Skip to main content

Advertisement

Log in

Epigenetic approach for angiostatic therapy: promising combinations for cancer treatment

  • Review Paper
  • Published:
Angiogenesis Aims and scope Submit manuscript

Abstract

Cancer cells are often dependent on epigenetic pathways for their survival. Consequently, drugs that target the epigenome, rather than the underlying DNA sequence, are currently attracting considerable attention. In recent years, the first epigenetic drugs have been approved for cancer chemotherapy, mainly for hematological applications. Limitations in single-drug efficacies have thus far limited their application in the treatment of solid tumors. Nevertheless, promising activity for these compounds has been suggested when combined with other, distinctly targeted agents. In this review, we discuss the anti-angiogenic activity of histone deacetylase and DNA methyltransferase inhibitors and their combinations with other targeted (anti-angiogenic) therapeutics in treatment of solid tumors. The role that these inhibitors play in the inhibition of tumor angiogenesis, particularly in combination with other targeted agents, and the advantages they present over broad acting anticancer agents, are critically discussed.

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
Fig. 3
Fig. 4

Similar content being viewed by others

Abbreviations

5FD:

5-Fluoro-2′-deoxycytidine

Ang-2:

Angiopoietin2

AML:

Acute myeloid leukemia

AZA:

Azacitidine, 5-AZA-CR, 5-azacytidine

bFGF:

Basic fibroblast growth factor

FGFR:

Fibroblast growth factor receptor

CAM:

Chorioallantoic membrane of the chicken embryo

CTCL:

Cutaneous T cell lymphoma

CYR61:

Cysteine-rich angiogenic inducer 61

DAC:

Decitabine, 5-AZA-2′-deoxycytidine, 5-AZA-CdR

DNMT:

DNA methyltransferase

E-cadherin:

Epithelial cadherin

EC:

Endothelial cells

EGCG:

Epigallocatechin gallate

EGF:

Epidermal growth factor

EGFR:

Epidermal growth factor receptor

eNOS:

Endothelial nitric oxide synthase (eNOS)

FAO:

Fatty acid oxidation

FSC:

Feedback system control

HAT:

Acetyltransferase

HDAC:

Histone deacetylase

HIF-1α:

Hypoxia-inducible factor 1 alpha

HUVEC:

Human umbilical vein endothelial cells

HYD:

Hydralazine

IC50 :

The half maximal inhibitory concentration

IGF1:

Insulin-like growth factor 1

IL:

Interleukin

KP1019:

trans-[Tetrachlorobis(1H-indazole)ruthenate(III)

miRNA:

Noncoding microRNA

MMP:

Matrix metalloproteinase

MTA1:

Metastasis-associated protein 1

mTOR:

Mechanistic target of rapamycin

MVD:

Microvessel density

NAMI-A:

trans-[Tetrachloro(dimethylsulfoxide)(imidazole)ruthenate(III)]

NOX4:

NADPH oxidase 4

NSCLC:

Non-small cell lung cancer

NuRD:

Nucleosome remodeling and deacetylase

OS:

Overall survival

p300-HAT:

p300 histone acetyltransferase

PBA:

Phenylbutyrate

PDGF-B:

Platelet-derived growth factor subunit B

PDGFR-B:

Platelet-derived growth factor subunit B receptor

PFS:

Progression-free survival

PVRL2:

Poliovirus receptor-related 2

RAPTA-C:

Ru(η6-p-cymene)(pta)Cl2

RAPTA-T:

Ru(η6-toluene)(pta)Cl2

RCC:

Renal cell carcinoma

SAHA:

Vorinostat

SAM:

S-adenosyl methionine

siRNA:

Small interfering RNA

SIRT:

Histone deacetylase sirtuins

TGF-β:

Transforming growth factor beta

TIMP3:

Tissue inhibitor of matrix metalloproteinase 3

Tie-2:

Angiopoietin 1 receptor

TKI:

Tyrosine kinase inhibitor

TSA:

Trichostatin A

TSG:

Tumor suppressor gene

TSP-1:

Thrombospondin 1

VEGF:

Vascular endothelial growth factor

VEGFR:

Vascular endothelial growth factor receptor

VE-cadherin:

Vascular endothelial cadherin

VHL:

Von Hippel–Lindau

VPA:

Valproic acid

WIF-1:

Wnt inhibitory factor 1

ZEB:

Zebularine

References

  1. Rodríguez-Paredes M, Esteller M (2011) Cancer epigenetics reaches mainstream oncology. Nat Med 17(3):330–339

    Article  PubMed  CAS  Google Scholar 

  2. Russo VEA, Martienssen RA, Riggs AD (1996) Epigenetic mechanisms of gene regulation. Cold Spring Harbor, New york

  3. Viré E, Brenner C, Deplus R, Blanchon L, Fraga M, Didelot C, Morey L, Van Eynde A, Bernard D, Vanderwinden J-M, Bollen M, Esteller M, Di Croce L, de Launoit Y, Fuks F (2006) The Polycomb group protein EZH2 directly controls DNA methylation. Nature 439(7078):871–874

    Article  PubMed  CAS  Google Scholar 

  4. Bird A (2002) DNA methylation patterns and epigenetic memory. Genes Dev 16(1):6–21

    Article  CAS  PubMed  Google Scholar 

  5. Peserico A, Simone C (2011) Physical and functional HAT/HDAC interplay regulates protein acetylation balance. J Biomed Biotechnol. doi:10.1155/2011/371832

    PubMed  Google Scholar 

  6. Whittaker SJ, Demierre MF, Kim EJ, Rook AH, Lerner A, Duvic M, Scarisbrick J, Reddy S, Robak T, Becker JC, Samtsov A, McCulloch W, Kim YH (2010) Final results from a multicenter, international, pivotal study of romidepsin in refractory cutaneous T-cell lymphoma. J Clin Oncol 28(29):4485–4491

    Article  CAS  PubMed  Google Scholar 

  7. Santini V, Melnick A, Maciejewski JP, Duprez E, Nervi C, Cocco L, Ford KG, Mufti G (2013) Epigenetics in focus: pathogenesis of myelodysplastic syndromes and the role of hypomethylating agents. Crit Rev Oncol Hematol 88(2):231–245

