5,693
Views
85
CrossRef citations to date
0
Altmetric
Review Article

Targeting tumour hypoxia to prevent cancer metastasis. From biology, biosensing and technology to drug development: the METOXIA consortium

, , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , & show all
Pages 689-721 | Received 21 Aug 2014, Accepted 15 Sep 2014, Published online: 27 Oct 2014

References

  • Young SD, Marshall RS, Hill RP. Hypoxia induces DNA overreplication and enhances metastatic potential of murine tumor cells. Proc Natl Acad Sci USA 1988;85:9533–7
  • Young SD, Hill RP. Effects of reoxygenation on cells from hypoxic regions of solid tumors: analysis of transplanted murine tumors for evidence of DNA overreplication. Cancer Res 1990;50:5031–8
  • Rofstad EK, Galappathi K, Mathiesen B, Ruud EB. Fluctuating and diffusion-limited hypoxia in hypoxia-induced metastasis. Clin Cancer Res 2007;13:1971–8
  • McKeown SR. Defining normoxia, physoxia and hypoxia in tumours-implications for treatment response. Br J Radiol 2014;87:20130676
  • Connett RJ, Honig CR, Gayeski TE, Brooks GA. Defining hypoxia: a systems view of VO2, glycolysis, energetics, and intracellular PO2. J Appl Physiol (Bethesda, MD) 1990;68:833–42
  • Cairns RA, Kalliomaki T, Hill RP. Acute (cyclic) hypoxia enhances spontaneous metastasis of KHT murine tumors. Cancer Res 2001;61:8903–8
  • Jiang BH, Semenza GL, Bauer C, Marti HH. Hypoxia-inducible factor 1 levels vary exponentially over a physiologically relevant range of O2 tension. Am J Physiol 1996;271:C1172–80
  • Mottram MC. Factor of importance in radiosensitivity of tumours. Br J Radiol 1936;9:606–14
  • Gray LH, Conger AD, Ebert M, et al. The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br J Radiol 1953;26:638–48
  • Thomlinson RH, Gray LH. The histological structure of some human lung cancers and the possible implications for radiotherapy. Br J Cancer 1955;9:539–49
  • Adams GE, Dewey DL. Hydrated electrons and radiobiological sensitisation. Biochem Biophys Res Commun 1963;12:473–7
  • Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metastasis Rev 2007;26:225–39
  • Lu X, Kang Y. Hypoxia and hypoxia-inducible factors: master regulators of metastasis. Clin Cancer Res 2010;16:5928–35
  • Ortiz-Barahona A, Villar D, Pescador N, et al. Genome-wide identification of hypoxia-inducible factor binding sites and target genes by a probabilistic model integrating transcription-profiling data and in silico binding site prediction. Nucleic Acids Res 2010;38:2332–45
  • Chen J. Regulation of tumor initiation and metastatic progression by Eph receptor tyrosine kinases. Adv Cancer Res 2012;114:1–20
  • Choudhry H, Schodel J, Oikonomopoulos S, et al. Extensive regulation of the non-coding transcriptome by hypoxia: role of HIF in releasing paused RNApol2. EMBO Rep 2014;15:70–6
  • Gómez-Maldonado L, Tiana M, Roche O, et al. EFNA3 long non-coding RNAs induced by hypoxia promote metastatic dissemination. Oncogene 2014. [Epub ahead of print]. doi: 10.1038/onc.2014.200
  • Shweiki D, Neeman M, Itin A, Keshet E. Induction of vascular endothelial growth factor expression by hypoxia and by glucose deficiency in multicell spheroids: implications for tumor angiogenesis. Proc Natl Acad Sci USA 1995;92:768–72
  • Bienes-Martinez R, Ordonez A, Feijoo-Cuaresma M, et al. Autocrine stimulation of clear-cell renal carcinoma cell migration in hypoxia via HIF-independent suppression of thrombospondin-1. Sci Rep 2012;2:788
  • Good DJ, Polverini PJ, Rastinejad F, et al. A tumor suppressor-dependent inhibitor of angiogenesis is immunologically and functionally indistinguishable from a fragment of thrombospondin. Proc Natl Acad Sci USA 1990;87:6624–8
  • Henkin J, Volpert OV. Therapies using anti-angiogenic peptide mimetics of thrombospondin-1. Expert Opin Therapeut Targets 2011;15:1369–86
  • Veliceasa D, Ivanovic M, Hoepfner FT, et al. Transient potential receptor channel 4 controls thrombospondin-1 secretion and angiogenesis in renal cell carcinoma. FEBS J 2007;274:6365–77
  • Zubac DP, Bostad L, Kihl B, et al. The expression of thrombospondin-1 and p53 in clear cell renal cell carcinoma: its relationship to angiogenesis, cell proliferation and cancer specific survival. J Urol 2009;182:2144–9
  • Villar D, Ortiz-Barahona A, Gomez-Maldonado L, et al. Cooperativity of stress-responsive transcription factors in core hypoxia-inducible factor binding regions. PLoS One 2012;7:e45708
  • Tiana M, Villar D, Perez-Guijarro E, et al. A role for insulator elements in the regulation of gene expression response to hypoxia. Nucleic Acids Res 2012;40:1916–27
  • Benej M, Pastorekova S, Pastorek J. Carbonic anhydrase IX: regulation and role in cancer. Sub-Cellular Biochem 2014;75:199–219
  • Wykoff CC, Beasley NJ, Watson PH, et al. Hypoxia-inducible expression of tumor-associated carbonic anhydrases. Cancer Res 2000;60:7075–83
  • Ditte P, Dequiedt F, Svastova E, et al. Phosphorylation of carbonic anhydrase IX controls its ability to mediate extracellular acidification in hypoxic tumors. Cancer Res 2011;71:7558–67
  • Sedlakova O, Svastova E, Takacova M, et al. Carbonic anhydrase IX, a hypoxia-induced catalytic component of the pH regulating machinery in tumors. Frontiers Physiol 2014;4:400
  • Svastova E, Witarski W, Csaderova L, et al. Carbonic anhydrase IX interacts with bicarbonate transporters in lamellipodia and increases cell migration via its catalytic domain. J Biol Chem 2012;287:3392–402
  • Csaderova L, Debreova M, Radvak P, et al. The effect of carbonic anhydrase IX on focal contacts during cell spreading and migration. Frontier Physiol 2013;4:271
  • Zatovicova M, Jelenska L, Hulikova A, et al. Carbonic anhydrase IX as an anticancer therapy target: preclinical evaluation of internalizing monoclonal antibody directed to catalytic domain. Curr Pharm Des 2010;16:3255–63
  • Winum JY, Scozzafava A, Montero JL, Supuran CT. Inhibition of carbonic anhydrase IX: a new strategy against cancer. Anti-Cancer Agents Med Chem 2009;9:693–702
  • Dubois L, Peeters S, Lieuwes NG, et al. Specific inhibition of carbonic anhydrase IX activity enhances the in vivo therapeutic effect of tumor irradiation. Radiother Oncol 2011;99:424–31
  • Lou Y, McDonald PC, Oloumi A, et al. Targeting tumor hypoxia: suppression of breast tumor growth and metastasis by novel carbonic anhydrase IX inhibitors. Cancer Res 2011;71:3364–76
  • Ditte Z, Ditte P, Labudova M, et al. Carnosine inhibits carbonic anhydrase IX-mediated extracellular acidosis and suppresses growth of HeLa tumor xenografts. BMC Cancer 2014;14:358
  • Fukuda R, Zhang H, Kim JW, et al. HIF-1 regulates cytochrome oxidase subunits to optimize efficiency of respiration in hypoxic cells. Cell 2007;129:111–22
  • Tello D, Balsa E, Acosta-Iborra B, et al. Induction of the mitochondrial NDUFA4L2 protein by HIF-1alpha decreases oxygen consumption by inhibiting complex I activity. Cell Metabol 2011;14:768–79
  • Balsa E, Marco R, Perales-Clemente E, et al. NDUFA4 is a subunit of complex IV of the mammalian electron transport chain. Cell Metabol 2012;16:378–86
  • Balsa E, Acosta-Iborra B, Tello D, et al. HIF-dependent vs. independent mechanisms that regulate Complex IV and oxygen consumption in hypoxia. 2014; submitted for publication
  • Wouters BG, Koritzinsky M. Hypoxia signalling through mTOR and the unfolded protein response in cancer. Nat Rev Cancer 2008;8:851–64
  • Koritzinsky M, Magagnin MG, van den Beucken T, et al. Gene expression during acute and prolonged hypoxia is regulated by distinct mechanisms of translational control. Embo J 2006;25:1114–25
  • Koritzinsky M, Seigneuric R, Magagnin MG, et al. The hypoxic proteome is influenced by gene-specific changes in mRNA translation. Radiother Oncol 2005;76:177–86
  • Bi M, Naczki C, Koritzinsky M, et al. ER stress-regulated translation increases tolerance to extreme hypoxia and promotes tumor growth. Embo J 2005;24:3470–81
  • Walter P, Ron D. The unfolded protein response: from stress pathway to homeostatic regulation. Science (New York) 2011;334:1081–6
  • Harding HP, Zhang Y, Zeng H, et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell 2003;11:619–33
  • Koritzinsky M, Levitin F, van den Beucken T, et al. Two phases of disulfide bond formation have differing requirements for oxygen. J Cell Biol 2013;203:615–27
  • Braakman I, Bulleid NJ. Protein folding and modification in the mammalian endoplasmic reticulum. Ann Rev Biochem 2011;80:71–99
  • Tu BP, Weissman JS. The FAD- and O(2)-dependent reaction cycle of Ero1-mediated oxidative protein folding in the endoplasmic reticulum. Mol Cell 2002;10:983–94
