Current research progress on hypoxia and tumor

Authors

  • Jafar Abdel-Hadi Department of Urology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
  • Adnan Mesmar Department of General Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China

DOI:

https://doi.org/10.18203/2349-3933.ijam20182002

Keywords:

HIF-1α, HIF-2α, Hypoxia, Tumor

Abstract

Hypoxia is the decrease of normal oxygen tension in tissues. It often occurs in the tissues of acute and chronic vascular diseases or lung diseases and cancerous lesions. Severe or persistent hypoxia can lead to the death of cell. The lack of oxygen in the tumor is mainly due to the absence or deficiency of normal tumor blood vessel function, which results in a decrease of the oxygen diffusion function within the tumor, eventually leading to a series of biochemical reactions. Although hypoxia is toxic on tumor cells, tumor cells still have a strong adaptability. In the hypoxic microenvironment of tumors, tumor cells can produce adaptive changes or genetic changes so that the tumor could survive in an oxygen-deficient environment, even proliferate. Finally, tumor progression will happen. This process visualizes malignant phenotype of the tumor and makes them more aggressive. The epithelial mesenchymal transition in the tumor microenvironment plays an important role in tumor cell migration, invasion, malignant progression and metastasis. Hypoxia leads to epithelial-mesenchymal transition in tumor tissue, reconstitution of extracellular matrix in tumors, and induction of resistance to antitumor drugs. There are many researches related to hypoxia and tumors. The research contents are complex. Targeted therapy for hypoxia is receiving more and more attention, but the overlapping of signal molecules due to the related mechanism between hypoxia and tumors. An effective hypoxia-targeted therapeutic drug that can be used clinically which requires hard work and arduous exploration from researchers.

References

Hansen JM, Coleman RL, Sood AK. Targeting the tumour microenvironment in ovarian cancer. Eur J Cancer. 2016;56:131-43.

Giakoustidis A, Mudan S, Hagemann T. Tumour microenvironment: overview with an emphasis microenvironment Colorectal Liver Metastasis Pathway. Cancer. 20158(3):177-86.

McKeown SR. Defining normoxia, physoxia and hypoxia in tumours-implications for treatment response. Br J Radiol. 2014;87(1035):20130676.

Thiery JP, Acloque H, Huang RY. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871-90.

Thews O, Wolloscheck T, Dillenburg W. Microenvironmental adaptation of experimental tumours to chronic vs acute hypoxia. Br J Cancer. 2004;91(6):1181-9.

Thomlinson RH, Gray LH. The histological structure of some human lung cancers and the possible implications for radiotherapy. Br J Cancer. 1955;9(4):539-49.

Chen WL, Wang CC, Lin YJ. Cycling hypoxia induces chemoresistance through the activation of reactive oxygen species-mediated B-cell lymphoma extra-long pathway in glioblastoma multiforme. J Transl Med. 2015;13:389.

Mujcic H, Hill RP, Koritzinsky M. Hypoxia signalling and the metastatic phenotype. Curr Molecular Med. 2014;14(5):565-79.

Sullivan R, Graham CH. Hypoxia-driven selection of the metastatic phenotype. Cancer Metastasis Rev. 2007;26(2):319-31.

Wong SC, Foster JG, Sharp TV. The hypoxic tumor microenvironment: driving the tumorigenesis of non-small-cell lung cancer. Future Oncol. 2014;10(16):2659-74.

Sotgia F, Martinez-Outschoorn U, Lisanti MP. Tumor microenvironment and metabolic synergy in breast cancers: critical importance of mitochondrial fuels and function. Semin Oncol. 2014;41(2):195-216.

Labiano S, Palazon A, Melero I. Immune response regulation in the tumor microenvironment by hypoxia. Semin Oncol. 2015;42(3):378-86.

Finlay DK, Rosenzweig E, Sinclair LV, Feijoo-Carnero C, Hukelmann JL, Rolf J, et al. PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8+ T cells. J Exp Med. 2012;209(13):2441-53.

Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41(1):49-61.

Carmeliet P, Jain RK. Molecular mechanisms and clinical applications of angiogenesis. Nature Biotech. 2011;473:298-307.

Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Sci. 2005;307:58-62.

Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell. 2014;26(5):605-22.

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.

Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature. 2014;513:559-63.