    Article  PubMed  Google Scholar 

  8. Nervi C, De Marinis E, Codacci-Pisanelli G (2015) Epigenetic treatment of solid tumours: a review of clinical trials. Clin Epigenet 7(1):127

    Article  CAS  Google Scholar 

  9. Mann J (2002) Natural products in cancer chemotherapy: past, present and future. Nat Rev Cancer 2(2):143–148

    Article  CAS  PubMed  Google Scholar 

  10. Rajesh E, Sankari LS, Malathi L, Krupaa JR (2015) Naturally occurring products in cancer therapy. J Pharm Bioallied Sci 7(Suppl 1):S181–S183

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Cherblanc FL, Davidson RWM, Di Fruscia P, Srimongkolpithak N, Fuchter MJ (2013) Perspectives on natural product epigenetic modulators in chemical biology and medicine. Nat Prod Rep 30(5):605

    Article  CAS  PubMed  Google Scholar 

  12. Suzuki T, Miyata N (2006) Epigenetic control using natural products and synthetic molecules. Curr Med Chem 13(8):935–958

    Article  CAS  PubMed  Google Scholar 

  13. Herman JG, Baylin SB (2003) Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med 349(21):2042–2054

    Article  CAS  PubMed  Google Scholar 

  14. Esteller M (2007) Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet 8(4):286–298

    Article  CAS  PubMed  Google Scholar 

  15. Boehm T, Folkman J, Browder T, O’Reilly MS (1997) Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Nature 390(6658):404–407

    Article  CAS  PubMed  Google Scholar 

  16. Griffioen AW, Molema G (2000) Angiogenesis: potentials for pharmacologic intervention in the treatment of cancer, cardiovascular diseases, and chronic inflammation. Pharmacol Rev 52(2):237–268

    CAS  PubMed  Google Scholar 

  17. Bergers G, Hanahan D (2008) Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 8(8):592–603

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. van Beijnum JR, Nowak-Sliwinska P, Huijbers EJM, Thijssen VL, Griffioen AW (2015) The great escape; the hallmarks of resistance to antiangiogenic therapy. Pharmacol Rev 67(2):441–461

    Article  PubMed  CAS  Google Scholar 

  19. Huijbers EJM, van Beijnum JR, Thijssen VL, Sabrkhany S, Nowak-Sliwinska P, Griffioen AW (2016) Role of the tumor stroma in resistance to anti-angiogenic therapy. Drug Resist Updates 25:26–37

    Article  Google Scholar 

  20. Bani MR, Decio A, Giavazzi R, Ghilardi C (2017) Contribution of tumor endothelial cells to drug resistance: anti-angiogenic tyrosine kinase inhibitors act as p-glycoprotein antagonists. Angiogenesis. doi:10.1007/s10456-017-9549-6

  21. Chun P (2015) Histone deacetylase inhibitors in hematological malignancies and solid tumors. Arch Pharm Res 38(6):933–949

    Article  CAS  PubMed  Google Scholar 

  22. Hellebrekers DMEI, Melotte V, Viré E, Langenkamp E, Molema G, Fuks F, Herman JG, Van Criekinge W, Griffioen AW, Van Engeland M (2007) Identification of epigenetically silenced genes in tumor endothelial cells. Cancer Res 67(9):4138–4148

    Article  CAS  PubMed  Google Scholar 

  23. Pazin MJ, Kadonaga JT (1997) What’s up and down with histone deacetylation and transcription? Cell 89(3):325–328

    Article  CAS  PubMed  Google Scholar 

  24. Gu W, Roeder RG (1997) Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain. Cell 90(4):595–606

    Article  CAS  PubMed  Google Scholar 

  25. Zhang W, Bieker JJ (1998) Acetylation and modulation of erythroid Kruppel-like factor (EKLF) activity by interaction with histone acetyltransferases. Proc Natl Acad Sci USA 95(17):9855–9860

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Dokmanovic M, Clarke C, Marks PA (2007) Histone deacetylase inhibitors: overview and perspectives. Mol Cancer Res 5(10):981–989

    Article  CAS  PubMed  Google Scholar 

  27. Mottet D, Bellahcène A, Pirotte S, Waltregny D, Deroanne C, Lamour V, Lidereau R, Castronovo V (2007) Histone deacetylase 7 silencing alters endothelial cell migration, a key step in angiogenesis. Circ Res 101(12):1237–1246

    Article  CAS  PubMed  Google Scholar 

  28. Gallinari P, Di Marco S, Jones P, Pallaoro M, Steinkühler C (2007) HDACs, histone deacetylation and gene transcription: from molecular biology to cancer therapeutics. Cell Res 17(3):195–211

    CAS  PubMed  Google Scholar 

  29. Fath DM, Kong X, Liang D, Lin Z, Chou A, Jiang Y, Fang J, Caro J, Sang N (2006) Histone deacetylase inhibitors repress the transactivation potential of hypoxia-inducible factors independently of direct acetylation of HIF-alpha. J Biol Chem 281(19):13612–13619

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Semenza GL (2010) Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 29(5):625–634

    Article  CAS  PubMed  Google Scholar 

  31. Verheul HMW, Salumbides B, Van Erp K, Hammers H, Qian DZ, Sanni T, Atadja P, Pili R (2008) Combination strategy targeting the hypoxia inducible factor-1 alpha with mammalian target of rapamycin and histone deacetylase inhibitors. Clin Cancer Res 14(11):3589–3597

    Article  CAS  PubMed  Google Scholar 

  32. Kim DH, Kim M, Kwon HJ (2003) Histone deacetylase in carcinogenesis and its inhibitors as anti-cancer agents. J Biochem Mol Biol 36(1):110–119

    CAS  PubMed  Google Scholar 

  33. Kim MS, Kwon HJ, Lee YM, Baek JH, Jang JE, Lee SWK, Moon EJ, Kim HS, Lee SWK, Chung HY, Kim CW, Kim KW (2001) Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat Med 7(4):437–443

    Article  PubMed  Google Scholar 

  34. Michaelis M, Suhan T, Cinatl J, Driever PH, Cinatl J (2004) Valproic acid and interferon-alpha synergistically inhibit neuroblastoma cell growth in vitro and in vivo. Int J Oncol 25(6):1795–1799

