Treatment Approach

In research published in 2004 and again in 2023, many contemporary cancer drugs were found to have 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 looking for novel ways to fight cancer to help improve the chances for long-term remission. One potentially promising way is to integrate repurposed medications and targeted supplements into conventional therapy. The rationale for combined interventions for cancer treatment is multifaceted:

  1. Increased efficacy: Combination therapies are often more effective than monotherapy. Using compounds that work through different mechanisms increases the likelihood of killing more cancer cells.
  2. Overcoming drug resistance: Cancer cells can become resistant to a single drug over time. Using a combination of repurposed medications and targeted supplements may reduce the likelihood of this happening, as the cancer cells would need to simultaneously develop resistance to multiple drugs.
  3. Addressing tumor heterogeneity: Tumors are often composed of different types of cells. A combination of drugs might more effectively target these different cell types.
  4. Synergistic effects: Some drugs may enhance the effects of others, leading to a greater overall impact than if the drugs were used individually.
  5. Targeting multiple pathways: Cancer cells often have alterations in multiple cellular pathways. Using a combination of compounds allows for the targeting of multiple pathways, which may lead to more effective treatment.

Targeting key vulnerabilities of cancer:

Cancer cells and their stem cell counterparts are marked by distinct metabolic characteristics and vulnerabilities, including an overreliance on aerobic glycolysis, known as the Warburg effect, and an increased generation of reactive oxygen species (ROS). These peculiarities fuel their rapid proliferation and create specific Achilles’ heels that may be exploited for potential therapeutic gain. By inducing energy restriction or manipulating ROS levels, alongside targeting the disrupted cell signaling pathways that enable cancer cells to grow uncontrollably and evade death, it becomes possible to eliminate these cells selectively. This approach, focusing on the metabolic and signaling vulnerabilities of cancer, offers a potentially promising avenue for targeting cancer initiation, proliferation, metastasis, immune evasion, and treatment resistance. The numerous scientific references at the bottom provide a solid foundation for the key points below.

Energy metabolism:

Cancer cells and cancer stem cells exhibit metabolic qualities that render them particularly susceptible to energy restriction. These cells typically rely on aerobic glycolysis, a less efficient form of energy production known as the Warburg effect, to fuel their rapid proliferation. This metabolic shift allows them to generate building blocks for new cells but also makes them heavily dependent on a constant supply of glucose and vulnerable to energy stress. Agents that induce a state of energy restriction may challenge the cancer cells’ metabolic flexibility and ability to sustain their rapid growth and division. This approach may lead to energy stress within the tumor microenviron­ment, selectively impairing cancer cells more than normal cells due to the former’s heightened energy demands and reduced metabolic flexibility. When energy availability is restricted, cancer cells struggle to main­tain their metabolic needs, leading to impaired growth and survival. 

Reactive oxygen species:

Cancer’s altered metabolism results in an increased generation of ROS, which, at controlled levels, can promote cancer cell proliferation and survival. However, when ROS levels become excessive, they can cause significant damage to cellular components, leading to overwhelming oxidative stress and subsequent cell death. Exploiting these vulnerabilities therapeutically offers a potentially promising strategy for targeting cancer cells and cancer stem cells. Agents that increase ROS levels beyond the threshold that cancer cells can manage or inhibit their antioxidant defenses may exacerbate oxidative damage, leading to cell death. 

Pro-survival signaling pathways:

In healthy cells, intricate cell signaling pathways act like traffic lights, regulating growth, division, and death. However, in cancer, mutations can jam these “lights,” triggering uncontrolled cell growth and spread. Aberrant signaling, often involving growth factors and receptor proteins, allows cancer cells to proliferate without proper checks, evade programmed cell death (apoptosis), and acquire traits like increased mobility and invasion. Fortunately, this dependence on altered signaling presents an potential therapeutic opportunity. By targeting specific components of these pathways, the goal is to disrupt the cancer’s communication network, hindering its ability to grow, resist therapy, and spread, offering a potentially promising avenue for cancer treatment and prevention.

In summary, therapeutically inducing energy restriction, manipulating reactive oxygen species (ROS) levels, and disrupting aberrant cell signaling pathways, our goal is to selectively target cancer cells and cancer stem cells for destruction. By focusing on these critical aspects, an integrated approach represents a potentially promising new direction for cancer treatment according to research.

