Author: Thomas Seyfried
Topics: Nutrition, physiology, fasting, ketogenic diet cancer, mitochondria
All information is attributed to the author. Except in the case where we may have misunderstood a concept and summarized it incorrectly. These notes are only for reference, and we always suggest reading from the source.
Preface
1. Images of Cancer
2. Confusion Surrounds the Origin of Cancer
3. Cancer Models
4. Energetics of Normal Cells and Cancer Cells
5. Respiratory Dysfunction in Cancer Cells
6. The Warburg Dispute
7. Is Respiration Normal in Cancer Cells?
8. Is Mitochondrial Glutamine Fermentation a Missing Link in the Metabolic Theory of Cancer?
9. Genes, Respiration, Viruses, and Cancer
10. Respiratory Insufficiency, the Retrograde Response, and the Origin of Cancer
11. Mitochondria: The Ultimate Tumor Suppressor
12. Abnormalities in Growth Control, Telomerase Activity, Apoptosis, and Angiogenesis Linked to Mitochondrial Dysfunction
13. Metastasis
14. Mitochondrial Respiratory Dysfunction and the Extrachromosomal Origin of Cancer
15. Nothing in Cancer Biology Makes Sense Except in the Light of Evolution
16. Cancer Treatment Strategies
17. Metabolic Management of Cancer
18. Patient Implementation of Metabolic Therapies for Cancer Management
19. Cancer Prevention
20. Case Studies and Personal Experiences in Using the Ketogenic Diet for Cancer Management
21. Conclusions
Abnormal cell growth (neoplasia) is the biological endpoint of the disease. Tumor cell invasion of surrounding tissues and spread to distant organs is the primary cause of morbidity and mortality in most cancer patients. This phenomenon is referred to as metastasis.
How Cancer Is Viewed
While cataloging genetic cancer defects is interesting, the defects often vary from one neoplastic cell to another within the same tumor.
Akt (v-Akt murine thymoma viral oncogene) or PKB (protein kinase-B) is a serine/threonine kinase that is involved in mediating various biological responses, such as inhibition of programmed cell death (apoptosis), stimulation of cell proliferation, and enhancement of tumor energy metabolism. Akt expression is generally greater in cancer cells than in normal cells. Although targeting Akt-related pathways is part of cancer drug development, the restriction of calorie intake will reduce Akt expression in tumors.
Angiogenesis involves the production of new blood vessels from existing blood vessels and involves interactions among numerous signaling molecules. Cancer therapies that target angiogenesis are thought to help manage the disease. Besides antiangiogenic cancer drugs such as bevacizumab (Avastin), simple calorie restriction effectively targets angiogenesis in tumors.
The following is a list of the mortality rate of different cancers:
Cancer genome projects are commendable for their technical achievement and have advanced the field of molecular biology, but have done little to defeat cancer. Dr. Linda Chin mentioned in her plenary lecture that improved genomic sequencing speed was a significant beneficiary of the cancer genome projects. Another benefit has been the increased number of jobs created in the biotechnology sector due to genome projects.
While gene-based targeted therapies could be effective against those few cancers that are inherited, and where all cells within the tumor have a common genetic defect, most cancers are not inherited through the germ line and few cancer cells have gene defects that are expressed in all cells of the tumor. Although almost 700 targeted therapies have been developed from the cancer genome projects, no patients with solid tumor have been cured from this strategy.
Most genetic defects found in tumors are “red herrings” that have diverted attention away from mitochondrial respiratory insufficiency, the central feature of the disease.
Hallmarks of Cancer
The Warburg Theory
Aerobic fermentation (the Warburg effect) is also a metabolic hallmark of most tumors whether they are solid or blood born. Aerobic fermentation involves elevated glucose uptake with lactic acid production in the presence of oxygen. Elevated glucose uptake and lactic acid production is a defining characteristic of most tumors and is the basis for tumor imaging using labeled glucose analogs.
Nearly all cancers express elevated fermentation, regardless of their tissue or cellular origin.
An increased dependency on energy through glucose fermentation (glycolysis) was viewed as an essential compensatory mechanism of energy production for cell viability following damage to respiration. If cells lose their ability to derive energy through respiration, then an alternative source of energy becomes essential for survival. Although aerobic glycolysis in cancer cells and anaerobic glycolysis in normal cells are similar in that lactate is produced under both situations, anaerobic glycolysis in normal cells arises from the absence of oxygen, whereas aerobic glycolysis in tumor cells arises as a consequence of both absence of oxygen and respiratory insufficiency.
The author and his colleagues expanded Warburg’s concept to include energy derived through amino acid fermentation and substrate level phosphorylation in the citric acid cycle, also known as the tricarboxylic acid (TCA) cycle. In other words, respiratory insufficiency leads to a dependency on nonoxidative phosphorylation for energy and survival. Substrate level phosphorylation, arising from respiratory insufficiency, is the single most common phenotype found in cancer regardless of tissue origin.
Respiratory insufficiency can arise from the cumulative effects of any number of environmental or genetic factors that alter mitochondrial function.
Critics argued that Warburg’s hypothesis on the origin of cancer did not address the role of tumor-associated mutations, the phenomenon of metastasis, nor did it link the molecular mechanisms of uncontrolled cell growth directly to impaired respiration.
Reassessment
It is widely assumed that the Warburg effect and the metabolic defects expressed in cancer cells arise primarily from genomic mutability selected during tumor progression. In other words, the abnormal energy metabolism in cancer arises as a secondary consequence of defects in oncogenes and tumor-suppressor genes. Emerging evidence, however, questions the genetic origin of cancer and suggests that cancer is primarily a metabolic disease as Warburg originally described.
Once established, genome instability contributes to further respiratory impairment, genome mutability, and tumor progression.
Seyfried contends that most of the gene defects in natural cancers arise as downstream effects of damaged mitochondrial function. His hypothesis is based on evidence that nuclear genome integrity is largely dependent on the cell having sufficient mitochondrial respiration, and that all cells require regulated energy homeostasis to maintain their differentiated state. While Warburg recognized the centrality of impaired respiration in the origin of cancer, his research did not explain how impaired mitochondrial function was connected to what is now recognized as the hallmarks of cancer. Moreover, he did not clearly describe how cancer cells appear to respire normally but have defective mitochondrial respiration.
Metastatic Models
Few good animal models exist for systemic metastasis. This is unfortunate, as systemic metastasis is the single-most serious aspect of the disease.
Once tumor cells leave their primary site and begin to show up in distant organs or tissues, effective management and long-term patient prognosis become uncertain. Most available cancer models, however, rarely show systemic metastatic behavior. Most tumor cells will rapidly grow when implanted under the skin (subcutaneously) or in an orthotopic site (tissue of origin) but will rarely show distal invasion or spread to multiple organ systems as is often seen in human disease.
Not every mouse inoculated with tumor cells develops metastatic cancer. Also, the time to systemic metastasis can vary significantly from one mouse to another. Models expressing these shortcomings are of limited value for the evaluation of new antimetastatic therapies.
Xenograft Models
Xenograft models involve growth of human tumor cells in nude mice or some other mice with a compromised innate and/or adaptive immune system. It is not possible to grow human tumors in mice that have normal T- and B-cell immunity because of antibody production and host tumor rejection. In addition, functional innate immunity derived from natural killer cells (NK), complement, etc., may contribute to tumor–host interactions. The normal mouse immune system will destroy implanted human cells. Most knowledgeable investigators in the cancer field know that xenograft models are not representative of the real-world situation.
A sharp border is seen between the tumor tissue and normal tissue in the noninvasive tumors, whereas no clear border is seen between the tumor tissue and the normal tissue in the metastatic cancers. Many xenograft models used in the cancer field are locally invasive, but rarely show systemic metastasis as seen in most human metastatic cancers.
Seyfried showed that human U87MG brain cancer cells express mouse carbohydrates on their surface when grown as a xenograft in immune-deficient mice.
More than 65% of the sialic acid composition on the U87 tumor cells consisted of the nine-carbon sugar, N-glycolylneuraminic acid. Humans, however, are unable to synthesize N-glycolylneuraminic acid because of mutation in the gene that encodes a common mammalian hydroxylase enzyme. The acquisition of mouse carbohydrates and lipids will likely occur in any human tumor grown in the body of a mouse or rat. N-Glycolylneuraminic acid also alters the characteristics of human embryonic stem cells when grown on nonhuman feeder cells. This has been a confounding variable in the stem cell field.
Many investigators believe that xenograft models are more representative of human disease than natural animal models of cancer simply because the tumor cells are derived from humans. Consequently, many cancer researchers use xenografts to demonstrate therapeutic effects. Many clinical drug trials have been initiated in patients on the basis of information generated from xenograft models. Many of these drugs are later discontinued because of lack of efficacy, unacceptable toxicity, or some combination of these.
Cell Culture Models
Migration of cancer cells through artificial extracellular matrix materials such as Matrigel or into scratches made on the surface of culture dishes has often been used to assess the invasive behavior of various cancer cells. Only a few studies compare and contrast the invasive and metastatic behavior of tumor cells in the artificial culture environment with their invasive behavior in the natural environment. The farther the model system is from the “real-world” situation, the more caution is required in relating the observations to what actually takes place in the human body.
The blood–brain barrier evolved over millions of years to prevent molecules in the serum from entering the brain. Astrocytes protect neurons from serum molecules and become quite reactive when exposed to serum. Yet, many investigators have studied the behavior of neural tumor cells cultured in growth media containing fetal cow serum.
Brain cancer cells expressing SDH1 mutations and other well-recognized phenotypes such as EGFR gene amplification in the in vivo environment could not be observed in cells cultured from the tumors. Indeed, tumor cells with SDH1 mutations that grow rapidly in vivo do not grow or survive in vitro.
Natural Models
The best cancer models, in his opinion, are those that arise naturally (spontaneously) and are grown orthotopically in their syngeneic hosts.
The spontaneous brain tumors in the inbred VM mouse strain represent a more natural model of metastatic cancer than any xenograft model. The tumor cells arising in the VM mouse strain manifest the full spectrum of growth characteristics seen in most human metastatic cancers. The VM model can be classified as a natural spontaneous model according to the criteria of Kerbel and coworkers. The metastatic VM tumors also share several features in common with the fusion hybrid metastatic mouse cancers described by Kerbel.
A good cancer model is one where the metastatic and invasive behavior of tumor cells is similar to that seen in human disease. A good model of metastasis should be one where cancer cells will invade locally from any implanted tissue site and readily spread to multiple organ systems within a short period of time (2–4 weeks). This is seen in the VM model.
Animal Charges as a Major Impediment to Cancer Research
The high cost of animal-maintenance charges (cage charges) is having a major negative impact on animal cancer research. It is becoming too expensive for many investigators to include animals in their research designs. In the past, animal charges were covered as part of the overhead costs on extramural grants. Currently, the animal charges are added to research grants as a “direct cost” line item. As institutions can charge “overhead” costs on direct cost items, animal charges have now become a convenient means of enhancing institutional revenue. In other words, animal charges have become a “cash cow” for university administrators.
The activities of the animal rights movement have led to excessive federal regulations that now impede animal research. For example, only five mice weighing less than 25 g can be housed in a standard mouse cage. How might these regulations relate to the natural housing of mice in the wild?
Problems with Tumor Histological Classification
Too often investigators will focus more on tumor cell classification than on the biological behavior of the tumor cells. The success or failure in adapting a new cancer model can sometimes depend on how the tumor is classified histologically. This is especially the case in the brain cancer field, where neuropathology has a dominant influence on the direction of research.
Even though there may be striking differences in appearance, growth rate, and biochemistry of certain tumors, they may develop a similar histological appearance. It is because of this those neuropathologists may have disagreements about the identification of a tumor sample.
According to Dr. Sanford Palay, the abnormalities in cytoarchitecture made tumor cell identification ambiguous at best. Diagnosis of most cancer types rests primarily on the subjective impressions of pathologists. While information on brain tumor classification might provide some insight into the tumor origin, it is not clear how tumor cell classification will influence therapy.
Good cancer models should be evaluated more for their in vivo growth behaviors than for their histological classification.
Personal Perspective on Cancer
His perspective is similar to that of Otto Warburg, who originally proposed that all cancer is a disease of respiration. Moreover, they found that many metastatic cancers share multiple properties with cells of myeloid origin. These are cells of the immune system such as macrophages and leukocytes. Macrophages and leukocytes are already mesenchymal cells genetically programmed to enter and exit tissues and to survive in hypoxic environments. These are hallmarks of most metastatic tumor cells. It is not necessary to view cancer as a complicated cybernetic system.
In order for cells to remain viable and to perform their genetically programmed functions, they must produce energy. Most of this energy is commonly stored in the terminal γ and β phosphates of adenosine triphosphate (ATP) and is released during the hydrolysis of their phosphoanhydride bonds. This energy is generally referred to as the free energy of activation or ATP hydrolysis. The standard energy of ATP hydrolysis under physiological conditions is known as DeltaG’ATP and is tightly regulated in all cells between −53 and −60 kJ/mol. G is the Gibbs free energy, Delta is the difference between two energy states, and prime represents the activated state.
Metabolic Homeostasis
Metabolic homeostasis within cells is dependent to a large extent on the energy supply to the membrane pumps. Hormones, such as insulin and glucagon, can regulate global system energy homeostasis in order to maintain steady energy balance within the cells of each organ. If the energy to the cellular pumps is interrupted, the cell begins to swell. Swelling results from increased Na+ and Ca2+ concentrations and decreased K+ concentration. Because the inside of the cell is more negative than the outside, Na+ and Ca2+ will naturally move down their concentration gradient from outside to inside. On the other hand, K+, which is more concentrated inside than outside, will flow down its concentration gradient. Most cell functions are linked either directly or indirectly to the plasma membrane potential and to the Na+/K+/Ca2+ gradients. Ready availability of ATP to the pumps maintains these ionic gradients. Global cellular dysfunction and ultimately organ and systems failure will arise if energy flow to the pumps is disrupted.
There are several sources of ATP synthesis that can be used to maintain membrane potential. The mitochondria produce most of the energy in normal mammalian cells.
Besides OxPhos, approximately 11% (4/36 total ATP molecules) of the total cellular energy is produced through substrate-level phosphorylation. Substrate-level phosphorylation involves the transfer of a free phosphate to ADP from a metabolic substrate to form ATP.
Two major metabolic pathways can produce ATP through substrate-level phosphorylation in mammalian cells and tissues:
Under normal physiological conditions, two ATP molecules are produced from glycolysis in the cytoplasm and two from the succinyl-CoA synthetase reaction in the mitochondrial matrix. In contrast to OxPhos, which involves oxygen and a membrane-regulated proton gradient, oxygen is not a requirement for ATP synthesis through substrate-level phosphorylation. A proton motive gradient could still operate, however, through a reverse action of the F1F0-ATPase. Stepien and colleagues have shown how the mitochondrial attached hexokinase II isoform provides glycolytic ATP to the mitochondria in order to maintain the proton motive gradient. This is important because it explains, in part, how tumor cells can produce energy and remain viable in hypoxia despite damage to mitochondrial structure and function.
The number of ATP molecules produced from TCA cycle, substrate-level phosphorylation would need to increase if OxPhos were insufficient to maintain energy homeostasis. This would be similar to the increase in the number of ATP molecules produced through glycolysis when OxPhos is reduced. Nonoxidative energy production through amino acid fermentation and substrate-level phosphorylation has been documented in developing mammalian embryos, in diving animals, and in heart and kidney tissue under hypoxia.
The Constancy of the DeltaG’ATP (GATP)
Veech and coworkers showed that the GATP of cells was empirically formalized and was measurable through the energies of ion distributions via the sodium pump and its linked transporters. The Gibbs–Donnan equilibrium describes the flow of ions across semipermeable membranes and is estimated using the Nernst equation. The Nernst equation can link the Gibbs free energy to the electric charge across a membrane.
A remarkable finding was the similarity of the GATP among cells with widely differing resting membrane potentials and mechanisms of energy production. For example, the GATP in heart, liver, and erythrocytes was approximately –56 kJ/mol despite having very different electrical potentials of –86, –56, and –6 mV.
Moreover, energy production in the heart and liver, which contain many mitochondria, is largely through OxPhos, whereas energy production in the erythrocyte, which contains no nucleus or mitochondria, is entirely through glycolysis. Despite the profound differences in resting membrane potentials and in mechanisms of energy production among these disparate cell types, they all express a similar free energy of ATP hydrolysis. These observations suggest that the balance of energy consumption and production is independent of the energy source and the amount of the total ATP produced.
The constancy of the GATP of approximately–56 kJ/mol is fundamental to cellular energy homeostasis and its relationship to cancer cell energy metabolism is critical. The maintenance of the GATP is the end point of both genetic and metabolic processes and any disturbance in this energy balance will compromise cell function and viability.
Cells can die from either too little or too much energy. Too little energy leads to cell death by either necrotic or apoptotic mechanisms. Overproduction of ATP, a polyanionic Donnan active material, disrupts the Gibbs–Donnan equilibrium, alters the function of membrane pumps, and inhibits respiration and viability. To maintain cellular energy balance, the mitochondrial F0F1-ATPase can sometimes run in reverse (hydrolyzing ATP). Additionally, some tumor cells release ATP into the extracellular milieu through the action of the p-glycoprotein, which is linked to glycolysis and is often overexpressed in tumors.
If OxPhos becomes compromised, energy production through substrate-level phosphorylation must be increased in order to maintain a stable free energy of ATP hydrolysis and cell viability. Alternatively, energy expenditure can be reduced to offset reduced energy production. Acute damage to respiratory function usually causes apoptotic or necrotic cell death due to membrane pump energy depletion. However, energy through substrate-level phosphorylation can gradually compensate for minor damage to OxPhos-derived energy over protracted periods. As tumors rarely occur following acute injury to respiration, considerable time is required for nonoxidative energy metabolism to displace OxPhos as the dominant energy generator in the cell.
Prolonged reliance on substrate-level phosphorylation for energy production in previously normally respiring cells produces genome instability, disorder, and increased proliferation, that is, the hallmarks of cancer. Entropy refers to the degree of disorder in systems and is the foundation of the second law of thermodynamics. Szent-Gyorgyi described cancer as a state of increased entropy, where randomness and disorder predominate. Protracted OxPhos insufficiency coupled with persistent compensatory fermentation increases entropy. Cells that do not increase fermentation energy to compensate for insufficient OxPhos simply die off and never become neoplastic. Adaptation to fermentation allows a cell to bypass mitochondrial-induced senescence. Cancer arises in those cells that bypass mitochondrial-induced senescence.
ATP Production in Normal Cells and Tumor Cells
Warburg considered the ascites cancer cells a better preparation than tumor tissue slices, as the ascites cells are not contaminated with nonneoplastic stromal cells that are present to various degrees in the tumor tissue slices. Stromal cells are expected to have normal metabolism and might therefore dilute the magnitude of metabolic deficiency in the neoplastic cells of the tumor. This can be problematic as they found that stromal cells in the form of tumor-associated macrophages (TAMs) can contribute significantly to the total cell population of some tumors.
Warburg evaluated the ascites cells maintained in a medium, which was supplemented only with glucose and bicarbonate. Later studies showed, however, that normal cell respiration dramatically increased in pure serum, whereas cancer cell respiration increased only slightly. The slight respiratory increase by the cancer cells most likely reflects the upper limits of their respiratory capacity. Under physiological conditions of pH and temperature, Warburg expressed his data on energy metabolism in the ascites cells as metabolic quotients (Q).
Donnelly and Scheffler who showed that the total ATP production is similar in respiration-deficient and in respiration-normal Chinese hamster fibroblasts. The respiration deficiency in these cells involved a defect in the NADH-coenzyme Q reductase. This defect significantly reduces the TCA cycle function and oxygen consumption.
Energy Production Through Glucose Fermentation
Warburg considered fermentation as the formation of lactate from glucose in the absence of oxygen. This type of energy is also produced in mammalian embryos and in our muscles during strenuous exercise. Instead of entering the TCA cycle for complete oxidation, pyruvate is reduced to lactate when oxygen levels are low. Lactate fermentation generates NAD+ as an oxidizing agent for glycolysis. The NAD+ can be used as an electron acceptor during the oxidation of dihydroxyacetone phosphate to 1,3-diphosphoglycerate, the reaction preceding the first substrate-level phosphorylation in glycolysis. Failure to regenerate cytoplasmic NAD+ reduces energy through glycolysis, which could compromise cell viability in the absence of energy through OxPhos or TCA cycle, substrate-level phosphorylation.
Lactate is basically metabolic waste from the incomplete oxidation of glucose and must be removed from the microenvironment as quickly as possible. Most lactate enters the blood stream where it is used to synthesize glucose in the liver through what is known as the Cori cycle. Lactate is simply excreted into the medium in cultured cells that are grown in glucose. This usually changes the color of the pH indicator dye (phenol red) from red to yellow. Once oxygen becomes available, glucose utilization and lactate production decrease due to the Pasteur effect.
Warburg was unaware of possible mitochondrial amino acid fermentation in tumor cells. While O2 exposure of tumor cells can decrease lactate production to some extent, lactate production from glucose is generally higher in tumor cells than in their normal cellular counterparts. Mitochondrial amino acid fermentation provides a possible missing metabolic link in Warburg’s theory. Mitochondrial amino acid fermentation obscures the boundaries between normal respiration and fermentation and can explain much of the controversy surrounding the Warburg theory.
Lactate accumulates as an end product of glucose fermentation. If OxPhos were normal in cancer cells, then lactate production would decrease in the presence of O2, as pyruvate would be effectively oxidized through the TCA cycle and would no longer be available for the lactic acid dehydrogenase (LDH) reaction. In contrast to normal cells, tumor cells continue to ferment glucose in the presence of oxygen. Cancer cells that rely more on glutamine than on glucose for energy production can produce ATP through nonoxidative processes in the mitochondria.
Pseudo respiration has all the characteristics of respiration, but does not involve ATP synthesis through OxPhos. Seyfried proposes that this apparent respiratory energy is derived from amino acid fermentation. Just as tumor cells ferment glucose in the presence of O2, some tumor cells also ferment glutamine and possibly other amino acids in the presence of elevated glucose and O2. Glucose and glutamine interact synergistically to drive tumor cell fermentation.
Unlike most normal mammalian cells, which balance energy production to energy output, energy balance is dysregulated in cancer cells because they do not suppress ATP turnover under anoxia. Rather, tumor cells seem to enhance ATP turnover under hypoxia.
Allen and Attwell showed that lactate was unable to replace glucose as a metabolic fuel for brain cells under normoxia or hypoxia. Lactate can, however, be metabolized to glucose through the Cori cycle, which can then be used to fuel tumor cell growth.
Mitochondrial membrane lipids are altered and energy production through OxPhos is compromised from simply growing dividing cells in culture. Unfortunately, many investigators fail to account for mitochondrial ATP production through amino acid fermentation and substrate-level phosphorylation in the mitochondria especially when high glucose levels are present. Consequently, monitoring extracellular pH as a marker for lactic acid production could be misleading especially if the cells are metabolizing glutamine and producing ammonia.
Glutaminolysis with or without Lactate Production
Glutamine can serve as a major source of metabolic fuel for generating ATP through TCA cycle, substrate-level phosphorylation when OxPhos is deficient. Glutamine is also anapleurotic in replenishing metabolites for the TCA cycle.
Glutamine is also a major energy fuel for cells of the immune system. As myeloid cells can be the origin of many metastatic cancers following fusion hybridizations, glutamine becomes an important fuel for driving metastasis. Indeed, targeting glutamine can significantly inhibit systemic metastasis.
McKeehan has first described glutaminolysis as the process by which glutamine metabolism produces carbon dioxide, pyruvate, and lactate through oxidative pathways. Under this scheme, malate would leave the mitochondria where it would be metabolized to pyruvate and then to lactate. McKeehan has not explained how malate would leave the mitochondria. Malate usually enters the mitochondria through the malate–aspartate shuttle, which is active in cancer cells.
The issue of lactate production from glutamine remains unsettled in tumor cells. Why is it important to know whether glutaminolysis is involved in cancer energy metabolism and whether glutamine carbons are found in lactate? This information can provide insight on the fate of tumor energy metabolites that can be used for energy and growth. Moreover, it will be important to determine if the metabolic changes in tumor cells are the cause or the consequence of the genetic changes that occur in the tumor cells. It is therefore important that we know the metabolic origin of lactate production in tumor cells.