    CAS  PubMed  Google Scholar 

  35. Michaelis M, Michaelis UR, Fleming I, Suhan T, Cinatl JJ, Blaheta RA, Hoffmann K, Kotchetkov R, Busse R, Nau H, Cinatl JJ (2004) Valproic acid inhibits angiogenesis in vitro and in vivo. Mol Pharmacol 65(3):520–527

    Article  CAS  PubMed  Google Scholar 

  36. Ellis L, Hammers H, Pili R (2009) Targeting tumor angiogenesis with histone deacetylase inhibitors. Cancer Lett 280(2):145–153

    Article  CAS  PubMed  Google Scholar 

  37. Kong X, Lin Z, Liang D, Fath D (2006) Histone deacetylase inhibitors induce VHL and ubiquitin-independent proteasomal degradation of hypoxia-inducible factor 1α. Mol Cell Biol 26(6):2019–2028

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hellebrekers DMEI et al (2006) Epigenetic regulation of tumor endothelial cell anergy: silencing of intercellular adhesion molecule-1 by histone modifications. Cancer Res 66(22):10770–10777

    Article  CAS  PubMed  Google Scholar 

  39. Srivastava RK, Kurzrock R, Shankar S (2010) MS-275 sensitizes TRAIL-resistant breast cancer cells, inhibits angiogenesis and metastasis, and reverses epithelial-mesenchymal transition in vivo. Mol Cancer Ther 9(12):3254–3266

    Article  CAS  PubMed  Google Scholar 

  40. Mie Lee Y, Kim S-H, Kim H-S, Jin Son M, Nakajima H, Jeong Kwon H, Kim K-W (2003) Inhibition of hypoxia-induced angiogenesis by FK228, a specific histone deacetylase inhibitor, via suppression of HIF-1alpha activity. Biochem Biophys Res Commun 300(1):241–246

    Article  PubMed  Google Scholar 

  41. Vieira-Coimbra M, Henrique R, Jeronimo C (2015) New insights on chromatin modifiers and histone post-translational modifications in renal cell tumours. Eur J Clin Invest 45:16–24

    Article  CAS  PubMed  Google Scholar 

  42. Chang S, Young BD, Li S, Qi X, Richardson JA, Olson EN (2006) Histone deacetylase 7 maintains vascular integrity by repressing matrix metalloproteinase 10. Cell 126(2):321–334

    Article  CAS  PubMed  Google Scholar 

  43. Kato H, Tamamizu-Kato S, Shibasaki F (2004) Histone deacetylase 7 associates with hypoxia-inducible factor 1alpha and increases transcriptional activity. J Biol Chem 279(40):41966–41974

    Article  CAS  PubMed  Google Scholar 

  44. Kaluza D, Kroll J, Gesierich S, Manavski Y, Boeckel J-N, Doebele C, Zelent A, Rossig L, Zeiher AM, Augustin HG, Urbich C, Dimmeler S (2013) Histone deacetylase 9 promotes angiogenesis by targeting the antiangiogenic microRNA-17-92 cluster in endothelial cells. Arterioscler Thromb Vasc Biol 33(3):533–543

    Article  CAS  PubMed  Google Scholar 

  45. Kaluza D, Kroll J, Gesierich S, Yao T-P, Boon RA, Hergenreider E, Tjwa M, Rossig L, Seto E, Augustin HG, Zeiher AM, Dimmeler S, Urbich C (2011) Class IIb HDAC6 regulates endothelial cell migration and angiogenesis by deacetylation of cortactin. EMBO J 30(20):4142–4156

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Hrgovic I, Doll M, Pinter A, Kaufmann R, Kippenberger S, Meissner M (2017) Histone deacetylase inhibitors interfere with angiogenesis by decreasing endothelial VEGFR-2 protein half-life in part via a VE-cadherin-dependent mechanism. Exp Dermatol 26(2):194–201

    Article  CAS  PubMed  Google Scholar 

  47. Calera MR, Venkatakrishnan A, Kazlauskas A (2004) VE-cadherin increases the half-life of VEGF receptor 2. Exp Cell Res 300(1):248–256

    Article  CAS  PubMed  Google Scholar 

  48. Hakami NY, Dusting GJ, Peshavariya HM (2016) Trichostatin A, a histone deacetylase inhibitor suppresses NADPH Oxidase 4-Derived Redox Signalling and Angiogenesis. J Cell Mol Med 20(10):1932–1944

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Deroanne CF, Bonjean K, Servotte S, Devy L, Colige A, Clausse N, Blacher S, Verdin E, Foidart J-M, Nusgens BV, Castronovo V (2002) Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene 21(3):427–436

    Article  CAS  PubMed  Google Scholar 

  50. Qian DZ, Wang X, Kachhap SK, Kato Y, Wei Y, Zhang L, Atadja P, Pili R (2004) The histone deacetylase inhibitor NVP-LAQ824 Inhibits angiogenesis and has a greater antitumor effect in combination with the vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584. Cancer Res 64(18):6626–6634

    Article  CAS  PubMed  Google Scholar 

  51. Potente M, Dimmeler S (2008) Emerging roles of SIRT1 in vascular endothelial homeostasis. Cell Cycle 7(14):2117–2122

    Article  CAS  PubMed  Google Scholar 

  52. Saunders LR, Verdin E (2007) Sirtuins: critical regulators at the crossroads between cancer and aging. Oncogene 26(37):5489–5504

    Article  CAS  PubMed  Google Scholar 

  53. Lappas M (2012) Anti-inflammatory properties of sirtuin 6 in human umbilical vein endothelial cells. Mediat Inflamm 2012:597514

    Article  Google Scholar 

  54. Zhang Q-J, Wang Z, Chen H-Z, Zhou S, Zheng W, Liu G, Wei Y-S, Cai H, Liu D-P, Liang C-C (2008) Endothelium-specific overexpression of class III deacetylase SIRT1 decreases atherosclerosis in apolipoprotein E-deficient mice. Cardiovasc Res 80(2):191–199

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Safe S, Kasiappan R (2016) Natural products as mechanism-based anticancer agents: Sp transcription factors as targets. Phyther Res 30(11):1723–1732

    Article  CAS  Google Scholar 

  56. Gerhauser C (2013) Cancer chemoprevention and nutri-epigenetics: state of the art and future challenges. Top Curr Chem 11(1):73–132

