Skip to main content
Log in

Tumor Hypoxia As an Enhancer of Inflammation-Mediated Metastasis: Emerging Therapeutic Strategies

  • Review Article
  • Published:
Targeted Oncology Aims and scope Submit manuscript

Abstract

Metastasis is the leading cause of cancer-related deaths. Recent research has implicated tumor inflammation as a promoter of metastasis. Myeloid, lymphoid, and mesenchymal cells in the tumor microenvironment promote inflammatory signaling amongst each other and together with cancer cells to modulate sustained inflammation, which may enhance cancer invasiveness. Tumor hypoxia, a state of reduced available oxygen present in the majority of solid tumors, acts as a prognostic factor for a worse outcome and is known to have a role in tumor inflammation through the regulation of inflammatory mediator signals in both cancer and neighboring cells in the microenvironment. Multiple methods to target tumor hypoxia have been developed and tested in clinical trials, and still more are emerging as the impacts of hypoxia become better understood. These strategies include mechanistic inhibition of the hypoxia inducible factor signaling pathway and hypoxia activated pro-drugs, leading to both anti-tumor and anti-inflammatory effects. This prompts a need for further research on the prevention of hypoxia-mediated inflammation in cancer. Hypoxia-targeting strategies seem to have the most potential for therapeutic benefit when combined with traditional chemotherapy agents. This paper will serve to summarize the role of the inflammatory response in metastasis, to discuss how hypoxia can enable or enhance inflammatory signaling, and to review established and emerging strategies to target the hypoxia-inflammation-metastasis axis.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Mehlen P, Puisieux A. Metastasis: a question of life or death. Nat Rev Cancer. 2006;6:449–58.

    Article  CAS  PubMed  Google Scholar 

  2. Chambers AF, Groom AC, MacDonald IC. Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2002;2:563–72.

    Article  CAS  PubMed  Google Scholar 

  3. Bald T, Quast T, Landsberg J, Rogava M, Glodde N, Lopez-Ramos D, et al. Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis in melanoma. Nature. 2014;507:109–13.

    Article  CAS  PubMed  Google Scholar 

  4. Hansen MT, Forst B, Cremers N, Quagliata L, Ambartsumian N, Grum-Schwensen B, et al. A link between inflammation and metastasis: serum amyloid A1 and A3 induce metastasis, and are targets of metastasis-inducing S100A4. Oncogene. 2015;34:424–35.

    Article  CAS  PubMed  Google Scholar 

  5. El Rayes T, Catena R, Lee S, Stawowczyk M, Joshi N, Fischbach C, et al. Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1. Proc Natl Acad Sci U S A. 2015;112:16000–5. 

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Hugo HJ, Saunders C, Ramsay RG, Thompson EW. New insights on COX-2 in chronic inflammation driving breast cancer growth and metastasis. J Mammary Gland Biol Neoplasia. 2015;20:109–19.

    Article  PubMed  Google Scholar 

  7. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–76.

    Article  CAS  PubMed  Google Scholar 

  8. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650–9.

    Article  CAS  PubMed  Google Scholar 

  9. Crusz SM, Balkwill FR. Inflammation and cancer: advances and new agents. Nat Rev Clin Oncol. 2015;12:584–96.

    Article  CAS  PubMed  Google Scholar 

  10. Hawinkels LJAC, Paauwe M, Verspaget HW, Wiercinska E, van der Zon JM, van der Ploeg K, et al. Interaction with colon cancer cells hyperactivates TGF-β signaling in cancer-associated fibroblasts. Oncogene. 2014;33:97–107.

    Article  CAS  PubMed  Google Scholar 

  11. Bonapace L, Coissieux M-M, Wyckoff J, Mertz KD, Varga Z, Junt T, et al. Cessation of CCL2 inhibition accelerates breast cancer metastasis by promoting angiogenesis. Nature. 2014;515:130–3.

    Article  CAS  PubMed  Google Scholar 

  12. Qian BZ. Inflammation fires up cancer metastasis. Semin Cancer Biol. 2017;47:170–6.

    Article  CAS  PubMed  Google Scholar 

  13. Fan QM, Jing YY, Yu GF, Kou XR, Ye F, Gao L, et al. Tumor-associated macrophages promote cancer stem cell-like properties via transforming growth factor-beta1-induced epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett. 2014;352:160–8.

    Article  CAS  PubMed  Google Scholar 

  14. Yu Y, Xiao C-H, Tan L-D, Wang Q-S, Li X-Q, Feng Y-M. Cancer-associated fibroblasts induce epithelial-mesenchymal transition of breast cancer cells through paracrine TGF-β signalling. Br J Cancer. 2014;110:724–32.

    Article  CAS  PubMed  Google Scholar 

  15. Qian BZ, Li J, Zhang H, Kitamura T, Zhang J, Campion LR, et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature. 2011;475:222–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Iannitti RG, Casagrande A, De Luca A, Cunha C, Sorci G, Riuzzi F, et al. Hypoxia promotes danger-mediated inflammation via receptor for advanced glycation end products in cystic fibrosis. Am J Respir Crit Care Med. 2013;188:1338–50.

    Article  CAS  PubMed  Google Scholar 

  17. Petrangeli E, Coroniti G, Brini AT, de Girolamo L, Stanco D, Niada S, et al. Hypoxia promotes the inflammatory response and Stemness features in visceral fat stem cells from obese subjects. J Cell Physiol. 2016;231:668–79.

    Article  CAS  PubMed  Google Scholar 

  18. Murdoch C, Muthana M, Lewis CE. Hypoxia regulates macrophage functions in inflammation. J Immunol. 2005;175:6257–63.

    Article  CAS  PubMed  Google Scholar 

  19. Pennacchietti S, Michieli P, Galluzzo M, Mazzone M, Giordano S, Comoglio PM. Hypoxia promotes invasive growth by transcriptional activation of the met protooncogene. Cancer Cell. 2003;3:347–61.

    Article  PubMed  Google Scholar 

  20. Azab AK, Hu J, Quang P, Azab F, Pitsillides C, Awwad R, et al. Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. Blood. 2012;119:5782–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rofstad EK, Rasmussen H, Galappathi K, Mathiesen B, Nilsen K, Graff BA. Hypoxia promotes lymph node metastasis in human melanoma xenografts by up-regulating the urokinase-type plasminogen activator receptor. Cancer Res. 2002;62:1847–53.

