Eight medications, six supplements, one purpose: Death of cancer cells and cancer stem cells

In pivotal research published in 2004 and again in 2023, many contemporary cancer drugs were shown to have minimal impact on enhancing the quality of life or prolonging the lives of those with advanced-stage cancer (click here and click here). A significant limitation of these drugs is their focus on a singular pathway, allowing tumor cells to mutate and develop resistance over time. Fortunately, forward-thinking scientists and physicians are pioneering new treatments. Presented below is a multifaceted and science-based protocol developed by Dr. Daniel Thomas. It combines diet and lifestyle, repurposed medications, and nutritional supplements to simultaneously target these 13 critical pathways of cancer cells and cancer stem cells:

  1. Energy metabolism: Unlike normal tissue, tumors are known for their rapid growth and cellular division. This requires substantial energy, leading to an increased demand for energy-providing nutrients such as glucose, glutamine, and fatty acids. Therefore, energy-restriction strategies that “starve” (deprive) cancer cells of the nutrients they need can make them less able to grow and divide and more vulnerable to treatment. Additionally, energy restriction can target cancer stem cells that are often resistant to conventional therapies.
  2. Oxidative stress: Cancer cells have high levels of potentially lethal oxidative stress; however, they also have robust antioxidant defenses, mainly through the upregulation of the glutathione and thioredoxin reductase systems. Disabling glutathione and thioredoxin reductase synthesis leads to oxidative stress beyond a manageable level for cancer cells. This suppresses cancer cell proliferation and promotes cell death. It also improves the immune destruction of cancer cells and sensitizes them to other therapies.
  3. Tumor hypoxia: Hypoxia, or low oxygen levels, is a common feature in tumors due to the rapid growth of cancer cells outpacing their blood supply. While this can be detrimental to normal cells, cancer cells are able to adapt and survive in hypoxic conditions. Hypoxia can even promote characteristics associated with malignancy and resistance to therapy. Therefore, targeting tumor hypoxia can potentially kill cancer cells directly or sensitize them to other treatments.
  4. Angiogenesis: This is the process of forming new blood vessels. It is crucial for tumor growth and metastasis because it supplies cancer cells with the necessary nutrients and oxygen. To restrict tumor growth, therapies targeting angiogenesis aim to further “starve” the tumor by reducing its blood supply.
  5. Chronic inflammation: This can promote the development and progression of cancer. Interleukin-6 (IL-6) is a critical player in the inflammatory process. Elevated levels of IL-6 in the tumor microenvironment can contribute to a chronic state of inflammation that supports cell proliferation, survival, and migration. Therefore, targeting chronic inflammation by reducing IL-6 can help to neutralize its tumor-promoting effects.
  6. Cell-cycle regulation: Cancer cells are characterized by uncontrolled growth and division, which is fundamentally due to aberrant cell-cycle regulation. These cells often have mutations that allow them to bypass the checkpoints that control cell division. Targeting cell-cycle regulation can potentially halt the proliferation of cancer cells.
  7. DNA repair: Cancer cells often exhibit genomic instability and have high rates of mutation that can fuel their progression. However, to survive, they rely on repair mechanisms to fix any potentially lethal (genotoxic) DNA damage. Inhibiting DNA repair in cancer cells can increase their mutation rate to lethal levels or make them more susceptible to therapies that cause DNA damage, such as radiation and certain types of chemotherapy.
  8. mTOR signaling: The mammalian target of rapamycin (mTOR) signaling pathway contributes to cancer progression by modulating essential cellular processes such as protein synthesis and cell survival. Hyperactivation of mTOR signaling is associated with various cancers, promoting tumor growth and metastasis by enhancing cell cycle progression, inhibiting apoptosis, and increasing the metabolic adaptability of cancer cells. Thus, mTOR serves as a promising therapeutic target.
  9. Inverted pH gradient: Cancer cells often show an inverted pH gradient, with a more acidic extracellular environment and more alkaline intracellular pH compared to normal cells. This inversion supports cancer cells in various ways, including promoting tumor motility, invasion, and metastasis, and contributing to treatment resistance. Targeting this aberrant pH regulation can disrupt these processes and hinder cancer’s ability to survive.
  10. Cysteine metabolism: Many cancer cells are highly dependent on the amino acid cysteine, which plays a crucial role in tumor proliferation and survival. By targeting cysteine metabolism, we may selectively compromise the viability of cancer cells while sparing normal cells that do not share cysteine dependency.
  11. Immune evasion: One of the hallmarks of cancer is its ability to evade the immune system. Cancer cells do this through a variety of mechanisms, such as downregulating proteins that make them recognizable to immune cells and creating an immunosuppressive tumor microenvironment. Therefore, targeting immune evasion is a key component of our protocol.
  12. Iron dependency: Cancer cells often exhibit heightened iron requirements to support their rapid proliferation. This increased dependency on iron makes them more susceptible to ferroptosis, a form of cell death triggered by the accumulation of excess lipid peroxides in the presence of iron. By promoting conditions conducive to ferroptosis, we can exploit this key vulnerability as a targeted approach to combat cancer.
  13. Cell-free chromatin particles: These are tiny fragments of DNA and histones without other cellular components that are released from dying cancer into the tumor microenvironment. These cell-free chromatin particles are readily internalized by surviving cancer cells and inflict DNA damage, inflammation, immune suppression, increased toxicity from chemotherapy, and ultimately, more cancer. Safely eradicating them is a crucial part of our protocol.

This protocol is innovative and comprehensive. It is potent but has a low side-effect profile. When combined with important dietary and lifestyle changes (click here), the protocol not only enhances the effectiveness of conventional cancer treatment but can also serve as a standalone treatment. The therapeutic rationale for each compound below is supported by the numerous scientific references listed at the bottom. 

