3389/fphys.2018.01182

(2018) Lv, H., Wang, C., Fang, T. et al.  “Vitamin C preferentially kills cancer stem cells in hepatocellular carcinoma via SVCT-2.” npj Precision Onc 2, 1 (2018) doi:10.1038/s41698-017-0044-8

(2018) Schoenfeld, Joshua D et al. “Pharmacological Ascorbate as a Means of Sensitizing Cancer Cells to Radio-Chemotherapy While Protecting Normal Tissue.” Seminars in radiation oncology vol. 29,1 (2019): 25-32. doi:10.1016/j.semradonc.2018.10.006

(2018) Mingay, M. et al. “Vitamin C-induced epigenomic remodeling in IDH1 mutant acute myeloid leukaemia.” Leukemia 32, 11–20 (2018).

(2018) Pires, Ana S et al. “Ascorbic Acid Chemosensitizes Colorectal Cancer Cells and Synergistically Inhibits Tumor Growth.” Frontiers in physiology vol. 9 911. 23 Jul. 2018, doi:10.3389/fphys.2018.00911

(2019) Blaszczak, Wiktoria et al. “Vitamin C as a Modulator of the Response to Cancer Therapy.” Molecules (Basel, Switzerland) vol. 24,3 453. 28 Jan. 2019, doi:10.3390/molecules24030453

(2019) Gonzalez, et al. “Increasing the Effectiveness of Intravenous Vitamin C as an Anticancer Agent.” International Society for Orthomolecular Medicine. Volume 34, Number 2, 2019.

(2019) Lee, et al. Effect of High-dose Vitamin C Combined With Anti-cancer Treatment on Breast Cancer Cells.” Anticancer Res. 2019 Feb;39(2):751-758. doi: 10.21873/anticanres.13172.

(2019) Gonzalez, et al. “High dose IV Vitamin C and Breast Cancer: A case report.” International Society for Orthomolecular Medicine. 32(6). 2019.https://isom.ca/article/high-dose-iv-vitamin-c-metastatic-breast-cancer-case-report/

(2019) Pena E, et al. “Increased expression of mitochondrial sodium-coupled ascorbic acid transporter-2 (mitSVCT2) as a central feature in breast cancer.” Free Radic Biol Med. 2019 May 1;135:283-292. doi: 10.1016/j.freeradbiomed.2019.03.015. Epub 2019 Mar 19.

(2019) Pawlowska, Elzbieta et al. “Pro- and Antioxidant Effects of Vitamin C in Cancer in correspondence to Its Dietary and Pharmacological Concentrations.” Oxidative medicine and cellular longevity vol. 2019 7286737. 24 Dec. 2019, doi:10.1155/2019/7286737

(2019) van Gorkom, Gwendolyn N Y et al. “The Effect of Vitamin C (Ascorbic Acid) in the Treatment of Patients with Cancer: A Systematic Review.” Nutrients vol. 11,5 977. 28 Apr. 2019, doi:10.3390/nu11050977

(2019) Das, A.B., Kakadia, P.M., Wojcik, D. et al. “Clinical remission following ascorbate treatment in a case of acute myeloid leukemia with mutations in TET2 and WT1.” Blood Cancer J. 9, 82 (2019). https://doi.org/10.1038/s41408-019-0242-4

(2020) Roa, Francisco J et al. “Therapeutic Use of Vitamin C in Cancer: Physiological Considerations.” Frontiers in pharmacology vol. 11 211. 3 Mar. 2020, doi:10.3389/fphar.2020.00211

(2020) Choi YK, Kang JI, Han S, et al. “L-Ascorbic Acid Inhibits Breast Cancer Growth by Inducing IRE/JNK/CHOP-Related Endoplasmic Reticulum Stress-Mediated p62/SQSTM1 Accumulation in the Nucleus.” Nutrients. 2020;12(5):1351. Published 2020 May 8. doi:10.3390/nu12051351

VITAMIN D

(2004) Holick MF. Vitamin D: importance in the prevention of cancers, type 1 diabetes, heart disease, and osteoporosis. Am J Clin Nutr. 2004; 79:362-71.