A more unified approach to cancer treatment:

 If one were to design an integrative approach to cancer treatment, it should have the following features:
 
  1. Minimal pill burden (i.e., fewer pills to take)
  2. Applicable to many cancer types and stages
  3. Well tolerated with low systemic toxicity
  4. Focuses on key vulnerabilities of cancer, especially energy restriction, excessive reactive oxygen species, and cell-signaling inhibition
  5. Targets bulk tumor cells and cancer stem cells (cancer stem cells are at the root of cancer initiation, metastasis, treatment resistance, and cancer recurrence)
  6. Instructs on proper diet and lifestyle
  7. Promotes immune destruction of cancer cells and removal of tumor debris
  8. Optimizes the gut microbiome to enhance the efficacy of conventional cancer treatment and reduce  treatment-related toxicity
  9. Provides guidance to help navigate the complex landscape of conventional cancer treatment options
 

Looking carefully at the published scientific literature (scroll down to see the extensive reference list), the following compounds, when taken together, helps to unify the features listed above:

  1. Anti-cancer diet and lifestyle (click here)
  2. D-mannose
  3. Docosahexaenoic acid
  4. High-ozonide ozonated oil
  5. Niclosamide (liposomal)
  6. Disulfiram (liposomal)
  7. Piperlongumine (liposomal)
  8. Broad spectrum probiotic + prebiotic

Click here to learn the details about the various anticancer mechanisms of action of the above items.

Niclosamide, disulfiram, and piperlongumine have demonstrated to have significant anti-cancer properties but face these significant challenges:

  1. Low water solubility limits absorption
  2. Rapid degradation in the bloodstream reduces systemic availability
  3. Poor cellular uptake restricts anti-cancer effects

Liposomalization may mitigate these issues by:

  1. Maximizing bioavailability: Liposomes enhance the solubility of niclosamide, disulfiram, and piperlongumine, thereby helping the drugs to be better absorbed into the systemic circulation.
  2. Extending the duration of action: The liposomal format may extend the duration of action through the sustained release of the drugs in the bloodstream to help promote a longer-lasting impact on cancer cells.
  3. Enhancing cellular uptake: Due to their structural similarity to cell membranes, liposomes can fuse with these membranes, better enabling them to transport the drugs directly into cancer cells. This process may result in elevated levels of the drugs inside the cells.

A more unified approach to fighting cancer combines energy restriction, ROS manipulation, and disruption of signaling pathways. Utilizing compounds like D-mannose, docosahexaenoic acid, high-ozonide ozonated oil, niclosamide, disulfiram, and piperlongumine, this strategy is designed to exploit cancer’s metabolic and cell-signaling anomalies. It seeks to reduce pill burden, enhance treatment efficacy across cancer types, reduce treatment toxicity, and target both cancer cells and cancer stem cells. This approach may enhance conventional treatments and boost the immune response against cancer, providing a potentially promising path to overcome metastasis, treatment resistance, and disease recurrence.

Here are the types of cancer that we treat:

  • Bladder
  • Brain
  • Breast
  • Cervical
  • Colorectal
  • Esophageal
  • Germinoma
  • Head & neck
  • Kidney
  • Leukemia
  • Liver
  • Lung
  • Lymphoma
  • Melanoma
  • Myeloma
  • Ovarian
  • Pancreatic
  • Prostate
  • Sarcoma
  • Stomach
  • Testicular
  • Thyroid
  • UterineStage

Treatment indications:

Indications for our treatment approach include the following:

  1. Patients seeking ways to potentially improve the effectiveness and/or reduce the side effects of conventional cancer treatment, including surgery, chemotherapy, radiation, targeted therapy, and immunotherapy.
  2. Patients who have failed or can no longer tolerate conventional treatment.
  3. Patients who do not qualify for a drug trial or failed to respond favorably to a trial drug.
  4. Patients wanting to prolong their remission with health-enhancing strategies.
  5. Patients seeking to improve their quality of life.

Patient care and treatment cost:

An oncologist typically manages the care of 250-500 patients simultaneously. In contrast, Dr. Thomas prioritizes highly individualized attention by capping his practice to no more than than 50 patients. This elevates the quality of his care and allows him the time needed for ongoing research om behalf of his patients.

The protocol 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.

The cost for our services is a flat rate of $2850 per month. This is less expensive than alternative cancer treatment often sought in Mexico, Central America, the Caribbean, and Europe. In these locations, clinics typically charge $7,000 to $20,000 weekly for medical treatment or $40,000 to $120,000 for highly-experimental procedures

Our monthly fee includes the cost of custom-compounded liposomal disulfiram and niclosamide. Other costs are for items available online (D-mannose, docosahexaenoic acid, high-ozonide ozonated oil, liposomal piperlongumine). These total around $500 per month. Treatment is maintained until remission or disease stabilization is achieved. Other than covering the cost of bloodwork, health insurance and Medicare do not pay for integrative cancer treatment.

References:

  1. Adorno-Cruz V, Kibria G, Liu X, Doherty M, Junk DJ, Guan D, Hubert C, Venere M, Mulkearns-Hubert E, Sinyuk M, Alvarado A, Caplan AI, Rich J, Gerson SL, Lathia J, Liu H. Cancer stem cells: targeting the roots of cancer, seeds of metastasis, and sources of therapy resistance. Cancer Res. 2015 Mar 15;75(6):924-9.
  2. Al Hadeethi S, El-Baba C, Araji K, Hayar B, Cheikh IA, El-Khoury R, Usta J, Darwiche N. Mannose Inhibits the Pentose Phosphate Pathway in Colorectal Cancer and Enhances Sensitivity to 5-Fluorouracil Therapy. Cancers (Basel). 2023 Apr 13;15(8):2268.
  3. Allensworth JL, Evans MK, Bertucci F, Aldrich AJ, Festa RA, Finetti P, Ueno NT, Safi R, McDonnell DP, Thiele DJ, Van Laere S, Devi GR. Disulfiram (DSF) acts as a copper ionophore to induce copper-dependent oxidative stress and mediate anti-tumor efficacy in inflammatory breast cancer. Mol Oncol. 2015 Jun;9(6):1155-68.

