GET THE APP

Acidity of Microenvironment as a Further Driver of Tumor Metaboli
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2017) Volume 8, Issue 1

Acidity of Microenvironment as a Further Driver of Tumor Metabolic Reprogramming

Silvia Peppicelli, Elena Andreucci, Jessica Ruzzolini, Francesca Margheri, Anna Laurenzana, Francesca Bianchini and Lido Calorini*
Department of Experimental and Clinical Biomedical Sciences, University of Florence, Istituto Toscano Tumori, Florence, Italy
*Corresponding Author: Lido Calorini, Dipartimento di Scienze Biomediche Sperimentali e Cliniche “Mario Serio”, Università di Firenze, Viale G.B. Morgagni, 50 I-50134 Firenze, Italy, Tel: 39.0552751286, Fax: 39.0552751281 Email:

Abstract

In the last decade, experimental research has intensely focused on metabolic reprogramming of tumor cells, which contributes to cancer cell adaptation and survival in different and hostile microenvironments. Metabolic reprogramming consists of the switch of tumor cells from aerobic or anaerobic glycolysis to oxidative phosphorylation. A comprehensive vision of the metabolic scenario involving functionally different tumor cell subpopulations was proposed as a necessary premise to the design of new strategies of diagnosis and therapy. Special focus has been put on the role of acidosis of certain tumor regions, a very important although frequently neglected aspect.
Despite the progresses in cancer therapy, the escaping of tumor cancer cells from host defense and relapse of disease still represent main issues in tumor-bearing patients. Indeed, malignant cells are provided with a tremendous plasticity that they exploit to survive, replicate and invade in stressed microenvironments. Such plasticity allows cancer cells to easily modify their properties, including metabolism, switching back and forth from aerobic or anaerobic glycolysis to oxidative phosphorylation (OxPhos). It is well ascertained that a suitable metabolic profile of cancer cells is necessary to sustain tumor growth, local invasion and distant colonization. Thus, cancer metabolism needs to be considered in view of the design of new strategies to control tumor progression.

Keywords: “Warburg effect”; Glucose level; Hypoxia; Hypoxia-inducible factor-1α; Acidosis; Oxidative phosphorylation

Tumor Microenvironment and Metabolic Reprogramming of Tumor Cells

Warburg effect

Proliferating tumor cells have been largely shown to adopt “aerobic glycolysis” as the main metabolic profile, the so-called “Warburg effect” [1]. Indeed, most cancer cells use huge amounts of glucose even when oxygen tension is high enough to sustain mitochondrial respiration in normal cells. A link between oncogenesis and glucose metabolism is the activating mutations in phosphoinositide 3-kinase (PI3K) or overexpression of the AKT oncogenes, promoting expression and localization of the high affinity glucose transporters on the plasma membrane [2]. This is followed by lactic acid fermentation in the cytosol and lactate export from the cell [3]. This alteration of glucose metabolism acquired practical importance in clinical settings following the development of 18-fluorodeoxyglucose-positron emission tomography imaging [4,5]. Furthermore, it is well recognized that a high serum level of lactate dehydrogenase (LDH) represents a biomarker of poor prognosis in different cancers [6].

When Warburg metabolism is favored, a relatively low level of pyruvate is metabolized in the mitochondria and the energy gain is only 2 ATP per molecule of glucose. Thus, fermentation of glucose to lactic acid is an inefficient pathway, but, very important, it is a fast energy supplier (about 100 folds faster than OxPhos). Moreover, the aerobic glycolytic phenotype confers a significant proliferative advantage as it ensures biomass formation and DNA duplication, crucial aspects of proliferation [7]. Indeed, the glycolytic breakdown of glucose generates a number of substrates which turn into “anabolic” precursors for the synthesis of different compounds, such as glucose-6- phosphate for glycogen and ribose 5-phosphate, dihydroxyacetone phosphate for triacylglyceride and phospholipids, and pyruvate for alanine and malate [8]. Nucleotides, amino acids and lipids are indeed synthesized during anabolic reactions [9]. Cancer cells try to reduce the last step of glycolysis that is catalyzed by pyruvate kinase (PK) by up regulating the low-activity dimeric isoform M2 of PK, and thereby facilitating the accumulation of metabolites upstream of pyruvate [10]. In this respect, intermediate components of the glycolytic pathway appear to be more significant than its final product, e.g. pyruvate. To replenish the TCA cycle, due to the limited supply of pyruvate, cancer cells increase the consumption of glutamine [11]. The particular attitude of proliferating cancer cells to use aerobic glycolysis favors the development of a microenvironment in which lactate produced by tumor cells can be taken up by normal stroma cells to regenerate pyruvate, which can be extruded to refuel cancer cells [12].

