Treatment Approach
Strategic Warfare Against Cancer: A Combined Defensive and Offensive Approach
In research published in 2004 and again in 2023, many contemporary cancer drugs were found to have a limited impact on enhancing the quality of life or prolonging the lives of those with metastatic cancer (click here and here). Such cancer is often associated with reduced survival rates because alternative approaches are limited when cancer cells evolve and acquire multi-drug resistance. New treatment approaches are needed to substantially improve outcomes. Fortunately, research scientists and forward-thinking physicians have been hard at work developing innovative approaches to fight cancer and improve the chances for long-term remission.
Our novel approach draws inspiration from an unexpected but powerful source: ancient military strategy. As Sun Tzu, the Chinese military strategist (544-496 BCE), wrote in his enduring treatise The Art of War, “Invincibility lies in the defense.” This principle, which has guided military campaigns for over two millennia, can be powerfully applied to cancer treatment through a strategic combination of defensive and offensive measures. Before launching a major offensive against cancer, we must first secure our position and weaken the enemy’s strongholds. In addition to important diet and lifestyle changes (click here), our approach involves a carefully sequenced deployment of two powerful therapeutic strategies: bicarbonate therapy as the defensive foundation, followed by enhanced high-ozonide oil as the offensive strike.
The Defensive Phase: Creating an Inhospitable Environment for Cancer
The defensive phase employs bicarbonate therapy to systematically weaken cancer’s position through multiple mechanisms. Cancer cells create and thrive in an acidic environment, and bicarbonate therapy neutralizes this acidic fortress, disrupting cancer’s protective barrier and forcing cancer cells to expend extra energy maintaining their preferred acidic state. Through this process, abnormal blood vessels in tumors are normalized, which improves oxygen delivery to the tumor site, enables more effective drug delivery, and reduces tumor hypoxia. Additionally, matrix metalloproteinases (enzymes that enable cancer spread) are inhibited, reducing cancer cells’ ability to break away and spread, effectively blocking escape routes and preventing cancer from establishing new colonies.
The neutralization of the acidic environment profoundly affects immune function and cancer stem cells. Acidic environments typically suppress immune cell function, but by neutralizing the acidity, T-cells and natural killer cells can operate effectively, allowing the immune system to better recognize and attack cancer cells, improving overall immune surveillance. Furthermore, the alkaline environment reduces the population of cancer stem cells, which are responsible for tumor regrowth and treatment resistance, effectively cutting off cancer’s ability to regenerate and diminish treatment resistance potential. Progress in this defensive phase can be monitored through urine pH, with a target of 7.5-8.0 signaling successful alkalinization and confirming defensive preparations are complete, giving the green light for proceeding to the offensive phase.
The Offensive Phase: Launching a Precision-Guided Attack
Once the defensive groundwork is laid, the offensive phase employs enhanced high-ozonide oil to deliver a powerful oxidative assault through multiple mechanisms. The primary attack mechanisms include direct targeting of cancer cell mitochondria, cardiolipin oxidation in mitochondrial membranes, triggering of apoptosis (programmed cell death), and calcium release causing cellular destruction. This core offensive strategy is enhanced by several compounds, each targeting specific cancer defense mechanisms. Sulfasalazine blocks glutathione synthesis, removing the primary antioxidant defense and making cancer cells vulnerable to oxidative stress. Auranofin disrupts the thioredoxin system, eliminating secondary antioxidant defense and further weakening cancer’s protective mechanisms. Chrysin suppresses Nrf2-mediated responses, preventing adaptation to oxidative stress and blocking cancer’s ability to develop new defenses. Niclosamide induces additional mitochondrial stress, promotes autophagy (cellular self-destruction), and creates multiple paths to cancer cell death.
The synergistic benefits of this two-phase approach are significant and multi-faceted. The defensive preparations optimize offensive impact, with multiple simultaneous attacks overwhelming cancer cells while drug delivery and effectiveness are maximized and immune system support is strengthened. Importantly, healthy cells maintain their natural defenses and are better equipped to handle oxidative stress, while the targeted nature of the attack spares healthy tissue, creating selective toxicity toward cancer cells. The approach also effectively prevents resistance through multiple simultaneous mechanisms that prevent adaptation, block escape routes, and disable survival mechanisms. This comprehensive strategy creates a “perfect storm” against cancer cells while protecting normal cells, with the carefully sequenced deployment of defensive and offensive measures maximizing the therapeutic impact while minimizing the potential for resistance or escape.
The success of this approach lies in its systematic nature and the synergy between its components. By first creating an inhospitable environment for cancer cells through bicarbonate therapy, then launching a powerful oxidative assault enhanced by multiple defensive system disablers, the strategy effectively corners cancer cells while protecting healthy tissue. The careful monitoring of progress through urine pH ensures that the offensive phase begins only when optimal conditions have been created, maximizing its effectiveness. This methodical, two-phase approach represents a sophisticated strategy that leverages our understanding of cancer cell biology and vulnerability to create a comprehensive treatment protocol with the potential for improved outcomes in cancer therapy.
Note: To review the anti-cancer mechanisms of action and scientific validation for each of the components in our strategy, please scroll down to the reference list below.
Treatment Indications
Based on this comprehensive approach, our treatment may be particularly beneficial for:
- Patients seeking to enhance the effectiveness and/or minimize the side effects of conventional cancer treatment, including surgery, chemotherapy, radiation, targeted therapy, hormone blockade, and immunotherapy.
- Patients who have not responded to or can no longer tolerate conventional treatment.