    Google Scholar 

  57. Sagar SM, Yance D, Wong RK (2006) Natural health products that inhibit angiogenesis: a potential source for investigational new agents to treat cancer—part 2. Curr Oncol 13(3):14–26

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Dhar S, Kumar A, Li K, Tzivion G, Levenson AS (2015) Resveratrol regulates PTEN/Akt pathway through inhibition of MTA1/HDAC unit of the NuRD complex in prostate cancer. Biochim Biophys Acta Mol Cell Res 1853(2):265–275

    Article  CAS  Google Scholar 

  59. Saenglee S, Jogloy S, Patanothai A, Leid M, Senawong T (2016) Cytotoxic effects of peanut phenolics possessing histone deacetylase inhibitory activity in breast and cervical cancer cell lines. Pharmacol Rep 68(6):1102–1110

    Article  CAS  PubMed  Google Scholar 

  60. Tseng SH, Lin SM, Chen JC, Su YH, Huang HY, Chen CK, Lin PY, Chen Y (2004) Resveratrol suppresses the angiogenesis and tumor growth of gliomas in rats. Clin Cancer Res 10(6):2190–2202

    Article  CAS  PubMed  Google Scholar 

  61. Raina K, Kumar S, Dhar D, Agarwal R (2016) Silibinin and colorectal cancer chemoprevention: a comprehensive review on mechanisms and efficacy. J Biomed Res 30(6):452–465

    PubMed  Google Scholar 

  62. Deep G, Kumar R, Nambiar DK, Jain AK, Ramteke AM, Serkova NJ, Agarwal C, Agarwal R (2016) Silibinin inhibits hypoxia-induced HIF-1α–mediated signaling, angiogenesis and lipogenesis in prostate cancer cells: in vitro evidence and in vivo functional imaging and metabolomics. Mol Carcinog 56(3):833–848

    Article  PubMed  CAS  Google Scholar 

  63. Jiang C, Agarwal R, Lü J (2000) Anti-angiogenic potential of a cancer chemopreventive flavonoid antioxidant, silymarin: inhibition of key attributes of vascular endothelial cells and angiogenic cytokine secretion by cancer epithelial cells. Biochem Biophys Res Commun 276(1):371–378

    Article  CAS  PubMed  Google Scholar 

  64. Singh RP, Sharma G, Dhanalakshmi S, Agarwal C, Agarwal R (2003) Suppression of advanced human prostate tumor growth in athymic mice by silibinin feeding is associated with reduced cell proliferation, increased apoptosis, and inhibition of angiogenesis. Cancer Epidemiol Biomark Prev 12(9):933–939

    CAS  Google Scholar 

  65. Singh T, Prasad R, Katiyar SK (2016) Therapeutic intervention of silymarin on the migration of non-small cell lung cancer cells is associated with the axis of multiple molecular targets including class 1 HDACs, ZEB1 expression, and restoration of miR-203 and E-cadherin expression. Am J Cancer Res 6(6):1287–1301

    PubMed  PubMed Central  Google Scholar 

  66. Laird PW, Jaenisch R (1996) The role of DNA methylation in cancer genetics and epigenetics. Annu Rev Genet 30(1):441–464

    Article  CAS  PubMed  Google Scholar 

  67. Watt F, Molloy PL (1988) Cytosine methylation prevents binding to DNA of a HeLa cell transcription factor required for optimal expression of the adenovirus major late promoter. Genes Dev 2(9):1136–1143

    Article  CAS  PubMed  Google Scholar 

  68. Hendrich B, Bird A (1998) Identification and characterization of a family of mammalian methyl-CpG binding proteins. Mol Cell Biol 18(11):6538–6547

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Boyes J, Bird A (1991) DNA methylation inhibits transcription indirectly via a methyl-CpG binding protein. Cell 64(6):1123–1134

    Article  CAS  PubMed  Google Scholar 

  70. Bird A, Nan X, Ng H-H, Johnson CA, Laherty CD, Turner BM, Eisenman RN (1998) Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393(6683):386–389

    Article  PubMed  CAS  Google Scholar 

  71. Okano M, Xie S, Li E (1998) Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nat Am Inc 19(3):219–220

    CAS  Google Scholar 

  72. Okano M, Bell DW, Haber DA, Li E (1999) DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99(3):247–257

    Article  CAS  PubMed  Google Scholar 

  73. Beard C, Li E, Jaenisch R (1995) Loss of methylation activates Xist in somatic but not in embryonic cells. Genes Dev 9(19):2325–2334

    Article  CAS  PubMed  Google Scholar 

  74. Bestor TH (1992) Activation of mammalian DNA methyltransferase by cleavage of a Zn binding regulatory domain. EMBO J 11(7):2611–2617

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Li E, Bestor TH, Jaenisch R (1992) Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69(6):915–926

    Article  CAS  PubMed  Google Scholar 

  76. Matouk CC, Marsden PA (2008) Epigenetic regulation of vascular endothelial gene expression. Circ Res 102(8):873–878

    Article  CAS  PubMed  Google Scholar 

  77. Şahin M, Şahin E, Gümüşlü S, Erdoǧan A, Gültekin M (2010) DNA methylation or histone modification status in metastasis and angiogenesis-related genes: a new hypothesis on usage of DNMT inhibitors and S-adenosylmethionine for genome stability. Cancer Metastasis Rev 29(4):655–676

    Article  PubMed  CAS  Google Scholar 

  78. Guo W, Dong Z, He M, Guo Y, Guo J, Chen Z, Yang Z, Kuang G (2010) Aberrant methylation of thrombospondin-1 and its association with reduced expression in gastric cardia adenocarcinoma. J Biomed Biotechnol. doi:10.1155/2010/721485

    Google Scholar 

  79. Zhang Y, Wang X, Xu B, Wang B, Wang Z, Liang Y, Zhou J, Hu J, Jiang B (2013) Epigenetic silencing of miR-126 contributes to tumor invasion and angiogenesis in colorectal cancer. Oncol Rep 30(4):1976–1984