(2006) Garland, C. F., Garland, F. C., Gorham, E. D., Lipkin, M., Newmark, H., Mohr, S. B., & Holick, M. F. (2006). The Role of Vitamin D in Cancer Prevention. American Journal of Public Health96(2), 252–261. http://doi.org/10.2105/AJPH.2004.045260

(2007) Wu K, Feskanich D, Fuchs CS, Willett WC, Hollis BW, Giovannucci EL. A nested case control study of plasma 25-hydroxyvitamin D concentrations and risk of colorectal cancer. J Natl Cancer Inst. 2007; 99:1120-9.

(2007) Garland CF, Gorham ED, Mohr SB, et al. Vitamin D and prevention of breast cancer: pooled analysis. J Steroid Biochem Mol Biol. 2007; 103:708-11.

(2007) Gorham ED, Garland CF, Garland FC, et al. Optimal vitamin D status for colorectal cancer prevention: a quantitative meta analysis. Am J Prev Med. 2007; 32:210-6.

(2008) Joyce Maalouf, Mona Nabulsi, Reinhold Vieth, Samantha Kimball, Rola El-Rassi, Ziyad Mahfoud, and Ghada El-Hajj Fuleihan. Short term and long term safety of weekly high dose vitamin D3 supplementation in school childrenJ. Clin. Endocrinol. Metab., Published April 29, 2008 DOI: 10.1210/jc.2007-2530

*(2009) Garland CF, Gorham ED, Mohr SB, Garland FC. Vitamin D for cancer prevention: global perspective. Ann Epidemiol. 2009; 19:468-83.

(2009) Yin L, Grandi N, Raum E, Haug U, Arndt V, Brenner H. Meta-analysis: longitudinal studies of serum vitamin D and colorectal cancer risk.  Aliment Pharmacol Ther. 2009; 30:113-25.

(2011) Takako Araki, Michael F. Holick, Bianca D. Alfonso, Esti Charlap, Carla M. Romero, Dahlia Rizk, Lisa G. Newman; Vitamin D Intoxication with Severe Hypercalcemia due to Manufacturing and Labeling Errors of Two Dietary Supplements Made in the United States, The Journal of Clinical Endocrinology & Metabolism, Volume 96, Issue 12, 1 December 2011, Pages 3603–3608, https://doi.org/10.1210/jc.2011-1443

(2013) Frangou C, et al. “25(OH) Vitamin D3 Affects Acute Myeloid Leukemia Cell Proliferation.” Blood (2013) 122 (21): 5026.  

(2015) Sharon Theimer.  Vitamin D Toxicity Rare in People Who Take Supplements, Mayo. https://newsnetwork.mayoclinic.org/discussion/vitamin-d-toxicity-rare-in-people-who-take-supplements-mayo-clinic-study-finds/ Clinic Study Finds

(2015) Park S, Lee DH, Jeon JY, Ryu J, Kim S, Kim JY, Park HS, Kim SI, Park BW. Serum 25-hydroxyvitamin D deficiency and increased risk of breast cancer among Korean women: a case-control study. Breast Cancer Res Treat. 2015 Jul;152(1):147-154.

(2016) Alison M. Mondul, Stephanie J. Weinstein, Kristin A. Moy, Satu Mannisto and Demetrius Albanes. Circulating 25-hydroxyvitamin D and prostate cancer survival. Cancer Epidemiol Biomarkers Prev January 25 2016 DOI: 10.1158/1055-9965.EPI-15-0991

(2017) Editorial. Low vitamin D levels as a risk factor for cancer. BMJ 2017;359:j4952

(2017) Seyedalipour, Fatere et al. “High Prevalence of Vitamin D Deficiency in Newly Diagnosed Acute Myeloid Leukemia Patients and Its Adverse Outcome.” International journal of hematology-oncology and stem cell research vol. 11,3 (2017): 209-216.

