Opportunities in the Metabolic Chaos of Cancer
Abstract
Cancer cells demonstrate the Warburg Effect. They ferment fuel by anaerobic glycolysis despite the presence of oxygen. This remarkable metabolic transformation at the onset of malignancy appears to be an innate adaptation to hypoxia. Cancer cells always continue some oxidative phosphorylation, and will die in a completely anoxic environment, but adapt to a crowded, compressed, and hypermetabolic situation by moving almost entirely to a metabolic phenotype that can operate without oxygen. Fermentation is not just an alternative way to obtain cellular energy – it provides increased biosynthesis of cellular components necessary to maintain exponential tumour growth. Anaerobic glycolysis is also a route to epigenetic alteration of the phenotype. Increased de novo synthesis includes not only structural cellular components, but also signaling molecules such as succinate that interact with both oncogene and tumour suppressor pathways. The mitochondria reprogram the cancer cell genetics to a new fermentative, fetal-like phenotype characterized by symmetrical mitosis. Targeting fermentation by cancer cells appears to have huge potential for healing and preventing cancer. Tumour growth and spread is linearly related to loss of, and damage to cancer cell mitochondria. Restoring mitochondria numbers and polarization, with redirection of cellular bioenergetics back towards oxidative phosphorylation, is possible. This tactic should result in lower rates of disease progression, invasion, metastasis, and relapses, particularly in common and deadly cancers which are grossly fermentative, such as of the breast, pancreas, liver, and colon.
Introduction
Cancer cells demonstrate the Warburg Effect (Koppenol et al 2011, Warburg 1956) – they ferment fuel by anaerobic glycolysis despite the presence of oxygen. This remarkable metabolic transformation at the onset of malignancy appears to be an innate adaptation to hypoxia. Surgeries and traumas create hypoxic zones. The Standard American Diet (SAD) creates a significant net acid residue compared to ancestral diets, reducing cytoplasmic oxygen retention (Frassetto et al 2001, Pizzorno 2012). Cancer cells always continue some oxidative phosphorylation, and will die in a completely anoxic environment, but adapt to a crowded, compressed, and hypermetabolic situation by moving almost entirely to a metabolic phenotype that can operate without oxygen. Reduced perfusion creates relative hypoxia, and the resulting shift to glycolytic metabolism (Fang et al 2008) generates high acid residue, which interacts with the pH gradient and resulting voltage gradient in mitochondria when moving protons necessary for ATP production by oxidative phosphorylation (Lewis et al 2002), reinforcing the adaptation to the hallmark cancer cell dependence on anaerobic glycolysis. Fermentation is not just an alternative way to obtain cellular energy, it provides increased biosynthesis of cellular components necessary to maintain exponential tumour growth and is a route to epigenetic alteration of the phenotype. Increased de novo synthesis includes not only structural cellular components, but also signaling molecules that interact with both oncogene and tumour suppressor pathways (Röhrig and Schulze 2016). The mitochondria reprogram the cancer cell genetics to a new fermentative, fetal-like phenotype (Seyfried 2012, Wallace 2012).
Targeting fermentation by cancer cells appears to have huge potential for healing and preventing cancer (Abdel-Wahab et al 2019, Bucay 2007, Frezza and Gottlieb 2009, Gogvadze et al 2008, Hanahan and Weinberg 2000, Hanahan and Weinberg 2011, Kulawiec et al 2009, Vander Heiden et al 2009, Wallace 2005, Yeung et al 2008). Tumour growth and spread is linearly related to loss of, and damage to, cancer cell mitochondria (Christofferson 2014). The landmark dichloroacetate (DCA) study of 2007 from the University of Alberta showed that restoring the malignant cell mitochondrial membrane potential can spark up oxidative metabolism, and this will arrest tumour growth (Bonnet et al 2007). Restoration of mitochondrial respiration must also influence the retrograde signaling and epigenetic reprogramming of cancer cells. This normalization of cellular bioenergetics should result in lower rates of disease progression, invasion, metastasis, and relapses, particularly in common and deadly cancers which are grossly fermentative, such as of the breast, pancreas, liver, and colon.
Possible Opportunities for Impacting Cancer Cell Metabolism
The number of cells in a given tissue remains constant once in the second trimester of gestation. Normal cells follow a mature growth pattern whereby cells do not reproduce, but if lost, are replaced from stem cells, in a process called asymmetrical mitosis. The stem cell makes a functional replacement differentiated cell, plus a new stem cell for future use. Cancer cells are reprogrammed to behave like the earlier fetal cells, with symmetrical mitosis generating masses of relatively undifferentiated cells. Cancer cells with this unlocked mitotic potential arise from a multitude of factors, such as radiation-induced mutations and DNA damage, toxic exposure such as to heavy metals (e.g. cadmium), or from bioactive chemicals such as pesticides. Inherited defects in DNA repair and other genetic issues account for perhaps 10% of cases. The Standard American Diet (SAD) leads to a dramatic increase in acid load and profound alterations in the sodium-potassium ratio (Frassetto et al 2001, Pizzorno 2013), which alters oxygen capacity of the cytoplasm. Mitochondrial and endoplasmic reticulum nutrient sensors adapt with changes to mitochondrial membrane function (Arnett 2010, De Saedeleer et al 2012, Fang et al 2008, Lemasters and Holmuhamedov 2006, Pizzorno 2012, Robey 2012). Mitochondria are also more sensitive and less able to repair DNA damage from most drugs and toxic substances (Cohen 2010, Darlington 1948). Cancers always have mitochondrial damage (Jurasunas 2006, Okouoyo et al 2004, Singh 2004, Varga et al 2015). The degree to which any cancer grows and spreads has a linear relationship to the number of lost or damaged mitochondria, and the shift to an increasing dependence on fermentation – burning fuel without oxygen (Christofferson 2014). Seyfried (Seyfried 2012, Seyfried et al 2014, Seyfried 2015, Seyfried et al 2017) and others have demonstrated that mitochondria are able to send retrograde signals to the nuclear DNA, reprogramming metabolism (Caino and Altieri 2016, Cerella et al 2015, Hung et al 2010, Ishikawa et al 2008, Jose et al 2011, Kaipparettu et al 2013, Ralph et al 2010, Wallace 2012). While anaerobic glycolysis is distinctly inferior to aerobic metabolism in terms of ATP energy production per molecule of fuel such as glucose, the cancer cells end up with normal amounts of energy by adaptations such as recruiting stromal cells to make ATP, a process called the Reverse Warburg Effect (Feron 2009, Sotgia et al 2011, Sotgia et al 2012).
