Cancer as a metabolic disease: implications for novel therapeutics

Thomas N Seyfried, Roberto E Flores, Angela M Poff, Dominic P D'Agostino, Thomas N Seyfried, Roberto E Flores, Angela M Poff, Dominic P D'Agostino

Abstract

Emerging evidence indicates that cancer is primarily a metabolic disease involving disturbances in energy production through respiration and fermentation. The genomic instability observed in tumor cells and all other recognized hallmarks of cancer are considered downstream epiphenomena of the initial disturbance of cellular energy metabolism. The disturbances in tumor cell energy metabolism can be linked to abnormalities in the structure and function of the mitochondria. When viewed as a mitochondrial metabolic disease, the evolutionary theory of Lamarck can better explain cancer progression than can the evolutionary theory of Darwin. Cancer growth and progression can be managed following a whole body transition from fermentable metabolites, primarily glucose and glutamine, to respiratory metabolites, primarily ketone bodies. As each individual is a unique metabolic entity, personalization of metabolic therapy as a broad-based cancer treatment strategy will require fine-tuning to match the therapy to an individual's unique physiology.

Figures

Fig. 1.
Fig. 1.
Role of the nucleus and mitochondria in the origin of tumors. This image summarizes the experimental evidence supporting a dominant role of the mitochondria in the origin of tumorigenesis as described previously (49). Normal cells are depicted in green with mitochondrial and nuclear morphology indicative of normal respiration and nuclear gene expression, respectively. Tumor cells are depicted in red with abnormal mitochondrial and nuclear morphology indicative of abnormal respiration and genomic instability. (1) Normal cells beget normal cells. (2) Tumor cells beget tumor cells. (3) Delivery of a tumor cell nucleus into a normal cell cytoplasm begets normal cells despite the persistence of tumor-associated genomic abnormalities. (4) Delivery of a normal cell nucleus into a tumor cell cytoplasm begets tumor cells or dead cells but not normal cells. The results suggest that tumors do not arise from nuclear genomic defects alone and that normal mitochondria can suppress tumorigenesis. Original diagram from Jeffrey Ling and Thomas N. Seyfried, with permission.
Fig. 2.
Fig. 2.
Typical ultrastructure of a normal mitochondrion and a mitochondrion from a human glioblastoma. Normal mitochondria contain elaborate cristae, which are extensions of the inner membrane and contain the protein complexes of the electron transport chain necessary for producing ATP through OxPhos. The mitochondrion from the glioblastoma (m) is enlarged and shows a near total breakdown of cristae (cristolysis) and an electron-lucent matrix. The absence of cristae in glioblastoma mitochondria indicates that OxPhos would be deficient. The arrow indicates an inner membrane fold. Bar: 0.33 μm. Method of staining: uranyl acetate/lead citrate. The glioblastoma multiforme mitochondrion was reprinted with permission from Journal of Electron Microscopy (94). The normal mitochondrion diagram was from http://academic.brooklyn.cuny.edu/biology/bio4fv/page/mito.htm.
Fig. 3.
Fig. 3.
Mitochondrial respiratory dysfunction as the origin of cancer. Cancer can arise from any number of non-specific events that damage the respiratory capacity of cells over time. The path to carcinogenesis will occur only in those cells capable of enhancing energy production through fermentation (substrate level phosphorylation, SLP). Despite the shift from respiration to SLP the ΔG′ of ATP hydrolysis remains fairly constant at approximately −56 kJ indicating that the energy from SLP compensates for the reduced energy from OxPhos. The mitochondrial stress response or retrograde signaling will initiate oncogene upregulation and tumor suppressor gene inactivation that are necessary to maintain viability of incipient cancer cells when respiration becomes unable to maintain energy homeostasis. Genomic instability will arise as a secondary consequence of protracted mitochondrial stress from disturbances in the intracellular and extracellular microenvironment. Metastasis arises from respiratory damage in cells of myeloid/macrophage origin (146). The degree of malignancy is linked directly to the energy transition from OxPhos to SLP. This scenario links all major cancer hallmarks to an extrachromosomal respiratory dysfunction (141). The T signifies an arbitrary threshold when the shift from OxPhos to SLP might become irreversible. Reprinted with modifications from (17).
Fig. 4.
Fig. 4.
Timeline of events following expression of K-Ras. The time axis depicts the various events after stimulation of the K-RAS gene expression. The findings of Huang et al. indicate that mitochondria-linked changes are observed around the time of the increase in the K-Ras protein (142). This is then followed by other changes, such as alteration in cell metabolism. Gene mutations would form as a downstream epiphenomenon of altered metabolism. Malignant transformation, documented by the colony-forming activity of the cells and their propensity to form tumors ensue much later. According to these findings, the Warburg effect (aerobic fermentation) arises as a consequence of K-RAS-induced respiratory injury. This timeline is in general agreement with Warburg’s central theory and with other similar findings that respiratory disturbance is an initial event in K-RAS-induced tumorigenesis (237–239). The timeline will be greatly protracted in vivo as shown from the Roskelley et al. (240) experiments. Reprinted from Neuzil et al. (143) with permission.
Fig. 5.
Fig. 5.
Relationship of circulating levels of glucose and ketones (β-hydroxybutyrate) to tumor management. The glucose and ketone values are within normal physiological ranges under fasting conditions in humans and will produce anti-angiogenic, anti-inflammatory and pro-apoptotic effects. We refer to this state as the zone of metabolic management. Metabolic stress will be greater in tumor cells than in normal cells when the whole body enters the metabolic zone. The values for blood glucose in mg/dl can be estimated by multiplying the mM values by 18. The glucose and ketone levels predicted for tumor management in human cancer patients are 3.1–3.8mM (55–65mg/dl) and 2.5–7.0mM, respectively. These ketone levels are well below the levels associated with ketoacidosis (blood ketone values greater than 15 mmol). Elevated ketones will protect the brain from hypoglycemia. Modified from a previous version (241).