(2018) Budhathoki Sanjeev, Hidaka Akihisa, Yamaji Taiki, Sawada Norie, Tanaka-Mizuno Sachiko, KuchibaAya et al. “Plasma 25-hydroxyvitamin D concentration and subsequent risk of total and site specific cancers in Japanese population: large case-cohort study within Japan Public Health Center-based Prospective Study cohort”. BMJ 2018;360 :k671

(2018) William B Grant. A Review of the Evidence Supporting the Vitamin D-Cancer Prevention Hypothesis in 2017.” Anticancer Research 38: 1121-1136 (2018)

(2019) Song, Dingli et al. “Vitamin D intake, blood vitamin D levels, and the risk of breast cancer: a dose-response meta-analysis of observational studies.” Aging vol. 11,24 (2019): 12708-12732. doi:10.18632/aging.102597

ZINC

(2013) Consolo, L., Melnikov, P., Cônsolo, F. et al. “Zinc supplementation in children and adolescents with acute leukemia.” Eur J Clin Nutr 67, 1056–1059 (2013). https://doi.org/10.1038/ejcn.2013.146

(2016) Pradhan R, et al.  “Protection against Methotrexate Induced Hepato-Renal Toxicity in Rats by Zinc and its Combination with Vitamin C and Vitamin E.” Med Safe Glo Heal 2016, 5:2 Research Article Open Access Med Safe Glo Heal, an open access journal Volume 5 • Issue 2 • 1000127 

(2018) Mehrzad, Valiollah et al. “Determination Relation of the Zinc Serum Level in Acute Leukemia Adult Patients with Mucositis and Neutropenic Prevalence before and after Treatment in Isfahan’ Seyed-Al-Shohada Hospital, 2012-2013.” Advanced biomedical research vol. 7 31. 16 Feb. 2018, doi:10.4103/abr.abr_7_15

(2019) Valadbeigi S, et al. ASSESSMENT OF TRACE ELEMENTS IN SERUM OF ACUTE LYMPHOBLASTIC AND MYELOID LEUKEMIA PATIENTS.” Exp Oncol 2019 41, 1, 69–71

CHEMOTHERAPY AGENTS

Riordan Clinic – IVC Protocol- https://riordanclinic.org/research-study/vitamin-c-research-ivc-protocol/

LOW DOSE (Metronomic Therapy) vs HIGH DOSE CHEMOTHERAPY 

(2016) Chan et al. “Metronomic chemotherapy prevents therapy-induced stromal activation and induction of tumor-initiating cells.” J. Exp. Med, November 2016 DOI: 10.1084/jem.20151665

(2016) “Study shows low-dose chemo keeps cancer under control.” February 24, 2016. https://medicalxpress.com/news/2016-02-low-dose-chemo-cancer.html

(2017) Xie, Xianhe et al. “Efficacy and Toxicity of Low-Dose versus Conventional-Dose Chemotherapy for Malignant Tumors: a Meta-Analysis of 6 Randomized Controlled Trials.” Asian Pacific journal of cancer prevention : APJCP vol. 18,2 479-484. 1 Feb. 2017, doi:10.22034/APJCP.2017.18.2.479

(2017) Bishnoi, R., Shah, C., Bejjanki, H. et al. “An alternative approach with a low dose and prolonged chemotherapy for palliative treatment of locally advanced, metastatic or recurrent squamous cell head and neck cancer.”  Appl Cancer Res 37, 43 (2017). https://doi.org/10.1186/s41241-017-0049-1

(2017) Cary Goldberg.  “When Cancer Can’t Be Cured, Low-Dose Chemo Aims To Keep It In Check.” https://www.wbur.org/commonhealth/2017/05/19/low-dose-chemo-cancer-treatment

(2019) Nicholas C Rohs, MD. “Lower-dose chemotherapy benefits older, frail patients with advanced gastroesophageal cancer.” HemOnc Today. May 16, 2019.

NATURAL TREATMENTS AND CHEMO/RADIATION/ IMMUNOTHERAPY 

(2011) Vollbracht C, Schneider B, Leendert V, et al. “Intravenous vitamin C administration improves quality of life in breast cancer patients during chemo-/radiotherapy and aftercare: results of a retrospective, multicentre, epidemiological cohort study in Germany.” In Vivo 25 (6): 983-90, 2011 Nov-Dec.