    CAS  PubMed  Google Scholar 

  22. Zhang H, Wong CCL, Wei H, Gilkes DM, Korangath P, Chaturvedi P, et al. HIF-1-dependent expression of angiopoietin-like 4 and L1CAM mediates vascular metastasis of hypoxic breast cancer cells to the lungs. Oncogene. 2012;31:1757–70.

    Article  CAS  PubMed  Google Scholar 

  23. Vaupel P, Mayer A, Hockel M. Tumor hypoxia and malignant progression. Methods Enzymol. 2004;23:335–54.

    Article  Google Scholar 

  24. Hockel M, Schlenger K, Aral B, Mitze M, Schaffer U, Vaupel P. Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. Cancer Res. 1996;56:4509–15.

    CAS  PubMed  Google Scholar 

  25. Höckel M, Knoop C, Schlenger K, Vorndran B, Bauβnann E, Mitze M, et al. Intratumoral pO2 predicts survival in advanced cancer of the uterine cervix. Radiother Oncol. 1993;26:45–50.

    Article  PubMed  Google Scholar 

  26. 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;12:5447–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang GL, Semenza GL. Purification and characterization of hypoxia-inducible factor 1. J Biol Chem. 1995;270:1230–7.

    Article  CAS  PubMed  Google Scholar 

  28. Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci U S A. 1995;92:5510–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Ema M, Taya S, Yokotani N, Sogawa K, Matsuda Y, Fujii-Kuriyama Y. A novel bHLH-PAS factor with close sequence similarity to hypoxia-inducible factor 1 regulates the VEGF expression and is potentially involved in lung and vascular development. Proc Natl Acad Sci U S A. 1997;94:4273–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tian H, McKnight SL, Russell DW. Endothelial PAS domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes Dev. 1997;11:72–82.

    Article  CAS  PubMed  Google Scholar 

  31. Hogenesch JB, Chan WK, Jackiw VH, Brown RC, Gu YZ, Pray-Grant M, et al. Characterization of a subset of the basic-helix-loop-helix-PAS superfamily that interacts with components of the dioxin signaling pathway. J Biol Chem. 1997;272:8581–93.

    Article  CAS  PubMed  Google Scholar 

  32. Semenza GL. HIF-1 and mechanisms of hypoxia sensing. Curr Opin Cell Biol. 2001;13:167–71.

    Article  CAS  PubMed  Google Scholar 

  33. Bruick RK, McKnight SL. A conserved family of prolyl-4-hydroxylases that modify HIF. Science. 2001;294:1337–40.

    Article  CAS  PubMed  Google Scholar 

  34. Epstein ACR, Gleadle JM, McNeill LA, Hewitson KS, O’Rourke J, Mole DR, 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.

    Article  CAS  PubMed  Google Scholar 

  35. Maxwell PH, Wlesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399:271–5.

    Article  CAS  PubMed  Google Scholar 

  36. Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, et al. Ubiquitination of hypoxia-inducible factor requires direct binding to the β-domain of the von Hippel - Lindau protein. Nat Cell Biol. 2000;2:423–7.

    Article  CAS  PubMed  Google Scholar 

  37. Semenza GL. HIF-1: mediator of physiological and pathophysiological responses to hypoxia. J Appl Physiol. 2000;88:1474–80.

    Article  CAS  PubMed  Google Scholar 

  38. Semenza GL. Regulation of oxygen homeostasis by hypoxia-inducible factor 1. Physiology. 2009;24:97–106.

    Article  CAS  PubMed  Google Scholar 

  39. Loboda A, Jozkowicz A, Dulak J. HIF-1 and HIF-2 transcription factors--similar but not identical. Mol Cells. 2010;29:435–42.

    Article  CAS  PubMed  Google Scholar 

  40. Krieg M, Haas R, Brauch H, Acker T, Flamme I, Plate KH. Up-regulation of hypoxia-inducible factors HIF-1α and HIF-2α under normoxic conditions in renal carcinoma cells by von Hippel-Lindau tumor suppressor gene loss of function. Oncogene. 2000;19:5435–43.

    Article  CAS  PubMed  Google Scholar 

  41. Stiehl DP, Jelkmann W, Wenger RH, Hellwig-Bürgel T. Normoxic induction of the hypoxia-inducible factor 1α by insulin and interleukin-1β involves the phosphatidylinositol 3-kinase pathway. FEBS Lett. 2002;512:157–62.

    Article  CAS  PubMed  Google Scholar 

  42. Hudson CC, Liu M, Chiang GG, Otterness DM, Loomis DC, Kaper F, et al. Regulation of hypoxia-inducible factor 1alpha expression and function by the mammalian target of rapamycin. Mol Cell Biol. 2002;22:7004–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gilkes DM, Semenza GL, Wirtz D. Hypoxia and the extracellular matrix: drivers of tumour metastasis. Nat Rev Cancer. 2014;14:430–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Casazza A, Laoui D, Wenes M, Rizzolio S, Bassani N, Mambretti M, et al. Impeding macrophage entry into hypoxic tumor areas by Sema3A/Nrp1 signaling blockade inhibits angiogenesis and restores antitumor immunity. Cancer Cell. 2013;24:695–709.

    Article  CAS  PubMed  Google Scholar 

  45. Gilkes DM, Bajpai S, Chaturvedi P, Wirtz D, Semenza GL. Hypoxia-inducible factor 1 (HIF-1) promotes extracellular matrix remodeling under hypoxic conditions by inducing P4HA1, P4HA2, and PLOD2 expression in fibroblasts. J Biol Chem. 2013;288:10819–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sullivan R, Pare GC, Frederiksen LJ, Semenza GL, Graham CH. Hypoxia-induced resistance to anticancer drugs is associated with decreased senescence and requires hypoxia-inducible factor-1 activity. Mol Cancer Ther. 2008;7:1961–73.

    Article  CAS  PubMed  Google Scholar 

  47. Rohwer N, Cramer T. Hypoxia-mediated drug resistance: novel insights on the functional interaction of HIFs and cell death pathways. Drug Resist Updat. 2011;14:191–201.

    Article  CAS  PubMed  Google Scholar 

  48. Samanta D, Gilkes DM, Chaturvedi P, Xiang L, Semenza GL. Hypoxia-inducible factors are required for chemotherapy resistance of breast cancer stem cells. Proc Natl Acad Sci U S A. 2014;111:E5429–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y. Cancer metastasis is accelerated through Immunosuppression during snail-induced EMT of cancer cells. Cancer Cell. 2009;15:195–206.