Fig. 6.
Fig. 6.
The KD and HBO2T are synergistic in reducing systemic metastatic cancer in the syngeneic VM mouse model. VM-M3/Fluc tumor cells were implanted subcutaneously and systemic organ metastasis was evaluated ex vivo using bioluminescent imaging as described previously (242,243). Tumor growth was slower in mice fed the KD than in mice fed a standard high carbohydrate diet. (A) Representative animals from each treatment group demonstrating tumor bioluminescence at day 21 after tumor cell inoculation. Treated animals showed less bioluminescence than controls with KD + HBO2T mice exhibiting a profound decrease in tumor bioluminescence compared with all groups. (B) Total body bioluminescence was measured weekly as a measure of tumor size; error bars represent standard error of the mean. KD + HBO2T mice exhibited significantly less tumor bioluminescence than control animals at week 3 (P < 0.01; two-tailed student’s t-test) and an overall trend of notably slower tumor growth than controls and other treated animals throughout the study. (C and D) Day 21 organ bioluminescence of standard high carbohydrate diet and KD + HBO2T animals (N = 8) demonstrated a trend of reduced metastatic tumor burden in animals receiving the combined therapy. Spleen bioluminescence was significantly decreased in KD + HBO2T mice (*P < 0.02; two-tailed student’s t-test). Reprinted with permission from Poff et al. (243).

References

    1. Sporn M.B. (1996). The war on cancer. Lancet, 347, 1377–1381
    1. Seyfried T.N. (2012). Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ
    1. Fidler I.J. (2003). The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat. Rev. Cancer, 3, 453–458
    1. Lazebnik Y. (2010). What are the hallmarks of cancer? Nat. Rev. Cancer, 10, 232–233
    1. Tarin D. (2011). Cell and tissue interactions in carcinogenesis and metastasis and their clinical significance. Semin. Cancer Biol., 21, 72–82
    1. Seyfried T.N. (2012). Confusion surrounds the origin of cancer. In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 15–29
    1. Hanahan D., et al. (2011). Hallmarks of cancer: the next generation. Cell, 144, 646–674
    1. Baker S.G., et al. (2007). Paradoxes in carcinogenesis: new opportunities for research directions. BMC Cancer, 7, 151.
    1. Soto A.M., et al. (2004). The somatic mutation theory of cancer: growing problems with the paradigm? Bioessays, 26, 1097–1107
    1. Vogelstein B., et al. (2013). Cancer genome landscapes. Science, 339, 1546–1558
    1. Alexandrov L.B., et al. (2013). Signatures of mutational processes in human cancer. Nature, 500, 415–421
    1. Soto A.M., et al. (2012). Is systems biology a promising approach to resolve controversies in cancer research? Cancer Cell Int., 12, 12.
    1. Seyfried T.N., et al. (2012). Is the restricted ketogenic diet a viable alternative to the standard of care for managing malignant brain cancer? Epilepsy Res., 100, 310–326
    1. Seyfried T.N. (2013). Cancer as a metabolic disease: implications for novel therapeutics. Amer. Assoc. Cancer Res. Education Book, 2013, 31–36
    1. Seyfried T.N., et al. (2011). Metabolic management of brain cancer. Biochim. Biophys. Acta, 1807, 577–594
    1. Seyfried T.N., et al. (2005). Targeting energy metabolism in brain cancer: review and hypothesis. Nutr. Metab. (Lond)., 2, 30.
    1. Seyfried T.N., et al. (2010). Cancer as a metabolic disease. Nutr. Metab. (Lond)., 7, 7.
    1. Seyfried T.N. (2012). Genes, respiration, viruses, and cancer. In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 145–176
    1. Stratton M.R. (2011). Exploring the genomes of cancer cells: progress and promise. Science, 331, 1553–1558
    1. Seyfried T.N. (2012). Nothing in cancer biology makes sense except in the light of evolution. In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 261–275
    1. Nowell P.C. (1976). The clonal evolution of tumor cell populations. Science, 194, 23–28
    1. Fojo T., et al. (2010). Biologically targeted cancer therapy and marginal benefits: are we making too much of too little or are we achieving too little by giving too much? Clin. Cancer Res., 16, 5972–5980
    1. Rosell R., et al. (2009). Customized treatment in non-small-cell lung cancer based on EGFR mutations and BRCA1 mRNA expression. PLoS One, 4, e5133.