- Patients who do not qualify for a drug trial or failed to respond favorably to a trial drug.
- Patients wanting to prolong their remission with health-enhancing strategies.
- Patients seeking to improve their quality of life.
Patient Care and Treatment Cost
Dr. Thomas ensures exceptional personalized care by limiting his practice to no more than 50 patients, in contrast to the typical oncology practice of 250-500 patients. This focused approach allows for both individualized attention and ongoing research to optimize patient outcomes. The treatment approach described above requires close medical oversight by Dr. Thomas. Following an initial physical examination in his office, patients commence their treatment. Monthly follow-up appointments are scheduled and can be conducted through telemedicine or in-person visits. Patients enjoy unrestricted email access to Dr. Thomas throughout their treatment, ensuring they receive continuous and responsive support.
Our comprehensive monthly fee of $1850 includes custom-synthesized oral liposomal niclosamide and provides continuous access to Dr. Thomas’s expertise. This represents a significant value compared to international treatment options, which often range from $7,000 to $20,000 weekly or $40,000 to $120,000 for experimental procedures with limited scientific validation. Other costs are for online items (bicarbonate, high-ozonide ozonated oil, and liposomal chrysin) and retail pharmacy items (auranofin and sulfasalazine). Treatment continues until achieving remission or disease stabilization, typically within a 12-18-month timeframe. Other than covering the cost of bloodwork, health insurance and Medicare do not pay for integrative cancer treatment.
References
Auranofin:
Mechanisms of action:
- Cell cycle arrest and apoptosis induction: In multiple myeloma, auranofin induces cell cycle arrest and apoptosis, reduces Mcl-1 expression, and down-regulates NF-κß activity.
- Oxidative stress induction: It increases reactive oxygen species (ROS) levels, leading to DNA damage and caspase-independent apoptosis, particularly in cells dependent on the Trx1 system.
- PI3K/AKT/mTOR pathway inhibition: Auranofin inhibits this pathway, essential for cell proliferation, apoptosis, and angiogenesis, affecting tumor growth and metastasis.
- Protein homeostasis disruption: It inhibits proteasome and deubiquitinases (dubs), inducing apoptosis in liver hepatocellular and breast cancer cells.
- FOXO3-dependent apoptosis: In ovarian cancer cells lacking p53, auranofin triggers apoptosis through FOXO3 activation, indicating a p53-independent pathway.
- IKK-β inhibition and NF-κß signaling modulation: Downregulates IKK-β, reducing NF-κß signaling and promoting apoptosis via FOXO3 nuclear translocation.
- Mitochondrial dysfunction: Leads to loss of mitochondrial membrane potential, resulting in cell death through apoptosis or necrosis.
- Abdalbari FH, Telleria CM. The gold complex auranofin: new perspectives for cancer therapy. Discov Oncol. 2021 Oct 20;12(1):42
- Cui XY, Park SH, Park WH. Anti-Cancer Effects of Auranofin in Human Lung Cancer Cells by Increasing Intracellular ROS Levels and Depleting GSH Levels. Molecules. 2022 Aug 15;27(16):5207.
- Cui XY, Park SH, Park WH. Auranofin inhibits the proliferation of lung cancer cells via necrosis and caspase‑dependent apoptosis. Oncol Rep. 2020 Dec;44(6):2715-2724.
- Fiskus W, Saba N, Shen M, Ghias M, Liu J, Gupta SD, Chauhan L, Rao R, Gunewardena S, Schorno K, Austin CP, Maddocks K, Byrd J, Melnick A, Huang P, Wiestner A, Bhalla KN. Auranofin induces lethal oxidative and endoplasmic reticulum stress and exerts potent preclinical activity against chronic lymphocytic leukemia. Cancer Res. 2014 May 1;74(9):2520-32.
- Gamberi T, Chiappetta G, Fiaschi T, Modesti A, Sorbi F, Magherini F. Upgrade of an old drug: Auranofin in innovative cancer therapies to overcome drug resistance and to increase drug effectiveness. Med Res Rev. 2022 May;42(3):1111-1146.
- Huang H, Liao Y, Liu N, Hua X, Cai J, Yang C, Long H, Zhao C, Chen X, Lan X, Zang D, Wu J, Li X, Shi X, Wang X, Liu J. Two clinical drugs deubiquitinase inhibitor auranofin and aldehyde dehydrogenase inhibitor disulfiram trigger synergistic anti-tumor effects in vitro and in vivo. Oncotarget. 2016 Jan 19;7(3):2796-808.
- Li H, Hu J, Wu S, Wang L, Cao X, Zhang X, Dai B, Cao M, Shao R, Zhang R, Majidi M, Ji L, Heymach JV, Wang M, Pan S, Minna J, Mehran RJ, Swisher SG, Roth JA, Fang B. Auranofin-mediated inhibition of PI3K/AKT/mTOR axis and anticancer activity in non-small cell lung cancer cells. Oncotarget. 2016 Jan 19;7(3):3548-58.
- Liu X, Wang W, Yin Y, Li M, Li H, Xiang H, Xu A, Mei X, Hong B, Lin W. A high-throughput drug screen identifies auranofin as a potential sensitizer of cisplatin in small cell lung cancer. Invest New Drugs. 2019 Dec;37(6):1166-1176.
- Nag D, Bhanja P, Riha R, Sanchez-Guerrero G, Kimler BF, Tsue TT, Lominska C, Saha S. Auranofin Protects Intestine against Radiation Injury by Modulating p53/p21 Pathway and Radiosensitizes Human Colon Tumor. Clin Cancer Res. 2019 Aug 1;25(15):4791-4807.