Repurposed medications:

  1. 2-deoxy-D-glucose (2DG): Inhibits glucose metabolism in cancer cells and cancer stem cells, leading to a reduction in cellular energy, an increase of oxidative stress, impaired biosynthesis of macromolecules (cellular building blocks), and ultimately apoptosis (programmed cell death). Promotes immune destruction of cancer cells by blocking immunosuppressive tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). This important medication is not available in any retail or compounding pharmacy. To make it available to our patients, we have it custom-synthesized by medicinal chemists and include it in the cost of treatment.
  2. Acarbose: Synergizes with 2DG to further inhibit glucose metabolism to induce oxidative stress and apoptosis.
  3. Aspirin: Speeds the removal of dead tumor cells by increasing resolvins to enhance macrophage phagocytosis, and inhibits platelet aggregation, thereby reducing metastasis by preventing circulating tumor cells from using platelets as a protective shield to hide from circulating immune cells.
  4. Disulfiram: Inhibits numerous molecular targets related to drug resistance, DNA repair, cancer stem cells, angiogenesis, and metastasis. Reduces IL-6 and synergizes with sulfasalazine to induce cytotoxic oxidative stress in cancer cells. Promotes immune destruction of cancer cells by promoting the proliferation and infiltration of cytotoxic T-cells (CTCs) and natural killer (NK) cells, and blocking immunosuppressive tumor-associated macrophages (TAMs).
  5. Metformin: Inhibits glutamine and fatty acid metabolism, downregulates oxidative phosphorylation, and inhibits mTOR signaling. This leads to a further reduction in cellular energy, an increase in metabolic stress, and ultimately the death of cancer cells and cancer stem cells.  Promotes immune destruction of cancer cells by promoting the proliferation and infiltration of cytotoxic T-cells (CTCs) and natural killer (NK) cells, and blocking immunosuppressive tumor-associated macrophages (TAMs), tumor-associated T-regulatory cells (Tregs), and myeloid-derived suppressor cells (MDSCs). Reduces the protein stability of SLC7A11 (also known as xCT), resulting in disruption of the protective antioxidant system of cancer cells by the depletion of glutathione and thioredoxin reductase.
  6. Niclosamide: Inhibits hypoxia-inducible factor 1-alpha (HIF-1α) which stands at the crossroads of tumor cell metabolism and cancer-driving hypoxia, angiogenesis, inflammation, and immune evasion. Inhibits other tumor-promoting pathways such as Wnt, KRAS, STAT3, Notch, mTOR, E2F, and Myc. Activates tumor-suppressor signaling such as p53, PP2A, and AMPK; blocks DNA repair; reduces IL-6; and induces intracellular acidification. Promotes immune destruction of cancer cells by blocking immunosuppressive myeloid-derived suppressor cells (MDSCs). Induces nutrient starvation in cancer cells by blocking macropinocytosis, a process by which cancer cells engulf large volumes of extracellular fluid, aiding in nutrient uptake to support their rapid growth and survival. Niclosamide is no longer available from US pharmacies. To make it available to our patients, we obtain it from reputable overseas pharmacies and include it in the cost of treatment.
  7. Sulfasalazine: Induces cysteine starvation in cancer cells by blocking the cystine transporter SLC7A11, resulting in disruption of the protective antioxidant system of cancer cells by the depletion of glutathione and thioredoxin reductase. Together with disulfiram, this induces irreversible toxicity caused by the accumulation of reactive oxygen species (ROS) and irreparable DNA damage, with subsequent cellular demise via apoptosis and ferroptosis (iron-mediated cell death). Furthermore, sulfasalazine promotes ferroptosis, reduces IL-6, and promotes the immune destruction of cancer cells by inhibiting immunosuppressive regulatory T-cells (Tregs).
  8. Syrosingopine: Disables glutathione synthesis and promotes intracellular acidification and extracellular alkalinization of cancer cells by inhibiting monocarboxylate transporters 1 and 4 (MCT1 and MCT4). In combination with metformin, syrosingopine facilitates apoptosis by inhibiting essential NAD+ production pathways. These pathways allow cancer cells to convert dietary nutrients such as glucose, glutamine, and fatty acids, and high-energy fuels like lactate, pyruvate, and ketone bodies sourced from neighboring cancer-associated fibroblasts (CAFs) into much-needed energy in the form of ATP (adenosine triphosphate). This important medication is not available in any retail or compounding pharmacy. To make it available to our patients, we have it custom-synthesized by medicinal chemists and include it in the cost of treatment.

Nutritional supplements:

  1. Apigenin: Promotes ferroptosis and inhibits angiogenesis, cell proliferation, invasion, and metastasis. Blocks immunosuppressive tumor-associated macrophages (TAMs) and tumor-associated T-regulatory cells (Tregs). When combined with metformin, promotes reactive oxygen species (ROS)-induced apoptosis of cancer cells.
  2. Copper gluconate (microdosed): Together with resveratrol (microdosed), eradicates cell-free chromatin particles.
  3. Ozonated oil: Inhibits angiogenesis and metastasis by increasing oxygen availability in the tumor microenvironment, decreases inflammation, activates apoptosis of cancer cells and cancer stem cells by depleting their antioxidants, promotes immune destruction of cancer cells by inhibiting tumor-associated macrophages (TAMs), and improves quality of life of those receiving chemotherapy or radiation therapy.
  4. Piperlongumine: Elevates oxidative stress of cancer cells and cancer stem cells by dually inhibiting glutathione and thioredoxin reductase synthesis; hampers critical cancer cell pathways by inhibiting NF-κB and STAT3 activation; disrupts cancer cell growth by inducing cell cycle arrest, promoting apoptosis and ferroptosis, and curbing angiogenesis; diminishes the metastatic potential of cancer cells; enhances the efficacy of conventional chemotherapy by increasing cancer cell sensitivity and inhibiting drug efflux pumps; decreases inflammation; directly binds to cysteine residues; induces autophagy; inhibits mTOR signaling; inhibits IL-6; and downregulates anti-apoptotic proteins. 
  5. Resveratrol (microdosed): Together with copper gluconate (microdosed), eradicates cell-free chromatin particles.
  6. Tributyrin: Suppresses cancer cell proliferation and induces apoptosis by upregulating and sequestering the p53 signaling protein in the nucleus.

The importance of eradicating cancer stem cells:

Healthy stem cells reside in the human body, and they play a crucial role in repairing damaged tissues. However, tumors also contain stem cells that contribute to the regeneration of cancerous tissue. In 1997, researchers first identified cancer stem cells, and since then, numerous studies have been conducted to better understand their biological properties.

The propor­tion of cancer stem cells in tumors is extremely low. They account for only 0.01-2% of the total tumor mass, but they are believed to constitute the origin of most tumors. Unlike the bulk tumor cells, cancer stem cells are not detectable using standard tumor biomarkers or imaging techniques. For anti-cancer therapy to be successful, both the bulk tumor cells and the cancer stem cells must be eliminated since cancer stem cells have the ability to regrow tumors repeatedly.

Cancer stem cells drive treatment failure, immune evasion, cancer progression, metastasis, and disease recurrence. Furthermore, they may be the root cause of the initial tumor formation. Although conventional cancer therapies can effectively target the bulk tumor cells, they are largely ineffective against cancer stem cells due to their highly resistant nature. In some cases, conventional treatments may even promote the proliferation of cancer stem cells. Cancer stem cells are a major obstacle to successful cancer treatment.