  • Rouschop KMA, van den Beucken T, Dubois L, et al. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. J Clin Invest 2010;120:127–41
  • Zhang HF, Bosch-Marce M, Shimoda LA, et al. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J Biol Chem 2008;283:10892–903
  • Papandreou I, Lim AL, Laderoute K, Denko NC. Hypoxia signals autophagy in tumor cells via AMPK activity, independent of HIF-1, BNIP3, and BNIP3L. Cell Death Differ 2008;15:1572–81
  • Klionsky DJ, Abeliovich H, Agostinis P, et al. Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy 2008;4:151–75
  • Ding WX, Ni HM, Gao WT, et al. Linking of autophagy to ubiquitin-proteasome system is important for the regulation of endoplasmic reticulum stress and cell viability. Am J Pathol 2007;171:513–24
  • Iwata A, Christianson JC, Bucci M, et al. Increased susceptibility of cytoplasmic over nuclear polyglutamine aggregates to autophagic degradation. Proc Natl Acad Sci USA 2005;102:13135–40
  • Rzymski T, Milani M, Pike L, et al. Regulation of autophagy by ATF4 in response to severe hypoxia. Oncogene 2010;29:4424–35
  • Schaaf MB, Cojocari D, Keulers TG, et al. The autophagy associated gene, ULK1, promotes tolerance to chronic and acute hypoxia. Radiother Oncol 2013;108:529–34
  • Pike LR, Singleton DC, Buffa F, et al. Transcriptional up-regulation of ULK1 by ATF4 contributes to cancer cell survival. Biochem J 2013;449:389–400
  • Rouschop KMA, Ramaekers CHMA, Schaaf MBE, et al. Autophagy is required during cycling hypoxia to lower production of reactive oxygen species. Radiother Oncol 2009;92:411–16
  • Dickhout JG, Carlisle RE, Jerome DE, et al. Integrated stress response modulates cellular redox state via induction of cystathionine gamma-lyase cross-talk between integrated stress response and thiol metabolism. J Biol Chem 2012;287:7603–14
  • Rouschop KM, Dubois Lj Fau, Keulers TG, et al. PERK/eIF2alpha signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS. Proc Natl Acad Sci USA 2013;110:4622–7
  • Fyles A, Milosevic M, Hedley D, et al. Tumor hypoxia has independent predictor impact only in patients with node-negative cervix cancer. J Clin Oncol 2002;20:680–7
  • Cairns RA, Hill RP. Acute hypoxia enhances spontaneous lymph node metastasis in an orthotopic murine model of human cervical carcinoma. Cancer Res 2004;64:2054–61
  • Mujcic H, Nagelkerke A Fau, Rouschop KMA, et al. Hypoxic activation of the PERK/eIF2alpha arm of the unfolded protein response promotes metastasis through induction of LAMP3. Clin Cancer Res 2013;19:6126–37
  • Mujcic H, Rzymski T, Rouschop KM, et al. Hypoxic activation of the unfolded protein response (UPR) induces expression of the metastasis-associated gene LAMP3. Radiother Oncol 2009;92:450–9
  • Greenman C, Stephens P, Smith R, et al. Patterns of somatic mutation in human cancer genomes. Nature 2007;446:153–8
  • Cancer Genome Atlas N. Comprehensive molecular portraits of human breast tumours. Nature 2012;490:61–70
  • Romero-Ramirez L, Cao HB, Nelson D, et al. XBP1 is essential for survival under hypoxic conditions and is required for tumor growth. Cancer Res 2004;64:5943–7
  • Cojocari D, Vellanki RN, Sit B, et al. New small molecule inhibitors of UPR activation demonstrate that PERK, but not IRE1 alpha signaling is essential for promoting adaptation and survival to hypoxia. Radiother Oncol 2013;108:541–7
  • Atkins C, Liu Q, Minthorn E, et al. Characterization of a novel PERK kinase inhibitor with antitumor and antiangiogenic activity. Cancer Res 2013;73:1993–2002
  • Wang H, Blais J, Ron D, Cardozo T. Structural determinants of PERK inhibitor potency and selectivity. Chem Biol Drug Design 2010;76:480–95
  • Cross BC, Bond PJ, Sadowski PG, et al. The molecular basis for selective inhibition of unconventional mRNA splicing by an IRE1-binding small molecule. Proc Natl Acad Sci USA 2012;109:E869–78
  • Fribley AM, Cruz PG, Miller JR, et al. Complementary cell-based high-throughput screens identify novel modulators of the unfolded protein response. J Biomol Screen 2011;16:825–35
  • Papandreou I, Denko NC, Olson M, et al. Identification of an Ire1alpha endonuclease specific inhibitor with cytotoxic activity against human multiple myeloma. Blood 2011;117:1311–14
  • Korennykh AV, Egea PF, Korostelev AA, et al. The unfolded protein response signals through high-order assembly of Ire1. Nature 2009;457:687–93
  • Wang L, Perera Bg Fau, Hari SB, et al. Divergent allosteric control of the IRE1alpha endoribonuclease using kinase inhibitors. Nat Chem Biol 2012;8:982–9
  • Axten JM, MedinaJr Fau, Feng Y, et al. Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-p yrrolo[2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK). J Med Chem 2012;55:7193–207
  • Sidrauski C, Acosta-Alvear D, Khoutorsky A, et al. Pharmacological brake-release of mRNA translation enhances cognitive memory. eLife 2013;2:e00498. doi: 10.7554/eLife.00498
  • Hawkins BJ, Madesh M, Kirkpatrick CJ, Fisher AB. Superoxide flux in endothelial cells via the chloride channel-3 mediates intracellular signaling. Mol Biol Cell 2007;18:2002–12
  • Han D, Antunes F, Canali R, et al. Voltage-dependent anion channels control the release of the superoxide anion from mitochondria to cytosol. J Biol Chem 2003;278:5557–63
  • Gorlach A, Kietzmann T. Superoxide and derived reactive oxygen species in the regulation of hypoxia-inducible factors. Methods Enzymol 2007;435:421–46
  • Hybertson BM, Gao B, Bose SK, McCord JM. Oxidative stress in health and disease: the therapeutic potential of Nrf2 activation. Mol Aspects Med 2011;32:234–46
  • Muller FL, Lustgarten MS, Jang Y, et al. Trends in oxidative aging theories. Free Radic Biol Med 2007;43:477–503
  • Gorlach A, Kietzmann T, Hess J. Redox signaling through NADPH oxidases: involvement in vascular proliferation and coagulation. Ann New York Acad Sci 2002;973:505–7
  • Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 2007;87:245–313
  • BelAiba RS, Djordjevic T, Petry A, et al. NOX5 variants are functionally active in endothelial cells. Free Radic Biol Med 2007;42:446–59
  • Lambeth JD, Kawahara T, Diebold B. Regulation of Nox and Duox enzymatic activity and expression. Free Radic Biol Med 2007;43:319–31
  • Petry A, Weitnauer M, Gorlach A. Receptor activation of NADPH oxidases. Antioxid Redox Signal 2010;13:467–87
  • Wang GL, Jiang BH, Semenza GL. Effect of altered redox states on expression and DNA-binding activity of hypoxia-inducible factor 1. Biochem Biophys Res Commun 1995;212:550–6
  • Huang LE, Arany Z, Livingston DM, Bunn HF. Activation of hypoxia-inducible transcription factor depends primarily upon redox-sensitive stabilization of its alpha subunit. J Biol Chem 1996;271:32253–9
  • Welsh SJ, Bellamy WT, Briehl MM, Powis G. The redox protein thioredoxin-1 (Trx-1) increases hypoxia-inducible factor 1alpha protein expression: Trx-1 overexpression results in increased vascular endothelial growth factor production and enhanced tumor angiogenesis. Cancer Res 2002;62:5089–95
  • Liu Q, Berchner-Pfannschmidt U, Moller U, et al. A Fenton reaction at the endoplasmic reticulum is involved in the redox control of hypoxia-inducible gene expression. Proc Natl Acad Sci USA 2004;101:4302–7
  • Wang M, Kirk JS, Venkataraman S, et al. Manganese superoxide dismutase suppresses hypoxic induction of hypoxia-inducible factor-1alpha and vascular endothelial growth factor. Oncogene 2005;24:8154–66
  • BelAiba RS, Djordjevic T, Bonello S, et al. Redox-sensitive regulation of the HIF pathway under non-hypoxic conditions in pulmonary artery smooth muscle cells. Biol Chem 2004;385:249–57
  • Bonello S, Zahringer C, BelAiba RS, et al. Reactive oxygen species activate the HIF-1alpha promoter via a functional NFkappaB site. Arterioscler Thromb Vasc Biol 2007;27:755–61
  • Diebold I, Flugel D, Becht S, et al. The hypoxia-inducible factor-2alpha is stabilized by oxidative stress involving NOX4. Antioxidants Redox Signal 2010;13:425–36
  • Gorlach A, Diebold I, Schini-Kerth VB, et al. Thrombin activates the hypoxia-inducible factor-1 signaling pathway in vascular smooth muscle cells: role of the p22(phox)-containing NADPH oxidase. Circulation Res 2001;89:47–54
  • Chandel NS, McClintock DS, Feliciano CE, et al. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: a mechanism of O2 sensing. J Biol Chem 2000;275:25130–8
  • Kietzmann T, Samoylenko A, Roth U, Jungermann K. Hypoxia-inducible factor-1 and hypoxia response elements mediate the induction of plasminogen activator inhibitor-1 gene expression by insulin in primary rat hepatocytes. Blood 2003;101:907–14
  • Tacchini L, De Ponti C, Matteucci E, et al. Hepatocyte growth factor-activated NF-kappaB regulates HIF-1 activity and ODC expression, implicated in survival, differently in different carcinoma cell lines. Carcinogenesis 2004;25:2089–100