Accumulation of succinate is a key aspect of metabolic dysregulation in carcinogenesis. This buildup of a Krebs cycle metabolite inhibits several α-ketoglutarate dioxygenases, thereby inducing a pseudohypoxia pathway (Teicher et al 2012), and via hypoxia inducible factors HIFαand HIF β, causes epigenetic reprogramming of genes regulating energy metabolism, angiogenesis and cell survival. Loss of succinate dehydrogenase enzyme (SDH) (aka Complex II) leads to reprogramming of cell metabolism to support glycolysis. Mutations of genes encoding for the succinate dehydrogenase complex are associated with familial paraganglioma, pheo-chromocytoma, renal cell carcinoma, gastrointestinal stromal tumors and, possibly, pituitary adenomas. SDHx-related tumors display the Warburg effect, driven by HIFα, which induces expression of GLUT1 and GLUT3, hexokinase 2, pyruvate kinase variant M2 (PKM2) and lactate dehydrogenase A (LDH-A), thereby stimulating the shift from oxidative phosphorylation to glycolysis. There is an overexpression of LDH-A which converts pyruvate to lactate, thereby recovering the NAD+ needed to maintain glycolysis, critically important for tumor proliferation. Inhibitors of GLUT1, pyruvate dehydrogenase kinase (eg DCA), and pyruvate carboxylase are potential targets for treatment of SDHx-associated tumors (Eijkelenkamp et al 2020). SDHx also triggers the epithelial to mesenchymal transition, cell migration and invasion (Dalla Pozza et al 2020). Anti-inflammatory tumour-associated macrophages (TAMs) secrete cytokine TGF-β that suppresses the transcription factor STAT1, decreasing the production of succinate dehydrogenase (SDH) enzyme in breast cancer cells (Gomez et al 2020). Melatonin may inhibit succinate accumulation (Gu et al 2020).
Central to the shift to a malignant phenotype is the down-regulation of phosphatases that dampen reactions and the up-regulation of kinases that stimulate metabolism and growth. The key metabolic controller pyruvate dehydrogenase kinase (PDK) creates a biochemical bottleneck, with a shift towards fermentation (Luo et al 2011, Pathania et al 2009). Under hypoxic conditions oxidative phosphorylation (OxiPhos) can no longer convert pyruvate to acetyl-CoA via the tricarboxylic acid (TCA) cycle. Anaerobic glycolysis uses lactate dehydrogenase (LDH) pathway to convert pyruvate to lactate, and lactate to pyruvate (da Veiga Moreira et al 2021).
The other hallmark of the malignant cell metabolism is glutaminolysis, the burning of glutamine as a fuel to generate TCA metabolites such as citrate, glutamate, pyruvate, and lactate, for the production of nucleic acids and other important cellular components needed by the cancer cells. For example, citrate can be used to make acetyl-CoA, which is needed to produce de novo lipids. These fatty acids are used structurally, but also as signalling molecules acting on oncogenes and tumour suppressor pathways (Röhrig and Schulze 2016). Normal cells use hexokinase one (HK1 or Hex I) on the mitochondrial membranes to move glucose, but cancer cells also make the unique hexokinase two (HK2 or Hex II) in abundance. HK2 takes ATP and glucose and produces glucose-6-phosphate. This precursor to lactate serves to lock on the throttle of glycolysis. Not only does this shift in biosynthesis of cellular components increase cell proliferation, it inhibits apoptosis, effectively immortalizing the malignant cells (Mathupala et al 2006).
In a landmark study in 2007 at the University of Alberta, it was demonstrated that dichloroacetate (DCA), a simple chlorinated vinegar, could inhibit PDK and shift mitochondria in cancer cells back to oxidative phosphorylation (Bonnet et al 2007). Rather than this pyruvate mimetic (Stockwin et al 2010) “giving the cancer more energy” as might have been imagined, the result was a dramatic decline in cancer growth. Unfortunately, premature hype of DCA (Coghlan 2007) lead to many people being harmed by self-prescribing on DCA obtained over the internet, overdosing (no human dose had been determined), or from using fraudulent or contaminated products. The first controlled human trial of DCA for cancer in 2010 showed significant toxicity to the peripheral nervous system, limiting its utility to brain and neurological cancers, which it rapidly saturates (Michelakis et al 2010). A smattering of published case studies has demonstrated some positive outcomes (Khan 2011, Khan 2012, Khan et al 2014, Khan et al 2017). DCA is highly neurotoxic when taken by mouth (Cornett 1999, Felitsyn 2007, Kaufmann 2006, Stacpoole 1998, Schaefer 2006).
The mechanism of action of DCA: DCA inhibits pyruvate dehydrogenase kinase, triggering an influx of acetyl-CoA into mitochondria. This drives more NADH into complex I. Superoxides that form are converted into hydrogen peroxide by manganese-super oxide dismutase. The H2O2 inhibits proton (H+) efflux, reducing mitochondrial membrane potential Dym, the proton-driving force → ATP. This opens the mitochondrial transition pore (MTP), inhibiting calcium ion entry via voltage-dependent channels. Reduced intra-mitochondrial calcium (Ca++) suppresses a tonic activation of nuclear factor of activated T-lymphocytes (NFAT). NFAT1 is a nuclear transcription activator, similar in action to activator protein 1 (AP-1) and nuclear factor kappa B (NFkB). This reduces Kv1.5 expression, increasing potassium ion K+ efflux, reducing inhibition of caspases, and finally triggering cancer cell apoptosis (Bonnet et al 2007). |
Fortunately, there are promising candidates for safer alternatives to DCA, as published in the first peer-reviewed paper on “mitochondrial rescue” as a potential cancer therapy (McKinney 2008), revised and republished in 2011 (McKinney 2011). The most important elements of this mitochondrial rescue protocol were R-alpha lipoic acid (ALA) (Abolhassani et al 2012, Baronzio et al 2012, Dörsam and Fahrer 2016) and thiamine – vitamin B1 (Babaei-Jadidi et al 2003, Comin-Anduix et al 2001). ALA activates pyruvate dehydrogenase (PDH) by inhibition of pyruvate dehydrogenase kinase (PDK), which results in an increased amount of pyruvate entering the tricarboxylic acid cycle. This drives the mitochondria towards oxidative phosphorylation and away from the Warburg effect anaerobic glycolysis on which cancer cells depend. Adjuncts to support R-ALA in normalizing the functionality of mitochondria include the Co-enzyme Q10 derivative PQQ (pyrroloquinoline quinone), thiamine derivative benfotiamine, acetyl-L-carnitine, grapeseed extract, and quercetin. Dr. Walter Lemmo’s discovery that DCA given by IV route was far less toxic than by mouth (private communication) revealed the potential to use DCA safely on non-neurological cancers. Later it was found that DCA and R-ALA could also be safely ingested by inhalation using a nebulizer (McKinney 2020B, McKinney 2020C). This metabolic approach was tried in many patients with advanced cancer, or cancers not responsive to the standard of care, with some good responses, including in some cases rapid tumor shrinkage. However, these anecdotal reports have yet to be fully tested in controlled studies or published under peer-review.