Cancer stem cells can migrate and settle in remote parts of the body, where they can lie dormant for extended periods, waiting for the right conditions to form new tumors. Though conventional treatments can shrink tumors, they cannot stop cancer relapse. In fact, even after all observable signs of cancer are gone, the existence of residual cancer stem cells increases the risk of eventually developing new and possibly more aggressive tumors.

It is not enough to solely eliminate the bulk tumor cells. To achieve long-term survival, cancer stem cells must also be eliminated. Doing so could potentially eliminate the entire cancer cell population, thereby stopping cancer metastasis and recurrence, which are the leading causes of cancer-related deaths. Components of the protocol that target cancer stem cells are 2DG, disulfiram, metformin, niclosamide, ozonated oil, piperlongumine, and sulfasalazine.

Targeting interleukin-6:

Interleukin-6 (IL-6) is a multifunctional cytokine that plays critical roles in various physiological and pathological processes. In the context of cancer, the microenvironmental IL-6 has been shown to contribute to several aspects of tumorigenesis, progression, and therapy resistance. Here’s a breakdown of the role that microenvironmental IL-6 plays in cancer:

  1. Tumor growth and proliferation: IL-6 can act in an autocrine manner on cancer cells and promote their growth. For instance, in multiple myeloma, malignant plasma cells produce and respond to IL-6, which promotes their survival.
  2. Inflammation: Chronic inflammation is recognized as a hallmark of cancer, and IL-6 is a key player in the inflammatory response. Elevated IL-6 levels in the tumor microenvironment can contribute to a chronic state of inflammation, which can drive the mutation rate in cells and promote tumorigenesis.
  3. Angiogenesis: IL-6 has been implicated in promoting angiogenesis, the formation of new blood vessels. Tumors need an extensive blood supply to grow, and by stimulating angiogenesis, IL-6 can facilitate tumor growth and progression.
  4. Invasion and metastasis: IL-6 can promote epithelial-to-mesenchymal transition (EMT), a process by which epithelial cells acquire a mesenchymal phenotype. This transition increases the invasiveness of cancer cells and their potential to metastasize to distant sites.
  5. Immune evasion: IL-6 can contribute to the suppression of the anti-tumor immune response. It can influence the polarization of tumor-associated macrophages (TAMs) towards an M2 (anti-inflammatory) phenotype, which tends to promote tumor progression and suppress T-cell responses. Moreover, IL-6 can further suppress the anti-tumor immune response by leading to the expansion of myeloid-derived suppressor cells (MDSCs).
  6. Treatment resistance: Elevated levels of IL-6 have been associated with resistance to various treatments, including chemotherapy and targeted therapies. For instance, in the context of breast cancer, IL-6 signaling has been shown to confer resistance to anti-HER2 therapies.

Because of the pivotal role that IL-6 plays in cancer biology, targeting the IL-6 signaling pathway is crucial. Components of the protocol that reduce IL-6 levels are disulfiram, niclosamide, piperlongumine, and sulfasalazine.

The immunology of cancer:

The immune system is intricately designed to identify and eliminate abnormal cells, including those that are cancerous. However, this natural defense mechanism can be subverted by the tumor microenvironment, which recruits and reprograms specific immune cells to create an immunosuppressive setting. Among these are tumor-associated macrophages (TAMs), T-regulatory cells (Tregs), and myeloid-derived suppressor cells (MDSCs), each of which contributes to cancer progression in unique but overlapping ways.

TAMs are often driven to differentiate from an M1 macrophage into an M2 macrophage that actively promotes immunosuppression. They accomplish this by secreting cytokines like IL-10 and TGF-β, which inhibit effector T-cell function. Additionally, TAMs are known to suppress dendritic cell maturation, making these antigen-presenting cells less effective in activating T-cells. TAMs also participate in recruiting and stimulating Tregs, which in turn amplifies the immunosuppressive signals in the tumor microenvironment. Beyond immunosuppression, TAMs play a role in angiogenesis by releasing factors such as vascular endothelial growth factor (VEGF) and facilitating tumor invasion and metastasis through the production of matrix metalloproteinases (MMPs). The components of the protocol that target TAMs are 2DG, apigenin, disulfiram, metformin, and ozonated oil.

Tregs further complement this immunosuppressive landscape. They are proficient in inhibiting effector T-cell activation by releasing their own set of immunosuppressive cytokines, such as IL-10 and TGF-β. Tregs can also exert their suppressive effects through direct cell-to-cell interactions with other immune entities like CD8+ T cells and dendritic cells. Additionally, Tregs engage in metabolic competition by consuming IL-2, a growth factor that is essential for the functioning of effector T cells. Some evidence even suggests a role for Tregs in supporting the immunosuppressive functions of TAMs. The components of the protocol that target Tregs are apigenin, metformin, and sulfasalazine.

MDSCs are another major player in this scenario. They have broad immunosuppressive capabilities, often producing high levels of arginase and reactive oxygen species (ROS) that impair T-cell receptor signaling and thus inhibit T-cell function. MDSCs also secrete cytokines such as IL-10 and TGF-β; and can induce the formation and activation of Tregs, contributing to a multi-layered suppression of anti-tumor immunity. Moreover, MDSCs are known to inhibit the activities of natural killer (NK) cells, adding another layer of complexity to the immunosuppressive environment. The components of the protocol that target MDSCs are 2DG, metformin, and niclosamide.

Iron dependency in cancer and the therapeutic potential of ferroptosis:

Cancer cells often exhibit profound iron uptake and retention due to their heightened metabolic demands for proliferation. This iron dependency accentuates the production of reactive oxygen species and lipid peroxides, especially in the presence of excess iron. As a result, these cells become more vulnerable to ferroptosis, a form of cell death driven by iron-dependent lipid peroxidation. By exploiting this increased susceptibility, therapies that induce or amplify ferroptosis can selectively target and eliminate iron-addicted cancer cells. Here’s why targeting ferroptosis is considered crucial in cancer treatment:

