922
Views
24
CrossRef citations to date
0
Altmetric
Review

The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy

, , &
Pages 83-92 | Published online: 11 Dec 2015

References

  • Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87–108.
  • Thomlinson RH, Gray LH. The histological structure of some human lung cancers and the possible implications for radiotherapy. Br J Cancer. 1955;9(4):539–549.
  • Semenza GL. Hypoxia, clonal selection, and the role of HIF-1 in tumor progression. Crit Rev Biochem Mol Biol. 2000;35(2):71–103.
  • Hockel M, Schlenger K, Knoop C, Vaupel P. Oxygenation of carcinomas of the uterine cervix: evaluation by computerized O2 tension measurements. Cancer Res. 1991;51(22):6098–6102.
  • Carreau A, El Hafny-Rahbi B, Matejuk A, Grillon C, Kieda C. Why is the partial oxygen pressure of human tissues a crucial parameter? Small molecules and hypoxia. J Cell Mol Med. 2011;15(6):1239–1253.
  • Vaupel P, Hockel M, Mayer A. Detection and characterization of tumor hypoxia using pO2 histography. Antiox Redox Signal. 2007; 9(8):1221–1235.
  • Muller M, Padberg W, Schindler E, et al. Renocortical tissue oxygen pressure measurements in patients undergoing living donor kidney transplantation. Anesth Analg. 1998;87(2):474–476.
  • Dings J, Meixensberger J, Jager A, Roosen K. Clinical experience with 118 brain tissue oxygen partial pressure catheter probes. Neurosurgery. 1998;43(5):1082–1095.
  • Hoffman WE, Charbel FT, Edelman G. Brain tissue oxygen, carbon dioxide, and pH in neurosurgical patients at risk for ischemia. Anesth Analg. 1996;82(3):582–586.
  • Cervos-Navarro J, Diemer NH. Selective vulnerability in brain hypoxia. Crit Rev Neurobiol. 1991;6(3):149–182.
  • Wenger RH, Kurtcuoglu V, Scholz CC, Marti HH, Hoogewijs D. Frequently asked questions in hypoxia research. Hypoxia. 2015(3):35–43.
  • Bayer C, Vaupel P. Acute versus chronic hypoxia in tumors: Controversial data concerning time frames and biological consequences. Strahlenther Onkol. 2012;188(7):616–627.
  • Vaupel P, Harrison L. Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist. 2004;9 Suppl 5:4–9.
  • Chaplin DJ, Olive PL, Durand RE. Intermittent blood flow in a murine tumor: radiobiological effects. Cancer Res. 1987;47(2):597–601.
  • Mazure NM, Pouyssegur J. Hypoxia-induced autophagy: cell death or cell survival? Curr Opin Cell Biol. 2010;22(2):177–180.
  • Rouschop KM, Ramaekers CH, Schaaf MB, et al. Autophagy is required during cycling hypoxia to lower production of reactive oxygen species. Radiother Oncol. 2009;92(3):411–416.
  • Hsieh CH, Lee CH, Liang JA, Yu CY, Shyu WC. Cycling hypoxia increases U87 glioma cell radioresistance via ROS induced higher and long-term HIF-1 signal transduction activity. Oncol Rep. 2010; 24(6):1629–1636.
  • Hsieh CH, Shyu WC, Chiang CY, Kuo JW, Shen WC, Liu RS. NADPH oxidase subunit 4-mediated reactive oxygen species contribute to cycling hypoxia-promoted tumor progression in glioblastoma multiforme. PloS One. 2011;6(9):e23945.
  • Kato Y, Yashiro M, Fuyuhiro Y, et al. Effects of acute and chronic hypoxia on the radiosensitivity of gastric and esophageal cancer cells. Anticancer Res. 2011;31(10):3369–3375.
  • Chaplin DJ, Durand RE, Olive PL. Acute hypoxia in tumors: implications for modifiers of radiation effects. Int J Radiat Oncol Biol Phys. 1986;12(8):1279–1282.
  • Bellot G, Garcia-Medina R, Gounon P, et al. Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol Cell Biol. 2009; 29(10):2570–2581.