  • Richard DE, Berra E, Pouyssegur J. Nonhypoxic pathway mediates the induction of hypoxia-inducible factor 1alpha in vascular smooth muscle cells. J Biol Chem 2000;275:26765–71
  • Diebold I, Petry A, Sabrane K, et al. The HIF1 target gene NOX2 promotes angiogenesis through urotensin-II. J Cell Sci 2012;125:956–64
  • Stiehl DP, Jelkmann W, Wenger RH, Hellwig-Burgel T. Normoxic induction of the hypoxia-inducible factor 1alpha by insulin and interleukin-1beta involves the phosphatidylinositol 3-kinase pathway. FEBS Lett 2002;512:157–62
  • Haddad JJ, Land SC. A non-hypoxic, ROS-sensitive pathway mediates TNF-alpha-dependent regulation of HIF-1alpha. FEBS Lett 2001;505:269–74
  • Diebold I, Djordjevic T, Hess J, Gorlach A. Rac-1 promotes pulmonary artery smooth muscle cell proliferation by upregulation of plasminogen activator inhibitor-1: role of NFkappaB-dependent hypoxia-inducible factor-1alpha transcription. Thromb Haemost 2008;100:1021–8
  • Petry A, BelAiba RS, Weitnauer M, Gorlach A. Inhibition of endothelial nitric oxyde synthase increases capillary formation via Rac1-dependent induction of hypoxia-inducible factor-1alpha and plasminogen activator inhibitor-1. Thromb Haemost 2012;108:849–62
  • Diebold I, Djordjevic T, Petry A, et al. Phosphodiesterase 2 mediates redox-sensitive endothelial cell proliferation and angiogenesis by thrombin via Rac1 and NADPH oxidase 2. Circ Res 2009;104:1169–77
  • Block K, Gorin Y. Aiding and abetting roles of NOX oxidases in cellular transformation. Nat Rev Cancer 2012;12:627–37
  • Moon EJ, Sonveaux P, Porporato PE, et al. NADPH oxidase-mediated reactive oxygen species production activates hypoxia-inducible factor-1 (HIF-1) via the ERK pathway after hyperthermia treatment. Proc Natl Acad Sci USA 2010;107:20477–82
  • Goyal P, Weissmann N, Grimminger F, et al. Upregulation of NAD(P)H oxidase 1 in hypoxia activates hypoxia-inducible factor 1 via increase in reactive oxygen species. Free Radic Biol Med 2004;36:1279–88
  • Xia C, Meng Q, Liu LZ, et al. Reactive oxygen species regulate angiogenesis and tumor growth through vascular endothelial growth factor. Cancer Res 2007;67:10823–30
  • Block K, Gorin Y, Hoover P, et al. NAD(P)H oxidases regulate HIF-2alpha protein expression. J Biol Chem 2007;282:8019–26
  • Maranchie JK, Zhan Y. NOX4 is critical for hypoxia-inducible factor 2-alpha transcriptional activity in von Hippel–Lindau-deficient renal cell carcinoma. Cancer Res 2005;65:9190–3
  • Block K, Gorin Y, New DD, et al. The NADPH oxidase subunit p22phox inhibits the function of the tumor suppressor protein tuberin. Am J Pathol 2010;176:2447–55
  • Gerald D, Berra E, Frapart YM, et al. JunD reduces tumor angiogenesis by protecting cells from oxidative stress. Cell 2004;118:781–94
  • Knowles HJ, Raval RR, Harris AL, Ratcliffe PJ. Effect of ascorbate on the activity of hypoxia-inducible factor in cancer cells. Cancer Res 2003;63:1764–8
  • Epstein AC, Gleadle JM, McNeill LA, et al. C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 2001;107:43–54
  • Page EL, Chan DA, Giaccia AJ, et al. Hypoxia-inducible factor-1alpha stabilization in nonhypoxic conditions: role of oxidation and intracellular ascorbate depletion. Mol Biol Cell 2008;19:86–94
  • Flashman E, Davies SL, Yeoh KK, Schofield CJ. Investigating the dependence of the hypoxia-inducible factor hydroxylases (factor inhibiting HIF and prolyl hydroxylase domain 2) on ascorbate and other reducing agents. Biochem J 2010;427:135–42
  • Mecinovic J, Chowdhury R, Flashman E, Schofield CJ. Use of mass spectrometry to probe the nucleophilicity of cysteinyl residues of prolyl hydroxylase domain 2. Anal Biochem 2009;393:215–21
  • Nytko KJ, Maeda N, Schlafli P, et al. Vitamin C is dispensable for oxygen sensing in vivo. Blood 2011;117:5485–93
  • Gorlach A. Regulation of HIF-1 at the transcriptional level. Curr Pharm Des 2009;15:3844–52
  • Rius J, Guma M, Schachtrup C, et al. NF-kappaB links innate immunity to the hypoxic response through transcriptional regulation of HIF-1alpha. Nature 2008;453:807–11
  • van Uden P, Kenneth NS, Rocha S. Regulation of hypoxia-inducible factor-1alpha by NF-kappaB. Biochem J 2008;412:477–84
  • Gorlach A, Bonello S. The cross-talk between NF-kappaB and HIF-1: further evidence for a significant liaison. Biochem J 2008;412:e17–19
  • Belaiba RS, Bonello S, Zahringer C, et al. Hypoxia up-regulates hypoxia-inducible factor-1alpha transcription by involving phosphatidylinositol 3-kinase and nuclear factor kappaB in pulmonary artery smooth muscle cells. Mol Biol Cell 2007;18:4691–7
  • Kracun D, Riess F, Kanchev I, et al. The beta3-integrin binding protein beta3-endonexin is a novel negative regulator of hypoxia-inducible factor-1. Antioxidant Redox Signal 2014;20:1964–76
  • Diebold I, Petry A, Djordjevic T, et al. Reciprocal regulation of Rac1 and PAK-1 by HIF-1alpha: a positive-feedback loop promoting pulmonary vascular remodeling. Antioxidant Redox Signal 2010;13:399–412
  • Fatrai S, Wierenga AT, Daenen SM, et al. Identification of HIF2alpha as an important STAT5 target gene in human hematopoietic stem cells. Blood 2011;117:3320–30
  • Zhong H, Chiles K, Feldser D, et al. Modulation of hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res 2000;60:1541–5
  • Laughner E, Taghavi P, Chiles K, et al. HER2 (neu) signaling increases the rate of hypoxia-inducible factor 1alpha (HIF-1alpha) synthesis: novel mechanism for HIF-1-mediated vascular endothelial growth factor expression. Mol Cell Biol 2001;21:3995–4004
  • Djordjevic T, Pogrebniak A, BelAiba RS, et al. The expression of the NADPH oxidase subunit p22phox is regulated by a redox-sensitive pathway in endothelial cells. Free Radic Biol Med 2005;38:616–30
  • Wenner CE. Targeting mitochondria as a therapeutic target in cancer. J Cell Physiol 2012;227:450–6
  • Page EL, Robitaille GA, Pouyssegur J, Richard DE. Induction of hypoxia-inducible factor-1alpha by transcriptional and translational mechanisms. J Biol Chem 2002;277:48403–9
  • Zhou J, Callapina M, Goodall GJ, Brune B. Functional integrity of nuclear factor kappaB, phosphatidylinositol 3′-kinase, and mitogen-activated protein kinase signaling allows tumor necrosis factor alpha-evoked Bcl-2 expression to provoke internal ribosome entry site-dependent translation of hypoxia-inducible factor 1alpha. Cancer Res 2004;64:9041–8
  • Nayak BK, Feliers D, Sudarshan S, et al. Stabilization of HIF-2alpha through redox regulation of mTORC2 activation and initiation of mRNA translation. Oncogene 2012;32:3147–55
  • Doerries C, Grote K, Hilfiker-Kleiner D, et al. Critical role of the NAD(P)H oxidase subunit p47phox for left ventricular remodeling/dysfunction and survival after myocardial infarction. Circulation Res 2007;100:894–903
  • Murdoch CE, Grieve DJ, Cave AC, et al. NADPH oxidase and heart failure. Curr Opin Pharmacol 2006;6:148–53
  • Kleikers PW, Wingler K, Hermans JJ, et al. NADPH oxidases as a source of oxidative stress and molecular target in ischemia/reperfusion injury. J Mol Med 2012;90:1391–406
  • Nair D, Dayyat EA, Zhang SX, et al. Intermittent hypoxia-induced cognitive deficits are mediated by NADPH oxidase activity in a murine model of sleep apnea. PLoS One 2011;6:e19847
  • Malec V, Gottschald OR, Li S, et al. HIF-1 alpha signaling is augmented during intermittent hypoxia by induction of the Nrf2 pathway in NOX1-expressing adenocarcinoma A549 cells. Free Radic Biol Med 2010;48:1626–35
  • Hsieh CH, Wu CP, Lee HT, et al. NADPH oxidase subunit 4 mediates cycling hypoxia-promoted radiation resistance in glioblastoma multiforme. Free Radic Biol Med 2012;53:649–58
  • Urao N, McKinney RD, Fukai T, Ushio-Fukai M. NADPH oxidase 2 regulates bone marrow microenvironment following hindlimb ischemia: role in reparative mobilization of progenitor cells. Stem Cells 2012;30:923–34
  • Prabhakar NR, Semenza GL. Adaptive and maladaptive cardiorespiratory responses to continuous and intermittent hypoxia mediated by hypoxia-inducible factors 1 and 2. Physiol Rev 2012;92:967–1003
  • Djordjevic T, BelAiba RS, Bonello S, et al. Human urotensin II is a novel activator of NADPH oxidase in human pulmonary artery smooth muscle cells. Arterioscler Thromb Vasc Biol 2005;25:519–25
  • Liu JQ, Zelko IN, Erbynn EM, et al. Hypoxic pulmonary hypertension: role of superoxide and NADPH oxidase (gp91phox). Am J Physiol Lung Cell Mol Physiol 2006;290:L2–10
  • Weissmann N, Zeller S, Schafer RU, et al. Impact of mitochondria and NADPH oxidases on acute and sustained hypoxic pulmonary vasoconstriction. Am J Respir Cell Mol Biol 2006;34:505–13