Therefore, dividing cancer cells, usually exposed to a relatively high oxygen tension, adopt a glycolytic metabolism, followed by increased glucose uptake and lactic acid production.

Hypoxia

Although the use of aerobic glycolysis leads to a lower oxygen usage, promoting oxygen availability for tumor cells located more distant from blood vessels [13], tumor growth within a disorganized vasculature generates an hypoxic microenvironment. Experimental studies have demonstrated that partial pressure of oxygen is near zero between 100 to 200 μm from a vessel (Figure 1), at the variance of glucose exhaustion which is longer, due to the higher diffusion rate of glucose [14]. Tumor cells need to adapt to the above change and this adaptation partly relies on the induction of hypoxia-inducible factor (HIF)-1α, a transcription factor finely regulated at a protein level in an oxygen-sensitive manner [15]. HIF-1α-related genes are critical in inducing reduction of cell proliferation combined with the development of a more aggressive and resistant phenotype, particularly potentiated by an elicited epithelial-to-mesenchymal transition (EMT) program [16]. Indeed, HIF-1α promotion causes loss of E-cadherin [16] and the expression of the met proto-oncogene and TWIST transcription factor [17]. HIF-1α also acts as a central player in tumor metabolism to support hypoxic tumor cell energy supply, driving an anaerobic glycolysis by upregulating glucose transporter isoform 1 (GLUT1), hexokinase (HK1 and HK2, which catalyze the initial and fundamental step of glycolysis), lactate dehydrogenase A (LDHA), and the lactate-extruding monocarboxylate transporter 4 (MCT4) [16,18]. Besides, carbonic anhydrase IX (CAIX) under the influence of HIF-1α prevents the acidification of intracellular pH of cells under hypoxia [19]. HIF-1α blocks pyruvate conversion into acetyl-CoA triggered by pyruvate dehydrogenase (PDH), by upregulating the PDH inhibitor PDH kinase 1 (PDK1) [20,21]. Thus, inhibiting PDH, HIF-1α compromises the entrance into the oxidative metabolism. HIF-1α can also be activated under normoxic conditions by oncogenic signaling pathways, including PI3K, and by mutations in tumor suppressor proteins, such as VHL (von Hippel Lindau), succinate dehydrogenase, fumarate hydratase [22]. c-Myc also cooperates with HIF-1α in the promotion of glycolysis inducing HK2 and PDK1 [23].

clinical-cellular-immunology-Metabolic-reprogramming

Figure 1: Metabolic reprogramming of tumor cells prompted by variability of tumor microenvironment. Data in part from Helmlinger et al. [72].

The result of this metabolic change is that tumor microenvironment is enriched with H+ and lactate generated by hypoxic cancer cells [24].

Acidosis of tumor microenvironment

Both aerobic and anaerobic glycolysis used by proliferating and hypoxic tumor cells cooperate in the generation of an acidic extracellular pH [25], now considered an important hallmark of cancer to be investigated. The use of a new pH-sensitive positron emission tomography (PET) radiotracer allows in vivo determination of tumor pH, which has been showed to be in the range of 6.4-7.0 for the most solid tumors [26]. We cannot exclude that a chaotic vasculature, low lymphatic drainage and high interstitial pressure contribute to the lowering of extracellular pH (pHe). It has been demonstrated that tumor cells are tolerant to a broad range of acidic pHe, while acidification of the microenvironment is toxic for adjacent normal cells [25].