Transamination Reactions
Glutamine enters the mitochondria where it is rapidly metabolized to glutamate by mitochondrial glutaminase. Glutamate is then metabolized to α-ketoglutarate through either a transamination reaction with aspartate or alanine as products or through the action of glutamate dehydrogenase.
In the glutamate dehydrogenase reaction, NH3 becomes a toxic by-product that must be eliminated. In the transamination reactions, on the other hand, OAA accepts the NH3 group to form either aspartate or alanine. Whether aspartate or alanine becomes the primary product of the transamination reaction depends upon the presence or absence of oxygen and malate, and whether or not respiration is sufficient or insufficient.
Seyfried considers that the transamination reactions would predominate over the glutamate dehydrogenase reaction in tumor cell mitochondria since the guanosine triphosphate (GTP), and also the ATP, formed through TCA cycle, substrate-level phosphorylation could inhibit the glutamate dehydrogenase reaction, thus reducing its activity.
Epigallocatechin gallate (EGCG), which inhibits the glutamate dehydrogenase, could be used to help determine if the α-ketoglutarate arises through the action of glutamate dehydrogenase or a transamination reaction. Some of the aspartate formed through transamination could also be used for malate production in the cytosol. This will depend to a large extent on the activity of the malate aspartate shuttle. The activity of the shuttle is correlated with glycolytic activity, that is, the greater the glycolysis, the greater is the shuttle activity.
TCA Cycle, Substrate-Level Phosphorylation
The α-ketoglutarate formed from glutamine enters the TCA cycle where it is decarboxylated and conjugated with coenzyme A to form succinyl-CoA. The α-ketoglutarate dehydrogenase catalyzes this reaction with the formation of NADH and CO2. The Succinyl-CoA formed is then oxidized to succinate. A histidine residue in the enzyme becomes phosphorylated. It is during this reaction that the histidine phosphate is transferred from the enzyme itself (succinyl-CoA synthetase) to either GDP or ADP to form GTP or ATP.
As NADH in the cytoplasm cannot directly enter the mitochondria due to impermeability and the lack of a mitochondrial membrane transporter, the malate–aspartate shuttle and the glycerol 3-phosphate shuttle are used to indirectly transport reducing equivalents from NADH in the cytoplasm to the mitochondria. The malate–aspartate shuttle normally delivers reducing equivalents from NADH in the cytoplasm to complex I of the electron transport chain (ETC). Consequently, the NADH is used to reduce OAA to malate in the cytoplasm. The cytoplasmic malate enters the mitochondria through the malate–aspartate shuttle where it is oxidized to OAA with the production of NAD+. Succinate is a major end product of anaerobic amino acid catabolism with alanine produced as a minor end product.
Tomitsuka and colleagues provide evidence that tumor cells express an active fumarate reductase reaction.
Cholesterol Synthesis and Hypoxia
Cholesterol is a major membrane lipid that must be synthesized for cancer cells to grow. Oxygen is required for the squalene monooxygenase reaction of cholesterol synthesis. They found that cultured metastatic VM-M3 tumor cells grow well with glucose and glutamine as the only metabolic fuels in normoxia without serum. These cells actively synthesize cholesterol from either glucose or glutamine. However, the cells die rapidly under hypoxia without added serum. It appears that serum is required for growth under hypoxia, but is not required for growth under normoxia. Serum contains, among many factors, high levels of cholesterol.
It is not necessary for the cells to synthesize cholesterol if they can get it free from the growth environment. Hence, the VM-M3 cells can grow in hypoxia as long as they have fermentable fuels and can obtain cholesterol from an external source.
Summary
All cells including tumor cells require a relatively constant level of usable ATP synthesis for maintaining viability. This appears to be a biological constant independent of cell origin or function. Energy metabolism in cancer cells differs markedly from that in normal cells. In contrast to normal cells, which generate most of their useable energy through OxPhos, cancer cells depend more heavily on fermentation reactions using nonoxidative, substrate-level phosphorylation for their ATP synthesis. Substrate-level phosphorylation occurs through glycolysis in the cytoplasm and through succinyl-CoA synthetase in the mitochondria.
Both glucose and glutamine can provide energy to cancer cells through substrate-level phosphorylation. Although tumor cell mitochondria might appear to respire, we refer to this as pseudo respiration since OxPhos is either reduced or absent. Tumor cells can grow in hypoxia as long as they have fermentable fuels and access to extracellular cholesterol. Respiration is the bioenergetic signature of normal cells; fermentation is the bioenergetic signature of cancer cells. Fermentation drives cancer cells whether or not oxygen is present.
OxPhos is the final stage of cellular respiration involving multiple coupled redox reactions where the energy contained in carbon–hydrogen bonds of food molecules is captured and conserved in the terminal phosphoanhydride bond of ATP. The process specifically involves: (i) the flow of electrons through a chain of membrane-bound carriers, (ii) the coupling of the downhill electron flow to an uphill transport of protons across a proton-impermeable membrane, thus conserving the free energy of fuel oxidation as a transmembrane electrochemical potential, and (iii) the synthesis of ATP from ADP+Pi through a membrane-bound enzymatic complex linked to the transmembrane flow of the protons down their concentration gradient.
Normal cells have structure, while most tumor cells are dysmorphic compared to normal cells. As the structural integrity of the mitochondria provides the energy needed to maintain cellular differentiation, it is necessary to consider the types of injuries that would reduce OxPhos in tumor cells.
Normal Mitochondria
The mitochondrion is bounded by two sets of membranes, a smooth outer membrane and an inner membrane that is arranged in folds, or cristae that extend into the interior matrix area of the organelle. The complexes of the electron transport chain (ETC), which contribute to energy through OxPhos are found in the mitochondrial cristae. The cristae are swollen cisterns or sacs, with multiple narrow tubular connections to the peripheral surface of the inner membrane, and to each other.
OxPhos requires the transport of electrons through proteins imbedded in the cristae. Mitochondria contain many enzymes within the matrix involved with activities including the citric acid and fatty acid cycles as well as calcium flux. Mitochondria also regulates intracellular calcium that can have global effects on numerous aspects of cell physiology. Mitochondria are self-replicating and contain their own DNA, RNA polymerase, transfer RNA, and ribosomes. Mitochondria are also dynamic organelles that can expand and contract and undergo fission and fusions in response to the metabolic state of the cell.
Morphological Defects in Tumor Cell Mitochondria
Numerous studies on patient and animal cancers show that tumor mitochondria differ from normal mitochondria in number, size, and shape. Pedersen summarized data from over 20 studies showing that the total number of mitochondria in tumor cells was significantly lower than the number in normal cells of origin. He also mentioned that the total respiratory capacity of tumor mitochondria was lower relative to that of normal cells. Carew and Huang also reviewed evidence suggesting that abnormalities in mitochondrial DNA could compromise mitochondrial function in tumor cells.
Bayley and Devilee showed aerobic glycolysis (Warburg effect) could be directly linked to mitochondrial respiratory injury in those tumors arising from inherited mutations in the genes for succinate dehydrogenases and fumarate hydratase. Besides these defects, any abnormalities in the number of mitochondria, in their ultrastructure and morphology, and in their response to changes in growth environment would predict some degree of respiratory dysfunction.
Proteomic Abnormalities in Tumor Cell Mitochondria
Roskelley and coworkers showed that cytochrome–oxidase activity was deficient in nearly all types of highly malignant cancers examined. The human malignancies included cancers of rectum, colon, kidney, breast, brain, prostate, stomach, skin, and testis. The same biochemical deficiencies were also found in well-established transplantable and induced tumors of rat mouse and rabbit. Moreover, carcinogenesis, both by a chemical agent and by a virus, produced the same energy defects in animal models. Their data from a variety of human and animal cancers clearly showed that all of the normal adult tissues displayed a high oxidative response, whereas all of the frankly malignant cancers displayed a poor oxidative response. Pedersen later provided a comprehensive review documenting the numerous protein defects in mitochondria from tumor cells.
Cuezva and colleagues also provide proteomic evidence for respiratory dysfunction in cancer. These investigators evaluated the relationship of glyceraldehyde-3-phosphate dehydrogenase (GAPHD) and the β-F1 ATPase in a broad spectrum of tumors including breast, colon, lung, and esophagus. GAPHD and the β-F1 ATPase are key enzymes needed to drive glycolysis and OxPhos, respectively. GAPHD consumes NAD+ and inorganic phosphate, Pi, to synthesize the energy-rich intermediate, 1,3-bisglycerophosphate with NADH + H+ as by-products. Elevated GAPHD activity indicates enhanced energy production through glycolysis. The β-subunit of the F1 ATPase is required for ATP synthesis through OxPhos.
Simonnet and colleagues have shown that respiratory impairment was significantly greater in patients with clear cell or high-grade renal tumors than in patients with low grade or benign renal tumors. Moreover, the respiratory impairment in these renal tumors was correlated with significant decreases in the content of ETC complexes II, III, and IV as well as with abnormal assembly of the complex V (the F1F0 ATPase).
Unwin and coworkers used a proteomic approach, based on two-dimensional gel electrophoresis and mass spectrometry, to compare the protein profiles of renal carcinoma tissue with tissue from patient matched normal kidney cortex. The most striking findings from their study were the decreased expression of several mitochondrial enzymes implicated in OxPhos and the increased expression of enzymes for glycolysis. The increased expression of the glycolytic enzymes was also associated with a parallel decrease in three of the enzymes catalyzing the reverse reactions of gluconeogenesis.
Lipidomic Abnormalities in Tumor Cell Mitochondria
The lipidome refers to the total content and composition of all lipids in a cell or cell organelle. Lipids maintain the integrity of biomembranes. Abnormalities in lipids can compromise mitochondrial function. The functions of ETC proteins are dependent to a considerable degree on the lipid composition of the inner mitochondrial membrane. Lipid abnormalities in the inner mitochondrial membrane will therefore alter OxPhos capabilities.
Cholesterol is a relatively minor lipid of the inner mitochondrial membrane of normal cells. Feo and colleagues have previously shown that the cholesterol/phospholipid ratio was significantly higher in mitochondria from hepatomas than in mitochondria from normal liver cells. As cholesterol reduces membrane fluidity, elevated levels of cholesterol would be expected to reduce the fluidity properties of mitochondrial membranes. In contrast to mitochondrial phospholipids in normal tissues, which contain an abundance of long-chain polyunsaturated fatty acids, phospholipids in tumor mitochondria are enriched in short-chain saturated or monounsaturated species.
Most importantly, they have found several abnormalities in the structure of cardiolipin (CL), the major lipid of the inner mitochondrial membrane.
Cardiolipin (CL): A Mitochondrial-Specific Lipid
CL (1,3-diphosphatidyl-sn-glycerol) is a complex, mitochondrial-specific phospholipid that regulates numerous enzyme activities, especially those related to OxPhos and coupled respiration. Several studies have shown that CL is essential for efficient oxidative energy production and mitochondrial function. CL is necessary for maintaining coupled mitochondria, and defects in CL can produce protein independent uncoupling. Hence, alterations in the content or composition of CL will alter cellular respiration.
CL contains two phosphate head groups, three glycerol moieties, and four fatty acyl chains and is primarily enriched in the inner mitochondrial membrane. Enrichment in the inner mitochondrial membrane makes CL a pivotal molecule for regulating cristae structure and OxPhos. CL binds complex I, III, IV, and V and stabilizes the super complexes (I/III/IV, I/III, and III/IV), demonstrating an absolute requirement of CL for the catalytic activity of these respiratory enzyme complexes.
CL restricts pumped protons within its head group domain, thus providing the structural basis for mitochondrial membrane potential and for supplying protons to the ATP synthase.
Respiratory complex proteins that interact with CL form hydrophobic amino acid grooves on their surface. These grooves accommodate the fatty acid chains of CL. Since long-chain carbon molecules appeared earlier in evolution than membrane proteins, it is likely that the grooves evolved to accommodate already existing lipid fatty acids. While the amino acid sequence of electron transport proteins is highly conserved across species, considerable variability occurs for the fatty acid sequences of CL. Although the respiratory protein structure is largely invariant, the fatty acid composition of CL can be modulated through changes in nutrition and the physiological environment. They have found that hypoxia could significantly modify brain CL fatty acid composition in VM mice. CL can modulate ETC activities without altering the primary sequence of amino acids. Hence, changes in CL content and composition can influence electron transport and ultimately the efficiency of OxPhos.
The activity of respiratory enzymes in complex I and complex III and their linked activities are directly related to CL content. The activities of the respiratory enzyme complexes are also dependent on the composition of the CL molecular species. Importantly, the degree of CL unsaturation is related to states 1–3 of respiration.
Cardiolipin and Abnormal Energy Metabolism in Tumor Cells
The CL content was significantly lower in the mitochondria from the CT-2A and the EPEN tumors than in the mitochondria from the normal control B6 mouse brain. In contrast to the B6 mouse brain, which contains about 100 molecular fatty acid species of CL distributed symmetrically over seven major groups, the VM mouse brain is unique in having only about 45 major CL molecular species and is missing molecular species in groups IV, V, and VII.
The CL content was significantly lower in the mitochondria from the VM-NM1 and the VM-M2 tumors than in the mitochondria from the control VM mouse brain. They found that the CL abnormalities in these tumors were associated with significant reductions in ETC activities consistent with the pivotal role of CL in maintaining the structural integrity of the inner mitochondrial membrane.
Abnormalities in the content and composition of CL could underlie the abnormal energy metabolism of these diverse brain tumors. This is the type of connection that Weinhouse considered essential for establishing the credibility of the Warburg theory.
Their findings are also consistent with earlier studies in rat hepatomas that show an increase in shorter chain saturated fatty acid content (palmitic and stearic) characteristic of immature CL. Their studies are also consistent with more recent findings in rhabdomyosarcoma, a type of muscle tumor, which show that the reduction in complex I activity was associated with CL abnormalities. Continued expression of immature CL would reduce efficient respiratory energy production. In light of what we know about the CL structure and respiratory function, it is difficult to conceive how mitochondrial OxPhos could function normally in tumors that express CL abnormalities.
They proposed that CL abnormalities could arise from either inherited cancer risk factors as seen in the VM mice or from numerous epigenetic and environmental cancer risk factors including inflammation, viruses, hypoxia, radiation, and so on.
γ -radiation is known to induce free radical CL fragmentation, which would compromise respiratory function. On the basis of these and other observations, we suggested that most tumors, regardless of cell origin, would contain abnormalities in CL composition and/or content. Regardless of whether the CL abnormalities are related to the cause of the tumor or arise during tumor progression, the CL abnormalities will significantly reduce the efficiency of mitochondrial OxPhos.
Mitochondrial immaturity would predict insufficient energy production through OxPhos. The association of CL abnormalities with an impaired respiratory function would be expected on the basis of the localization and role of CL in ETC activities.
Complicating Influence of the In Vitro Growth Environment on Cardiolipin Composition and Energy Metabolism
CL composition of the cultured cells was composed largely of immature CL containing shorter chain saturated or monounsaturated species indicative of failed remodeling. These findings indicate that the in vitro growth environment produced abnormalities in CL remodeling. A failure to remodel CL reduces efficient energy production through OxPhos.
Abnormalities in CL can reduce ATP production through OxPhos. Abnormalities in CL can arise through the process of tumorigenesis and from the growth of mammalian cells in the in vitro environment. Caution should be used in comparing energy metabolism in nontumorigenic cells and tumorigenic cells grown in tissue culture environments that do not replicate the growth conditions of the in vivo environment.
Mitochondrial Uncoupling and Cancer
Uncoupling involves dissipation of the mitochondrial proton motive gradient. Uncoupling can produce heat rather than ATP. Mitochondrial uncoupling occurs during cold acclimation in mammals and is mediated, at least in part, by uncoupling proteins.
While reduced oxygen uptake can be indicative of reduced OxPhos, increased oxygen uptake may or may not be indicative of increased OxPhos and ATP production.
Cancer Cell Heat Production and Uncoupled Mitochondria
Mitochondria of brown adipose are naturally uncoupled so that oxidation of substrates in these cells produces heat rather than a proton motive gradient for ATP synthesis. As heat production is a characteristic of uncoupled mitochondria, it would be important to know if heat production is greater in more tumorigenic cells than in less tumorigenic cells.
The greater heat production in the less differentiated cells supports the hypothesis that mitochondrial uncoupling is greater in cancer cells that are more malignant than in those that are less malignant.
Lymphoma cell heat production was significantly greater in patients who died within two years of diagnosis than in patients who survived more than two years following diagnosis. Similar results were obtained in a thermal analysis of breast cancer in that prognosis was worse for patients with warmer tumors than for those with cooler tumors. Zhao and coworkers also used a thermocouple to show that tumor malignancy was positively correlated with tumor temperature.
As uncoupling leads to heat production, it is possible that the increased heat production in the more aggressive tumors is due to mitochondrial uncoupling. It is also interesting that heat production was correlated with increased glucose consumption and lactic acid production in human leukemia cells. This supports Warburg’s theory that aerobic fermentation compensates for insufficient respiration.
Evidence of respiratory injury would be obvious in the cancer cells that express reduced mitochondrial adenosine triphosphate (ATP) production in association with decreased oxygen consumption, since O2 is necessary for ATP synthesis through OxPhos. However, oxygen consumption is not reduced in some cancer cells. Indeed, O2 consumption increases with increased malignancy in some tumor cells. Warburg has attributed this phenomenon to defects in the coupling of respiration to ATP production. In other words, some cancer cells produce CO2 and consume O2, but produce insufficient energy through respiration.
Disruption of the proton motive gradient, with reduced efficiency of ATP production, would naturally require a compensatory mechanism of energy production to prevent cell death. Warburg emphasized that fermentation, involving the catabolism of glucose to lactic acid, was the compensatory mechanism responsible for energy production in cancer cells. Obviously, this process would need to increase substantially in tumor cells to compensate for the lost ATP production through OxPhos. If respiratory energy production is compromised, there “must” be some compensatory mechanism of energy production to maintain cell viability. Without compensation, the cell would die from energy failure.
Sidney Weinhouse’s Criticisms of the Warburg Theory
Weinhouse was troubled by findings showing that oxygen consumption and CO2 production was high in many cancerous tissues and cells. How could cancer cells consume oxygen and produce CO2 if their respiration were irreversibly damaged? He felt that many tumor cells could metabolize fatty acids for energy, thus producing CO2.
He concluded that, “the available evidence indicates to me that high glycolysis occurs, despite quantitatively and qualitatively normal occurrence of carbon and electron transport. This can mean only that glucose catabolism is so rapid in tumors that the normal channels for disposal of pyruvic acid are over-loaded”.
Alan Aisenberg’s Criticisms of the Warburg Theory
Despite reviewing and documenting numerous studies that supported Warburg’s theory, Aisenberg concluded that evidence supporting Warburg’s views did not exist and that the entire concept of respiratory injury as the origin of cancer must remain just a hypothesis.
Basically, the data not supporting Warburg’s theory were mostly obtained from experimental systems unrepresentative of conditions occurring in vivo. Warburg’s generalization that all tumors show a high rate of anaerobic glycolysis was strongly supported by the data presented.
Oxygen suppresses anaerobic glycolysis completely in most normal tissues. Oxygen, however, does not completely suppress aerobic glycolysis in tumor cells, which involves the continued expression of glycolysis in the presence of oxygen, that is, the Warburg effect.
The presence of normal cells in the tumor tissue could mask some of the metabolic abnormalities of the tumor cells. This is why the best comparisons should be between (i) tumor tissue and normal tissue from which the tumors are derived and (ii) normal cells and tumor cells derived from the same cell clone. The continued production of lactic acid in the presence of O2, indicative of an inefficient Pasteur effect, persists in tumor cells. In contrast to the effects of O2 in normal cells and tissues, O2 is unable to completely suppress glycolysis in tumor cells. The incomplete O2 suppression of glycolysis in tumor cells suggests that these cells have impaired respiration. On the other hand, some would argue that the persistent aerobic glycolysis in tumor cells results from damaged glycolysis regulation rather than damaged respiration itself.
Sidney Colowick’s Assessment of the Aisenberg Monograph
Colowick emphasized that, besides denying damaged respiration in tumors, Aisenberg had also attempted to refute Warburg’s theory using several arguments. First, that many carcinogens were not inhibitors of respiration.
Second, Aisenberg mentioned that the highest concentration of protein-bound carcinogen was present in cytoplasm, but admitted that mitochondria could take up carcinogenic dyes. In fact, Aisenberg cited Potter’s view that the specific lesion in tumors may be due to the deletion of mitochondria rather than alteration of soluble enzymes.
Third, Aisenberg doubted that X-ray induction of tumors should be attributed to damage of mitochondrial respiration, since X rays can damage nuclear morphology. It is not clear, however, why the X rays could not also damage mitochondria as Warburg and others had suggested. X rays can damage mitochondria.
Fourth, Aisenberg doubted the interpretation of Goldblatt and Cameron, who had shown that intermittent anoxia could transform normal fibroblasts into malignant cells. Just because malignancy could be produced in culture without anaerobiosis does not rule out the possibility that anaerobiosis, too, could cause malignancy.
Apples and Oranges
Many experiments compare respiration in one tumor cell with that in another tumor cell of different origin without also including nontumorigenic, cell specific controls. Other experiments compare respiration of cultured tumor cells with respiration of tumor tissue or with tissue slices.
Growth of cells in high glucose media deficient in respiratory nutrients will suppress normal respiration, potentially masking differences in respiration between cancer cells and normal cells.
The energy of respiration is associated with the intact structure of mitochondrial membranes and the differentiated state, whereas glycolysis is associated with reduced structure (mostly soluble enzymes in the cytoplasm) and the dedifferentiated state. Inferiority was therefore linked to the disorganized organelle structure. It is well documented that the mitochondria in most if not all cancer cells differ in structure from those present in normal cells.
Pseudo-Respiration
There is a plethora of papers in the scientific literature suggesting that cancer cells respire based on data showing that they eject H+, transport electrons, consume O2, release CO2, and produce ATP in mitochondria. Seyfried described earlier how O2 consumption can increase in cancer cells that express uncoupled OxPhos (electron transport is not coupled to ATP synthesis). Abnormalities in cardiolipin content and composition can induce protein independent uncoupling as mentioned in Chapter 5. Tumor cells contain abnormalities in cardiolipin.
Cells with greater tumorigenic potential consume more oxygen and yet exhibit less oxygen-dependent ATP synthesis than cells with lower tumorigenic potential. Ramanathan goes on to suggest that such cells might use the mitochondrial electron transport chain and oxidative phosphorylation for reasons other than ATP synthesis.
Cancer cells can synthesize ATP in their mitochondria through nonoxidative processes involving amino acid fermentation. This energy is generated through the action of succinyl-CoA synthetase and a reversal of the succinate dehydrogenase, where fumarate becomes the oxidizing agent. In other words, the cells consume O2 but do not produce ATP exclusively through OxPhos but rather also produce ATP through mitochondrial fermentation involving substrate-level phosphorylation.
Human tumor cells grown in mice acquire unique mouse lipids, which cannot be synthesized by the human cells. Tumor cells obtain cholesterol from serum. Cholesterol is needed for cell proliferation. Oxygen is needed for cholesterol synthesis. Serum can provide cholesterol in the absence of oxygen. Serum cholesterol can sustain tumor cell growth in the absence of oxygen.
It is helpful to grow the cells under at least two conditions, one under normal oxygen (normoxia) and another under hypoxia (<0.5% oxygen). Many consider that 20% O2 is normoxia. However, normoxia in tissue is only about 5–9% O2. In addition to using hypoxic conditions, he also thinks glycolytic inhibitors and anti-respiration drugs should be used as additional control groups. However, some of these drugs can produce toxic effects unrelated to OxPhos inhibition. On the other hand, hypoxia could influence mitochondrial function independent of OxPhos inhibition.
They found that the absence of CO2 can have toxic effects on cells. Cells grown in pure N2 without CO2 die from rapid acidity. Consequently, several observations are required as evidence that cancer cells use OxPhos for energy production.
Many cancer cells can use both glucose and glutamine for maintaining viability. Energy from glucose fermentation is derived mostly through glycolysis. Energy from glutamine can be derived from OxPhos or from fermentation. Glutamine fermentation can provide energy from either substrate-level phosphorylation at the succinyl-CoA synthetase step in the TCA cycle or from the fumarate reductase reaction also in the mitochondria.
If glucose is not available for glycolysis or the pentose phosphate pathway, then viability cannot be maintained through these processes. If glutamine maintains cell viability through TCA cycle metabolism and OxPhos, then hypoxia should inhibit ATP synthesis and kill cells that are grown in glutamine alone or in mixtures of glutamine or galactose. Galactose is not generally fermented well in cancer cells. If glutamine maintains viability in the absence of O2, or in the presence of cyanide, then the process is not likely to involve OxPhos, which requires O2 and cytochrome c for ATP production.