    Article  CAS  PubMed  Google Scholar 

  80. Chan SL, Cui Y, van Hasselt A, Li H, Srivastava G, Jin H, Ng KM, Wang Y, Lee KY, Tsao GSW, Zhong S, Robertson KD, Rha SY, Chan ATC, Tao Q (2007) The tumor suppressor Wnt inhibitory factor 1 is frequently methylated in nasopharyngeal and esophageal carcinomas. Lab Investig 87(7):644–650

    Article  CAS  PubMed  Google Scholar 

  81. Bo BL, Eun JL, Eun HJ, Chun HK, Dong KC, Sang YS, Park J, Kim DH (2009) Aberrant methylation of APC, MGMT, RASSF2A, and Wif-1 genes in plasma as a biomarker for early detection of colorectal cancer. Clin Cancer Res 15(19):6185–6191

    Article  CAS  Google Scholar 

  82. Veeck J, Wild PJ, Fuchs T, Schüffler PJ, Hartmann A, Knüchel R, Dahl E (2009) Prognostic relevance of Wnt-inhibitory factor-1 (WIF1) and Dickkopf-3 (DKK3) promoter methylation in human breast cancer. BMC Cancer. doi:10.1186/1471-2407-9-217

    PubMed  PubMed Central  Google Scholar 

  83. Lee SM, Park JY, Kim DS (2013) Wif1 hypermethylation as unfavorable prognosis of non-small cell lung cancers with EGFR mutation. Mol Cells 36(1):69–73

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Lambiv WL, Vassallo I, Delorenzi M, Shay T, Diserens AC, Misra A, Feuerstein B, Murat A, Migliavacca E, Hamou MF, Sciuscio D, Burger R, Domany E, Stupp R, Hegi ME (2011) The Wnt inhibitory factor 1 (WIF1) is targeted in glioblastoma and has a tumor suppressing function potentially by induction of senescence. Neuro Oncol 13(7):736–747

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Skurk C, Maatz H, Rocnik E, Bialik A, Force T, Walsh K (2005) Glycogen-synthase kinase3beta/beta-catenin axis promotes angiogenesis through activation of vascular endothelial growth factor signaling in endothelial cells. Circ Res 96(3):308–318

    Article  CAS  PubMed  Google Scholar 

  86. Carmeliet P, Jain RK (2011) Molecular mechanisms and clinical applications of angiogenesis. Nature 473(7347):298–307

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Ramachandran I, Thavathiru E, Ramalingam S, Natarajan G, Mills WK, Benbrook DM, Zuna R, Lightfoot S, Reis A, Anant S, Queimado L (2012) Wnt inhibitory factor 1 induces apoptosis and inhibits cervical cancer growth, invasion and angiogenesis in vivo. Oncogene 31(22):2725–2737

    Article  CAS  PubMed  Google Scholar 

  88. Saito Y, Friedman JM, Chihara Y, Egger G, Chuang JC, Liang G (2009) Epigenetic therapy upregulates the tumor suppressor microRNA-126 and its host gene EGFL7 in human cancer cells. Biochem Biophys Res Commun 379(3):726–731

    Article  CAS  PubMed  Google Scholar 

  89. van Beijnum JR, Giovannetti E, Poel D, Nowak-Sliwinska P, Griffioen AW (2017) miRNAs: micro-managers of anti-cancer combination therapies. Angiogenesis. doi:10.1007/s10456-017-9545-x

  90. Lindner DJ, Wu Y, Haney R, Jacobs BS, Fruehauf JP, Tuthill R, Borden EC (2013) Thrombospondin-1 expression in melanoma is blocked by methylation and targeted reversal by 5-Aza-deoxycytidine suppresses angiogenesis. Matrix Biol 32(2):123–132

    Article  CAS  PubMed  Google Scholar 

  91. Mirochnik Y, Kwiatek A, Volpert O (2008) Thrombospondin and apoptosis: molecular mechanisms and use for design of complementation treatments. Curr Drug Targets 9(10):851–862

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Jueliger S, Lyons J, Cannito S, Pata I, Pata P, Shkolnaya M, Lo Re O, Peyrou M, Villarroya F, Pazienza V, Rappa F, Cappello F, Azab M, Taverna P, Vinciguerra M (2016) Efficacy and epigenetic interactions of novel DNA hypomethylating agent guadecitabine (SGI-110) in preclinical models of hepatocellular carcinoma. Epigenetics 11(10):709–720

    Article  PubMed Central  Google Scholar 

  93. Shih YP, Liao YC, Lin Y, Lo SH (2010) DLC1 negatively regulates angiogenesis in a paracrine fashion. Cancer Res 70(21):8270–8275

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Griffioen AW (2008) Anti-angiogenesis: making the tumor vulnerable to the immune system. Cancer Immunol Immunother 57(10):1553–1558

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Hellebrekers DMEI, Jair K-W, Viré E, Eguchi S, Hoebers NTH, Fraga MF, Esteller M, Fuks F, Baylin SB, van Engeland M, Griffioen AW (2006) Angiostatic activity of DNA methyltransferase inhibitors. Mol Cancer Ther 5(2):467–475

    Article  CAS  PubMed  Google Scholar 

  96. Alexius-Lindgren M, Andersson E, Lindstedt I, Engström W (2014) The RECK gene and biological malignancy-its significance in angiogenesis and inhibition of matrix metalloproteinases. Anticancer Res 34(8):3867–3874

    CAS  PubMed  Google Scholar 

  97. Shian S-G, Kao Y-R, Wu FY-H, Wu C-W (2003) Inhibition of invasion and angiogenesis by zinc-chelating agent disulfiram. Mol Pharmacol 64(5):1076–1084

    Article  PubMed  Google Scholar 

  98. Murai R, Yoshida Y, Muraguchi T, Nishimoto E, Morioka Y, Kitayama H, Kondoh S, Kawazoe Y, Hiraoka M, Uesugi M, Noda M (2010) A novel screen using the Reck tumor suppressor gene promoter detects both conventional and metastasis-suppressing anticancer drugs. Oncotarget 1(4):252–264

    Article  PubMed  PubMed Central  Google Scholar 

  99. Graça I, Sousa EJ, Costa-Pinheiro P, Vieira FQ, Torres-Ferreira J, Martins MG, Henrique R, Jerónimo C (2014) Anti-neoplastic properties of hydralazine in prostate cancer. Oncotarget 5(15):5950–5964