  1. Treatment-resistant tumors: Some tumors are resistant to conventional therapies that aim to induce apoptosis. Ferroptosis offers an alternative mechanism to induce cell death in these resistant cancer cells.
  2. Cancer specificity: Normal cells have robust antioxidant defenses to prevent lipid peroxidation, making them relatively resistant to ferroptosis. In contrast, certain cancer cells, due to their metabolic profiles, are more susceptible to ferroptotic cell death. Therefore, targeting ferroptosis can be more selective for cancer cells.
  3. Tumor microenvironment: The tumor microenvironment, especially in solid tumors, is often deprived of nutrients and oxygen, making it susceptible to oxidative stress. By targeting ferroptosis, one can exploit this vulnerability of the tumor microenvironment.
  4. Synergistic effects: Combining ferroptosis-inducing agents with other treatments, such as chemotherapy, radiation, or immunotherapy, might result in synergistic anti-tumor effects. This can potentially lower the doses of conventional therapies, reducing side effects.
  5. Inhibition of cancer stem cells: Preliminary studies have suggested that inducing ferroptosis can target cancer stem cells, which are believed to be the root cause of tumor initiation, metastasis, and therapeutic resistance.
  6. Targeting specific mutations: Some tumors with specific genetic mutations (e.g., tumors with mutations in p53) have shown increased sensitivity to ferroptosis. Targeting ferroptosis can be a strategy to exploit these genetic vulnerabilities.
  7. Metastasis prevention: There’s evidence to suggest that inducing ferroptosis can prevent cancer metastasis, which is a significant cause of cancer-related deaths.
  8. Overcoming drug resistance: Inducing ferroptosis can potentially overcome resistance to targeted therapies. For instance, certain tumors that develop resistance to EGFR inhibitors may still be susceptible to ferroptosis inducers.

The components of the protocol that promote ferroptosis are apigenin, piperlongumine, and sulfasalazine.

Targeting cell-free chromatin particles: A new frontier in cancer therapy:

In a tumor, even without treatment, a substantial number of cancer cells spontaneously die each day. This is due to the high rate of cell proliferation and the unstable nature of cancer cells, which often have defects in their DNA repair mechanisms and can be more prone to apoptosis (programmed cell death) or necrosis (cell death due to injury or disease). Scientists discovered that when cancer cells die, either spontaneously or from treatment such as chemotherapy, cell-free chromatin particles are released into the tumor microenvironment where they are readily internalized by surviving cancer cells and inflict DNA damage, inflammation, immune suppression, increased toxicity from chemotherapy, and ultimately, more cancer.

Our DNA is stored on our chromosomes as densely packed units called chromatin which contains DNA and proteins called histones. This configuration prevents the long strands of DNA from becoming tangled and plays an important role in stabilizing the DNA during cell division, preventing DNA damage, and maintaining normal gene expression. Cell-free chromatin particles are composed of tiny fragments of DNA and histones without other cellular components. These particles are released from cancer cells undergoing apoptosis—the mechanism a cell undertakes for self-destruction, activated when the cell senses significant self-damage.

Scientists also found that cell-free chromatin particles are released into the bloodstream from the billions of normal cells that die each day as we age. These particles readily enter healthy cells and integrate into their DNA. This leads to DNA damage, mitochondrial dysfunction, activation of apoptotic and inflammatory pathways, and impairment of mitochondrial function. This process triggers a vicious cycle and ongoing chain reaction that perpetuates the aging process and increases the risk of cancer.

Some scientists believe that the lifelong assault on healthy cells by internalized cell-free chromatin particles may not only be the chief underlying cause of the aging process and cancer, but also may act as a global instigator of other age-associated disorders, including cardiovascular disease, diabetes, sepsis, and Alzheimer’s disease.

Fortunately, scientists found a way to safely neutralize cell-free chromatin particles by using minute and specific oral doses of resveratrol and copper to generate oxygen radicals in the stomach. These radicals are readily absorbed into the circulation and permeate the extracellular spaces of the tumor microenvironment and deactivate cell-free chromatin particles. In an animal model, this was found to downregulate several biological hallmarks of aging and neurodegeneration. In a human model using patients with advanced oral cancer, it was found that deactivating cell-free chromatin particles downregulated the hallmarks of cancer, as well as five immune checkpoint (suppressor) proteins. 

Pill burden:

Taking too many pills can compromise quality of life, incur extra expenses, and potentially harm kidney, liver, or heart function. With this in mind, we begin our approach with these six carefully selected compounds, which are designed to comprehensively target the twelve key pathways essential for both cancer cells and cancer stem cells, as outlined above:

  1. Aspirin
  2. Copper gluconate (microdosed)
  3. Metformin
  4. Ozonated oil
  5. Piperlongumine
  6. Resveratrol (microdosed)
  7. Syrosingopine

Treatment cost:

The average oncologist manages between 250 and 500 patients. Dr. Thomas limits his caseload to fewer than 50. This allows him to provide highly attentive care and to dedicate time to vital research that directly benefits his patients. The treatment protocol necessitates close medical supervision, management, and ongoing support by Dr. Thomas. After an initial in-office physical examination, treatment begins. Follow-up visits are conducted via telemedicine and yearly physical exams are performed. While under his care, patients have unlimited email access to Dr. Thomas.

The cost for our services is a flat rate of $2850 per month. This is less expensive than alternative cancer treatment in Mexico or Europe. Clinics there typically charge $7,000 to $20,000 per week (click here and scroll down to FAQs and click “What are the costs of stage 4 cancer treatments at a clinic?”). Despite their high costs, many of these clinics lack a more comprehensive understanding of tumor biology, which could pose a threat if they fail to target key vulnerabilities of cancer.

Our monthly fee includes the 2DG, niclosamide, and syrosingopine. Other costs are for the medications available at the local pharmacy (acarbose, aspirin, disulfiram, metformin, sulfasalazine); and supplements available online (apigenin, copper gluconate, ozonated oil, piperlongumine, resveratol, tributyrin). These total between $300 and $400 per month. Treatment is maintained until remission or disease stabilization is achieved. This usually takes 6-18 months. Afterward, we employ a simpler and less expensive protocol. For more information, click here.

Scientific references:

Aghaee F, Pirayesh Islamian J, Baradaran B. Enhanced radiosensitivity and chemosensitivity of breast cancer cells by 2-deoxy-d-glucose in combination therapy. J Breast Cancer. 2012 Jun;15(2):141-7.

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.

Amin S, Lux A, O’Callaghan F. The journey of metformin from glycaemic control to mTOR inhibition and the suppression of tumour growth. Br J Clin Pharmacol. 2019 Jan;85(1):37-46.

Ashrafizadeh M, Bakhoda MR, Bahmanpour Z, Ilkhani K, Zarrabi A, Makvandi P, Khan H, Mazaheri S, Darvish M, Mirzaei H. Apigenin as Tumor Suppressor in Cancers: Biotherapeutic Activity, Nanodelivery, and Mechanisms With Emphasis on Pancreatic Cancer. Front Chem. 2020 Oct 15;8:829.