Hypoxia or rapid inhibition of OxPhos should quickly kill respiring cells. The sodium pump becomes the cell’s dominant energy sink during hypoxia. If OxPhos is shut down due to the absence of O2, then the cell will swell and die due to depletion of energy for the sodium pump. Death will happen quickly, especially if glucose is not available as an alternative energy substrate to produce ATP through glycolysis.
If hypoxia does not kill the cell, the cell is obviously generating energy through mechanisms other than OxPhos. Lactate cannot provide energy to normal cells, if OxPhos is shut down. If lactate provides energy to tumor cells, then this is not likely to involve OxPhos, but rather fermentation. Lactate could be oxidized to pyruvate, which could then be fermented in the mitochondria through substrate-level phosphorylation. If glucose is removed from the media and the cell continues to survive in hypoxia, then OxPhos is not likely the mechanism of survival. It is important to mention, however, that Molina et al. reported that lactate might serve as a metabolic fuel for subsets of breast cancer cells.
Ketone bodies and fatty acids can provide alternative metabolic fuels to glutamine for mitochondrial ATP synthesis. As these alternative fuels also require O2 for metabolism, death should occur quickly for any cell in the absence of both glucose and O2, especially if ketone bodies and fatty acids are the only available fuels. If cells maintain viability in O2 using either ketone bodies or fatty acids as the only energy substrates, then these cells are likely using OxPhos for survival.
If viability and lactate production are similar for tumor cells grown in the presence or absence or O2 in minimal media with glucose and glutamine as the only energy metabolites, then the cells are likely deriving their energy from fermentation rather than from OxPhos. If cancer cells are using OxPhos, then viability should be significantly lower in the absence than in the presence of O2. If cancer cells are using OxPhos, then viability should be significantly lower in the absence than in the presence of KCN, which inhibits cytochrome c and OxPhos.
OxPhos Origin of ATP in Cancer Cells Reevaluated
Reitzer et al. presented evidence showing that glutamine, rather than glucose or other sugars, was the major energy source for cancerous HeLa cells. According to Google, this paper has been cited almost 600 times since its publication in 1979. Although the evidence presented in this paper indicates that glutamine is a major energy substrate for HeLa cells, the authors did not prove that the energy derived from glutamine actually came through coupled OxPhos, despite their suggestion that it did.
The Reitzer et al. experiments are incomplete as they lack critical control experiments (cells grown in complete media under pure N2 and a cell viability assay), showing that their experimental conditions were not toxic to the cells. Without these experiments and without inclusion of longer-term viability data (longer than 2 h), it is not possible to conclude that glutamine oxidation provides ATP by OxPhos. Seyfried’s findings indicate that pure N2 and absence of CO2 is toxic to cultured tumor cells grown in complete medium (containing serum, glutamine, and glucose) and cannot be used to assess whether OxPhos is operational.
What About OxPhos Expression in Other Tumors?
Wong and colleagues described evidence indicating that respiration was mostly normal in mitochondria isolated from tissues of ovarian cancer patients. Although these investigators did not evaluate respiration in control mitochondria isolated from normal ovarian tissue, they mentioned that ATP production and the specific activities of succinate, malate, and glutamate dehydrogenases were comparable to values reported in human skeletal muscle, heart, and liver. However, a careful examination of the data from their Table shows that the rate of ATP production was markedly lower in ovarian and peritoneal cancers (mean of 37 nmol/min/mg) than in skeletal muscle (mean of 265 nmol/min/mg) when succinate was used as a substrate. Similar observations were obtained when TMPD+ascorbate was used as substrate.
On the basis of these data, it is not clear how these investigators could conclude that the TCA cycle was functional and that mitochondrial OxPhos was competent in these ovarian cancer tissues.
The Pederson Review on Tumor Mitochondria and the Bioenergetics of Cancer Cells
Although there are a variety of reasons why many investigators rejected Warburg’s central hypothesis that damage to respiration is the origin of cancer, most of the reasons are unsupported by evidence. In none of the cited works arguing against the Warburg theory has the investigators excluded mitochondrial amino acid fermentation and substrate-level phosphorylation as an alternative to OxPhos for mitochondrial energy production.
Amino Acid Fermentation Can Maintain Cellular Energy Homeostasis During Anoxia
Mitochondrial amino acid fermentation is known to maintain metabolic homeostasis under hypoxia in several species of diving animals. Mitochondrial amino acid fermentation can also maintain metabolic homeostasis in the heart and kidney under low glucose and low O2 conditions.
While it is well known that glucose can be fermented, less is known about amino acid fermentation. Lactate is the by-product of glucose fermentation, whereas succinate, alanine, and aspartate are by-products of glutamine or amino acid fermentation under hypoxia. The expression of lactate in the presence of O2 is abnormal and would indicate that the cells are fermenting. The degree of fermentation is positively correlated with the degree of malignant growth. Also, the less is the respiration, the greater the fermentation. Under anoxia, fumarate can replace O2 as an electron acceptor. If the cells consume oxygen, it is unlikely that succinate would accumulate. Under high glucose, amino acid fermentation can occur whether or not succinate accumulates. Hence, it is important to account for the multiple variables required to assure that cells are actually using OxPhos alone or are using some combination of OxPhos and mitochondrial substrate-level phosphorylation to maintain their viability.
Evidence Suggesting that Metastatic Mouse Cells Derive Energy from Glutamine Fermentation
Seyfried showed that tumor cell viability and ATP production were robust in either anoxia or cyanide as long as both glucose and glutamine were present in the media. Since anoxia (95% N2, 5% CO2) or cyanide (an inhibitor of complex IV respiration) inhibits OxPhos, the robust synergy seen for glucose and glutamine is unlikely due to significant energy from OxPhos.
They propose that the glucose/glutamine energy synergy observed in their metastatic mouse cells arises from linked fermentation redox couples in the cytoplasm and mitochondria that synthesize ATP largely through nonoxidative substrate-level phosphorylation.
Fermentation Energy Pathways Can Drive Cancer Cell Viability Under Hypoxia
The malate–aspartate and glycerol 3-phosphate shuttles can link the redox couples in cytoplasm and mitochondria. This linkage is consistent with evidence showing high expression of these shuttle systems in various cancer cells. Shuttle expression in tumor cells, however, depends in part on whether cells can grow in the presence or absence of glucose. In addition to the shuttles, the mitochondrial fumarate reductase pathway is also thought to produce ATP under certain hypoxic conditions. NADH serves as the electron and proton donor, whereas fumarate serves as the ultimate electron and proton acceptor with succinate as an end product.
According to their model, simultaneous glutamine and glucose fermentation would maintain cancer cell viability in those environments where oxygen is limited. It remains to be determined, however, if glutamine can also be fermented in tumor cells in the presence of oxygen.
Succinate accumulation is indicative of amino acid fermentation under hypoxia. It is not yet clear if the succinate detected in the tumor cells under aerobic conditions from the NMR experiments results from glutamine fermentation. Succinate should not accumulate in cells that respire. It is also possible that glutamine is oxidized under aerobic conditions but is fermented under hypoxia. Glutamine could also be metabolized under hypoxia through anaerobic respiration involving uncoupled electron transport. Elevated glucose levels would suppress OxPhos through a Crabtree effect, thus allowing the possibility of glutamine fermentation under normoxia. It would be difficult to distinguish glutamine respiration from glutamine fermentation under normoxia since both processes would involve electron transfer and TCA cycle activity.
Glutamine fermentation, occurring under high glucose conditions, will generate considerable energy through substrate-level phosphorylation and possibly through the fumarate reductase reaction. Neither process involves OxPhos but would still require uncoupled electron transport. ATP uptake into the mitochondria from the cytoplasm and electron transport would be needed to drive the F1-F0-ATPase in reverse in order to maintain a proton motive gradient.
Seyfried believes this situation would be present in those highly glycolytic tumor cells where the hexokinase-2 becomes attached to the outer mitochondrial membrane as described by Pedersen. The ATP needed to drive the ATP synthase in reverse under hypoxia would come almost exclusively from glucose and glutamine fermentation. Hence, targeting glucose and glutamine could effectively shut down energy metabolism in many cancers that depend on these metabolites for energy.
Tumor cells survive in hypoxia not because they have a growth advantage over normal cells but because they can ferment organic molecules. Organic molecules become O2 surrogates in accepting electrons. Cancer cells not only ferment glucose, as Warburg first showed, but they might also ferment glutamine and possibly other amino acids in the mitochondria under hypoxia and when glucose levels are high under normoxia. Unlike normal cells that can switch back to OxPhos when O2 becomes available, most tumor cells depend on fermentation metabolism whether or not O2 is present in the environment. Tumor cells adapt to fermentation because their OxPhos is insufficient to maintain energy homeostasis. Fermentation adaptation underlies the pathology of cancer.
The difference between glutamine oxidation and glutamine fermentation is that the latter does not couple the proton motive gradient to ATP production.
Competing Explanations for the Metabolic Origin of Cancer
The first hypothesis is that of Weinhouse, which considers that cancer cells express aerobic glycolysis despite having normal respiratory function.
The second hypothesis suggests that elevated glycolysis suppresses respiration in cancer cells. Under this hypothesis, cancer respiration is considered repressed, but the repression arises secondary to the appearance of aerobic glycolysis. In other words, many of the abnormalities seen in tumor mitochondria structure and function would arise as effects rather than the cause of aerobic glycolysis.
Summary
Cancer is a disease of abnormal energy metabolism. In order to survive insufficient respiration, tumor cells have adapted to energy production through fermentation. Powerful synergy is established between fermentation redox couples in the cytoplasm and mitochondria. These redox couples are linked through shuttle systems that drive tumor cell energy metabolism using glucose and glutamine as fermentable metabolic fuels. Adaptation to fermentation allows tumor cells to survive and grow in hypoxic environments.
Does Cancer Have a Genetic Origin?
With the exception of imatinib (Gleevec), which targets the Abelson (ABL) proto-oncogene receptor tyrosine kinase, little success has been found to date for other targeted therapies.
How was it possible for the gene theory to gain precedence over Warburg’s metabolic theory for the origin of cancer?
Theodor Boveri, Aneuploidy, and the Genetic Origin of Cancer
The gene theory of cancer originated with Theodor Boveri’s suggestion in 1914 that cancer could arise from defects in the segregation of chromosomes during cell division.
As chromosomal instability in the form of aneuploidy (extra chromosomes, missing chromosomes, or broken chromosomes) is present in many tumor tissues, it was not too much of a reach to extend these observations to somatic mutations within individual genes including oncogenes and tumor suppressor genes.
Boveri’s hypothesis on the role of chromosomes in the origin of malignancy was based primarily on his observations of chromosome behavior in nematodes (Ascaris) and sea urchins (Paracentrotus) and on von Hansemann’s earlier observations of chromosome behavior in tumors. Hence, the founder of the genetic theory of cancer appears not to have directly studied the disease.
Inconsistencies with the Genetic Origin of Cancer
The most damning evidence against the gene theory comes from nuclear/cytoplasmic transfer experiments. Gene and chromosomal defects can, however, contribute to the respiratory insufficiency in tumor cells, thus solidifying the insufficiency once it occurs. Aneuploidy can disrupt respiratory function, thus forcing cells to rely more heavily on fermentation for energy.
Many correctly surmise that it is easier to get papers published and grants funded in hot areas than in areas not considered hot. Cancer is one of the few fields where research areas are consistently hot, but progress toward the cure is consistently cold.
Respiratory Insufficiency as the Origin of Cancer
Metabolic studies in a variety of human cancers have previously shown that loss of respiratory function precedes the appearance of malignancy and aerobic glycolysis (the Warburg effect).
Roskelley and coworkers also illustrated this fact in their studies of various animal and human tumor tissues. They used two chemical systems to assess the respiratory function in tumor tissue and in the normal host tissue from which the tumor arose.
These systems included the following:
These enzyme systems provide the main pathway by which oxygen is fed to the vital combustive processes occurring in most normal cells. These pathways therefore represented a physiologic unit for evaluating the likelihood that a given tissue is neoplastic. It was clear from their findings that the respiratory function was seriously impaired in human cancer tissue in comparison to the respiratory function in normal non-diseased host tissues.
It is now recognized that carcinogenic hydrocarbons, aflatoxin, viruses, and X rays all damage mitochondrial function and energy metabolism in similar way. This is interesting since chemical carcinogens and viruses also activate oncogenes in a similar way, suggesting that oncogene activation follows mitochondrial damage. While γ -radiation causes mutations, it is the effect of radiation on mitochondrial respiration that causes cancer.
Cancer is not a collection of many different diseases, but is rather a singular disease of respiratory insufficiency regardless of the tissue origin or cellular composition. While cancer cells arising in one organ will look morphologically different from cancer cells arising in another organ, they all suffer from a common malady, that is, respiratory insufficiency with compensatory fermentation.
Germline Mutations, Damaged Respiration, and Cancer
In general, cancer-causing germline mutations are rare and contribute to only about 5–7% of all cancers. Although mitochondrial function is impaired in all tumor cells, it remains unclear how these impairments relate to the large number of somatic mutations and chromosomal abnormalities found in tumors.
The risk for paraganglioma involves mutations in the succinate dehydrogenase (SDH) gene, whereas risk for leiomyomatosis and renal cell carcinoma involves mutations in the fumarate hydratase (FH, fumarase) gene. Mutations in the von Hippel–Lindau (VHL) tumor suppressor gene enhance risk for the VHL syndrome involving a predisposition to renal clear cell cancer (RCC), retinal and central nervous system hemangioblastomas, pancreatic cysts, and adrenal tumors (pheochromocytomas). The VHL tumor suppressor gene targets the mitochondria. It is important to recognize that these and similar mutations directly impair mitochondrial energy production leading to increased glycolysis and the Warburg effect.
Inherited Mutations in p53 and Damaged Respiration
Rare inherited mutations in the p53 tumor suppressor gene increase risk for cancers of the Li Fraumeni syndrome. The spectrum of cancers identified in this syndrome includes breast carcinomas, soft tissue sarcomas, brain tumors, osteosarcoma, leukemia, and adrenocortical carcinoma.
Hwang and coworkers have shown that p53 regulates mitochondrial respiration through its transcriptional target gene synthesis of cytochrome c oxidase 2 (SCO2). Most importantly, these investigators have shown that genome stability is dependent on OxPhos. These findings are also consistent with the earlier findings of Singh and coworkers showing that mitochondrial energy metabolism is impaired in human cancer cells containing defects in p53. Genome stability is dependent on OxPhos, while mutations in p53 influence cancer susceptibility through disturbance in mitochondrial OxPhos. Werner syndrome, a disease of rapid aging and cancer predisposition, can also be linked to abnormalities in p53 and defective mitochondrial function. Hence, the guardian function of p53 appears to reside in its ability to maintain sufficient OxPhos activity.
A recent commentary in Science has suggested that the tumor suppressor genes p73 and p63 could serve along with p53 as “brothers in arms against cancer”. Although p73 also appears to function in the mitochondria like p53, germline mutations in p73 are not associated with increased cancer risk. As no effective cancer therapies have yet emerged from attempts to manipulate p53 in tumor cells, it is unlikely that effective therapies will emerge from attempts to manipulate either p73 or p63 in tumor cells.
Inherited Mutations in BRCA1 and Damaged Respiration?
Coene and coworkers show that several anti-BRCA1 antibodies colocalize with mitochondrial staining in a variety of normal and cancer cell lines. BRCA1 was mostly localized to the mitochondrial matrix possibly in association with mtDNA (mitochondrial DNA). About 20% of BRCA1 staining was also found in the inner mitochondrial membrane, suggesting involvement in multiple mitochondrial functions. Like the BRCA1 tumor suppressor protein, a mitochondrial location was also reported for the adenomatous polyposis coli (APC) tumor suppressor protein, which is mutated in most of the colon cancers. These findings raise the possibility that mutations in the BRCA1 and APC genes influence cancer susceptibility through alterations of mitochondrial function and OxPhos efficiency.
Inherited Mutations in RB and Damaged Respiration
The tumor suppressor protein, RB (retinoblastoma), regulates cell cycle exit and is dysregulated in numerous cancers. Germline mutations in the RB gene cause familial forms of retinal tumors. Recent studies indicate that the RB protein regulates mitochondrial biogenesis and the control of cell differentiation. In other words, defects in RB alter mitochondrial function, thus sustaining cell proliferation while preventing differentiation. Normal mitochondrial function is required for maintaining cellular differentiation and quiescence. These findings also link the action of oncogene-induced cell senescence through effects on mitochondria and RB activity. Abnormalities in ATP production through OxPhos are linked to aerobic glycolysis in tumors with RB abnormalities.
Xeroderma Pigmentosum and Damaged Respiration
Enhanced susceptibility to skin cancers is seen in patients who inherit the autosomal recessive gene for xeroderma pigmentosum (XP). XP involves defects in nuclear DNA repair, thus enhancing susceptibility to cancer in skin cells and neural defects in brain cells. This disease is often used to support the hypothesis that cancer is a genetic disease since defects in genomic stability are the linchpin for the gene theory of cancer. However, studies from Rothe and coworkers have shown that XP mutations alter mitochondrial energy production. Altered mitochondrial ATP production is consistent with other studies showing that mitochondrial morphology and structure is also abnormal in XP patients and fibroblasts.
Fredrich’s Ataxia and Damaged Respiration
Friedrich’s ataxia involves reduced expression of the mitochondrial protein frataxin, which regulates OxPhos and mitochondrial ATP production. It appears that frataxin directs the intramitochondrial synthesis of iron/sulfur clusters needed for electron transport.
Ristow and colleagues have shown that targeted disruption of hepatic frataxin expression in mice causes impaired mitochondrial function and tumor growth.
Somatic Mutations and Cancer
Most of the gene defects found in cancers are not inherited, but arise sporadically, as do most mutations in the p53 gene. While germline mutations can increase the risk of some rare cancers, most cancer mutations are somatic and will contribute more to the progression than to the origin of most cancers. It is interesting to note, however, that somatic mutations occur only rarely in cells and tissues.
Loeb and colleagues had initially proposed that the multiple mutations found in tumor cells resulted from mutations in genes responsible for maintaining the fidelity of DNA synthesis or the adequacy of DNA repair. More specifically, mutations in genomic caretakers underlie genomic stability and the large number of somatic mutations found in cancer. Mutations in these genes would then trigger an explosion of new mutations throughout the genome.
As the integrity of the nuclear genome is dependent on the fidelity of OxPhos, respiratory insufficiency becomes the real mutator phenotype. On the other hand, a normal respiratory function can suppress tumorigenicity.
Mutations in the p53 caretaker gene are not expressed in all common human malignancies, suggesting a more complicated involvement of this and other genome guardians in carcinogenesis. Although p53 mutations are considered to be common in human glioblastoma multiforme, no defects in the p53 gene are found in about 60% of these tumors. While numerous genetic abnormalities have been described in most human cancers, no specific mutation is reliably diagnostic of any specific type of tumor.
Although common somatic mutations occur in some tumors, it is unlikely that these mutations are expressed in every individual cell of the tumor due to cellular and genetic heterogeneity. It is interesting that progression of malignant gliomas is generally slower in patients with chromosome 1p/19q co-deletions, promoter hypermethylation of the O6-methylguanine methyltransferase (MGMT) gene, or with mutations in the gene for IDH1. Should we consider these as “good” mutations since tumors containing these mutations grow slower than tumors not containing these mutations? GBM patients with IDH1 mutations live slightly longer than patients without this mutation. Mutations in this gene could inhibit mitochondrial amino acid fermentation, thus disrupting the glucose/glutamine synergy.
Considering the complexity of metabolic flux, genetic heterogeneity, and gene–environmental interactions, caution should be used in assuming that targeting any specific mutation or signaling pathway will have a major effect on tumor growth or patient survival. Sandra Yin’s piece in Medscape Medical News made this point clear. It should be no surprise that attempts to restore p53 guardian function in cancer patients have met with little success.
Revisiting the Oncogene Theory
According to Michael Stratton, the key evidence supporting the oncogene theory arose from studies showing that the introduction of total genomic DNA from human cancers into normal NIH3T3 cells could transform them into cancer cells. He cited the paper from Krontriris and Cooper as providing this evidence. However, high molecular weight DNA from only 2 out of 24 cancers, both bladder cancers, was able to transform the NIH3T3 cells into cancer cells.
Viral infection can damage mitochondria. No information was presented showing that mitochondria were normal or unaffected in the transformed cells. This might be difficult, however, since fermentation is elevated in the NIH3T3, suggesting that they suffer from some type of respiratory insufficiency. This led Rubin to agree with Leslie Foulds’ conclusions that epigenetic phenomena contribute in part to the transformation of normal cells including NIH3T3 cells. Mitochondria represent an extrachromosomal epigenetic system.
Warburg noted that acute damage to respiration is more likely to cause cell death than to cause cancer. Only those cells capable of upregulating fermentation to compensate for chronic mitochondrial damage can become tumor cells. Moiseeva et al. have shown that mitochondrial dysfunction, not a defect in glucose consumption, is the underlying cause of the bioenergetic defects of Ras-senescent cells. A defect in tumor cell glucose consumption would be expected if tumorigenesis were dependent on the damage to the regulation of glycolysis as suggested by Koppenol and Dang.
Elevated fermentation allows cells to bypass senescence, thus enhancing the likelihood of oncogenic transformation. Several research groups provide compelling evidence showing that oncogene transformation increases ROS expression and damages mitochondria. Lee and coworkers have shown that transfection of human diploid cells with V12Ras significantly increased the damage to oxygen species in mitochondria, whereas Weinberg and colleagues have shown that mitochondrial ROS generation and damage to complex III was essential for K-Ras-induced cell proliferation and tumorigenesis. Moreover, Yang and colleagues have shown that H-Ras transformation of mouse fibroblasts damaged respiration, thus forcing the cells into a glycolytic metabolism. This is notable since activated Ras has been proposed to induce MYC activity and to enhance non-hypoxic levels of HIF-1α. As MYC and HIF-1 drive glycolysis, their upregulation would be necessary to prevent senescence following respiratory damage.
MYC-induced damage to mitochondrial structure and respiratory function caused some cardiomyocytes to die, but caused other cardiomyocytes to reenter the cell cycle and proliferate. There is also emerging evidence that the c-RAF oncogene targets the mitochondria, which produces ROS and damages mitochondrial physiology. Elevated Hif-1α expression follows RAF-c-induced mitochondrial damage. Hif-1α upregulates glucose transport and multiple glycolytic pathways. Hence, oncogenes can sometimes target and damage mitochondrial function.
These findings indicate that respiratory dysfunction is an effector pathway of oncogene-induced senescence. An upregulation of glycolysis following respiratory insufficiency will prevent senescence leading to cell-cycle reentry and proliferation, that is, the initiating events in tumorigenesis.
Mitochondrial Mutations and the Absence or Presence of Cancer
If defective mitochondrial respiratory function is the origin of all cancers, why are cancers rare in those people that inherit mutations damaging mitochondrial respiration? For example, mutations in Cu/Zn superoxide dismutase (SOD) gene, which disturbs respiratory function, are associated with familial amyotrophic lateral sclerosis. However, cancer is rare in patients with ALS (amyotrophic lateral sclerosis).
Most of the inherited mutations that affect respiratory chain function and the TCA cycle are homozygous and cause profound damage to multiple organ systems. Inherited mutations are found in all cells, whereas the mitochondrial defects in cancer cells are found only in the cancer cells. Also, some individuals that inherit mitochondrial mutations do not live long enough to get cancer, for example, those with Barth syndrome involving abnormalities in cardiolipin remodeling. Second, those mutations that alter the TCA cycle function and cause cancer, that is, mutations in the SDH and FH genes, are generally heterozygous and do not affect the physiology of multiple organ systems. Homozygous mutations in these genes are associated with neurodegeneration rather than cancer. Neurodegeneration is also seen in those individuals with heterozygous mutations in the SOD gene.
Douglas Wallace suggests that mutations producing mitochondrial ROS rather than energy impairment are the missing link to cancer. However, mitochondrial ROS kills dopaminergic cells in Parkinson’s disease without producing cancer.