As well as fixing the damaged mitochondria in the cancer cell, it is also possible to increase the number of mitochondria, known as mitogenesis. One tactic for increasing mitogenesis is to increase intracellular nitric oxide, which can be accomplished with grapeseed extract proanthocyanidins (Vitseva et al 2005). Another mitogenic tactic is to activate adenosine monophosphate kinase (AMPK) (Chaube and Bhat 2016), which we may achieve with aerobic exercise, resveratrol, curcumin, quercetin, metformin, berberine, or green tea EGCG (McKinney 2020B, McKinney 2020C). Mitogenesis will also increase if peroxisome proliferator-activated receptor (PPAR) gamma γ (Jamwal et al 2021) coactivator 1α (PGC-1α) is inhibited with fermented wheat germ extract (FWGE) or red wine. PGC-1α is a transcriptional coactivator of the fusion mediator mitofusin-2, which is modulated by resveratrol. Also potentially effective would be increasing the levels of the “fountain of youth” sirtuin protein SIRT1, an NAD-dependent histone deacetylase, by caloric restriction, resveratrol, quercetin, or exercise (Archer 2013, Shigenaga and Ames 1993).
Another key part of the malignant mitochondrial metabolic puzzle is how to inhibit Hexokinase II (HK2 or Hex II), to further shift away from fermentation (DeWaal et al 2018, Mathupala et al 2006). HK2 binds to mitochondrial porin, redirecting mitochondrial ATP to phosphorylate glucose and driving glycolysis (Wallace 2005). Dr. Davis Lamson, ND, and others such as Israel and Schwarz in France put forward several candidate inhibitors, including hydroxycitrate from Garcinia cambogia (Guais et al 2012, Israel and Schaeffer 1988, Israel and Schwartz 2011, Schwartz et al 2010, Schwartz et al 2012, Schwartz et al 2013, Schwartz et al 2014). Clinical responses to hydroxycitrate have not been robust. In pre-clinical studies, lectins from Solomon’s seal herb (Polygonatum spp.) strongly inhibit HK2 (Wang et al 2011, Zhang et al 2017) and GLUT2 (Wang et al 2018). The target lectins are most abundant in the leaf, explaining why the whole-herb extracts seem to be much more bioactive in oncology than the root extracts commonly used for arthritis. The bioflavonoid quercetin is also a candidate Hex II inhibitor (Graziani 1977). Itraconazole is an antifungal drug that is being repurposed as a Hex II inhibitor (Gu et al 2016).
Another opportunity to intervene in the fermentation phenotype in cancer is how it regulates the highly acidic and toxic lactate output. Lactate can behave as a hypoxia mimetic factor capable of activating transcription factor HIF-1 in normoxic cancer cells, a key step in angiogenesis (Koukourakis et al 2005, Koukourakis et al 2006), and in triggering cancer cells to develop stem cell properties by which they become virulently malignant. Lactate over-production has many negative consequences (Walenta et al 2000, Walenta et al 2004), including reducing tumour antigenicity to dendritic immune cells (Gottfried et al 2006). Cancer cells are distinctly more alkaline on the inside than normal cells of the same type (Hao et al 2018), although the environment right around them is highly acidic (Newell et al 1993). This localized pH stress is an obvious therapeutic target (Huber et al 2017, Martin et al 2012, McCarty and Whitaker 2010, Pilon-Thomas et al 2016) but one that has proven elusive to overcome in practice. Laypeople as well as doctors have assumed from a cursory view of Warburg’s findings that a net alkaline residue diet, and alkaline therapies such as intravenous bicarbonate, will address this problem, but clinical results have been marginal (Martin et al 2012, Wenzel and Daniel 2004). Alkalizing increases patient quality of life but has little to no impact on progression of the disease. Rather than halting lactate production, or neutralizing it in the periphery of the tumour, it may be best to keep it in the cancer cell. In malignant cells there is a dramatic up-regulation of proton efflux pumps, shifting the acidic lactate from the cancer cell cytoplasm out into the pericellular spaces. This opens up the possibility of killing cancer cells by inhibiting these proton-coupled mono-carboxylate transporters, especially MCT1 and MCT4 (Benjamin et al 2018, Colen et al 2011, Payen et al 2020, Pérez-Escuredo et al 2016, Sun et al 2020). Quercetin appears to be the most promising intervention to address this tactic (Batiha et al 2020, Jones et al 2017, Ikegawa et al 2002).
Ketogenic diets are a relatively new concept in cancer care (Harper and Drewery 2019, Katz and Edelson 2017, McKinney 2020A, McKinney 2020C, Poff et al 2015, Seyfried et al 2009, Winters et al 2017). Very high fat, moderate protein, and very low carbohydrate ketogenic diets take advantage of the ability of normal cell mitochondria to adapt to ketones as fuel, while cancer cell mitochondria cannot do so (Davis 2017, de Cabo and Mattson 2019, Khodabakhshi et al 2020, Lee and Longo 2011, Longo and Fontana 2010). Some possible adjuncts to the ketogenic diet are supplemental ketones such as β-hydroxybutyrate, acetoacetate or 1,3,butane-diol, Metformin, berberine, Poly-MVA™ lipoic acid-palladium complex, hyperbaric oxygen therapy, and vitamin A retinol (Anderson 2018, Stengler and Anderson 2018). Fasting was once a widely used tool for healing, and research is now returning this dietary intervention to the clinical practice. Fasting – near, full, or intermittent – positively influences tolerance of SOC (standard of care) oncology therapies, and increases survival, in part by destressing mitochondria (Lee et al 2010, Lee and Longo 2011, Longo and Fontana 2010, Lv et al 2014, Safdie et al 2009, Simone et al 2018).
Discussion
Cancer cells adopt a phenotype largely dependent on anaerobic glycolysis. The cell is epigenetically reprogrammed to a fetal growth pattern. Since the cell has once undergone a shift from the fetal growth pattern (symmetrical mitosis) to the mature normal cell replacement model (asymmetrical mitosis), it is possible a mechanism exists to move the cancer cell from symmetrical mitosis to the normal asymmetrical pattern. Mitochondria in cancer cells drive the metabolic shifts and retrograde signalling to the genome, and their numbers and functionality correspond to the aggressiveness of the disease. Since the aberrant metabolism of cancer cell mitochondria can be corrected, it suggests a possible path to actually healing the cancer cell. Natural medicine candidates with pre-clinical and limited clinical evidence demonstrating the ability to correct cancer cell metabolism and mitochondria include quercetin (Batiha et al 2020, Zhang et al 2005, Das et al 2020, Jones et al 2017, Kothan et al 2004, Langner et al 2019, Ikegawa et al 2002, Suolinna et al 1975), R-alpha lipoic acid (da Veiga Moreira et al 2021, Humphries and Szweda1998, Korotchkina et al 2004, Moffa et al 2019, Wenzel et al 2005), thiamine (Parkhomenko et al 1987, Sheline et al 2002), acetyl-L-carnitine (Hoang et al 2007, Wenzel et al 2005), phloretin (Abdel-Wahab et al 2019, Choi 2019, Tang and Gong 2021), grapeseed proanthocyanidins (Nguyen and Pandey 2019), Coenzyme Q10 (Berbel-Garcia et al 2004, Smith et al 2008), Solomon’s seal (Polygonatum spp.) lectins, and blackseed (Nigella sativa) thymoquinone (Tavakkoli et al 2017). A ketogenic diet appears to be supportive of this positive metabolic shift, as does fasting.
Common clinical doses of candidate substances for metabolic regulation:
- Acetyl-L-carnitine 1000mg tid.
- Alpha lipoic acid 300mg bid R-ALA or 600mg bid if DL-ALA.