  4. Balamurugan K. HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int J Cancer. 2016 Mar 1;138(5):1058-66.

  5. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014 Mar 27;157(1):121-41. 

  6. Beyer MP, Videla LA, Farías C, Valenzuela R. Potential Clinical Applications of Pro-Resolving Lipids Mediators from Docosahexaenoic Acid. Nutrients. 2023 Jul 26;15(15):3317.
  7. Bougnoux P, Hajjaji N, Ferrasson MN, Giraudeau B, Couet C, Le Floch O. Improving outcome of chemotherapy of metastatic breast cancer by docosahexaenoic acid: a phase II trial. Br J Cancer. 2009 Dec 15;101(12):1978-85.
  8. Brar SS, Grigg C, Wilson KS, Holder WD Jr, Dreau D, Austin C, Foster M, Ghio AJ, Whorton AR, Stowell GW, Whittall LB, Whittle RR, White DP, Kennedy TP. Disulfiram inhibits activating transcription factor/cyclic AMP-responsive element binding protein and human melanoma growth in a metal-dependent manner in vitro, in mice and in a patient with metastatic disease. Mol Cancer Ther. 2004 Sep;3(9):1049-60.
  9. Bu W, Wang Z, Meng L, Li X, Liu X, Chen Y, Xin Y, Li B, Sun H. Disulfiram inhibits epithelial-mesenchymal transition through TGFβ-ERK-Snail pathway independently of Smad4 to decrease oral squamous cell carcinoma metastasis. Cancer Manag Res. 2019 May 1;11:3887-3898.
  10. Cao HZ, Yang WT, Zheng PS. Cytotoxic effect of disulfiram/copper on human cervical cancer cell lines and LGR5-positive cancer stem-like cells. BMC Cancer. 2022 May 9;22(1):521.
  11. Chen W, Lian W, Yuan Y, Li M. The synergistic effects of oxaliplatin and piperlongumine on colorectal cancer are mediated by oxidative stress. Cell Death Dis. 2019 Aug 8;10(8):600.

  12. Chen YJ, Kuo CC, Ting LL, Lu LS, Lu YC, Cheng AJ, Lin YT, Chen CH, Tsai JT, Chiou JF. Piperlongumine inhibits cancer stem cell properties and regulates multiple malignant phenotypes in oral cancer. Oncol Lett. 2018 Feb;15(2):1789-1798.
  13. 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.
  14. Choi SA, Choi JW, Wang KC, Phi JH, Lee JY, Park KD, Eum D, Park SH, Kim IH, Kim SK. Disulfiram modulates stemness and metabolism of brain tumor initiating cells in atypical teratoid/rhabdoid tumors. Neuro Oncol. 2015 Jun;17(6):810-21.
  15. Colas S, Paon L, Denis F, Prat M, Louisot P, Hoinard C, Le Floch O, Ogilvie G, Bougnoux P. Enhanced radiosensitivity of rat autochthonous mammary tumors by dietary docosahexaenoic acid. Int J Cancer. 2004 Apr 10;109(3):449-54.
  16. Deng YL, Liu R, Cai ZD, Han ZD, Feng YF, Cai SH, Chen QB, Zhu JG, Zhong WD. Mannose inhibits the growth of prostate cancer through a mitochondrial mechanism. Asian J Androl. 2022 Sep-Oct;24(5):540-548.
  17. Dhanalakshmi M, Sruthi D, Jinuraj KR, Das K, Dave S, Andal NM, Das J. Mannose: a potential saccharide candidate in disease management. Med Chem Res. 2023;32(3):391-408.
  18. Dhillon H, Chikara S, Reindl KM. Piperlongumine induces pancreatic cancer cell death by enhancing reactive oxygen species and DNA damage. Toxicol Rep. 2014;1:309-318.