    Article  PubMed  PubMed Central  Google Scholar 

  100. Chavez-Blanco A, Perez-Plasencia C, Perez-Cardenas E, Carrasco-Legleu C, Rangel-Lopez E, Segura-Pacheco B, Taja-Chayeb L, Trejo-Becerril C, Gonzalez-Fierro A, Candelaria M, Cabrera G, Duenas-Gonzalez A (2006) Antineoplastic effects of the DNA methylation inhibitor hydralazine and the histone deacetylase inhibitor valproic acid in cancer cell lines. Cancer Cell Int. doi:10.1186/1475-2867-6-2

    Google Scholar 

  101. Zhang Q, Lin Z, Yin X, Tang L, Luo H, Li H, Zhang Y, Luo W (2016) In vitro and in vivo study of hydralazine, a potential anti-angiogenic agent. Eur J Pharmacol 779:138–146

    Article  CAS  PubMed  Google Scholar 

  102. Li X, Feng Y, Liu J, Feng X, Zhou K, Tang X (2013) Epigallocatechin-3-gallate inhibits IGF-I-stimulated lung cancer angiogenesis through downregulation of HIF-1α and VEGF expression. J Nutrigenet Nutrigenom 6(3):169–178

    Article  CAS  Google Scholar 

  103. Sakamoto Y, Terashita N, Muraguchi T, Fukusato T, Kubota S (2013) Effects of epigallocatechin-3-gallate (EGCG) on A549 lung cancer tumor growth and angiogenesis. Biosci Biotechnol Biochem 77(9):1799–1803

    Article  CAS  PubMed  Google Scholar 

  104. Gu J, Makey KL, Tucker KB, Chinchar E, Mao X, Pei I, Thomas EY, Miele L (2013) EGCG, a major green tea catechin suppresses breast tumor angiogenesis and growth via inhibiting the activation of HIF-1alpha and NFkappaB, and VEGF expression. Vasc Cell 5(1):9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Kato K, Long NK, Makita H, Toida M, Yamashita T, Hatakeyama D, Hara A, Mori H, Shibata T (2008) Effects of green tea polyphenol on methylation status of RECK gene and cancer cell invasion in oral squamous cell carcinoma cells. Br J Cancer 99(4):647–654

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Kunnumakkara AB, Anand P, Aggarwal BB (2008) Curcumin inhibits proliferation, invasion, angiogenesis and metastasis of different cancers through interaction with multiple cell signaling proteins. Cancer Lett 269(2):199–225

    Article  CAS  PubMed  Google Scholar 

  107. Arbiser JL, Klauber N, Rohan R, van Leeuwen R, Huang MT, Fisher C, Flynn E, Byers HR (1998) Curcumin is an in vivo inhibitor of angiogenesis. Mol Med 4(6):376–383

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Du L, Xie Z, Wu LC, Chiu M, Lin J, Chan KK, Liu S, Liu Z (2012) Reactivation of RASSF1A in breast cancer cells by curcumin. Nutr Cancer Int J 64(8):1228–1235

    Article  CAS  Google Scholar 

  109. Mirza S, Sharma G, Parshad R, Gupta SD, Pandya P, Ralhan R (2013) Expression of DNA methyltransferases in breast cancer patients and to analyze the effect of natural compounds on DNA methyltransferases and associated proteins. J Breast Cancer 16(1):23–31

    Article  PubMed  PubMed Central  Google Scholar 

  110. Teiten MH, Dicato M, Diederich M (2013) Curcumin as a regulator of epigenetic events. Mol Nutr Food Res 57(9):1619–1629

    Article  CAS  PubMed  Google Scholar 

  111. Alexandrow MG, Song LJ, Altiok S, Gray J, Haura EB, Kumar NB (2012) Curcumin: a novel Stat3 pathway inhibitor for chemoprevention of lung cancer. Eur J Cancer Prev 21(5):407–412

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Niu G, Wright KL, Huang M, Song L, Haura E, Turkson J, Zhang S, Wang T, Sinibaldi D, Coppola D, Heller R, Ellis LM, Karras J, Bromberg J, Pardoll D, Jove R, Yu H (2002) Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene 21(13):2000–2008

    Article  CAS  PubMed  Google Scholar 

  113. Wei D, Le X, Zheng L, Wang L, Frey JA, Gao AC, Peng Z, Huang S, Xiong HQ, Abbruzzese JL, Xie K (2003) Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis. Oncogene 22(3):319–329

    Article  CAS  PubMed  Google Scholar 

  114. Lee H, Zhang P, Herrmann A, Yang C, Xin H, Wang Z, Hoon DSB, Forman SJ, Jove R, Riggs AD, Yu H (2012) Acetylated STAT3 is crucial for methylation of tumor-suppressor gene promoters and inhibition by resveratrol results in demethylation. Proc Natl Acad Sci 109(20):7765–7769

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Huang L, Hu B, Ni J, Wu J, Jiang W, Chen C, Yang L, Zeng Y, Wan R, Hu G, Wang X (2016) Transcriptional repression of SOCS3 mediated by IL-6/STAT3 signaling via DNMT1 promotes pancreatic cancer growth and metastasis. J Exp Clin Cancer Res 35(1):27

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Stahl A, Joyal JS, Chen J, Sapieha P, Juan AM, Hatton CJ, Pei DT, Hurst CG, Seaward MR, Krah NM, Dennison RJ, Greene ER, Boscolo E, Panigrahy D, Smith LEH (2012) SOCS3 is an endogenous inhibitor of pathologic angiogenesis. Blood 120(14):2925–2929

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Dhillon N, Aggarwal BB, Newman RA, Wolff RA, Kunnumakkara AB, Abbruzzese JL, Ng CS, Badmaev V, Kurzrock R (2008) Phase II trial of curcumin in patients with advanced pancreatic cancer. Clin Cancer Res 14(14):4491–4499

    Article  CAS  PubMed  Google Scholar 

  118. Licona C, Spaety M-E, Capuozzo A, Ali M, Santamaria R, Armant O, Delalande F, Van Dorsselaer A, Cianferani S, Spencer J, Pfeffer M, Mellitzer G, Gaiddon C (2017) A ruthenium anticancer compound interacts with histones and impacts differently on epigenetic and death pathways compared to cisplatin. Oncotarget 8(2):2568–2584