- May also be administered by IV 150mg twice a week, or by nebulization 50-100mg bid.
- Berberine 300-500mg bid Coenzyme Q10 – 300mg qd of ubiquinone or 100mg of ubiquinol.
- Curcumin -120mg bid of TheraCurmin™ or bid water-soluble curcumin extract.
- Fermented wheat germ extract (FWGE) 9g qd – bid.
- Grapeseed extract 250mg bid.
- Hydroxycitrate from Garcinia cambogia 1000mg bid.
- Phloretin 1000mg bid.
- Quercetin 1000mg bid, less if in liposomal format, delivering up to 140mg bid of dihydroquercetin.
- Red wine 1 glass daily.
- Resveratrol 250mg bid.
- Sodium bicarbonate – up to ½tsp bid.
- Solomon’s seal aerial herb Polygonatum spp. (minus seeds which are toxic) 5:1 tincture – 30 drops (½tsp) bid.
- Thymoquinone, from blackseed Nigella sativa, 100-200mg bid.
- Vitamin A retinol 25000IU retinol or 30000IU palmitate daily, but lower to 5,000IU if ALT or AST are over 300.
- Vitamin B1 – 100-200mg bid as thiamine, or 160mg bid as Benfotiamine™.
References
Abdel-Wahab AF, Mahmoud W, Al-Harizy RM. Targeting glucose metabolism to suppress cancer progression: prospective of anti-glycolytic cancer therapy. Pharmacol Res. 2019 Dec;150:104511.
Abolhassani M, Guais A, Sanders E, Campion F, Fichtner I, Bonte J, Baronzio G, Fiorentini G, Israël M, Schwartz L. Screening of well-established drugs targeting cancer metabolism: reproducibility of the efficacy of a highly effective drug combination in mice. Invest New Drugs. 2012 Aug;30(4):1331-1342.
Anderson PS. Metabolic Therapies in Advanced “Salvage” Cancer Cases. Townsend Letter. 2018;Aug/Sept:39-43.
Archer SL. Mitochondrial dynamics–mitochondrial fission and fusion in human diseases. N Engl J Med. 2013 Dec 5;369(23):2236-2251.
Arnett TR. Acidosis, hypoxia and bone. Arch Biochem Biophys. 2010 Nov 1;503(1):103-109.
Babaei-Jadidi R, Karachalias N, Ahmed N, Battah S, Thornalley PJ. Prevention of incipient diabetic nephropathy by high-dose thiamine and benfotiamine. Diabetes. 2003 Aug;52(8):2110-2120.
Batiha GE, Beshbishy AM, Ikram M, Mulla ZS, El-Hack MEA, Taha AE, Algammal AM, Elewa YHA. The Pharmacological Activity, Biochemical Properties, and Pharmacokinetics of the Major Natural Polyphenolic Flavonoid: Quercetin. Foods. 2020 Mar 23;9(3):374.
Baronzio G, Laurent S, Elisabetta C, Guais A, Sanders E, Delépine N, Fiorentini G. Early clinical and toxicological results of a combination of natural glycolysis inhibitors (METABLOC TM) on cancer patients. Biomedical Research. 2012;23:219-222.
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.
Berbel-Garcia A, Barbera-Farre JR, Etessam JP, Salio AM, Cabello A, Gutierrez-Rivas E, Campos Y. Coenzyme Q 10 improves lactic acidosis, strokelike episodes, and epilepsy in a patient with MELAS (mitochondrial myopathy, encephalopathy, lactic acidosis, and strokelike episodes). Clin Neuropharmacol. 2004 Jul-Aug;27(4):187-191.
Bonnet S, Archer SL, Allalunis-Turner J, Haromy A, Beaulieu C, Thompson R, Lee CT, Lopaschuk GD, Puttagunta L, Bonnet S, Harry G, Hashimoto K, Porter CJ, Andrade MA, Thebaud B, Michelakis ED. A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell. 2007 Jan;11(1):37-51.
Bucay AH. The biological significance of cancer: mitochondria as a cause of cancer and the inhibition of glycolysis with citrate as a cancer treatment. Med Hypotheses. 2007;69(4):826-828.
Caino MC, Altieri DC. Molecular Pathways: Mitochondrial Reprogramming in Tumor Progression and Therapy. Clin Cancer Res. 2016 Feb 1;22(3):540-545.
Cerella C, Gaigneaux A, Dicato M, Diederich M. Antagonistic role of natural compounds in mTOR-mediated metabolic reprogramming. Cancer Lett. 2015 Jan 28;356(2 Pt A):251-262.
Chaube B, Bhat MK. AMPK, a key regulator of metabolic/energy homeostasis and mitochondrial biogenesis in cancer cells. Cell Death Dis. 2016 Jan 14;7(1):e2044.
Choi BY. Biochemical Basis of Anti-Cancer-Effects of Phloretin-A Natural Dihydrochalcone. Molecules. 2019 Jan 13;24(2):278.
Christofferson T. Tripping Over the Truth: The Return of the Metabolic Theory of Cancer Illuminates a New and Hopeful Path to a Cure. CreateSpace Independent Publishing Platform. Scotts Valley, California. 2014.
Coghlan A. Cheap, Safe Drug Kills Most Cancers, New Scientist January 2007.
Cohen BH. Pharmacologic effects on mitochondrial function. Dev Disabil Res Rev. 2010;16(2):189-199.
Colen CB, Shen Y, Ghoddoussi F, Yu P, Francis TB, Koch BJ, Monterey MD, Galloway MP, Sloan AE, Mathupala SP. Metabolic targeting of lactate efflux by malignant glioma inhibits invasiveness and induces necrosis: an in vivo study. Neoplasia. 2011 Jul;13(7):620-632.
Comín-Anduix B, Boren J, Martinez S, Moro C, Centelles JJ, Trebukhina R, Petushok N, Lee WN, Boros LG, Cascante M. The effect of thiamine supplementation on tumour proliferation. A metabolic control analysis study. Eur J Biochem. 2001 Aug;268(15):4177-4182.
Cornett R, James MO, Henderson GN, Cheung J, Shroads AL, Stacpoole PW. Inhibition of glutathione S-transferase zeta and tyrosine metabolism by dichloroacetate: a potential unifying mechanism for its altered biotransformation and toxicity. Biochem Biophys Res Commun. 1999 Sep 7;262(3):752-756.
Dalla Pozza E, Dando I, Pacchiana R, Liboi E, Scupoli MT, Donadelli M, Palmieri M. Regulation of succinate dehydrogenase and role of succinate in cancer. Semin Cell Dev Biol. 2020 Feb;98:4-14.
Darlington CD. The plasmagene theory of the origin of cancer. Br J Cancer. 1948 Jun;2(2):118-126.
Das SS, Hussain A, Verma PRP, Imam SS, Altamimi MA, Alshehri S, Singh SK. Recent Advances in Liposomal Drug Delivery System of Quercetin for Cancer Targeting: A Mechanistic Approach. Curr Drug Deliv. 2020;17(10):845-860.
da Veiga Moreira J, De Staercke L, César Martínez-Basilio P, Gauthier-Thibodeau S, Montégut L, Schwartz L, Jolicoeur M. Hyperosmolarity Triggers the Warburg Effect in Chinese Hamster Ovary Cells and Reveals a Reduced Mitochondria Horsepower. Metabolites. 2021 May 26;11(6):344.