  19. Ewaschuk JB, Newell M, Field CJ. Docosahexanoic acid improves chemotherapy efficacy by inducing CD95 translocation to lipid rafts in ER(-) breast cancer cells. Lipids. 2012 Nov;47(11):1019-30.
  20. Falls-Hubert KC, Butler AL, Gui K, Anderson M, Li M, Stolwijk JM, Rodman SN 3rd, Solst SR, Tomanek-Chalkley A, Searby CC, Sheffield VC, Sandfort V, Schmidt H, McCormick ML, Wels BR, Allen BG, Buettner GR, Schultz MK, Spitz DR. Disulfiram causes selective hypoxic cancer cell toxicity and radio-chemo-sensitization via redox cycling of copper. Free Radic Biol Med. 2020 Apr;150:1-11.
  21. Farooq MA, Aquib M, Khan DH, Hussain Z, Ahsan A, Baig MMFA, Wande DP, Ahmad MM, Ahsan HM, Jiajie J, Wang B. Recent advances in the delivery of disulfiram: a critical analysis of promising approaches to improve its pharmacokinetic profile and anticancer efficacy. Daru. 2019 Dec;27(2):853-862.
  22. Gao X, Huang H, Pan C, Mei Z, Yin S, Zhou L, Zheng S. Disulfiram/Copper Induces Immunogenic Cell Death and Enhances CD47 Blockade in Hepatocellular Carcinoma. Cancers (Basel). 2022 Sep 28;14(19):4715.
  23. Gonzalez PS, O’Prey J, Cardaci S, Barthet VJA, Sakamaki JI, Beaumatin F, Roseweir A, Gay DM, Mackay G, Malviya G, Kania E, Ritchie S, Baudot AD, Zunino B, Mrowinska A, Nixon C, Ennis D, Hoyle A, Millan D, McNeish IA, Sansom OJ, Edwards J, Ryan KM. Mannose impairs tumour growth and enhances chemotherapy. Nature. 2018 Nov;563(7733):719-723.
  24. Guo W, Zhang X, Lin L, Wang H, He E, Wang G, Zhao Q. The disulfiram/copper complex induces apoptosis and inhibits tumour growth in human osteosarcoma by activating the ROS/JNK signalling pathway. J Biochem. 2021 Oct 11;170(2):275-287.
  25. Hamdoun S, Jung P, Efferth T. Drug Repurposing of the Anthelmintic Niclosamide to Treat Multidrug-Resistant Leukemia. Front Pharmacol. 2017 Mar 10;8:110.
  26. Harada Y, Mizote Y, Suzuki T, Hirayama A, Ikeda S, Nishida M, Hiratsuka T, Ueda A, Imagawa Y, Maeda K, Ohkawa Y, Murai J, Freeze HH, Miyoshi E, Higashiyama S, Udono H, Dohmae N, Tahara H, Taniguchi N. Metabolic clogging of mannose triggers dNTP loss and genomic instability in human cancer cells. Elife. 2023 Jul 18;12:e83870.
  27. Hatamipour M, Jaafari MR, Momtazi-Borojeni AA, Ramezani M, Sahebkar A. Nanoliposomal Encapsulation Enhances In Vivo Anti-Tumor Activity of Niclosamide against Melanoma. Anticancer Agents Med Chem. 2019;19(13):1618-1626.
  28. 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.

  29. Huang LH, Chung HY, Su HM. Docosahexaenoic acid reduces sterol regulatory element binding protein-1 and fatty acid synthase expression and inhibits cell proliferation by inhibiting pAkt signaling in a human breast cancer MCF-7 cell line. BMC Cancer. 2017 Dec 28;17(1):890.
  30. 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.
  31. Iljin K, Ketola K, Vainio P, Halonen P, Kohonen P, Fey V, Grafström RC, Perälä M, Kallioniemi O. High-throughput cell-based screening of 4910 known drugs and drug-like small molecules identifies disulfiram as an inhibitor of prostate cancer cell growth. Clin Cancer Res. 2009 Oct 1;15(19):6070-8.

  32. Infantino V, Santarsiero A, Convertini P, Todisco S, Iacobazzi V. Cancer Cell Metabolism in Hypoxia: Role of HIF-1 as Key Regulator and Therapeutic Target. Int J Mol Sci. 2021 May 27;22(11):5703.