Acidosis and lactic acidosis, generated by protons and lactate extrusion in the extracellular milieu, correlates with a poorer clinical prognosis [27,28]. We have demonstrated that acidity reduces proliferation together with stimulation of apoptosis resistance and an EMT profile in melanoma cells [29]. The EMT phenotype acquired by acidic melanoma cells was characterized by an increased invasiveness and the ability to promote lung colonization of non-acidic counterpart of cells, disclosing a new cooperation among different tumor cell subpopulations [30]. Robey et al. reported that orally-administered bicarbonate to tumor-bearing mice reduces cell dissemination, providing the evidence that increasing tumor pHe to a physiological level is crucial to abrogate progression to metastasis [30]. The acidic tumor microenvironment also represents a major driving force of resistant phenotype to chemo- and radio-therapy [31,32]. Indeed, both the uptake and efficacy of weak base drugs are reduced in low pH environment [33]. Moreover, lactate by itself has been found to induce motility, angiogenesis, radio-resistance and immune escaping, all features contributing to disease progression [34]. Beyond the aggressive features that acidosis confers to cancer cells, recent discoveries show that local acidity, either generated by high proton level or lactic acid, plays a role in tumor metabolic reprogramming, driving cancer cells from glycolysis to OxPhos [35]. Thanks to aerobic and anaerobic glycolytic pathways adopted by proliferating and hypoxic tumor cells, a residual oxygen tension in tissues may sustain an OxPhos metabolism, possibly at a “reduced rate”. It is known that a residual 3.5 mmHg tension of O2 may permit mitochondrial activity (Figure 1) [36]. OxPhos is considered a major sink for protons able to control internal pH, quite different from protons generated during ATP hydrolysis produced by glycolysis, which are not consumed and generate acidosis. A low pHe generates a low pH of intracellular milieu able to contribute to the metabolic switch to OxPhos, inhibiting the HIF-1α-dependent program through a PI3K/Akt/mTOR signal transduction pathway [37]. In accordance with this finding, it has been recently found that pHe influences hypoxia-related gene expression to a large extent [38,39]. Low pHe suppresses the hypoxia induced up-regulation of PDK1, the effector of PDH inhibition and entry into mitochondrial respiration [40,41]. Breast cancer cells, grown in an acidic medium, also express a reduced glycolysis and lactate production and an increased respiratory metabolism [42]. More recently, we proved that both acidosis and lactic acidosis induce OxPhos metabolism in melanoma cells and, importantly, metformin, a mitochondrial complex I inhibitor, selectively targets and kills acidic cancer cells in a dose-dependent manner [35]. Further, glucose exhaustion caused by glycolytic cells and accumulation of AMP-activated protein-kinase favor the metabolic shift of acidic cells toward OxPhos. When low-pH-adapted cells were grown in a high glucose medium, they maintain a higher pyruvate oxidation, indicating that Crabtree effect is not applicable to acidic cells [43]. To sustain resistance to apoptosis, migration and distant dissemination, all energy-demanding processes, and in the absence of a sufficient glucose supply (already used by proliferating and hypoxic cells), acidic cells use alternative substrates such as lactate, free fatty acids and amino acids. Accordingly, acidosis increases both glutaminolysis and fatty acid β- oxidation in oxidative tumor cells contributing to the entry of metabolic intermediates into the TCA cycle and ATP generation [42]. Lactate also promotes glutamine uptake and metabolism in oxidative cancer cells [44].

LeBleu et al. [45] demonstrated that the bioenergetic phenotype of circulating tumor cells, endowed with a high migratory ability and EMT profile, uses OxPhos. This is of the utmost importance in view of our finding that acidic melanoma cells using an OxPhos metabolism are able to promote lung colonization of non-acidic/glycolytic melanoma cells intravenously injected into immunodeficient mice [29]. We may speculate that, when tumor cells reach their target organ thanks to the ability of acidic subpopulations to lodge and resist to new stressors, non-acidic/glycolytic cell subpopulations start to proliferate leading to a new colony. It is also possible that acidic/OxPhos cells, influenced by an adequate blood supply, which may remove protons and lactate from the microenvironment, reprogram their metabolic profile back to aerobic glycolysis, a key to proliferation. EMT plays a fundamental role during development as well as tumor metastasis. Liu et al., studying head and neck squamous cell carcinoma (HNSCC), found that histone H3K9 methyltransferase G9a is essential for the EMT-mediated metastasis [46]. Actually, G9a forms a complex with Snail and mediates Snail-induced transcriptional repression of Ecadherin and EMT [47]. G9a is also essential for the induction of EMT and cancer stem cells (CSC)-like properties in HNSCC [48]. Since recent observation indicates that apart from establishment and maintenance of H3K9me2, G9a also plays a critical role in the maintenance of DNA methylation at certain loci [49], both the epigenetic mechanisms that associated with histone and DNA methylation might be involved in the maintenance of the EMT-mediated metastasis ability in tumor cells. Therefore, G9a might be a promising target for identifying a subset of cancer cells that display CSC-like profile and it might be of interest investigating the expression levels of G9a in cancer cells undergoing the metabolic reprogramming.