    Article  CAS  PubMed  Google Scholar 

  51. Liu VC, Wong LY, Jang T, Shah AH, Park I, Yang X, et al. Tumor evasion of the immune system by converting CD4+CD25- T cells into CD4+CD25+ T regulatory cells: role of tumor-derived TGF-beta. J Immunol. 2007;178:2883–92.

    Article  CAS  PubMed  Google Scholar 

  52. Mitchem JB, Brennan DJ, Knolhoff BL, Belt BA, Zhu Y, Sanford DE, et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013;73:1128–41.

    Article  CAS  PubMed  Google Scholar 

  53. Yang L, Zhang Y. Tumor-associated macrophages: from basic research to clinical application. J Hematol Oncol. 2017;10:58.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers. 2014;6:1670–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Sierra-Filardi E, Nieto C, Dominguez-Soto A, Barroso R, Sanchez-Mateos P, Puig-Kroger A, et al. CCL2 shapes macrophage polarization by GM-CSF and M-CSF: identification of CCL2/CCR2-dependent gene expression profile. J Immunol. 2014;192:3858–67.

    Article  CAS  PubMed  Google Scholar 

  56. Zhou W, Ke SQ, Huang Z, Flavahan W, Fang X, Paul J, et al. Periostin secreted by glioblastoma stem cells recruits M2 tumour-associated macrophages and promotes malignant growth. Nat Cell Biol. 2015;17:170–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Kitamura T, Qian B-Z, Soong D, Cassetta L, Noy R, Sugano G, et al. CCL2-induced chemokine cascade promotes breast cancer metastasis by enhancing retention of metastasis-associated macrophages. J Exp Med. 2015;212:1043–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Su S, Liu Q, Chen J, Chen J, Chen F, He C, et al. A positive feedback loop between mesenchymal-like cancer cells and macrophages is essential to breast cancer metastasis. Cancer Cell. 2014;25:605–20.

    Article  PubMed  CAS  Google Scholar 

  59. Klune JR, Dhupar R, Cardinal J, Billiar TR, Tsung A. HMGB1: endogenous danger signaling. Mol Med. 2008;14:476–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wei B, Yao M, Xing C, Wang W, Yao J, Hong Y, et al. The neutrophil lymphocyte ratio is associated with breast cancer prognosis : an updated systematic review and meta-analysis. Onco Targets Ther. 2016;9:5567–75.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Pang W, Lou N, Jin C, Hu C, Arvine C, Zhu G, et al. Combination of preoperative platelet/lymphocyte and neutrophil/lymphocyte rates and tumor-related factors to predict lymph node metastasis in patients with gastric cancer. Eur J Gastroenterol Hepatol. 2016;28:493–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Wculek SK, Malanchi I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature. 2015;528:413–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gabitass RF, Annels NE, Stocken DD, Pandha HA, Middleton GW. Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol Immunother. 2011;60:1419–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Huang B, Lei Z, Zhao J, Gong W, Liu J, Chen Z, et al. CCL2/CCR2 pathway mediates recruitment of myeloid suppressor cells to cancers. Cancer Lett. 2007;252:86–92.

    Article  CAS  PubMed  Google Scholar 

  65. Obermajer N, Muthuswamy R, Odunsi K, Edwards RP, Kalinski P. PGE 2-induced CXCL 12 production and CXCR4 expression controls the accumulation of human MDSCs in ovarian cancer environment. Cancer Res. 2011;71:7463–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Oh K, Lee OY, Shon SY, Nam O, Ryu PM, Seo MW, et al. A mutual activation loop between breast cancer cells and myeloid-derived suppressor cells facilitates spontaneous metastasis through IL-6 trans-signaling in a murine model. Breast Cancer Res. 2013;15:R79.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, et al. Abrogation of TGFβ signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell. 2008;13:23–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Augsten M. Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment. Front Oncol. 2014;4:62.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Ha SY, Yeo S-Y, Xuan Y, Kim S-H. The prognostic significance of cancer-associated fibroblasts in esophageal Squamous cell carcinoma. PLoS One. 2014;9:e99955.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Comito G, Giannoni E, Segura CP, Barcellos-de-Souza P, Raspollini MR, Baroni G, et al. Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression. Oncogene. 2014;33:2423–31.

    Article  CAS  PubMed  Google Scholar 

  71. Nagasaki T, Hara M, Nakanishi H, Takahashi H, Sato M, Takeyama H. Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour-stroma interaction. Br J Cancer. 2014;110:469–78.

    Article  CAS  PubMed  Google Scholar 

  72. Jayatilaka H, Tyle P, Chen JJ, Kwak M, Ju J, Kim HJ, et al. Synergistic IL-6 and IL-8 paracrine signalling pathway infers a strategy to inhibit tumour cell migration. Nat Commun. 2017;8:15584.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449:557–63.

    Article  CAS  PubMed  Google Scholar 

  74. Lu X, Kang Y. Hypoxia and hypoxia-inducible factors: master regulators of metastasis. Clin Cancer Res. 2010;16:5928–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Gilkes DM. Implications of hypoxia in breast cancer metastasis to bone. Int J Mol Sci. 2016;17:1669.

    Article  PubMed Central  CAS  Google Scholar 

  76. Gilkes DM, Semenza GL. Role of hypoxia-inducible factors in breast cancer metastasis. Future Oncol. 2013;9:1623–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Chan DA, Giaccia AJ. Hypoxia, gene expression, and metastasis. Cancer Metastasis Rev. 2007;26:333–9.

    Article  CAS  PubMed  Google Scholar 

  78. Mimeault M, Batra SK. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer- and metastasis-initiating cells. J Cell Mol Med. 2013;17:30–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Samanta D, Park Y, Andrabi SA, Shelton LM, Gilkes DM, Semenza GL. PHGDH expression is required for mitochondrial redox homeostasis, breast cancer stem cell maintenance, and lung metastasis. Cancer Res. 2016;76:4430–42.