    1. McLeod H.L. (2013). Cancer pharmacogenomics: early promise, but concerted effort needed. Science, 339, 1563–1566
    1. Hardy P.A., et al. . (2005) Reappraisal of the Hansemann-Boveri hypothesis on the origin of tumors. Cell Biol. Int., 29, 983–992
    1. Gibbs W.W. (2003). Untangling the roots of cancer. Sci. Am., 289, 56–65
    1. Hameroff S.R. (2004). A new theory of the origin of cancer: quantum coherent entanglement, centrioles, mitosis, and differentiation. Biosystems., 77, 119–136
    1. Manchester K. (1997). The quest by three giants of science for an understanding of cancer. Endeavour, 21, 72–76
    1. Wolf U. (1974). Theodor Boveri and his book, on the problem of the origin of malignant tumors. In German J. (ed.) Chromosomes and Cancer, John Wiley & Sons, New York, pp. 1–20
    1. Duesberg P., et al. (2000). Aneuploidy, the somatic mutation that makes cancer a species of its own. Cell Motil. Cytoskeleton, 47, 81–107
    1. Salk J.J., et al. (2010). Mutational heterogeneity in human cancers: origin and consequences. Annu. Rev. Pathol., 5, 51–75
    1. Cairns J. (1981). The origin of human cancers. Nature, 289, 353–357
    1. Loeb L.A. (2001). A mutator phenotype in cancer. Cancer Res., 61, 3230–3239
    1. Whitman R.C. (1919). Somatic mutations as a factor in the production of cancer; a critical review of von Hansemanns’s theory of anaplasia in light of modern knowledge of genetics. J. Cancer Res., 4, 181–202
    1. Nigro J.M., et al. (1989). Mutations in the p53 gene occur in diverse human tumour types. Nature, 342, 705–708
    1. Fearon E.R., et al. (1990). A genetic model for colorectal tumorigenesis. Cell, 61, 759–767
    1. Knudson A.G. (2002). Cancer genetics. Am. J. Med. Genet., 111, 96–102
    1. Wagner R.P. (1999). Anecdotal, historical and critical commentaries on genetics. Rudolph Virchow and the genetic basis of somatic ecology. Genetics, 151, 917–920
    1. Darlington C.D. (1948) The plasmagene theory of the origin of cancer. Br. J. Cancer, 2, 118–126
    1. Koura M., et al. (1982). Suppression of tumorigenicity in interspecific reconstituted cells and cybrids. Gann, 73, 574–580
    1. Israel B.A., et al. (1987). Cytoplasmic suppression of malignancy. In Vitro Cell. Dev. Biol., 23, 627–632
    1. Shay J.W., et al. (1988). Cytoplasmic suppression of tumorigenicity in reconstructed mouse cells. Cancer Res., 48, 830–833
    1. Howell A.N., et al. (1978). Tumorigenicity and its suppression in cybrids of mouse and Chinese hamster cell lines. Proc. Natl Acad. Sci. U. S. A., 75, 2358–2362
    1. Jonasson J., et al. (1977). The analysis of malignancy by cell fusion. VIII. Evidence for the intervention of an extra-chromosomal element. J. Cell Sci., 24, 255–263
    1. McKinnell R.G., et al. (1969). Transplantation of pluripotential nuclei from triploid frog tumors. Science, 165, 394–396
    1. Mintz B., et al. (1975). Normal genetically mosaic mice produced from malignant teratocarcinoma cells. Proc. Natl Acad. Sci. U. S. A., 72, 3585–3589
    1. Li L., et al. (2003). Mouse embryos cloned from brain tumors. Cancer Res., 63, 2733–2736
    1. Hochedlinger K., et al. (2004). Reprogramming of a melanoma genome by nuclear transplantation. Genes Dev., 18, 1875–1885
    1. Seyfried T.N. (2012). Mitochondria: the ultimate tumor suppressor. In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 195–205
    1. Kaipparettu B.A., et al. (2013). Crosstalk from non-cancerous mitochondria can inhibit tumor properties of metastatic cells by suppressing oncogenic pathways. PLoS One, 8, e61747.
    1. Elliott R.L., et al. . (2012) Mitochondria organelle transplantation: introduction of normal epithelial mitochondria into human cancer cells inhibits proliferation and increases drug sensitivity. Breast Cancer Res. Treat., 136, 347–354
    1. Israel B.A., et al. (1988). Cytoplasmic mediation of malignancy. In Vitro Cell. Dev. Biol., 24, 487–490
    1. Petros J.A., et al. (2005). mtDNA mutations increase tumorigenicity in prostate cancer. Proc. Natl Acad. Sci. U. S. A., 102, 719–724
    1. Warburg O. (1931). The Metabolism of Tumours. Richard R. Smith, New York
    1. Warburg O. (1956). On the origin of cancer cells. Science, 123, 309–314
    1. Pedersen P.L. (2007). Warburg, me and hexokinase 2: multiple discoveries of key molecular events underlying one of cancers’ most common phenotypes, the “Warburg Effect”, i.e., elevated glycolysis in the presence of oxygen. J. Bioenerg. Biomembr., 39, 211–222
    1. Warburg O. (1956). On respiratory impairment in cancer cells. Science, 124, 269–270
    1. Warburg O. (1969). Revidsed Lindau lectures: the prime cause of cancer and prevention - Parts 1 & 2. In Burk D. (ed.) Meeting of the Nobel-Laureates. K.Triltsch, Lindau, Lake Constance, Germany
    1. Racker E. (1972) Bioenergetics and the problem of tumor growth. Am. Sci., 60, 56–63
    1. Nelson D.L., et al. (2008) Lehninger Principles of Biochemistry W. H. Freeman, New York
    1. Cuezva J.M., et al. (2002). The bioenergetic signature of cancer: a marker of tumor progression. Cancer Res., 62, 6674–6681
    1. Acebo P., et al. (2009). Cancer abolishes the tissue type-specific differences in the phenotype of energetic metabolism. Transl. Oncol., 2, 138–145