- Nakaya A, Sagawa M, Muto A, Uchida H, Ikeda Y, Kizaki M. The gold compound auranofin induces apoptosis of human multiple myeloma cells through both down-regulation of STAT3 and inhibition of NF-κß activity. Leuk Res. 2011 Feb;35(2):243-9.
- Park SH, Lee JH, Berek JS, Hu MC. Auranofin displays anticancer activity against ovarian cancer cells through FOXO3 activation independent of p53. Int J Oncol. 2014 Oct;45(4):1691-8.
- Varghese E, Büsselberg D. Auranofin, an anti-rheumatic gold compound, modulates apoptosis by elevating the intracellular calcium concentration ([ca2+]I) in mcf-7 breast cancer cells. Cancers (Basel). 2014 Nov 6;6(4):2243-58.
- Wang H, Bouzakoura S, de Mey S, Jiang H, Law K, Dufait I, Corbet C, Verovski V, Gevaert T, Feron O, Van den Berge D, Storme G, De Ridder M. Auranofin radiosensitizes tumor cells through targeting thioredoxin reductase and resulting overproduction of reactive oxygen species. Oncotarget. 2017 May 30;8(22):35728-35742.
- Zou P, Chen M, Ji J, Chen W, Chen X, Ying S, Zhang J, Zhang Z, Liu Z, Yang S, Liang G. Auranofin induces apoptosis by ROS-mediated ER stress and mitochondrial dysfunction and displayed synergistic lethality with piperlongumine in gastric cancer. Oncotarget. 2015 Nov 3;6(34):36505-21.
Bicarbonate:
Mechanisms of action:
- Neutralization of tumor acidity: Bicarbonate buffers the acidic tumor microenvironment, increasing extracellular pH. This disrupts the acidosis that supports cancer progression, metastasis, and therapy resistance.
- Inhibition of cancer cell invasion and metastasis: Acidic conditions facilitate extracellular matrix degradation, aiding cancer cell migration. Bicarbonate reduces extracellular acidity, impairing the invasiveness and metastatic potential of cancer cells.
- Enhancement of immune response and reduction of tumor microenvironment immunosuppression: Acidosis suppresses immune cell functions, including T-cell activation and cytokine production, while lactate accumulation directly inhibits cytotoxic T lymphocytes. Bicarbonate neutralizes acidity and reduces lactate levels, restoring immune activity and enhancing responses to immunotherapies such as anti-CTLA-4 and anti-PD1.
- Improvement of chemotherapy efficacy: Acidic environments reduce the effectiveness of weak-base chemotherapeutic drugs. Bicarbonate neutralizes tumor acidity, improving drug delivery and efficacy.
- Reversal of metabolic advantages of cancer cells: Cancer cells rely on the Warburg effect (aerobic glycolysis), producing lactic acid and creating an acidic microenvironment. Bicarbonate disrupts this metabolic adaptation, limiting cancer cell survival and proliferation.
- Reduction of hypoxia-driven tumor adaptations: Hypoxic and acidic conditions drive tumor metabolic reprogramming and aggressive behavior. By increasing pH, bicarbonate disrupts these adaptive mechanisms.
- Suppression of acid-sensitive pathways: Bicarbonate interferes with the function of acid-regulating proteins such as carbonic anhydrase IX, monocarboxylate transporters, and proton pumps, which are critical for cancer cell survival.
- Restoration of cellular processes: Acidosis disrupts key cellular processes like adhesion, proliferation, apoptosis, and histone acetylation. Bicarbonate restores these processes by maintaining a more alkaline intracellular and extracellular pH.
Citations:
- Alfarouk KO, Verduzco D, Rauch C, Muddathir AK, Adil HH, Elhassan GO, Ibrahim ME, David Polo Orozco J, Cardone RA, Reshkin SJ, Harguindey S. Glycolysis, tumor metabolism, cancer growth and dissemination. A new pH-based etiopathogenic perspective and therapeutic approach to an old cancer question. Oncoscience. 2014 Dec 18;1(12):777-802.
- Boedtkjer E, Pedersen SF. The Acidic Tumor Microenvironment as a Driver of Cancer. Annu Rev Physiol. 2020 Feb 10;82:103-126.
- Bogdanov A, Verlov N, Bogdanov A, Burdakov V, Semiletov V, Egorenkov V, Volkov N, Moiseyenko V. Tumor alkalization therapy: Misconception or good therapeutics perspective? The case of malignant ascites. Front Oncol. 2024;14:1342802.
- Fais S, Venturi G, Gatenby B. Microenvironmental acidosis in carcinogenesis and metastases: new strategies in prevention and therapy. Cancer Metastasis Rev. 2014 Dec;33(4):1095-108.
- Gillies RJ, Pilot C, Marunaka Y, Fais S. Targeting acidity in cancer and diabetes. Biochim Biophys Acta Rev Cancer. 2019 Apr;1871(2):273-280.
- Hamaguchi R, Isowa M, Narui R, Morikawa H, Okamoto T, Wada H. How Does Cancer Occur? How Should It Be Treated? Treatment from the Perspective of Alkalization Therapy Based on Science-Based Medicine. Biomedicines. 2024 Sep 26;12(10):2197.
- Hamaguchi R, Isowa M, Narui R, Morikawa H, Wada H. Clinical review of alkalization therapy in cancer treatment. Front Oncol. 2022 Sep 14;12:1003588.