Critical Evaluation of Pathogenic Mitochondrial DNA Mutations in Tumors
Salas and coworkers have shown that much of the evidence for pathogenic mtDNA mutations in human tumor cells was largely due to artifacts of data interpretation or methodologies used in mtDNA analysis. In order to prove that mtDNA mutations contribute to the OxPhos deficiency in tumor cells, it is necessary to isolate and purify mitochondria from the tumor tissue and from the normal tissue of the patient, and then sequence the entire genome of the purified mtDNA of the tumor and normal tissue. Many studies of mtDNA mutations in human tumor tissue fail to include all of the necessary controls to exclude misinformation.
As the mitochondrial genome is highly redundant, it is unlikely that many cancers arise directly from mtDNA mutations due to the multiple copies of normal alleles in the mitochondrial genome. However, some cancers could arise if mutations are expressed in all copies of the circular mitochondrial genome or where the entire mitochondrial genome is depleted, as Singh and coworkers have recently described.
It is possible that the mtDNA content is lower in the tumor cells than in normal cells. Depletion of mtDNA increases the expression of uncoupling proteins (UCPs). Activation of mitochondrial UCPs, especially uncoupling protein 2 (UCP-2), has been detected in a broad range of tumor cells. Normal cells activate UCP-2 in response to elevated glucose levels in order to reduce mitochondrial membrane hyperpolarization. UCP activation can also help reduce ROS, which arises from elevated glucose levels. UCP-2 activation in tumor cells can be an attempt to regulate oxidative stress following OxPhos damage and mtDNA depletion.
Viral Infection, Damaged Respiration, and the Origin of Cancer
Viruses have long been recognized as the cause of some cancers. About 15% of human cancers are caused by tumor viruses. Kofman and colleagues recently reviewed substantial information linking viral infections to the origin of malignant gliomas. It is interesting that several cancer-associated viruses or their protein products localize to, or accumulate in, the mitochondria. Viral alteration of mitochondrial function could potentially disrupt energy metabolism, thus altering the expression of tumor suppressor genes and oncogenes over time. Viruses that affect mitochondrial function and increase cancer risk include the Rous sarcoma virus (src), Epstein–Barr virus (EBV), Kaposi’s sarcoma-associated herpes virus (KSHV), human papilloma virus (HPV), hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), human cytomegalovirus (HCMV), and human T-cell leukemia virus type 1 (HTLV-1).
Although viral disruption of mitochondrial function will kill many cells through apoptosis following an acute infection, those infected cells that can upregulate fermentation through substrate-level phosphorylation will survive and potentially produce a neoplasm following chronic infection.
Siddiqui and colleagues have shown that the HBV-encoded protein HBx, which enhances the risk of hepatocellular carcinoma, disrupts the mitochondrial proton motive gradient. The HBx protein also blocks ubiquitination of HIF-1α thus increasing HIF-1α stability and activity in a hypoxia-independent manner.
Although it is assumed that these retroviruses cause cancer by nuclear DNA insertion and oncogene upregulation, considerable evidence indicates that either the viruses themselves or their protein products damage OxPhos, leading to respiratory insufficiency. Viral infections can cause cancer through damage to cellular respiration.
HIV and Cancer Risk
People infected with HIV have a substantially higher risk of contracting some types of cancers than uninfected people of the same age. Three of these cancers are known as acquired immunodeficiency syndrome (AIDS)-defining cancers or AIDS-defining malignancies: Kaposi sarcoma, non-Hodgkin lymphoma, and cervical cancer. People infected with HIV are about 800 times more likely than uninfected people to be diagnosed with Kaposi sarcoma, at least seven times more likely to be diagnosed with non-Hodgkin lymphoma, and, among women, at least three times more likely to be diagnosed with cervical cancer. In addition, people infected with HIV are also at higher risk for anal cancer, Hodgkin lymphoma, liver cancer, and lung cancer.
The explanation given for increased cancer risk in HIV infection is that the infection weakens the immune system and reduces the body’s ability to destroy cancer cells and fight infections that may lead to cancer. This explanation does not address where all these cancer cells came from in the HIV-infected patients. This explanation is also not connected to a molecular mechanism. Chronic viral infections can cause inflammation. Inflammation damages OxPhos, thus shifting energy metabolism to fermentation. OxPhos insufficiency is the origin of cancer regardless of the tissue involved.
Summary
Many germline cancer mutations damage OxPhos. Many known carcinogenic agents damage cellular respiration while also producing nuclear genomic instability. Oncogene activation and tumor suppressor gene inactivation are necessary changes in order to drive fermentation when OxPhos is insufficient. These changes are effects rather than causes of the disease.
The Retrograde (RTG) Response: An Epigenetic System Responsible for Nuclear Genomic Stability
The RTG response is the general term used for mitochondria-to-nuclear signaling and involves cellular responses to changes in respiration and the functional state of mitochondria. The RTG response is initiated following interruption in the respiratory energy production. Genomic stability is dependent on the integrity of the mitochondrial function. If respiratory insufficiency is not corrected, the RTG response will persist, thus producing the Warburg effect, genomic instability, and the path to tumorigenesis.
Although DNA methylation and histone modification are considered to be one type of epigenetic mechanism, the mitochondrion as an extrachromosomal element is the predominant driver of epigenetic control within the cell. Mitochondria maintain cellular differentiation through well-established nuclear cytoplasmic interactions.
Jazwinski and colleagues have recently shown that the RTG metabolic stress response in yeast is similar to the NF-kB metabolic stress response in humans. Expression of multiple nuclear genes controlling energy metabolism is profoundly altered following impairment in mitochondrial energy homeostasis. Respiratory insufficiency can arise from abnormalities in mtDNA, the TCA cycle, the electron transport chain, or in the proton motive gradient of the inner membrane.
The RTG response evolved in eukaryotic microorganisms to maintain cell viability following periodic disruption of respiratory ATP production. This mostly involves an energy transition from OxPhos to substrate-level phosphorylation including glycolysis and amino acid fermentation.
According to Seyfried’s hypothesis, the RTG response would include upregulation of networks needed for nonoxidative energy metabolism.
The structural organization of the cell including its morphology and genome integrity is dependent on sufficient respiration. The maintenance of structure and genome integrity is dependent on the regulatory elements of the RTG response. Although the RTG response evolved to protect cell viability following transient disruption of respiration, a prolonged RTG response will lead to genomic instability and disorder.
Three main regulatory elements define the RTG response in yeast, including the Rtg2 signaling protein and the Rtg1/Rtg3 transcriptional factor complex (both are basic helix-loop-helix-leucine zippers). Rtg2 contains an N-terminal, ATP-binding motif that senses changes in mitochondrial ATP production. Rtg2 also regulates the function and cellular localization of the heterodimeric Rtg1/Rtg3 complex.
The RTG response is turned “on” following insufficient energy production through OxPhos. In the on state, cytoplasmic Rtg2 disengages the Rtg1/Rtg3 complex through a dephosphorylation of Rtg3. The Rtg1 and Rtg3 proteins then individually enter the nucleus where Rtg3 binds to R box sites, Rtg1 reengages Rtg3, and transcription and signaling commences for multiple energy and antiapoptotic related genes and proteins to include MYC, TOR, Ras, CREB, NF-kB, and CHOP.
The primary role of the RTG response is to coordinate the synthesis of ATP through glycolysis alone or through a combination of glycolysis and glutamine metabolism when respiration becomes insufficient to maintain energy homeostasis. The RTG response would be essential for maintaining a stable GATP for cell viability during periods when OxPhos is impaired. A prolonged RTG response, however, would leave the nuclear genome vulnerable to instability and mutability.
The upregulation of oncogenes (Myc, Ras, Akt, Hif-1, etc.) becomes necessary to derive tumor energy through fermentation. Respiratory insufficiency coupled with compensatory fermentation also increases levels of (i) cytoplasmic calcium; (ii) the multidrug resistance phenotype; (iii) production of reactive oxygen species (ROS); and (iv) abnormalities in iron–sulfur complexes. Together these changes would further accelerate aberrant RTG signaling and genome mutability.
The expression of matrix metalloproteinase 2 (MMP2) is elevated in cells with mtDNA deficiency. MMP2 and other metalloproteases are elevated in association with chronic inflammation. Since mtDNA deficiency and ROS production reduces mitochondrial respiration, it is not unreasonable to speculate that MMP2 expression would also be elevated following any number of insults to mitochondrial respiration. Elevated MMP expression is the phenotype seen in activated macrophages, which hybridize with neoplastic epithelial to form cancer cells with a high metastatic potential. ROS production associated with inflammation would also activate the RTG response as previously seen in other systems. Hence, the RTG response can be linked to both the initiation and the progression of carcinogenesis.
Human Myc/Max transcription factor complex shows interesting homologies to the yeast Rtg1/Rtg3 proteins. MYC is a member of the basic, helix-loop-helix-leucine zipper family of transcription factors as are Rtg1/Rtg3. MYC upregulation is also necessary for the induction of genes needed for glycolysis and glutamine metabolism. Although there is currently no known counterpart in higher eukaryotes for the Rtg2 protein as a sensor of mitochondrial dysfunction and a transducer of mitochondrial signals that activate Rtg1/3-like transcription factors, there is recognized conservation of the NF-kB stress response in humans and the RTG stress response in yeast.
The yeast RTG response also shares interesting functional homologies with the mTor, Akt, and RAS signaling pathways.
Epigenetics involves more than just DNA methylation, genomic imprinting, and histone modification. Mitochondrial function is also epigenetic. It is interesting that inherited defects in p53 can damage OxPhos, leading to genomic instability. Hwang and colleagues showed that efficient mitochondrial respiration is essential for maintaining genomic stability in environments where oxygen is present.
The multiple carcinogenic effects of the Myc oncogene can also be linked to OxPhos damage. While the RTG response evolved to protect cells from acute energy failure, a persistent RTG response associated with insufficient respiration can eventually initiate genomic instability and tumorigenesis. Hence, chronic respiratory insufficiency together with an activated RTG response is the gateway to cellular disorder and the origin of neoplasia regardless of whether genetic or environmental factors initiate the response.
Inflammation Injures Cellular Respiration
It is not clear how inflammation specifically causes cancer. It is known that inflammation associated with sepsis or LPS impairs mitochondrial respiration. Sepsis is an acute inflammatory condition that can lead to systemic organ failure and death. In contrast to inflammation from sepsis and LPS, which induce acute mitochondrial failure and cell death, the inflammation that causes many cancers is chronic. Chronic inflammation will produce protracted mitochondrial damage.
Viewed collectively, these findings indicate that respiratory damage links inflammation to carcinogenesis. Chronic inflammation, which enhances the expression of nitric oxide and TGF-β, damages respiration. Most cells that suffer respiratory damage die. According to Warburg’s theory, tumors arise only from those cells that are capable of increasing fermentation in order to compensate for insufficient respiration. Enhanced fermentation prevents senescence.
Hypoxia-Inducible Factor (HIF) Stability is Required for the Origin of Cancer
Mammalian cells increase the expression of HIF-1α in response to transient hypoxia. HIF-1α is rapidly degraded under normoxia, but becomes stabilized under hypoxia.
The rapid degradation of HIF-1α in oxygen is regulated by oxygen-dependent prolyl hydroxylases (PHDs). PHDs hydroxylate prolyl residues in an oxygen-dependent degradation domain. The inhibition of PHDs stabilizes Hif-1α even in the presence of oxygen. HIF-1α stabilization under aerobic conditions can be linked to respiratory insufficiency through abnormalities in calcium homeostasis, ROS generation, NF-kB signaling, accumulation of TCA cycle metabolites (succinate and fumarate), and oncogenic viral infections. Genomic instability arises, in part, through ROS production and “prolonged” HIF-1α stabilization under aerobic conditions. This process would be linked to the RTG system.
Gottlieb and colleagues indicate that certain energy metabolites, such as succinate, α-ketoglutarate, and fumarate, can stabilize HIF-1α in the presence of oxygen. Succinate and fumarate are also products of amino acid fermentation. Hence, succinate and fumarate, arising through glutamine fermentation, could contribute to inhibited PHDs and the stabilization of Hif-1α. As Hif-1α expression regulates multiple genes needed for glycolysis, Hif-1α stabilization would be important for maintaining fermentation energy production through substrate-level phosphorylation following deficiency in OxPhos. It is important to recognize that respiratory insufficiency is ultimately responsible for Hif-1α stabilization in cancer cells.
Mitochondria and the Mutator Phenotype
Singh and the Jazwinski groups provide compelling evidence that mitochondrial dysfunction, operating largely through the epigenetic RTG response (mitochondrial stress signaling), can underlie the mutator phenotype of tumor cells. Chromosomal instability, expression of gene mutations, and the tumorigenic phenotype are significantly greater in human cells with mtDNA depletion than in cells with normal mtDNA.
Singh and coworkers have shown that mtDNA depletion downregulates the expression of the apurinic/apyrimidinic endonuclease (APE1). APE1 is a redox sensitive multifunctional endonuclease that regulates DNA transcription and repair. In other words, the function of this DNA repair enzyme is dependent on the mitochondrial function.
Regardless of the process by which mitochondria get damaged, respiratory insufficiency in tumor cells as an initial event in carcinogenesis can account for the eventual genomic instability seen in cancer cells. Hence, the elevated mutation rates, gross chromosomal rearrangements, and alterations in chromosome number observed in tumor cells can be linked to impaired mitochondrial respiration.
Besides APE1, other DNA repair proteins are downregulated in association with mtDNA depletion and OxPhos insufficiency, including p53 and SMC4.
Even different tumors within the same cancer type could appear to represent different diseases when evaluated at the genomic level. When evaluated at the metabolic level, however, most cancers and tumors are alike in expressing respiratory insufficiency and elevated fermentation. Impaired mitochondrial function can induce abnormalities in tumor suppressor genes and oncogenes. For example, an impaired mitochondrial function can induce abnormalities in p53 activation, while abnormalities in p53 expression and regulation can further impair mitochondrial respiration.
Calcium Homeostasis, Aneuploidy, and Mitochondrial Dysfunction
Calcium homeostasis is dependent on mitochondrial function and the integrity of the proton motive gradient of the inner mitochondrial membrane. Calcium homeostasis is essential for the fidelity of cell division to include spindle and microtubule assemblies, sister chromosome separation, and cytokinesis. In light of the important role of the mitochondria in maintaining intracellular calcium flux, any disturbances in cytoplasmic calcium homeostasis, arising as a consequence of insufficient respiration, will contribute to abnormalities in chromosomal segregation during mitosis. In other words, nondisjunction and mitotic defects can arise from changes in the intracellular calcium flux, which is ultimately determined by the health status of the mitochondria and the sufficiency of OxPhos.
We now know that calcium flux maintains the fidelity of mitosis and that inappropriate attachment of kinetochores to spindle microtubules can undermine the fidelity of chromosome segregation during mitosis, which leads eventually to aneuploidy. As calcium flux regulates these processes, disturbances in calcium flux can lead to chromosome imbalances during cell division. The integrity of the proton motive gradient of the inner mitochondrial membrane is largely responsible for the intracellular calcium flux. Consequently, damage to mitochondrial respiration and the integrity of this membrane will ultimately contribute to chromosome imbalances and aneuploidy.
We also know that ROS, arising from any number of environmental insults including tissue inflammation, damage the proton motive gradient. These insults include X rays, chemicals, or viruses. Samper and colleagues and Seoane et al. have clearly shown that mitochondrial stress, arising through ROS, causes genomic instability including aneuploidy.
Seyfried proposes that aneuploidy and the numerous somatic mutations and other genomic aberrations found in cancer cells ultimately arise as a consequence of damage to mitochondrial proteins, lipids, and mtDNA. This damage dissipates the proton motive gradient, leading to elevated fermentation and an imbalance in the cellular calcium flux. The vast number of genomic changes identified in sporadic cancers ultimately arises as a consequence of mitochondrial dysfunction and respiratory insufficiency.
Mitochondrial Dysfunction and Loss of Heterozygosity (LOH)
A normal phenotype can be maintained in the heterozygous state for most recessive genes. Loss of function or deletion of the single normal allele, however, will prevent production of any normal product from that gene. The loss of heterozygosity (LOH) in relationship to the origin of cancer is often referred to as the Knudson hypothesis and originates with Alfred Knudson, who had first developed the idea that LOH in critical genes such as p53 and RB could predispose individuals to cancer. This concept is now a well-accepted mechanism for the somatic mutation theory of cancer.
Recent studies in yeast indicate that damage to the inner mitochondrial membrane potential, following mtDNA depletion, induces mitochondrial dysfunction and LOH in the nuclear genome. Yeast colonies formed following mtDNA depletion varied in size, but eventually expressed improved growth following repeated passaging despite the continued absence of their mtDNA. Remarkably, these clones were unable to respire and had slower growth than cells with intact mtDNA. After 30 h of growth, however, they formed colonies that grew faster, and displayed fewer nuclear LOH events than cells within the first 30 h following loss of mtDNA.
Tissue Inflammation, Damaged Respiration, and Cancer
Sonnenschein and Soto argued that disruption of tissue organization and structure, rather than random somatic mutations, could give rise to cancers. They describe this process as the tissue organizational field theory (TOFT) of carcinogenesis. This concept is based on evidence that cancer-provoking agents disturb the tridimensional organization of tissue architecture, thus disturbing positional and historical information embodied in the morphogenetic field.
ROS damages mitochondrial proteins, lipids, and nuclear DNA. ROS arise from chronic inflammation, which also disrupts tissue morphogenetic fields. Chronic disruption of the tissue morphogenetic field (microenvironment) would ultimately impair respiratory function in cells within the field. Acquired abnormalities in mitochondrial function would produce a type of vicious cycle where insufficient mitochondrial energy production initiates genome instability and mutability, which then promotes further mitochondrial dysfunction and energy impairment, and so on, in a cumulative way. This would ultimately be seen as a gross disturbance in the structural organization of the local tissue and eventually as a carcinoma. An increased dependency on fermentation energy for cell survival would follow each round of metabolic and genetic damage, thus initiating uncontrolled cell growth with the eventual formation of a malignant neoplasm. Hence, the well-documented, tumor-associated abnormalities and genomic instability seen in cancer can arise as a consequence of the progressive impairment of OxPhos.
Mitochondrial Suppression of Tumorigenicity
All other characteristics of cancer arise either directly or indirectly from insufficient respiration. It is also clear that genomic instability and the vast numbers of gene and chromosome defects seen in tumor cells can arise as secondary consequences of protracted respiratory insufficiency. Genome instability is linked to mitochondrial dysfunction through the retrograde signaling system. If all cancer arises from mitochondrial dysfunction, then replacement of damaged mitochondria with normal mitochondria should prevent cancer. In other words, mitochondria producing sufficient respiration should suppress tumor growth regardless of the numbers and types of mutations or aneuploidy present.
Energy derived from substrate level phosphorylation (including the Warburg effect and amino acid fermentation) will persist in the presence of insufficient respiration. Up-regulation of oncogenes and down regulation of tumor suppressor genes is necessary to maintain fermentation when mitochondria fail to produce sufficient energy through respiration. While the mutator phenotype of cancer can be linked to impaired mitochondrial function, substantial evidence also exists showing that normal mitochondrial function suppresses tumorigenesis.
If respiratory insufficiency is the origin of cancer, then tumor nuclei should not induce malignancy when placed in cytoplasm containing respiration competent normal mitochondria. Alternatively, if mitochondrial dysfunction is the origin of cancer, normal nuclei should be unable to prevent tumorigenesis when placed into the tumor cytoplasm.
Normal Mitochondria Suppress Tumorigenesis in Cybrids
Tumorigenicity can be suppressed when cytoplasm from enucleated normal cells is fused with nucleated tumor cells to form cybrids. Cybrids contain a single nucleus and mixtures of cytoplasm from two different cells.
The findings and conclusions of Israel and Schaeffer that cytoplasmic factors suppress tumorigenicity were also strongly supported by the findings of Shay and Werbin. These investigators also discussed the various factors that could influence the success or failure of cybrid experiments designed to uncover cytoplasmic suppressors of tumorigenicity. These factors included, (i) the relative amounts of tumorigenic and nontumorigenic cytoplasm in cybrids; (ii) the time interval that cybrids are passaged prior to testing their tumorigenicity; (iii) whether mutagenesis with carcinogens was used to introduce genetic markers on the cells; and (iv) the specific cell combinations used. They were not surprised that some investigators could obtain varying results if the various factors were not monitored carefully.
The findings from the Jonasson and Harris study were remarkable for several reasons:
Surprisingly, Jonasson and Harris excluded a mitochondrial origin in preference to a centrosome origin for the suppression effect. This decision was based on the findings of others who showed that no human mitochondrial DNA or proteins were found in human mouse cybrids. However, new studies in transmissible cancers show that tumor mitochondria can integrate with normal mitochondria in some tumors.
Paul Saxon and colleagues showed that the microcell transfer of chromosome 11 could suppress tumorigenicity in HeLa cells. They concluded that chromosome 11 contained a tumor suppressor gene. These findings are interesting and also suggest an interaction between chromosome 11 and the mitochondria. It is possible that a gene on chromosome 11 facilitates mitochondrial respiration thus suppressing tumorigenicity in the HeLa cells. It is also interesting that neuroblastoma and Wilms tumor are associated with defects on chromosome 11.
Evidence From rho0 Cells
Singh and coworkers also provided evidence for the role of mitochondria in the suppression of tumorigenicity by showing that exogenous transfer of wild-type mitochondria to cells with depleted mitochondria DNA (rho0 cells) could reverse the altered expression of the APE1 DNA repair protein and the tumorigenic phenotype. The efficiency of APE1-mediated DNA repair is dependent on the sufficiency of mitochondrial respiration. The rho0 cells have deficient respiration because they lack mtDNA, which is necessary for normal respiration. Consequently, transfer of normal mtDNA to rho0 cells will restore respiration, turn off the RTG response, and prevent genomic instability.
The authors concluded that mtDNA mutations play an important role in the etiology of prostate cancer and that cancer can be best defined as a type of mitochondrial disease.
Normal Mitochondria Suppress Tumorigenesis In Vivo
It is also well documented that nuclei from cancer cells can be reprogrammed to form normal tissues when transplanted into normal cytoplasm despite the continued presence of the tumor-associated genomic defects in the cells of the derived tissues.
Herpes viruses have an intimate attachment with mitochondria that causes dysfunctional respiration. Hence, the suppression of tumorigenesis in the Lucke frog tumor is likely due to the replacement of virus-damaged mitochondria with normal mitochondria.
Normal Mouse Cytoplasm Suppresses Tumorigenic Phenotypes
Nuclei from mouse medulloblastoma (a brain tumor thought to arise from cerebellar granule cells) could direct normal development when transplanted into enucleated somatic cells. These investigators concluded that somatic nuclear transfer into normal cytoplasm suppressed the tumorigenic phenotype. Moreover, transplanted medulloblastoma nuclei gave rise to post-implantation embryos that underwent tissue differentiation and early stages of organogenesis. Remarkably, no malignancies were observed in any of the recipient mice, and normal proliferation control was observed in cultured blastocysts.
The findings from the Lucke frog and mouse medulloblastoma experiments were further supported from the work of Konrad Hochedlinger, Rudy Jaenisch and colleagues at MIT. These investigators showed that the nuclei of many cancer cells including pancreatic cancer and melanoma were able to support preimplantation mouse development into normal-appearing blastocysts without signs of abnormal proliferation.
Enhanced Differentiation and Suppressed Tumorigenicity in the Liver Microenvironment
Grisham and colleagues reported that two aneuploid liver tumor cell lines, which formed aggressive tumors when grown subcutaneously, did not form tumors when grown in the liver. The tumors became morphologically differentiated when they were transplanted and grown in the liver. It is likely the normal mitochondria in the fused cell hybrids are responsible for enhanced differentiation and suppressed tumorigenicity.
Respiration is required for the emergence and maintenance of differentiation, while loss of respiration leads to glycolysis, dedifferentiation, and unbridled proliferation.
Summary of Nuclear-Cytoplasmic Transfer Experiments
According to Warburg’s theory, it would be expected that the presence of normal mitochondria in tumor cells would restore the cellular redox status, turn off the mitochondrial stress response, and reduce or eliminate the need for fermentation to maintain viability. In rephrasing, normal mitochondrial function maintains the differentiated state thereby suppressing carcinogenesis, whereas dysfunctional mitochondria can enhance dedifferentiation thereby facilitating carcinogenesis.
(a) Normal cells beget normal cells. (b) Tumor cells beget tumor cells. (c) Delivery of a tumor cell nucleus into a normal cell cytoplasm begets normal cells despite the persistence of tumor-associated genomic abnormalities. (d) Delivery of a normal cell nucleus into a tumor cell cytoplasm begets tumor cells or dead cells, but not normal cells. The results show that nuclear genomic defects alone cannot cause tumors and that normal mitochondria can suppress tumorigenesis.