Davis E. Fight Cancer With a Ketogenic Diet – Using a Low-Carb, Fat-Burning Diet as Metabolic Therapy, 3rd Ed. Gutsy Badger. 2017.
de Cabo R, Mattson MP. Effects of Intermittent Fasting on Health, Aging, and Disease. N Engl J Med. 2019 Dec 26;381(26):2541-2551.
De Saedeleer CJ, Copetti T, Porporato PE, Verrax J, Feron O, Sonveaux P. Lactate activates HIF-1 in oxidative but not in Warburg-phenotype human tumor cells. PLoS One. 2012;7(10):e46571.
DeWaal D, Nogueira V, Terry AR, Patra KC, Jeon SM, Guzman G, Au J, Long CP, Antoniewicz MR, Hay N. Hexokinase-2 depletion inhibits glycolysis and induces oxidative phosphorylation in hepatocellular carcinoma and sensitizes to metformin. Nat Commun. 2018 Jan 31;9(1):446.
Dörsam B, Fahrer J. The disulfide compound α-lipoic acid and its derivatives: A novel class of anticancer agents targeting mitochondria. Cancer Lett. 2016 Feb 1;371(1):12-19.
Eijkelenkamp K, Osinga TE, Links TP, van der Horst-Schrivers ANA. Clinical implications of the oncometabolite succinate in SDHx-mutation carriers. Clin Genet. 2020 Jan;97(1):39-53.
Fang JS, Gillies RD, Gatenby RA. Adaptation to hypoxia and acidosis in carcinogenesis and tumor progression. Semin Cancer Biol. 2008 Oct;18(5):330-337.
Felitsyn N, Stacpoole PW, Notterpek L. Dichloroacetate causes reversible demyelination in vitro: potential mechanism for its neuropathic effect. J Neurochem. 2007 Jan;100(2):429-436.
Feron O. Pyruvate into lactate and back: from the Warburg effect to symbiotic energy fuel exchange in cancer cells. Radiother Oncol. 2009 Sep;92(3):329-333.
Frassetto L, Morris RC Jr, Sellmeyer DE, Todd K, Sebastian A. Diet, evolution and aging–the pathophysiologic effects of the post-agricultural inversion of the potassium-to-sodium and base-to-chloride ratios in the human diet. Eur J Nutr. 2001 Oct;40(5):200-213.
Frezza C, Gottlieb E. Mitochondria in cancer: not just innocent bystanders. Semin Cancer Biol. 2009 Feb;19(1):4-11.
Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria in cancer cells: what is so special about them? Trends Cell Biol. 2008 Apr;18(4):165-173.
Gómez V, Eykyn TR, Mustapha R, Flores-Borja F, Male V, Barber PR, Patsialou A, Green R, Panagaki F, Li CW, Fruhwirth GO, Ros S, Brindle KM, Ng T. Breast cancer-associated macrophages promote tumorigenesis by suppressing succinate dehydrogenase in tumor cells. Sci Signal. 2020 Oct 6;13(652):eaax4585.
Gottfried E, Kunz-Schughart LA, Ebner S, Mueller-Klieser W, Hoves S, Andreesen R, Mackensen A, Kreutz M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood. 2006 Mar 1;107(5):2013-2021.
Graziani Y. Bioflavonoid regulation of ATPase and hexokinase activity in Ehrlich ascites cell mitochondria. Biochim Biophys Acta. 1977 May 11;460(2):364-373.
Gu C, Yang H, Chang K, Zhang B, Xie F, Ye J, Chang R, Qiu X, Wang Y, Qu Y, Wang J, Li M. Melatonin alleviates progression of uterine endometrial cancer by suppressing estrogen/ubiquitin C/SDHB-mediated succinate accumulation. Cancer Lett. 2020 Apr 28;476:34-47.
Gu JY, Mavis L, Barth C, Hernandez-Ilizaliturri M. Itraconazole, an Oral Antifungal Drug, Is Active in Chemotherapy Resistant B-Cell Non-Hodgkin Lymphoma and Enhances the Anti-Tumor Activity of Chemotherapy Agents. Blood. 2016;128:5138.
Guais A, Baronzio G, Sanders E, Campion F, Mainini C, Fiorentini G, Montagnani F, Behzadi M, Schwartz L, Abolhassani M. Adding a combination of hydroxycitrate and lipoic acid (METABLOC™) to chemotherapy improves effectiveness against tumor development: experimental results and case report. Invest New Drugs. 2012 Feb;30(1):200-211.
Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000 Jan 7;100(1):57-70.
Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011 Mar 4;144(5):646-674.
Hao G, Xu ZP, Li L. Manipulating extracellular tumour pH: an effective target for cancer therapy. RSC Adv. 2018 Jun 19;8(39):22182-22192.
Harper DG, Drewery D. BioDiet: The Scientifically Proven, Ketogenic Way to Lose Weight and Improve Your Health. Page Two Books. Vancouver, British Columbia. 2019.
Hoang BX, Graeme Shaw D, Pham P, Levine S. Restoration of cellular energetic balance with L-carnitine in the neuro-bioenergetic approach for cancer prevention and treatment. Med Hypotheses. 2007;69(2):262-272.
Huber V, Camisaschi C, Berzi A, Ferro S, Lugini L, Triulzi T, Tuccitto A, Tagliabue E, Castelli C, Rivoltini L. Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation. Semin Cancer Biol. 2017 Apr;43:74-89.
Humphries KM, Szweda LI. Selective inactivation of alpha-ketoglutarate dehydrogenase and pyruvate dehydrogenase: reaction of lipoic acid with 4-hydroxy-2-nonenal. Biochemistry. 1998 Nov 10;37(45):15835-15841.
Hung WY, Wu CW, Yin PH, Chang CJ, Li AF, Chi CW, Wei YH, Lee HC. Somatic mutations in mitochondrial genome and their potential roles in the progression of human gastric cancer. Biochim Biophys Acta. 2010 Mar;1800(3):264-270.
Ikegawa T, Ohtani H, Koyabu N, Juichi M, Iwase Y, Ito C, Furukawa H, Naito M, Tsuruo T, Sawada Y. Inhibition of P-glycoprotein by flavonoid derivatives in adriamycin-resistant human myelogenous leukemia (K562/ADM) cells. Cancer Lett. 2002 Mar 8;177(1):89-93.
Ishikawa K, Takenaga K, Akimoto M, Koshikawa N, Yamaguchi A, Imanishi H, Nakada K, Honma Y, Hayashi J. ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science. 2008 May 2;320(5876):661-664.
Israel BA, Schaeffer WI. Cytoplasmic mediation of malignancy. In Vitro Cell Dev Biol. 1988 May;24(5):487-490.
Israel M, Schwartz L. On the metabolic origin of cancer: substances that target tumor metabolism. Biomedical Research-tokyo, 2011A;22:0.
