Skip to main content

Therapeutic Strategies to Block the Hypoxic Response

  • Chapter
  • First Online:
Hypoxia and Cancer Metastasis

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 1136))

Abstract

Patients with the low levels of O2 (hypoxia) in their primary tumors have a higher risk for metastasis and death, indicating a need to therapeutically inhibit the effectors of hypoxia. Many strategies have been developed and investigated to block the hypoxic response. For example, inhibitors of HIF-1 and HIF-2 function by altering the transcription, translation, dimerization, nuclear translocation, DNA-binding, or ubiquitination of the HIF proteins. Hypoxia-activated prodrugs inhibit the hypoxic response through hypoxia-mediated reduction of an inactive, or minimally active, chemical to a cytotoxic agent. Most hypoxia-activated prodrugs function by inducing DNA damage, but others with more novel functions, including prodrugs that release EGFR/HER2 inhibitors also exist. Despite the existence of many therapeutics to combat the hypoxic response, there has been very little success in late phase clinical trials, potentially due to a lack of biomarkers that can be used to stratify patients who would benefit from a hypoxia-targeted therapy.

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

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 109.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 139.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 139.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Hockel M et al (1996) Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. Cancer Res 56(19):4509–4515

    CAS  PubMed  Google Scholar 

  2. Hockel M et al (1993) Intratumoral pO2 predicts survival in advanced cancer of the uterine cervix. Radiother Oncol 26(1):45–50

    Article  CAS  PubMed  Google Scholar 

  3. Semenza GL (2003) Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3(10):721–732

    Article  CAS  PubMed  Google Scholar 

  4. Hu Y, Liu J, Huang H (2013) Recent agents targeting HIF-1alpha for cancer therapy. J Cell Biochem 114(3):498–509

    Article  CAS  PubMed  Google Scholar 

  5. Stiehl DP et al (2002) Normoxic induction of the hypoxia-inducible factor 1alpha by insulin and interleukin-1beta involves the phosphatidylinositol 3-kinase pathway. FEBS Lett 512(1–3):157–162

    Article  CAS  PubMed  Google Scholar 

  6. Hudson CC et al (2002) Regulation of hypoxia-inducible factor 1alpha expression and function by the mammalian target of rapamycin. Mol Cell Biol 22(20):7004–7014

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. DiGiacomo JW, Gilkes DM (2018) Tumor hypoxia as an enhancer of inflammation-mediated metastasis: emerging therapeutic strategies. Target Oncol 13(2):157–173

    Article  PubMed  Google Scholar 

  8. Ban HS et al (2016) Hypoxia-inducible factor (HIF) inhibitors: a patent survey (2011–2015). Expert Opin Ther Pat 26(3):309–322

    Article  CAS  PubMed  Google Scholar 

  9. Li SH et al (2008) A novel mode of action of YC-1 in HIF inhibition: stimulation of FIH-dependent p300 dissociation from HIF-1{alpha}. Mol Cancer Ther 7(12):3729–3738

    Article  CAS  PubMed  Google Scholar 

  10. Carroll CE et al (2013) The anticancer agent YC-1 suppresses progestin-stimulated VEGF in breast cancer cells and arrests breast tumor development. Int J Oncol 42(1):179–187

    Article  CAS  PubMed  Google Scholar 

  11. Shin DH et al (2007) Preclinical evaluation of YC-1, a HIF inhibitor, for the prevention of tumor spreading. Cancer Lett 255(1):107–116

    Article  CAS  PubMed  Google Scholar 

  12. Harada H et al (2009) Treatment regimen determines whether an HIF-1 inhibitor enhances or inhibits the effect of radiation therapy. Br J Cancer 100(5):747–757

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wakiyama K et al (2017) Low-dose YC-1 combined with glucose and insulin selectively induces apoptosis in hypoxic gastric carcinoma cells by inhibiting anaerobic glycolysis. Sci Rep 7(1):12653

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Kung AL et al (2004) Small molecule blockade of transcriptional coactivation of the hypoxia-inducible factor pathway. Cancer Cell 6(1):33–43

    Article  CAS  PubMed  Google Scholar 

  15. Staab A et al (2007) Effects of HIF-1 inhibition by chetomin on hypoxia-related transcription and radiosensitivity in HT 1080 human fibrosarcoma cells. BMC Cancer 7:213