  • Rofstad EK, Gaustad JV, Egeland TA, Mathiesen B, Galappathi K. Tumors exposed to acute cyclic hypoxic stress show enhanced angiogenesis, perfusion and metastatic dissemination. Int J Cancer. 2010;127(7):1535–1546.
  • Chan N, Koritzinsky M, Zhao H, et al. Chronic hypoxia decreases synthesis of homologous recombination proteins to offset chemoresistance and radioresistance. Cancer Res. 2008;68(2):605–614.
  • Luoto KR, Kumareswaran R, Bristow RG. Tumor hypoxia as a driving force in genetic instability. Genome Integr. 2013;4(1):5.
  • Kondo A, Safaei R, Mishima M, Niedner H, Lin X, Howell SB. Hypoxia-induced enrichment and mutagenesis of cells that have lost DNA mismatch repair. Cancer Res. 2001;61(20):7603–7607.
  • Pires IM, Bencokova Z, Milani M, et al. Effects of acute versus chronic hypoxia on DNA damage responses and genomic instability. Cancer Res. 2010;70(3):925–935.
  • Brurberg KG, Graff BA, Olsen DR, Rofstad EK. Tumor-line specific pO(2) fluctuations in human melanoma xenografts. Int J Radiat Oncol Biol Phys. 2004;58(2):403–409.
  • Kimura H, Braun RD, Ong ET, et al. Fluctuations in red cell flux in tumor microvessels can lead to transient hypoxia and reoxygenation in tumor parenchyma. Cancer Res. 1996;56(23):5522–5528.
  • Agani F, Jiang BH. Oxygen-independent regulation of HIF-1: novel involvement of PI3K/AKT/mTOR pathway in cancer. Curr Cancer Drug Targets. 2013;13(3):245–251.
  • Courtnay R, Ngo DC, Malik N, Ververis K, Tortorella SM, Karagiannis TC. Cancer metabolism and the Warburg effect: the role of HIF-1 and PI3K. Mol Biol Rep. 2015;42(4):841–851.
  • Seta KA, Spicer Z, Yuan Y, Lu G, Millhorn DE. Responding to hypoxia: lessons from a model cell line. Sci STKE. 2002;2002(146):re11.
  • Sanchez A, Tripathy D, Yin X, et al. p38 MAPK: a mediator of hypoxia-induced cerebrovascular inflammation. J Alzheimers Dis. 2012;32(3):587–597.
  • Minet E, Arnould T, Michel G, et al. ERK activation upon hypoxia: involvement in HIF-1 activation. FEBS Lett. 2000;468(1):53–58.
  • Koong AC, Chen EY, Giaccia AJ. Hypoxia causes the activation of nuclear factor kappa B through the phosphorylation of I kappa B alpha on tyrosine residues. Cancer Res. 1994;54(6):1425–1430.
  • Salmena L, Carracedo A, Pandolfi PP. Tenets of PTEN tumor suppression. Cell. 2008;133(3):403–414.
  • Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol. 1992; 12(12):5447–5454.
  • Bruick RK, McKnight SL. A conserved family of prolyl-4-hydroxylases that modify HIF. Science. 2001;294(5545):1337–1340.
  • Maxwell PH, Wiesener MS, Chang GW, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399(6733):271–275.
  • van Uden P, Kenneth NS, Rocha S. Regulation of hypoxia-inducible factor-1alpha by NF-kappaB. Biochem J. 2008;412(3):477–484.
  • Kilic-Eren M, Boylu T, Tabor V. Targeting PI3K/Akt represses Hypoxia inducible factor-1alpha activation and sensitizes Rhabdomyosarcoma and Ewing’s sarcoma cells for apoptosis. Cancer Cell Int. 2013;13:36.
  • Richard DE, Berra E, Gothie E, Roux D, Pouyssegur J. p42/p44 mitogen-activated protein kinases phosphorylate hypoxia-inducible factor 1alpha (HIF-1alpha) and enhance the transcriptional activity of HIF-1. J Biol Chem. 1999;274(46):32631–32637.
  • Royds JA, Dower SK, Qwarnstrom EE, Lewis CE. Response of tumour cells to hypoxia: role of p53 and NFkB. Mol Pathol. 1998;51(2):55–61.