    PubMed  Google Scholar 

  119. Nazarov AA, Baquié M, Nowak-Sliwinska P, Zava O, van Beijnum JR, Groessl M, Chisholm DM, Ahmadi Z, McIndoe JS, Griffioen AW, van den Bergh H, Dyson PJ (2013) Synthesis and characterization of a new class of anti-angiogenic agents based on ruthenium clusters. Sci Rep 3:1485

    Article  PubMed  CAS  Google Scholar 

  120. Clavel CM, Păunescu E, Nowak-Sliwinska P, Griffioen AW, Scopelliti R, Dyson PJ (2014) Discovery of a highly tumor-selective organometallic ruthenium(II)-arene complex. J Med Chem 57(8):3546–3558

    Article  CAS  PubMed  Google Scholar 

  121. Allardyce CS, Dyson PJ (2016) Metal-based drugs that break the rules. Dalton Trans 45(8):3201–3209

    Article  CAS  PubMed  Google Scholar 

  122. Singh V, Azad GK, Mandal P, Amarendar Reddy M, Tomar RS (2014) Anti-cancer drug KP1019 modulates epigenetics and induces DNA damage response in Saccharomyces cerevisiae. FEBS Lett 588(6):1044–1052

    Article  CAS  PubMed  Google Scholar 

  123. Leijen S, Burgers SA, Baas P, Pluim D, Tibben M, Van Werkhoven E, Alessio E, Sava G, Beijnen JH, Schellens JHM (2015) Phase I/II study with ruthenium compound NAMI-A and gemcitabine in patients with non-small cell lung cancer after first line therapy. Investig New Drugs 33(1):201–214

    Article  CAS  Google Scholar 

  124. Rademaker-Lakhai JM, Van Den Bongard D, Pluim D, Beijnen JH, Schellens JHM (2004) A phase I and pharmacological study with imidazolium-trans-DMSO-imidazole-tetrachlororuthenate, a novel ruthenium anticancer agent. Clin Cancer Res 10:3717–3727

    Article  CAS  PubMed  Google Scholar 

  125. Hartinger CG, Jakupec MA, Zorbas-Seifried S, Groessl M, Egger A, Berger W, Zorbas H, Dyson PJ, Keppler BK (2008) KP1019, a new redox-active anticancer agent—preclinical development and results of a clinical phase I study in tumor patients. Chem Biodivers 5(10):2140–2155

    Article  CAS  PubMed  Google Scholar 

  126. Trondl R, Heffeter P, Kowol CR, Jakupec MA, Berger W, Keppler BK (2014) NKP-1339, the first ruthenium-based anticancer drug on the edge to clinical application. Chem Sci 5(8):2925

    Article  CAS  Google Scholar 

  127. Morbidelli L, Donnini S, Filippi S, Messori L, Piccioli F, Orioli P, Sava G, Ziche M (2003) Antiangiogenic properties of selected ruthenium(III) complexes that are nitric oxide scavengers. Br J Cancer 88(9):1484–1491

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Pintus G, Tadolini B, Posadino AM, Sanna B, Debidda M, Bennardini F, Sava G, Ventura C (2002) Inhibition of the MEK/ERK signaling pathway by the novel antimetastatic agent NAMI-A down regulates c-myc gene expression and endothelial cell proliferation. Eur J Biochem 269(23):5861–5870

    Article  CAS  PubMed  Google Scholar 

  129. Vacca A, Bruno M, Boccarelli A, Coluccia M, Ribatti D, Bergamo A, Garbisa S, Sartor L, Sava G (2002) Inhibition of endothelial cell functions and of angiogenesis by the metastasis inhibitor NAMI-A. Br J Cancer 86(6):993–998

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Hartinger CG, Dyson PJ (2009) Bioorganometallic chemistry–from teaching paradigms to medicinal applications. Chem Soc Rev 38(2):391–401

    Article  CAS  PubMed  Google Scholar 

  131. Smith GS, Therrien B (2011) Targeted and multifunctional arene ruthenium chemotherapeutics. Dalton Trans 40(41):10793

    Article  CAS  PubMed  Google Scholar 

  132. Süss-Fink G (2014) Water-soluble arene ruthenium complexes: from serendipity to catalysis and drug design. J Organomet Chem 751:2–19

    Article  CAS  Google Scholar 

  133. Murray BS, Babak MV, Hartinger CG, Dyson PJ (2016) The development of RAPTA compounds for the treatment of tumors. Coord Chem Rev 306:86–114

    Article  CAS  Google Scholar 

  134. Scolaro C, Bergamo A, Brescacin L, Delfino R, Cocchietto M, Laurenczy G, Geldbach TJ, Sava G, Dyson PJ (2005) In vitro and in vivo evaluation of ruthenium(II)-arene PTA complexes. J Med Chem 48:4161–4171

    Article  CAS  PubMed  Google Scholar 

  135. Weiss A, Berndsen RH, Dubois M, Müller C, Schibli R, Griffioen AW, Dyson PJ, Nowak-Sliwinska P (2014) In vivo anti-tumor activity of the organometallic ruthenium(II)-arene complex [Ru(η6-p-cymene)Cl2 (pta)] (RAPTA-C) in human ovarian and colorectal carcinomas. Chem Sci 5(12):4742–4748

    Article  CAS  Google Scholar 

  136. Nowak-Sliwinska P, Van Beijnum JR, Casini A, Nazarov AA, Wagnieres G, Van Den Bergh H, Dyson PJ, Griffioen AW (2011) Organometallic ruthenium(II) arene compounds with antiangiogenic activity. J Med Chem 54(11):3895–3902

    Article  CAS  PubMed  Google Scholar 

  137. Shoemaker RH (2006) The NCI60 human tumour cell line anticancer drug screen. Nat Rev Cancer 6(10):813–823

    Article  CAS  PubMed  Google Scholar 

  138. Adhireksan Z, Davey GE, Campomanes P, Groessl M, Clavel CM, Yu H, Nazarov AA, Yeo CHF, Ang WH, Dröge P, Rothlisberger U, Dyson PJ, Davey CA (2014) Ligand substitutions between ruthenium-cymene compounds can control protein versus DNA targeting and anticancer activity. Nat Commun 5:3462