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Kessler J et al (2010) HIF-1alpha inhibition by siRNA or chetomin in human malignant glioma cells: effects on hypoxic radioresistance and monitoring via CA9 expression. BMC Cancer 10:605

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kong D et al (2005) Echinomycin, a small-molecule inhibitor of hypoxia-inducible factor-1 DNA-binding activity. Cancer Res 65(19):9047–9055

    Article  CAS  PubMed  Google Scholar 

  18. Vlaminck B et al (2007) Dual effect of echinomycin on hypoxia-inducible factor-1 activity under normoxic and hypoxic conditions. FEBS J 274(21):5533–5542

    Article  CAS  PubMed  Google Scholar 

  19. Park JY et al (2004) Echinomycin and a novel analogue induce apoptosis of HT-29 cells via the activation of MAP kinases pathway. Pharmacol Res 50(2):201–207

    Article  CAS  PubMed  Google Scholar 

  20. Marshall ME et al (1993) Phase II trial of echinomycin for the treatment of advanced renal cell carcinoma. A Southwest Oncology Group study. Investig New Drugs 11(2–3):207–209

    Article  CAS  Google Scholar 

  21. Chang AY et al (1998) A randomized phase II trial of echinomycin, trimetrexate, and cisplatin plus etoposide in patients with metastatic nonsmall cell lung carcinoma: an Eastern Cooperative Oncology Group Study (E1587). Cancer 82(2):292–300

    Article  CAS  PubMed  Google Scholar 

  22. Gradishar WJ et al (1995) A phase II clinical trial of echinomycin in metastatic soft tissue sarcoma. An illinois Cancer Center Study. Investig New Drugs 13(2):171–174

    Article  CAS  Google Scholar 

  23. Lee K et al (2009) Acriflavine inhibits HIF-1 dimerization, tumor growth, and vascularization. Proc Natl Acad Sci U S A 106(42):17910–17915

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wong CC et al (2012) Inhibitors of hypoxia-inducible factor 1 block breast cancer metastatic niche formation and lung metastasis. J Mol Med (Berl) 90(7):803–815

    Article  CAS  Google Scholar 

  25. Chaturvedi P et al (2013) Hypoxia-inducible factor-dependent breast cancer-mesenchymal stem cell bidirectional signaling promotes metastasis. J Clin Invest 123(1):189–205

    CAS  PubMed  Google Scholar 

  26. Koh MY et al (2008) Molecular mechanisms for the activity of PX-478, an antitumor inhibitor of the hypoxia-inducible factor-1alpha. Mol Cancer Ther 7(1):90–100

    Article  CAS  PubMed  Google Scholar 

  27. Palayoor ST et al (2008) PX-478, an inhibitor of hypoxia-inducible factor-1alpha, enhances radiosensitivity of prostate carcinoma cells. Int J Cancer 123(10):2430–2437

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhao T et al (2015) Inhibition of HIF-1alpha by PX-478 enhances the anti-tumor effect of gemcitabine by inducing immunogenic cell death in pancreatic ductal adenocarcinoma. Oncotarget 6(4):2250–2262

    Article  PubMed  Google Scholar 

  29. Zhu Y et al (2017) Inhibition of HIF-1alpha by PX-478 suppresses tumor growth of esophageal squamous cell cancer in vitro and in vivo. Am J Cancer Res 7(5):1198–1212

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Welsh S et al (2004) Antitumor activity and pharmacodynamic properties of PX-478, an inhibitor of hypoxia-inducible factor-1alpha. Mol Cancer Ther 3(3):233–244

    CAS  PubMed  Google Scholar 

  31. Tibes R et al (2010) Results from a phase I, dose-esclation study of PX-478, an orally available inhibitor of HIF-1alpha. J Clin Oncol 28(15(Suppl.)):3076

    Article  Google Scholar 

  32. Greenberger LM et al (2008) A RNA antagonist of hypoxia-inducible factor-1alpha, EZN-2968, inhibits tumor cell growth. Mol Cancer Ther 7(11):3598–3608

    Article  CAS  PubMed  Google Scholar 

  33. Jeong W et al (2014) Pilot trial of EZN-2968, an antisense oligonucleotide inhibitor of hypoxia-inducible factor-1 alpha (HIF-1alpha), in patients with refractory solid tumors. Cancer Chemother Pharmacol 73(2):343–348