  • Tafani M, Pucci B, Russo A, et al. Modulators of HIF1alpha and NFkB in Cancer Treatment: Is it a Rational Approach for Controlling Malignant Progression? Front Pharmacol. 2013;4:13.
  • 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(33):25130–25138.
  • Movafagh S, Crook S, Vo K. Regulation of hypoxia-inducible factor-1a by reactive oxygen species: new developments in an old debate. J Cell Biochem. 2015;116(5):696–703.
  • Mateo J, Garcia-Lecea M, Cadenas S, Hernandez C, Moncada S. Regulation of hypoxia-inducible factor-1alpha by nitric oxide through mitochondria-dependent and -independent pathways. Biochem J. 2003;376(Pt 2):537–544.
  • Semenza GL. Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy. Trends Pharmacol Sci. 2012;33(4):207–214.
  • Bertout JA, Patel SA, Simon MC. The impact of O2 availability on human cancer. Nat Rev Cancer. 2008;8(12):967–975.
  • Muz B, de la Puente P, Azab F, Luderer M, Azab AK. The role of hypoxia and exploitation of the hypoxic environment in hematologic malignancies. Mol Cancer Res. 2014;12(10):1347–1354.
  • Siemeister G, Weindel K, Mohrs K, Barleon B, Martiny-Baron G, Marme D. Reversion of deregulated expression of vascular endothelial growth factor in human renal carcinoma cells by von Hippel-Lindau tumor suppressor protein. Cancer Res. 1996;56(10):2299–2301.
  • Zagzag D, Krishnamachary B, Yee H, et al. Stromal cell-derived factor-1alpha and CXCR4 expression in hemangioblastoma and clear cell-renal cell carcinoma: von Hippel-Lindau loss-of-function induces expression of a ligand and its receptor. Cancer Res. 2005;65(14):6178–6188.
  • Iyer NV, Kotch LE, Agani F, et al. Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1 alpha. Genes Dev. 1998;12(2):149–162.
  • Iliopoulos O, Levy AP, Jiang C, Kaelin WG Jr, Goldberg MA. Negative regulation of hypoxia-inducible genes by the von Hippel-Lindau protein. Proc Natl Acad Sci U S A. 1996;93(20):10595–10599.
  • Semenza GL. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J Clin Invest. 2013;123(9):3664–3671.
  • Guo K, Searfoss G, Krolikowski D, et al. Hypoxia induces the expression of the pro-apoptotic gene BNIP3. Cell Death Differ. 2001;8(4):367–376.
  • An WG, Kanekal M, Simon MC, Maltepe E, Blagosklonny MV, Neckers LM. Stabilization of wild-type p53 by hypoxia-inducible factor 1 alpha. Nature. 1998;392(6674):405–408.
  • Roberts AM, Watson IR, Evans AJ, Foster DA, Irwin MS, Ohh M. Suppression of hypoxia-inducible factor 2alpha restores p53 activity via Hdm2 and reverses chemoresistance of renal carcinoma cells. Cancer Res. 2009;69(23):9056–9064.
  • Krishnamachary B, Zagzag D, Nagasawa H, et al. Hypoxia-inducible factor-1-dependent repression of E-cadherin in von Hippel-Lindau tumor suppressor-null renal cell carcinoma mediated by TCF3, ZFHX1A, and ZFHX1B. Cancer Res. 2006;66(5):2725–2731.
  • Staller P, Sulitkova J, Lisztwan J, Moch H, Oakeley EJ, Krek W. Chemokine receptor CXCR4 downregulated by von Hippel-Lindau tumour suppressor pVHL. Nature. 2003;425(6955):307–311.
  • Grabmaier K, MC AdW, Verhaegh GW, Schalken JA, Oosterwijk E. Strict regulation of CAIX(G250/MN) by HIF-1alpha in clear cell renal cell carcinoma. Oncogene. 2004;23(33):5624–5631.
  • Erler JT, Bennewith KL, Nicolau M, et al. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature. 2006;440(7088):1222–1226.
  • Krishnamachary B, Berg-Dixon S, Kelly B, et al. Regulation of colon carcinoma cell invasion by hypoxia-inducible factor 1. Cancer Res. 2003;63(5):1138–1143.
  • Azab AK, Hu J, Quang P, et al. Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. Blood. 2012;119(24):5782–5794.