- Hamaguchi R, Ito T, Narui R, Morikawa H, Uemoto S, Wada H. Effects of Alkalization Therapy on Chemotherapy Outcomes in Advanced Pancreatic Cancer: A Retrospective Case-Control Study. In Vivo. 2020 Sep-Oct;34(5):2623-2629.
- Hamaguchi R, Narui R, Wada H. Effects of Alkalization Therapy on Chemotherapy Outcomes in Metastatic or Recurrent Pancreatic Cancer. Anticancer Res. 2020 Feb;40(2):873-880.
- Han JH, Jeong SH, Yuk HD, Jeong CW, Kwak C, Ku JH. Acidic Urine Is Associated With Poor Prognosis of Upper Tract Urothelial Carcinoma. Front Oncol. 2022 Jan 24;11:817781.
- Isowa M, Hamaguchi R, Narui R, Morikawa H, Okamoto T, Wada H. Potential of Alkalization Therapy for the Management of Metastatic Pancreatic Cancer: A Retrospective Study. Cancers (Basel). 2024;16(1):61.
- Peppicelli S, Andreucci E, Ruzzolini J, Margheri F, Laurenzana A, et al. (2017) Acidity of Microenvironment as a Further Driver of Tumor Metabolic Reprogramming. J Clin Cell Immunol 8: 485.
- Pilon-Thomas S, Kodumudi KN, El-Kenawi AE, Russell S, Weber AM, Luddy K, Damaghi M, Wojtkowiak JW, Mulé JJ, Ibrahim-Hashim A, Gillies RJ. Neutralization of Tumor Acidity Improves Antitumor Responses to Immunotherapy. Cancer Res. 2016 Mar 15;76(6):1381-90.
- Wada H, Hamaguchi R, Narui R, Morikawa H. Meaning and significance of alkalization therapy for cancer. Front Oncol. 2022;12:920843.
- Ward C, Meehan J, Gray ME, Murray AF, Argyle DJ, Kunkler IH, Langdon SP. The impact of tumour pH on cancer progression: strategies for clinical intervention. Explor Target Antitumor Ther. 2020;1(2):71-100.
- Welch AA, Mulligan A, Bingham SA, Khaw KT. Urine pH is an indicator of dietary acid-base load, fruit and vegetables and meat intakes: results from the European Prospective Investigation into Cancer and Nutrition (EPIC)-Norfolk population study. Br J Nutr. 2008 Jun;99(6):1335-43.
- Worsley CM, Veale RB, Mayne ES. The acidic tumour microenvironment: Manipulating the immune response to elicit escape. Hum Immunol. 2022 May;83(5):399-408.
- Yang M, Zhong X, Yuan Y. Does baking soda function as a magic bullet for patients with cancer? A mini review. Integr Cancer Ther. 2020;19:1534735420922579.4
Chrysin:
Mechanisms of action:
- Suppressing Nrf2-mediated defenses: Chrysin enhances the effectiveness of ROS-mediated cancer treatments by suppressing the Nrf2-mediated antioxidant response in cancer cells, preventing them from upregulating defenses against oxidative stress, thereby increasing their vulnerability to ROS-induced damage and improving sensitivity to pro-oxidative therapies.
- Induction of apoptosis: Chrysin activates apoptotic pathways, often involving caspase activation and the inhibition of anti-apoptotic proteins such as Bcl-2. It induces apoptosis through both intrinsic and extrinsic pathways, enhancing cancer cell susceptibility to programmed cell death.
- Cell cycle arrest: It disrupts the cell cycle by regulating cyclins and cyclin-dependent kinases (CDKs), effectively halting cancer cell proliferation.
- Inhibition of angiogenesis: Chrysin downregulates hypoxia-inducible factor-1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF), which are critical for tumor angiogenesis.
- Anti-inflammatory effects: By suppressing NF-κB activity and reducing levels of pro-inflammatory cytokines such as TNF-α and IL-1β, chrysin creates an unfavorable environment for cancer progression.
- Anti-metastatic properties: Chrysin inhibits epithelial-to-mesenchymal transition (EMT), reducing cancer cell migration and invasion. It also downregulates metalloproteinases (MMPs) involved in metastasis.
- Regulation of autophagy: Chrysin affects autophagy through pathways such as CDK1/ULK1, modulating the tumor microenvironment and cancer cell survival mechanisms.
- Sensitization to chemotherapy: Chrysin sensitizes cancer cells to chemotherapeutic agents by overcoming drug resistance mechanisms, enhancing the efficacy of combination therapies.
- Inhibition of pro-survival signaling pathways: It targets critical signaling pathways like PI3K/Akt, STAT3, and MAPK, which are involved in cancer cell survival and proliferation.
- Suppression of tumor-associated macrophages (TAMs): By inhibiting TAM-mediated autophagy and their pro-tumor effects, chrysin reduces the supportive role of the tumor microenvironment.
- Epigenetic modulation: Chrysin exhibits inhibitory effects on histone deacetylases (HDACs) and influences the expression of microRNAs involved in cancer progression.
- Fu B, Xue J, Li Z, Shi X, Jiang BH, Fang J. Chrysin inhibits expression of hypoxia-inducible factor-1alpha through reducing hypoxia-inducible factor-1alpha stability and inhibiting its protein synthesis. Mol Cancer Ther. 2007 Jan;6(1):220-6.
- Khoo BY, Chua SL, Balaram P. Apoptotic effects of chrysin in human cancer cell lines. Int J Mol Sci. 2010 May 19;11(5):2188-99.