To add complexity to the metabolic reprogramming of tumor cells, a metabolic symbiosis among different tumor cell subpopulations, generating and using lactate, may also take place and contribute to tumor cell dissemination [50]. A metabolic cross-talk has been demonstrated, called “reverse Warburg effect”, among cancer-associated fibroblasts (CAF) and prostate or breast cancer cells, identifying a scenario in which lactate generated by glycolytic CAF is used by tumor cells using a respiratory metabolism [51,52]. In addition, we have identified a possible metabolic cross-talk between acidic mesenchymal stem cells (MSC) and melanoma cells. We demonstrated that acidic MSC undergo a metabolic reprogramming to OxPhos, which in turn may favor the removal of lactate produced by glycolytic melanoma cells [53]. Lactate taken up by acidic stromal cells may work reducing acidification of tumor microenvironment and supporting tumor growth.

Overall, whereas the Warburg effect confers growth advantage to cancer cells when oxygen and glucose supply is sufficient, the OxPhos phenotype promoted by lactic acidosis prompts the use of alternative substrates and renders cancer cells more resistant and competent to progress locally and at a distance.

Perspective in diagnosis and therapy of acidic tumor microenvironment

Several findings indicate that measuring pHe in tumors might acquire a great importance for the characterization of tumor aggressiveness. Indeed, the influence that acidic microenvironment exerts on tumor progression and relapse, due to the reduced sensitivity of acidic cancer cells to radio- and chemotherapy [13], led to test several non-invasive optical imaging methods to monitor in vivo tumor pHe. Positron emission tomography was used for the imaging of acidic prostate tumors [54]. Experience was also made using electron paramagnetic resonance spectroscopy [55] and magnetic resonance (MR) spectroscopy [56]. Quite recently, Chen described a method, termed acido-CEST (chemical exchange saturation transfer) MR imaging, to provide information of tumor pHe in preclinical settings of breast carcinoma [57].

On the meantime, many efforts were put in developing combined therapeutic strategies to target the acidic subpopulations of tumors. Among these, are the alkalization of tumor microenvironment with the use of systemic buffers, such as sodium bicarbonate (NaHCO3) [30,58], or use of proton pump inhibitors, such as esomeprazole, which leads to the normalization of pHe [59]. A comprehensive review of pH regulation in tumors is reported by Neri and Supuran [60]. An additional attractive target of acidic cancer cells is CAIX, which is overexpressed in acidic cancer cells in a HIF-1α-independent manner [61]. Several inhibitors have been developed, such as sulfonamides, sulfamates, and sulfamides that bind to Zn2+ ion-containing catalytic site of the enzyme, thus blocking its function [62]. These compounds are currently under investigation to test in vivo efficacy. Treatment of mice transplanted with CAIX-positive mammary tumor cells with novel CAIX specific inhibitors resulted in a significant reduction of tumor growth and metastatic dissemination [63]. Further, weaklyacidic drugs are designed to reach solid tumors in order to release toxins within the acidic microenvironment [64]. Additional delivery systems to selectively reach and target the acidic core of cancers have been developed. Among them, pH- sensitive liposomes, that release chemotherapeutical substances at an acidic pH [65], and the pH low insertion peptides (pHLIP), that consist of soluble peptides able to bind cell membrane in a pH-dependent way [66]. Another interesting strategy is to target the plasticity of cancer metabolic reprogramming with anti-metabolic drugs. Recently, different therapeutic approaches have been developed based on the targeting of OxPhos metabolism of cancer cells by inhibiting tumour-specific alterations of mitochondrial metabolism or by stimulating mitochondrial membrane permeabilization [67,68]. Among the several drugs tested, metformin, one of the most widely used antidiabetic agents, was found to be efficient in various cancers, such as prostate, breast, lung and pancreas [69]. Its anticancer effect is mediated through the activation of LKB1- AMPK pathway, resulting in the inhibition of the mammalian target of rapamycin (mTOR) that is associated with resistance to anticancer drugs [70]. Also, acidic tumor cells, including melanoma cells, seem to be responsive to metformin treatment [35]. Using osteosarcoma as a cancer model, Quattrini et al. [71] found that metformin amplifies the effects of cisplatin, pointing to a novel approach to therapy of tumors that is the combination of conventional therapy with metabolic inhibitors.