Growth Signaling Abnormalities and Limitless Replicative Potential
A central concept in linking abnormalities of growth signaling and replicative potential to impaired energy metabolism is in recognizing that proliferation, rather than quiescence, is the default state of both microorganisms and metazoans. The default state of the cell is the condition under which cells are found when they are freed from any active control. Respiring cells in mature organ systems are largely quiescent because their replicative potential is under negative control through the action of normal mitochondrial function. Respiration maintains differentiation and quiescence. In addition, tumor suppressor genes such as p53 and the retinoblastoma protein, pRB, can also help to maintain quiescence.
In addition to facilitating the metabolism of glucose and glutamine through fermentation and substrate-level phosphorylation, MYC and Ras also stimulate cell proliferation. Part of this mechanism also includes inactivation of pRB, the function of which is dependent on mitochondrial activities and the cellular redox state. Disruption of the pRB signaling pathway will contribute to cell proliferation and neoplasia. Cell proliferation is linked to fermentation, whereas quiescence is linked to respiration.
The greater the loss of mitochondrial function, the greater the induction of the RTG response and the greater the longevity (bud production) in yeast. As mitochondrial energy efficiency declines with age, fermentation energy becomes necessary to compensate for insufficient respiration. A reliance on fermentation becomes essential if a cell is to remain alive. A greater dependency on substrate-level phosphorylation will induce oncogene expression and unbridled proliferation, which could underlie in part the enhanced longevity in yeast. When this process occurs in mammalian cells, however, the phenomenon is referred to as neoplasia or new growth.
Linking Telomerase Activity to Cellular Energy and Cancer
Telomerase is a ribonucleoprotein enzyme complex associated with cellular immortality through telomere maintenance. Telomerase is activated in about 90% of human cancers, suggesting a role in tumorigenesis. Evidence suggests that mitochondrial dysfunction could underlie the relocation of telomerase from the mitochondria, where it seems to have a protective role, to the nucleus, where it maintains telomere integrity necessary for limitless replicative potential. Interestingly, telomerase activity is high during early embryonic development when cell proliferation is high, but telomerase activity is low in adult tissues where most cells are differentiated and quiescent.
Telomerase activity is high in normal cells or tumor cells that primarily use fermentation for energy, but the activity is low or nonexistent in nontumorigenic differentiated cells that primarily use OxPhos for energy. These findings suggest that the energy state of the cells dictates the activity level of telomerase. Elevated telomerase activity in tumor cells would therefore be an effect rather than a cause of cancer.
Evasion of Programmed Cell Death (Apoptosis)
Damage to mitochondrial energy production is one type of insult that can trigger the apoptotic cascade, which ultimately involves release of mitochondrial cytochrome c, activation of intracellular caspases, and death. In contrast to normal cells, acquired resistance to apoptosis is a hallmark of most types of cancer cells. The evasion of apoptosis is a predictable physiological response for tumor cells that primarily use fermentation and substrate-level phosphorylation for energy production following respiratory damage during the protracted process of carcinogenesis. Only those cells capable of making the gradual energy transition from respiration to fermentation in response to respiratory insufficiency will be able to evade apoptosis. Cells unable to make this energy transition will die and thus never become tumor cells.
Numerous findings indicate that the genes and signaling pathways needed to upregulate and sustain fermentation are themselves antiapoptotic. For example, sustained glycolysis or glutamine fermentation requires participation of mTOR, MYC, Ras, HIF-1a, and the IGF-1/PI3K/Akt signaling pathways.
Sustained Vascularity (Angiogenesis)
Angiogenesis is required for most tumors to grow beyond an approximate size of 0.2–2.0 mm. This is necessary to provide the tumor with essential energy nutrients, including glucose and glutamine, and to remove toxic tumor waste products such as lactic acid and ammonia.
In addition to its role in upregulating glycolysis in response to hypoxia, HIF-1α is also the main transcription factor for vascular endothelial growth factor (VEGF), which stimulates angiogenesis. HIF-1α is part of the IGF-1/PI3K/Akt signaling pathway that also indirectly influences the expression of βfibroblast growth factor (FGF), another key angiogenesis growth factor.
The sustained vascularity of tumors can be linked mechanistically to the metabolic requirements of fermentation and substrate-level phosphorylation necessary for tumor cell survival.
Metastasis Overview
Metastasis is the term used to describe the spread of cancer cells from the primary tumor to surrounding tissues and to distant organs and is the primary cause of cancer morbidity and mortality. It is estimated that metastasis is responsible for about 90% of cancer deaths. Although systemic metastasis is responsible for 90% of cancer deaths, most research in cancer does not involve metastasis in the in vivo state.
In order to complete the metastatic cascade, cancer cells must detach from the primary tumor, intravasate into the circulatory and lymphatic systems, evade immune attack, extravasate at distant capillary beds, and invade and proliferate in distant organs. Metastatic cells also establish a microenvironment that facilitates angiogenesis and proliferation, resulting in macroscopic, malignant secondary tumors.
The key phenotype of metastasis is that the tumor cells spread naturally from the primary tumor site to secondary locations. Nevertheless, numerous investigators use intravenous tumor cell injection models to study metastasis. While these models can provide information on tumor cell survival in circulation, it is not clear if this information is relevant to survival of naturally metastatic tumor cells.
Cellular Origin of Metastasis
Epithelial to Mesenchymal Transition (EMT):
Stem Cell Origin of Metastasis:
Myeloid Cells as the Origin of Metastasis:
Macrophages and Metastasis
Macrophages are among the most versatile cells of the body with respect to their ability to migrate, to change shape, and to secrete growth factors and cytokines. These macrophage behaviors are also the recognized behaviors of metastatic cells. Macrophages manifest two distinct polarization phenotypes: the classically activated (M1 phenotype) and the alternatively activated (M2 phenotype). Macrophages acquire the M1 phenotype in response to proinflammatory molecules and release inflammatory cytokines, reactive oxygen species, and nitric oxide. In contrast, macrophages acquire the M2 phenotype in response to anti-inflammatory molecules such as IL-4, IL-13, and IL-10 and to apoptotic cells. M2 macrophages promote tissue remodeling and repair but are immunosuppressive and poor antigen presenters. Although the M1 and M2 macrophages play distinct roles during tumor initiation and malignant progression, macrophage–epithelial cell fusions can involve either activation state.
M1 macrophages facilitate the early stages of tumorigenesis through the creation of an inflammatory microenvironment that can produce nuclear and mitochondrial damage. However, TAM can also undergo a phenotypic switch to the M2 phenotype during tumor progression. The TAM population comprising M2 macrophages scavenge cellular debris, promote tumor growth, and enhance angiogenesis. M2 macrophages also fuse with tumor cells and are considered facilitators of metastasis.
Several human metastatic cancers express multiple molecular and behavioral characteristics of macrophages, including phagocytosis, cell–cell fusion, and antigen expression. Tarin also considers the expression of osteopontin (OPN) and CD44 as important in the regulatory gene group/network associated with metastasis. This is interesting as there is strong evidence that both OPN and CD44 are expressed in monocytes and macrophages under various physiological and pathological states.
Macrophages express most hallmarks of metastatic tumor cells when responding to tissue injury or disease. For example, monocytes (derived from hematopoietic bone marrow cells) extravasate from the vasculature and are recruited to the wound via cytokines released from the damaged tissue. Within the wound, monocytes differentiate into alternatively activated macrophages and dendritic cells where they release a variety of proangiogenic molecules, including vascular endothelial growth factor, fibroblast growth factor, and platelet-derived growth factor. M2 macrophages also actively phagocytize dead cells and cellular debris. Occasionally, macrophages undergo homotypic fusion resulting in multinucleated giant cells with increased phagocytic capacity. Following these wound healing activities, macrophages intravasate back into the circulation where they travel to the lymph nodes to participate in the immune response.
Phagocytosis: A Shared Behavior of Macrophages and Metastatic Cells
Phagocytosis involves the engulfment and ingestion of extracellular material and is a specialized behavior of M2 macrophages and other professional phagocytes. This process is essential for maintaining tissue homeostasis by clearing apoptotic cells, cellular debris, and invading pathogens. Like M2 macrophages, many malignant tumor cells are phagocytic both in vitro and in vivo.
Fais and colleagues provided dramatic evidence of tumor cell phagocytosis in showing how malignant melanoma cells eat T-cells. This is remarkable as T-cells are thought to target and kill tumor cells.
There is also evidence that some tumor cells can eat NK cells. If macrophage-derived metastatic cells can eat T-cells and possibly NK cells, then it is possible that immune therapies involving these cells might not be effective for long-term management of some metastatic cancers.
Melanocytes are the resident macrophages of the skin. Expression of cathepsins B and D are elevated in the phagocytic melanoma cells just as they are in malignant melanomas. These tumor cell phagocytic/cannibalistic behaviors are not to be confused with autophagy; a cellular self-digestion process often associated with starvation conditions. Many human cancers and some murine cancers can phagocytize other tumor cells, erythrocytes, leukocytes, platelets, dead cells, as well as extracellular particles. Hence, the characteristics of phagocytosis appear similar in resident skin macrophages and in malignant melanoma.
Phagocytic Cancers:
Metastatic Behavior of the RAW 264.7 Mouse Macrophages:
Fusogenicity: A Shared Behavior of Macrophages and Metastatic Cells:
Fusogenic Cancers:
Myeloid Biomarkers Expressed in Tumor Cells:
Cathepsins, Ezrin, and E-Cadherin:
Anemia and Increased Hepcidin in Metastatic Cancer:
Carcinoma of Unknown Primary Origin
Carcinoma of unknown primary (CUP) origin is a systemic metastatic disease without an identifiable primary tumor and is often associated with poor prognosis. Approximately 5% of all newly diagnosed cancers are classified as CUP. These cancers are often classified as adenocarcinomas, squamous cell carcinomas, poorly differentiated carcinoma, and neuro-endocrine carcinomas. It is thought that these cancers metastasize before the primary tumor has had time to develop into a macroscopic lesion. Signet-ring cells were found in some CUP, indicating that subsets of these cancers exhibit phagocytic behavior like other metastatic cancers. Aneuploidy was identified in 70% of CUP adenocarcinomas but was not found in about 30% of the tumors. Aneuploidy can arise in part from cell fusion events. Survival was better in patients with aneuploid tumors than with diploid tumors, showing that patients with diploid tumors do not have a more favorable prognosis. This is interesting and is consistent with findings that aneuploidy actually slows cell growth. Owing to their high aggressiveness, they suggested that some CUPs could arise from macrophage fusion hybrids.
Many Metastatic Cancers Express Multiple Macrophage Properties
Many phagocytic or fusogenic tumors also express myeloid antigens, further supporting a myeloid origin of these metastatic cancers. It is important to mention that the myeloid properties are expressed in the tumor cells themselves and should not be confused with myeloid properties expressed in TAM, which are also present in the tumors but are not part of the neoplastic cell population.
The macrophage cell fusion explanation of metastasis does not require the induction and reversion of extremely complicated gene regulatory systems.
Linking Metastasis to Mitochondrial Dysfunction
When permanent respiratory damage occurs in cells of myeloid origin, including hematopoietic stem cells and their fusion hybrids, metastasis would be a potential outcome. Numerous studies indicate that mitochondria from a broad range of metastatic cancers are abnormal and incapable of generating energy through normal respiration.
Mitochondrial damage can arise in any cell within the inflammatory microenvironment of the incipient tumor, including TAM, homotypic fusion hybrids of hematopoietic cells, or heterotypic fusion hybrids of macrophages and neoplastic epithelial cells. The end result would be cells with metastatic potential. Although metastatic cells will differ in their morphology from one organ system to the next, they all suffer from insufficient respiration. The origin of metastatic cancer from myeloid cells and fusion hybrids can explain the substantial morphological and genetic diversity seen among different tumor types. It is clear that metastasis can arise in macrophage fusion hybrids that sustain irreversible mitochondrial damage.
What About the Tumor Suppressive Effects of Mitochondria?
Revisiting the “Seed and Soil” Hypothesis of Metastasis
Metastatic tumor cells do not invade distant organs randomly, but invade in a nonrandom pattern with lung, liver, and bone as primary sites of metastases.
Macrophages enter and engraft tissues in a nonrandom manner. Macrophages are genetically programmed to exist in circulation and to preferentially enter various tissues during wound healing and the replacement of resident myeloid cells. Some macrophage populations in the liver are regularly replaced with bone marrow-derived monocytic cells, whereas other macrophage populations are more permanent and require fewer turnovers. It is reasonable to assume that metastatic cancer cells derived from macrophages or fusions of monocytic cells with epithelial cells will also preferentially home to those tissues that naturally require regular replacement of resident macrophages.
Macrophage turnover should be greater in tissues such as liver and lung where the degree of bacterial exposure and the wear and tear on the resident macrophage populations is considerable. This could explain why these organs are a preferred soil of many metastatic cancer cells. Bone marrow should also be a common target of metastatic cells because this site is the origin of the hematopoietic stem cells, which give rise to myeloid cells. Liver, lung, and bone are also preferential sites for metastatic spread for the VM mouse tumor cells.
Because the metastatic cells express insufficient respiration with compensatory fermentation, these cells will enter their default state of proliferation, as would any neoplastic cell. In addition to those organs receiving high macrophage turnover, macrophages also target sites of inflammation and injury. This is interesting in light of findings showing that metastatic cancer cells from lung and breast can appear in the mouth following recent tooth extraction or along needle tracts following biopsy. An unhealed wound is an ideal “soil” for macrophage infiltration. This phenomenon is referred to as inflammatory oncotaxis and can explain in part the seed and soil hypothesis. If metastasis were a metabolic disease of myeloid cells, then the appearance of metastatic cells in recent tooth extraction or wounds would not be unexpected.
Revisiting the Mesenchymal Epithelial Transition (MET)
In contrast to the EMT, the MET involves proliferation and expression of epithelial characteristics following extravasation, invasion, and proliferation at distant cites. The MET is considered a reversibility of the EMT. How is it possible that a series of somewhat random somatic mutations orchestrate the sophisticated series of behaviors associated with the EMT and then have most of these behaviors reversed during the MET? An origin from myeloid cells provides a more credible explanation of metastasis.
Metastatic cells arising from myeloid cell fusions would retain the genetic architecture necessary for entering and exiting the circulation at recognized sites. It is not necessary to construct complicated mutation-based regulatory systems to explain these phenomena. Macrophages naturally enter and exit the circulatory and lymphatic systems. The circulatory system is not a “hostile” environment for cells in the macrophage lineage. These cells also express the cell-surface adhesion molecules (selectins) necessary for extravasation at designated organs. They already express the batteries of metalloproteases necessary for degradation of basement membranes and invasion. When these capabilities occur together with impaired respiration, dysregulated proliferation would be an expected outcome. While these properties certainly implicate myeloid cells as the origin of metastatic cells, the fusogenic properties of myeloid cells can also explain how metastatic cells can recapitulate the epithelial characteristics of the primary tumor at secondary growth sites.
Wong and colleagues showed how macrophage/epithelial cell hybrids could recapitulate phenotypes of epithelial cells, while retaining the properties of macrophages. It is clear that phenotypes of epithelial cells and macrophages can be maintained in fusion hybrids of macrophages and intestinal epithelial tumor cells. Moreover, these characteristics are passed on to daughter cells through somatic inheritance.
Fusions of activated macrophages with epithelial cells in the primary tumor microenvironment will bestow the capability of the fused cells to degrade basement membranes, to enter and exit the circulatory and lymphatic systems, and to recapitulate the epithelial characteristics of the primary tumor at distant secondary sites. The dysregulated growth at secondary sites is the consequence of damaged respiration in these cells. Hence, the origin of metastatic cells from macrophage fusion hybrids with dysfunctional mitochondria can explain the phenomenon of metastasis.
Genetic Heterogeneity in Cancer Metastasis
Considerable genetic heterogeneity is observed by comparing tumor tissue from primary growth sites with tissue from distant metastases. Genetic heterogeneity is seen not only between patients with similar tumor histopathology but also for the tumors growing at different sites within the same patient.
Almost every type of genetic heterogeneity imaginable from point mutations to major genomic rearrangements can be found in metastatic and highly invasive cancers, including those from breast, brain, and pancreas. The mostly nonuniform distribution of mutations in these tumors is consistent with findings that each neoplastic cell within a given tumor can have a profile of changes uniquely different from any other cell within the tumor. Moreover, if the spread of metastatic cells to some organs (such as liver and lung) occurs earlier than spread to other organs, it is possible that genetic heterogeneity would be greater in these organs than in organs that receive metastatic cells later in the disease progression. This is expected if the number of divisions is greater for tumor cells that arrive earlier in these organs than for tumor cells that arrive later in other organs. This could explain why genomic heterogeneity is more diverse in some organs than in other organs or in the primary tumor.
The richness is the likely consequence of damaged respiration in populations of fusion hybrids that differ from each other in genetic architecture. A nonuniform or random distribution of mutations arises from the migration of these hybrid cells to other organs. The driver of the metastatic phenomenon is not a gene but is respiratory insufficiency in macrophages or their fusion hybrids. The gene mutations arise as downstream epiphenomena.
Fusion hybrid hypothesis of cancer cell metastasis. According to their hypothesis, metastatic cancer cells arise following fusion hybridization between neoplastic epithelial cells and myeloid cells (macrophages). Macrophages are known to invade in situ carcinoma as if it were an unhealed wound. This creates a protracted inflammatory microenvironment leading to fusion hybridization between the neoplastic epithelial cell and the macrophage. Fusion hybridization can explain the phenomenon of EMT without invoking new mutations. Inflammation damages mitochondria leading to enhanced fermentation and acidification of the microenvironment. Mitochondrial damage becomes the driver for the neoplastic transformation of the epithelial cell and of the fusion hybrids. As macrophages are already mesenchymal cells that naturally possess the capability to enter (intravasate) and exit (extravasate) the circulation, the neoplastic fusion hybrid will behave as a rogue macrophage. The fusogenic properties of macrophage cells can also explain how metastatic cells can recapitulate the epithelial characteristics of the primary tumor at secondary micrometastatic growth sites. This process can explain the phenomenon of MET without invoking a mutation suppression mechanism.
Transmissible Metastatic Cancers
The best known is the canine transmissible venereal tumors and the Tasmanian devil tumor disease (DFTD). These tumors will often spread from the primary site of contact to distant organs. The metastatic behavior of these transmissible tumors is basically the same as that seen for the nontransmissible human metastatic cancers.
Murchison et al. have recently shown that DFTD originated from cells expressing Schwann cell and epithelial characteristics. It is important to mention that hematopoietic bone marrow cells can elaborate Schwann cell-like phenotypes in injury conditions.
Like all cancers, mitochondrial dysfunction and respiratory insufficiency would be the expected driver phenotype of these transmissible cancers. However, Tasmanian devils living in the western part of the island are resistant to the disease. It appears that the resistance results from a unique DNA polymorphism in the mitochondria of these animals. This is interesting in light of findings showing that transmissible cancers will occasionally acquire mitochondria from the host.
The Absence of Metastases in Crown-Gall Plant Tumors
Crown-gall tumors arise from bacterial infections that enter damaged areas of the plant leading to plant cell proliferation. The mechanisms by which bacteria induce crown-gall disease in plants are similar to those by which viruses induce tumors in animals. Defects in mitochondrial morphology and energy metabolism were later described in crown-gall tumors.
Crown-gall tumors express four of the Hanahan and Weinberg hallmarks of cancer, that is, self-sufficiency in growth signals, insensitivity to growth inhibitory (antigrowth) signals, evasion of programmed cell death (apoptosis), and limitless replicative potential. However, these tumors do not express invasion or metastasis. With the exception of metastasis and invasion, the abnormalities in growth and physiology are similar in crown-gall disease and in animal tumors.
The crown-gall tumors do not metastasize because they do not have macrophages or myeloid cells as part of their immune system. The findings in crown-gall are also consistent with Tarin’s hypothesis, “that metastasis cannot occur until an organism has evolved the genes for lymphocyte trafficking.”
Chapter Summary
Seyfried proposes that the metastatic mesenchymal phenotype arises primarily from respiratory damage in macrophages or in epithelial–macrophage fusion hybrids. Inflammation and radiation damage enhances hybridization, while also damaging mitochondrial function over time. It is my opinion that the myeloid origin of metastasis is the most compelling explanation for the origin of metastasis and tumor progression.
He discussed in Chapters 7 and 8, amino acid fermentation and anaerobic respiration in tumor mitochondria can give the appearance that aerobic respiration is normal when, in fact, it is not. In Chapter 13, he discussed how mitochondrial dysfunction can account for the phenomenon of metastasis in macrophage fusion hybrids and, in Chapters 7 and 8, how amino acid fermentation might simulate OxPhos. This evidence more strongly supports cancer as a metabolic disease than as a genetic disease.
Connecting the Links
Any unspecific condition that damages a cell’s respiratory capacity, but is not severe enough to kill the cell, can potentially initiate the path to a malignant cancer. Reduced respiratory capacity could arise from damage to any mitochondrial protein, lipid, or mtDNA. Some of the many unspecific conditions that can damage a cell’s respiratory capacity thus initiating carcinogenesis include inflammation, carcinogens, radiation (ionizing or ultraviolet), intermittent hypoxia, rare germline mutations, viral infections, and age.
Inflammation has long been recognized in the initiation and promotion of cancer. Inflammation produces ROS and elevates TGF-β, which damages mitochondria while disrupting tissue morphogenetic fields.
Besides producing mutations, carcinogens also produce ROS. Carcinogens, in addition to causing mutations, disrupt OxPhos and cause permanent injury to mitochondria. It is this effect of the carcinogen on mitochondrial energy production rather than its mutagenic effect that primarily initiates cancer. It is unfortunate that the Ames tests focused only on the mutagenic effects of carcinogens rather than on the mitochondrial damaging effects of these compounds.
Radiation not only causes mutations, but also injures mitochondria. Radiation causes necrotic cell death and inflammation. It is the production of ROS and the injurious effect of radiation on OxPhos that causes cancer. While radiation can certainly kill cancer cells, radiation can also initiate cancer through its effect on the mitochondrial energy production.
Similar to inflammation, hypoxia produces high levels of ROS in the microenvironment, which will damage mitochondrial respiratory capacity thus facilitating cancer initiation and progression.
The accumulation of ROS with age damages mitochondrial respiratory energy production. If mitochondrial damage underlies the origin of cancer according to his central hypothesis, then it is predictable that cancer risk should increase with age.
Rare germline mutations increase cancer risk through a direct effect on mitochondrial function. Hence, the plethora of nonspecific factors known to increase the risk of cancer can all be linked to the disease through their protracted and deleterious effects on mitochondrial function, which leads to respiratory insufficiency.
Addressing the Oncogenic Paradox
Chronic injury to the structure and function of mitochondria, which impairs respiration, will activate the mitochondrial RTG response within the damaged cell. The RTG response is an epigenetic system that upregulates those genes needed to derive energy through fermentation. Fermentation involves SLP through glycolysis in the cytoplasm and through amino acid fermentation in the mitochondria. Uncorrected mitochondrial damage will require continuous compensatory energy through fermentation involving SLP in order to maintain the GATP of approximately −56 kJ/mol. This standard energy of ATP hydrolysis is essential for cell viability. This ATP hydrolysis remains mostly constant regardless of whether the ATP is synthesized through respiration or fermentation.
Although fermentation energy can temporally compensate for disruptions to respiration in order to maintain cell viability, persistent energy production through fermentation can compromise cellular differentiation. Tumor cells require energy production through fermentation because their mitochondrial respiration is insufficient to maintain energy homeostasis. If their respiration was sufficient, fermentation would not persist. Confusion arises from amino acid fermentation, which can simulate properties of normal respiration. Cancer cells appear to respire while also fermenting glucose (aerobic glycolysis). Hence, tumor cells differ from normal cells because they generate a significant amount of energy through fermentation.
Tumor progression is linked to irreversible respiratory damage with fermentation becoming the permanent compensatory energy source for the tumor cells. The change in shape of mitochondrial cristae from convoluted- to smooth illustrates the transition from respiration to fermentation. Persistent and cumulative mitochondrial damage underlies the initiation and progression of cancer.
It is only when fermentation compensates for the greater part of total cellular energy production that tumor progression becomes irreversible according to their model. Tumor progression can be reversed, however, as long as some functional mitochondrial respiration remains. Mitochondrial enhancement therapies can help restore impaired respiration.