  33. 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.
  34. Jalali Motlagh N, Wang C, Kuellenberg EG, Wojtkiewicz GR, Schmidt S, Chen JW. D-Mannose Slows Glioma Growth by Modulating Myeloperoxidase Activity. Cancers (Basel). 2021 Dec 18;13(24):6360.
  35. 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.
  36. Jeong DW, Lee S, Chun YS. How cancer cells remodel lipid metabolism: strategies targeting transcription factors. Lipids Health Dis. 2021 Nov 14;20(1):163.
  37. Jeong S, Jing K, Kim N, Shin S, Kim S, Song KS, Heo JY, Park JH, Seo KS, Han J, Wu T, Kweon GR, Park SK, Park JI, Lim K. Docosahexaenoic acid-induced apoptosis is mediated by activation of mitogen-activated protein kinases in human cancer cells. BMC Cancer. 2014 Jul 3;14:481.
  38. Jia Y, Huang T. Overview of Antabuse® (Disulfiram) in Radiation and Cancer Biology. Cancer Manag Res. 2021 May 20;13:4095-4101.
  39. Jiang H, Li AM, Ye J. The magic bullet: Niclosamide. Front Oncol. 2022 Nov 21;12:1004978.
  40. Jiao Y, Hannafon BN, Zhang RR, Fung KM, Ding WQ. Docosahexaenoic acid and disulfiram act in concert to kill cancer cells: a mutual enhancement of their anticancer actions. Oncotarget. 2017 Mar 14;8(11):17908-17920.
  41. Jin H, Liu X, Liu HX. Biological function, regulatory mechanism, and clinical application of mannose in cancer. Biochim Biophys Acta Rev Cancer. 2023 Nov;1878(6):188970.
  42. 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.
  43. Kang KS, Wang P, Yamabe N, Fukui M, Jay T, Zhu BT. Docosahexaenoic acid induces apoptosis in MCF-7 cells in vitro and in vivo via reactive oxygen species formation and caspase 8 activation. PLoS One. 2010 Apr 22;5(4):e10296.
  44. Kang X, Jadhav S, Annaji M, Huang CH, Amin R, Shen J, Ashby CR Jr, Tiwari AK, Babu RJ, Chen P. Advancing Cancer Therapy with Copper/Disulfiram Nanomedicines and Drug Delivery Systems. Pharmaceutics. 2023 May 23;15(6):1567.
  45. Kannappan V, Ali M, Small B, Rajendran G, Elzhenni S, Taj H, Wang W, Dou QP. Recent Advances in Repurposing Disulfiram and Disulfiram Derivatives as Copper-Dependent Anticancer Agents. Front Mol Biosci. 2021 Sep 17;8:741316.
  46. Karsa M, Xiao L, Ronca E, Bongers A, Spurling D, Karsa A, Cantilena S, Mariana A, Failes TW, Arndt GM, Cheung LC, Kotecha RS, Sutton R, Lock RB, Williams O, de Boer J, Haber M, Norris MD, Henderson MJ, Somers K. FDA-approved disulfiram as a novel treatment for aggressive leukemia. J Mol Med (Berl). 2024 Apr;102(4):507-519.
  47. 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.
  48. Khan NA, Nishimura K, Aires V, Yamashita T, Oaxaca-Castillo D, Kashiwagi K, Igarashi K. Docosahexaenoic acid inhibits cancer cell growth via p27Kip1, CDK2, ERK1/ERK2, and retinoblastoma phosphorylation. J Lipid Res. 2006 Oct;47(10):2306-13.
  49. Kim JY, Cho Y, Oh E, Lee N, An H, Sung D, Cho TM, Seo JH. Disulfiram targets cancer stem-like properties and the HER2/Akt signaling pathway in HER2-positive breast cancer. Cancer Lett. 2016 Aug 28;379(1):39-48.
  50. 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.
  51. 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.
  52. Kung FP, Lim YP, Chao WY, Zhang YS, Yu HI, Tai TS, Lu CH, Chen SH, Li YZ, Zhao PW, Yen YP, Lee YR. Piperlongumine, a Potent Anticancer Phytotherapeutic, Induces Cell Cycle Arrest and Apoptosis In Vitro and In Vivo through the ROS/Akt Pathway in Human Thyroid Cancer Cells. Cancers (Basel). 2021 Aug 24;13(17):4266.

  53. Lavy M, Gauttier V, Poirier N, Barillé-Nion S, Blanquart C. Specialized Pro-Resolving Mediators Mitigate Cancer-Related Inflammation: Role of Tumor-Associated Macrophages and Therapeutic Opportunities. Front Immunol. 2021 Jun 30;12:702785.
  54. 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.
  55. Li J, Sharkey CC, King MR. Piperlongumine and immune cytokine TRAIL synergize to promote tumor death. Sci Rep. 2015 May 18;5:9987.

  56. Li P, Sun Q, Bai S, Wang H, Zhao L. Combination of the cuproptosis inducer disulfiram and anti-PD-L1 abolishes NSCLC resistance by ATP7B to regulate the HIF-1 signaling pathway. Int J Mol Med. 2024 Feb;53(2):19.
  57. Li Y, Chen F, Chen J, Chan S, He Y, Liu W, Zhang G. Disulfiram/Copper Induces Antitumor Activity against Both Nasopharyngeal Cancer Cells and Cancer-Associated Fibroblasts through ROS/MAPK and Ferroptosis Pathways. Cancers (Basel). 2020 Jan 6;12(1):138.

  58. 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.
  59. Lian S, Xia Y, Nguyen TT, Ung TT, Yoon HJ, Kim NH, Kim KK, Jung YD. Docosahexaenoic Acid Inhibits Tumor Promoter-Induced Urokinase-Type Plasminogen Activator Receptor by Suppressing PKCδ- and MAPKs-Mediated Pathways in ECV304 Human Endothelial Cells. PLoS One. 2016 Sep 21;11(9):e0163395.
  60. Lin H, Sun F, Li T, Zhang Y, Guo X, Li M, Liang M, Zhou X, Fang Z. Disulfiram enhances chemotherapeutic effects of doxorubicin liposomes against human hepatocellular carcinoma via activating ROS-induced cell stress response pathways. Cancer Chemother Pharmacol. 2022 Dec;90(6):455-465.
  61. Lin TH, Kuo CH, Zhang YS, Chen PT, Chen SH, Li YZ, Lee YR. Piperlongumine Induces Cellular Apoptosis and Autophagy via the ROS/Akt Signaling Pathway in Human Follicular Thyroid Cancer Cells. Int J Mol Sci. 2023 Apr 28;24(9):8048.