Motz GT, Coukos G. Deciphering and reversing tumor immune suppression. Immunity. 2013;39:61-73.

Finger EC, Giaccia AJ. Hypoxia, inflammation, and the tumor microenvironment in metastatic disease. Cancer Metastasis Rev. 2010;29:285-93.

Lanning RM, Vakoc BJ, Tyrrell JA. Three-dimensional microscopy of the tumor microenvironment in vivo using optical frequency domain imaging. Nat Med. 2009;15:1219-23.

Jain RK. Determinants of tumor blood flow: a review. Cancer Res. 1988;48:2641-58.

Stoll BR, Padera TP, Tooredman JB. Pathology: cancer cells compress intratumour vessels. Nature. 2004;427:695.

Sckell A, Helmlinger G, Dellian M. Acid production in glycolysis-impaired tumors provides new insights into tumor metabolism. Clin Cancer Res. 2002;8:1284-91.

Cohen T, Gitay-Goren H, Sharon R, Shibuya M, Halaban R, Levi BZ, et al. VEGF121, a vascular endothelial growth factor (VEGF) isoform lacking heparin binding ability, requires cell surface heparan sulfates for efficient binding to the VEGF receptors of human melanoma cells. J Biol Chem. 1995;270(19):11322-6.

Ferrara N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev. 2004;25(4):581-611.

Kowalski J, Gerber HP, Sherman D, Eberhard DA, Ferrara N. Complete inhibition of rhabdomyosarcoma xenograft growth and neovascularization requires blockade of both tumor and host vascular endothelial growth factor. Cancer Res. 2000;60(22):6253-8.

Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat Med. 2001;7(9):987-9.

Zage PE, Nilsson MB, Zeng L, Xu L, Cascone T, Wu HK, et al. Multiple receptor tyrosine kinases regulate HIF-1 alpha and HIF-2 alpha in normoxia and hypoxiain neuroblastoma: implications for antiangiogenic mechanisms of multikinase inhibitors. Oncogene. 2010;29(20):2938-49.

Fredlund E, Holmquist-Mengelbier L, Lofstedt T, Noguera R, Navarro S, Nilsson H, et al. Recruitment of HIF-lalpha and HIF-2alpha to common target genes is differentially regulated in neuroblastoma: HIF-2alpha promotes an aggressive phenotype. Cancer Cell. 2006;10(5):413-23.

Yeger H, Das B, Tsuchida R, Torkin R, Gee MF, Thorner PS, et al. A hypoxia-driven vascular endothelial growth factor/Fltl autocrine loop interactswith hypoxia-inducible factor-lalpha through mitogen-activated proteinkinase/extracellular signal-regulated kinase 1/2 pathway in neuroblastoma. Cancer Res. 2005;65(16):7267-75.

Wang L, Tang N, Esko J, Giordano FJ, Huang Y, Gerber HP, et al. Loss of HIF-lalpha inendothelial cells disrupts a hypoxia-driven VEGF autocrine loop necessary for tumorigenesis. Cancer Cell. 2004;6(5):485-495.

Ferrara N. Role of vascular endothelial growth factor in physiologic and patho-logic angiogenesis: therapeutic implications. Semin Oncol. 2002;29(6):10-4.

Dias S, Santos SC. Internal and external autocrine VEGF/KDR loops regulate survival of subsets of acute leukaemia through distinct signalling pathways. Blood. 2004;103(10):3883-9.

Makino Y, Nakamura H, Okamoto K. TCR engagement increases hypoxia-inducible factor-lalpha protein synthesis via rapamycin-sensitive pathway under hypoxic conditions in human peripheral T cells. J Immunol. 2005;174(12):7592-5.

Caldwell CC, Thiel M, Kreth S. Targeted deletion of HIF-lalpha gene in T cells prevents their inhibition in hypoxic inflamed tissues and improves septic mice survival. PLoS One. 2007;2(9):e853.

Peng X, Facciabene A, Hagemann IS. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T (reg) cells. Nature. 2011;475(7335):226-30.

Noman MZ, Hasmim M, Messai Y. Cutting edge: hypoxia-induced nanog favours the intratumoral infiltration of regulatory T cells and macrophages via direct regulation of TGF-beta. J Immunol. 2013;191(12):5802-6.