  • Diebold I, Petry A, Hess J, Gorlach A. The NADPH oxidase subunit NOX4 is a new target gene of the hypoxia-inducible factor-1. Mol Biol Cell 2010;21:2087–96
  • Mittal M, Roth M, Konig P, et al. Hypoxia-dependent regulation of nonphagocytic NADPH oxidase subunit NOX4 in the pulmonary vasculature. Circulation Res 2007;101:258–67
  • Li J, Wang JJ, Yu Q, et al. Inhibition of reactive oxygen species by Lovastatin downregulates vascular endothelial growth factor expression and ameliorates blood-retinal barrier breakdown in db/db mice: role of NADPH oxidase 4. Diabetes 2010;59:1528–38
  • Kieninger J DA, Aravindalochanan K, Jobst G, et al. Amperometric oxygen sensor array with novel chronoamperometric protocols for hypoxic tumor cell cultures. TRANSDUCERS and EUROSENSORS ‘07 – 4th International Conference on Solid-State Sensors, Actuators and Microsystems 4300531; Lyon; pp. 1907–10
  • Ebbesen P, Pettersen EO, Gorr TA, et al. Taking advantage of tumor cell adaptations to hypoxia for developing new tumor markers and treatment strategies. J Enzyme Inhib Med Chem 2009;24:1–39
  • Kieninger J, Aravindalochanan K, Sandvik JA, et al. Pericellular oxygen monitoring with integrated sensor chips for reproducible cell culture experiments. Cell Prolif 2014;47:180–8
  • Jobst G, Moser I, Varahram M, et al. Thin-film microbiosensors for glucose-lactate monitoring. Analy Chem 1996;68:3173–9
  • Schneider BL, Kulesz-Martin M. Destructive cycles: the role of genomic instability and adaptation in carcinogenesis. Carcinogenesis 2004;25:2033–44
  • Chen EI, Hewel J, Krueger JS, et al. Adaptation of energy metabolism in breast cancer brain metastases. Cancer Res 2007;67:1472–86
  • Halliwell B. Oxidative stress and cancer: have we moved forward? Biochem J 2007;401:1–11
  • Swartz HM, Gutierrez PL. Free radical increases in cancer: evidence that there is not a real increase. Science (New York) 1977;198:936–8
  • Hafeman DG, Parce JW, McConnell HM. Light-addressable potentiometric sensor for biochemical systems. Science (New York) 1988;240:1182–5
  • Owicki JC, Parce JW. Biosensors based on the energy metabolism of living cells: the physical chemistry and cell biology of extracellular acidification. Biosens Bioelectron 1992;7:255–72
  • Hafner F. Cytosensor Microphysiometer: technology and recent applications. Biosens Bioelectron 2000;15:149–58
  • Eklund SE TD, Kozlov E, Prokop A, Cliffel DE. A microphysiometer for simultaneous measurement of changes in extracellular glucose, lactate, oxygen, and acidification rate. Anal Chem 2004;76:519–27
  • Eklund SE SR, Wikswo J, Baudenbacher F, Prokop A. Multianalyte microphysiometry as a tool in metabolomics and systems biology. J Electroanal Chem 2006;587:333–9
  • Ehret R, Baumann W, Brischwein M, et al. Monitoring of cellular behaviour by impedance measurements on interdigitated electrode structures. Biosens Bioelectron 1997;12:29–41
  • Lehmann M, Baumann W, Brischwein M, et al. Non-invasive measurement of cell membrane associated proton gradients by ion-sensitive field effect transistor arrays for microphysiological and bioelectronical applications. Biosens Bioelectron 2000;15:117–24
  • Lehmann M, Baumann W, Brischwein M, et al. Simultaneous measurement of cellular respiration and acidification with a single CMOS ISFET. Biosens Bioelectron 2001;16:195–203
  • Wolf B, Brischwein M, Baumann W, et al. Monitoring of cellular signalling and metabolism with modular sensor-technique: the PhysioControl-Microsystem (PCM). Biosens Bioelectron 1998;13:501–9
  • Brischwein M, Motrescu ER, Cabala E, et al. Functional cellular assays with multiparametric silicon sensor chips. Lab Chip 2003;3:234–40
  • Thedinga E, Kob A, Holst H, et al. Online monitoring of cell metabolism for studying pharmacodynamic effects. Toxicol Appl Pharmacol 2007;220:33–44
  • Mestres P, Morguet A. The Bionas technology for anticancer drug screening. Expert Opin Drug Discov 2009;4:785–97
  • Weltin A, Slotwinski K, Kieninger J, et al. Cell culture monitoring for drug screening and cancer research: a transparent, microfluidic, multi-sensor microsystem. Lab Chip 2014;14:138–46
  • Hogan C, Simons S, Zhang H, Burdick D. Living with irresolute cell lines in an automated world. J Assoc Lab Automat 2008;13:159–67
  • Kempner ME, Felder RA. A review of cell culture automation. J Assoc Lab Automat 2002;7:56–62
  • Triaud F, Clenet DH, Cariou Y, et al. Evaluation of automated cell culture incubators. J Assoc Lab Automat 2003;8:82–6
  • Thomas RJ, Chandra A, Hourd PC, Williams DJ. Cell culture automation and quality engineering: a necessary partnership to develop optimized manufacturing processes for cell-based therapies. JALA - J Assoc Lab Automat 2008;13:152–8
  • Franscini N, Wuertz K, Patocchi-Tenzer I, et al. Development of a novel automated cell isolation, expansion, and characterization platform. J Lab Automat 2011;16:204–13
  • Jain S, Sondervan D, Rizzu P, et al. The complete automation of cell culture: improvements for high-throughput and high-content screening. J Biomol Screen 2011;16:932–9
  • Ngibuini M. Reducing biomanufacturing bottlenecks. Genetic Eng Biotechnol News 2013;33:34–5
  • Maddox CB, Rasmussen L, White EL. Adapting cell-based assays to the high-throughput screening platform: problems encountered and lessons learned. J Assoc Lab Automat 2008;13:168–73
  • Su X, Theberge AB, January CT, Beebe DJ. Effect of microculture on cell metabolism and biochemistry: do cells get stressed in microchannels? Analy Chem 2013;85:1562–70
  • Kuwae S, Ohda T, Tamashima H, et al. Development of a fed-batch culture process for enhanced production of recombinant human antithrombin by Chinese hamster ovary cells. J Biosci Bioeng 2005;100:502–10
  • Yeo D, Kiparissides A, Cha JM, et al. Improving embryonic stem cell expansion through the combination of perfusion and bioprocess model design. PLoS ONE 2013;8
  • Adams T, Noack U, Frick T, et al. Increasing efficiency in protein supply and cell production by combining single-use bioreactor technology and perfusion: an off-the-shelf, single-use perfusion system enables high-density cell culture with high viability and high protein yield. BioPharm Int 2011;24:s4–11
  • Sciences GEHL. Application note 29-0051-80 AA and 28-9606-57 AC. Uppsala: GE Healthcare Life Sciences; 2012
  • Hu S, Deng L, Wang H, et al. Bioprocess development for the production of mouse-human chimeric anti-epidermal growth factor receptor vIII antibody C12 by suspension culture of recombinant Chinese hamster ovary cells. Cytotechnology 2011;63:247–58
  • Kleman GL, Chalmers JJ, Luli GW, Strohl WR. Glucose-stat, a glucose-controlled continuous culture. Appl Environ Microbiol 1991;57:918–23
  • Moser I, Jobst G, Urban GA. Biosensor arrays for simultaneous measurement of glucose, lactate, glutamate, and glutamine. Biosens Bioelectron 2002;17:297–302
  • Moser I, Jobst G. Pre-calibrated biosensors for single-use applications. Chemie-Ingenieur-Technik 2013;85:172–8
  • Poon E, Harris AL, Ashcroft M. Targeting the hypoxia-inducible factor (HIF) pathway in cancer. Expert Rev Mol Med 2009;11:e26. doi: 10.1017/S1462399409001173
  • Kioi M, Vogel H, Schultz G, et al. Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J Clin Invest 2010;120:694–705
  • Chau NM, Rogers P, Aherne W, et al. Identification of novel small molecule inhibitors of hypoxia-inducible factor-1 that differentially block hypoxia-inducible factor-1 activity and hypoxia-inducible factor-1alpha induction in response to hypoxic stress and growth factors. Cancer Res 2005;65:4918–28
  • Rapisarda A, Hollingshead M, Uranchimeg B, et al. Increased antitumor activity of bevacizumab in combination with hypoxia inducible factor-1 inhibition. Mol Cancer Ther 2009;8:1867–77
  • Rogers JL, Bayeh L, Scheuermann TH, et al. Development of inhibitors of the PAS-B domain of the HIF-2alpha transcription factor. J Med Chem 2013;56:1739–47
  • Kung AL, Zabludoff SD, France DS, et al. Small molecule blockade of transcriptional coactivation of the hypoxia-inducible factor pathway. Cancer Cell 2004;6:33–43
  • Miranda E, Nordgren IK, Male AL, et al. A cyclic peptide inhibitor of HIF-1 heterodimerization that inhibits hypoxia signaling in cancer cells. J Am Chem Soc 2013;135:10418–25
  • Baker LC, Boult JK, Walker-Samuel S, et al. The HIF-pathway inhibitor NSC-134754 induces metabolic changes and anti-tumour activity while maintaining vascular function. Br J Cancer 2012;106:1638–47
  • Carroll VA, Ashcroft M. Regulation of angiogenic factors by HDM2 in renal cell carcinoma. Cancer Res 2008;68:545–52
  • Hickin JA, Ahmed A, Fucke K, et al. The synthesis and structure revision of NSC-134754. Chem Commun (Camb) 2014;50:1238–40