Bagherpoor AJ, Shameem M, Luo X, Seelig D, Kassie F. Inhibition of lung adenocarcinoma by combinations of sulfasalazine (SAS) and disulfiram-copper (DSF-Cu) in cell line models and mice. Carcinogenesis. 2023 Jun 24;44(4):291-303.

Benhar M, Shytaj IL, Stamler JS, Savarino A. Dual targeting of the thioredoxin and glutathione systems in cancer and HIV. J Clin Invest. 2016 May 2;126(5):1630-9.

Benjamin D, Robay D, Hindupur SK, Pohlmann J, Colombi M, El-Shemerly MY, Maira SM, Moroni C, Lane HA, Hall MN. Dual Inhibition of the Lactate Transporters MCT1 and MCT4 Is Synthetic Lethal with Metformin due to NAD+ Depletion in Cancer Cells. Cell Rep. 2018 Dec 11;25(11):3047-3058.e4.

Bent EH, Millán-Barea LR, Zhuang I, Goulet DR, Fröse J, Hemann MT. Microenvironmental IL-6 inhibits anti-cancer immune responses generated by cytotoxic chemotherapy. Nat Commun. 2021 Oct 28;12(1):6218.

Bhullar D, Commisso C. Stopping the fat: Repurposing an antidepressant for cancer treatment. J Exp Med. 2023 Mar 6;220(3):e20222097.

Bonifácio VDB, Pereira SA, Serpa J, Vicente JB. Cysteine metabolic circuitries: druggable targets in cancer. Br J Cancer. 2021 Mar;124(5):862-879.

Buyse C, Joudiou N, Warscotte A, Richiardone E, Mignion L, Corbet C, Gallez B. Evaluation of Syrosingopine, an MCT Inhibitor, as Potential Modulator of Tumor Metabolism and Extracellular Acidification. Metabolites. 2022 Jun 17;12(6):557.

Calderon JJ, Prieto K, Lasso P, Fiorentino S, Barreto A. Modulation of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment by Natural Products. Arch Immunol Ther Exp (Warsz). 2023 Jul 6;71(1):17.

Chen SY, Huang HY, Lin HP, Fang CY. Piperlongumine induces autophagy in biliary cancer cells via reactive oxygen species-activated Erk signaling pathway. Int J Mol Med. 2019 Nov;44(5):1687-1696.

Cheong JH, Park ES, Liang J, Dennison JB, Tsavachidou D, Nguyen-Charles C, Wa Cheng K, Hall H, Zhang D, Lu Y, Ravoori M, Kundra V, Ajani J, Lee JS, Ki Hong W, Mills GB. Dual inhibition of tumor energy pathway by 2-deoxyglucose and metformin is effective against a broad spectrum of preclinical cancer models. Mol Cancer Ther. 2011 Dec;10(12):2350-62.

Chiang CF, Chao TT, Su YF, Hsu CC, Chien CY, Chiu KC, Shiah SG, Lee CH, Liu SY, Shieh YS. Metformin-treated cancer cells modulate macrophage polarization through AMPK-NF-κB signaling. Oncotarget. 2017 Mar 28;8(13):20706-20718.

Chonov DC, Ignatova MMK, Ananiev JR, Gulubova MV. IL-6 Activities in the Tumour Microenvironment. Part 1. Open Access Maced J Med Sci. 2019 Jul 20;7(14):2391-2398.

Ciavardelli D, Rossi C, Barcaroli D, Volpe S, Consalvo A, Zucchelli M, De Cola A, Scavo E, Carollo R, D’Agostino D, Forlì F, D’Aguanno S, Todaro M, Stassi G, Di Ilio C, De Laurenzi V, Urbani A. Breast cancer stem cells rely on fermentative glycolysis and are sensitive to 2-deoxyglucose treatment. Cell Death Dis. 2014 Jul 17;5(7):e1336.

Clavo B, Cánovas-Molina A, Ramallo-Fariña Y, Federico M, Rodríguez-Abreu D, Galván S, Ribeiro I, Marques da Silva SC, Navarro M, González-Beltrán D, Díaz-Garrido JA, Cazorla-Rivero S, Rodríguez-Esparragón F, Serrano-Aguilar P. Effects of Ozone Treatment on Health-Related Quality of Life and Toxicity Induced by Radiotherapy and Chemotherapy in Symptomatic Cancer Survivors. Int J Environ Res Public Health. 2023 Jan 13;20(2):1479.

Combs JA, DeNicola GM. The Non-Essential Amino Acid Cysteine Becomes Essential for Tumor Proliferation and Survival. Cancers (Basel). 2019 May 16;11(5):678.

Cosialls E, El Hage R, Dos Santos L, Gong C, Mehrpour M, Hamaï A. Ferroptosis: Cancer Stem Cells Rely on Iron until “to Die for” It. Cells. 2021 Nov 2;10(11):2981.

Doherty JR, Yang C, Scott KE, Cameron MD, Fallahi M, Li W, Hall MA, Amelio AL, Mishra JK, Li F, Tortosa M, Genau HM, Rounbehler RJ, Lu Y, Dang CV, Kumar KG, Butler AA, Bannister TD, Hooper AT, Unsal-Kacmaz K, Roush WR, Cleveland JL. Blocking lactate export by inhibiting the Myc target MCT1 Disables glycolysis and glutathione synthesis. Cancer Res. 2014 Feb 1;74(3):908-20.

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.

Finisguerra V, Dvorakova T, Formenti M, Van Meerbeeck P, Mignion L, Gallez B, Van den Eynde BJ. Metformin improves cancer immunotherapy by directly rescuing tumor-infiltrating CD8 T lymphocytes from hypoxia-induced immunosuppression. J Immunother Cancer. 2023 May;11(5):e005719.

Fong D, Christensen CT, Chan MM. Targeting Cancer Stem Cells with Repurposed Drugs to Improve Current Therapies. Recent Pat Anticancer Drug Discov. 2021;16(2):136-160.

Gao C, Fang L, Zhang H, Zhang WS, Li XO, Du SY. Metformin Induces Autophagy via the AMPK-mTOR Signaling Pathway in Human Hepatocellular Carcinoma Cells. Cancer Manag Res. 2020 Jul 14;12:5803-5811.

Gilligan MM, Gartung A, Sulciner ML, Norris PC, Sukhatme VP, Bielenberg DR, Huang S, Kieran MW, Serhan CN, Panigrahy D. Aspirin-triggered proresolving mediators stimulate resolution in cancer. Proc Natl Acad Sci U S A. 2019 Mar 26;116(13):6292-6297.