In addition to the fermentation of glucose to lactate, which is needed to drive glycolysis, many cancer cells might also ferment glutamine in the mitochondria. It is the fermentation of glucose and glutamine that primarily drives tumor progression and makes tumor cells unresponsive to most conventional therapies.
Most of the gene changes associated with tumor progression arise as direct or indirect consequences of insufficient respiration and elevated fermentation. Oncogene upregulation coupled with tumor-suppressor-gene downregulation becomes necessary in order to increase those metabolic pathways needed for fermentation. If the oncogenes needed to drive cellular fermentation are not expressed then the cell will die from energy failure. Oncogene expression is essential to maintain cell viability following respiratory damage.
Succinate produced through mitochondrial glutamine fermentation could be responsible, in part, for stabilization of Hif-1α. Hif-1α is required for maintaining elevated glucose uptake and glycolysis. Respiratory injury becomes the driver for the gene regulatory changes needed for increasing compensatory energy production through fermentation. Insufficient respiration drives oncogene expression, not the reverse.
As DNA repair mechanisms are dependent on the efficiency of respiratory energy production, the continued impairment of respiration will gradually undermine nuclear genome integrity leading to a mutator phenotype and the plethora of somatic mutations identified in tumor cells. Specifically, the integrity of the nuclear genome is dependent on normal cellular respiration. When cellular respiration becomes compromised, genomic instability increases. Activation of oncogenes, inactivation of tumor-suppressor genes, and aneuploidy will be the natural consequences of protracted mitochondrial dysfunction. These gene abnormalities will contribute to accumulative mitochondrial dysfunction while also enhancing those energy pathways needed to upregulate and sustain fermentation energy. The greater the dependency on fermentation and SLP over time, the greater will be the degree of malignancy.
As respiration is necessary for maintaining cellular differentiation, loss of respiration leads to dedifferentiation and a return to the default state of proliferation. Szent-Gyorgyi considered this cellular state as that which existed in the α-period in the history of life on the planet.
The first three cancer hallmarks are consequences of the cell’s return to its mode of existence during the α-period. This would naturally involve increased aerobic glycolysis and resistance to apoptosis. A large number of fermenting cells will also produce an excess of lactate and succinate. This would naturally produce an acidic microenvironment. Angiogenesis is a natural response to wound healing and to the metabolic state in the tumor microenvironment. All of these cancer hallmarks arise as a consequence of insufficient respiration and tumor cell fermentation.
According to the recent commentary of Lazebnik, all hallmarks of cancer with the exception of invasion and metastasis can be found in benign tumors or nonmetastatic cancers. Seyfried also mentioned in Chapter 13 that four of the five hallmarks of cancer are also found in the crown-gall tumors of plants. In contrast to animal cancers, crown-gall tumors do not invade or develop metastases. Hence, it is the hallmark of invasion and metastasis that primarily makes cancer the deadly disease that it is.
As an alternative to the EMT explanation of metastasis, Seyfried showed in Chapter 13 how macrophage fusion hybridization with neoplastic epithelial cells can logically account for all characteristics of the metastatic cascade. Many of the gene expression profiles observed in metastatic cancers are similar to those associated with the function of macrophages or other fusogenic cells of the immune system. Damaged respiration in these fusion hybrids can account for the invasive and metastatic properties found in cancer cells.
Is Cancer Many Diseases or a Singular Disease of Energy Metabolism?
If all cancer cells suffer from respiratory insufficiency, then respiratory insufficiency becomes the central hallmark of the disease. The current view of cancer as a hodgepodge of many diseases is fundamentally inaccurate in view of the central defect of the disease. Cancer appears as many diseases only if viewed from its histological appearance and from its genomic changes. Seyfried considers the histological appearance and gene expression profiles of cancer cells as “red herrings.” When viewed in the light of energy metabolism, cancer is a singular disease of respiratory insufficiency.
Revisiting Growth Advantage of Tumor Cells, Mutations, and Evolution
John Cairns has proposed that carcinogens induce mutations in genes that further enhance mutagenesis. These mutations were considered to produce cells with greater fitness and adaptability than normal cells. According to Cairns’ view, natural selection becomes a liability during cancer progression by selecting dangerous mutations that confer an increased survival advantage on a cell.
According to Seyfried’s view, cancer cells proliferate and survive not because of their genomic instability, but because of their respiratory insufficiency. Respiratory insufficiency enhances fermentation, destabilizes the genome, and causes entry into the default state of unbridled proliferation. Unlike mammalian embryos, which would abort if they expressed the types of mutations found in cancer cells, cancer cells survive and grow with these mutations. The mutations are not lethal and are tolerated because the cancer cells depend more on fermentation than on respiration for energy. Glycolysis-derived pyruvate also enhances the p-glycoprotein activity. The p-glycoprotein is responsible for pumping toxic drugs out of cells, and, when activated, makes tumor cells resistant to most chemotherapy. This is often referred to as the multidrug resistance (MDR)phenotype, which is glycolysis dependent. Hence, this aspect of cancer cell drug resistance arises as a consequence of the glycolytic phenotype.
Aborted development, rather than neoplasia, arises from the epigenetic reprogramming of tumor nuclei. That normal tissues can be derived from the nuclei of cancer cells provides compelling evidence against the notion that somatic mutations are the origin of cancer or drive the disease. There is no conceivable explanation in Darwin’s doctrine that could account for the enhanced survival of organisms that express multiple types and kinds of deleterious mutations.
Cells adapted to ferment glucose and glutamine can survive better in hypoxic environments than cells that require respiration for survival. Adaptation to fermentation is the consequence of protracted damage to respiration during the initiation and progression of cancers. Fermentation energy is primal energy. Fermentation is linked to unbridled proliferation. Somatic mutations do not drive this process, but arise as the result of the process.
Also, somatic mutations and aneuploidy reduce rather than enhance the rate of tumor growth. Support of the inhibitory effects of mutations on tumor growth comes from the well-documented findings that glioma progression is generally slower in patients with chromosome 1p/19q codeletions, promoter hypermethylation of the O6-methylguanine methyltransferase (MGMT) gene, or mutations in the gene for isocitrate dehydrogenase 1 (IDH1).
Work from the Amon and the Compton groups shows that aneuploidy retards cell growth. These findings are also inconsistent with the view that mutations enhance the fitness and growth advantage of tumor cells. The paradox is resolved once cancer becomes recognized as a mitochondrial metabolic disease rather than as a genetic disease.
Normal cells of any tissue follow their genetically scripted program for the differentiated state. Rapid growth is generally not part of this scripted program. If, however, growth becomes necessary for tissue repair, then the normal cells can grow fast. The dysregulated growth of tumor cells arises as a consequence of their abnormal metabolic state involving enhanced fermentation. Fermentation is linked to unbridled proliferation. Unbridled proliferation is the default state of metazoan cells when released from active negative control.
It is now recognized that Ras oncogenes damage the mitochondria to induce cell senescence. The transition from respiration to fermentation allows tumor cells to circumvent the Ras-induced senescence checkpoint. This also allows the tumor cells to survive despite high production of oxidative stress. The origin of Ras-induced oncogenic transformation is damaged mitochondria. Nuclear genome integrity unravels as a result of protracted respiratory dysfunction.
Unbridled proliferation existed during the oxygen-sparse α period of species evolution. This was also a highly reduced state where the ancient pathways of fermentation predominated in driving cell physiology. The appearance of oxygen gave rise to the oxidized state and the emergence of respiration. The emergence of respiration facilitated greater complexity in biological systems.
Respiration largely maintains the differentiated state of metazoan cells. Irreversible damage to respiration, coincident with a rise in fermentation, would unlock the toolkit of preexisting adaptations needed to survive in low oxygen environments.
Tumor Cell Fitness in Light of the Evolutionary Theory of Rick Potts
Potts, a paleoanthropologist at the Smithsonian Institution, has suggested that the evolutionary success of our species has been due largely to the germ line inheritance of traits that have bestowed adaptive versatility. Adaptability was defined in terms of (i) the ability of an organism to persist through major environmental shifts, (ii) to spread to new habitats, and (iii) to respond in novel ways to its surroundings. These characteristics were honed over millions of years and have enabled humans to adapt rapidly to abrupt changes in the physical environment including changes in moisture, temperature, food resources, and so on. The adaptability to abrupt environmental change is a property of the genome, which was selected for in order to ensure survival under environmental extremes.
The success in dealing with environmental stress and disease is therefore dependent on the integrated action of all cells in the organism. Further, this integrated action depends on the flexibility of each cell’s genome, which responds to both internal and external signals according to the needs of the organism. More specifically, only those cells that possess flexibility in nutrient utilization will be able to survive under nutrient stress. Environmental forcing has therefore selected those genomes that are most capable of adapting to change in order to maintain metabolic homeostasis.
According to Darwin and Potts, mutations that bestow a selective advantage are those that will enhance survival under environmental stress. If the multiple pathogenic mutations, chromosomal rearrangements, and mitochondrial abnormalities confer a fitness or survival advantage to tumor cells, then survival under environmental stress should be better in tumor cells than in normal cells. This is not what actually happens when the hypothesis is tested.
Because tumor cells ferment rather than respire, they are dependent on the availability of fermentable fuels (glucose and glutamine). Normal cells shift metabolism from glucose to ketone bodies and fats when placed under energy stress. This is dependent on genomic stability.
Ketone bodies and fats are nonfermentable fuels in mammalian cells. Tumor cells have difficulty in using ketone bodies and fats for fuel when glucose is reduced. Because tumor cells lack genomic stability, they are less able than normal cells to adapt to changes in the metabolic environment. The survival of such cells will be counterproductive for the survival of society and will be eliminated for the good of society.
Cancer cells survive and multiply only in physiological environments that provide fuels necessary for fermentation through substrate-level phosphorylation. If these fuels become restricted, tumor cells will have difficulty in surviving and growing regardless of their complement of genomic changes. Multiple genetic defects will reduce genomic flexibility, thus increasing the likelihood of cell death under environmental stress. Regardless of when or how genomic defects become involved in the initiation or progression of tumors, these defects can be exploited for the destruction or management of the tumor.
Cancer Development and Lamarckian Inheritance
Lamarck’s theory of evolution, involving the use and disuse of organs and the inheritance of acquired characters, can better explain the origin and progression of cancer than Darwin’s views. According to Lamarck, the environment produces changes in biological structures. Through adaptation and differential use, these changes lead to modifications in the structures. The modifications would then be passed on to successive generations as acquired traits.
The degree of use or disuse has shaped biological evolution along with the inheritance of acquired adaptability. Lamarck’s ideas could also accommodate a dominant role for epigenetics and horizontal gene transfer, as factors that could facilitate progression. Epigenetic mechanisms in the form of cell fusion and horizontal gene transfer also contribute to cancer progression and metastasis.
Considering the dynamic behavior of mitochondria involving regular fusions and fissions, abnormalities in mitochondrial structure and function can be rapidly disseminated throughout the cellular mitochondrial network and passed along to daughter cells somatically through cytoplasmic inheritance. The degree of mitochondrial respiratory function becomes progressively less with each cell division as adaptability to fermentation increases.
The most malignant cancer cells would sustain the near-complete replacement of their respiration with fermentation. This process could be viewed as Lamarckian inheritance. Although Lamarck considered the inheritance of acquired characteristics as enhancing biological complexity and perfection, the opposite effect would occur in adapting his evolutionary concepts to cancer progression. More specifically, a Lamarckian view can account for the escalation situation of biological chaos and the nonuniform accumulation of mutations seen during cancer progression.
Can Teleology Explain Cancer?
Teleology involves design with a purpose and is the cornerstone of arguments involving intelligent design or creationism. A teleological explanation for complicated phenomena assumes that the system examined has an intended purpose and was designed to accomplish that purpose. Evolution operates without purpose, but rather by genetic chance and environmental necessity.
Cybernetic-type diagrams, which are sometimes used to describe the complexity of cancer signaling systems, can also be considered as teleological explanations. Although teleological explanations might appear appealing on the surface, they muddle mechanistic explanations for the phenomena under investigation. Intentions and purpose are not assumed in mechanistic explanations. Descriptions of cancer cells as being motivated, choosing to do something, or having an agenda are examples of teleology. It is unlikely that teleological explanations will provide much insight into the origin or progression of cancer.
Current Status of Cancer Treatment
At present, surgery, chemo-, and radiation therapy are the standard procedures used for treating most malignant cancers. While these therapies can certainly provide long-term management of benign or nonmetastatic tumors, they have been less effective in providing long-term control of many advanced metastatic cancers.
Not only is the cure rate poor for most malignant metastatic cancers, but many current therapies can actually exacerbate the disease. Chemo- and radiation therapy sicken and weaken patients, thus increasing their susceptibility to infections and diseases. Although these procedures could resolve the disease over a short term (months to years), they can enhance systemic physiological disorder over the long term. Enhanced entropy accelerates the aging process thus reducing longevity. It is not clear how many cancer patients die from their disease or die from the toxic effects of the therapies used to treat their disease.
The US Food and Drug Administration (FDA) has recently approved the immunotherapy drug ipilimumab, “ipi”, for treatment of malignant melanoma. The adverse effects of ipilimumab can include severe diarrhea, colitis (colon inflammation), and endocrine disruption. These adverse effects are generally treated with steroids. Indeed, the steroid drug, dexamethasone, is widely used to suppress nausea and vomiting in many cancer patients who receive toxic chemotherapy. Steroids significantly elevate blood glucose levels thus enhancing tumor cell survival and drug resistance. While only 3 out of 540 persons who received ipilimumab became cancer free, 14 persons died from the drug treatment.
The BRAF kinase inhibitor vemurafenib received approval from the FDA for treatment of those melanoma patients whose tumors contain the V600E mutation in the V-RAF murine sarcoma viral homolog B1 (BRAF) oncogene.
Any cancer immunotherapy that also produces chills and fever in patients could be effective in causing tumor regression. Fever places global stress on the body, which will indirectly target energy metabolism. Cancer cells are less adaptable to hyperthermia energy stress than normal cells. William Coley long ago showed that cancer regression could follow vaccine-induced fever. It will therefore be important to determine if the therapeutic action of expensive new immune therapies arises through a gene-mediated mechanism or through the simple induction of fever.
A recent study showed that the adverse effects of rash and diarrhea were correlated with very modest increase in survival of brain cancer patients treated with Gefitinib, a small molecule inhibitor of the epidermal growth factor receptor (EGFR) tyrosine kinase. Without appropriate control groups for rash and diarrhea, it is difficult to interpret such findings. In other words, was the modest increase in survival due to the effects of Gefitinib or was it due to the effects of rash and diarrhea? As many cancer therapies are toxic to cells and tissues, toxicity has become the norm rather than the exception for new cancer therapies.
The BATTLE therapy is considered to be personalized because it uses a cocktail of drugs that target various tumor cell growth factor receptors such as the EGFR and other tumor cell molecular defects. In other words, the BATTLE therapeutic strategy is structured on the view that cancer is a genetic disease. However, we know from the work of Loeb, Stratton, and others that the molecular abnormalities found in tumors are likely to differ from one tumor cell to the next within most malignant neoplasms.
As metastatic melanoma and most metastatic cancers arise from macrophages with defective energy metabolism, it is my opinion that targeting glucose and glutamine under energy restriction will be a more effective long-term therapy than any of the current drugs used to treat these cancers.
The “Standard of Care” for Glioblastoma Management
In contrast to other metastatic cancers, GBM usually kills patients before the disease shows systemic metastasis. As most metastatic cancers, including GBM, arise from respiratory damage in cells of myeloid origin, the problems encountered in treatment strategies will be common to all or most metastatic cancers.
As for many malignant cancers, the current standard of care for GBM includes maximum surgical resection, radiation therapy, and chemotherapy. Almost 99% of GBM patients receive perioperative corticosteroids (dexamethasone) as part of the standard of care, which is sometimes extended throughout the course of the disease.
GBM is notoriously heterogeneous in cellular composition consisting of tumor stem cells, mesenchymal cells, and host stromal cells. The number of cells with characteristics of macrophages/monocytes in GBM can sometimes equal the number of tumor cells. These cells are sometimes referred to as tumor-associated macrophages (TAMs), but their origin from either stromal cells or neoplastic cells remains ambiguous. They recently suggested that many cells that appear as TAM are actually part of the neoplastic cell population. TAMs contribute to tumor progression through the release of proinflammatory and proangiogenic factors. Many of the neoplastic cells in GBM are highly migratory and invade the brain well beyond the main tumor mass making complete surgical resections exceedingly rare. Despite the best available treatments, only about 5–10% of GBM patients become long-term survivors (36 months).
Glucose and glutamine are the prime fuels for driving the growth of most malignant cancers. Glucose is necessary for nearly all brain functions under normal physiological conditions. Tumor cells metabolize glutamine to glutamate, which is then metabolized to α-ketoglutarate for further metabolism within the mitochondria. In contrast to extracranial tissues, where glutamine is the most available amino acid, glutamine is tightly regulated in the brain through its involvement in the glutamate–glutamine cycle of neurotransmission.
Glutamate is a major excitatory neurotransmitter that must be cleared rapidly following synaptic release in order to prevent excitotoxic damage to neurons. Glial cells possess transporters for the clearance of extracellular glutamate, which is then metabolized to glutamine for delivery back to neurons. Neurons metabolize the glutamine to glutamate, which is then repackaged into synaptic vesicles for future release. The glutamine–glutamate cycle maintains low extracellular levels of both glutamate and glutamine in normal neural parenchyma. Disruption of the cycle can provide neoplastic GBM cells access to glutamine. Besides serving as a metabolic fuel for neoplastic tumor cells, glutamine is also an important fuel for cells of myeloid origin, that is, macrophages, monocytes, and microglia. As long as GBM cells have access to glucose and glutamine, the tumor will grow, making long-term management difficult or impossible.
In contrast to normal glia, some neoplastic glioma cells secrete glutamate. Glioma glutamate secretion is thought to contribute, in part, to neuronal excitotoxicity and tumor expansion. Neurotoxicity from mechanical trauma (surgery), radiotherapy, and chemotherapy will increase extracellular glutamate levels thus contributing to tumor progression.
Radiation and chemotherapies induce necrosis and inflammation, both of which will increase tissue glutamate levels. Local astrocytes rapidly clear extracellular glutamate metabolizing it to glutamine for release to neurons. In the presence of dead or dying neurons, surviving tumor cells and TAM will use astrocyte-derived glutamine for their energy and growth.
Radiation damage to tumor cell mitochondria will hasten a dependence on glucose and glutamine for growth and survival of neoplastic cells.
High dose glucocorticoids (dexamethasone) are generally prescribed to reduce radiation-associated brain swelling and tumor edema. It is well documented, however, that dexamethasone significantly elevates blood glucose levels.
TAMs will respond to the local tumor environment as if it were an unhealed wound thus releasing proangiogenic growth factors. What develops then is an escalating situation of biological chaos, where the intrinsic properties of TAM to heal wounds enhance the capacity of brain tumor cells to proliferate, invade, and self-renew. High glucose levels together with unrestricted glutamine availability will provide the energy necessary for driving the escalating situation within the tumor.
The current standard of care for GBM creates the “perfect storm” of adverse effects that guarantees the demise of most patients.
As cells with properties of macrophages are naturally programmed to ferment glucose and glutamine, it is possible that bevacizumab, cediranib, and other antiangiogenic drug therapies select for those cells that are invasive and can survive in hypoxic microenvironments. Such cells would become less dependent on a supporting vasculature for survival. These tumor cell capabilities would render bevacizumab and other similar antiangiogenic drugs fruitless as effective treatments for GBM management.
While the current standard of care for GBM can increase patient survival over a shorter term (months), this therapeutic program will ultimately accelerate GBM energy metabolism and progression. Any therapy that enhances tumor cell energy metabolism runs the risk of reducing long-term patient survival.
DER is produced from a total reduction in dietary nutrients and differs from starvation in that DER reduces total calorie energy intake without causing anorexia or malnutrition. As a natural therapy, DER improves health, prevents tumor formation, and reduces inflammation. Reduced calorie intake is ideally suited as a therapy for reducing tumor growth without the adverse effects associated with conventional cancer therapies. Indeed, fasting can reduce the toxic effects of some chemotherapies.
Is it Dietary Content or Dietary Composition That Primarily Reduces Tumor Growth?
Dietary Energy Reduction and Therapeutic Fasting in Rodents and Humans
The DER-induced inhibition of brain tumor growth in mice is directly correlated with reduced levels of glucose and elevated levels of ketone bodies. Ketone bodies [β-hydroxybutyrate (β-OHB) and acetoacetate] become an alternative fuel for tissue energy metabolism when glucose levels are reduced as would occur during consumption of very low-calorie diets or water-only fasting.
Acetone is also a by-product of ketone synthesis, but acetone is not used for energy and is released in the breath or urine. β-OHB is the major circulating ketone body and is used primarily for energy metabolism when glucose levels become reduced. Although β-OHB is metabolized to acetoacetate in most tissues except liver, tissue uptake from the circulation is faster for β-OHB than for acetoacetate. A greater number of surface receptors for β-OHB than for acetoacetate might account for the more rapid uptake of β-OHB.
Ketogenic Diets
KDs were developed originally to manage epileptic seizures in children, but they are also effective in managing brain cancers, especially when administered in reduced amounts to reduce glucose levels. KD consumption can also lower blood glucose levels in some people. This is usually due to self-restriction because of diet unpalatability. Additionally, the high fat composition of the KD can reduce overall consumption through an effect on expression the cholecystokinin peptide. Intestinal cells release cholecystokinin in response to high fat diets. Cholecystokinin activates vagal sensory neurons to inhibit feeding behavior.
The efficacy of the KetoCal KD is optimal for brain cancer management when the ratio of dietary fats to combined carbohydrate/protein is 4:1.
The KD reduced growth and vascularization in mouse astrocytoma, but only when the diet was administered in reduced amounts that also lowered body weights. The KD had no therapeutic efficacy against tumor growth when consumed ad libitum (AL) or in unrestricted amounts.
When the KD is fed to mice in unrestricted amounts, blood glucose levels remain high and ketones are largely excreted in the urine. They clearly showed, however, that blood ketones were higher in tumor-bearing mice under DER than under AL feeding. Under DER, ketones are retained in the body for use in metabolism rather than excreted in the urine.
It is also important to mention that Dr. Adrienne Scheck and colleagues reported growth inhibition of the mouse GL261 glioma cells from a KD fed to mice in unrestricted amounts. These findings suggest that some tumors might be susceptible to KD growth inhibition without CR or glucose reduction. Drs Scheck and Mohammed Abdelwahab also reported at the 2011 meeting of the American Association of Cancer Research (AACR) that the KD can improve survival in mice receiving radiation therapy for brain cancer. However, the anticarcinogenic effects of the KD will probably be best when the diet is consumed in lower amounts rather than in higher amounts, as the unrestricted consumption of the KD can produce adverse events due to the high fat content of the diet.
Ketones are retained in the body when glucose levels are low. Ketones serve as an energy substitute for glucose. If glucose is not reduced as in the KC-UR groups, then most ketones will be excreted in the urine. This is why it is better to measure blood ketone levels than to measure urine ketones as an indicator of ketosis. Cancer cells are placed under metabolic stress when glucose levels are reduced and ketone levels are elevated. The therapeutic action of ketones is best when blood glucose levels are low.
DER, which lowers glucose and elevates ketone bodies, improves mitochondrial respiratory function and glutathione redox state in normal cells. Glutathione plays an important role in protecting cells and tissues from oxygen radical damage. Reactive oxygen species (ROS), which are elevated in many cancer cells, can damage DNA, lipids, and proteins. Ketone bodies can also protect normal cells from damage associated with aggressive tumor growth through a variety of neuroprotective mechanisms, including elevated glutathione levels. Hence, the natural elevation of blood ketones accompanying reduced food intake or consumption of KDs can reduce the pathological effects of a broad range of diseases, especially cancer. Ketone body metabolism also reduces inflammation.
Glucagon and Insulin
Glucagon is responsible for the elevation of ketone bodies in the blood. Glucagon becomes elevated during food restriction. Besides stimulating fat breakdown, glucagon also stimulates the synthesis of glucose from stored proteins and fats in order to maintain a basal level of glucose in blood. The glycerol part of the triglycerides is used for glucose synthesis. A basal glucose level is also needed to maintain brain function. However, ketone bodies will gradually replace glucose as the major fuel for the brain and other tissues. Ketone bodies allow the brain to maintain normal function under hypoglycemia. In addition to ketone bodies, fatty acids released from stored triglycerides (body fat) will become a predominant fuel for most tissues except the brain, which primarily burns ketone bodies for energy under low glucose conditions.