    Article  CAS  PubMed  Google Scholar 

  80. Schwab LP, Peacock DL, Majumdar D, Ingels JF, Jensen LC, Smith KD, et al. Hypoxia-inducible factor 1α promotes primary tumor growth and tumor-initiating cell activity in breast cancer. Breast Cancer Res. 2012;14:R6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Kai AKL, Chan LK, Lo RCL, Lee JMF, Wong CCL, Wong JCM, et al. Down-regulation of TIMP2 by HIF-1α/miR-210/HIF-3α regulatory feedback circuit enhances cancer metastasis in hepatocellular carcinoma. Hepatology. 2016;64:473–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Ju JA, Godet I, Ye IC, Byun J, Jayatilaka H, Lee SJ, et al. Hypoxia selectively enhances Integrin α 5 β 1 receptor expression in breast cancer to promote metastasis. Mol Cancer Res. 2017;15:723–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kaidi A, Qualtrough D, Williams AC, Paraskeva C. Direct transcriptional up-regulation of cyclooxygenase-2 by hypoxia-inducible factor (HIF)-1 promotes colorectal tumor cell survival and enhances HIF-1 transcriptional activity during hypoxia. Cancer Res. 2006;66:6683–91.

    Article  CAS  PubMed  Google Scholar 

  84. Tsujii M, Kawano S, DuBois RN. Cyclooxygenase-2 expression in human colon cancer cells increases metastatic potential. Proc Natl Acad Sci U S A. 1997;94:3336–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Singh B, Berry JA, Shoher A, Ayers GD, Wei C, Lucci A. COX-2 involvement in breast cancer metastasis to bone. Oncogene. 2007;26:3789–96.

    Article  CAS  PubMed  Google Scholar 

  86. Murdoch C, Giannoudis A, Lewis CE. Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood. 2004;104:2224–34.

    Article  CAS  PubMed  Google Scholar 

  87. Cramer T, Yamanishi Y, Clausen BE, Förster I, Pawlinski R, Mackman N, et al. HIF-1α is essential for myeloid cell-mediated inflammation. Cell. 2003;112:645–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Chaturvedi P, Gilkes DM, Takano N, Semenza GL. Hypoxia-inducible factor-dependent signaling between triple-negative breast cancer cells and mesenchymal stem cells promotes macrophage recruitment. Proc Natl Acad Sci U S A. 2014;111:E2120–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Semba H, Takeda N, Isagawa T, Sugiura Y, Honda K, Wake M, et al. HIF-1α-PDK1 axis-induced active glycolysis plays an essential role in macrophage migratory capacity. Nat Commun. 2016;7:11635.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, DeNardo DG, et al. Macrophage expression of hypoxia-inducible factor-1α suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70:7465–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Tao L-L, Shi S-J, Chen L-B, Huang G-C. Expression of monocyte chemotactic protein-1/CCL2 in gastric cancer and its relationship with tumor hypoxia. World J Gastroenterol. 2014;20:4421–7.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Mojsilovic-Petrovic J, Callaghan D, Cui H, Dean C, Stanimirovic DB, Zhang W. Hypoxia-inducible factor-1 (HIF-1) is involved in the regulation of hypoxia-stimulated expression of monocyte chemoattractant protein-1 (MCP-1/CCL2) and MCP-5 (Ccl12) in astrocytes. J Neuroinflammation. 2007;4:12.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Snodgrass RG, Boß M, Zezina E, Weigert A, Dehne N, Fleming I, et al. Hypoxia potentiates palmitate-induced pro-inflammatory activation of primary human macrophages. J Biol Chem. 2016;291:413–24.

    Article  CAS  PubMed  Google Scholar 

  94. Almendros I, Wang Y, Becker L, Lennon FE, Zheng J, Coats BR, et al. Intermittent hypoxia-induced changes in tumor-associated macrophages and tumor malignancy in a mouse model of sleep apnea. Am J Respir Crit Care Med. 2014;189:593–601.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Tripathi C, Tewari BN, Kanchan RK, Baghel KS, Nautiyal N, Shrivastava R, et al. Macrophages are recruited to hypoxic tumor areas and acquire a pro-angiogenic M2-polarized phenotype via hypoxic cancer cell derived cytokines Oncostatin M and Eotaxin. Oncotarget. 2014;5:5350–68.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Ye LY, Chen W, Bai XL, Xu XY, Zhang Q, Xia XF, et al. Hypoxia-induced epithelial-to-mesenchymal transition in hepatocellular carcinoma induces an immunosuppressive tumor microenvironment to promote metastasis. Cancer Res. 2016;76:818–30.

    Article  CAS  PubMed  Google Scholar 

  97. Laoui D, Van Overmeire E, Di Conza G, Aldeni C, Keirsse J, Morias Y, et al. Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population. Cancer Res. 2014;74:24–30.

    Article  CAS  PubMed  Google Scholar 

  98. Campbell EL, Bruyninckx WJ, Kelly CJ, Glover LE, McNamee EN, Bowers BE, et al. Transmigrating neutrophils shape the mucosal microenvironment through localized oxygen depletion to influence resolution of inflammation. Immunity. 2014;40:66–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Egners A, Erdem M, Cramer T. The response of macrophages and neutrophils to hypoxia in the context of cancer and other inflammatory diseases. Mediat Inflamm. 2016;2016:2053646.

    Article  CAS  Google Scholar 

  100. D’Ignazio L, Bandarra D, Rocha S. NF-κB and HIF crosstalk in immune responses. FEBS J. 2016;283:413–24.

    Article  PubMed  CAS  Google Scholar 

  101. Tohme S, Yazdani HO, Al-Khafaji AB, Chidi AP, Loughran P, Mowen K, et al. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 2016;76:1367–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Chiu DKC, Xu IMJ, Lai RKH, Tse APW, Wei LL, Koh HY, et al. Hypoxia induces myeloid-derived suppressor cell recruitment to hepatocellular carcinoma through chemokine (C-C motif) ligand 26. Hepatology. 2016;64:797–813.

    Article  CAS  PubMed  Google Scholar 

  103. Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211:781–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Corzo CA, Condamine T, Lu L, Cotter MJ, Youn J-I, Cheng P, et al. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207:2439–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment : expect the unexpected. J Clin Investig. 2015;125:3356–64.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Chafe SC, Lou Y, Sceneay J, Vallejo M, Hamilton MJ, McDonald PC, et al. Carbonic anhydrase IX promotes myeloid-derived suppressor cell mobilization and establishment of a metastatic niche by stimulating G-CSF production. Cancer Res. 2015;75:996–1008.

    Article  CAS  PubMed  Google Scholar 

  107. Nakamura H, Makino Y, Okamoto K, Poellinger L, Ohnuma K, Morimoto C, et al. TCR engagement increases hypoxia-inducible factor-1 alpha protein synthesis via rapamycin-sensitive pathway under hypoxic conditions in human peripheral T cells. J Immunol. 2005;174:7592–9.