Gu Y, Niu X, Yin L, Wang Y, Yang Y, Yang X, Zhang Q, Ji H. Enhancing Fatty Acid Catabolism of Macrophages Within Aberrant Breast Cancer Tumor Microenvironment Can Re-establish Antitumor Function. Front Cell Dev Biol. 2021 Apr 15;9:665869.

Guo W, Chen S, Li C, Xu J, Wang L. Application of Disulfiram and its Metabolites in Treatment of Inflammatory Disorders. Front Pharmacol. 2022 Feb 2;12:795078.

Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K. Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res. 2009 Oct 1;69(19):7507-11.

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.

Jeong CH, Ryu H, Kim DH, Cheng WN, Yoon JE, Kang S, Han SG. Piperlongumine Induces Cell Cycle Arrest via Reactive Oxygen Species Accumulation and IKKβ Suppression in Human Breast Cancer Cells. Antioxidants (Basel). 2019 Nov 14;8(11):553.

Jeong YK, Kim MS, Lee JY, Kim EH, Ha H. Metformin Radiosensitizes p53-Deficient Colorectal Cancer Cells through Induction of G2/M Arrest and Inhibition of DNA Repair Proteins. PLoS One. 2015 Nov 23;10(11):e0143596.

Jian SL, Chen WW, Su YC, Su YW, Chuang TH, Hsu SC, Huang LR. Glycolysis regulates the expansion of myeloid-derived suppressor cells in tumor-bearing hosts through prevention of ROS-mediated apoptosis. Cell Death Dis. 2017 May 11;8(5):e2779.

Jiang H, Li AM, Ye J. The magic bullet: Niclosamide. Front Oncol. 2022 Nov 21;12:1004978.

Kang J, Lee D, Lee KJ, Yoon JE, Kwon JH, Seo Y, Kim J, Chang SY, Park J, Kang EA, Park SJ, Park JJ, Cheon JH, Kim TI. Tumor-Suppressive Effect of Metformin via the Regulation of M2 Macrophages and Myeloid-Derived Suppressor Cells in the Tumor Microenvironment of Colorectal Cancer. Cancers (Basel). 2022 Jun 10;14(12):2881.

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.

Kerkhove L, Geirnaert F, Rifi AL, Law KL, Gutiérrez A, Oudaert I, Corbet C, Gevaert T, Dufait I, De Ridder M. Repurposing Sulfasalazine as a Radiosensitizer in Hypoxic Human Colorectal Cancer. Cancers (Basel). 2023 Apr 18;15(8):2363.

Kim MY, Jung AR, Shin D, Kwon H, Cho HJ, Ha US, Hong SH, Lee JY, Kim SW, Park YH. Niclosamide exerts anticancer effects through inhibition of the FOXM1-mediated DNA damage response in prostate cancer. Am J Cancer Res. 2021 Jun 15;11(6):2944-2959.

Kirolikar S, Prasannan P, Raghuram GV, Pancholi N, Saha T, Tidke P, Chaudhari P, Shaikh A, Rane B, Pandey R, Wani H, Khare NK, Siddiqui S, D’souza J, Prasad R, Shinde S, Parab S, Nair NK, Pal K, Mittra I. Prevention of radiation-induced bystander effects by agents that inactivate cell-free chromatin released from irradiated dying cells. Cell Death Dis. 2018 Nov 15;9(12):1142.

Koltai, Tomas. (2017). Finding a Role for 2-Deoxyglucose in Cancer Treatment.

Koppula P, Zhuang L, Gan B. Cystine transporter SLC7A11/xCT in cancer: ferroptosis, nutrient dependency, and cancer therapy. Protein Cell. 2021 Aug;12(8):599-620.

Krakauer T. Sulfasalazine attenuates staphylococcal enterotoxin B-induced immune responses. Toxins (Basel). 2015 Feb 13;7(2):553-9.

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.

Lei Y, Yi Y, Liu Y, et al. Metformin targets multiple signaling pathways in cancer. Chin J Cancer. 2017;36(1):17.

Leung WH, Shih JW, Chen JS, Mokgautsi N, Wei PL, Huang YJ. Preclinical Identification of Sulfasalazine’s Therapeutic Potential for Suppressing Colorectal Cancer Stemness and Metastasis through Targeting KRAS/MMP7/CD44 Signaling. Biomedicines. 2022 Feb 4;10(2):377.

Liang L, Huang M, Xiao Y, Zen S, Lao M, Zou Y, Shi M, Yang X, Xu H. Inhibitory effects of niclosamide on inflammation and migration of fibroblast-like synoviocytes from patients with rheumatoid arthritis. Inflamm Res. 2015 Apr;64(3-4):225-33.

Liang Y, Zhong Q, Ma R, Ni Z, Thakur K, Zhang J, Wei Z. Apigenin, a natural flavonoid, promotes autophagy and ferroptosis in human endometrial carcinoma Ishikawa cells in vitro and in vivo. Food Science and Human Wellness, Vol. 12, No. 6, 2023, pp. 2242-2251.

Liotti F, Marotta M, Melillo RM, Prevete N. The Impact of Resolution of Inflammation on Tumor Microenvironment: Exploring New Ways to Control Cancer Progression. Cancers (Basel). 2022 Jul 8;14(14):3333

Liu F, Zhou Q, Jiang HF, Zhang TT, Miao C, Xu XH, Wu JX, Yin SL, Xu SJ, Peng JY, Gao PP, Cao X, Pan F, He X, Chen XQ. 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. 2023 May 10;42(1):118..

Liu S, Liu X, Zhang C, Shan W, Qiu X. T-Cell Exhaustion Status Under High and Low Levels of Hypoxia-Inducible Factor 1α Expression in Glioma. Front Pharmacol. 2021 Jul 9;12:711772.

Liu Y, White KA, Barber DL. Intracellular pH Regulates Cancer and Stem Cell Behaviors: A Protein Dynamics Perspective. Front Oncol. 2020 Aug 26;10:1401.

Luongo M, Brigida AL, Mascolo L, Gaudino G. Possible Therapeutic Effects of Ozone Mixture on Hypoxia in Tumor Development. Anticancer Res. 2017 Feb;37(2):425-435.

Makhov P, Golovine K, Teper E, Kutikov A, Mehrazin R, Corcoran A, Tulin A, Uzzo RG, Kolenko VM. Piperlongumine promotes autophagy via inhibition of Akt/mTOR signalling and mediates cancer cell death. Br J Cancer. 2014 Feb 18;110(4):899-907.