Fatty acid metabolism can generate heat, which can interfere with normal brain function. The brain does not dissipate heat as well as other organs due to skull confinement. More heat is generated from metabolism of fatty acids than from ketones, as fatty acid metabolism can increase expression of uncoupling proteins.
Ketone metabolism is, therefore, more energetically efficient than fatty acid metabolism since less heat is released from ketone body metabolism than from fatty acid metabolism. The use of fatty acids and ketone bodies as metabolic fuel arises in large part from the inability of the liver and kidneys to synthesize enough glucose (gluconeogenesis) to maintain metabolic homeostasis under periods of prolonged food restriction.
As insulin stimulates glycolysis, insulin can stimulate the growth of those tumors that depend on glucose and glycolysis. Blood insulin levels become low in the absence of food intake because blood glucose levels become low. Hence, insulin and glucagon regulate metabolic homeostasis in the presence and absence of food, respectively.
Basal Metabolic Rate
The basal metabolic rate (BMR) is the energy required for maintaining body temperature, blood circulation, cellular respiration, and glandular activities under conditions of rest. It is important to recognize that the physiological response to DER in rodents is not the same as in humans due to differences in BMR. The BMR is about sevenfold greater in the mouse than in the human. Consequently, the ability to maintain metabolic homeostasis under food restriction is far greater in humans than in rodents. The health benefits documented in mice under 40% dietary restriction (DR) can be realized in humans under very low-calorie intake (400–500 kcal) or with water-only therapeutic fasting.
Alternatively, these health benefits can also be achieved using the restricted KD, which increases circulating levels of ketone bodies, while maintaining low blood glucose levels. The KD-R can replace a therapeutic fast for cancer management.
Additionally, recent studies by Kashiwaya, Veech, and coworkers show that diets supplemented with ketone esters could also be effective in reducing blood glucose and glutamine, while elevating ketone levels.
Dominic D’Augostino is also evaluating new formulations of ketone esters that might eventually replace some aspects of the KD-R as cancer therapy.
Ketones and Glucose
Only those cells with normal mitochondrial respiratory capacity can effectively use ketone bodies for energy, as ketones cannot be effectively metabolized for energy without an intact electron transport chain or with the mitochondrial enzymes needed to metabolize ketones. They found that ketone bodies are unable to maintain tumor cell viability in the absence of either glucose or glutamine. The inability of ketone bodies alone to maintain tumor cell viability has also been found in human glioma cells. Moreover, ketone bodies can actually be toxic to some tumor cells even in the presence of glucose, for example, neuroblastoma. Many human and mouse tumors also express deficiencies in the key enzymes needed to process β-OHB to acetyl CoA. More specifically, many tumor cells cannot effectively use ketone bodies to fuel their growth or maintain their survival.
Metabolic Management of Brain Cancer Using the KD
A mildly restricted KD, which lowers glucose and elevates ketone bodies, could reduce glycolytic energy metabolism in brain tumors. More recently they published a case report showing that a KD and therapeutic fasting could also help manage glioblastoma growth in an older female patient. KD can improve the quality of life of some cancer patients. It is his opinion that the therapeutic effects would be even greater if the KD were administered in restricted amounts with drugs that target glucose and glutamine.
Glucose Accelerates Tumor Growth
Glucose fuels tumor cell glycolysis and provides precursors for the pentose phosphate shunt as well as for glutamate synthesis.
As glucose levels fall, tumor size (weight) and growth rate falls. As mentioned earlier, hyperglycemia is directly linked to poor prognosis in humans with malignant brain cancer and is connected to the rapid growth of most malignant cancers.
Glucose Regulates Blood Levels of Insulin and Insulin-Like Growth Factor 1
IGF-1 is a cell surface receptor linked to rapid tumor growth. The association of elevated plasma IGF-1 levels with rapid tumor growth rate is due primarily to high circulating glucose levels. Just as DER lowers insulin levels, DER also lowers IGF-1 levels. This happens because DER lowers blood glucose levels. Glucose drives tumor cell energy metabolism, while IGF-1 drives tumor cell growth through the IGF-1/PI3K/Akt/hypoxia-inducible factor-1α (HIF- 1α) signaling pathway.
The transition from glucose to ketone bodies requires multiple changes in gene expression and metabolic adjustments. These adjustments readily occur in normal cells of the body, as the transition from glucose to ketones is an evolutionarily conserved adaptation to food restriction. Genes for glucose metabolism and glycolysis are downregulated, while genes for respiration are upregulated. Insufficient respiration and genomic instability will prevent adaptive versatility of tumor cells, thus contributing to their elimination.
As most tumor cells require increased glycolysis for growth and survival, a transition from glucose energy to ketone energy places considerable metabolic stress on tumor cells. Tumor cells are metabolically challenged compared to normal cells and cannot effectively use ketone bodies for energy. Treatments that reduce glucose while elevating ketones will place more stress on tumor cells than on normal cells. Hence, an energy transition from glucose to ketone bodies becomes a rational therapeutic strategy to tumor management that enhances the metabolic efficiency of normal cells, while targeting the metabolically challenged tumor cells.
Dietary Energy Reduction is Antiangiogenic
The challenge is to target tumor angiogenesis without harming patients or reducing the quality of life. Payton Rous first suggested that the restriction of food intake inhibited tumor growth by delaying tumor vascularity from the host. Angiogenesis involves neovascularization or the formation of new capillaries from existing blood vessels and is associated with the processes of tissue inflammation, wound healing, and tumorigenesis.
Biomarkers for angiogenesis, including IGF-1 and vascular endothelial growth factor (VEGF), were significantly lower in all tumors when grown under DER than when grown under unrestricted or AL conditions. DER also reduces angiogenesis in prostate and mammary cancer. DER of KDs is also antiangiogenic, indicating that the antiangiogenic effects are related to the amount rather than the composition of the diet.
Compared to bevacizumab (Avastin), which targets angiogenesis, while producing adverse effects and enhancing tumor cell invasion, DER targets angiogenesis, while improving general health and inhibiting tumor cell invasion.
Dietary Energy Reduction Targets Abnormal Tumor Vessels
It is important to mention that blood vessel structure and function are different in the tumor microenvironment than in normal microenvironment. Puchowicz et al. showed that diet-induced ketosis increases capillary density in normal rat brain. In contrast to normal tissue vasculature, tumor blood vessels express leakiness and immaturity (absence of a pericyte smooth muscle sheath). DER reduces the abnormal vasculature in tumors. Ivan Urits, recently found that DER enhances expression of α-smooth muscle actin (α-SMA) in the tumor vasculature. A restoration of blood vessel integrity should reduce local inflammation arising from vessel leakiness. Hence, DER has a marked effect on the structure and function of tumor blood vessels that reduces tumor growth.
KD-R enhances brain delivery of NB-DNJ (N-butyldeoxynojirimycin), a small imino sugar molecule that inhibits ganglioside biosynthesis. Hence, DER or restricted KDs target abnormal tumor blood vessels, while enhancing formation of normal vasculature.
Dietary Energy Reduction is Proapoptotic
The DER-induced reduction in brain tumor growth was also associated with significant elevation in TUNEL-positive cells (apoptosis). TUNEL is an acronym for “terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate biotin nick end labeling.” DNA begins to disintegrate in a specific way in cells that die from programmed cell death or apoptosis.
Apoptotic cell death differs from necrotic cell death, which is usually associated with inflammation. Apoptotic tumor cell death would therefore be less provocative to the tumor microenvironment than would necrotic cell death, as tissue inflammation is less during apoptosis than during necrosis. This is important since the current standard of care for many cancers often involve radiation therapy together with toxic chemotherapy that causes inflammation and necrotic tumor cell death.
Phosphorylation and inactivation of BAD (BCL2 agonist of cell death) and procaspase-9 mediate, in part, the antiapoptotic actions of Akt (protein kinase B) activation. BAD transmits proapoptotic signals generated during glucose deprivation. Jeremy Marsh and Purna Mukherjee found that BAD was constitutively phosphorylated in mouse astrocytoma compared with contralateral normal brain and showed that DER suppressed BAD phosphorylation and increased procaspase-9/-3 cleavage. BAD stimulates apoptosis by forming heterodimers with and by inactivating the antiapoptotic proteins Bcl-2 and Bcl-xL. DER is known to reduce Bcl-2 and Bcl-xL expression and to increase the expression of Bax, Apaf-1, caspase-9, and caspase-3 in experimental carcinomas.
Their studies suggest that DER inhibits tumor growth by inducing mitochondrial-dependent apoptosis mediated by the dephosphorylation of BAD. These findings are consistent with evidence that DER is proapoptotic in malignant astrocytomas and support evidence that BAD coordinates glucose/IGF-1 homeostasis and the induction of apoptosis. They also showed that reduced glucose availability and IGF-1 expression play a key role in suppressing Akt and in mediating the proapoptotic effects of DER in the CT-2A mouse astrocytoma that is PTEN (phosphatase and tensin homologue)/TSC2-(tuberous sclerosis complex 2) deficient.
The upregulation of metabolic pathways, involving c-Myc, Hif-1α, and so on, will inhibit apoptosis. If energy from glycolysis is reduced, then many tumor cells will die or growth arrest from catastrophic energy failure. Tumor cells have difficulty growing once their access to glucose and glutamine becomes limited.
Dietary Energy Reduction is Anti-Inflammatory
Inflammation not only initiates tumorigenesis but also drives tumor progression. Inflammation damages OxPhos, which is the origin of many cancers. Nuclear factor kappa B (NF-κB) is a transcription factor largely responsible for enhancing tissue inflammation. Phosphorylation and activation of NF-κB results in the transactivation of many genes, including those encoding cyclooxygenase-2 (COX-2) and allograft inflammatory factor-1 (AIF-1), both of which are primarily expressed by inflammatory and malignant cancer cells within the tumor microenvironment. Activated NF-κB translocates to the nucleus, binds to DNA, and then activates a number of proinflammatory molecules, including COX-2, tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, IL-8, and Matrix metallopeptidase 9 (MMP-9). COX-2 enhances inflammation and promotes tumor cell survival.
Purna Mukherjee and former undergraduate student, Tiernan Mulrooney, found that the p65 subunit of NF-κB was expressed constitutively in mouse astrocytoma compared with contralateral normal brain tissue. NF-κB also activates mitochondrial glutaminase, which hydrolyzes glutamine to glutamate. Glutamate is used as an energy metabolite for tumor growth and, when secreted, can enhance tumor progression. Inhibition of NF-κB activation would reduce rapid tumor growth and progression in part through inhibition of glutamine metabolism. As NF-κB-mediated inflammation is common to most malignant cancers, any therapy that reduces expression of NF-κB should be effective in managing cancer.
Targeting Energy Metabolism in Advanced Cancer
Although many studies showed that DER could reduce tumor progression when initiated soon after tumor implantation, fewer studies have evaluated the influence of DER when tumors are already advanced and are heavily inflamed and vascularized.
Payton Rous showed that underfeeding could slow advanced subcutaneous adenocarcinoma implants in mice. Seyfried showed that late-onset DER (i.e., DER initiated 10 days after tumor implantation rather than only 2–3 days after) could reduce the growth of large tumors. Late-onset DER also delayed malignant progression and significantly extended mouse survival. These effects were associated with changes in metabolic biomarkers, including blood glucose and lactate levels. They showed that expression of these biomarkers was linked to the downregulation of the IGF-I/Akt/Hif-1α signaling pathway.
DER-induced reduction in glycolysis is associated with declines in both circulating glucose and lactate levels as well as in the expression of HIF-1α and the type 1 glucose transporter (GLUT1). These reductions were also associated with a reduction in signaling through the IGF-I/Akt pathway. Reduced glycolytic energy could increase ROS-related cell death in tumor cells, while reducing ROS levels in normal cells. Normal cells switch to ketone bodies for energy under low glucose. Ketone metabolism reduces ROS production in normal cells and is neuroprotective. DER reduces availability of a prime fuel (glucose) needed for tumor metabolism, while elevating availability of the prime fuel (ketones) needed to maintain energy homeostasis in normal cells during energy stress.
Differential Response of Normal Cells and Tumor Cells to Energy Stress
GLUT1 expression is significantly higher in astrocytoma cells than in normal brain cells under AL feeding conditions. Unrestricted food availability maintains high blood glucose, which enhances tumor growth. GLUT1 expression is higher in astrocytoma under AL feeding conditions than under DER. DER downregulates GLUT1 expression in tumor tissue. In contrast to tumor tissue, DER upregulates GLUT1 expression in normal tissue. These findings show that GLUT1 expression behaves in opposite ways to glucose availability in normal cells and tumor cells.
By upregulating GLUT1 expression during DER, normal cells are better able than tumor cells to acquire available glucose. This, coupled with their ability to metabolize ketones, would make normal cells more fit than tumor cells under energy stress. Normal cells have evolved to survive and maintain energy homeostasis under conditions of energy stress. Respiratory insufficiency and genomic instability make tumor cells less fit than normal cells. Insufficient respiration makes tumor cells dependent on fermentation energy for survival and growth. Reduced glucose availability targets glycolysis.
Reduction in IGF-1 expression can be lethal to glycolysis-dependent tumor cells but not harmful to normal cells. Recent studies show that dietary energy restriction enhances phosphorylation of adenosine monophosphate kinase (AMPK), which induces apoptosis in glycolytic-dependent astrocytoma cells but protects normal brain cells from death.
Dietary Energy Reduction is Anti-invasive in Experimental Glioblastoma
The total percentage of proliferating tumor cells (Ki-67-positive cells) within the primary tumor and the total number of blood vessels was also significantly lower in the DER-treated mice than in the mice fed AL, indicating that reduced caloric intake is also antiproliferative and antiangiogenic in this tumor. These findings indicate that DER can inhibit proliferation and invasion of tumor cells throughout much of the brain. Considering that invasion is primarily responsible for patient death, he suggests that survival will be increased in those patients who would use DER therapies to treat their brain tumors.
His findings with DER therapy in the invasive glioblastoma model contrast with those in patients treated with bevacizumab. Bevacizumab appears to enhance glioma invasion without reducing Ki-67-positive tumor cells. His findings in mice suggest that DER could be a more effective antiangiogenic therapy than bevacizumab for brain cancer management in humans. Moreover, the therapeutic efficacy of DER was not associated with diarrhea or other adverse effects, as occurs with the potent epidermal growth factor receptor (EGFR) inhibitor, gefitinib. In contrast, DER enhances general health.
Findings indicate that the anti-invasive properties of DER can be due in large part to reductions in tumor cell proliferation, glycolysis, inflammation, and angiogenic factors in both the tumor cells and in the tumor microenvironment.
Influence of Growth Site and Host on Tumor Progression
DER significantly reduces the growth of a PTEN-deficient malignant mouse astrocytoma and the human U87-MG glioma, which have PI3K activation. DER also reduces the growth of the mouse ependymal-cell brain tumor (EPEN). They have not yet found a brain tumor that is resistant to the growth inhibitory effects of DER when implanted in the orthotopic site.
Implications of Dietary Energy Reduction or Anticancer Therapeutics
Dr. Epstein mentioned that loss or distorted taste (dysgeusia), muscle cramps, and weight loss were common side effects in patients treated with the new hedgehog inhibitor, GDC-449, for nonmelanoma skin cancer. 20% of treated patients discontinued GDC-0449 because of the side effects. Termination of treatment was associated with weight gain and tumor recurrence. Since no controls for dysgeusia, muscle cramps, and weight loss were included in the study design, it is not clear if the therapeutic effects observed were due to GDC-449 or to the side effects of the drug.
As some drugs may reduce food assimilation, active body weight controls must be evaluated together with pair-fed controls. Unfortunately, many scientific reports of new cancer drugs or therapies fail to include all the necessary control groups needed to distinguish specific from nonspecific effects.
Targeting Glucose
Dietary energy restriction specifically targets the IGF-1/PI3K/Akt/HIF-1α signaling pathway, which underlies several cancer hallmarks to include cell proliferation, evasion of apoptosis, and angiogenesis. DER also causes a simultaneous downregulation of multiple genes and metabolic pathways regulating glycolysis. In addition, subsets of tumors have inherited or acquired mutations in the TCA cycle genes. Such mutations are expected to limit the function of the TCA cycle, thus increasing the glycolytic dependence of these tumors. Tumors with these types of mutations could be especially vulnerable to management through DER. Hence, DER can be considered a broad-spectrum, nontoxic metabolic therapy that inhibits multiple signaling pathways required for progression of malignant tumors regardless of tissue origin.
In addition to DER, several small molecules that target aerobic glycolysis or tumor cell energy metabolism are under consideration as novel tumor therapeutics:
Toxicity can become an issue as some of these compounds target pathways other than glycolysis or nucleotide synthesis and high dosages are sometimes required to achieve therapeutic efficacy in vivo. A reformulation of resveratrol was discontinued as a therapy for multiple myeloma because some patients developed kidney failure. This is unfortunate, as resveratrol itself has many health benefits that can improve longevity, reduce inflammation, and help lower blood glucose.
Brain damage could arise if glucose is targeted without using ketones as a compensatory fuel. It should be possible to effectively target and kill tumor cells using a combination of CR mimetics and the restricted KD.
Metformin
Metformin lowers blood glucose levels by targeting class IIa histone deacetylases in liver. Because of its ability to lower blood glucose levels, metformin has also been considered for treating glucose availability to tumor cells.
In addition to inhibiting gluconeogenesis, metformin also acts like insulin in facilitating glucose uptake into cells. Glucose transporters are upregulated in tumor cells. One side effect of metformin is lactic acidosis. Lactate is produced from glycolysis. Glycolysis drives tumor growth. It is therefore possible that metformin could enhance tumor cell glycolysis in some cancer patients.
Claffey and coworkers showed that metformin had only a marginal effect on the growth of primary breast cancer (triple-negative 66cl4 tumor cells from Balb/c mice), but had no significant effect on the metastatic spread of the tumor cells from fat pads to lung.
While metformin can be effective in reducing very high blood glucose levels in diabetic and obese patients, it’s uncertain if metformin will be effective against brain tumor growth in patients.
In contrast to metformin, somatostatin might be more effective in lowering blood glucose levels for cancer patients without producing toxicity. Somatostatin targets glucagon naturally to lower blood glucose levels. Further studies are needed to assess the therapeutic effects of somatostatin, especially when combined with the KD-R.
Synergistic Interaction of the Restricted Ketogenic Diet (KD-R) and 2-Deoxyglucose (2-DG)
Although DER is effective in reducing tumor growth and invasion, this therapeutic approach alone is unlikely to completely eradicate all types of malignant cancers.
He thinks 3-BP and possibly dichloroacetate could be even more effective cancer therapies if combined with if combined with the KD-R. Sometimes novel therapies can be effective without being toxic or expensive.
On the basis of his findings and those from the Longo group, it is clear that therapeutic fasting and DER can enhance the antitumor effects of chemotherapy and help patients tolerate the adverse effects of chemotherapy. The administration of antiglycolytic drugs together with energy-restricted diets could act as a powerful double “metabolic punch” for the rapid killing of glycolysis-dependent tumor cells. Combinations of CR mimetics with the restricted KD could open new avenues in cancer drug development, as many drugs that might have minimal therapeutic efficacy or high toxicity when administered alone could become therapeutically relevant and less toxic when combined with energy-restricted diets.
Can Synergy Occur with the KD-R and Hyperbaric Oxygen Therapy?
We know that the KD-R is an effective therapeutic strategy for managing a broad range of cancers. We also know that synergy occurs between the KD-R and 2-DG for brain cancer management. These observations beg the question as to how the KD-R might work if combined with hyperbaric oxygen therapy (HBO). HBO involves the treatment of subjects at 1–2 atmospheres in 100% oxygen.
HBO increases oxygen content of tissues and has been used to facilitate wound healing as well as treating several tumor types, including lung, breast, and glioma. It appears that many types of tumor cells are susceptible to the hyperoxia produced by HBO. Dominic D’Agostino mentioned to him that HBO “explodes” mitochondria in cultured tumor cells. This would kill any tumor cell with marginal respiratory activity or is dependent on glutamine for mitochondrial fermentation.
Like DER, hyperoxia targets tumor angiogenesis, while increasing tumor cell apoptosis. Stuhr and colleagues showed that HBO reduces micro-vessel density in gliomas to a similar extent.
Besides reducing angiogenesis and increasing apoptosis, HBO also appears to target inflammation. Viewed together, these findings suggest that HBO and DER target tumor energy metabolism in similar ways. Both the KD-R and HBO target tumor glycolysis. The KD-R reduces glucose availability, while HBO targets hexokinase II. Hexokinase II is attached to the mitochondria and plays a significant role in stimulating glycolysis.
Besides supporting glycolysis, the pentose phosphate pathway also provides metabolites for DNA synthesis and NADPH for lipid synthesis. NADPH is essential for maintaining catalase activity. Catalase is needed to metabolize H2O2 to water and O2. Tumor cells have excessive ROS due to respiratory dysfunction. NADPH depletion would therefore increase the vulnerability of tumor cells to ROS through linkage to catalase reduction. The KD-R would reduce levels of NADPH through the pentose phosphate pathway, thus reducing catalase activity, while HBO would elevate ROS, thus increasing risk of ROS-induced death. Ketones protect against ROS damage in cells with normal respiration because ketone metabolism in mitochondria oxidizes the coenzyme Q couple, thus decreasing the Q semiquinone, a major source of radical production in cells.
Targeting Glutamine
Glutamine is a major energy metabolite for many tumor cells and especially for cells of hematopoietic or myeloid lineage. This is important as cells of myeloid lineage are considered the origin of many metastatic cancers. Moreover, glutamine is necessary for the synthesis of those cytokines involved in cancer cachexia, including TNF-α, IL-1, and IL-6. This further indicates a metabolic linkage between metastatic cancer and myeloid cells, for example, macrophages.
Phenylbutyrate is a relatively nontoxic drug that can lower systemic glutamine levels in humans, but mice are unable to metabolize phenylbutyrate to phenylacetate. To lower blood glutamine levels in humans, phenylbutyrate must be first metabolized to phenylacetate. Phenylacetate then binds glutamine and is excreted in the urine as phenylacetylglutamine.
DON is a glutamine antagonist that inhibits glutamine metabolism. DON was effective in reducing colon and lung tumor growth in patients when administered with the glutamine-depleting enzyme PEG-PGA.
Glutamine Targeting Inhibits Systemic Metastasis
Although DER reduces blood glucose levels, it does not reduce blood glutamine levels. Indeed, blood glutamine levels might increase under DER in mice, as moderate physical activity can increase blood glutamine. Mice increase physical activity food foraging under DER.
They found that the DON prevented metastatic spread to the liver, lung, and kidney. In addition, they examined liver histology because liver becomes heavily infiltrated with VM-M3 cells. Indeed, liver metastasis was found in 100% of the control mice. Liver is also a common site for many metastatic human cancers. Histological analysis confirmed the lack of tumor cells in the liver of the DON-treated mice in comparison to the control AL nontreated mouse and control and CR-treated groups.
Interestingly, the DON-treated mice showed metastasis to the spleen. The spleen is recognized as a reservoir for monocytes and may represent a sanctuary for the myeloid-like metastatic cells.
Previous studies showed increases in glutaminase activity in the spleens of tumor-bearing mice. Glutaminase is the first enzyme involved in glutamine metabolism.
They found that DON treatment, either alone or in combination with CR, significantly reduced tumor growth and metastasis. Moreover, less DON was used to achieve therapeutic effect in the DON+CR mice than in the mice treated with DON alone. They reduced the DON doses for the mice in order to reduce potential toxicities or extreme energy stress.
The mice treated with DON+CR were active throughout the study and maintained a healthy body weight. Interestingly, the mice on DON treatment alone showed a more adverse response to drug treatment than did the mice on DON+CR. The mice treated with DON alone dropped body weight and were lethargic over the last 3 days of the study. Toxicity in the mice treated with DON alone became more evident as the study progressed. Toxicity was reduced and survival was enhanced when DON was administered together with CR, as a lower drug dosage was needed to achieve therapeutic effect. Moreover, the incidence of metastasis to spleen was significantly lower in the DON+CR mice than in the mice treated with DON alone.
Glutamine restriction can increase glucose metabolism. This should not be a problem, as the KD-R targets glucose metabolism.