    Article  CAS  PubMed  Google Scholar 

  34. Zhang H et al (2008) Digoxin and other cardiac glycosides inhibit HIF-1alpha synthesis and block tumor growth. Proc Natl Acad Sci U S A 105(50):19579–19586

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Zhang H et al (2012) HIF-1-dependent expression of angiopoietin-like 4 and L1CAM mediates vascular metastasis of hypoxic breast cancer cells to the lungs. Oncogene 31(14):1757–1770

    Article  CAS  PubMed  Google Scholar 

  36. Jhaveri K, Modi S (2015) Ganetespib: research and clinical development. Onco Targets Ther 8:1849–1858

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Isaacs JS et al (2002) Hsp90 regulates a von Hippel Lindau-independent hypoxia-inducible factor-1 alpha-degradative pathway. J Biol Chem 277(33):29936–29944

    Article  CAS  PubMed  Google Scholar 

  38. Xiang L et al (2014) 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 (Berl) 92(2):151–164

    Article  CAS  Google Scholar 

  39. Nagaraju GP et al (2013) Antiangiogenic effects of ganetespib in colorectal cancer mediated through inhibition of HIF-1alpha and STAT-3. Angiogenesis 16(4):903–917

    Article  PubMed  Google Scholar 

  40. Thakur MK et al (2016) A phase II trial of ganetespib, a heat shock protein 90 (Hsp90) inhibitor, in patients with docetaxel-pretreated metastatic castrate-resistant prostate cancer (CRPC)-a prostate cancer clinical trials consortium (PCCTC) study. Investig New Drugs 34(1):112–118

    Article  CAS  Google Scholar 

  41. Jhaveri K et al (2014) A phase II open-label study of ganetespib, a novel heat shock protein 90 inhibitor for patients with metastatic breast cancer. Clin Breast Cancer 14(3):154–160

    Article  CAS  PubMed  Google Scholar 

  42. Mabjeesh NJ et al (2002) Geldanamycin induces degradation of hypoxia-inducible factor 1alpha protein via the proteosome pathway in prostate cancer cells. Cancer Res 62(9):2478–2482

    CAS  PubMed  Google Scholar 

  43. Alqawi O, Moghaddas M, Singh G (2006) Effects of geldanamycin on HIF-1alpha mediated angiogenesis and invasion in prostate cancer cells. Prostate Cancer Prostatic Dis 9(2):126–135

    Article  CAS  PubMed  Google Scholar 

  44. Koga F, Tsutsumi S, Neckers LM (2007) Low dose geldanamycin inhibits hepatocyte growth factor and hypoxia-stimulated invasion of cancer cells. Cell Cycle 6(11):1393–1402

    Article  CAS  PubMed  Google Scholar 

  45. Welsh SJ et al (2002) 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 62(17):5089–5095

    CAS  PubMed  Google Scholar 

  46. Kim YH et al (2011) Antitumor agent PX-12 inhibits HIF-1alpha protein levels through an Nrf2/PMF-1-mediated increase in spermidine/spermine acetyl transferase. Cancer Chemother Pharmacol 68(2):405–413

    Article  CAS  PubMed  Google Scholar 

  47. Welsh SJ et al (2003) The thioredoxin redox inhibitors 1-methylpropyl 2-imidazolyl disulfide and pleurotin inhibit hypoxia-induced factor 1alpha and vascular endothelial growth factor formation. Mol Cancer Ther 2(3):235–243

    CAS  PubMed  Google Scholar 

  48. Baker AF et al (2006) The antitumor thioredoxin-1 inhibitor PX-12 (1-methylpropyl 2-imidazolyl disulfide) decreases thioredoxin-1 and VEGF levels in cancer patient plasma. J Lab Clin Med 147(2):83–90

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ramanathan RK et al (2011) A randomized phase II study of PX-12, an inhibitor of thioredoxin in patients with advanced cancer of the pancreas following progression after a gemcitabine-containing combination. Cancer Chemother Pharmacol 67(3):503–509