    Article  CAS  PubMed  Google Scholar 

  108. Chouaib S, Noman MZ, Kosmatopoulos K, Curran MA. Hypoxic stress: obstacles and opportunities for innovative immunotherapy of cancer. Oncogene. 2017;36:439–45.

    Article  CAS  PubMed  Google Scholar 

  109. Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, et al. Control of TH17/Treg balance by hypoxia-inducible factor 1. Cell. 2011;146:772–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Ikejiri A, Nagai S, Goda N, Kurebayashi Y, Osada-Oka M, Takubo K, et al. Dynamic regulation of Th17 differentiation by oxygen concentrations. Int Immunol. 2012;24:137–46.

    Article  CAS  PubMed  Google Scholar 

  111. Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR, et al. HIF1α-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of T H 17 and T reg cells. J Exp Med. 2011;208:1367–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Chaturvedi P, Gilkes DM, Wong CCL, Kshitiz, Luo W, Zhang H, et al. Hypoxia-inducible factor-dependent breast cancer-mesenchymal stem cell bidirectional signaling promotes metastasis. J Clin Invest. 2013;123:189–205.

    Article  CAS  PubMed  Google Scholar 

  113. Chen L, Tredget EE, Wu PYG, Wu Y, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One. 2008;3:e1886.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  114. Chen L, Xu Y, Zhao J, Zhang Z, Yang R, Xie J, et al. Conditioned medium from hypoxic bone marrow-derived mesenchymal stem cells enhances wound healing in mice. PLoS One. 2014;9:e96161.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  115. Ammirante M, Shalapour S, Kang Y, Jamieson CAM, Karin M. Tissue injury and hypoxia promote malignant progression of prostate cancer by inducing CXCL13 expression in tumor myofibroblasts. Proc Natl Acad Sci U S A. 2014;111:14776–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Hung S-P, Yang M-H, Tseng K-F, Lee OK. Hypoxia-induced secretion of TGF-β1 in Mesenchymal stem cell promotes breast cancer cell progression. Cell Transplant. 2013;22:1869–82.

    Article  PubMed  Google Scholar 

  117. De Francesco EM, Lappano R, Santolla MF, Marsico S, Caruso A, Maggiolini M. HIF-1α/GPER signaling mediates the expression of VEGF induced by hypoxia in breast cancer associated fibroblasts (CAFs). Breast Cancer Res. 2013;15:R64.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Giannoni E, Bianchini F, Calorini L, Chiarugi P. Cancer associated fibroblasts exploit reactive oxygen species through a Proinflammatory signature leading to epithelial Mesenchymal transition and Stemness. Antioxid Redox Signal. 2011;14:2361–71.

    Article  CAS  PubMed  Google Scholar 

  119. Jeong H-J, Hong S-H, Park R-K, Shin T, An N-H, Kim H-M. Hypoxia-induced IL-6 production is associated with activation of MAP kinase, HIF-1, and NF-kappaB on HEI-OC1 cells. Hear Res. 2005;207:59–67.

    Article  CAS  PubMed  Google Scholar 

  120. Tamm M, Bihl M, Eickelberg O, Stulz P, Perruchoud AP, Roth M. Hypoxia-induced interleukin-6 and interleukin-8 production is mediated by platelet-activating factor and platelet-derived growth factor in primary human lung cells. Am J Respir Cell Mol Biol. 1998;19:653–61.

    Article  CAS  PubMed  Google Scholar 

  121. Bao B, Ahmad A, Kong D, Ali S, Azmi AS, Li Y, et al. Hypoxia induced aggressiveness of prostate cancer cells is linked with deregulated expression of VEGF, IL-6 and miRNAs that are attenuated by CDF. PLoS One. 2012;7:e43726.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Xu L, Xie K, Mukaida N, Matsushima K, Fidler IJ. Hypoxia-induced elevation in interleukin-8 expression by human ovarian carcinoma cells. Cancer Res. 1999;59:5822–9.

    CAS  PubMed  Google Scholar 

  123. Singh JK, Simões BM, Howell SJ, Farnie G, Clarke RB. Recent advances reveal IL-8 signaling as a potential key to targeting breast cancer stem cells. Breast Cancer Res. 2013;15:210.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. Falanga V, Su Wen Qian V, Danielpour D, Katz MH, Roberts AB, Sporn MB. Hypoxia Upregulates the synthesis of TGF-β1 by human dermal fibroblasts. J Invest Dermatol. 1991;97:634–7.

    Article  CAS  PubMed  Google Scholar 

  125. Klempt ND, Sirimanne E, Gunn AJ, Klempt M, Singh K, Williams C, et al. Hypoxia-ischemia induces transforming growth factor beta 1 mRNA in the infant rat brain. Brain Res Mol Brain Res. 1992;13:93–101.

    Article  CAS  PubMed  Google Scholar 

  126. Liu F, Ai F, Tian L, Liu S, Zhao L, Wang X. Infliximab enhances the therapeutic effects of 5-fluorouracil resulting in tumor regression in colon cancer. Onco Targets Ther. 2016;9:5999–6008.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Guida E, Stewart A. Influence of hypoxia and glucose deprivation on tumour necrosis factor-alpha and granulocyte-macrophage colony-stimulating factor expression in human cultured monocytes. Cell Physiol Biochem. 1998;8:75–88.

    Article  CAS  PubMed  Google Scholar 

  128. Yoshida S, Ono M, Shono T, Izumi H, Ishibashi T, Suzuki H, et al. Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis. Mol Cell Biol. 1997;17:4015–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Ghosh S, Paul A, Sen E. Tumor necrosis factor alpha-induced hypoxia-inducible factor 1α-β-catenin Axis regulates major Histocompatibility complex class I gene activation through chromatin remodeling. Mol Cell Biol. 2013;33:2718–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Olenyuk BZ, Zhang G-J, Klco JM, Nickols NG, Kaelin WG, Dervan PB. Inhibition of vascular endothelial growth factor with a sequence-specific hypoxia response element antagonist. Proc Natl Acad Sci U S A. 2004;101:16768–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Kondo S, Kubota S, Shimo T, Nishida T, Yosimichi G, Eguchi T, et al. Connective tissue growth factor increased by hypoxia may initiate angiogenesis in collaboration with matrix metalloproteinases. Carcinogenesis. 2002;23:769–76.