  • Yang J, Ahmed A, Poon E, et al. Small-molecule activation of p53 blocks hypoxia-inducible factor 1alpha and vascular endothelial growth factor expression in vivo and leads to tumor cell apoptosis in normoxia and hypoxia. Mol Cell Biol 2009;29:2243–53
  • Ahmed A, Yang J, Maya-Mendoza A, et al. Pharmacological activation of a novel p53-dependent S-phase checkpoint involving CHK-1. Cell Death Dis 2011;2:e160. doi:10.1038/cddis.2011.42
  • Yang J, Ahmed A, Ashcroft M. Activation of a unique p53-dependent DNA damage response. Cell Cycle 2009;8:1630–2
  • Vousden KH, Lu X. Live or let die: the cell's response to p53. Nat Rev Cancer 2002;2:594–604
  • Bardos JI, Chau NM, Ashcroft M. Growth factor-mediated induction of HDM2 positively regulates hypoxia-inducible factor 1alpha expression. Mol Cell Biol 2004;24:2905–14
  • Supiot S, Hill RP, Bristow RG. Nutlin-3 radiosensitizes hypoxic prostate cancer cells independent of p53. Mol Cancer Ther 2008;7:993–9
  • Vassilev LT. MDM2 inhibitors for cancer therapy. Trends Mol Med 2007;13:23–31
  • Vassilev LT, Vu BT, Graves B, et al. In vivo activation of the p53 pathway by small-molecule antagonists of MDM2. Science 2004;303:844–8
  • Secchiero P, Corallini F, Gonelli A, et al. Antiangiogenic activity of the MDM2 antagonist nutlin-3. Circ Res 2007;100:61–9
  • LaRusch GA, Jackson MW, Dunbar JD, et al. Nutlin3 blocks vascular endothelial growth factor induction by preventing the interaction between hypoxia inducible factor 1alpha and Hdm2. Cancer Res 2007;67:450–4
  • Ahmed A, Yang J, Maya-Mendoza A, et al. Pharmacological activation of a novel p53-dependent S-phase checkpoint involving CHK-1. Cell Death Dis 2011;2:e160. doi:10.1038/cddis.2011.42
  • Selvarajah J, Nathawat K, Moumen A, et al. Chemotherapy-mediated p53-dependent DNA damage response in clear cell renal cell carcinoma: role of the mTORC1/2 and hypoxia-inducible factor pathways. Cell Death Dis 2013;4:e865. doi: 10.1038/cddis.2013.395
  • Kaelin WG Jr. Treatment of kidney cancer: insights provided by the VHL tumor-suppressor protein. Cancer 2009;115:2262–72
  • Shen C, Kaelin WG Jr. The VHL/HIF axis in clear cell renal carcinoma. Semin Cancer Biol 2013;23:18–25
  • Roe JS, Kim H, Lee SM, et al. p53 stabilization and transactivation by a von Hippel-Lindau protein. Mol Cell 2006;22:395–405
  • Maxwell PH, Wiesener MS, Chang GW, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 1999;399:271–5
  • Pelicano H, Martin DS, Xu RH, Huang P. Glycolysis inhibition for anticancer treatment. Oncogene 2006;25:4633–46
  • Maity A, Tuttle SW. 2-Deoxyglucose and radiosensitization: teaching an old DOG new tricks? Cancer Biol Ther 2006;5:824–6
  • Ellinghaus P, Heisler I, Unterschemmann K, et al. BAY 87-2243, a highly potent and selective inhibitor of hypoxia-induced gene activation has antitumor activities by inhibition of mitochondrial complex I. Cancer Med 2013;2:611–24
  • Yang J, Staples O, Thomas LW, et al. Human CHCHD4 mitochondrial proteins regulate cellular oxygen consumption rate and metabolism and provide a critical role in hypoxia signaling and tumor progression. J Clin Invest 2012;122:600–11
  • Gupta GP, Massague J. Cancer metastasis: building a framework. Cell 2006;127:679–95
  • Pastorekova S, Parkkila S, Parkkila AK, et al. Carbonic anhydrase IX, MN/CA IX: analysis of stomach complementary DNA sequence and expression in human and rat alimentary tracts. Gastroenterology 1997;112:398–408
  • Gieling RG, Parker CA, De Costa LA, et al. Inhibition of carbonic anhydrase activity modifies the toxicity of doxorubicin and melphalan in tumour cells in vitro. J Enzym Inhib Med Chem 2013;28:360–9
  • Potter CP, Harris AL. Diagnostic, prognostic and therapeutic implications of carbonic anhydrases in cancer. Br J Cancer 2003;89:2–7
  • Neri D, Supuran CT. Interfering with pH regulation in tumours as a therapeutic strategy. Nat Rev Drug Discov 2011;10:767–77
  • Svastova E, Hulikova A, Rafajova M, et al. Hypoxia activates the capacity of tumor-associated carbonic anhydrase IX to acidify extracellular pH. FEBS Lett 2004;577:439–45
  • Dubois L, Douma K, Supuran CT, et al. Imaging the hypoxia surrogate marker CA IX requires expression and catalytic activity for binding fluorescent sulfonamide inhibitors. Radiother Oncol: journal of the European Society for Therapeutic Radiology and Oncology 2007;83:367–73
  • Dubois L, Lieuwes NG, Maresca A, et al. Imaging of CA IX with fluorescent labelled sulfonamides distinguishes hypoxic and (re)-oxygenated cells in a xenograft tumour model. Radiother Oncol 2009;92:423–8
  • Ahlskog JK, Dumelin CE, Trussel S, et al. In vivo targeting of tumor-associated carbonic anhydrases using acetazolamide derivatives. Bioorg Med Chem Lett 2009;19:4851–6
  • Buller F, Steiner M, Frey K, et al. Selection of carbonic anhydrase IX inhibitors from one million DNA-encoded compounds. ACS Chem Biol 2011;6:336–44
  • Gieling RG, Babur M, Mamnani L, et al. Antimetastatic effect of sulfamate carbonic anhydrase IX inhibitors in breast carcinoma xenografts. J Med Chem 2012;55:5591–600
  • Rami M, Dubois L, Parvathaneni NK, et al. Hypoxia-targeting carbonic anhydrase IX inhibitors by a new series of nitroimidazole-sulfonamides/sulfamides/sulfamates. J Med Chem 2013;56:8512–20
  • Dubois L, Peeters SG, van Kuijk SJ, et al. Targeting carbonic anhydrase IX by nitroimidazole based sulfamides enhances the therapeutic effect of tumor irradiation: a new concept of dual targeting drugs. Radiother Oncol 2013;108:523–8
  • Overgaard J, Hansen HS, Overgaard M, et al. A randomized double-blind phase III study of nimorazole as a hypoxic radiosensitizer of primary radiotherapy in supraglottic larynx and pharynx carcinoma. Results of the Danish Head and Neck Cancer Study (DAHANCA) Protocol 5-85. Radiother Oncol 1998;46:135–46
  • Casey JR, Grinstein S, Orlowski J. Sensors and regulators of intracellular pH. Nat Rev Mol Cell Biol 2010;11:50–61
  • Parks SK, Chiche J, Pouyssegur J. Disrupting proton dynamics and energy metabolism for cancer therapy. Nat Rev Cancer 2013;13:611–23
  • Chiche J, Ilc K, Laferriere J, et al. Hypoxia-inducible carbonic anhydrase IX and XII promote tumor cell growth by counteracting acidosis through the regulation of the intracellular pH. Cancer Res 2009;69:358–68
  • Swietach P, Hulikova A, Vaughan-Jones RD, Harris AL. New insights into the physiological role of carbonic anhydrase IX in tumour pH regulation. Oncogene 2010;29:6509–21
  • Cummins EP, Selfridge AC, Sporn PH, et al. Carbon dioxide-sensing in organisms and its implications for human disease. Cell Mol Life Sci 2014;71:831–45
  • Doyen J, Parks SK, Marcie S, et al. Knock-down of hypoxia-induced carbonic anhydrases IX and XII radiosensitizes tumor cells by increasing intracellular acidosis. Frontier Oncol 2012;2:199. doi: 10.3389/fonc.2012.00199. eCollection 2012
  • Doherty JR, Yang C, Scott KE, et al. Blocking lactate export by inhibiting the Myc target MCT1 disables glycolysis and glutathione synthesis. Cancer Res 2014;74:908–20
  • Le Floch R, Chiche J, Marchiq I, et al. CD147 subunit of lactate/H + symporters MCT1 and hypoxia-inducible MCT4 is critical for energetics and growth of glycolytic tumors. Proc Natl Acad Sci USA 2011;108:16663–8
  • Murray CM, Hutchinson R, Bantick JR, et al. Monocarboxylate transporter MCT1 is a target for immunosuppression. Nat Chem Biol 2005;1:371–6
  • Polanski R, Hodgkinson CL, Fusi A, et al. Activity of the monocarboxylate transporter 1 inhibitor AZD3965 in small cell lung cancer. Clin Cancer Res 2014;20:926–37
  • Chiche J, Le Fur Y, Vilmen C, et al. In vivo pH in metabolic-defective Ras-transformed fibroblast tumors: key role of the monocarboxylate transporter, MCT4, for inducing an alkaline intracellular pH. Int J Cancer 2012;130:1511–20
  • Lutz NW, Le Fur Y, Chiche J, et al. Quantitative in vivo characterization of intracellular and extracellular pH profiles in heterogeneous tumors: a novel method enabling multiparametric pH analysis. Cancer Res 2013;73:4616–28
  • Halestrap AP. Monocarboxylic acid transport. Compreh Physiol 2013;3:1611–43
  • Gottfried E, Kunz-Schughart LA, Ebner S, et al. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood 2006;107:2013–21
  • Biswas S, Aggarwal M, Guzel O, et al. Conformational variability of different sulfonamide inhibitors with thienyl-acetamido moieties attributes to differential binding in the active site of cytosolic human carbonic anhydrase isoforms. Bioorg Med Chem 2011;19:3732–8
  • Scozzafava A, Supuran CT. Carbonic anhydrase inhibitors. Arylsulfonylureido- and arylureido-substituted aromatic and heterocyclic sulfonamides: towards selective inhibitors of carbonic anhydrase isozyme I. J Enzym Inhib 1999;14:343–63