    1. Weinhouse S. (1956). On respiratory impairment in cancer cells. Science, 124, 267–269
    1. Koppenol W.H., et al. (2011). Otto Warburg’s contributions to current concepts of cancer metabolism. Nat. Rev. Cancer, 11, 325–337
    1. Stellingwerff T., et al. . (2006) Hyperoxia decreases muscle glycogenolysis, lactate production, and lactate efflux during steady-state exercise. Am. J. Physiol. Endocrinol. Metab., 290, E1180–E1190
    1. Gatenby R.A., et al. (2004). Why do cancers have high aerobic glycolysis? Nat. Rev. Cancer, 4, 891–899
    1. Pedersen P.L. (1978). Tumor mitochondria and the bioenergetics of cancer cells. Prog. Exp. Tumor Res., 22, 190–274
    1. Chevrollier A., et al. (2005) ANT2 expression under hypoxic conditions produces opposite cell-cycle behavior in 143B and HepG2 cancer cells. Mol. Carcinog., 42, 1–8
    1. Wijburg F.A., et al. (1989) Studies on the formation of lactate and pyruvate from glucose in cultured skin fibroblasts: implications for detection of respiratory chain defects. Biochem. Int., 19, 563–570
    1. Tiefenthaler M., et al. (2001) Increased lactate production follows loss of mitochondrial membrane potential during apoptosis of human leukaemia cells. Br. J. Haematol., 114, 574–580
    1. Donnelly M., et al. (1976). Energy metabolism in respiration-deficient and wild type Chinese hamster fibroblasts in culture. J. Cell. Physiol., 89, 39–51
    1. Fiske B.P., et al. (2012) Seeing the Warburg effect in the developing retina. Nat. Cell Biol., 14, 790–791
    1. Prichard J., et al. (1991) Lactate rise detected by 1H NMR in human visual cortex during physiologic stimulation. Proc. Natl Acad. Sci. U. S. A., 88, 5829–5831
    1. Fox P.T., et al. (1988). Nonoxidative glucose consumption during focal physiologic neural activity. Science, 241, 462–464
    1. Krasnow N., et al. (1962) Myocardial lactate and pyruvate metabolism. J. Clin. Invest., 41, 2075–2085
    1. Burk D., et al. (1956). On respiratory impairment in cancer cells. Science, 124, 270–272
    1. Burk D., et al. (1967). On the significance of glucolysis for cancer growth, with special reference to Morris rat hepatomas. J. Natl Cancer Inst., 38, 839–863
    1. Vaupel P., et al. (2012) Availability, not respiratory capacity governs oxygen consumption of solid tumors. Int. J. Biochem. Cell Biol., 44, 1477–1481
    1. Moreno-Sánchez R., et al. (2007). Energy metabolism in tumor cells. FEBS J., 274, 1393–1418
    1. Haq R., et al. (2013) Oncogenic BRAF regulates oxidative metabolism via PGC1α and MITF. Cancer Cell, 23, 302–315
    1. Weinhouse S. (1976). The Warburg hypothesis fifty years later. Z. Krebsforsch. Klin. Onkol. Cancer Res. Clin. Oncol., 87, 115–126
    1. Ferreira L.M. (2010). Cancer metabolism: the Warburg effect today. Exp. Mol. Pathol., 89, 372–380
    1. Hall A., et al. (2013). Dysfunctional oxidative phosphorylation makes malignant melanoma cells addicted to glycolysis driven by the (V600E)BRAF oncogene. Oncotarget, 4, 584–599
    1. Hochachka P.W., et al. (2002). Biochemical Adaptation: Mechanism and Process in Physiological Evolution. Oxford Press, New York
    1. Seyfried T.N. (2012). Is respiration normal in cancer cells? In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 119–132
    1. Ramanathan A., et al. (2005). Perturbational profiling of a cell-line model of tumorigenesis by using metabolic measurements. Proc. Natl Acad. Sci. U. S. A., 102, 5992–5997
    1. Seyfried T.N. (2012). Is mitochondrial glutamine fermentation a missing link in the metabolic theory of cancer? In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 133–144
    1. Chinopoulos C., et al. (2010) Forward operation of adenine nucleotide translocase during F0F1-ATPase reversal: critical role of matrix substrate-level phosphorylation. FASEB J., 24, 2405–2416
    1. Phillips D., et al. (2009). Succinyl-CoA synthetase is a phosphate target for the activation of mitochondrial metabolism. Biochemistry, 48, 7140–7149
    1. Schwimmer C., et al. (2005). Increasing mitochondrial substrate-level phosphorylation can rescue respiratory growth of an ATP synthase-deficient yeast. J. Biol. Chem., 280, 30751–30759
    1. Seyfried T.N. (2012). Respiratory dysfunction in cancer cells. In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 73–105
    1. Arismendi-Morillo G. (2011) Electron microscopy morphology of the mitochondrial network in gliomas and their vascular microenvironment. Biochim. Biophys. Acta, 1807, 602–608
    1. Arismendi-Morillo G. (2009). Electron microscopy morphology of the mitochondrial network in human cancer. Int. J. Biochem. Cell Biol., 41, 2062–2068
    1. Arismendi-Morillo G.J., et al. (2008). Ultrastructural mitochondrial pathology in human astrocytic tumors: potentials implications pro-therapeutics strategies. J. Electron Microsc. (Tokyo)., 57, 33–39
    1. Galluzzi L., et al. (2010) Mitochondrial gateways to cancer. Mol. Aspects Med., 31, 1–20
    1. Kiebish M.A., et al. (2009). In vitro growth environment produces lipidomic and electron transport chain abnormalities in mitochondria from non-tumorigenic astrocytes and brain tumours. ASN Neuro, 1, e00011.