    Article  CAS  PubMed  Google Scholar 

  132. Shimo T, Kubota S, Kondo S, Nakanishi T, Sasaki A, Mese H, et al. Connective tissue growth factor as a major angiogenic agent that is induced by hypoxia in a human breast cancer cell line. Cancer Lett. 2001;174:57–64.

    Article  CAS  PubMed  Google Scholar 

  133. Eguchi D, Ikenaga N, Ohuchida K, Kozono S, Cui L, Fujiwara K, et al. Hypoxia enhances the interaction between pancreatic stellate cells and cancer cells via increased secretion of connective tissue growth factor. J Surg Res. 2013;181:225–33.

    Article  CAS  PubMed  Google Scholar 

  134. Lu H, Kojima K, Battula VL, Korchin B, Shi Y, Chen Y, et al. Targeting connective tissue growth factor (CTGF) in acute lymphoblastic leukemia preclinical models: anti-CTGF monoclonal antibody attenuates leukemia growth. Ann Hematol. 2014;93:485–92.

    Article  CAS  PubMed  Google Scholar 

  135. Kondo S, Kubota S, Mukudai Y, Moritani N, Nishida T, Matsushita H, et al. Hypoxic regulation of stability of connective tissue growth factor/CCN2 mRNA by 3′-untranslated region interacting with a cellular protein in human chondrosarcoma cells. Oncogene. 2006;25:1099–110.

    Article  CAS  PubMed  Google Scholar 

  136. Kundu N, Fulton AM. Selective cyclooxygenase (COX)-1 or COX-2 inhibitors control metastatic disease in a murine model of breast cancer. Cancer Res. 2002;62:2343–6.

    CAS  PubMed  Google Scholar 

  137. Wu G, Mannam AP, Wu J, Kirbis S, Shie J-L, Chen C, et al. Hypoxia induces myocyte-dependent COX-2 regulation in endothelial cells: role of VEGF. Am J Physiol Heart Circ Physiol. 2003;285:H2420–9.

    Article  CAS  PubMed  Google Scholar 

  138. Condamine T, Ramachandran I, Youn J-I, Gabrilovich DI. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu Rev Med. 2015;66:97–110.

    Article  CAS  PubMed  Google Scholar 

  139. Hu Y, Liu J, Huang H. Recent agents targeting HIF-1α for cancer therapy. J Cell Biochem. 2013;114:498–509.

    Article  CAS  PubMed  Google Scholar 

  140. Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3:721–32.

    Article  CAS  PubMed  Google Scholar 

  141. Ban HS, Uto Y, Won M, Nakamura H. Hypoxia-inducible factor (HIF) inhibitors: a patent survey (2011-2015). Expert Opin Ther Pat. 2016;26:309–22.

    Article  CAS  PubMed  Google Scholar 

  142. Carroll CE, Liang Y, Benakanakere I, Besch-Williford C, Hyder SM. The anticancer agent YC-1 suppresses progestin-stimulated VEGF in breast cancer cells and arrests breast tumor development. Int J Oncol. 2013;42:179–87.

    Article  CAS  PubMed  Google Scholar 

  143. Wakiyama K, Kitajima Y, Tanaka T, Kaneki M, Yanagihara K, Aishima S, et al. Low-dose YC-1 combined with glucose and insulin selectively induces apoptosis in hypoxic gastric carcinoma cells by inhibiting anaerobic glycolysis. Sci Rep. 2017;7:12653.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Huang YT, Pan SL, Guh JH, Chang YL, Lee FY, Kuo SC, et al. YC-1 suppresses constitutive nuclear factor-kappaB activation and induces apoptosis in human prostate cancer cells. Mol Cancer Ther. 2005;4:1628–35.

    Article  CAS  PubMed  Google Scholar 

  145. Hutt DM, Roth DM, Vignaud H, Cullin C, Bouchecareilh M. The histone deacetylase inhibitor, vorinostat, represses hypoxia inducible factor 1 alpha expression through translational inhibition. PLoS One. 2014;9:e106224.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist. 2007;12:1247–52.

    Article  CAS  PubMed  Google Scholar 

  147. Jhaveri K, Modi S. Ganetespib: research and clinical development. Onco Targets Ther. 2015;8:1849–58.

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Lee K, Kim HM. A novel approach to cancer therapy using PX-478 as a HIF-1α inhibitor. Arch Pharm Res. 2011;34:1583–5.

    Article  CAS  PubMed  Google Scholar 

  149. Zhao T, Ren H, Jia L, Chen J, Xin W, Yan F, et al. Inhibition of HIF-1α by PX-478 enhances the anti-tumor effect of gemcitabine by inducing immunogenic cell death in pancreatic ductal adenocarcinoma. Oncotarget. 2015;6:2250–62.

    PubMed  Google Scholar 

  150. Zhu Y, Zang Y, Zhao F, Li Z, Zhang J, Fang L, et al. Inhibition of HIF-1α by PX-478 suppresses tumor growth of esophageal squamous cell cancer in vitro and in vivo. Am J Cancer Res. 2017;7:1198–212.

    PubMed  PubMed Central  Google Scholar 

  151. Greenberger LM, Horak ID, Filpula D, Sapra P, Westergaard M, Frydenlund HF, et al. A RNA antagonist of hypoxia-inducible factor-1alpha, EZN-2968, inhibits tumor cell growth. Mol Cancer Ther. 2008;7:3598–608.

    Article  CAS  PubMed  Google Scholar 

  152. Jeong W, Rapisarda A, Park SR, Kinders RJ, Chen A, Melillo G, et al. Pilot trial of EZN-2968, an antisense oligonucleotide inhibitor of hypoxia-inducible factor-1 alpha (HIF-1α), in patients with refractory solid tumors. Cancer Chemother Pharmacol. 2014;73:343–8.

    Article  CAS  PubMed  Google Scholar 

  153. Wigerup C, Påhlman S, Bexell D. Therapeutic targeting of hypoxia and hypoxia-inducible factors in cancer. Pharmacol Ther. 2016;164:152–69.

    Article  CAS  PubMed  Google Scholar 

  154. Chen W, Hill H, Christie A, Kim MS, Holloman E, Pavia-Jimenez A, et al. Targeting renal cell carcinoma with a HIF-2 antagonist. Nature. 2016;539:112–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Neesse A, Frese KK, Bapiro TE, Nakagawa T, Sternlicht MD, Seeley TW, et al. CTGF antagonism with mAb FG-3019 enhances chemotherapy response without increasing drug delivery in murine ductal pancreas cancer. Proc Natl Acad Sci U S A. 2013;110:12325–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Picozzi VJ, Pipas JM, Koong A, Giaccia A, Bahary N, Krishnamurthi SS, et al. FG-3019, a human monoclonal antibody to connective tissue growth factor (CTGF), with gemcitabine/erlotinib (G/E) in patients with locally advanced or metastatic pancreatic ductal adenocarcinoma (PDAC). J Clin Oncol. 2014;32:8–11.