- Liu X, Zhang X, Shao Z, Zhong X, Ding X, Wu L, Chen J, He P, Cheng Y, Zhu K, Zheng D, Jing J, Luo T. Pyrotinib and chrysin synergistically potentiate autophagy in HER2-positive breast cancer. Signal Transduct Target Ther. 2023 Dec 18;8(1):463.
- Moghadam ER, Ang HL, Asnaf SE, Zabolian A, Saleki H, Yavari M, Esmaeili H, Zarrabi A, Ashrafizadeh M, Kumar AP. Broad-Spectrum Preclinical Antitumor Activity of Chrysin: Current Trends and Future Perspectives. Biomolecules. 2020 Sep 27;10(10):1374.
- Raina R, Bhatt R, Hussain A. Chrysin targets aberrant molecular signatures and pathways in carcinogenesis (Review). World Acad Sci J. 2024 Jun;6:45.
- Salari N, Faraji F, Jafarpour S, Faraji F, Rasoulpoor S, Dokaneheifard S, Mohammadi M. Anti-cancer Activity of Chrysin in Cancer Therapy: a Systematic Review. Indian J Surg Oncol. 2022 Dec;13(4):681-690.
- Sood A, Mehrotra A, Sharma U, Aggarwal D, Singh T, Shahwan M, Jairoun AA, Rani I, Ramniwas S, Tuli HS, Yadav V, Kumar M. Advancements and recent explorations of anti-cancer activity of chrysin: from molecular targets to therapeutic perspective. Explor Target Antitumor Ther. 2024;5(3):477-494.
- Tang X, Luo X, Wang X, Zhang Y, Xie J, Niu X, Lu X, Deng X, Xu Z, Wu F. Chrysin Inhibits TAMs-Mediated Autophagy Activation via CDK1/ULK1 Pathway and Reverses TAMs-Mediated Growth-Promoting Effects in Non-Small Cell Lung Cancer. Pharmaceuticals (Basel). 2024 Apr 17;17(4):515.
- Talebi M, Talebi M, Farkhondeh T, Simal-Gandara J, Kopustinskiene DM, Bernatoniene J, Samarghandian S. Emerging cellular and molecular mechanisms underlying anticancer indications of chrysin. Cancer Cell Int. 2021 Apr 13;21(1):214.
High-ozonide oil (HOO):
Mechanisms of action:
- Re-activation of intrinsic apoptosis: HOO oxidizes mitochondrial membranes in cancer cells by exploiting structural differences in cardiolipin (a key phospholipid in mitochondrial membranes). In cancer cells, cardiolipin’s altered structure due to lack of cytochrome c binding creates gaps that allow HOO access, while normal cells’ intact cardiolipin structure blocks HOO. This selective targeting triggers release of cytochrome c and calcium, leading to apoptosis specifically in cancer cells.
- Inhibition of tumor-associated macrophage activation: HOO inhibits the oxidative burst and inflammatory cytokine release from macrophages that typically support tumor growth.
- Increase of oxygen availability in tumor tissue: HOO releases oxygen species inside cancer tissue, counteracting the hypoxic environment that triggers angiogenesis and metastasis.
- Competition with mitochondrial fat oxidation pathway: HOO may compete with fatty acid oxidation, which provides energy to cancer cells. Its catabolism leads to oxidative stress, mitochondrial damage, and apoptosis.
- Targeting cancer stem cells: HOO depletes the high antioxidant levels in cancer stem cells, reversing their chemo/radioresistance.
- Anti-inflammatory effects at the systemic level: HOO induces anti-inflammatory effects without immunosuppression by inhibiting macrophage oxidative burst.
Citations:
- Baeza-Noci J, Pinto-Bonilla R. Systemic Review: Ozone: A Potential New Chemotherapy. Int J Mol Sci. 2021 Oct 30;22(21):11796.
- Izzotti A, Fracchia E, Rosano C, Comite A, Belgioia L, Sciacca S, Khalid Z, Congiu M, Colarossi C, Blanco G, Santoro A, Chiara M, Pulliero A. Efficacy of High-Ozonide Oil in Prevention of Cancer Relapses Mechanisms and Clinical Evidence. Cancers (Basel). 2022 Feb 24;14(5):1174.
- Li Y, Pu R. Ozone Therapy for Breast Cancer: An Integrative Literature Review. Integr Cancer Ther. 2024 Jan-Dec;23:15347354241226667.
Niclosamide:
Mechanisms of action:
- Selective toxicity against p53-deficient cells: Niclosamide preferentially impairs the growth of p53-deficient cells and p53 mutant patient-derived ovarian xenografts, highlighting its targeted action against cancer cells with specific genetic vulnerabilities.
- Mitochondrial uncoupling and energy metabolism interference: It induces mitochondrial uncoupling, affects mitochondrial function, and disrupts energy metabolism in cancer cells, leading to metabolic stress, cell death through apoptosis, and autophagy.
- Induction of apoptosis and cell cycle arrest: Niclosamide triggers apoptosis in cancer cells, disrupts cell cycle progression, and enhances cleavage of caspase-9, caspase-3, and PARP1, contributing to the inhibition of tumor cell growth.
- Alteration of metabolome profile and arachidonic acid accumulation: It alters the metabolic landscape, particularly leading to the accumulation of arachidonic acid in p53-deficient cells, which is implicated in apoptosis.
- Perturbation of Ca²⁺ homeostasis: By triggering intracellular calcium fluxes, niclosamide affects calcium-dependent processes, leading to changes in cell signaling and metabolism.