  • Carta F, Garaj V, Maresca A, et al. Sulfonamides incorporating 1,3,5-triazine moieties selectively and potently inhibit carbonic anhydrase transmembrane isoforms IX, XII and XIV over cytosolic isoforms I and II: solution and X-ray crystallographic studies. Bioorg Med Chem 2011;19:3105–19
  • Garaj V, Puccetti L, Fasolis G, et al. Carbonic anhydrase inhibitors: synthesis and inhibition of cytosolic/tumor-associated carbonic anhydrase isozymes I, II, and IX with sulfonamides incorporating 1,2,4-triazine moieties. Bioorg Med Chem Lett 2004;14:5427–33
  • Garaj V, Puccetti L, Fasolis G, et al. Carbonic anhydrase inhibitors: novel sulfonamides incorporating 1,3,5-triazine moieties as inhibitors of the cytosolic and tumour-associated carbonic anhydrase isozymes I, II and IX. Bioorg Med Chem Lett 2005;15:3102–8
  • Pacchiano F, Carta F, McDonald PC, et al. Ureido-substituted benzenesulfonamides potently inhibit carbonic anhydrase IX and show antimetastatic activity in a model of breast cancer metastasis. J Med Chem 2011;54:1896–902
  • Pacchiano F, Aggarwal M, Avvaru BS, et al. Selective hydrophobic pocket binding observed within the carbonic anhydrase II active site accommodate different 4-substituted-ureido-benzenesulfonamides and correlate to inhibitor potency. Chem Commun (Camb) 2010;46:8371–3
  • McDonald PC, Winum JY, Supuran CT, Dedhar S. Recent developments in targeting carbonic anhydrase IX for cancer therapeutics. Oncotarget 2012;3:84–97
  • Carta F, Aggarwal M, Maresca A, et al. Dithiocarbamates: a new class of carbonic anhydrase inhibitors. Crystallographic and kinetic investigations. Chem Commun (Camb) 2012;48:1868–70
  • Carta F, Aggarwal M, Maresca A, et al. Dithiocarbamates strongly inhibit carbonic anhydrases and show antiglaucoma action in vivo. J Med Chem 2012;55:1721–30
  • Maresca A, Carta F, Vullo D, Supuran CT. Dithiocarbamates strongly inhibit the beta-class carbonic anhydrases from Mycobacterium tuberculosis. J Enzym Inhib Med Chem 2013;28:407–11
  • Monti SM, Maresca A, Viparelli F, et al. Dithiocarbamates are strong inhibitors of the beta-class fungal carbonic anhydrases from Cryptococcus neoformans, Candida albicans and Candida glabrata. Bioorg Med Chem Lett 2012;22:859–62
  • Kramer N, Walzl A, Unger C, et al. In vitro cell migration and invasion assays. Mutat Res 2013;752:10–24
  • Francia G, Cruz-Munoz W, Man S, et al. Mouse models of advanced spontaneous metastasis for experimental therapeutics. Nat Rev Cancer 2011;11:135–41
  • Cawthorne C, Burrows N, Gieling RG, et al. [18F]-FLT positron emission tomography can be used to image the response of sensitive tumors to PI3-kinase inhibition with the novel agent GDC-0941. Mol Cancer Ther 2013;12:819–28
  • Iorns E, Drews-Elger K, Ward TM, et al. A new mouse model for the study of human breast cancer metastasis. PLoS One 2012;7:e47995
  • Parkkila S, Rajaniemi H, Parkkila AK, et al. Carbonic anhydrase inhibitor suppresses invasion of renal cancer cells in vitro. Proc Natl Acad Sci USA 2000;97:2220–4
  • Gieling RG, Williams KJ. Carbonic anhydrase IX as a target for metastatic disease. Bioorg Med Chem 2013;21:1470–6
  • Winum JY, Carta F, Ward C, et al. Ureido-substituted sulfamates show potent carbonic anhydrase IX inhibitory and antiproliferative activities against breast cancer cell lines. Bioorg Med Chem Lett 2012;22:4681–5
  • Rademakers SE, Span PN, Kaanders JH, et al. Molecular aspects of tumour hypoxia. Mol Oncol 2008;2:41–53
  • Burrows N, Resch J, Cowen RL, et al. Expression of hypoxia-inducible factor 1 alpha in thyroid carcinomas. Endocrine-Relat Cancer 2010;17:61–72
  • Lee SL, Rouhi P, Dahl Jensen L, et al. Hypoxia-induced pathological angiogenesis mediates tumor cell dissemination, invasion, and metastasis in a zebrafish tumor model. Proc Natl Acad Sci USA 2009;106:19485–90
  • Rouhi P, Jensen LD, Cao Z, et al. Hypoxia-induced metastasis model in embryonic zebrafish. Nat Protocol 2010;5:1911–18
  • Cao R, Ji H, Feng N, et al. Collaborative interplay between FGF-2 and VEGF-C promotes lymphangiogenesis and metastasis. Proc Natl Acad Sci USA 2012;109:15894–9
  • Cao R, Bjorndahl MA, Religa P, et al. PDGF-BB induces intratumoral lymphangiogenesis and promotes lymphatic metastasis. Cancer Cell 2004;6:333–45
  • Cao R, Lim S, Ji H, et al. Mouse corneal lymphangiogenesis model. Nat Protocol 2011;6:817–26
  • Zamboni WC, Torchilin V, Patri AK, et al. Best practices in cancer nanotechnology: perspective from NCI nanotechnology alliance. Clin Cancer Res 2012;18:3229–41
  • Petsko GA. When failure should be the option. BMC Biol 2010;8:61
  • Meehan J WC, Supuran C, Langdon SP, Kunkler IH. The effects of novel carbonic anhydrase inhibitors on the proliferation and invasion of breast and ovarian cancer cells. J Pathol 2013;231:36–45
  • Huang M, Shen A, Ding J, Geng M. Molecularly targeted cancer therapy: some lessons from the past decade. Trends Pharmacol Sci 2014;35:41–50
  • Daniel VC, Marchionni L, Hierman JS, et al. A primary xenograft model of small-cell lung cancer reveals irreversible changes in gene expression imposed by culture in vitro. Cancer Res 2009;69:3364–73
  • Langdon SP. Isolation and culture of ovarian cancer cell lines. Method Mol Med 2004;88:133–9
  • Navin N, Kendall J, Troge J, et al. Tumour evolution inferred by single-cell sequencing. Nature 2011;472:90–4
  • Tannock IF, Rotin D. Acid pH in tumors and its potential for therapeutic exploitation. Cancer Res 1989;49:4373–84
  • Harrison DK, Vaupel P. Heterogeneity in tissue oxygenation: from physiological variability in normal tissues to pathophysiological chaos in malignant tumours. Adv Exp Med Biol 2014;812:25–31
  • Debnath J, Brugge JS. Modelling glandular epithelial cancers in three-dimensional cultures. Nat Rev Cancer 2005;5:675–88
  • Weigelt B, Bissell MJ. Unraveling the microenvironmental influences on the normal mammary gland and breast cancer. Semin Cancer Biol 2008;18:311–21
  • Gillet JP, Calcagno AM, Varma S, et al. Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistance. Proc Natl Acad Sci USA 2011;108:18708–13
  • Mehta JP, O'Driscoll L, Barron N, et al. A microarray approach to translational medicine in breast cancer: how representative are cell line models of clinical conditions? Anticancer Res 2007;27:1295–300
  • dit Faute MA, Laurent L, Ploton D, et al. Distinctive alterations of invasiveness, drug resistance and cell-cell organization in 3D-cultures of MCF-7, a human breast cancer cell line, and its multidrug resistant variant. Clin Exp Metastasis 2002;19:161–8
  • Yamada KM, Cukierman E. Modeling tissue morphogenesis and cancer in 3D. Cell 2007;130:601–10
  • Pampaloni F, Reynaud EG, Stelzer EH. The third dimension bridges the gap between cell culture and live tissue. Nat Rev Mol Biol 2007;8:839–45
  • Schmeichel KL, Bissell MJ. Modeling tissue-specific signaling and organ function in three dimensions. J Cell Sci 2003;116:2377–88
  • Kumar HR, Zhong X, Hoelz DJ, et al. Three-dimensional neuroblastoma cell culture: proteomic analysis between monolayer and multicellular tumor spheroids. Pediatr Surg Int 2008;24:1229–34
  • Griffith LG, Swartz MA. Capturing complex 3D tissue physiology in vitro. Nat Rev Mol Cell Biol 2006;7:211–24
  • Hirschhaeuser F, Menne H, Dittfeld C, et al. Multicellular tumor spheroids: an underestimated tool is catching up again. J Biotechnol 2010;148:3–15
  • Lin RZ, Chang HY. Recent advances in three-dimensional multicellular spheroid culture for biomedical research. Biotechnol J 2008;3:1172–84
  • Provenzano PP, Inman DR, Eliceiri KW, et al. Collagen density promotes mammary tumor initiation and progression. BMC Med 2008;6:11
  • Ramaswamy S, Ross KN, Lander ES, Golub TR. A molecular signature of metastasis in primary solid tumors. Nat Genet 2003;33:49–54
  • Friedrich J, Ebner R, Kunz-Schughart LA. Experimental anti-tumor therapy in 3-D: spheroids--old hat or new challenge? Int J Radiat Biol 2007;83:849–71
  • Minchinton AI, Tannock IF. Drug penetration in solid tumours. Nat Rev Cancer 2006;6:583–92
  • Swietach P, Patiar S, Supuran CT, et al. The role of carbonic anhydrase 9 in regulating extracellular and intracellular ph in three-dimensional tumor cell growths. J Biol Chem 2009;284:20299–310
  • McIntyre A, Patiar S, Wigfield S, et al. Carbonic anhydrase IX promotes tumor growth and necrosis in vivo and inhibition enhances anti-VEGF therapy. Clin Cancer Res 2012;18:3100–11
  • Kamb A. What's wrong with our cancer models? Nat Rev Drug Discov 2005;4:161–5
  • Harrington KJ, Billingham LJ, Brunner TB, et al. Guidelines for preclinical and early phase clinical assessment of novel radiosensitisers. Br J Cancer 2011;105:628–39