    Article  CAS  PubMed  Google Scholar 

  50. Baker AF et al (2013) A phase IB trial of 24-hour intravenous PX-12, a thioredoxin-1 inhibitor, in patients with advanced gastrointestinal cancers. Investig New Drugs 31(3):631–641

    Article  CAS  Google Scholar 

  51. Befani CD et al (2012) Bortezomib represses HIF-1alpha protein expression and nuclear accumulation by inhibiting both PI3K/Akt/TOR and MAPK pathways in prostate cancer cells. J Mol Med (Berl) 90(1):45–54

    Article  CAS  Google Scholar 

  52. Kane RC et al (2003) Velcade: U.S. FDA approval for the treatment of multiple myeloma progressing on prior therapy. Oncologist 8(6):508–513

    Article  PubMed  Google Scholar 

  53. Kaluz S, Kaluzova M, Stanbridge EJ (2006) Proteasomal inhibition attenuates transcriptional activity of hypoxia-inducible factor 1 (HIF-1) via specific effect on the HIF-1alpha C-terminal activation domain. Mol Cell Biol 26(15):5895–5907

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Zhu K et al (2009) Control of HIF-1alpha expression by eIF2 alpha phosphorylation-mediated translational repression. Cancer Res 69(5):1836–1843

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Abd-Aziz N, Stanbridge EJ, Shafee N (2015) Bortezomib attenuates HIF-1- but not HIF-2-mediated transcriptional activation. Oncol Lett 10(4):2192–2196

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Veschini L et al (2007) Hypoxia-inducible transcription factor-1 alpha determines sensitivity of endothelial cells to the proteosome inhibitor bortezomib. Blood 109(6):2565–2570

    Article  CAS  PubMed  Google Scholar 

  57. Fels DR et al (2008) Preferential cytotoxicity of bortezomib toward hypoxic tumor cells via overactivation of endoplasmic reticulum stress pathways. Cancer Res 68(22):9323–9330

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Rapisarda A et al (2004) Topoisomerase I-mediated inhibition of hypoxia-inducible factor 1: mechanism and therapeutic implications. Cancer Res 64(4):1475–1482

    Article  CAS  PubMed  Google Scholar 

  59. Puppo M et al (2008) Topotecan inhibits vascular endothelial growth factor production and angiogenic activity induced by hypoxia in human neuroblastoma by targeting hypoxia-inducible factor-1alpha and -2alpha. Mol Cancer Ther 7(7):1974–1984

    Article  CAS  PubMed  Google Scholar 

  60. Calvani M et al (2006) Hypoxic induction of an HIF-1alpha-dependent bFGF autocrine loop drives angiogenesis in human endothelial cells. Blood 107(7):2705–2712

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Rapisarda A et al (2004) Schedule-dependent inhibition of hypoxia-inducible factor-1alpha protein accumulation, angiogenesis, and tumor growth by topotecan in U251-HRE glioblastoma xenografts. Cancer Res 64(19):6845–6848

    Article  CAS  PubMed  Google Scholar 

  62. Kummar S et al (2011) Multihistology, target-driven pilot trial of oral topotecan as an inhibitor of hypoxia-inducible factor-1alpha in advanced solid tumors. Clin Cancer Res 17(15):5123–5131

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Hutt DM et al (2014) The histone deacetylase inhibitor, Vorinostat, represses hypoxia inducible factor 1 alpha expression through translational inhibition. PLoS One 9(8):e106224

    Article  PubMed  PubMed Central  Google Scholar 

  64. Zhang C et al (2017) Vorinostat suppresses hypoxia signaling by modulating nuclear translocation of hypoxia inducible factor 1 alpha. Oncotarget 8(34):56110–56125

    PubMed  PubMed Central  Google Scholar 

  65. Mann BS et al (2007) FDA approval summary: vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 12(10):1247–1252

    Article  CAS  PubMed  Google Scholar 

  66. Pili R et al (2017) Combination of the histone deacetylase inhibitor vorinostat with bevacizumab in patients with clear-cell renal cell carcinoma: a multicentre, single-arm phase I/II clinical trial. Br J Cancer 116(7):874–883

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Lee YH et al (2016) Synthesis and biological evaluation of 1,2-dithiol-3-thiones and pyrrolo[1,2-a]pyrazines as novel hypoxia inducible factor-1 (HIF-1) inhibitor. Bioorg Med Chem 24(12):2843–2851