  • Conway EM, Collen D, Carmeliet P. Molecular mechanisms of blood vessel growth. Cardiovasc Res. 2001;49(3):507–521.
  • Carmeliet P. Angiogenesis in life, disease and medicine. Nature. 2005;438(7070):932–936.
  • de la Puente P, Muz B, Azab F, Azab AK. Cell Trafficking of Endothelial Progenitor Cells in Tumor Progression. Clin Cancer Res. 2013; 19(3):3360–3368.
  • Krock BL, Skuli N, Simon MC. Hypoxia-induced angiogenesis: good and evil. Genes Cancer. 2011;2(12):1117–1133.
  • Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature. 2011;473(7347):298–307.
  • Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285(21):1182–1186.
  • Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350(23):2335–2342.
  • Mountzios G, Pentheroudakis G, Carmeliet P. Bevacizumab and micrometastases: revisiting the preclinical and clinical rollercoaster. Pharmacol Ther. 2014;141(2):117–124.
  • Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307(5706):58–62.
  • Semenza GL. Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene. 2010;29(5):625–634.
  • Rohwer N, Lobitz S, Daskalow K, et al. HIF-1alpha determines the metastatic potential of gastric cancer cells. Br J Cancer. 2009; 100(5):772–781.
  • Muz B, de la Puente P, Azab F, Ghobrial IM, Azab AK. Hypoxia promotes dissemination and colonization in new bone marrow niches in Waldenstrom macroglobulinemia. Mol Cancer Res. 2015;13(2):263–272.
  • Cairns RA, Hill RP. Acute hypoxia enhances spontaneous lymph node metastasis in an orthotopic murine model of human cervical carcinoma. Cancer Res. 2004;64(6):2054–2061.
  • Cairns RA, Kalliomaki T, Hill RP. Acute (cyclic) hypoxia enhances spontaneous metastasis of KHT murine tumors. Cancer Res. 2001;61(24):8903–8908.
  • Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7(2):131–142.
  • Mulholland DJ, Kobayashi N, Ruscetti M, et al. Pten loss and RAS/MAPK activation cooperate to promote EMT and metastasis initiated from prostate cancer stem/progenitor cells. Cancer Res. 2012;72(7):1878–1889.
  • Kim K, Lu Z, Hay ED. Direct evidence for a role of beta-catenin/LEF-1 signaling pathway in induction of EMT. Cell Biol Int. 2002; 26(5):463–476.
  • Hsu M, Andl T, Li G, Meinkoth JL, Herlyn M. Cadherin repertoire determines partner-specific gap junctional communication during melanoma progression. J Cell Sci. 2000;113(Pt 9):1535–1542.
  • Manotham K, Tanaka T, Matsumoto M, et al. Transdifferentiation of cultured tubular cells induced by hypoxia. Kidney Int. 2004;65(3):871–880.
  • Tian M, Neil JR, Schiemann WP. Transforming growth factor-beta and the hallmarks of cancer. Cell Signal. 2011;23(6):951–962.
  • Tamara Marie-Egyptienne D, Lohse I, Hill RP. Cancer stem cells, the epithelial to mesenchymal transition (EMT) and radioresistance: Potential role of hypoxia. Cancer Lett. 2013;341(1):63–72.
  • Greenberger LM, Horak ID, Filpula D, et al. A RNA antagonist of hypoxia-inducible factor-1alpha, EZN-2968, inhibits tumor cell growth. Mol Cancer Ther. 2008;7(11):3598–3608.
  • Hu J, Handisides DR, Van Valckenborgh E, et al. Targeting the multiple myeloma hypoxic niche with TH-302, a hypoxia-activated prodrug. Blood. 2010;116(9):1524–1527.
  • 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(9):3364–3376.
  • Erler JT, Bennewith KL, Cox TR, et al. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 2009;15(1):35–44.
  • Azab AK, Azab F, Blotta S, et al. RhoA and Rac1 GTPases play major and differential roles in stromal cell-derived factor-1-induced cell adhesion and chemotaxis in multiple myeloma. Blood. 2009; 114(3):619–629.
  • Azab AK, Runnels JM, Pitsillides C, et al. CXCR4 inhibitor AMD3100 disrupts the interaction of multiple myeloma cells with the bone marrow microenvironment and enhances their sensitivity to therapy. Blood. 2009;113(18):4341–4351.