- Inhibition of various signaling pathways: Niclosamide downregulates critical cancer signaling pathways, including NF-κB, Wnt/β-catenin, Notch, ROS, mTORC1, STAT3, and HIF-1α, suppressing cancer cell energy metabolism, proliferation, growth, and survival.
- Targeting cancer stem cells: Niclosamide targets cancer stem cells, addressing the challenges of tumor recurrence and metastasis.
- Inhibition of tumor growth and metastasis: It exhibits anti-metastatic properties, inhibiting cancer cell migration, invasion, and overall tumor growth.
- Enhancing chemo and radio sensitivity: Niclosamide improves the sensitivity of cancer cells to chemotherapy and radiotherapy, making it a valuable adjunct in cancer treatment.
- Synergistic effects with cancer therapies: Demonstrating synergy with existing cancer treatments, niclosamide can potentiate the therapeutic effects against cancer.
- Modulation of epigenetic regulation: It influences epigenetic mechanisms, altering gene expression related to cancer survival and resistance.
- Altering tumor microenvironment: Niclosamide may improve the therapeutic response by modulating the tumor microenvironment and affecting the cancer’s immune evasion capabilities.
- Induction of lipid oxygenation genes in wild-type p53 cells: In cells with functional p53, niclosamide induces genes involved in lipid metabolism, which helps in counteracting the metabolic stress imposed by the drug.
- Inhibition of glutathione synthase (GS): Inhibits GS, reducing glutathione levels and increasing oxidative stress, contributing to cell stress and death.
- Downregulation of nuclear factor of activated T-cells (NFAT) activity: Decreases NFAT activity, impacting cell growth and viability.
- Decreased runt-related transcription factor 2 (RUNX2) expression: RUNX2’s central role in the growth and spread of various cancers makes it an attractive therapeutic target.
Citations:
- Cheng B, Morales LD, Zhang Y, Mito S, Tsin A. Niclosamide induces protein ubiquitination and inhibits multiple pro-survival signaling pathways in the human glioblastoma U-87 MG cell line. PLoS One. 2017 Sep 6;12(9):e0184324.
- Hamdoun S, Jung P, Efferth T. Drug Repurposing of the Anthelmintic Niclosamide to Treat Multidrug-Resistant Leukemia. Front Pharmacol. 2017 Mar 10;8:110.
- Hsu CW, Huang R, Khuc T, Shou D, Bullock J, Grooby S, Griffin S, Zou C, Little A, Astley H, Xia M. Identification of approved and investigational drugs that inhibit hypoxia-inducible factor-1 signaling. Oncotarget. 2016 Feb 16;7(7):8172-83.
- Huang M, Qiu Q, Zeng S, Xiao Y, Shi M, Zou Y, Ye Y, Liang L, Yang X, Xu H. Niclosamide inhibits the inflammatory and angiogenic activation of human umbilical vein endothelial cells. Inflamm Res. 2015 Dec;64(12):1023-32.
- Jeengar MK, Kumar S, Shrivastava S, P SN et al. Niclosamide exerts anti-tumor activity through generation of reactive oxygen species and by suppression of Wnt/ β-catenin signaling axis in HGC-27, MKN-74 human gastric cancer cells. Asia-Pac J Oncol 2020.
- Jiang H, Li AM, Ye J. The magic bullet: Niclosamide. Front Oncol. 2022 Nov 21;12:1004978.
- Jin Y, Lu Z, Ding K, Li J, Du X, Chen C, Sun X, Wu Y, Zhou J, Pan J. Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: inactivation of the NF-kappaB pathway and generation of reactive oxygen species. Cancer Res. 2010 Mar 15;70(6):2516-27.
- Kaushal JB, Bhatia R, Kanchan RK, Raut P, Mallapragada S, Ly QP, Batra SK, Rachagani S. Repurposing Niclosamide for Targeting Pancreatic Cancer by Inhibiting Hh/Gli Non-Canonical Axis of Gsk3β. Cancers (Basel). 2021 Jun 22;13(13):3105.
- Kulthawatsiri T, Kittirat Y, Phetcharaburanin J, Tomacha J, Promraksa B, Wangwiwatsin A, Klanrit P, Titapun A, Loilome W, Namwat N. Metabolomic analyses uncover an inhibitory effect of niclosamide on mitochondrial membrane potential in cholangiocarcinoma cells. PeerJ. 2023 Nov 22;11:e16512.
- Kumar R, Coronel L, Somalanka B, Raju A, Aning OA, An O, Ho YS, Chen S, Mak SY, Hor PY, Yang H, Lakshmanan M, Itoh H, Tan SY, Lim YK, Wong APC, Chew SH, Huynh TH, Goh BC, Lim CY, Tergaonkar V, Cheok CF. Mitochondrial uncoupling reveals a novel therapeutic opportunity for p53-defective cancers. Nat Commun. 2018 Sep 26;9(1):3931.
- Lee MC, Chen YK, Hsu YJ, Lin BR. Niclosamide inhibits the cell proliferation and enhances the responsiveness of esophageal cancer cells to chemotherapeutic agents. Oncol Rep. 2020 Feb;43(2):549-561.
- Li Y, Li PK, Roberts MJ, Arend RC, Samant RS, Buchsbaum DJ. Multi-targeted therapy of cancer by niclosamide: A new application for an old drug. Cancer Lett. 2014 Jul 10;349(1):8-14.