    Article  CAS  PubMed  Google Scholar 

  68. Koivunen P et al (2016) The 2-oxoglutarate analog 3-oxoglutarate decreases normoxic hypoxia-inducible factor-1alpha in cancer cells, induces cell death, and reduces tumor xenograft growth. Hypoxia (Auckl) 4:15–27

    Article  Google Scholar 

  69. Wang L et al (2015) Novel chalcone derivatives as hypoxia-inducible factor (HIF)-1 inhibitor: synthesis, anti-invasive and anti-angiogenic properties. Eur J Med Chem 89:88–97

    Article  CAS  PubMed  Google Scholar 

  70. Shen C, Kaelin WG Jr (2013) The VHL/HIF axis in clear cell renal carcinoma. Semin Cancer Biol 23(1):18–25

    Article  CAS  PubMed  Google Scholar 

  71. Holmquist-Mengelbier L et al (2006) Recruitment of HIF-1alpha and HIF-2alpha to common target genes is differentially regulated in neuroblastoma: HIF-2alpha promotes an aggressive phenotype. Cancer Cell 10(5):413–423

    Article  CAS  PubMed  Google Scholar 

  72. Wallace EM et al (2016) A small-molecule antagonist of HIF2alpha is efficacious in preclinical models of renal cell carcinoma. Cancer Res 76(18):5491–5500

    Article  CAS  PubMed  Google Scholar 

  73. Xu J et al (2017) Increasing AR by HIF-2alpha 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 8(10):e3095

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Courtney KD et al (2018) Phase I dose-escalation trial of PT2385, a first-in-class hypoxia-inducible factor-2alpha antagonist in patients with previously treated advanced clear cell renal cell carcinoma. J Clin Oncol 36(9):867–874

    Article  CAS  PubMed  Google Scholar 

  75. Wong TW et al (2018) PT2977, a novel HIF-2a antagonist, has potent antitumor activity and remodels the immunosuppressive tumor microenvironment in clear cell renal cell cancer. Mol Targets Cancer Ther 17(1(Suppl.))

    Google Scholar 

  76. Papadopoulos KP et al (2018) A first-in-human phase 1 dose-escalation trial of the oral HIF-2a inhibitor PT2977 in patients with advanced solid tumors. J Clin Oncol 36(15(Suppl.)):2508

    Article  Google Scholar 

  77. Sun DR et al (2018) Exploring the inhibition mechanism on HIF-2 by inhibitor PT2399 and 0X3 using molecular dynamics simulations. J Mol Recognit 31:e2730

    Article  PubMed  CAS  Google Scholar 

  78. Cho H et al (2016) On-target efficacy of a HIF-2alpha antagonist in preclinical kidney cancer models. Nature 539(7627):107–111

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Chen W et al (2016) Targeting renal cell carcinoma with a HIF-2 antagonist. Nature 539(7627):112–117

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Guise CP et al (2014) Bioreductive prodrugs as cancer therapeutics: targeting tumor hypoxia. Chin J Cancer 33(2):80–86

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Wilson WR, Hay MP (2011) Targeting hypoxia in cancer therapy. Nat Rev Cancer 11(6):393–410

    Article  CAS  PubMed  Google Scholar 

  82. Brown JM, Wang LH (1998) Tirapazamine: laboratory data relevant to clinical activity. Anticancer Drug Des 13(6):529–539

    CAS  PubMed  Google Scholar 

  83. Reddy SB, Williamson SK (2009) Tirapazamine: a novel agent targeting hypoxic tumor cells. Expert Opin Investig Drugs 18(1):77–87

    Article  CAS  PubMed  Google Scholar 

  84. Mistry IN et al (2017) Clinical advances of hypoxia-activated prodrugs in combination with radiation therapy. Int J Radiat Oncol Biol Phys 98(5):1183–1196

    Article  CAS  PubMed  Google Scholar 

  85. DiSilvestro PA et al (2014) Phase III randomized trial of weekly cisplatin and irradiation versus cisplatin and tirapazamine and irradiation in stages IB2, IIA, IIB, IIIB, and IVA cervical carcinoma limited to the pelvis: a Gynecologic Oncology Group Study. J Clin Oncol 32(5):458–464