  • Azab AK, Weisberg E, Sahin E, et al. The influence of hypoxia on CML trafficking through modulation of CXCR4 and E-cadherin expression. Leukemia. 2013;27(4):961–964.
  • Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nat Rev Cancer. 2011;11(6):393–410.
  • Munshi NC, Anderson KC. Minimal residual disease in multiple myeloma. J Clin Oncol. 2013;31(20):2523–2526.
  • Rohwer N, Cramer T. Hypoxia-mediated drug resistance: novel insights on the functional interaction of HIFs and cell death pathways. Drug Resist Updat. 2011;14(3):191–201.
  • Gray LH, Conger AD, Ebert M, Hornsey S, Scott OC. The concentration of oxygen dissolved in tissues at the time of irradiation as a factor in radiotherapy. Br J Radiol. 1953;26(312):638–648.
  • Vaupel P, Kelleher DK, Hockel M. Oxygen status of malignant tumors: pathogenesis of hypoxia and significance for tumor therapy. Semin Oncol. 2001;28(2 Suppl 8):29–35.
  • Das B, Tsuchida R, Malkin D, Koren G, Baruchel S, Yeger H. Hypoxia enhances tumor stemness by increasing the invasive and tumorigenic side population fraction. Stem Cells. 2008;26(7):1818–1830.
  • Abraham J, Salama NN, Azab AK. The role of P-glycoprotein in drug resistance in multiple myeloma. Leuk Lymphoma. 2015;56(1):26–33.
  • Cosse JP, Michiels C. Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression. Anticancer Agents Med Chem. 2008;8(7):790–797.
  • Pawlik TM, Keyomarsi K. Role of cell cycle in mediating sensitivity to radiotherapy. Int J Radiat Oncol Biol Phys. 2004;59(4):928–942.
  • Muz B, de la Puente P, Azab F, Luderer M, Azab AK. Hypoxia promotes stem cell-like phenotype in multiple myeloma cells. Blood Cancer J. 2014;4:e262.
  • Bao S, Wu Q, McLendon RE, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756–760.
  • Aebersold DM, Burri P, Beer KT, et al. Expression of hypoxia-inducible factor-1alpha: a novel predictive and prognostic parameter in the radiotherapy of oropharyngeal cancer. Cancer Res. 2001; 61(7):2911–2916.
  • Koukourakis MI, Giatromanolaki A, Sivridis E, et al. Hypoxia-inducible factor (HIF1A and HIF2A), angiogenesis, and chemoradiotherapy outcome of squamous cell head-and-neck cancer. Int J Radiat Oncol Biol Phys. 2002;53(5):1192–1202.
  • Moeller BJ, Cao Y, Vujaskovic Z, Li CY, Haroon ZA, Dewhirst MW. The relationship between hypoxia and angiogenesis. Semin Radiat Oncol. 2004;14(3):215–221.
  • Unruh A, Ressel A, Mohamed HG, et al. The hypoxia-inducible factor-1 alpha is a negative factor for tumor therapy. Oncogene. 2003; 22(21):3213–3220.
  • Schwartz DL, Powis G, Thitai-Kumar A, et al. The selective hypoxia inducible factor-1 inhibitor PX-478 provides in vivo radiosensitization through tumor stromal effects. Mol Cancer Ther. 2009; 8(4):947–958.
  • Bertout JA, Majmundar AJ, Gordan JD, et al. HIF2alpha inhibition promotes p53 pathway activity, tumor cell death, and radiation responses. Proc Natl Acad Sci U S A. 2009;106(34):14391–14396.
  • Sounni NE, Noel A. Targeting the tumor microenvironment for cancer therapy. Clin Chem. 2013;59(1):85–93.
  • Casazza A, Di Conza G, Wenes M, Finisguerra V, Deschoemaeker S, Mazzone M. Tumor stroma: a complexity dictated by the hypoxic tumor microenvironment. Oncogene. 2014;33(14):1743–1754.
  • Baudry N, Danialou G, Boczkowski J, Vicaut E. In vivo study of the effect of systemic hypoxia on leukocyte-endothelium interactions. Am J Respir Crit Care Med. 1998;158(2):477–483.