Conclusions

In order to block local and distant dissemination of tumor cells, it is of critical importance to target the acidic regions of tumors to revert the aggressive phenotype that acidity confers to cancer cells. In view of the growing knowledge on tumor metabolism and its reprogramming stimulated by an acidic microenvironment, metabolism of acidic cancer cells may represent an interesting target to improve diagnosis and therapy of tumors.

Acknowledgement

We acknowledge the “Istituto Toscano Tumori” and Ente Cassa di Risparmio di Firenze for their financial support.

References

  1. Schulze A, Harris AL (2012) How cancer metabolism is tuned for proliferation and vulnerable to disruption.  Nature 491: 364-373.
  2. Hirschey MD, DeBerardinis RJ, Diehl AM, Drew JE, Frezza C, et al. (2015) Dysregulated metabolism contributes to oncogenesis. Semin Cancer Biol 35: S129-150.
  3. Gatenby RA, Gillies RJ (2004) Why do cancers have high aerobic glycolysis?  Nat Rev Cancer 4: 891-899.
  4. Zhang X, Lin Y, Gillies RJ (2010) Tumor pH and its measurement.  J Nucl Med 51: 1167-1170.
  5. Delbeke D, Coleman RE, Guiberteau MJ, Brown ML, Royal HD, et al. (2006) Procedure guideline for tumor imaging with 18F-FDG PET/CT 1.0.  J Nucl Med 47: 885-895.
  6. Manola J, Atkins M, Ibrahim J, Kirkwood J (2000) Prognostic factors in metastatic melanoma: a pooled analysis of Eastern Cooperative Oncology Group trials. J ClinOncol 18: 3782-3793.
  7. Vander Heiden MG, Cantley LC, Thompson CB (2009) Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324: 1029-1033.
  8. Kroemer G, Pouyssegur J (2008) Tumor cell metabolism: cancer's Achilles' heel.  Cancer Cell 13: 472-482.
  9. Lunt SY, Vander Heiden MG (2011) Aerobic glycolysis: meeting the metabolic requirements of cell proliferation.  Annu Rev Cell DevBiol 27: 441-464.
  10. Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, et al. (2008) The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 452: 230-233.
  11. DeBerardinis RJ, Mancuso A, Daikhin E, Nissim I, Yudkoff M, et al. (2007) Beyond aerobic glycolysis: transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. ProcNatlAcadSci U S A 104: 19345-19350.
  12. Koukourakis MI, Giatromanolaki A, Harris AL, Sivridis E (2006) Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: a metabolic survival role for tumor-associated stroma. Cancer Res 66: 632-637.
  13. Böhme I, Bosserhoff AK (2016) Acidic tumor microenvironment in human melanoma.  Pigment Cell Melanoma Res 29: 508-523.
  14. Brahimi-Horn MC, Pouysségur J (2007) Oxygen, a source of life and stress.  FEBS Lett 581: 3582-3591.
  15. Denko NC (2008) Hypoxia, HIF1 and glucose metabolism in the solid tumour.  Nat Rev Cancer 8: 705-713.
  16. Pouysségur J, Dayan F, Mazure NM (2006) Hypoxia signalling in cancer and approaches to enforce tumour regression.  Nature 441: 437-443.
  17. Yang MH, Wu KJ (2008) TWIST activation by hypoxia inducible factor-1 (HIF-1): implications in metastasis and development.  Cell Cycle 7: 2090-2096.
  18. Semenza GL (2009) Regulation of cancer cell metabolism by hypoxia-inducible factor 1.  Semin Cancer Biol 19: 12-16.
  19. Swietach P, Hulikova A, Vaughan-Jones RD, Harris AL (2010) New insights into the physiological role of carbonic anhydrase IX in tumour pH regulation.  Oncogene 29: 6509-6521.