  62. Liu F, Xu X, Li C, Li C, Li Y, Yin S, Yu S, Chen XQ. Mannose synergizes with chemoradiotherapy to cure cancer via metabolically targeting HIF-1 in a novel triple-negative glioblastoma mouse model. Clin Transl Med. 2020 Nov;10(7):e226.
  63. Liu F, Zhou Q, Jiang Hf. et al. Piperlongumine conquers temozolomide chemoradiotherapy resistance to achieve immune cure in refractory glioblastoma via boosting oxidative stress-inflamation-CD8+-T cell immunity. J Exp Clin Cancer Res 42, 118 (2023).

  64. Liu P, Kumar IS, Brown S, Kannappan V, Tawari PE, Tang JZ, Jiang W, Armesilla AL, Darling JL, Wang W. Disulfiram targets cancer stem-like cells and reverses resistance and cross-resistance in acquired paclitaxel-resistant triple-negative breast cancer cells. Br J Cancer. 2013 Oct 1;109(7):1876-85.
  65. Liu P, Wang Z, Brown S, Kannappan V, Tawari PE, Jiang W, Irache JM, Tang JZ, Armesilla AL, Darling JL, Tang X, Wang W. Liposome encapsulated Disulfiram inhibits NFκB pathway and targets breast cancer stem cells in vitro and in vivo. Oncotarget. 2014 Sep 15;5(17):7471-85.
  66. Lu C, Li X, Ren Y, Zhang X. Disulfiram: a novel repurposed drug for cancer therapy. Cancer Chemother Pharmacol. 2021 Feb;87(2):159-172.

  67. 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.
  68. 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.

  69. Lu X, Xu C, Xu Z, Lu C, Yang R, Zhang F, Zhang G. Piperlongumine inhibits the growth of non-small cell lung cancer cells via the miR-34b-3p/TGFBR1 pathway. BMC Complement Med Ther. 2021 Jan 7;21(1):15.

  70. Lunt S, Fendt S. Metabolism – A cornerstone of cancer initiation, progression, immune evasion and treatment response. Current Opinion in Systems Biology 8 (2018): 67-72.
  71. 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.
  72. Manzi L, Costantini L, Molinari R, Merendino N. Effect of Dietary ω-3 Polyunsaturated Fatty Acid DHA on Glycolytic Enzymes and Warburg Phenotypes in Cancer. Biomed Res Int. 2015;2015:137097.
  73. Maralbashi S, Aslan C, Kahroba H, Asadi M, Soltani-Zangbar MS, Haghnavaz N, Jadidi F, Salari F, Kazemi T. Docosahexaenoic acid (DHA) impairs hypoxia-induced cellular and exosomal overexpression of immune-checkpoints and immunomodulatory molecules in different subtypes of breast cancer cells. BMC Nutr. 2024 Mar 4;10(1):41.

  74. 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.
  75. Nan F, Sun Y, Liang H, Zhou J, Ma X, Zhang D. Mannose: A Sweet Option in the Treatment of Cancer and Inflammation. Front Pharmacol. 2022 May 13;13:877543.
  76. Newell M, Baker K, Postovit LM, Field CJ. A Critical Review on the Effect of Docosahexaenoic Acid (DHA) on Cancer Cell Cycle Progression. Int J Mol Sci. 2017 Aug 17;18(8):1784.
  77. Ni YL, Chien PJ, Hsieh HC, Shen HT, Lee HT, Chen SM, Chang WW. Disulfiram/Copper Suppresses Cancer Stem Cell Activity in Differentiated Thyroid Cancer Cells by Inhibiting BMI1 Expression. Int J Mol Sci. 2022 Oct 31;23(21):13276.
  78. 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.

  79. Parama D, Rana V, Girisa S, Verma E, Daimary UD, Thakur KK, Kumar A, Kunnumakkara AB. The promising potential of piperlongumine as an emerging therapeutics for cancer. Explor Target Antitumor Ther. 2021;2(4):323-354.
  80. Park M, Kim H. Anti-cancer Mechanism of Docosahexaenoic Acid in Pancreatic Carcinogenesis: A Mini-review. J Cancer Prev. 2017 Mar;22(1):1-5.
  81. Park YM, Go YY, Shin SH, Cho JG, Woo JS, Song JJ. Anti-cancer effects of disulfiram in head and neck squamous cell carcinoma via autophagic cell death. PLoS One. 2018 Sep 13;13(9):e0203069.
  82. Parveen S, Kumar S, Pal S, Yadav NP, Rajawat J, Banerjee M. Enhanced therapeutic efficacy of Piperlongumine for cancer treatment using nano-liposomes mediated delivery. Int J Pharm. 2023 Aug 25;643:123212.