- Lu L, Dong J, Wang L, Xia Q, Zhang D, Kim H, Yin T, Fan S, Shen Q. Activation of STAT3 and Bcl-2 and reduction of reactive oxygen species (ROS) promote radioresistance in breast cancer and overcome of radioresistance with niclosamide. Oncogene. 2018 Sep;37(39):5292-5304.
- Lu W, Lin C, Roberts MJ, Waud WR, Piazza GA, Li Y. Niclosamide suppresses cancer cell growth by inducing Wnt co-receptor LRP6 degradation and inhibiting the Wnt/β-catenin pathway. PLoS One. 2011;6(12):e29290.
- Luo F, Luo M, Rong QX, Zhang H, Chen Z, Wang F, Zhao HY, Fu LW. Niclosamide, an antihelmintic drug, enhances efficacy of PD-1/PD-L1 immune checkpoint blockade in non-small cell lung cancer. J Immunother Cancer. 2019 Sep 11;7(1):245.
- Mathew M, Sivaprakasam S, Dharmalingam-Nandagopal G, Sennoune SR, Nguyen NT, Jaramillo-Martinez V, Bhutia YD, Ganapathy V. Induction of Oxidative Stress and Ferroptosis in Triple-Negative Breast Cancer Cells by Niclosamide via Blockade of the Function and Expression of SLC38A5 and SLC7A11. Antioxidants (Basel). 2024 Feb 27;13(3):291.
- Mito S, Cheng B, Garcia BA, Yee Ooi X, Gonzalez D, Ruiz TC, Elisarraras FX, Tsin A. SAR study of niclosamide derivatives for neuroprotective function in SH-SY5Y neuroblastoma. Bioorg Med Chem Lett. 2023 Nov 15;96:129498.
- Pan JX, Ding K, Wang CY. Niclosamide, an old antihelminthic agent, demonstrates antitumor activity by blocking multiple signaling pathways of cancer stem cells. Chinese Journal of Cancer. 2012 Apr;31(4):178-184.
- Ren J, Wang B, Wu Q, Wang G. Combination of niclosamide and current therapies to overcome resistance for cancer: New frontiers for an old drug. Biomed Pharmacother. 2022 Nov;155:113789.
- Sennoune SR, Nandagopal GD, Ramachandran S, Mathew M, Sivaprakasam S, Jaramillo-Martinez V, Bhutia YD, Ganapathy V. Potent Inhibition of Macropinocytosis by Niclosamide in Cancer Cells: A Novel Mechanism for the Anticancer Efficacy for the Antihelminthic. Cancers (Basel). 2023 Jan 26;15(3):759.
- Shangguan F, Liu Y, Ma L, Qu G, Lv Q, An J, Yang S, Lu B, Cao Q. Niclosamide inhibits ovarian carcinoma growth by interrupting cellular bioenergetics. J Cancer. 2020 Mar 13;11(12):3454-3466.
- Suliman MA, Zhang Z, Na H, Ribeiro AL, Zhang Y, Niang B, Hamid AS, Zhang H, Xu L, Zuo Y. Niclosamide inhibits colon cancer progression through downregulation of the Notch pathway and upregulation of the tumor suppressor miR-200 family. Int J Mol Med. 2016 Sep;38(3):776-84.
- Tanaka T, Asano T, Okui T, Kuraoka S, Singh SA, Aikawa M, Aikawa E. Computational Screening Strategy for Drug Repurposing Identified Niclosamide as Inhibitor of Vascular Calcification. Front Cardiovasc Med. 2022 Jan 20;8:826529.
- Wang C, Zhou X, Xu H, Shi X, Zhao J, Yang M, Zhang L, Jin X, Hu Y, Li X, Xiao X, Liao M. Niclosamide Inhibits Cell Growth and Enhances Drug Sensitivity of Hepatocellular Carcinoma Cells via STAT3 Signaling Pathway. J Cancer. 2018 Oct 18;9(22):4150-4155.
- Wang LH, Xu M, Fu LQ, Chen XY, Yang F. The Antihelminthic Niclosamide Inhibits Cancer Stemness, Extracellular Matrix Remodeling, and Metastasis through Dysregulation of the Nuclear β-catenin/c-Myc axis in OSCC. Sci Rep. 2018 Aug 24;8(1):12776.
- Wang YC, Chao TK, Chang CC, Yo YT, Yu MH, Lai HC. Drug screening identifies niclosamide as an inhibitor of breast cancer stem-like cells. PLoS One. 2013 Sep 18;8(9):e74538.
- Wang Z, Ren J, Du J, Wang H, Liu J, Wang G. Niclosamide as a Promising Therapeutic Player in Human Cancer and Other Diseases. Int J Mol Sci. 2022 Dec 17;23(24):16116.
- Xiang M, Chen Z, Yang D, Li H, Zuo Y, Li J, Zhang W, Zhou H, Jiang D, Xu Z, Yu Z. Niclosamide enhances the antitumor effects of radiation by inhibiting the hypoxia-inducible factor-1α/vascular endothelial growth factor signaling pathway in human lung cancer cells. Oncol Lett. 2017 Aug;14(2):1933-1938.
- Yeh LT, Lin CW, Lu KH, Hsieh YH, Yeh CB, Yang SF, Yang JS. Niclosamide Suppresses Migration and Invasion of Human Osteosarcoma Cells by Repressing TGFBI Expression via the ERK Signaling Pathway. Int J Mol Sci. 2022 Jan 1;23(1):484.
- Zhang Q, Yang Z, Hao X, Dandreo LJ, He L, Zhang Y, Wang F, Wu X, Xu L. Niclosamide improves cancer immunotherapy by modulating RNA-binding protein HuR-mediated PD-L1 signaling. Cell Biosci. 2023 Oct 17;13(1):192.