  20. Kim JW, Tchernyshyov I, Semenza GL, Dang CV (2006) HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab 3: 177-185.
  21. Papandreou I, Cairns RA, Fontana L, Lim AL, Denko NC (2006) HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab 3: 187-197.
  22. Cairns RA, Harris I, McCracken S, Mak TW (2011) Cancer cell metabolism.  Cold Spring HarbSymp Quant Biol 76: 299-311.
  23. Dang CV, Kim JW, Gao P, Yustein J (2008) The interplay between MYC and HIF in cancer.  Nat Rev Cancer 8: 51-56.
  24. Chiche J, Brahimi-Horn MC, Pouysségur J (2010) Tumour hypoxia induces a metabolic shift causing acidosis: a common feature in cancer.  J Cell Mol Med 14: 771-794.
  25. Walenta S, Wetterling M, Lehrke M, Schwickert G, Sundfør K, et al. (2000) High lactate levels predict likelihood of metastases, tumor recurrence, and restricted patient survival in human cervical cancers. Cancer Res 60: 916-921.
  26. Reshetnyak YK, Yao L, Zheng S, Kuznetsov S, Engelman DM, et al. (2011) Measuring tumor aggressiveness and targeting metastatic lesions with fluorescent pHLIP. Mol Imaging Biol 13: 1146-1156.
  27. Mazzio EA, Boukli N, Rivera N, Soliman KF (2012) Pericellular pH homeostasis is a primary function of the Warburg effect: inversion of metabolic systems to control lactate steady state in tumor cells. Cancer Sci 103: 422-432.
  28. Dhup S, Dadhich RK, Porporato PE, Sonveaux P (2012) Multiple biological activities of lactic acid in cancer: influences on tumor growth, angiogenesis and metastasis. Current Pharmaceutical Design 18: 1319-1330.
  29. Peppicelli S, Bianchini F, Torre E, Calorini L (2014) Contribution of acidic melanoma cells undergoing epithelial-tomesenchymal transition to aggressiveness of non-acidic melanoma cells. ClinExp Metastasis 31: 423-433.
  30. Robey IF, Baggett BK, Kirkpatrick ND, Roe DJ, Dosescu J, et al. (2009) Bicarbonate increases tumor pH and inhibits spontaneous metastases. Cancer Research 69: 2260-2268.
  31. Thews O, Gassner B, Kelleher DK, Schwerdt G, Gekle M (2006) Impact of extracellular acidity on the activity of P-glycoprotein and the cytotoxicity of chemotherapeutic drugs.  Neoplasia 8: 143-152.
  32. Rottinger EM, Mendonca M (1982) Radioresistance secondary to low pH in human glial cells and Chinese hamster ovary cells. Int J RadiatOncolBiolPhys8: 1309-1314.
  33. Taylor S, Spugnini EP, Assaraf YG, Azzarito T, Rauch C, et al. (2015) Microenvironment acidity as a major determinant of tumor chemoresistance: Proton pump inhibitors (PPIs) as a novel therapeutic approach. Drug ResistUpdat 23: 69-78.
  34. Hirschhaeuser F, Sattler UG, Mueller-Klieser W (2011) Lactate: a metabolic key player in cancer.  Cancer Res 71: 6921-6925.
  35. Peppicelli S, Toti A, Giannoni E, Bianchini F, Margheri F, et al. (2016) Metformin is also effective on lactic acidosis-exposed melanoma cells switched to oxidative phosphorylation. Cell Cycle 15: 1908-1918.
  36. Weinberg SE, Chandel NS (2015) Targeting mitochondria metabolism for cancer therapy.  Nat ChemBiol 11: 9-15.
  37. Kahn BB, Alquier T, Carling D, Hardie DG (2005) AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab 1: 15-25.