Targeting Phagocytosis
The phagocytic activity of metastatic melanoma cells is significantly increased when the cells are grown under low glucose conditions, suggesting that metastatic cells use phagocytosis as a way to “feed” when nutrient supplies are low.
Glucose and glutamine can be derived from lysosomal digestion of phagocytosed materials. Shelton showed that the metastatic VM tumor cells could produce lactate when grown in Matrigel in minimal media containing no glucose. Cells grown in the absence of Matrigel produced no lactate and died, indicating that the Matrigel provided fermentable energy metabolites for the metastatic cells.
The antimalarial drug, chloroquine, might be useful in circumventing this problem. Chloroquine reduces the pH within lysosomes. Chloroquine also has demonstrated therapeutic efficacy against human brain cancer and experimental pancreatic cancer. As many highly invasive and metastatic cancers can be derived from naturally phagocytic myeloid cells, chloroquine could be effective in reducing lysosomal-based activities, for example, autophagy and phagocytosis.
Targeting the Microenvironment
The microenvironment maintains tissue architecture, thus inhibiting cell growth and suppressing the malignant phenotype of cancer cells. Growth factors and cytokines released by fibroblasts and macrophages, cells programmed to heal wounds, can actually provoke chronic inflammation and tumor progression. Part of the wound healing process also involves degradation of the extracellular matrix and enhancement of angiogenesis, which further contribute to tumor progression.
Kari and coworkers previously showed that CR significantly reduced the inflammatory properties of alveolar lung macrophages in response to Streptococcus infection. The transition in energy metabolism from glucose to ketones is powerfully anti-inflammatory to the tumor microenvironment. This is one reason why the KD is under consideration for use in numerous neurological and neurodegenerative diseases where inflammation is part of the pathology.
Dietary Energy Reduction as a Mitochondrial Enhancement Therapy (MET)
The transition from glucose metabolism to ketone metabolism will reduce tissue inflammation and ROS, while enhancing the metabolic efficiency of the mitochondria. MET is a more appealing term for cancer management in humans because it eliminates terms involving dietary restrictions. MET is also more accurate, as this term addresses the mechanism by which the therapy actually works.
Introduction
Most cancers, regardless of tissue origin, depend on fermentation energy for growth and survival. Glucose and glutamine are the major fermentable fuels for most cancer cells. Restriction of these fuels becomes a viable therapeutic strategy for management of most, if not all, cancers.
Dietary energy reduction (DER) creates a metabolic environment that targets tumor cell energy metabolism. DER creates a physiological environment where competition for available nutrients increases among all cells.
While drugs mimicking the global therapeutic effects of DER would certainly be optimal, no drugs are presently known that can produce these effects. Most calorie-restriction mimetic drugs fail to elevate ketones, which will protect normal cells from hypoglycemia.
Guidelines for Implementing the Restricted Ketogenic Diet as a Treatment Strategy for Cancer
Phase 1: Initiation:
Phase 1 of the treatment strategy would require cancer patients to gradually lower their circulating glucose levels while concurrently elevating their circulating ketone (β-hydroxybutyrate, β-OHB) levels. The procedures used for measuring blood glucose and ketone levels in cancer patients are essentially the same as those that would be used by individuals with diabetes.
Patients can measure their blood glucose three times per day, preferably before breakfast and about 2 h after lunch and dinner. It is essential for cancer patients to keep accurate records in order to identify any foods that might spike blood glucose levels.
Although it is better to measure ketone levels in blood than in urine (5–7), it may aid compliance to track urine ketones at frequent intervals during the early stages of implementation or until patients become familiar with the procedures for using the blood ketone meter. Thereafter, urine testing may be used in addition to blood testing as an additional measure of dietary compliance. As finger blood withdrawal is more easily tolerated in adults than in children, the protocol can be modified for children (less frequent or modified testing). There might also be blood glucose monitors available that do not require blood extraction for analysis.
Blood glucose ranges between 3.0 and 3.5 mM (55–65 mg/dl) and β-OHB ranges between 4 and 7 mM should be effective for reducing tumor growth in most patients. These values are within normal physiological ranges of glucose and ketones in humans and, based on our findings in mice, should have antiangiogenic, anti-inflammatory, and proapoptotic effects.
Ketones that are not used for energy are excreted in the urine. This happens when the KD is consumed in unrestricted or elevated amounts. When the KD is consumed in small amounts, the ketones generated are retained because they are needed to supply normal cells with energy as glucose availability decreases.
It is important for patients and physicians to recognize that circulating ketone levels will rarely exceed 7–9 mM in most nondiabetic states. Although elevated ketone bodies are often associated with diabetic states, ketone body elevation in people with normal physiology is considered “good medicine” and is therapeutic for a broad range of cardiac, neurological, and neurodegenerative diseases. It is unlikely that the KD-R or ketone ester supplementation will elevate blood ketones to pathological levels (greater than 15–20 mM) in most cancer patients with normal physiology.
Patients in good health should start the therapy with a water-only fast. Therapeutic fasting will lower blood glucose and will elevate blood ketones to the therapeutic ranges within 48–72 h. While this degree of food abstinence might sound draconian to some people, fasting for 2–3 days should not be difficult for those individuals in good physical health and who are already familiar with the health benefits of fasting. Fasting is often used to initiate the KD as a therapy for managing refractory seizures in children with epilepsy.
A gradual introduction of the KD without fasting might be necessary for those patients who are fragile or in poor health. For those patients not conducting a water-only fast, a restriction of carbohydrates to <12 g/day and a limitation of protein to about 0.8–1.2 g/kg of body weight/day is one way to potentially enter the therapeutic ranges for blood glucose and ketones. All cancer patients will need to carefully measure the amounts of food consumed each day.
Consumption of fats and oils can be used to make up the balance of energy needs. This approach will, however, require longer periods of time (perhaps several weeks) to reach the therapeutic ranges.
Once patients get their blood glucose levels to 55–65 mg/dl range and get their blood ketones to the 3–5 mM range, they can then maintain this metabolic state using various ketogenic diets and caloric adjustments. As the KD can have a diuretic effect, it is best to avoid diuretic drugs such as Lasix. Electrolyte levels should also be monitored or replenished if needed while on the diet for extended periods.
Consumption of the KD in unrestricted amounts will prevent blood glucose from reaching the reduced levels needed to target tumor progression and can have adverse effects for some patients. All cancer patients and their physicians should know that “less is better” when it comes to using the KD for managing cancer growth.
It is helpful for cancer patients to keep accurate food records during diet implementation. This information should be shared with health-care professionals who are experienced in implementing very low carbohydrate therapies. In order to maintain compliance, patients can use the “KetoCalculator”.
VARIABILITY IN CALORIC ADJUSTMENTS AND WEIGHT LOSS
Some patients might achieve the therapeutic glucose/ketone range without significant weight reduction, while other patients might require significant weight reduction to achieve the metabolic state.
The weight loss associated with the KD-R is part of a metabolically appropriate response to calorie restriction. In contrast, the weight loss often seen in cancer patients following radiation or chemotherapy is due to toxicity and to the effects of the therapy on appetite.
Some cancer patients are given high caloric drinks to prevent weight loss from toxic cancer therapies. Worse yet, some cancer patients are given steroids to reduce nausea and vomiting from toxic chemotherapy. It is his view that high energy drinks and steroids will help rescue some cancer cells from the therapies used to kill them.
The therapeutic efficacy of the metabolic KD therapy will be enhanced if blood glucose can be maintained in the low ranges (55–65 mg/dl). Given the wide variations in age, body type, weight, and metabolic status that we are likely to encounter in humans as compared to mouse models, he anticipates the need to individualize the degree of caloric restriction to lower glucose and to elevate ketones to ranges that will retard cancer progression.
SYMPTOMS OF GLUCOSE WITHDRAWAL
Some patients might experience light-headedness, nausea, headache, and so on in the first few days of the KD-R, especially if they initiate the therapy with a multiday fast. These symptoms are generally transient and are associated more with glucose withdrawal than with adverse effects of the diet. Evidence suggests that the human brain can become addicted to glucose from a lifelong consumption of energy-dense foods of low nutritional value.
Glucose withdrawal symptoms can be greater in those individuals who have never fasted than in individuals who have experience with fasting. As most people in modern industrial societies do not practice therapeutic fasting as a lifestyle, glucose withdrawal symptoms will likely be encountered in most patients who attempt the KD-R as cancer therapy. These symptoms could also be greater in older individuals than in younger ones. Indeed, fasting might not be possible in some older people who have lived a food-rich life of excess.
When compared to the debilitating effects of conventional chemotherapies and radiation, however, the symptoms associated with the KD-R are relatively mild and will pass after 2–3 days for most people. Nevertheless, glucose withdrawal symptoms and the feeling of hunger are simply too uncomfortable for some people regardless of the potential therapeutic benefits.
The KD-R should be used only for those patients who are motivated, disciplined, and healthy enough to make the necessary changes to diet and lifestyle. Unfortunately, many cancer patients are either incapable or are unwilling to meet these requirements.
A recent study in rats suggests that diets supplemented with ketone esters might produce physiological effects similar to those for the KD-R, but without significant food restriction. However, administration of ketone esters has not yet been tested in cancer patients. It would be important to evaluate the influence of ketone esters on blood glucose and ketones during therapeutic fasting.
The KD-R will not be effective in the presence of dexamethasone (decadron) or other steroid medications. Patients using steroids with the KD-R are unable to lower glucose to the therapeutic ranges. Steroid medications prevent glucose levels from falling into the therapeutic zone and therefore antagonize the therapeutic effects of KD-R. While steroids can rapidly mitigate some aspects of the tumor-related symptoms over the short term (paralysis, edema, appetite, etc.), chronic steroidal use will ultimately accelerate the growth of surviving tumor cells and thus the demise of cancer patients.
ROLE OF EXERCISE
Exercise during the fast should be fine, as long as the exercise is not too vigorous. Vigorous exercise will increase blood glucose due to muscle release of lactate and amino acids including glutamine. Moreover, excessive exercise will activate circulating monocytes that will leave the blood and enter the tumor.
Moderate exercise will not stress
the body’s immune system and should have therapeutic benefit.
Phase 2: Surgery:
This option will not be possible for those patients in a critical condition at the time of presentation. Dietary energy reduction and the KD-R will reduce tumor vascularization and inflammation and will more clearly delineate tumor tissue from the surrounding normal tissue.
The urge to resect malignant tumors as soon as possible after diagnosis may not be in the best interests of all patients and could actually exacerbate disease progression by inducing inflammation in the microenvironment.
Phase 3: Maintenance:
Phase three of the treatment strategy is designed to maintain metabolic pressure on surviving tumor cells.
Diet cycling for cancer patients could involve weekly transitions from calorically KD-R to nutritious, low calorie, and low glycemic diets. Patients should continue monitoring their blood glucose and ketone levels for as long as possible or until disease resolution is achieved.
In order to significantly extend patient survival, he recommends combining the KD-R therapy with drugs that also target glucose and glutamine. The KD-R can be administered together with 2-DG (30–40 mg/kg) and with phenylbutyrate (15 g/day) as a diet drug cocktail for targeting both glucose and glutamine in cancer patients. 2-DG will target glucose metabolism and glycolysis, while phenylbutyrate will help lower circulating glutamine levels.
Phenylbutyrate is metabolized to phenylacetate, which binds to glutamine for elimination in the urine. According to Henri Brunengraber, glycerol phenylbutyrate could be more effective in reducing systemic glutamine than sodium phenylbutyrate (buphenyl), as nontoxic dosing can be higher for glycerol phenylbutyrate than for buphenyl (personal communication). The drug AN-113 might access the brain better than phenylbutyrate and could therefore be more effective than phenylbutyrate in reducing brain levels of glutamine.
Interestingly, the glutamine analog drug, 6-diazo-5-oxo-l-norleucine (DON) appears to have less toxicity in humans than in mice. In contrast to the DON toxicity, they found when treating mice with metastatic cancer that DON, used with a glutaminase inhibitor, was well tolerated in patients with advanced colon and lung cancer.
Cachexia
In glioblastoma patients that express higher levels of IL-6, may be a biomarker of cachexia. Other procachexia molecules such as proteolysis-inducing factors are released from the tumor cells into the circulation and contribute to the cachexia phenotype. The KD-R will reduce inflammation and expression of IL-6. IL-6 also increases expression of hepcidin, which contributes to the anemia seen in many cancer patients (67). By killing the fermenting tumor cells that produce pro-cachexia molecules, the KD-R can potentially reduce tumor cachexia.
Summary
The objective of this new therapeutic strategy is to change the metabolic environment of the tumor and the host. Access to glucose and glutamine within the tumor microenvironment provides neoplastic tumor cells with fermentable fuels necessary for their survival and growth. The low carbohydrate, high fat ketogenic diet KD will reduce circulating glucose levels and will elevate circulating levels of ketone bodies especially when consumed in restricted amounts.
The therapeutic efficacy of the KD-R against malignant cancers can be enhanced when combined with drugs that also target or reduce access to glucose and glutamine. A use protocol is presented to help oncologists and cancer patients implement the KD-R as a treatment strategy. Although the KD-R is less toxic and potentially more effective in managing advanced cancers than the conventional standard of care, considerable patient education, motivation, and discipline will be necessary for implementing this therapy.
The incidence of cancer can be significantly reduced by avoiding exposure to agents or conditions that provoke tissue inflammation, such as smoking, excessive alcohol consumption, carcinogenic chemicals, ionizing radiation, and obesity.
Elevated levels of inflammation biomarkers (IL-6, IL-8, C-reactive protein, etc.) predict increased risk of cancer. Chronic inflammation, regardless of its origin, damages tissue morphogenetic fields and the epithelial and mesenchymal cells within the field. Most importantly, inflammation damages cellular mitochondria, thus reducing the efficiency of OxPhos. Reduced OxPhos efficiency initiates a mitochondrial stress response (RTG signaling) within cells.
RTG signaling is needed to upregulate either glycolysis in the cytoplasm or amino acid fermentation in the mitochondria. Only those cells that can enhance their fermentation in response to respiratory damage will survive. Cells incapable of enhancing fermentation will die from energy failure. As mitochondrial function maintains the differentiated state, cells that upregulate fermentation for survival are at increased risk of becoming less differentiated and ultimately transformed.
Prolonged reliance on fermentation destabilizes the nuclear genome, thus initiating the path to carcinogenesis and frank neoplasia. Inflammation damages cellular respiration; damaged respiration is the origin of cancer.
Vaccines against oncogenic viruses can also reduce the incidence of some cancers, as viruses can damage mitochondria in infected tissues. It is known that avoidance of cancer risk factors, which produce chronic inflammation and mitochondrial damage, will reduce the incidence of at least 80% of all cancers.
Cell Phones and Cancer
The cell phone is now considered to be a carcinogen in the same category as chloroform, formaldehyde, and lead. The risk of developing brain cancer from cell phone use will depend on gene-environmental interactions similar to the risk factors for developing any cancer.
Cell phones produce what is called extremely low frequency electromagnetic fields (ELF-EMF). These frequencies are in the range of those found in microwave ovens and television transmitters. Persistent tissue exposure to ELF-EMF produces thermogenesis (heat) in affected areas. While the increased temperature is slight, frequent and prolonged temperature shifts can influence CNS energy metabolism. Tissue thermogenesis will activate macrophages that then release inflammatory cytokines. These cytokines will induce inflammation in the tissue microenvironment, thus disturbing the integrity of the tissue morphogenetic field.
Mutations arise as an epiphenomenon of persistent fermentation, which ultimately arises from insufficient cellular respiration. The cell phone risk for cancer should be viewed in terms of respiratory insufficiency and disturbed energy metabolism in exposed cells rather than in terms of DNA damage and mutations. Hence, cell phone use can be linked to cancer risk through inflammation and injury to respiration in those individuals that are prone to focal CNS inflammation from increased temperature.
Alzheimer’s Disease and Cancer Risk
It appears that the risk of cancer in persons with Alzheimer’s disease (AD) is significantly less than that in persons without the disease.
It is known that AD is a type of hypometabolic disorder. Loss of appetite with accompanying reduced body weight and blood glucose levels are seen in many patients with AD. As glucose drives tumor cell growth, hypometabolism and reduced glucose would create a type of calorie-restricted environment. Such an environment would naturally inhibit tumor initiation and growth. In contrast to calorie restriction, however, the hypometabolism in AD is not associated with elevated ketone bodies.
Ketone Metabolism Reduces Cancer Risk
The metabolism of ketone bodies protects the mitochondria from inflammation and damaging reactive oxygen species (ROS). ROS production increases naturally with age and damages cellular proteins, lipids, and nucleic acids. Accumulation of ROS decreases the efficiency of mitochondrial energy production, thus requiring compensatory fermentation. Cancer risk increases with age and accumulation of ROS. Ketone metabolism enhances mitochondrial function, thus preventing fermentation. Ketone body metabolism, especially when glucose levels are reduced, will go far in preventing genomic instability and reducing cancer risk.
The origin of mitochondrial ROS comes largely from the spontaneous reaction of molecular oxygen (O2) with the semiquinone radical of coenzyme Q, that is, QH. This interaction will generate the superoxide radical O−2. Coenzyme Q is a hydrophobic molecule that resides in the inner mitochondrial membrane and is essential for electron transfer. Ketone body metabolism increases the ratio of the oxidized form to the fully reduced form of coenzyme Q (CoQ/CoQH2). Oxidation of the coenzyme Q couple reduces the amount of the semiquinone radical, thus decreasing the probability of superoxide production. Ketone body metabolism reduces ROS and enhances mitochondrial energy efficiency, thus reducing cancer risk.
In addition to reducing ROS, ketone body metabolism also increases the reduced form of glutathione since the cytosolic-free NADP+/NADPH concentration couple is in near equilibrium with the glutathione couple. More specifically, ketone body metabolism facilitates destruction of hydrogen peroxide. The reduction of free radicals through ketone body metabolism helps maintain the inner mitochondrial membrane integrity. This enhances the energy efficiency of mitochondria. As ROS also induces tissue inflammation, reduced ROS will reduce tissue inflammation. Ketone bodies are not only a more efficient metabolic fuel than glucose but also possess anti-inflammatory potential.
Mitochondrial Enhancement Therapy
Consumption of foods containing the active groups of respiratory enzymes (iron salts, riboflavin, nicotinamide, and pantothenic acid) will be effective in maintaining health when combined with dietary energy restriction. Vitamin D is also known to enhance mitochondrial efficiency.
Reducing blood glucose levels through DER facilitates ketone body uptake and metabolism for use as an alternative respiratory fuel. It is important to remember that tumor cells cannot effectively use ketone bodies for energy because of their injury to respiration. The metabolism of ketone bodies increases succinate dehydrogenase activity while enhancing the overall efficiency of energy production through respiration. The supplementation of DER with ketone esters could be even more effective as respiratory enhancement therapy.
The drug 1,3- butanediol could also help elevate ketone bodies to reduce inflammation and cancer.
Specifically, DER and ketone body metabolism delays entropy. Entropy is the bioenergetic signature of cancer. Entropy refers to the degree of disorder in systems and is the foundation of the second law of thermodynamics. Szent-Gyorgyi has described cancer as an increased state of entropy, where randomness and disorder predominate. As cancer is a disease of accelerated entropy, DER targets the very essence of the disease.
Therapeutic Fasting and Cancer Prevention
Humans have evolved to function for prolonged periods in the absence of food. Herbert Shelton described how most adults in good general health can function normally after fasting (water only) for as long as 30–40 days.
George Cahill and Oliver Owen have also shown that many overweight people could be fasted for prolonged periods (months) without adverse effects. Owen and Cahill were also the first to show that ketone bodies become the major fuel for the brain during periods of starvation.
“Danjiki” is the Japanese term for therapeutic fasting and is known to produce numerous health benefits including prevention of cancer. Humans are capable of conducting prolonged fasts without harm.
Although the terms fasting and starvation are often used interchangeably, they represent different physiological states. Starvation is a pathological state where the body suffers from energy imbalance and is deprived of key minerals and vitamins necessary for maintaining metabolic homeostasis. Fasting, on the other hand, is therapeutic and maintains metabolic homeostasis. Vitamins A, D, E, and K are stored in liver and body fat, and are released slowly during fasting. Minerals are stored in the bones and are also released slowly during fasting. Only the water-soluble vitamins C and B-complex vitamins would require supplementation after a 10–14-day fast.
Blood Glucose and Ketone Levels During Fasting
Caffeine can prevent glucose levels from entering the therapeutic zone necessary to target the energy metabolism of tumor cells. Herbert Shelton argues against coffee consumption during fasting.
Autophagy and Autolytic Cannibalism: A Thermodynamic Approach to Cancer Prevention
Autophagy is the process by which cells break down and recycle energy-rich molecules from inefficient organelles. The deficient organelles fuse with endosomes or lysosomes for the digestive process. Autolytic cannibalism is the process by which the body digests whole cells and tissues that are metabolically inefficient relative to normal healthy cells and tissues. Both processes can occur under DER.
The body temperature is lower during therapeutic fasting or DER than during unrestricted feeding. In order to maintain temperature, the body will metabolize the stored energy (fat) or dysplastic tissue.
The metabolism of ketones spares protein and protects the brain. Only those cells with flexible genomes, honed through millions of years of environmental forcing and variability selection, can readily transition from one energy state to another.
Tumor cells will be less able to survive energy stress than normal cells, thus allowing the normal cells to use the energy metabolites of the dysplastic tissue for maintaining body heat and organ homeostasis. In other words, the body cannibalizes dysplastic tissue through autolytic processes in order to supply the normal cells with energy. This process would occur, however, only under conditions of energy stress. Under conditions of energy excess, cancer cells would persist in the body and possibly thrive.
How Long Should People Fast or Remain on the Restricted Ketogenic Diet to Prevent Cancer?
The length of therapeutic fasting or administration of the KD-R for cancer prevention could vary from one person to the next. In general, a 7-day, water-only fast done once per year would be sufficient for the body to consume dysplastic or precancerous tissue. It usually requires 2–3 days for the blood glucose to reach the therapeutic levels of 55–65 mg/dl and for ketones to reach the 3–5 mmol therapeutic levels. Once the body reaches this metabolic state, autophagy and autolytic cannibalism will begin purging the body of neoplastic tissue.
For those individuals incapable of conducting longer fasts, several shorter fasts (2–3 days) done two to three times per year should also be effective in preventing cancer. A ketogenic diet consumed for 1 week should also be an effective cancer-prevention strategy as long as the blood glucose and ketones are maintained within the therapeutic ranges.
One key point was the difficulty encountered in having cancer patients maintain their blood glucose levels in the therapeutic ranges. Although the KD was capable of elevating circulating ketone levels, it was less effective in maintaining reduced blood glucose levels. As Beth Zupec-Kania mentioned in her comments, it is likely that certain medications might prevent glucose from reaching therapeutic levels necessary to kill tumor cells. This is especially the case for those patients taking steroids, which prevent glucose levels from entering the therapeutic zone. It is unclear if seizure medications or certain chemotherapies also prevent glucose levels from reaching the therapeutic zone. Most of his healthy students who have fasted or used the KD had no trouble reaching the therapeutic zones of glucose and ketones.
He agrees with Dr. Moore that the KD-R should have its greatest potential in managing lower grade tumors when used alone or when combined with nontoxic drugs that also target tumor cell energy metabolism.
The stability of chromosome number and the integrity of the genome are dependent on the integrity OxPhos. Spindle assembly and the fidelity of chromosomal segregation during mitosis are dependent on the energy of OxPhos. Injury to cellular respiration with compensatory fermentation will cause genomic instability including aneuploidy and mutations. It is the efficiency of mitochondrial respiration that maintains cellular differentiation and prevents tumorigenesis and dedifferentiation.
Metastatic tumor cells arise from respiratory damage to myeloid cells, which are already mesenchymal. Many of the biomarkers expressed in metastatic cancer cells are also expressed in macrophages. While epithelial tumor cells proliferate rapidly, they do not generally metastasize unless they fuse with a cell of myeloid origin. Tissue biomarkers of myeloid cells are expressed in many metastatic cancer cells.
According to his view, metastatic cancer cells arise from cells of the immune system (macrophages). While it might be difficult to induce nonneoplastic macrophages to recognize neoplastic macrophages as foreign invaders, it might be easier to eliminate the metastatic cells of immune origin by targeting their energy metabolism and capacity for phagocytosis.
Cancer cells will escape as long as they can maintain their ability to ferment. Fermentation energy (glycolysis) underlies drug resistance. If tumor cells cannot ferment, they will die.
MAJOR CONCLUSIONS