Jamwal S, Blackburn JK, Elsworth JD. PPARγ/PGC1α signaling as a potential therapeutic target for mitochondrial biogenesis in neurodegenerative disorders. Pharmacol Ther. 2021 Mar;219:107705.
Jones RS, Parker MD, Morris ME. Quercetin, Morin, Luteolin, and Phloretin Are Dietary Flavonoid Inhibitors of Monocarboxylate Transporter 6. Mol Pharm. 2017 Sep 5;14(9):2930-2936.
Jose C, Bellance N, Rossignol R. Choosing between glycolysis and oxidative phosphorylation: a tumor’s dilemma? Biochim Biophys Acta. 2011 Jun;1807(6):552-561.
Jurasunas S. Mitochondria and cancer. Townsend Letter: The Examiner of Alternative Medicine. 2006;277-278:83.
Kaipparettu BA, Ma Y, Park JH, Lee TL, Zhang Y, Yotnda P, Creighton CJ, Chan WY, Wong LJ. Crosstalk from non-cancerous mitochondria can inhibit tumor properties of metastatic cells by suppressing oncogenic pathways. PLoS One. 2013 May 9;8(5):e61747.
Katz R, Edelson M. The Cancer-Fighting Kitchen, Nourishing Big Flavor Recipes for Cancer Treatment and Recovery, 2nd Edition. Penguin Random House. London, England. 2017.
Kaufmann P, Engelstad K, Wei Y, Jhung S, Sano MC, Shungu DC, Millar WS, Hong X, Gooch CL, Mao X, Pascual JM, Hirano M, Stacpoole PW, DiMauro S, De Vivo DC. Dichloroacetate causes toxic neuropathy in MELAS: a randomized, controlled clinical trial. Neurology. 2006 Feb 14;66(3):324-330.
Khan A. Use of oral dichloroacetate for palliation of leg pain arising from metastatic poorly differentiated carcinoma: a case report. J Palliat Med. 2011 Aug;14(8):973-9777.
Khan A. Case Report of Long-Term Complete Remission of Metastatic Renal Squamous Cell Carcinoma After Palliative Radiotherapy and Adjuvant Dichloroacetate. Adv. Cancer Res. Treat. 2012;2012:441895.
Khan A, Marier D, Marsden E, Andrews D, Eliaz I. A novel form of dichloroacetate therapy for patients with advanced cancer: a report of 3 cases. Altern Ther Health Med. 2014 Oct;20 Suppl 2:21-28.
Khan A, Andrews D, Shainhouse J, Blackburn AC. Long-term stabilization of metastatic melanoma with sodium dichloroacetate. World J Clin Oncol. 2017 Aug 10;8(4):371-377.
Khodabakhshi A, Akbari ME, Mirzaei HR, Mehrad-Majd H, Kalamian M, Davoodi SH. Feasibility, Safety, and Beneficial Effects of MCT-Based Ketogenic Diet for Breast Cancer Treatment: A Randomized Controlled Trial Study. Nutr Cancer. 2020;72(4):627-634.
Koppenol WH, Bounds PL, Dang CV. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat Rev Cancer. 2011 May;11(5):325-337.
Korotchkina LG, Sidhu S, Patel MS. R-lipoic acid inhibits mammalian pyruvate dehydrogenase kinase. Free Radic Res. 2004 Oct;38(10):1083-1092.
Kothan S, Dechsupa S, Leger G, Moretti JL, Vergote J, Mankhetkorn S. Spontaneous mitochondrial membrane potential change during apoptotic induction by quercetin in K562 and K562/adr cells. Can J Physiol Pharmacol. 2004 Dec;82(12):1084-1090.
Koukourakis MI, Giatromanolaki A, Simopoulos C, Polychronidis A, Sivridis E. Lactate dehydrogenase 5 (LDH5) relates to up-regulated hypoxia inducible factor pathway and metastasis in colorectal cancer. Clin Exp Metastasis. 2005;22(1):25-30.
Koukourakis MI, Giatromanolaki A, Sivridis E, Gatter KC, Harris AL; Tumour Angiogenesis Research Group. Lactate dehydrogenase 5 expression in operable colorectal cancer: strong association with survival and activated vascular endothelial growth factor pathway–a report of the Tumour Angiogenesis Research Group. J Clin Oncol. 2006 Sep 10;24(26):4301-4308.
Kulawiec M, Owens KM, Singh KK. Cancer cell mitochondria confer apoptosis resistance and promote metastasis. Cancer Biol Ther. 2009 Jul;8(14):1378-1385.
Langner E, Lemieszek MK, Rzeski W. Lycopene, sulforaphane, quercetin, and curcumin applied together show improved antiproliferative potential in colon cancer cells in vitro. J Food Biochem. 2019 Apr;43(4):e12802.
Lee C, Safdie FM, Raffaghello L, Wei M, Madia F, Parrella E, Hwang D, Cohen P, Bianchi G, Longo VD. Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res. 2010 Feb 15;70(4):1564-1572.
Lee C, Longo VD. Fasting vs dietary restriction in cellular protection and cancer treatment: from model organisms to patients. Oncogene. 2011 Jul 28;30(30):3305-3316.
Lemasters JJ, Holmuhamedov E. Voltage-dependent anion channel (VDAC) as mitochondrial governator–thinking outside the box. Biochim Biophys Acta. 2006 Feb;1762(2):181-190.
Lewis J, Morgan DO, Raff M. Molecular Biology of the Cell, 4th Edition. Garland Publishing. New York, USA. 2002.
Longo VD, Fontana L. Calorie restriction and cancer prevention: metabolic and molecular mechanisms. Trends Pharmacol Sci. 2010 Feb;31(2):89-98.
Luo W, Hu H, Chang R, Zhong J, Knabel M, O’Meally R, Cole RN, Pandey A, Semenza GL. Pyruvate kinase M2 is a PHD3-stimulated coactivator for hypoxia-inducible factor 1. Cell. 2011 May 27;145(5):732-744.
Lv M, Zhu X, Wang H, Wang F, Guan W. Roles of caloric restriction, ketogenic diet and intermittent fasting during initiation, progression and metastasis of cancer in animal models: a systematic review and meta-analysis. PLoS One. 2014 Dec 11;9(12):e115147.
Martin NK, Robey IF, Gaffney EA, Gillies RJ, Gatenby RA, Maini PK. Predicting the safety and efficacy of buffer therapy to raise tumour pHe: an integrative modelling study. Br J Cancer. 2012 Mar 27;106(7):1280-1287.
Mathupala SP, Ko YH, Pedersen PL. Hexokinase II: cancer’s double-edged sword acting as both facilitator and gatekeeper of malignancy when bound to mitochondria. Oncogene. 2006 Aug 7;25(34):4777-4786.
McCarty MF, Whitaker J. Manipulating tumor acidification as a cancer treatment strategy. Altern Med Rev. 2010 Sep;15(3):264-272.
McKinney N. Ketogenic Diet and Ketosis Supplements: Metabolic Therapeutics in Primary Care, A Clinical Handbook. Nutritional Fundamentals for Health. Montreal, Canada. 2020A.