  83. Pizato N, Luzete BC, Kiffer LFMV, Corrêa LH, de Oliveira Santos I, Assumpção JAF, Ito MK, Magalhães KG. Omega-3 docosahexaenoic acid induces pyroptosis cell death in triple-negative breast cancer cells. Sci Rep. 2018 Jan 31;8(1):1952.
  84. Qiu C, Zhang X, Huang B, Wang S, Zhou W, Li C, Li X, Wang J, Yang N. Disulfiram, a Ferroptosis Inducer, Triggers Lysosomal Membrane Permeabilization by Up-Regulating ROS in Glioblastoma. Onco Targets Ther. 2020 Oct 20;13:10631-10640.
  85. Rae C, Tesson M, Babich JW, Boyd M, Sorensen A, Mairs RJ. The role of copper in disulfiram-induced toxicity and radiosensitization of cancer cells. J Nucl Med. 2013 Jun;54(6):953-60.
  86. 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.
  87. Samanta S. Potential Impacts of Prebiotics and Probiotics on Cancer Prevention. Anticancer Agents Med Chem. 2022;22(4):605-628.
  88. 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.
  89. Sha J, Cao D, Cui R, Xia L, Hua X, Lu Y, Han S. Mannose Impairs Lung Adenocarcinoma Growth and Enhances the Sensitivity of A549 Cells to Carboplatin. Cancer Manag Res. 2020 Nov 3;12:11077-11083.
  90. Shan K, Feng N, Zhu D, Qu H, Fu G, Li J, Cui J, Chen H, Wang R, Qi Y, Chen YQ. Free docosahexaenoic acid promotes ferroptotic cell death via lipoxygenase dependent and independent pathways in cancer cells. Eur J Nutr. 2022 Dec;61(8):4059-4075.
  91. 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.
  92. Skrott Z, Mistrik M, Andersen KK, Friis S, Majera D, Gursky J, Ozdian T, Bartkova J, Turi Z, Moudry P, Kraus M, Michalova M, Vaclavkova J, Dzubak P, Vrobel I, Pouckova P, Sedlacek J, Miklovicova A, Kutt A, Li J, Mattova J, Driessen C, Dou QP, Olsen J, Hajduch M, Cvek B, Deshaies RJ, Bartek J. Alcohol-abuse drug disulfiram targets cancer via p97 segregase adaptor NPL4. Nature. 2017 Dec 14;552(7684):194-199.

  93. Song EA, Kim H. Docosahexaenoic Acid Induces Oxidative DNA Damage and Apoptosis, and Enhances the Chemosensitivity of Cancer Cells. Int J Mol Sci. 2016 Aug 3;17(8):1257.
  94. Sulciner ML, Serhan CN, Gilligan MM, Mudge DK, Chang J, Gartung A, Lehner KA, Bielenberg DR, Schmidt B, Dalli J, Greene ER, Gus-Brautbar Y, Piwowarski J, Mammoto T, Zurakowski D, Perretti M, Sukhatme VP, Kaipainen A, Kieran MW, Huang S, Panigrahy D. Resolvins suppress tumor growth and enhance cancer therapy. J Exp Med. 2018 Jan 2;215(1):115-140.
  95. 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.

  96. Tanaka G, Inoue K, Shimizu T, Akimoto K, Kubota K. Dual pharmacological inhibition of glutathione and thioredoxin systems synergizes to kill colorectal carcinoma stem cells. Cancer Med. 2016 Sep;5(9):2544-57.

  97. Tripathi SK, Biswal BK. Piperlongumine, a potent anticancer phytotherapeutic: Perspectives on contemporary status and future possibilities as an anticancer agent. Pharmacol Res. 2020 Jun;156:104772.

  98. Viola-Rhenals M, Patel KR, Jaimes-Santamaria L, Wu G, Liu J, Dou QP. Recent Advances in Antabuse (Disulfiram): The Importance of its Metal-binding Ability to its Anticancer Activity. Curr Med Chem. 2018 Feb 12;25(4):506-524.
  99. 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.
  100. Wang H, Jiang H, Corbet C, de Mey S, Law K, Gevaert T, Feron O, De Ridder M. Piperlongumine increases sensitivity of colorectal cancer cells to radiation: Involvement of ROS production via dual inhibition of glutathione and thioredoxin systems. Cancer Lett. 2019 May 28;450:42-52.
  101.  Wang L, Yu Y, Zhou C, Wan R, Li Y. Anticancer effects of disulfiram: a systematic review of in vitro, animal, and human studies. Syst Rev. 2022 Jun 2;11(1):109.
  102. 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.
  103. Wang Q, Zhu T, Miao N, Qu Y, Wang Z, Chao Y, Wang J, Wu W, Xu X, Xu C, Xia L, Wang F. Disulfiram bolsters T-cell anti-tumor immunity through direct activation of LCK-mediated TCR signaling. EMBO J. 2022 Aug 16;41(16):e110636.
  104. Wang X, Guo W, Jia L, Xie L, Kiang J, Wang Y, Wang F, Lin Z, Wang E, Zhang Y, Huang P, Sun T, Zhang X, Bian Z, Tang T, Guo J, Ferrone S. Turning anecdotal irradiation-induced anti-cancer immune responses into reproducible in situ cancer vaccines via disulfiram/copper-mediated enhanced immunogenic cell death of breast cancer cells. Res Sq [Preprint]. 2023 Aug 18:rs.3.rs-3195392.
  105. Wang Y, Wang JW, Xiao X, Shan Y, Xue B, Jiang G, He Q, Chen J, Xu HG, Zhao RX, Werle KD, Cui R, Liang J, Li YL, Xu ZX. Piperlongumine induces autophagy by targeting p38 signaling. Cell Death Dis. 2013 Oct 3;4(10):e824.