  • Vargo-Gogola T, Rosen JM. Modelling breast cancer: one size does not fit all. Nat Rev Cancer 2007;7:659–72
  • Dean JL, McClendon AK, Hickey TE, et al. Therapeutic response to CDK4/6 inhibition in breast cancer defined by ex vivo analyses of human tumors. Cell Cycle (Georgetown, Tex) 2012;11:2756–61
  • Sausville EA, Burger AM. Contributions of human tumor xenografts to anticancer drug development. Cancer Res 2006;66:3351–4, discussion 4
  • Boedigheimer MJ, Freeman DJ, Kiaei P, et al. Gene expression profiles can predict panitumumab monotherapy responsiveness in human tumor xenograft models. Neoplasia (New York) 2013;15:125–32
  • Politi K, Pao W. How genetically engineered mouse tumor models provide insights into human cancers. J Clin Oncol 2011;29:2273–81
  • Sharpless NE, Depinho RA. The mighty mouse: genetically engineered mouse models in cancer drug development. Nat Rev Drug Discov 2006;5:741–54
  • Olive KP, Jacobetz MA, Davidson CJ, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science (New York) 2009;324:1457–61
  • Graves EE, Vilalta M, Cecic IK, et al. Hypoxia in models of lung cancer: implications for targeted therapeutics. Clin Cancer Res 2010;16:4843–52
  • Maity A, Koumenis C. Location, location, location-makes all the difference for hypoxia in lung tumors. Clin Cancer Res 2010;16:4685–7
  • Singh M, Lima A, Molina R, et al. Assessing therapeutic responses in Kras mutant cancers using genetically engineered mouse models. Nat Biotech 2010;28:585–93
  • Chen Z, Cheng K, Walton Z, et al. A murine lung cancer co-clinical trial identifies genetic modifiers of therapeutic response. Nature 2012;483:613–17
  • Moding EJ, Kastan MB, Kirsch DG. Strategies for optimizing the response of cancer and normal tissues to radiation. Nat Rev Drug Discov 2013;12:526–42
  • Fu L, Wang G, Shevchuk MM, et al. Generation of a mouse model of Von Hippel-Lindau kidney disease leading to renal cancers by expression of a constitutively active mutant of HIF1alpha. Cancer Res 2011;71:6848–56
  • Morgan MA, Parsels LA, Zhao L, et al. Mechanism of radiosensitization by the Chk1/2 inhibitor AZD7762 involves abrogation of the G2 checkpoint and inhibition of homologous recombinational DNA repair. Cancer Res 2010;70:4972–81
  • DeRose YS, Wang G, Lin YC, et al. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat Med 2011;17:1514–20
  • Whiteford CC, Bilke S, Greer BT, et al. Credentialing preclinical pediatric xenograft models using gene expression and tissue microarray analysis. Cancer Res 2007;67:32–40
  • Zhao X, Liu Z, Yu L, et al. Global gene expression profiling confirms the molecular fidelity of primary tumor-based orthotopic xenograft mouse models of medulloblastoma. Neuro-Oncology 2012;14:574–83
  • Reyal F, Guyader C, Decraene C, et al. Molecular profiling of patient-derived breast cancer xenografts. Breast Cancer Res 2012;14:R11
  • Ding L, Ellis MJ, Li S, et al. Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature 2010;464:999–1005
  • Siolas D, Hannon GJ. Patient-derived tumor xenografts: transforming clinical samples into mouse models. Cancer Res 2013;73:5315–19
  • Katz E, Sims AH, Sproul D, et al. Targeting of Rac GTPases blocks the spread of intact human breast cancer. Oncotarget 2012;3:608–19
  • Ward C MJ, Harrison DJ, Kunkler IH, Langdon SP. An ex-vivo tumour model using core biopsy explants validate carbonic anhydrase IX (CAIX) as a therapeutic target in breast cancer. J Pathol 2013;231:24–30
  • van der Kuip H, Murdter TE, Sonnenberg M, et al. Short term culture of breast cancer tissues to study the activity of the anticancer drug taxol in an intact tumor environment. BMC Cancer 2006;6:86
  • Steeg PS. Tumor metastasis: mechanistic insights and clinical challenges. Nat Med 2006;12:895–904
  • MacDonald IC, Groom AC, Chambers AF. Cancer spread and micrometastasis development: quantitative approaches for in vivo models. BioEssays 2002;24:885–93
  • Tentler JJ, Tan AC, Weekes CD, et al. Patient-derived tumour xenografts as models for oncology drug development. Nat Rev Clin Oncol 2012;9:338–50
  • Singh M, Ferrara N. Modeling and predicting clinical efficacy for drugs targeting the tumor milieu. Nat Biotechnol 2012;30:648–57
  • Nardella C, Lunardi A, Patnaik A, et al. The APL paradigm and the “co-clinical trial” project. Cancer Discov 2011;1:108–16
  • Lambin P, Petit SF, Aerts HJ, et al. The ESTRO Breur Lecture 2009. From population to voxel-based radiotherapy: exploiting intra-tumour and intra-organ heterogeneity for advanced treatment of non-small cell lung cancer. Radiother Oncol 2010;96:145–52
  • Thorwarth D, Eschmann SM, Paulsen F, Alber M. Hypoxia dose painting by numbers: a planning study. Int J Radiat Oncol Biol Phys 2007;68:291–300
  • Bollineni VR, Wiegman EM, Pruim J, et al. Hypoxia imaging using positron emission tomography in non-small cell lung cancer: implications for radiotherapy. Cancer Treat Rev 2012;38:1027–32
  • Mortensen LS, Johansen J, Kallehauge J, et al. FAZA PET/CT hypoxia imaging in patients with squamous cell carcinoma of the head and neck treated with radiotherapy: results from the DAHANCA 24 trial. Radiother Oncol 2012;105:14–20
  • Busk M, Jakobsen S, Horsman MR, et al. PET imaging of tumor hypoxia using 18F-labeled pimonidazole. Acta Oncol (Stockholm, Sweden) 2013;52:1300–7
  • Dubois LJ, Lieuwes NG, Janssen MH, et al. Preclinical evaluation and validation of [18F]HX4, a promising hypoxia marker for PET imaging. Proc Natl Acad Sci USA 2011;108:14620–5
  • van Loon J, Janssen MH, Ollers M, et al. PET imaging of hypoxia using [18F]HX4: a phase I trial. Eur J Nucl Med Mol Imag 2010;37:1663–8
  • Doss M, Zhang JJ, Belanger MJ, et al. Biodistribution and radiation dosimetry of the hypoxia marker 18F-HX4 in monkeys and humans determined by using whole-body PET/CT. Nucl Med Commun 2010;31:1016–24
  • Zegers CM, van Elmpt W, Wierts R, et al. Hypoxia imaging with [(1)(8)F]HX4 PET in NSCLC patients: defining optimal imaging parameters. Radiother Oncol 2013;109:58–64
  • Horsman MR, Mortensen LS, Petersen JB, Busk M, Overgaard J. Imaging hypoxia to improve radiotherapy outcome. Nat Rev Clin Oncol 2012;9:674–87
  • Betts GN, Eustace A, Patiar S, et al. Prospective technical validation and assessment of intra-tumour heterogeneity of a low density array hypoxia gene profile in head and neck squamous cell carcinoma. Eur J Cancer 2013;49:156–65
  • Winter SC, Buffa FM, Silva P, et al. Relation of a hypoxia metagene derived from head and neck cancer to prognosis of multiple cancers. Cancer Res 2007;67:3441–9
  • Buffa FM, Harris AL, West CM, Miller CJ. Large meta-analysis of multiple cancers reveals a common, compact and highly prognostic hypoxia metagene. Br J Cancer 2010;102:428–35
  • Curtis C, Shah SP, Chin SF, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 2012;486:346–52
  • TCGA. Comprehensive molecular portraits of human breast tumours. Nature 2012;490:61–70
  • Jubb AM, Buffa FM, Harris AL. Assessment of tumour hypoxia for prediction of response to therapy and cancer prognosis. J Cell Mol Med 2010;14:18–29
  • Eustace A, Mani N, Span PN, et al. A 26-gene hypoxia signature predicts benefit from hypoxia-modifying therapy in laryngeal cancer but not bladder cancer. Clin Cancer Res 2013;19:4879–88
  • Ghazoui Z, Buffa FM, Dunbier AK, et al. Close and stable relationship between proliferation and a hypoxia metagene in aromatase inhibitor-treated ER-positive breast cancer. Clin Cancer Res 2011;17:3005–12
  • Camps C, Buffa FM, Colella S, et al. hsa-miR-210 Is induced by hypoxia and is an independent prognostic factor in breast cancer. Clin Cancer Res 2008;14:1340–8
  • McCormick RI, Blick C, Ragoussis J, et al. miR-210 is a target of hypoxia-inducible factors 1 and 2 in renal cancer, regulates ISCU and correlates with good prognosis. Br J Cancer 2013;108:1133–42
  • Chan SY, Zhang YY, Hemann C, et al. MicroRNA-210 controls mitochondrial metabolism during hypoxia by repressing the iron-sulfur cluster assembly proteins ISCU1/2. Cell Metabol 2009;10:273–84
  • Favaro E, Ramachandran A, McCormick R, et al. MicroRNA-210 regulates mitochondrial free radical response to hypoxia and krebs cycle in cancer cells by targeting iron sulfur cluster protein ISCU. PLoS One 2010;5:e10345
  • Buffa FM, Camps C, Winchester L, et al. microRNA-associated progression pathways and potential therapeutic targets identified by integrated mRNA and microRNA expression profiling in breast cancer. Cancer Res 2011;71:5635–45
  • Rothe F, Ignatiadis M, Chaboteaux C, et al. Global microRNA expression profiling identifies MiR-210 associated with tumor proliferation, invasion and poor clinical outcome in breast cancer. PLoS One 2011;6:e20980