(2014) Ma Y, Chapman J, Levine M, et al. “High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy.” Sci Transl Med 6 (222): 222ra18, 2014.

(2018) Nauman, Gina et al. “Systematic Review of Intravenous Ascorbate in Cancer Clinical Trials.” Antioxidants (Basel, Switzerland) vol. 7,7 89. 12 Jul. 2018, doi:10.3390/antiox7070089

(2018) Anitra Carr and John Cook. Intravenous Vitamin C for Cancer Therapy – Identifying the Current Gaps in Our Knowledge.” Front. Physiol., 23 August 2018 | https://doi.org/10.3389/fphys.2018.01182.  Human trials have shown no adverse effects from combining IVC with a number of different chemotherapeutic agents (e.g., carboplatin, paclitaxel, decitabine, cytarabine, aclarubicin, gemcitabine, erlotinib, and temozolomide.

(2018) Ong C.P. High Dose Vitamin C and Low Dose Chemo Treatment Background Information”. J Cancer Sci. 2018;5(1): 4. Research has shown that intravenous high dose vitamin C in cancer therapy cannot be dismissed but that it warrants further investigation. Recent studies have made more inroads into the understanding of the role of vitamin C, bolstering more the use of the therapy by practitioners in complementary and alternative medicine. The efficaciousness of a new high dose vitamin C and low-dose chemo therapy (HiCLoChemo) reported here indicates a quantum leap in cancer treatment, which borders on the miraculous. The results of the preliminary trial on 20 patients suffering from end stage metastatic cancers, including those of the liver, lung, breast, pancreas, uterus, brain glioma, and prostate, show a remission of cancer, some complete, as confirmed by the PET/ CT images before and after the treatment protocol. The components of the treatment are not new and their variational combinations have also been used before. The key difference lies in the application of the HiCLoChemo protocol, where the vitamin C infused first, facilitates the delivery of the chemo drugs at the microenvironment of the tumor. The paper discusses the vital role of vitamin C and factors that lead to the inducement of the immune system being recruited to join in the cancer battle. The aim is to enlist cancer researchers, oncologists and cancer treatment centers to undertake clinical trials of the HiCLoChemo protocol. With confirmation of the findings, the treatment will represent a game-changer as it offers a treatment for terminal cancers that promises cancer remission at affordable cost, which is sorely needed.

ARSENIC TRIOXIDE

(2020) Wei Tian, et al.  Ascorbic Acid Sensitizes Colorectal Carcinoma to the Cytotoxicity of Arsenic Trioxide via Promoting Reactive Oxygen Species-Dependent Apoptosis and Pyroptosis.” Front. Pharmacol., 21 February 2020

AZACITIDINE

(2019) Gillberg, L., Ørskov, A.D., Nasif, A. et al. “Oral vitamin C supplementation to patients with myeloid cancer on azacitidine treatment: Normalization of plasma vitamin C induces epigenetic changes.” Clin Epigenet 11, 143 (2019). https://doi.org/10.1186/s13148-019-0739-5

BORTEZOMIB
(2011) Nakano, et al. “Delayed treatment with vitamin C and N-acetyl-L-cysteine protects Schwann cells without compromising the anti-myeloma activity of bortezomib.” Int J Hematol. 2011 Jun;93(6):727-735. doi: 10.1007/s12185-011-0850-7. Epub 2011 Apr 28.

(2020) Takács, A., Lajkó, E., Láng, O. et al. “Alpha-lipoic acid alters the antitumor effect of bortezomib in melanoma cells in vitro.” Sci Rep 10, 14287 (2020). https://doi.org/10.1038/s41598-020-71138-z

CAPECITABINE (XELODA)

(2015) Hoffer, L John et al. “High-dose intravenous vitamin C combined with cytotoxic chemotherapy in patients with advanced cancer: a phase I-II clinical trial.” PloS one vol. 10,4 e0120228. 7 Apr. 2015, doi:10.1371/journal.pone.0120228