    1. Kiebish M.A., et al. (2008). Cardiolipin and electron transport chain abnormalities in mouse brain tumor mitochondria: lipidomic evidence supporting the Warburg theory of cancer. J. Lipid Res., 49, 2545–2556
    1. Gonzalez M.J., et al. (2012) The bio-energetic theory of carcinogenesis. Med. Hypotheses, 79, 433–439
    1. Benard G., et al. (2008) Ultrastructure of the mitochondrion and its bearing on function and bioenergetics. Antioxid. Redox Signal., 10, 1313–1342
    1. Shapovalov Y., et al. (2011) Mitochondrial dysfunction in cancer cells due to aberrant mitochondrial replication. J. Biol. Chem., 286, 22331–22338
    1. Alirol E., et al. (2006). Mitochondria and cancer: is there a morphological connection? Oncogene, 25, 4706–4716
    1. Oudard S., et al. (1997). Gliomas are driven by glycolysis: putative roles of hexokinase, oxidative phosphorylation and mitochondrial ultrastructure. Anticancer Res., 17, 1903–1911
    1. Wu S.B., et al. (2012). AMPK-mediated increase of glycolysis as an adaptive response to oxidative stress in human cells: implication of the cell survival in mitochondrial diseases. Biochim. Biophys. Acta, 1822, 233–247
    1. Fulda S., et al. (2010) Targeting mitochondria for cancer therapy. Nat. Rev. Drug Discov., 9, 447–464
    1. Shapovalov Y., et al. (2011) Mitochondrial dysfunction in cancer cells due to aberrant mitochondrial replication. J. Biol. Chem., 286, 22331–22338
    1. Chance B., et al. (1959). Spectroscopic evidence of metabolic control. Science, 129, 700–708
    1. Colowick S.P. (1961). The status of Warburg’s theory of glycolysis and respiration in tumors. Quart. Rev. Biol., 36, 273–276
    1. Cairns R.A., et al. (2011) Regulation of cancer cell metabolism. Nat. Rev. Cancer, 11, 85–95
    1. Ward P.S., et al. (2012). Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell, 21, 297–308
    1. Vander Heiden M.G., et al. (2009). Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science, 324, 1029–1033
    1. Rossignol R., et al. (2004). Energy substrate modulates mitochondrial structure and oxidative capacity in cancer cells. Cancer Res., 64, 985–993
    1. Chevrollier A., et al. (2011) Adenine nucleotide translocase 2 is a key mitochondrial protein in cancer metabolism. Biochim. Biophys. Acta, 1807, 562–567
    1. Jahnke V.E., et al. (2010). Evidence for mitochondrial respiratory deficiency in rat rhabdomyosarcoma cells. PLoS One, 5, e8637.
    1. Schild L., et al. (2012) Composition of molecular cardiolipin species correlates with proliferation of lymphocytes. Exp. Biol. Med. (Maywood)., 237, 372–379
    1. Kocherginsky N. (2009). Acidic lipids, H(+)-ATPases, and mechanism of oxidative phosphorylation. Physico-chemical ideas 30 years after P. Mitchell’s Nobel Prize award. Prog. Biophys. Mol. Biol., 99, 20–41
    1. Claypool S.M., et al. (2012) The complexity of cardiolipin in health and disease. Trends Biochem. Sci., 37, 32–41
    1. Zu X.L., et al. (2004). Cancer metabolism: facts, fantasy, and fiction. Biochem. Biophys. Res. Commun., 313, 459–465
    1. Wang T., et al. (1976). Aerobic glycolysis during lymphocyte proliferation. Nature, 261, 702–705
    1. Lunt S.Y., et al. (2011) Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol., 27, 441–464
    1. Papandreou I., et al. (2011) Anticancer drugs that target metabolism: is dichloroacetate the new paradigm? Int. J. Cancer, 128, 1001–1008
    1. Burk D., et al. (1941). Metabolism of butter yellow rat liver cancers. Canc. Res., 1, 733–734
    1. Simek J., et al. (1965). Effect of glucose administered in vivo or in vitro on the respiratory quotient of rat liver tissue after partial hepatectomy. Nature, 207, 761–762
    1. Diatlovitskaia E.V., et al. (1972). [Cardiolipins of mitochondria and microsomes of Jensen’s sarcoma]. Dokl. Akad. Nauk SSSR, 206, 737–739
    1. Diatlovitskaia E.V., et al. (1976). [Positional distribution of fatty acids in the phospholipids of regenerating rat liver]. Biokhimiia., 41, 538–542
    1. Crabtree H.G. (1929). Observations on the carbohydrate metabolism of tumours. Biochem. J., 23, 536–545
    1. Redman E.K., et al. (2013) Role of p90(RSK) in regulating the Crabtree effect: implications for cancer. Biochem. Soc. Trans., 41, 124–126
    1. Díaz-Ruiz R., et al. (2008) Mitochondrial oxidative phosphorylation is regulated by fructose 1,6-bisphosphate. A possible role in Crabtree effect induction? J. Biol. Chem., 283, 26948–26955
    1. Diaz-Ruiz R., et al. (2011) The Warburg and Crabtree effects: on the origin of cancer cell energy metabolism and of yeast glucose repression. Biochim. Biophys. Acta, 1807, 568–576
    1. Holleran A.L., et al. (1995) Glutamine metabolism in AS-30D hepatoma cells. Evidence for its conversion into lipids via reductive carboxylation. Mol. Cell. Biochem., 152, 95–101
    1. Fendt S.M., et al. (2013) Reductive glutamine metabolism is a function of the α-ketoglutarate to citrate ratio in cells. Nat. Commun., 4, 2236.
    1. Pollard P.J., et al. (2003). The TCA cycle and tumorigenesis: the examples of fumarate hydratase and succinate dehydrogenase. Ann. Med., 35, 632–639
    1. Gottlieb E., et al. (2005). Mitochondrial tumour suppressors: a genetic and biochemical update. Nat. Rev. Cancer, 5, 857–866
    1. Seyfried T.N. (2012). Cancer models. In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 31–46
    1. Ecsedy J.A., et al. (1999). Expression of mouse sialic acid on gangliosides of a human glioma grown as a xenograft in SCID mice. J. Neurochem., 73, 254–259
    1. Chou H.H., et al. (1998). A mutation in human CMP-sialic acid hydroxylase occurred after the Homo-Pan divergence. Proc. Natl Acad. Sci. U. S. A., 95, 11751–11756
    1. Martin M.J., et al. (2005). Human embryonic stem cells express an immunogenic nonhuman sialic acid. Nat. Med., 11, 228–232
    1. Davies B., et al. (1993) Physiological parameters in laboratory animals and humans. Pharm. Res., 10, 1093–1095
    1. Mahoney L.B., et al. (2006). Caloric restriction in C57BL/6J mice mimics therapeutic fasting in humans. Lipids Health Dis., 5, 13.