Sulfasalazine:
Mechanisms of action:
- Targeting xCT antiporter and redox homeostasis to induce oxidative stress: Sulfasalazine inhibits the xCT cystine/glutamate antiporter, reducing cystine uptake essential for intracellular glutathione (GSH) synthesis. This disruption of cystine availability depletes GSH, leading to oxidative stress through increased reactive oxygen species (ROS). The imbalance in redox homeostasis undermines cancer cells’ antioxidant defenses, triggering nutrient starvation, autophagic processes, apoptotic cell death, and heightened susceptibility to oxidative damage.
- Reduction of tumor growth across multiple cancers: Sulfasalazine suppresses growth in various cancers, including prostate cancer, hepatocellular carcinoma (HCC), lymphoma, and breast cancer, both in vitro and in vivo.
- Selective cytotoxicity: Sulfasalazine preferentially affects cancer cells due to their higher dependency on extracellular cystine, sparing normal cells and minimizing toxicity.
- Targeting cancer stem cells: Sulfasalazine effectively eliminates cancer stem-like cells by disrupting redox balance, reducing tumor recurrence and aggressiveness.
- Suppression of chemotherapy resistance: By depleting GSH, sulfasalazine enhances the efficacy of chemotherapeutic agents, overcoming glutathione-mediated drug resistance.
- Synergistic effects with other treatments: Sulfasalazine enhances the efficacy of ROS-inducing therapies, such as vitamin C and chemotherapeutic drugs, by complementing their mechanisms of action.
- Inhibition of tumor microenvironment support: Sulfasalazine disrupts the tumor microenvironment by reducing macrophage-mediated cysteine supply and impairing stromal cell support, starving cancer cells of critical nutrients.
- Induction of autophagy and apoptosis: Sulfasalazine-mediated ROS accumulation activates autophagic pathways and apoptotic signaling, contributing to cancer cell death.
- Reduction of metastasis and invasiveness: By inhibiting xCT-related signaling, sulfasalazine reduces cancer cell invasiveness and metastatic potential, particularly in aggressive cancers.
Citations:
- Cramer SL, Saha A, Liu J, Tadi S, Tiziani S, Yan W, Triplett K, Lamb C, Alters SE, Rowlinson S, Zhang YJ, Keating MJ, Huang P, DiGiovanni J, Georgiou G, Stone E. Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth. Nat Med. 2017 Jan;23(1):120-127.
- Doxsee DW, Gout PW, Kurita T, Lo M, Buckley AR, Wang Y, Xue H, Karp CM, Cutz JC, Cunha GR, Wang YZ. Sulfasalazine-induced cystine starvation: potential use for prostate cancer therapy. Prostate. 2007 Feb 1;67(2):162-71.
- Gout PW, Buckley AR, Simms CR, Bruchovsky N. Sulfasalazine, a potent suppressor of lymphoma growth by inhibition of the x(c)- cystine transporter: a new action for an old drug. Leukemia. 2001 Oct;15(10):1633-40.
- Gout PW, Simms CR, Robertson MC. In vitro studies on the lymphoma growth-inhibitory activity of sulfasalazine. Anticancer Drugs. 2003 Jan;14(1):21-9.
- Guo W, Zhao Y, Zhang Z, Tan N, Zhao F, Ge C, Liang L, Jia D, Chen T, Yao M, Li J, He X. Disruption of xCT inhibits cell growth via the ROS/autophagy pathway in hepatocellular carcinoma. Cancer Lett. 2011 Dec 15;312(1):55-61.
- Lay JD, Hong CC, Huang JS, Yang YY, Pao CY, Liu CH, Lai YP, Lai GM, Cheng AL, Su IJ, Chuang SE. Sulfasalazine suppresses drug resistance and invasiveness of lung adenocarcinoma cells expressing AXL. Cancer Res. 2007 Apr 15;67(8):3878-87.
- Lo M, Ling V, Low C, Wang YZ, Gout PW. Potential use of the anti-inflammatory drug, sulfasalazine, for targeted therapy of pancreatic cancer. Curr Oncol. 2010 Jun;17(3):9-16.
- Lo M, Wang YZ, Gout PW. The x(c)- cystine/glutamate antiporter: a potential target for therapy of cancer and other diseases. J Cell Physiol. 2008 Jun;215(3):593-602.
- Shin CS, Mishra P, Watrous JD, Carelli V, D’Aurelio M, Jain M, Chan DC. The glutamate/cystine xCT antiporter antagonizes glutamine metabolism and reduces nutrient flexibility. Nat Commun. 2017 Apr 21;8:15074.
- Thanee M, Padthaisong S, Suksawat M, Dokduang H, Phetcharaburanin J, Klanrit P, Titapun A, Namwat N, Wangwiwatsin A, Sa-Ngiamwibool P, Khuntikeo N, Saya H, Loilome W. Sulfasalazine modifies metabolic profiles and enhances cisplatin chemosensitivity on cholangiocarcinoma cells in in vitro and in vivo models. Cancer Metab. 2021 Mar 16;9(1):11.
- Zheng Z, Luo G, Shi X, Long Y, Shen W, Li Z, Zhang X. The Xc– inhibitor sulfasalazine improves the anti-cancer effect of pharmacological vitamin C in prostate cancer cells via a glutathione-dependent mechanism. Cell Oncol (Dordr). 2020 Feb;43(1):95-1069Re