CARBOPLATIN

(2014) Yan Ma, et al. “Pharmacological use of ascorbate acid (Vitamin C) enhanced chemosensitivity of ovarian cancer in preclinical models and reduced chemotherapy-associated toxicity in patients.” Science Translational Medicine 05 Feb 2014: 222RA18

(2015) Hoffer, L John et al. “High-dose intravenous vitamin C combined with cytotoxic chemotherapy in patients with advanced cancer: a phase I-II clinical trial.” PloS one vol. 10,4 e0120228. 7 Apr. 2015, doi:10.1371/journal.pone.0120228

(2018) Carr, Anitra C, and John Cook. “Intravenous Vitamin C for Cancer Therapy – Identifying the Current Gaps in Our Knowledge.” Frontiers in physiology vol. 9 1182. 23 Aug. 2018, doi:10.3389/fphys.2018.01182 (gemcitabine, paclitaxel, carboplatin, melphalan, carfilzomib, bortezomib, cisplatin, and temozolomide)

(2018) Klimant, E. et al. “Intravenous vitamin C in the supportive care of cancer patients: a review and rational approach.” Current Oncology, [S.l.], v. 25, n. 2, p. 139-148, apr. 2018. ISSN 1718-7729. Available at: <https://current-oncology.com/index.php/oncology/article/view/3790/2712>. Date accessed: 06 feb. 2020. doi:http://dx.doi.org/10.3747/co.25.3790.

CISPLATIN

(1997) Smyth JF, et al. “Glutathione reduces the toxicity and improves quality of life of women diagnosed with ovarian cancer treated with cisplatin: results of a double-blind, randomized trial.” Ann Oncol. 1997 Jun;8(6):569-73. PubMed PMID: 9261526.

(2012) Meher U. Nessa, et al. “Combinations of Resveratrol, Cisplatin and Oxaliplatin Applied to Human Ovarian Cancer Cells.” Anticancer Research. 32: 53-60 (2012)

CYTARABINE

(2017) Huihui Zhao, et al. “The Synergy of Vitamin C with Decitabine Activates TET2 in Leukemic Cells and Significantly Improves Overall Survival in Elderly Patients with Acute Myeloid Leukemia.” Blood (2017) 130 (Supplement 1): 1339. https://doi.org/10.1182/blood.V130.Suppl_1.1339.1339

(2018) Dinicola S, et al.  “Natural products – alpha-lipoic acid and acetyl-L-carnitine – in the treatment of chemotherapy-induced peripheral neuropathy.” European Review for Medical and Pharmacological Sciences. 2018; 22: 4739-4754.

(2019) Das, A.B., Kakadia, P.M., Wojcik, D. et al. “Clinical remission following ascorbate treatment in a case of acute myeloid leukemia with mutations in TET2 and WT1.” Blood Cancer J. 9, 82 (2019). https://doi.org/10.1038/s41408-019-0242-4

(2020) Muluken Altaye Ayza, et al.  The Role of Antioxidants in Ameliorating Cyclophosphamide-Induced Cardiotoxicity.” Oxidative Medicine and Cellular Longevity. https://www.hindawi.com/journals/omcl/2020/4965171/

DOXORUBICIN (Adriamycin)

(1996) Kurbacher, et al. “Ascorbic acid (vitamin C) improves the antineoplastic activity of doxorubicin, cisplatin, and paclitaxel in human breast carcinoma cells in vitro.” Cancer letters. 103. 183-9. 10.1016/0304-3835(96)04212-7.

(2014) Guerriero, Eliana, et al. Vitamin C Effect on Mitoxantrone-Induced Cytotoxicity in Human Breast Cancer Cell Lines.” PLoS ONE 9(12): e115287. doi:10.1371/journal.pone. 0115287

(2014) Michaud, Christie, Byron Murray.  “Reduction in Toxicity of Doxorubicin by ᾀ-Tocopherol Succinate (Vitamin E) ad Ascorbic Acid (vitamin C) in Human Ademocarcinoma cells.” Journal of Undergraduate Cells.  Jan 30, 2014.