  106. 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.
  107. 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.
  108. 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.
  109. Xu Y, Lu L, Luo J, Wang L, Zhang Q, Cao J, Jiao Y. Disulfiram Alone Functions as a Radiosensitizer for Pancreatic Cancer Both In Vitro and In Vivo. Front Oncol. 2021 Sep 23;11:683695.
  110. Yang Y, Xu Y, Zhao C, Zhang L, Nuerbol A, Wang L, Jiao Y. Pronounced Enhancement in Radiosensitization of Esophagus Cancer Cultivated in Docosahexaenoic Acid via the PPAR –γ Activation. Front Med (Lausanne). 2022 Jul 22;9:922228.
  111. 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.
  112. Yin Y, Sui C, Meng F, Ma P, Jiang Y. The omega-3 polyunsaturated fatty acid docosahexaenoic acid inhibits proliferation and progression of non-small cell lung cancer cells through the reactive oxygen species-mediated inactivation of the PI3K /Akt pathway. Lipids Health Dis. 2017 May 3;16(1):87.
  113. Yip NC, Fombon IS, Liu P, Brown S, Kannappan V, Armesilla AL, Xu B, Cassidy J, Darling JL, Wang W. Disulfiram modulated ROS-MAPK and NFκB pathways and targeted breast cancer cells with cancer stem cell-like properties. Br J Cancer. 2011 May 10;104(10):1564-74.
  114. Yue H, Tian Y, Zhao Z, Bo Y, Guo Y, Wang J. Comparative Study of Docosahexaenoic Acid with Different Molecular Forms for Promoting Apoptosis of the 95D Non-Small-Cell Lung Cancer Cells in a PPARγ-Dependent Manner. Mar Drugs. 2022 Sep 23;20(10):599.
  115. Zeng M, Wu B, Wei W, Jiang Z, Li P, Quan Y, Hu X. Disulfiram: A novel repurposed drug for cancer therapy. Chin Med J (Engl). 2024 Jan 26.
  116. Zha J, Chen F, Dong H, Shi P, Yao Y, Zhang Y, Li R, Wang S, Li P, Wang W, Xu B. Disulfiram targeting lymphoid malignant cell lines via ROS-JNK activation as well as Nrf2 and NF-kB pathway inhibition. J Transl Med. 2014 Jun 11;12:163.
  117. Zhang H, Chen H, Yin S, Fan L, Jin C, Zhao C, Hu H. Docosahexaenoic acid reverses PD-L1-mediated immune suppression by accelerating its ubiquitin-proteasome degradation. J Nutr Biochem. 2023 Feb;112:109186.
  118. Zhang Q, Chen W, Lv X, Weng Q, Chen M, Cui R, Liang G, Ji J. Piperlongumine, a Novel TrxR1 Inhibitor, Induces Apoptosis in Hepatocellular Carcinoma Cells by ROS-Mediated ER Stress. Front Pharmacol. 2019 Oct 14;10:1180.

  119. 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.
  120. Zhang R, Tian Z, Xu Y, Lv L. D-mannose promotes the degradation of IDH2 through upregulation of RNF185 and suppresses breast cancer. Nutr Metab (Lond). 2024 Jan 2;21(1):5.
  121. Zhang R, Yang Y, Dong W, Lin M, He J, Zhang X, Tian T, Yang Y, Chen K, Lei QY, Zhang S, Xu Y, Lv L. D-mannose facilitates immunotherapy and radiotherapy of triple-negative breast cancer via degradation of PD-L1. Proc Natl Acad Sci U S A. 2022 Feb 22;119(8):e2114851119.
  122. Zhang S, Zong Y, Chen L, Li Q, Li Z, Meng R. The immunomodulatory function and antitumor effect of disulfiram: paving the way for novel cancer therapeutics. Discov Oncol. 2023 Jun 16;14(1):103.
  123. Zhao P, Qu J, Wu A, Wang S, Tang X, Ou A, Zhang J, Xu Y, Zhao Q, Huang Y. Anti-alcoholism drug disulfiram for targeting glioma energy metabolism using BBB-penetrating delivery of fixed-dose combination. Nano Today, Volume 44, 2022.