Noman MZ Hasmim M, Lauriol J. Hypoxia-dependent inhibition of tumor cell susceptibility to CTL-mediated lysis involves NANOG induction in target cells. J Immunol. 2011;187(8):4031-9.

Kjaergaard J, Sitkovsky MV, Lukashev D. Hypoxia-adenosinergic immuno suppression: tumor protection by T regulatory cells and cancerous tissue hypoxia. Clin Cancer Res. 2008;14(19):5947-52.

Curtis JD Chang CH, Maggi LB. Posttranscriptional control of T cell effect or function by aerobic glycolysis. Cell. 2013;153(6):1239-51.

Nejfelt MK, Semenza GL, Chi SM, Antonarakis SE. Hypoxia-inducible nuclear factors bind to an enhancer element located 3‘to the human erythropoietin gene. Proc Natl Acad Sci USA. 1991;88:5680-4.

Dunning SP, Goldberg MA, Bunn HF. Regulation of the erythropoietin gene: evidence that the oxygen sensor is a hemeprotein. Sci. 1988;242:1412-5.

Pongratz I, Kallio PJ, Gradin K, McGuire J, Poellinger L. Activation of hypoxia-inducible factor 1 a: post transcriptional regulation and conformational change by recruitment of the Arnt transcription factor. Proc Natl Acad Sci USA. 1997;94:5667-5672.

Masoud GN, Li W. HIF-1α pathway: role, regulation and intervention for cancer therapy. Acta Pharm Sin B. 2015;5(5):378-89.

McKnight SL, Tian H, Russell DW. Endothelial PAS domain protein 1 (EPAS 1), a transcription factor selectively expressed in endothelial cells. Genes Dev. 1997;11:72-82.

Jurgensen JS, Wiesener MS, Rosenberger C, Scholze CK, Horstrup JH, Warnecke C, et al. Widespread hypoxia-inducible expression of HIF-2alpha in distinct cell populations of different organs. FASEB J. 2003;17:271-3.

Turley H, Talks KL, Gatter KC, Maxwell PH, Pugh CW, Ratcliffe PJ. The expression and distribution of the hypoxia-inducible factors HIF-la and HIF-2a in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol. 2000;157:411-21.

Mazzone M, Fraisl P, Schmidt T, Carmeliet P. Regulation of angiogenesis by oxygen and metabolism. Dev Cell. 2009;16:167-79.

Simon MC, Keith B. Hypoxia-inducible factors, stem cells, and cancer. Cell. 2007;129:465-72.

Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;28(5):1182-6.

Lee SL, Rouhi P, Cao Z, Hedlund EM, Jensen LD, Cao Y. Pathological angiogenesis facilitates tumor cell dissemination and metastasis. Cell Cycle. 2010;9:913-7.

Laoui D, Casazza A, Wenes M, Rizzolio S, Bassani N, Mambretti M, et al. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrpl signalling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell. 2013;24:695-709.

Semenza GL. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J Clin Invest. 2013;123:3664-71.

Wu WS. The signalling mechanism of ROS in tumor progression. Cancer Metastasis Rev. 2006;2(5):695-705.

Sendoel A, Kohler I, Fellmann C. HIF-1 antagonizes p53-mediated apoptosis through a secreted neuronal tyrosinase. Nature. 2010;465(7298):577-83.

Brown LM, Cowen RL, Debray C, Eustace A, Erler, JT, Sheppard FC. Reversing hypoxic cell chemoresistance in vitro using genetic and small molecule approaches targeting hypoxia inducible factor-1. Mol Pharmacol. 2006;69:411-8.

Ressel AU, Mohamed A, Johnson HG, Nadrowitz RS, Richter R, Katschinski E, et al. The hypoxia-inducible factor-1 alpha is a negative factor for tumor therapy. Oncogene. 2003;22:3213-20.

Chen L, Feng PM, Li SF, Long D, Cheng JQ, Lu YR, et al. Effect of hypoxia-inducible factor-1 alpha silencing on the sensitivity of human brain glioma cells to doxorubicin and etoposide. Neurochem. Res. 2009;34:984-90.

Roberts AM, Watson LR, 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;69:9056-64.

Downloads

Published

2018-05-22

Issue

Section

Review Articles