    Google Scholar 

  157. Zheng Y, Basel D, Chow SO, Fong-Yee C, Kim S, Buttgereit F, et al. Targeting IL-6 and RANKL signaling inhibits prostate cancer growth in bone. Clin Exp Metastasis. 2014;31:921–33.

    Article  CAS  PubMed  Google Scholar 

  158. Zheng Y, Chow SO, Boernert K, Basel D, Mikuscheva A, Kim S, et al. Direct crosstalk between cancer and osteoblast lineage cells fuels metastatic growth in bone via auto-amplification of IL-6 and RANKL signaling pathways. J Bone Miner Res. 2014;29:1938–49.

    Article  CAS  PubMed  Google Scholar 

  159. Deisseroth A, Ko CW, Nie L, Zirkelbach JF, Zhao L, Bullock J, et al. FDA approval: Siltuximab for the treatment of patients with multicentric castleman disease. Clin Cancer Res. 2015;21:950–4.

    Article  CAS  PubMed  Google Scholar 

  160. Voorhees PM, Manges RF, Sonneveld P, Jagannath S, Somlo G, Krishnan A, et al. A phase 2 multicentre study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with relapsed or refractory multiple myeloma. Br J Haematol. 2013;161:357–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Miguel JS, Weisel K, Moreau P, Lacy M, Song K, Delforge M, et al. Pomalidomide plus low-dose dexamethasone versus high-dose dexamethasone alone for patients with relapsed and refractory multiple myeloma (MM-003): a randomised, open-label, phase 3 trial. Lancet Oncol. 2013;14:1055–66.

    Article  CAS  PubMed  Google Scholar 

  162. Hoy SM. Pomalidomide: a review in relapsed and refractory multiple myeloma. Drugs. 2017;77:1897–908.

    Article  CAS  PubMed  Google Scholar 

  163. D’Amato RJ, Lentzsch S, Anderson KC, Rogers MS. Mechanism of action of thalidomide and 3-aminothalidomide in multiple myeloma. Semin Oncol. 2001;28:597–601.

    Article  PubMed  CAS  Google Scholar 

  164. Baran N, Konopleva M. Molecular pathways: hypoxia-activated prodrugs in cancer therapy. Clin Cancer Res. 2017;23:2382–90.

    Article  CAS  PubMed  Google Scholar 

  165. Hoang NTH, Kadonosono T, Kuchimaru T, Kizaka-Kondoh S. Hypoxia-inducible factor-targeting prodrug TOP3 combined with gemcitabine or TS-1 improves pancreatic cancer survival in an orthotopic model. Cancer Sci. 2016;107:1151–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Nesbitt H, Byrne NM, Williams N, Ming L, Worthington J, Errington RJ, et al. Targeting hypoxic prostate tumours using the novel hypoxia-activated prodrug OCT1002 inhibits expression of genes associated with malignant progression. Clin Cancer Res. 2017;23:1797–808.

    Article  CAS  PubMed  Google Scholar 

  167. Nesbitt H, Worthington J, Errington RJ, Patterson LH, Smith PJ, McKeown SR, et al. The unidirectional hypoxia-activated prodrug OCT1002 inhibits growth and vascular development in castrate-resistant prostate tumors. Prostate. 2017:1539–47.

  168. Pan SL, Guh JH, Peng CY, Chang YL, Cheng FC, Chang JH, et al. A potential role of YC-I on the inhibition of cytokine release in peripheral blood mononuclear leukocytes and endotoxemic mouse models. Thromb Haemost. 2005;93:940–8.

    CAS  PubMed  Google Scholar 

  169. Palmieri D, Lockman PR, Thomas FC, Hua E, Herring J, Hargrave E, et al. Vorinostat inhibits brain metastatic colonization in a model of triple-negative breast cancer and induces DNA double-strand breaks. Clin Cancer Res. 2009;15:6148–57.

    Article  CAS  PubMed  Google Scholar 

  170. Munster PN, Thurn KT, Thomas S, Raha P, Lacevic M, Miller A, et al. A phase II study of the histone deacetylase inhibitor vorinostat combined with tamoxifen for the treatment of patients with hormone therapy-resistant breast cancer. Br J Cancer. 2011;104:1828–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Leoni F, Zaliani A, Bertolini G, Porro G, Pagani P, Pozzi P, et al. The antitumor histone deacetylase inhibitor suberoylanilide hydroxamic acid exhibits antiinflammatory properties via suppression of cytokines. Proc Natl Acad Sci U S A. 2002;99:2995–3000.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Choi Y, Park S-K, Kim HM, Kang JS, Yoon YD, Han SB, et al. Histone deacetylase inhibitor KBH-A42 inhibits cytokine production in RAW 264.7 macrophage cells and in vivo endotoxemia model. Exp. Mol. Med. 2008;40:574–81.

    CAS  Google Scholar 

  173. Leoni F, Fossati G, Lewis EC, Lee J-K, Porro G, Pagani P, et al. The Histone Deacetylase inhibitor ITF2357 reduces production of pro-inflammatory cytokines in vitro and systemic inflammation in vivo. Mol Med. 2005;11:1–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Tamura Y, Torigoe T, Kutomi G, Hirata K, Sato N. New paradigm for intrinsic function of heat shock proteins as endogenous ligands in inflammation and innate immunity. Curr Mol Med. 2012;12:1198–206.

    Article  CAS  PubMed  Google Scholar 

  175. Xiang L, Gilkes DM, Chaturvedi P, Luo W, Hu H, Takano N, et al. Ganetespib blocks HIF-1 activity and inhibits tumor growth, vascularization, stem cell maintenance, invasion, and metastasis in orthotopic mouse models of triple-negative breast cancer. J Mol Med. 2014;92:151–64.