McKinney N. Mitochondria Rescue (Possibly) Heals Cancer? Naturopathic Doctor News & Review.2008;4(5):10-11.
McKinney N. Mitochondrial Rescue –Turning Cancer Cells Off, Integrative Healthcare Practitioners. 2011;4(4):78-82.
McKinney N. Mitochondria: The Power to Energize Healing of Chronic, Degenerative Diseases and Aging, A Clinical Handbook. Nutritional Fundamentals for Health. Montreal, Canada. 2020B.
McKinney N. Naturopathic Oncology-An Encyclopedic Guide for Patients and Physicians, Fourth edition. Liaison Press. Canada. 2020C.
Michelakis ED, Sutendra G, Dromparis P, Webster L, Haromy A, Niven E, Maguire C, Gammer TL, Mackey JR, Fulton D, Abdulkarim B, McMurtry MS, Petruk KC. Metabolic modulation of glioblastoma with dichloroacetate. Sci Transl Med. 2010 May 12;2(31):31ra34.
Moffa S, Improta I, Rocchetti S, Mezza T, Giaccari A. Potential cause-effect relationship between insulin autoimmune syndrome and alpha lipoic acid: Two case reports. Nutrition. 2019 Jan;57:1-4.
Newell K, Franchi A, Pouysségur J, Tannock I. Studies with glycolysis-deficient cells suggest that production of lactic acid is not the only cause of tumor acidity. Proc Natl Acad Sci U S A. 1993 Feb 1;90(3):1127-1131.
Nguyen C, Pandey S. Exploiting Mitochondrial Vulnerabilities to Trigger Apoptosis Selectively in Cancer Cells. Cancers (Basel). 2019 Jun 29;11(7):916.
Okouoyo S, Herzer K, Ucur E, Mattern J, Krammer PH, Debatin KM, Herr I. Rescue of death receptor and mitochondrial apoptosis signaling in resistant human NSCLC in vivo. Int J Cancer. 2004 Feb 10;108(4):580-587.
Parkhomenko IuM, Chernysh IIu, Churilova TIa, Khalmuradov AG. Vliianie tiaminfosfatov na aktivnost’ reguliatornykh fermentov piruvatdegidrogenaznogo kompleksa [Effect of thiamine phosphates on the activity of regulatory enzymes of the pyruvate dehydrogenase complex]. Ukr Biokhim Zh (1978). 1987 Sep-Oct;59(5):49-54.
Pathania D, Millard M, Neamati N. Opportunities in discovery and delivery of anticancer drugs targeting mitochondria and cancer cell metabolism. Adv Drug Deliv Rev. 2009 Nov 30;61(14):1250-1275.
Payen VL, Mina E, Van Hée VF, Porporato PE, Sonveaux P. Monocarboxylate transporters in cancer. Mol Metab. 2020 Mar;33:48-66.
Pérez-Escuredo J, Van Hée VF, Sboarina M, Falces J, Payen VL, Pellerin L, Sonveaux P. Monocarboxylate transporters in the brain and in cancer. Biochim Biophys Acta. 2016 Oct;1863(10):2481-2497.
Pilon-Thomas S, Kodumudi KN, El-Kenawi AE, Russell S, Weber AM, Luddy K, Damaghi M, Wojtkowiak JW, Mulé JJ, Ibrahim-Hashim A, Gillies RJ. Neutralization of Tumor Acidity Improves Antitumor Responses to Immunotherapy. Cancer Res. 2016 Mar 15;76(6):1381-1390.
Pizzorno JE. Mitochondria, the Foundation of Health and Energy (lecture). Kelowna, BC. July 26, 2013.
Pizzorno JE. pH: Diet Induced Cellular Acidosis (lecture). Kelowna, BC. July 20, 2012.
Poff AM, Ward N, Seyfried TN, Arnold P, D’Agostino DP. Non-Toxic Metabolic Management of Metastatic Cancer in VM Mice: Novel Combination of Ketogenic Diet, Ketone Supplementation, and Hyperbaric Oxygen Therapy. PLoS One. 2015 Jun 10;10(6):e0127407.
Ralph SJ, Rodríguez-Enríquez S, Neuzil J, Saavedra E, Moreno-Sánchez R. The causes of cancer revisited: “mitochondrial malignancy” and ROS-induced oncogenic transformation – why mitochondria are targets for cancer therapy. Mol Aspects Med. 2010 Apr;31(2):145-170.
Robey IF. Examining the relationship between diet-induced acidosis and cancer. Nutr Metab (Lond). 2012 Aug 1;9(1):72.
Röhrig F, Schulze A. The multifaceted roles of fatty acid synthesis in cancer. Nat Rev Cancer. 2016 Nov;16(11):732-749.
Safdie FM, Dorff T, Quinn D, Fontana L, Wei M, Lee C, Cohen P, Longo VD. Fasting and cancer treatment in humans: A case series report. Aging (Albany NY). 2009 Dec 31;1(12):988-1007.
Schaefer AM. Trial of dichloroacetate in MELAS: toxicity overshadows the assessment of potential benefit. Neurology. 2006 Feb 14;66(3):302-303.
Schwartz L, Abolhassani M, Guais A, Sanders E, Steyaert JM, Campion F, Israël M. A combination of alpha lipoic acid and calcium hydroxycitrate is efficient against mouse cancer models: preliminary results. Oncol Rep. 2010 May;23(5):1407-1416.
Schwartz L, Buhler L, Icard P, Lincet H, Steyaert JM. Metabolic treatment of cancer: intermediate results of a prospective case series. Anticancer Res. 2014 Feb;34(2):973-980.
Schwartz L, Guais A, Israël M, Junod B, Steyaert JM, Crespi E, Baronzio G, Abolhassani M. Tumor regression with a combination of drugs interfering with the tumor metabolism: efficacy of hydroxycitrate, lipoic acid and capsaicin. Invest New Drugs. 2013 Apr;31(2):256-264.
Schwartz L, Summa M,Steyaert JM, Adeline Guais-Vergne A,Baronzio GF. New Cancer Paradigm and New Treatment: The Example of METABLOC. Conference of the International Clinical Hyperthermia Society 2012. 2013;2013:827686.
Seyfried BT, Kiebish M, Marsh J, Mukherjee P. Targeting energy metabolism in brain cancer through calorie restriction and the ketogenic diet. J Cancer Res Ther. 2009 Sep;5 Suppl 1:S7-15.
Seyfried T. Cancer As A Metabolic Disease – On the Origin, Management and Prevention of Cancer. John Wiley and Sons. Hoboken, New Jersey. 2012.
Seyfried TN, Flores RE, Poff AM, D’Agostino DP. Cancer as a metabolic disease: implications for novel therapeutics. Carcinogenesis. 2014 Mar;35(3):515-527.
Seyfried TN. Cancer as a Mitochondrial Metabolic Disease. Hypothesis and Theory. 2015.
Seyfried TN, Yu G, Maroon JC, D’Agostino DP. Press-pulse: a novel therapeutic strategy for the metabolic management of cancer. Nutr Metab (Lond). 2017 Feb 23;14:19.