  38. Sørensen BS, Alsner J, Overgaard J, Horsman MR (2007) Hypoxia induced expression of endogenous markers in vitro is highly influenced by pH.RadiotherOncol 83: 362-366.
  39. Sørensen BS, Hao J, Overgaard J, Vorum H, Honoré B, et al. (2005) Influence of oxygen concentration and pH on expression of hypoxia induced genes.  RadiotherOncol 76: 187-193.
  40. Sørensen BS, Busk M, Overgaard J, Horsman MR, Alsner J (2015) Simultaneous Hypoxia and Low Extracellular pH Suppress Overall Metabolic Rate and Protein Synthesis In vitro.  PLoS One 10: e0134955.
  41. Tang X, Lucas JE, Chen JL, LaMonte G, Wu J, et al (2012) Functional interaction between responses to lactic acidosis and hypoxia regulates genomic transcriptional outputs. Cancer Res 72: 491-502.
  42. Lamonte G, Tang X, Chen JL, Wu J, Ding CK, et al. (2013) Acidosis induces reprogramming of cellular metabolism to mitigate oxidative stress.  Cancer Metab 1: 23.
  43. Burd R, Wachsberger PR, Biaglow JE, Wahl ML, Lee I, et al. (2001) Absence of Crabtree effect in human melanoma cells adapted to growth at low pH: reversal by respiratory inhibitors. Cancer Res 61:5630-5635.
  44. Pérez-Escuredo J, Dadhich RK, Dhup S, Cacace A, Van Hée VF, et al. (2016) Lactate promotes glutamine uptake and metabolism in oxidative cancer cells.  Cell Cycle 15: 72-83.
  45. LeBleu VS, O'Connell JT, Gonzalez Herrera KN, Wikman H, Pantel K, et al. (2014) PGC-1a mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat Cell Biol 16: 992-1003.
  46. Liu S, Ye D, Guo W, Yu W, He Y, et al. (2015) G9a is essential for EMT-mediated metastasis and maintenance of cancer stem cell-like characters in head and neck squamous cell carcinoma.  Oncotarget 6: 6887-6901.
  47. Duvall-Noelle N, Karwandyar A, Richmond A, Raman D (2016) LASP-1: a nuclear hub for the UHRF1-DNMT1-G9a-Snail1 complex.  Oncogene 35: 1122-1133.
  48. Lin Y, Dong C, Zhou BP (2014) Epigenetic regulation of EMT: the Snail story.  Curr Pharm Des 20: 1698-1705.
  49. Zhang T, Termanis A, Özkan B, Bao XX, Culley J, et al. (2016) G9a/GLP Complex Maintains Imprinted DNA Methylation in Embryonic Stem Cells.  Cell Rep 15: 77-85.
  50. Semenza GL (2008) Tumor metabolism: cancer cells give and take lactate.  J Clin Invest 118: 3835-3837.
  51. Whitaker-Menezes D, Martinez-Outschoorn UE, Lin Z, Ertel A, Flomenberg N, et al. (2011) Evidence for a stromal-epithelial "lactate shuttle" in human tumors: MCT4 is a marker of oxidative stress in cancer-associated fibroblasts. Cell Cycle 10: 1772-1783.
  52. Fiaschi T, Marini A, Giannoni E, Taddei ML, Gandellini P, et al. (2012) Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay. Cancer Res 72: 5130-5140.
  53. Peppicelli S, Bianchini F, Toti A, Laurenzana A, Fibbi G, et al. (2015) Extracellular acidity strengthens mesenchymal stem cells to promote melanoma progression. Cell Cycle 14: 3088-3100.
  54. Vavere AL, Biddlecombe GB, Spees WM, Garbow JR, Wijesinghe D, et al. (2009) A novel technology for the imaging of acidic prostate tumors by positron emission tomography. Cancer Res 69: 4510-4516.
  55. Ahmad R, Potter LC, Khramtsov VV (2012) Spectral modeling for accelerated pH spectroscopy using EPR.  J MagnReson 218: 86-92.