  • Kourembanas S, Bernfield M. Hypoxia and endothelial-smooth muscle cell interactions in the lung. Am J Respir Crit Care Med. 1994; 11(4):373–374.
  • Michiels C, Arnould T, Remacle J. Endothelial cell responses to hypoxia: initiation of a cascade of cellular interactions. Biochim Biophys Acta. 2000;1497(1):1–10.
  • Verdegem D, Moens S, Stapor P, Carmeliet P. Endothelial cell metabolism: parallels and divergences with cancer cell metabolism. Cancer Metab. 2014;2:19.
  • Azab AK, Sahin I, Moschetta M, et al. CXCR7-dependent angiogenic mononuclear cell trafficking regulates tumor progression in multiple myeloma. Blood. 2014;124(12):1905–1914.
  • Semenza GL. Cancer-stromal cell interactions mediated by hypoxia-inducible factors promote angiogenesis, lymphangiogenesis, and metastasis. Oncogene. 2013;32(35):4057–4063.
  • Lee CT, Mace T, Repasky EA. Hypoxia-driven immunosuppression: a new reason to use thermal therapy in the treatment of cancer? Int J Hyperthermia. 2010;26(3):232–246.
  • Noman MZ, Hasmim M, Messai Y, et al. Hypoxia: a key player in anti-tumor immune response. A Review in the Theme: Cellular Responses to Hypoxia. Am J Physiol Cell Physiol. 2015;309(9):C569–C579.
  • Siemens DR, Hu N, Sheikhi AK, et al. Hypoxia increases tumor cell shedding of MHC class I chain-related molecule: role of nitric oxide. Cancer Res. 2008;68(12):4746–4753.
  • Yang M, Ma C, Liu S, et al. Hypoxia skews dendritic cells to a T helper type 2-stimulating phenotype and promotes tumour cell migration by dendritic cell-derived osteopontin. Immunology. 2009;128 (1 Suppl):e237–e249.
  • Sitkovsky MV. T regulatory cells: hypoxia-adenosinergic suppression and re-direction of the immune response. Trends Immunol. 2009; 30(3):102–108.
  • Rampling R, Cruickshank G, Lewis AD, Fitzsimmons SA, Workman P. Direct measurement of pO2 distribution and bioreductive enzymes in human malignant brain tumors. Int J Radiat Oncol Biol Phys. 1994;29(3):427–431.
  • Vaupel P, Schlenger K, Knoop C, Hockel M. Oxygenation of human tumors: evaluation of tissue oxygen distribution in breast cancers by computerized O2 tension measurements. Cancer Res. 1991; 51(12):3316–3322.
  • Lawrentschuk N, Poon AM, Foo SS, et al. Assessing regional hypoxia in human renal tumours using 18F-fluoromisonidazole positron emission tomography. BJU Int. 2005;96(4):540–546.
  • Leary TS, Klinck JR, Hayman G, Friend P, Jamieson NV, Gupta AK. Measurement of liver tissue oxygenation after orthotopic liver transplantation using a multiparameter sensor. A pilot study. Anaesthesia. 2002;57(11):1128–1133.
  • Brooks AJ, Eastwood J, Beckingham IJ, Girling KJ. Liver tissue partial pressure of oxygen and carbon dioxide during partial hepatectomy. Br J Anaesth. 2004;92(5):735–737.
  • Le QT, Chen E, Salim A, et al. An evaluation of tumor oxygenation and gene expression in patients with early stage non-small cell lung cancers. Clin Cancer Res. 2006;12(5):1507–1514.
  • Koong AC, Mehta VK, Le QT, et al. Pancreatic tumors show high levels of hypoxia. Int J Radiat Oncol Biol Phys. 2000;48(4):919–922.
  • Graffman S, Bjork P, Ederoth P, Ihse I. Polarographic pO2 measurements of intra-abdominal adenocarcinoma in connection with intraoperative radiotherapy before and after change of oxygen concentration of anaesthetic gases. Acta Oncol. 2001;40(1):105–107.
  • Kallinowski F, Eble MJ, Buhr HJ, Wannenmacher M, Herfarth C. Intraoperative radiotherapy for primary and recurrent rectal cancer. Eur J Surg Oncol. 1995;21(2):191–194.