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Cai TY et al (2014) Tirapazamine sensitizes hepatocellular carcinoma cells to topoisomerase I inhibitors via cooperative modulation of hypoxia-inducible factor-1alpha. Mol Cancer Ther 13(3):630–642

    Article  CAS  PubMed  Google Scholar 

  87. Hicks KO et al (2010) Pharmacokinetic/pharmacodynamic modeling identifies SN30000 and SN29751 as tirapazamine analogues with improved tissue penetration and hypoxic cell killing in tumors. Clin Cancer Res 16(20):4946–4957

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Wang J et al (2014) Identification of one-electron reductases that activate both the hypoxia prodrug SN30000 and diagnostic probe EF5. Biochem Pharmacol 91(4):436–446

    Article  CAS  PubMed  Google Scholar 

  89. Gu Y et al (2017) Reductive metabolism influences the toxicity and pharmacokinetics of the hypoxia-targeted benzotriazine di-oxide anticancer agent SN30000 in mice. Front Pharmacol 8:531

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Saggar JK, Tannock IF (2014) 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 134(11):2726–2734

    Article  CAS  PubMed  Google Scholar 

  91. Liapis V et al (2016) Anticancer efficacy of the hypoxia-activated prodrug evofosfamide (TH-302) in osteolytic breast cancer murine models. Cancer Med 5(3):534–545

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Sun JD et al (2016) Comparison of hypoxia-activated prodrug evofosfamide (TH-302) and ifosfamide in preclinical non-small cell lung cancer models. Cancer Biol Ther 17(4):371–380

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Zhang L et al (2016) Combined antitumor therapy with metronomic topotecan and hypoxia-activated prodrug, evofosfamide, in neuroblastoma and rhabdomyosarcoma preclinical models. Clin Cancer Res 22(11):2697–2708

    Article  CAS  PubMed  Google Scholar 

  94. Ganjoo KN et al (2011) 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 80(1–2):50–56

    Article  CAS  PubMed  Google Scholar 

  95. Chawla SP et al (2014) 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 32(29):3299–3306

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Tap WD et al (2017) Doxorubicin plus evofosfamide versus doxorubicin alone in locally advanced, unresectable or metastatic soft-tissue sarcoma (TH CR-406/SARC021): an international, multicentre, open-label, randomised phase 3 trial. Lancet Oncol 18(8):1089–1103

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Cutsem EV et al (2016) MAESTRO: A randomized, double-blind phase III study of evofosfamide (Evo) in combination with gemcitabine (Gem) in previously untreated patients (pts) with metastatic or locally advanced unresectable pancreatic ductal adenocarcinoma (PDAC). J Clin Oncol 34(15 (Suppl.)):4007

    Article  Google Scholar 

  98. Silva S et al (2015) Preclinical efficacy of tarloxotinib bromide (TH-4000), a hypoxia-activated EGFR/HER2 inhibitor: rationale for clinical evaluation in EGFR mutant, T790M-negative NSCLC following progression on EGFR-TKI therapy. Mol Cancer Ther 14(12 (Suppl.))

    Google Scholar 

  99. Patterson AV et al (2015) TH-4000, a hypoxia-activated EGFR/Her2 inhibitor to treat EGFR-TKI resistant T790M-negative NSCLC. J Clin Oncol 33(15 (Suppl.))

    Google Scholar 

  100. Hendriks HR et al (1993) EO9: a novel bioreductive alkylating indoloquinone with preferential solid tumour activity and lack of bone marrow toxicity in preclinical models. Eur J Cancer 29A(6):897–906

    Article  CAS  PubMed  Google Scholar 

  101. Adams GE et al (1992) Bioreductive drugs as post-irradiation sensitizers: comparison of dual function agents with SR 4233 and the mitomycin C analogue EO9. Int J Radiat Oncol Biol Phys 22(4):717–720

    Article  CAS  PubMed  Google Scholar 

  102. Phillips RM, Hendriks HR, Peters GJ (2013) EO9 (Apaziquone): from the clinic to the laboratory and back again. Br J Pharmacol 168(1):11–18

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. van der Heijden AG et al (2006) Phase II marker lesion study with intravesical instillation of apaziquone for superficial bladder cancer: toxicity and marker response. J Urol 176(4 Pt 1):1349–1353. discussion 1353