(2017) Akolkar, et al. Vitamin C mitigates oxidative/nitrosative stress and inflammation in doxorubicin-induced cardiomyopathy.” Am J Physiol Heart Circ Physiol. 2017 Oct 1;313(4):H795-H809. 

(2017) Bober, P., Alexovic, M., Talian, I. et al. “Proteomic analysis of the vitamin C effect on the doxorubicin cytotoxicity in the MCF-7 breast cancer cell line.”  J Cancer Res Clin Oncol 143, 35–42 (2017). 

(2017) Gauri Akolkar, et al. “Doxorubicin-induced nitrosative stress is mitigated by vitamin C via the modulation of nitric oxide synthases.” Am J Physiol Cell Physiol 312: C418–C427, 2017.

(2018) Yi, et al. Glutathione-triggered dual release of doxorubicin and camptothecin for highly efficient synergistic anticancer therapy.” Colloids Surf B Biointerfaces. 2018 Sep 1;169:273-279. doi: 10.1016/j.colsurfb.2018.05.025. Epub 2018 May 17.

(2018) Zhen Lui, et al. Switching off the interactions between graphene oxide and doxorubicin using vitamin C: combining simplicity and efficiency in drug delivery.” Journal of Materials Chemistry B. Issue 8, 2018. “As a result, improvement of DOX delivery from the GO surface can be achieved using vitamin C.”

(2019) Shen, et al.  Is the combinational administration of doxorubicin and glutathione a reasonable proposal?” Acta Pharmacologica Sinica volume 40, pages699–709 (2019).  Avoid glutathione supplements with doxo).

(2019) Kunter, L, et al. “The effect of neopterin alone or in combination with doxorubicin, cisplatin and Vitamin C on the viability of different hepatocellular carcinoma cell lines.” Eurasian Journal of Biological and Chemical Sciences 2 (2019): 115-119. “We found that the addition of cisplatin to the combination of neopterin and vitamin C, causes a greater decrease in cell viability of SNU-449 cells compared to the dual therapy with neopterin and vitamin C, while the addition of doxorubicin to the same dual therapy, leads to a decrease in the effectivity of it.”

(2020) Zhengzhong Wu,et al. Exogenous Vitamin C-Triggered Surface Charge Conversion of pH/Reduction-Responsive Micelles for the Enhanced Tumor-Specific Activity of Loaded Doxorubicin.”  Molecular Pharmaceutics 2020 17 (3), 954-964

FOLFOX

(2018) William J Mullally, Awis Naeem, Colum Dennehy, Caitriona Goggin, Jack Patrick Gleeson, John Patrick Greene, Mohammed Adam Ibrahim Dawod, Seamus O’Reilly, Deirdre O’Mahony, Derek Gerard Power, and Richard Martin Bambury. “Safety and tolerability of adjuvant FOLFOX vs. CAPOX in colon cancer: A real-world experience.” Journal of Clinical Oncology 2018 36:15

(2019) Wang, F., He, M., Wang, Z. et al. “Phase I study of high-dose ascorbic acid with mFOLFOX6 or FOLFIRI in patients with metastatic colorectal cancer or gastric cancer.” BMC Cancer 19, 460 (2019). https://doi.org/10.1186/s12885-019-5696-z

GEMCITIBINE- no known adverse reaction with Vit C

(2012) Monti DA, et al. “Phase I evaluation of intravenous ascorbic acid in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer.” PLoS One. 2012;7(1):e29794. doi: 10.1371/journal.pone.0029794. Epub 2012 Jan 17.

(2013) Welsh JL, Wagner BA, van’t Erve TJ, et al. “Pharmacological ascorbate with gemcitabine for the control of metastatic and node-positive pancreatic cancer (PACMAN): results from a phase I clinical trial.” Cancer Chemother Pharmacol 71 (3): 765-75, 2013.

(2016) Monti, et al.  “Intravenous vitamin C in combination with gemcitabine and erlotinib in subjects with metastatic pancreatic cancer.” DOI: 10.1200/jco.2011.29.15_suppl.e14547. Journal of Clinical Oncology 2. Published online September 22, 2016.9, no. 15_suppl 

(2017) Polireddy, et al. High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study.” Sci Rep. 2017 Dec 7;7(1):17188. doi: 10.C1038/s41598-017-17568-8.