    Article  CAS  PubMed  Google Scholar 

  176. Lilja A, Weeden CE, McArthur K, Nguyen T, Donald A, Wong ZX, et al. HSP90 inhibition suppresses lipopolysaccharide-induced lung inflammation in vivo. PLoS One. 2015;10:e0114975.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  177. Rice JW, Veal JM, Fadden RP, Barabasz AF, Partridge JM, Barta TE, et al. Small molecule inhibitors of Hsp90 potently affect inflammatory disease pathways and exhibit activity in models of rheumatoid arthritis. Arthritis Rheum. 2008;58:3765–75.

    Article  CAS  PubMed  Google Scholar 

  178. Sun K, Halberg N, Khan M, Magalang UJ, Scherer PE. Selective inhibition of hypoxia-inducible factor 1α ameliorates adipose tissue dysfunction. Mol Cell Biol. 2013;33:904–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Fell SM, Lu W, Kung AL, Schlisio S. The 2-oxoglutarate analog 3-oxoglutarate decreases normoxic hypoxia-inducible factor-1 α in cancer cells, induces cell death, and reduces tumor xenograft growth. Hypoxia. 2016;4:15–27.

    PubMed  PubMed Central  Google Scholar 

  180. Wang L, Chen G, Lu X, Wang S, Han S, Li Y, et al. Novel chalcone derivatives as hypoxia-inducible factor (HIF)-1 inhibitor: synthesis, anti-invasive and anti-angiogenic properties. Eur J Med Chem. 2015;89:88–97.

    Article  CAS  PubMed  Google Scholar 

  181. Wallace EM, Rizzi JP, Han G, Wehn PM, Cao Z, Du X, et al. A small-molecule antagonist of HIF2α is efficacious in preclinical models of renal cell carcinoma. Cancer Res. 2016;76:5491–500.

    Article  CAS  PubMed  Google Scholar 

  182. Xu J, Zheng L, Chen J, Sun Y, Lin H, Jin R, et al. Increasing AR by HIF-2α inhibitor (PT-2385) overcomes the side-effects of sorafenib by suppressing hepatocellular carcinoma invasion via alteration of pSTAT3, pAKT and pERK signals. Cell Death Dis. 2017;8:e3095.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Bhola NE, Balko JM, Dugger TC, Kuba MG, Sánchez V, Sanders M, et al. TGF-β inhibition enhances chemotherapy action against triple-negative breast cancer. J Clin Invest. 2013;123:1348–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Wan X, Li Z-G, Yingling JM, Yang J, Starbuck MW, Ravoori MK, et al. Effect of transforming growth factor beta (TGF-β) receptor I kinase inhibitor on prostate cancer bone growth. Bone. 2012;50:695–703.

    Article  CAS  PubMed  Google Scholar 

  185. Finger EC, Cheng C-F, Williams TR, Rankin EB, Bedogni B, Tachiki L, et al. CTGF is a therapeutic target for metastatic melanoma. Oncogene. 2014;33:1093–100.

    Article  CAS  PubMed  Google Scholar 

  186. Luo X, Fu Y, Loza AJ, Murali B, Leahy KM, Ruhland MK, et al. Stromal-initiated changes in the bone promote metastatic niche development. Cell Rep. 2016;14:82–92.

    Article  CAS  PubMed  Google Scholar 

  187. Guise CP, Mowday AM, Ashoorzadeh A, Yuan R, Lin WH, Wu DH, et al. Bioreductive prodrugs as cancer therapeutics: targeting tumor hypoxia. Chin J Cancer. 2014;33:80–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Yeh JJ, Kim WY. Targeting tumor hypoxia with hypoxia-activated prodrugs. J Clin Oncol. 2015;33:1505–8.

    Article  CAS  PubMed  Google Scholar 

  189. Saggar JK, Tannock IF. Activity of the hypoxia-activated pro-drug TH-302 in hypoxic and perivascular regions of solid tumors and its potential to enhance therapeutic effects of chemotherapy. Int J Cancer. 2014;134:2726–34.

    Article  CAS  PubMed  Google Scholar 

  190. Lohse I, Rasowski J, Cao P, Pintilie M, Do T, Tsao MS, et al. Targeting hypoxic microenvironment of pancreatic xenografts with the hypoxia-activated prodrug TH-302. Oncotarget. 2016;7:33571–80.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Liapis V, Zysk A, DeNichilo M, Zinonos I, Hay S, Panagopoulos V, et al. Anticancer efficacy of the hypoxia-activated prodrug evofosfamide is enhanced in combination with proapoptotic receptor agonists against osteosarcoma. Cancer Med. 2017;6:2164–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Zhang X, Wojtkowiak JW, Martinez GV, Cornnell HH, Hart CP, Baker AF, et al. MR imaging biomarkers to monitor early response to hypoxia-activated prodrug TH-302 in pancreatic cancer xenografts. PLoS One. 2016;11:e0155289.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  193. Hunter FW, Wouters BG, Wilson WR. Hypoxia-activated prodrugs: paths forward in the era of personalised medicine. Br J Cancer. 2016;114:1071–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Ganjoo KN, Cranmer LD, Butrynski JE, Rushing D, Adkins D, Okuno SH, et al. A phase i study of the safety and pharmacokinetics of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. Oncology. 2011;80:50–6.

    Article  CAS  PubMed  Google Scholar 

  195. Chawla SP, Cranmer LD, Van Tine BA, Reed DR, Okuno SH, Butrynski JE, et al. Phase II study of the safety and antitumor activity of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. J Clin Oncol. 2014;32:3299–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Zhang R, Song X, Liang C, Yi X, Song G, Chao Y, et al. Catalase-loaded cisplatin-prodrug-constructed liposomes to overcome tumor hypoxia for enhanced chemo-radiotherapy of cancer. Biomaterials. 2017;138:13–21.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We acknowledge the researchers who work in the field of hypoxia, inflammation, and metastasis, and apologize for any work that was not cited due to space constraints.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniele M. Gilkes.

Ethics declarations

Funding

Work in the author’s lab is supported by the NCI (R00-CA181352 and U54-CA210173), Susan G. Komen Foundation (CCR17483484), and pilot funds from The Breast Cancer Research Foundation and The JKTG foundation. D.M. Gilkes is a 2016 V Scholar awardee (V2016–024).

Conflict of Interest

The authors declare no conflict of interest.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

DiGiacomo, J.W., Gilkes, D.M. Tumor Hypoxia As an Enhancer of Inflammation-Mediated Metastasis: Emerging Therapeutic Strategies. Targ Oncol 13, 157–173 (2018). https://doi.org/10.1007/s11523-018-0555-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11523-018-0555-4

Navigation