  • Halle C, Andersen E, Lando M, et al. Hypoxia-induced gene expression in chemoradioresistant cervical cancer revealed by dynamic contrast-enhanced MRI. Cancer Res 2012;72:5285–95
  • Milosevic M, Warde P, Menard C, et al. Tumor hypoxia predicts biochemical failure following radiotherapy for clinically localized prostate cancer. Clin Cancer Res 2012;18:2108–14
  • Ragnum HB, Roe K, Holm R, et al. Hypoxia-independent downregulation of hypoxia-inducible factor 1 targets by androgen deprivation therapy in prostate cancer. Int J Radiat Oncol Biol Phys 2013;87:753–60
  • Mabjeesh NJ, Willard MT, Frederickson CE, et al. Androgens stimulate hypoxia-inducible factor 1 activation via autocrine loop of tyrosine kinase receptor/phosphatidylinositol 3′-kinase/protein kinase B in prostate cancer cells. Clin Cancer Res 2003;9:2416–25
  • Horii K, Suzuki Y, Kondo Y, et al. Androgen-dependent gene expression of prostate-specific antigen is enhanced synergistically by hypoxia in human prostate cancer cells. Mol Cancer Res 2007;5:383–91
  • Overgaard J, Eriksen JG, Nordsmark M, Group DHaNC. Plasma osteopontin, hypoxia, and response to the hypoxia sensitiser nimorazole in radiotherapy of head and neck cancer: results from the DAHANCA 5 randomised double-blind placebo-controlled trial. Lancet Oncol 2005;6:757–64
  • Rischin D, Hicks RJ, Fisher R, et al. Prognostic significance of [18F]-misonidazole positron emission tomography-detected tumor hypoxia in patients with advanced head and neck cancer randomly assigned to chemoradiation with or without tirapazamine: a substudy of Trans-Tasman Radiation Oncology Group Study 98.02. J Clin Oncol 2006;24:2098–104
  • Toustrup K, Sorensen BS, Nordsmark M, et al. Development of a hypoxia gene expression classifier with predictive impact for hypoxic modification of radiotherapy in head and neck cancer. Cancer Res 2011;71:5923–31
  • Toustrup K, Sorensen BS, Lassen P, et al. Gene expression classifier predicts for hypoxic modification of radiotherapy with nimorazole in squamous cell carcinomas of the head and neck. Radiother Oncol 2012;102:122–9
  • Sorensen BS, Busk M, Olthof N, et al. Radiosensitivity and effect of hypoxia in HPV positive head and neck cancer cells. Radiother Oncol 2013;108:500–5
  • Winther M, Alsner J, Tramm T, Nordsmark M. Hypoxia-regulated gene expression and prognosis in loco-regional gastroesophageal cancer. Acta Oncol (Stockholm, Sweden) 2013;52:1327–35
  • Bertout JA, Patel SA, Simon MC. The impact of O2 availability on human cancer. Nat Rev Cancer 2008;8:967–75
  • Casazza A, Di Conza G, Wenes M, et al. Tumor stroma: a complexity dictated by the hypoxic tumor microenvironment. Oncogene 2014;33:1743–54
  • Martini M, Vecchione L, Siena S, et al. Targeted therapies: how personal should we go? Nat Rev Clin Oncol 2012;9:87–97
  • Folkvord S, Flatmark K, Dueland S, et al. Prediction of response to preoperative chemoradiotherapy in rectal cancer by multiplex kinase activity profiling. Int J Radiat Oncol Biol Phys 2010;78:555–62
  • Saelen MG, Flatmark K, Folkvord S, et al. Tumor kinase activity in locally advanced rectal cancer: angiogenic signaling and early systemic dissemination. Angiogenesis 2011;14:481–9
  • Ree AH, Kristensen AT, Saelen MG, et al. Tumor phosphatidylinositol-3-kinase signaling and development of metastatic disease in locally advanced rectal cancer. PLoS One 2012;7:e50806
  • Roe K, Bratland A, Vlatkovic L, et al. Hypoxic tumor kinase signaling mediated by STAT5A in development of castration-resistant prostate cancer. PLoS One 2013;8:e63723
  • Tahiri A, Roe K, Ree AH, et al. Differential inhibition of ex-vivo tumor kinase activity by vemurafenib in BRAF(V600E) and BRAF wild-type metastatic malignant melanoma. PLoS One 2013;8:e72692
  • Ramachandran A, Betts G, Bhana S, et al. An in vivo hypoxia metagene identifies the novel hypoxia inducible factor target gene SLCO1B3. Eur J Cancer 2013;49:1741–51
  • van Baardwijk A, Tome WA, van Elmpt W, et al. Is high-dose stereotactic body radiotherapy (SBRT) for stage I non-small cell lung cancer (NSCLC) overkill? A systematic review. Radiother Oncol 2012;105:145–9
  • Ashworth A, Rodrigues G, Boldt G, Palma D. Is there an oligometastatic state in non-small cell lung cancer? A systematic review of the literature. Lung Cancer (Amsterdam, Netherlands) 2013;82:197–203
  • Cheruvu P, Metcalfe SK, Metcalfe J, et al. Comparison of outcomes in patients with stage III versus limited stage IV non-small cell lung cancer. Radiat Oncol (London, England) 2011;6:80. doi:10.1186/1748-717X-6-80
  • Dahele M, Senan S. The role of stereotactic ablative radiotherapy for early-stage and oligometastatic non-small cell lung cancer: evidence for changing paradigms. Cancer Res Treat 2011;43:75–82
  • Hasselle MD, Haraf DJ, Rusthoven KE, et al. Hypofractionated image-guided radiation therapy for patients with limited volume metastatic non-small cell lung cancer. J Thorac Oncol 2012;7:376–81
  • Higginson DS, Chen RC, Tracton G, et al. The impact of local and regional disease extent on overall survival in patients with advanced stage IIIB/IV non-small cell lung carcinoma. Int J Radiat Oncol Biol Phys 2012;84:e385–92
  • Marks LB, Saynak M, Christodouleas JP. Stage III vs. stage IV lung cancer: “crossing a great divide”. Lung Cancer (Amsterdam, Netherlands) 2010;67:1–3
  • Milano MT, Katz AW, Zhang H, Okunieff P. Oligometastases treated with stereotactic body radiotherapy: long-term follow-up of prospective study. Int J Radiat Oncol Biol Phys 2012;83:878–86
  • Tree AC, Khoo VS, Eeles RA, et al. Stereotactic body radiotherapy for oligometastases. Lancet Oncol 2013;14:e28–37
  • De Ruysscher D, Wanders R, van Baardwijk A, et al. Radical treatment of non-small-cell lung cancer patients with synchronous oligometastases: long-term results of a prospective phase II trial (Nct01282450). J Thorac Oncol 2012;7:1547–55
  • De Bock K, Mazzone M, Carmeliet P. Antiangiogenic therapy, hypoxia, and metastasis: risky liaisons, or not? Nat Rev Clin Oncol 2011;8:393–404
  • Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med 2011;17:1359–70
  • Cooke VG, LeBleu VS, Keskin D, et al. Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell 2012;21:66–81
  • Petersen SH, Harling H, Kirkeby LT, et al. Postoperative adjuvant chemotherapy in rectal cancer operated for cure. Cochrane Database Syst Rev 2012;3:CD004078
  • Bosset JF, Calais G, Mineur L, et al. Fluorouracil-based adjuvant chemotherapy after preoperative chemoradiotherapy in rectal cancer: long-term results of the EORTC 22921 randomised study. Lancet Oncol 2014;15:184–90
  • Fass L. Imaging and cancer: a review. Mol Oncol 2008;2:115–52
  • Orloff J, Douglas F, Pinheiro J, et al. The future of drug development: advancing clinical trial design. Nat Rev Drug Discov 2009;8:949–57
  • Vogelzang NJ, Benowitz SI, Adams S, et al. Clinical cancer advances 2011: Annual report on progress against cancer from the American society of clinical oncology. J Clin Oncol 2012;30:88–109
  • Lambin P, Roelofs E, Reymen B, et al. ‘Rapid Learning health care in oncology’ – an approach towards decision support systems enabling customised radiotherapy'. Radiother Oncol 2013;109:159–64
  • Maitland ML, Schilsky RL. Clinical trials in the era of personalized oncology. Cancer J Clin 2011;61:365–81
  • Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. New Engl J Med 2012;366:883–92
  • Bachtiary B, Boutros PC, Pintilie M, et al. Gene expression profiling in cervical cancer: an exploration of intratumor heterogeneity. Clin Cancer Res 2006;12:5632–40
  • Boyd CA, Benarroch-Gampel J, Sheffield KM, et al. 415 patients with adenosquamous carcinoma of the pancreas: a population-based analysis of prognosis and survival. J Surg Res 2012;174:12–9
  • Milosevic MF, Fyles AW, Wong R, et al. Interstitial fluid pressure in cervical carcinoma: within tumor heterogeneity, and relation to oxygen tension. Cancer 1998;82:2418–26
  • Lambin P, van Stiphout RG, Starmans MH, et al. Predicting outcomes in radiation oncology – multifactorial decision support systems. Nat Rev Clin Oncol 2013;10:27–40
  • (a) Supuran CT. Inhibition of carbonic anhydrase IX as a novel anticancer mechanism. World J Clin Oncol 2012;3:98–103. (b) Supuran CT. Structure-based drug discovery of carbonic anhydrase inhibitors. J Enzyme Inhib Med Chem 2012;27;759–72
  • Supuran CT. Carbonic anhydrases: from biomedical applications of the inhibitors and activators to biotechnological use for CO(2) capture. J Enzyme Inhib Med Chem 2013;28;229–32

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.