Man CH, Mercier FE, Liu N, Dong W, Stephanopoulos G, Jiang L, Jung Y, Lin CP, Leung AYH, Scadden DT. Proton export alkalinizes intracellular pH and reprograms carbon metabolism to drive normal and malignant cell growth. Blood. 2022 Jan 27;139(4):502-522.

Mittra I, Pal K, Pancholi N, Shaikh A, Rane B, Tidke P, Kirolikar S, Khare NK, Agrawal K, Nagare H, Nair NK. Prevention of chemotherapy toxicity by agents that neutralize or degrade cell-free chromatin. Ann Oncol. 2017 Sep 1;28(9):2119-2127.

 

Mittra I, Samant U, Sharma S, Raghuram GV, Saha T, et al. Cell-free chromatin from dying cancer cells integrate into genomes of bystander healthy cells to induce DNA damage and inflammation. Cell Death Disc. 3: 17015.

Mostafavi S, Zalpoor H, Hassan ZM. The promising therapeutic effects of metformin on metabolic reprogramming of cancer-associated fibroblasts in solid tumors. Cell Mol Biol Lett. 2022 Jul 22;27(1):58.

Nagane M, Kanai E, Shibata Y, Shimizu T, Yoshioka C, Maruo T, Yamashita T. Sulfasalazine, an inhibitor of the cystine-glutamate antiporter, reduces DNA damage repair and enhances radiosensitivity in murine B16F10 melanoma. PLoS One. 2018 Apr 12;13(4):e0195151.

Narang VS, Pauletti GM, Gout PW, Buckley DJ, Buckley AR. Sulfasalazine-induced reduction of glutathione levels in breast cancer cells: enhancement of growth-inhibitory activity of Doxorubicin. Chemotherapy. 2007;53(3):210-7.

Obaid QA, Khudair KK, Al-Shammari AM. Glucose deprivation using 2-deoxyglucose and acarbose induce metabolic oxidative stress and apoptosis in female mice bearing breast cancer. Biochimie. 2022 Apr;195:59-66.

Omar HA, Berman-Booty L, Kulp SK, Chen CS. Energy restriction as an antitumor target. Future Oncol. 2010 Nov;6(11):1675-9.

O’Neill S, Porter RK, McNamee N, Martinez VG, O’Driscoll L. 2-Deoxy-D-Glucose inhibits aggressive triple-negative breast cancer cells by targeting glycolysis and the cancer stem cell phenotype. Sci Rep. 2019 Mar 7;9(1):3788.

Ortega JF, de Conti A, Tryndyak V, Furtado KS, Heidor R, Horst MA, Fernandes LH, Tavares PE, Pogribna M, Shpyleva S, Beland FA, Pogribny IP, Moreno FS. Suppressing activity of tributyrin on hepatocarcinogenesis is associated with inhibiting the p53-CRM1 interaction and changing the cellular compartmentalization of p53 protein.

Pan JX, Ding K, Wang CY. Niclosamide, an old antihelminthic agent, demonstrates antitumor activity by blocking multiple signaling pathways of cancer stem cells. Chin J Cancer. 2012 Apr;31(4):178-84.

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.

Pecqueur C, Oliver L, Oizel K, Lalier L, Vallette FM. Targeting metabolism to induce cell death in cancer cells and cancer stem cells. Int J Cell Biol. 2013;2013:805975.

Pilankar A, Singhavi H, Raghuram GV, Siddiqui S, Khare NK, Jadhav V, Tandel H, Pal K, Bhattacharjee A, Chaturvedi P, Mittra I. A pro-oxidant combination of resveratrol and copper down-regulates hallmarks of cancer and immune checkpoints in patients with advanced oral cancer: Results of an exploratory study (RESCU 004). Front Oncol. 2022 Sep 16;12:1000957.

 

Raghuram GV, Chaudhary S, Johari S, Mittra I. Illegitimate and Repeated Genomic Integration of Cell-Free Chromatin in the Aetiology of Somatic Mosaicism, Ageing, Chronic Diseases and Cancer. Genes (Basel). 2019 May 28;10(6):407.

 

Raghuram GV, Tripathy B, Avadhani BK, Shabrish S, Khare NK, Lopes R, Pal K, Mittra I. Cell-free chromatin particles released from dying cells inflict mitochondrial damage and ROS production in living cells. bioRxiv 2021.12.30.47452.

Ralph SJ, Pritchard R, Rodríguez-Enríquez S, Moreno-Sánchez R, Ralph RK. Hitting the Bull’s-Eye in Metastatic Cancers-NSAIDs Elevate ROS in Mitochondria, Inducing Malignant Cell Death. Pharmaceuticals (Basel). 2015 Feb 13;8(1):62-106.

Repas J, Zupin M, Vodlan M, Veranič P, Gole B, Potočnik U, Pavlin M. Dual Effect of Combined Metformin and 2-Deoxy-D-Glucose Treatment on Mitochondrial Biogenesis and PD-L1 Expression in Triple-Negative Breast Cancer Cells. Cancers (Basel). 2022 Mar 5;14(5):1343.

Saladini S, Aventaggiato M, Barreca F, Morgante E, Sansone L, Russo MA, Tafani M. Metformin Impairs Glutamine Metabolism and Autophagy in Tumour Cells. Cells. 2019 Jan 14;8(1):49.

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.

Shabrish S, Pal K, Khare N,  Satsangi D, Pilankar A,  Jadhav V, Shinde S, Raphael N,  Sriram G, Relestina Lopes, Raghuram G,  Tandel H,  Mittra I. Cell-free chromatin particles activate immune checkpoints in human T cells: Implications for cancer therapy. bioRxiv 2023.06.09.544311.

Shafaee A, Pirayesh Islamian J, Zarei D, Mohammadi M, Nejati-Koshki K, Farajollahi A, Aghamiri SMR, Rahmati Yamchi M, Baradaran B, Asghari Jafarabadi M. Induction of Apoptosis by a Combination of 2-Deoxyglucose and Metformin in Esophageal Squamous Cell Carcinoma by Targeting Cancer Cell Metabolism. Iran J Med Sci. 2019 Mar;44(2):99-107.

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.

Shutt DC, O’Dorisio MS, Aykin-Burns N, Spitz DR. 2-deoxy-D-glucose induces oxidative stress and cell killing in human neuroblastoma cells. Cancer Biol Ther. 2010 Jun 1;9(11):853-61.

Singh R, Gupta V, Kumar A, Singh K. 2-Deoxy-D-Glucose: A Novel Pharmacological Agent for Killing Hypoxic Tumor Cells, Oxygen Dependence-Lowering in Covid-19, and Other Pharmacological Activities. Adv Pharmacol Pharm Sci. 2023 Mar 2;2023:9993386.