    1. Tentler J.J., et al. (2012) Patient-derived tumour xenografts as models for oncology drug development. Nat. Rev. Clin. Oncol., 9, 338–350
    1. Chaparro R.J., et al. (2006). Nonobese diabetic mice express aspects of both type 1 and type 2 diabetes. Proc. Natl Acad. Sci. U. S. A., 103, 12475–12480
    1. Seyfried T.N. (2012). Mitochondrial respiratory dysfunction and the extrachromosomal origin of cancer. In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 253–259
    1. Hu Y., et al. (2012). K-ras(G12V) transformation leads to mitochondrial dysfunction and a metabolic switch from oxidative phosphorylation to glycolysis. Cell Res., 22, 399–412
    1. Neuzil J., et al. (2012) K-Ras and mitochondria: dangerous liaisons. Cell Res., 22, 285–287
    1. Szent-Györgyi A. (1977). The living state and cancer. Proc. Natl Acad. Sci. U. S. A., 74, 2844–2847
    1. Pawelek J.M., et al. (2008). The cancer cell–leukocyte fusion theory of metastasis. Adv. Cancer Res., 101, 397–444
    1. Seyfried T.N., et al. (2013) On the origin of cancer metastasis. Crit. Rev. Oncog., 18, 43–73
    1. Powell A.E., et al. (2011). Fusion between Intestinal epithelial cells and macrophages in a cancer context results in nuclear reprogramming. Cancer Res., 71, 1497–1505
    1. Pawelek J.M. (2005). Tumour-cell fusion as a source of myeloid traits in cancer. Lancet Oncol., 6, 988–993
    1. Malkin D., et al. (1990). Germ line p53 mutations in a familial syndrome of breast cancer, sarcomas, and other neoplasms. Science, 250, 1233–1238
    1. Funes J.M., et al. (2007). Transformation of human mesenchymal stem cells increases their dependency on oxidative phosphorylation for energy production. Proc. Natl Acad. Sci. U. S. A., 104, 6223–6228
    1. Sung H.J., et al. (2011). Mitochondrial respiration protects against oxygen-associated DNA damage. Nat Commun, 1, 1–8
    1. Lago C.U., et al. (2011) p53, aerobic metabolism, and cancer. Antioxid. Redox Signal., 15, 1739–1748
    1. Matoba S., et al. (2006). p53 regulates mitochondrial respiration. Science, 312, 1650–1653
    1. Zhou S., et al. (2003). Mitochondrial impairment in p53-deficient human cancer cells. Mutagenesis, 18, 287–292
    1. Lee A.C., et al. (1999). Ras proteins induce senescence by altering the intracellular levels of reactive oxygen species. J. Biol. Chem., 274, 7936–7940
    1. Weinberg F., et al. (2010). Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl Acad. Sci. U. S. A., 107, 8788–8793
    1. Yang D., et al. (2010). Impairment of mitochondrial respiration in mouse fibroblasts by oncogenic H-RAS(Q61L). Cancer Biol. Ther., 9, 122–133
    1. Moiseeva O., et al. (2009). Mitochondrial dysfunction contributes to oncogene-induced senescence. Mol. Cell. Biol., 29, 4495–4507
    1. Seyfried T.N. (2012). Respiratory insufficiency, the retrograde response, and the origin of cancer. In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 177–194
    1. Guha M., et al. (2013) Mitochondrial retrograde signaling at the crossroads of tumor bioenergetics, genetics and epigenetics. Mitochondrion, 13, 577–591
    1. Singh K.K., et al. (2005). Inter-genomic cross talk between mitochondria and the nucleus plays an important role in tumorigenesis. Gene, 354, 140–146
    1. Jazwinski S.M. (2005). The retrograde response links metabolism with stress responses, chromatin-dependent gene activation, and genome stability in yeast aging. Gene, 354, 22–27
    1. Nargund A.M., et al. (2012). Mitochondrial import efficiency of ATFS-1 regulates mitochondrial UPR activation. Science, 337, 587–590
    1. Al Mamun A.A., et al. (2012). Identity and function of a large gene network underlying mutagenic repair of DNA breaks. Science, 338, 1344–1348
    1. Dang C.V. (2010). Glutaminolysis: supplying carbon or nitrogen or both for cancer cells? Cell Cycle, 9, 3884–3886
    1. Bissell M.J., et al. (2011). Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat. Med., 17, 320–329
    1. Gatenby R.A., et al. (2007). Glycolysis in cancer: a potential target for therapy. Int. J. Biochem. Cell Biol., 39, 1358–1366
    1. Husain Z., et al. (2013) Tumor-derived lactate modifies antitumor immune response: effect on myeloid-derived suppressor cells and NK cells. J. Immunol., 191, 1486–1495
    1. Liu E.T. (2013). Grappling with cancer. Science, 339, 1493.
    1. Pennisi E. (2013). Steering cancer genomics into the fast lane. Science, 339, 1540–1542
    1. Stratton M.R., et al. (2009). The cancer genome. Nature, 458, 719–724
    1. Crespi B., et al. (2005). Evolutionary biology of cancer. Trends Ecol. Evol., 20, 545–552
    1. Merlo L.M., et al. (2006). Cancer as an evolutionary and ecological process. Nat. Rev. Cancer, 6, 924–935
    1. Davies P.C., et al. (2011). Cancer tumors as Metazoa 1.0: tapping genes of ancient ancestors. Phys. Biol., 8, 015001.
    1. Mayer E. (1982). Evolution before Darwin. In The Growth of Biological Thought: Diversity, Evolution, and Inheritance. Belknap Harvard, Cambridge, MA, pp. 343–362