HYDROXYUREA- no known adverse reaction with Vit C

 

METHOTREXATE

(2014) Yiang, Giou-Teng et al. “Vitamin C enhances anticancer activity in methotrexate-treated Hep3B hepatocellular carcinoma cells.” Oncology reports vol. 32,3 (2014): 1057-63. doi:10.3892/or.2014.3289

(2016) Pradhan R, et al.  “Protection against Methotrexate Induced Hepato-Renal Toxicity in Rats by Zinc and its Combination with Vitamin C and Vitamin E.” Med Safe Glo Heal 2016, 5:2 Research Article Open Access Med Safe Glo Heal, an open access journal Volume 5 • Issue 2 • 1000127 

(2017) Wu CW, Liu HC, Yu YL, Hung YT, Wei CW, Yiang GT. “Combined treatment with vitamin C and methotrexate inhibits triple-negative breast cancer cell growth by increasing H2O2 accumulation and activating caspase-3 and p38 pathways.” Oncol Rep. 2017;37(4):2177-2184. doi:10.3892/or.2017.5439

(2017) Mehrzadi S, Fatemi I, Esmaeilizadeh M, Ghaznavi H, Kalantar H, Goudarzi M. “Hepatoprotective effect of berberine against methotrexate induced liver toxicity in rats.” Biomed Pharmacother. 2018;97:233-239. doi:10.1016/j.biopha.2017.10.113

NIVOLUMAB

(2019) Cusato, Jessica et al. “Influence of Vitamin D in Advanced Non-Small Cell Lung Cancer Patients Treated with Nivolumab.” Cancers vol. 11,1 125. 21 Jan. 2019, doi:10.3390/cancers11010125

OXALIPLATIN

(1986) Midander J, Deschavanne PJ, Debieu D, Malaise EP, Revesz L. “Reduced repair of potentially lethal radiation damage in glutathione synthetase-deficient human fibroblasts after X-irradiation.” Int J Radiat Biol Relat Stud Phys Chem Med. 1986 Mar;49(3):403-13. PMID: 3485589

(2002) Gedlicka, et al. Effective Treatment of Oxaliplatin-Induced Cumulative Poly-Neuropathy with Alpha-Lipoic Acid.”  J. Clin. Oncol. 2002; 20 (5): 3359-3361.

(2002) Cascinu S, et al. “Neuroprotective effect of reduced glutathione on oxaliplatin-based chemotherapy in advanced colorectal cancer: a randomized, double-blind, placebo-controlled trial.” J Clin Oncol. 2002 Aug 15;20(16):3478-83. PubMed PMID: 12177109.

(2009) Milla P, et al. “Administration of reduced glutathione in FOLFOX4 adjuvant treatment for colorectal cancer: effect on oxaliplatin pharmacokinetics, Pt-DNA adduct formation, and neurotoxicity.” Anticancer Drugs. 2009 Jun;20(5):396-402. PubMed PMID: 19287306.

(2009) Pujari G, Berni A, Palitti F, Chatterjee A. “Influence of glutathione levels on radiation-induced chromosomal DNA damage and repair in human peripheral lymphocytes.” Mutat Res. 2009 JunJul;677(1-2):109-10. PMID: 19386243

(2012) Meher U. Nessa, et al. “Combinations of Resveratrol, Cisplatin and Oxaliplatin Applied to Human Ovarian Cancer Cells.” Anticancer Research. 32: 53-60 (2012)

PACLITAXEL/ DOXORUBICIN

(1996) Kurbacher, et al. “Ascorbic acid (vitamin C) improves the antineoplastic activity of doxorubicin, cisplatin, and paclitaxel in human breast carcinoma cells in vitro.” Cancer letters. 103. 183-9. 10.1016/0304-3835(96)04212-7.