Sheline CT, Choi EH, Kim-Han JS, Dugan LL, Choi DW. Cofactors of mitochondrial enzymes attenuate copper-induced death in vitro and in vivo. Ann Neurol. 2002 Aug;52(2):195-204.
Shigenaga MK, Ames BN. Oxidants and mitogenesis as causes of mutation and cancer: the influence of diet. Basic Life Sci. 1993;61:419-436.
Simone BA, Palagani A, Strickland K, Ko K, Jin L, Lim MK, Dan TD, Sarich M, Monti DA, Cristofanilli M, Simone NL. Caloric restriction counteracts chemotherapy-induced inflammation and increases response to therapy in a triple negative breast cancer model. Cell Cycle. 2018;17(13):1536-1544.
Singh KK. Mitochondrial dysfunction is a common phenotype in aging and cancer. Ann N Y Acad Sci. 2004 Jun;1019:260-264.
Smith RA, Adlam VJ, Blaikie FH, Manas AR, Porteous CM, James AM, Ross MF, Logan A, Cochemé HM, Trnka J, Prime TA, Abakumova I, Jones BA, Filipovska A, Murphy MP. Mitochondria-targeted antioxidants in the treatment of disease. Ann N Y Acad Sci. 2008 Dec;1147:105-111.
Sotgia F, Martinez-Outschoorn UE, Lisanti MP. Mitochondrial oxidative stress drives tumor progression and metastasis: should we use antioxidants as a key component of cancer treatment and prevention? BMC Med. 2011 May 23;9:62.
Sotgia F, Whitaker-Menezes D, Martinez-Outschoorn UE, Flomenberg N, Birbe RC, Witkiewicz AK, Howell A, Philp NJ, Pestell RG, Lisanti MP. Mitochondrial metabolism in cancer metastasis: visualizing tumor cell mitochondria and the “reverse Warburg effect” in positive lymph node tissue. Cell Cycle. 2012 Apr 1;11(7):1445-1454.
Stacpoole PW, Henderson GN, Yan Z, James MO. Clinical pharmacology and toxicology of dichloroacetate. Environ Health Perspect. 1998 Aug;106 Suppl 4(Suppl 4):989-94.
Stengler M, Anderson P. Outside the Box Cancer Therapies – Alternative Therapies That Treat and Prevent Cancer. Hay House. Carlsbad, California. 2018.
Stockwin LH, Yu SX, Borgel S, Hancock C, Wolfe TL, Phillips LR, Hollingshead MG, Newton DL. Sodium dichloroacetate selectively targets cells with defects in the mitochondrial ETC. Int J Cancer. 2010 Dec 1;127(11):2510-2519.
Sun X, Wang M, Wang M, Yao L, Li X, Dong H, Li M, Sun T, Liu X, Liu Y, Xu Y. Role of Proton-Coupled Monocarboxylate Transporters in Cancer: From Metabolic Crosstalk to Therapeutic Potential. Front Cell Dev Biol. 2020 Jul 17;8:651.
Suolinna EM, Buchsbaum RN, Racker E. The effect of flavonoids on aerobic glycolysis and growth of tumor cells. Cancer Res. 1975 Jul;35(7):1865-1872.
Tang J, Gong Y. Synergistic Effect of Phloretin Combined With Radiotherapy on Lung Cancer. International Journal of Radiation Oncology, Biology, Physics. 2021;111(3):e238.
Tavakkoli A, Mahdian V, Razavi BM, Hosseinzadeh H. Review on Clinical Trials of Black Seed (Nigella sativa) and Its Active Constituent, Thymoquinone. J Pharmacopuncture. 2017 Sep;20(3):179-193.
Teicher BA, Linehan WM, Helman LJ. Targeting cancer metabolism. Clin Cancer Res. 2012 Oct 15;18(20):5537-45.
Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009 May 22;324(5930):1029-1033.
Varga ZV, Ferdinandy P, Liaudet L, Pacher P. Drug-induced mitochondrial dysfunction and cardiotoxicity. Am J Physiol Heart Circ Physiol. 2015 Nov;309(9):H1453-1467.
Vitseva O, Varghese S, Chakrabarti S, Folts JD, Freedman JE. Grape seed and skin extracts inhibit platelet function and release of reactive oxygen intermediates. J Cardiovasc Pharmacol. 2005 Oct;46(4):445-451.
Walenta S, Wetterling M, Lehrke M, Schwickert G, Sundfør K, Rofstad EK, Mueller-Klieser W. High lactate levels predict likelihood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers. Cancer Res. 2000 Feb 15;60(4):916-921.
Walenta S, Mueller-Klieser WF. Lactate: mirror and motor of tumor malignancy. Semin Radiat Oncol. 2004 Jul;14(3):267-274.
Wallace DC. Mitochondria and cancer: Warburg addressed. Cold Spring Harb Symp Quant Biol. 2005;70:363-374.
Wallace D. Mitochondria and cancer. Nat Rev Cancer. 2012;12:685–698.
Wang SY, Yu QJ, Bao JK, Liu B. Polygonatum cyrtonema lectin, a potential antineoplastic drug targeting programmed cell death pathways. Biochem Biophys Res Commun. 2011 Mar 25;406(4):497-500.
Wang H, Fowler MI, Messenger DJ, Terry LA, Gu X, Zhou L, Liu R, Su J, Shi S, Ordaz-Ortiz JJ, Lian G, Berry MJ, Wang S. Homoisoflavonoids Are Potent Glucose Transporter 2 (GLUT2) Inhibitors: A Potential Mechanism for the Glucose-Lowering Properties of Polygonatum odoratum. J Agric Food Chem. 2018 Mar 28;66(12):3137-3145.
Warburg O. On the origin of cancer cells. Science. 1956 Feb 24;123(3191):309-314.
Wenzel U, Daniel H. Early and late apoptosis events in human transformed and non-transformed colonocytes are independent on intracellular acidification. Cell Physiol Biochem. 2004;14(1-2):65-76.
Wenzel U, Nickel A, Daniel H. Increased carnitine-dependent fatty acid uptake into mitochondria of human colon cancer cells induces apoptosis. J Nutr. 2005 Jun;135(6):1510-1514.
Winters N, Higgins K, Turner K. The Metabolic Approach to Cancer: Integrating Deep Nutrition, The Ketogenic Diet and Non-Toxic Bio-Individualized Therapies. Chelsea Green. Vermont, USA. 2017.
Yeung SJ, Pan J, Lee MH. Roles of p53, MYC and HIF-1 in regulating glycolysis – the seventh hallmark of cancer. Cell Mol Life Sci. 2008 Dec;65(24):3981-3999.
Zhang H, Du X, Sun TT, Wang CL, Li Y, Wu SZ. Lectin PCL inhibits the Warburg effect of PC3 cells by combining with EGFR and inhibiting HK2. Oncol Rep. 2017 Mar;37(3):1765-1771.
Zhang XM, Chen J, Xia YG, Xu Q. Apoptosis of murine melanoma B16-BL6 cells induced by quercetin targeting mitochondria, inhibiting expression of PKC-alpha and translocating PKC-delta. Cancer Chemother Pharmacol. 2005 Mar;55(3):251-262.