    Article  PubMed  CAS  Google Scholar 

  104. Hendricksen K et al (2009) Two-year follow-up of the phase II marker lesion study of intravesical apaziquone for patients with non-muscle invasive bladder cancer. World J Urol 27(3):337–342

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Phillips RM (2016) Targeting the hypoxic fraction of tumours using hypoxia-activated prodrugs. Cancer Chemother Pharmacol 77(3):441–457

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Patterson AV et al (2007) Mechanism of action and preclinical antitumor activity of the novel hypoxia-activated DNA cross-linking agent PR-104. Clin Cancer Res 13(13):3922–3932

    Article  CAS  PubMed  Google Scholar 

  107. Benito J et al (2011) Pronounced hypoxia in models of murine and human leukemia: high efficacy of hypoxia-activated prodrug PR-104. PLoS One 6(8):e23108

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Jameson MB et al (2010) A phase I trial of PR-104, a nitrogen mustard prodrug activated by both hypoxia and aldo-keto reductase 1C3, in patients with solid tumors. Cancer Chemother Pharmacol 65(4):791–801

    Article  CAS  PubMed  Google Scholar 

  109. McKeage MJ et al (2011) A phase I trial of PR-104, a pre-prodrug of the bioreductive prodrug PR-104A, given weekly to solid tumour patients. BMC Cancer 11:432

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. McKeage MJ et al (2012) PR-104 a bioreductive pre-prodrug combined with gemcitabine or docetaxel in a phase Ib study of patients with advanced solid tumours. BMC Cancer 12:496

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Patterson LH (2002) Bioreductively activated antitumor N-oxides: the case of AQ4N, a unique approach to hypoxia-activated cancer chemotherapy. Drug Metab Rev 34(3):581–592

    Article  CAS  PubMed  Google Scholar 

  112. Lalani AS et al (2007) Selective tumor targeting by the hypoxia-activated prodrug AQ4N blocks tumor growth and metastasis in preclinical models of pancreatic cancer. Clin Cancer Res 13(7):2216–2225

    Article  CAS  PubMed  Google Scholar 

  113. Nishida CR, Lee M, de Montellano PR (2010) Efficient hypoxic activation of the anticancer agent AQ4N by CYP2S1 and CYP2W1. Mol Pharmacol 78(3):497–502

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Hoang NT et al (2016) Hypoxia-inducible factor-targeting prodrug TOP3 combined with gemcitabine or TS-1 improves pancreatic cancer survival in an orthotopic model. Cancer Sci 107(8):1151–1158

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Zhang R et al (2017) Catalase-loaded cisplatin-prodrug-constructed liposomes to overcome tumor hypoxia for enhanced chemo-radiotherapy of cancer. Biomaterials 138:13–21

    Article  CAS  PubMed  Google Scholar 

  116. Nesbitt H et al (2017) Targeting hypoxic prostate tumors using the novel hypoxia-activated prodrug OCT1002 inhibits expression of genes associated with malignant progression. Clin Cancer Res 23(7):1797–1808

    Article  CAS  PubMed  Google Scholar 

  117. Nesbitt H et al (2017) The unidirectional hypoxia-activated prodrug OCT1002 inhibits growth and vascular development in castrate-resistant prostate tumors. Prostate 77(15):1539–1547

    Article  CAS  PubMed  Google Scholar 

  118. Hunter FW, Wouters BG, Wilson WR (2016) Hypoxia-activated prodrugs: paths forward in the era of personalised medicine. Br J Cancer 114(10):1071–1077

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Zhou J et al (2004) PI3K/Akt is required for heat shock proteins to protect hypoxia-inducible factor 1alpha from pVHL-independent degradation. J Biol Chem 279(14):13506–13513

    Article  CAS  PubMed  Google Scholar 

  120. Zhong H et al (2000) 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 60(6):1541–1545

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniele M. Gilkes .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2019 Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

DiGiacomo, J.W., Gilkes, D.M. (2019). Therapeutic Strategies to Block the Hypoxic Response. In: Gilkes, D. (eds) Hypoxia and Cancer Metastasis. Advances in Experimental Medicine and Biology, vol 1136. Springer, Cham. https://doi.org/10.1007/978-3-030-12734-3_10

Download citation

Publish with us

Policies and ethics