(2014) Ma Y, Chapman J, Levine M, et al. “High-dose parenteral ascorbate enhanced chemosensitivity of ovarian cancer and reduced toxicity of chemotherapy.” Sci Transl Med 6 (222): 222ra18, 2014.

(2018) Anitra Carr and John Cook. “Intravenous Vitamin C for Cancer Therapy – Identifying the Current Gaps in Our Knowledge.” Frontiers in physiology.  August 23, 2018. https://www.frontiersin.org/articles/10.3389/fphys.2018.01182/full

Paclitaxel overview- https://www.medicines.org.uk/emc/product/3891/smpc

SORAFENIB

(2016) Yassem M Saleh and Waleed Hammam. “Efficacy and Safety of Sorafenib in Patients with Advanced Hepatocellular Cancer.”  Journal of Cancer and Tumor International. 3(1): 1-7, 2016. doi: 10.9734/JCTI/2016/22142

(2016) Potenza N, Mosca N, Zappavigna S, Castiello F, Panella M, Ferri C, Vanacore D, Giordano A, Stiuso P, Caraglia M, Russo A. “MicroRNA-125a-5p is a downstream effector of sorafenib in its antiproliferative activity toward human hepatocellular carcinoma cells.” J Cell Physiol. 2016 Dec 16. doi: 10.1002/jcp.25744.

(2016) Lauren Rouleau, Anil Noronha Antony, Sara Bisetto, Andrew Newberg, Cataldo Doria, Mark Levine, Daniel A. Monti, and Jan B. Hoek  “Synergistic effects of ascorbate and sorafenib in hepatocellular carcinoma: New insights into ascorbate cytotoxicity.” Free Radic Biol Med . 2016 June ; 95: 308–322. doi:10.1016/j.freeradbiomed.2016.03.031.

(2017) Yan-jing Zhu & Bo Zheng. “New knowledge of the mechanisms of sorafenib resistance in liver cancer.” Acta Pharmacologica Sinica.  (2017) 38, 614-622.  Dio: 10.1038/aps.2017.5

TAMOXIFEN

(2014) Subramani, et al. “Vitamin C suppresses cell death in MCF-7 human breast cancer cells induced by tamoxifen.” Journal of Cellular and Molecular Medicine. 2014 Feb;18(2):305-13. doi: 10.1111/jcmm.12188. Epub 2013 Nov 25.

TEMOZOLOMIDE

(2012) Yuan Y, et al. “Resveratrol enhances the antitumor effects of temozolomide in glioblastoma via ROS-dependent AMPK-TSCmtor signaling pathway.” CNS Neurosci Ther 2012;00:1-11.

(2012) Shih CM, et al. “Resveratrol enhances the therapeutic effect of temozolomide against malignant glioma in vitro and in vivo by inhibiting autophagy.” Free Radical Biol Med 2012;52:377-91.

(2015) Filho AZ, et al. “Autophagy inhibition improves the efficacy of curcumin/temozolomide combination therapy in glioblastomas.” Cancer Lett 2015;358:220-31.

(2015) Zhang ZS, et al. “Dihydroartemisinin increases temozolomide efficacy in glioma cells by inducing autophagy.” Oncol Lett 2015;10:379-83

(2016) Bak DH. “Autophagy enhancement contributes to the synergistic effect of vitamin D in temozolomide-based glioblastoma chemotherapy.” Exp Ther Med 2016;11:2153-62

(2018) McConnell, Diane D et al. “Do Anti-Oxidants Vitamin D3, Melatonin, and Alpha-Lipoic Acid Have Synergistic Effects with Temozolomide on Cultured Glioblastoma Cells?.” Medicines (Basel, Switzerland) vol. 5,2 58. 20 Jun. 2018, doi:10.3390/medicines5020058

Clinical Trials with Vitamin C and Standard of Care oncology

Pazopanib Hydrochloride with or without Ascorbic Acid in Treating Patients with Kidney Cancer That Is Metastatic or Cannot Be Removed by Surgery

(2018) Anitra Carr and John Cook. Intravenous Vitamin C for Cancer Therapy – Identifying the Current Gaps in Our Knowledge.” Front. Physiol., 23 August 2018