    1. Handel A.E., et al. (2010). Is Lamarckian evolution relevant to medicine? BMC Med. Genet., 11, 73.
    1. Koonin E.V., et al. (2009). Is evolution Darwinian or/and Lamarckian? Biol. Direct, 4, 42.
    1. Smiraglia D.J., et al. (2008). A novel role for mitochondria in regulating epigenetic modification in the nucleus. Cancer Biol. Ther., 7, 1182–1190
    1. Minocherhomji S., et al. (2012) Mitochondrial regulation of epigenetics and its role in human diseases. Epigenetics, 7, 326–334
    1. Feinberg A.P., et al. (2004). The history of cancer epigenetics. Nat. Rev. Cancer, 4, 143–153
    1. Pawelek J.M. (2000). Tumour cell hybridization and metastasis revisited. Melanoma Res., 10, 507–514
    1. Huysentruyt L.C., et al. (2010). Perspectives on the mesenchymal origin of metastatic cancer. Cancer Metastasis Rev., 29, 695–707
    1. Holmgren L., et al. (1999). Horizontal transfer of DNA by the uptake of apoptotic bodies. Blood, 93, 3956–3963
    1. Dziurzynski K., et al. ; HCMV and Gliomas Symposium (2012) Consensus on the role of human cytomegalovirus in glioblastoma. Neuro. Oncol., 14, 246–255
    1. Detmer S.A., et al. (2007). Functions and dysfunctions of mitochondrial dynamics. Nat. Rev. Mol. Cell Biol., 8, 870–879
    1. Xu R.H., et al. (2005). Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res., 65, 613–621
    1. VanItallie T.B., et al. (2003). Ketones: metabolism’s ugly duckling. Nutr. Rev., 61, 327–341
    1. Drenick E.J., et al. (1972) Resistance to symptomatic insulin reactions after fasting. J. Clin. Invest., 51, 2757–2762
    1. Veech R.L. (2004). The therapeutic implications of ketone bodies: the effects of ketone bodies in pathological conditions: ketosis, ketogenic diet, redox states, insulin resistance, and mitochondrial metabolism. Prostaglandins. Leukot. Essent. Fatty Acids, 70, 309–319
    1. Krebs H.A., et al. (1971). The role of ketone bodies in caloric homeostasis. Adv. Enzyme Reg., 9, 387–409
    1. Bonuccelli G., et al. (2010). Ketones and lactate “fuel” tumor growth and metastasis: evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle, 9, 3506–3514
    1. Seyfried T.N. (2012). Metabolic management of cancer. In Cancer As a Metabolic Disease: On the Origin, Management, and Prevention of Cancer. John Wiley & Sons, Hoboken, NJ, pp. 291–354
    1. Maurer G.D., et al. (2011). Differential utilization of ketone bodies by neurons and glioma cell lines: a rationale for ketogenic diet as experimental glioma therapy. BMC Cancer, 11, 315.
    1. Skinner R., et al. (2009). Ketone bodies inhibit the viability of human neuroblastoma cells. J. Pediatr. Surg., 44, 212–6; discussion 216
    1. Seyfried T.N., et al. (2003). Role of glucose and ketone bodies in the metabolic control of experimental brain cancer. Br. J. Cancer, 89, 1375–1382
    1. Jiang Y.S., et al. (2013) Caloric restriction reduces edema and prolongs survival in a mouse glioma model. J. Neurooncol., 114, 25–32
    1. Mukherjee P., et al. (2004). Antiangiogenic and proapoptotic effects of dietary restriction on experimental mouse and human brain tumors. Clin. Cancer Res., 10, 5622–5629
    1. Mukherjee P., et al. (2002). Dietary restriction reduces angiogenesis and growth in an orthotopic mouse brain tumour model. Br. J. Cancer, 86, 1615–1621
    1. Mulrooney T.J., et al. (2011). Influence of caloric restriction on constitutive expression of NF-κB in an experimental mouse astrocytoma. PLoS One, 6, e18085.
    1. Simone B.A., et al. (2013). Selectively starving cancer cells through dietary manipulation: methods and clinical implications. Future Oncol., 9, 959–976
    1. Stafford P., et al. (2010). The ketogenic diet reverses gene expression patterns and reduces reactive oxygen species levels when used as an adjuvant therapy for glioma. Nutr. Metab. (Lond)., 7, 74.
    1. Fine E.J., et al. (2012). Targeting insulin inhibition as a metabolic therapy in advanced cancer: a pilot safety and feasibility dietary trial in 10 patients. Nutrition, 28, 1028–1035
    1. Zuccoli G., et al. (2010). Metabolic management of glioblastoma multiforme using standard therapy together with a restricted ketogenic diet: Case Report. Nutr. Metab. (Lond)., 7, 33.
    1. Nebeling L.C., et al. (1995). Effects of a ketogenic diet on tumor metabolism and nutritional status in pediatric oncology patients: two case reports. J. Am. Coll. Nutr., 14, 202–208
    1. Zhou W., et al. (2007). The calorically restricted ketogenic diet, an effective alternative therapy for malignant brain cancer. Nutr. Metab. (Lond)., 4, 5.
    1. Gottschalk S., et al. (2004) Imatinib (STI571)-mediated changes in glucose metabolism in human leukemia BCR-ABL-positive cells. Clin. Cancer Res., 10, 6661–6668
    1. Zhao Y., et al. (2011). Overcoming trastuzumab resistance in breast cancer by targeting dysregulated glucose metabolism. Cancer Res., 71, 4585–4597
    1. Marsh J., et al. (2008). Akt-dependent proapoptotic effects of dietary restriction on late-stage management of a phosphatase and tensin homologue/tuberous sclerosis complex 2-deficient mouse astrocytoma. Clin. Cancer Res., 14, 7751–7762
    1. Veech R.L., et al. (2001). Ketone bodies, potential therapeutic uses. IUBMB Life, 51, 241–247
    1. Chen Y., et al. (2009). Oxygen consumption can regulate the growth of tumors, a new perspective on the Warburg effect. PLoS One, 4, e7033.