Smith TA, Phyu SM. Metformin Decouples Phospholipid Metabolism in Breast Cancer Cells. PLoS One. 2016 Mar 9;11(3):e0151179.

Talabnin C, Talabnin K, Wongkham S. Enhancement of piperlongumine chemosensitivity by silencing heme oxygenase-1 expression in cholangiocarcinoma cell lines. Oncol Lett. 2020 Sep;20(3):2483-2492.

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.

Tao DL, Tassi Yunga S, Williams CD, McCarty OJT. Aspirin and antiplatelet treatments in cancer. Blood. 2021 Jun 10;137(23):3201-3211.

Terashima Y, Toda E, Itakura M, Otsuji M, Yoshinaga S, Okumura K, Shand FHW, Komohara Y, Takeda M, Kokubo K, Chen MC, Yokoi S, Rokutan H, Kofuku Y, Ohnishi K, Ohira M, Iizasa T, Nakano H, Okabe T, Kojima H, Shimizu A, Kanegasaki S, Zhang MR, Shimada I, Nagase H, Terasawa H, Matsushima K. Targeting FROUNT with disulfiram suppresses macrophage accumulation and its tumor-promoting properties. Nat Commun. 2020 Jan 30;11(1):609.

Turdo A, Porcelli G, D’Accardo C, Franco SD, Verona F, Forte S, Giuffrida D, Memeo L, Todaro M, Stassi G. Metabolic Escape Routes of Cancer Stem Cells and Therapeutic Opportunities. Cancers (Basel). 2020 May 31;12(6):1436.

Villalobos-Ayala K, Ortiz Rivera I, Alvarez C, Husain K, DeLoach D, Krystal G, Hibbs ML, Jiang K, Ghansah T. Apigenin Increases SHIP-1 Expression, Promotes Tumoricidal Macrophages and Anti-Tumor Immune Responses in Murine Pancreatic Cancer. Cancers (Basel). 2020 Dec 4;12(12):3631. 

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.

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.

Warkad MS, Kim CH, Kang BG, Park SH, Jung JS, Feng JH, Inci G, Kim SC, Suh HW, Lim SS, Lee JY. Metformin-induced ROS upregulation as amplified by apigenin causes profound anticancer activity while sparing normal cells. Sci Rep. 2021 Jul 7;11(1):14002.

Wu Z, Zhang C, Najafi M. Targeting of the tumor immune microenvironment by metformin. J Cell Commun Signal. 2022 Sep;16(3):333-348.

Xi H, Kurtoglu M, Lampidis TJ. The wonders of 2-deoxy-D-glucose. IUBMB Life. 2014 Feb;66(2):110-21.

Xi H, Kurtoglu M, Liu H, Wangpaichitr M, You M, Liu X, Savaraj N, Lampidis TJ. 2-Deoxy-D-glucose activates autophagy via endoplasmic reticulum stress rather than ATP depletion. Cancer Chemother Pharmacol. 2011 Apr;67(4):899-910.

Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, Kang R, Tang D. Ferroptosis: process and function. Cell Death Differ. 2016 Mar;23(3):369-79.

Yamaguchi Y, Kasukabe T, Kumakura S. Piperlongumine rapidly induces the death of human pancreatic cancer cells mainly through the induction of ferroptosis. Int J Oncol. 2018 Mar;52(3):1011-1022.

Yan X, Qi M, Li P, Zhan Y, Shao H. Apigenin in cancer therapy: anti-cancer effects and mechanisms of action. Cell Biosci. 2017 Oct 5;7:50.

Yang J, Zhou Y, Xie S, Wang J, Li Z, Chen L, Mao M, Chen C, Huang A, Chen Y, Zhang X, Khan NUH, Wang L, Zhou J. Metformin induces Ferroptosis by inhibiting UFMylation of SLC7A11 in breast cancer. J Exp Clin Cancer Res. 2021 Jun 23;40(1):206.

Ye T, Xiong Y, Yan Y, Xia Y, Song X, Liu L, Li D, Wang N, Zhang L, Zhu Y, Zeng J, Wei Y, Yu L. The anthelmintic drug niclosamide induces apoptosis, impairs metastasis and reduces immunosuppressive cells in breast cancer model. PLoS One. 2014 Jan 8;9(1):e85887.

Yu Q, Xu M, Yu F, Jin Y. CD4(+)CD25(+) regulatory T cells as a therapeutic target in rheumatoid arthritis. Cent Eur J Immunol. 2014;39(1):100-3.

Yue D, Zhang D, Shi X, Liu S, Li A, Wang D, Qin G, Ping Y, Qiao Y, Chen X, Wang F, Chen R, Zhao S, Wang L, Zhang Y. Chloroquine Inhibits Stemness of Esophageal Squamous Cell Carcinoma Cells Through Targeting CXCR4-STAT3 Pathway. Front Oncol. 2020 Mar 13;10:311.

Zhang DF, Yang ZC, Chen JQ, Jin XX, Qiu YD, Chen XJ, Shi HY, Liu ZG, Wang MS, Liang G, Zheng XH. Piperlongumine inhibits migration and proliferation of castration-resistant prostate cancer cells via triggering persistent DNA damage. BMC Complement Med Ther. 2021 Jul 6;21(1):195.

Zhang F, Aft RL. Chemosensitizing and cytotoxic effects of 2-deoxy-D-glucose on breast cancer cells. J Cancer Res Ther. 2009 Sep;5 Suppl 1:S41-3.

Zhang Q, Wang J, Yadav DK, Bai X, Liang T. Glucose Metabolism: The Metabolic Signature of Tumor Associated Macrophage. Front Immunol. 2021 Jun 29;12:702580.

Zhang Q, Zhu B, Li Y. Resolution of Cancer-Promoting Inflammation: A New Approach for Anticancer Therapy. Front Immunol. 2017 Feb 2;8:71.

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.

Zhao Y, Xiao Z, Chen W, Yang J, Li T, Fan B. Disulfiram sensitizes pituitary adenoma cells to temozolomide by regulating O6-methylguanine-DNA methyltransferase expression. Mol Med Rep. 2015 Aug;12(2):2313-22.

Zhu X, Chen HH, Gao CY, Zhang XX, Jiang JX, Zhang Y, Fang J, Zhao F, Chen ZG. Energy metabolism in cancer stem cells. World J Stem Cells. 2020 Jun 26;12(6):448-461.

Updated October 3, 2023