  56. Gallagher FA, Kettunen MI, Day SE, Hu DE, Ardenkjaer-Larsen JH, et al. (2008) Magnetic resonance imaging of pH in vivo using hyperpolarized 13C-labelled bicarbonate. Nature 453: 940-943.
  57. Chen LQ, Howison CM, Jeffery JJ, Robey IF, Kuo PH, et al. (2014) Evaluations of extracellular pH within in vivo tumors using acidoCEST MRI.  MagnReson Med 72: 1408-1417.
  58. Pilon-Thomas S, Kodumudi KN, El-Kenawi AE, Russell S, Weber AM, et al. (2016) Neutralization of Tumor Acidity Improves Antitumor Responses to Immunotherapy. Cancer Res 76: 1381-1390.
  59. De Milito A, Canese R, Marino ML, Borghi M, Iero M, et al. (2010) pH-dependent antitumor activity of proton pump inhibitors against human melanoma is mediated by inhibition of tumor acidity. Int J Cancer 127: 207-219.
  60. Neri D, Supuran CT (2011) Interfering with pH regulation in tumours as a therapeutic strategy.  Nat Rev Drug Discov 10: 767-777.
  61. Ihnatko R, Kubes M, Takacova M, Sedlakova O, Sedlak J, et al. (2006) Extracellular acidosis elevates carbonic anhydrase IX in human glioblastoma cells via transcriptional modulation that does not depend on hypoxia. Int J Oncol 29: 1025-1033.
  62. Supuran CT (2008) Development of small molecule carbonic anhydrase IX inhibitors.  BJU Int 101 Suppl 4: 39-40.
  63. Lou Y, McDonald PC, Oloumi A, Chia S, Ostlund C, et al. (2011) Targeting tumor hypoxia: suppression of breast tumor growth and metastasis by novel carbonic anhydrase IX inhibitors.  Cancer Res 71: 3364-3376.
  64. Gullotti E, Yeo Y (2009) Extracellularly activated nanocarriers: a new paradigm of tumor targeted drug delivery.  Mol Pharm 6: 1041-1051.
  65. Karanth H, Murthy RS (2007) pH-sensitive liposomes--principle and application in cancer therapy.  J Pharm Pharmacol 59: 469-483.
  66. Deacon JC, Engelman DM, Barrera FN (2015) Targeting acidity in diseased tissues: mechanism and applications of the membrane-inserting peptide, pHLIP. Arch BiochemBiophys 565: 40-48.
  67. Fulda S, Galluzzi L, Kroemer G (2010) Targeting mitochondria for cancer therapy.  Nat Rev Drug Discov 9: 447-464.
  68. Yang Y, Karakhanova S, Hartwig W, D'Haese JG, Philippov PP, et al. (2016) Mitochondria and Mitochondrial ROS in Cancer: Novel Targets for Anticancer Therapy.  J Cell Physiol 231: 2570-2581.
  69. Leone A, Di Gennaro E, Bruzzese F, Avallone A, Budillon A (2014) New perspective for an old antidiabetic drug: metformin as anticancer agent.  Cancer Treat Res 159: 355-376.
  70. Cerezo M, Tomic T, Ballotti R, Rocchi S (2015) Is it time to test biguanide metformin in the treatment of melanoma?  Pigment Cell Melanoma Res 28: 8-20.
  71. Quattrini I, Conti A, Pazzaglia L, Novello C, Ferrari S, et al. (2014) Metformin inhibits growth and sensitizes osteosarcoma cell lines to cisplatin through cell cycle modulation. Oncol Rep 31: 370-375.
  72. Helmlinger G, Yuan F, Dellian M, Jain RK (1997) Interstitial pH and pO2 gradients in solid tumors in vivo: high-resolution measurements reveal a lack of correlation.  Nat Med 3: 177-182.
Citation: Peppicelli S, Andreucci E, Ruzzolini J, Margheri F, Laurenzana A, et al. (2017) Acidity of Microenvironment as a Further Driver of Tumor Metabolic Reprogramming. J Clin Cell Immunol 8:485.

Copyright: © 2017 Peppicelli S, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top