    1. Spitz D.R., et al. (2000). Glucose deprivation-induced oxidative stress in human tumor cells. A fundamental defect in metabolism? Ann. N. Y. Acad. Sci., 899, 349–362
    1. Harrison L., et al. (2004). Hypoxia and anemia: factors in decreased sensitivity to radiation therapy and chemotherapy? Oncologist, 9 (suppl. 5), 31–40
    1. Allen B.G., et al. (2013) Ketogenic diets enhance oxidative stress and radio-chemo-therapy responses in lung cancer xenografts. Clin. Cancer Res., 19, 3905–3913
    1. Abdelwahab M.G., et al. (2012). The ketogenic diet is an effective adjuvant to radiation therapy for the treatment of malignant glioma. PLoS One, 7, e36197.
    1. Seyfried T.N., et al. (2010). Does the existing standard of care increase glioblastoma energy metabolism? Lancet Oncol., 11, 811–813
    1. Yuneva M. (2008). Finding an “Achilles’ heel” of cancer: the role of glucose and glutamine metabolism in the survival of transformed cells. Cell Cycle, 7, 2083–2089
    1. DeBerardinis R.J., et al. (2010). Q’s next: the diverse functions of glutamine in metabolism, cell biology and cancer. Oncogene, 29, 313–324
    1. Lazova R., et al. (2013). A melanoma brain metastasis with a donor-patient hybrid genome following bone marrow transplantation: first evidence for fusion in human cancer. PLoS One, 8, e66731.
    1. Newsholme P. (2001). Why is L-glutamine metabolism important to cells of the immune system in health, postinjury, surgery or infection? J. Nutr., 131(suppl. 9), 2515S–2522S; discussion 2523S.
    1. Mates J.M., et al. (2012). Glutaminase isoenzymes as key regulators in metabolic and oxidative stress against cancer. Current Mol. Med., 12, 1–21
    1. Shelton L.M., et al. (2010). Glutamine targeting inhibits systemic metastasis in the VM-M3 murine tumor model. Int. J. Cancer, 127, 2478–2485
    1. Lim V., et al. (2009). Glutamine prevents DMBA-induced squamous cell cancer. Oral Oncol., 45, 148–155
    1. Vincent M.D. (2011) Cancer: beyond speciation. Adv. Cancer Res., 112, 283–350
    1. Arens N.C., et al. (2008). Press-pulse: a general theory of mass extinction? Paleobiology, 34, 456–471
    1. Ko Y.H., et al. (2004). Advanced cancers: eradication in all cases using 3-bromopyruvate therapy to deplete ATP. Biochem. Biophys. Res. Commun., 324, 269–275
    1. Bonnet S., et al. (2007). A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell, 11, 37–51
    1. Marsh J., et al. (2008). Drug/diet synergy for managing malignant astrocytoma in mice: 2-deoxy-D-glucose and the restricted ketogenic diet. Nutr. Metab. (Lond)., 5, 33.
    1. Michaelis M., et al. (2009). The story of human cytomegalovirus and cancer: increasing evidence and open questions. Neoplasia, 11, 1–9
    1. Yu Y., et al. (2013) Genome-wide identification and characterization of polygalacturonase genes in Cucumis sativus and Citrullus lanatus. Plant Physiol. Biochem., 74C, 263–275
    1. Bozidis P., et al. (2010) Trafficking of UL37 proteins into mitochondrion-associated membranes during permissive human cytomegalovirus infection. J. Virol., 84, 7898–7903
    1. Williamson C.D., et al. (2009) Access of viral proteins to mitochondria via mitochondria-associated membranes. Rev. Med. Virol., 19, 147–164
    1. Dziurzynski K., et al. (2011) Glioma-associated cytomegalovirus mediates subversion of the monocyte lineage to a tumor propagating phenotype. Clin. Cancer Res., 17, 4642–4649
    1. Munzarová M., et al. (1991). Do some malignant melanoma cells share antigens with the myeloid monocyte lineage? Neoplasma, 38, 401–405
    1. Söderberg-Nauclér C., et al. (2013) Survival in patients with glioblastoma receiving valganciclovir. N. Engl. J. Med., 369, 985–986
    1. Clarke K., et al. (2012) Kinetics, safety and tolerability of ®-3-hydroxybutyl ®-3-hydroxybutyrate in healthy adult subjects. Regul. Toxicol. Pharmacol., 63, 401–408
    1. D’Agostino D.P., et al. (2013) Therapeutic ketosis with ketone ester delays central nervous system oxygen toxicity seizures in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol., 304, R829–R836
    1. Lu W., et al. (2012). Novel role of NOX in supporting aerobic glycolysis in cancer cells with mitochondrial dysfunction and as a potential target for cancer therapy. PLoS Biol., 10, e1001326.
    1. de Groof A.J., et al. (2009). Increased OXPHOS activity precedes rise in glycolytic rate in H-RasV12/E1A transformed fibroblasts that develop a Warburg phenotype. Mol. Cancer, 8, 54.
    1. Baracca A., et al. (2010) Mitochondrial Complex I decrease is responsible for bioenergetic dysfunction in K-ras transformed cells. Biochim. Biophys. Acta, 1797, 314–323
    1. Roskelley R.C., et al. (1943). Studies in cancer. VII. Enzyme deficiency in human and experimental cancer. J. Clin. Invest., 22, 743–751
    1. Seyfried T.N., et al. (2008). Targeting energy metabolism in brain cancer with calorically restricted ketogenic diets. Epilepsia, 49 Suppl 8, 114–116
    1. Huysentruyt L.C., et al. (2008). Metastatic cancer cells with macrophage properties: evidence from a new murine tumor model. Int. J. Cancer, 123, 73–84
    1. Poff A.M., et al. (2013). The ketogenic diet and hyperbaric oxygen therapy prolong survival in mice with systemic metastatic cancer. PLoS One, 8, e65522.

Source: PubMed

3
Sottoscrivi