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  139. Nowak-Sliwinska P, Segura T, Iruela-Arispe ML (2014) The chicken chorioallantoic membrane model in biology, medicine and bioengineering. Angiogenesis 4:779–804

    Article  CAS  Google Scholar 

  140. Lee RFS, Escrig S, Croisier M, Clerc-Rosset S, Knott GW, Meibom A, Davey CA, Johnsson K, Dyson PJ (2015) NanoSIMS analysis of an isotopically labelled organometallic ruthenium(II) drug to probe its distribution and state in vitro. Chem Commun (Camb) 51(92):16486–16489

    Article  CAS  Google Scholar 

  141. Wu B, Ong MS, Groessl M, Adhireksan Z, Hartinger CG, Dyson PJ, Davey CA (2011) A ruthenium antimetastasis agent forms specific histone protein adducts in the nucleosome core. Chemistry 17(13):3562–3566

    Article  CAS  PubMed  Google Scholar 

  142. Adhireksan Z, Palermo G, Riedel T, Ma Z, Muhammad R, Röthlisberger U, Dyson PJ, Davey CA (2017) Allosteric cross-talk in chromatin can mediate drug-drug synergy. Nat Commun. doi:10.1038/ncomms14860

  143. Weiss A, Nowak-Sliwinska P (2016) Current trends in multidrug optimization. J Lab Autom. doi:10.1177/2211068216682338

    PubMed  Google Scholar 

  144. Nowak-Sliwinska P, Weiss A, Ding X, Dyson PJ, van den Bergh H, Griffioen AW, Ho C-M (2016) Optimization of drug combinations using feedback system control. Nat Protoc 11(2):302–315. doi:10.1177/2472630316682338

    Article  CAS  PubMed  Google Scholar 

  145. Weiss A, Ding X, van Beijnum JR, Wong I, Wong TJ, Berndsen RH, Dormond O, Dallinga M, Shen L, Schlingemann RO, Pili R, Ho C-M, Dyson PJ, van den Bergh H, Griffioen AW, Nowak-Sliwinska P (2015) Rapid optimization of drug combinations for the optimal angiostatic treatment of cancer. Angiogenesis 18(3):233–244

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Oronsky BT, Oronsky AL, Lybeck M, Oronsky NC, Scicinski JJ, Carter C, Day RM, Rodriguez Orengo JF, Rodriguez-Torres M, Fanger GF, Reid TR (2015) Episensitization: defying time’s arrow. Front Oncol. doi:10.3389/fonc.2015.00134

    Google Scholar 

  147. Spencer J, Amin J, Wang M, Packham G, Alwi SSS, Tizzard GJ, Coles SJ, Paranal RM, Bradner JE, Heightman TD (2011) Synthesis and biological evaluation of JAHAs: ferrocene-based histone deacetylase inhibitors. ACS Med Chem Lett 2(5):358–362

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Librizzi M, Longo A, Chiarelli R, Amin J, Spencer J, Luparello C (2012) Cytotoxic effects of Jay Amin hydroxamic acid (JAHA), a ferrocene-based class i histone deacetylase inhibitor, on triple-negative MDA-MB231 breast cancer cells. Chem Res Toxicol 25(11):2608–2616

    Article  CAS  PubMed  Google Scholar 

  149. Beisel C, Paro R (2011) Silencing chromatin: comparing modes and mechanisms. Nat Rev Genet 12(2):123–135

    Article  CAS  PubMed  Google Scholar 

  150. Robertson KD, Ait-Si-Ali S, Yokochi T, Wade PA, Jones PL, Wolffe AP (2000) DNMT1 forms a complex with Rb, E2F1 and HDAC1 and represses transcription from E2F-responsive promoters. Nat Genet 25(3):338–342

    Article  CAS  PubMed  Google Scholar 

  151. Rountree MR, Bachman KE, Baylin SB (2000) DNMT1 binds HDAC2 and a new co-repressor, DMAP1, to form a complex at replication foci. Nat Genet 25(3):269–277

    Article  CAS  PubMed  Google Scholar 

  152. Shiva Shankar TV, Willems L (2014) Epigenetic modulators mitigate angiogenesis through a complex transcriptomic network. Vasc Pharmacol 60(2):57–66

    Article  CAS  Google Scholar 

  153. Chen WY, Townes TM (2000) Molecular mechanism for silencing virally transduced genes involves histone deacetylation and chromatin condensation. Proc Natl Acad Sci USA 97(1):377–382

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Liu X, Luo M, Wu K (2012) Epigenetic interplay of histone modifications and DNA methylation mediated by HDA6. Plant Signal Behav 7(6):633–635

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Bachman KE, Park BH, Rhee I, Rajagopalan H, Herman JG, Baylin SB, Kinzler KW, Vogelstein B (2003) Histone modifications and silencing prior to DNA methylation of a tumor suppressor gene. Cancer Cell 3(1):89–95

    Article  CAS  PubMed  Google Scholar 

  156. Selker EU (1998) Trichostatin A causes selective loss of DNA methylation in Neurospora. Proc Natl Acad Sci USA 95(16):9430–9435

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Zhou Q, Agoston AT, Atadja P, Nelson WG, Davidson NE (2008) Inhibition of histone deacetylases promotes ubiquitin-dependent proteasomal degradation of DNA methyltransferase 1 in human breast cancer cells. Mol Cancer Res 6(5):873–883

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Vaissière T, Sawan C, Herceg Z (2008) Epigenetic interplay between histone modifications and DNA methylation in gene silencing. Mutat Res 659(1–2):40–48

    Article  PubMed  CAS  Google Scholar 

  159. Cedar H, Bergman Y (2009) Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet 10(5):295–304

    Article  CAS  PubMed  Google Scholar 

  160. Lyu T, Jia N, Wang J, Yan X, Yu Y, Lu Z, Bast RC, Hua K, Feng W (2013) Expression and epigenetic regulation of angiogenesis-related factors during dormancy and recurrent growth of ovarian carcinoma. Epigenetics 8(12):1330–1346

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Gros C, Fahy J, Halby L, Dufau I, Erdmann A, Gregoire JM, Ausseil F, Vispé S, Arimondo PB (2012) DNA methylation inhibitors in cancer: recent and future approaches. Biochimie 94(11):2280–2296

    Article  CAS  PubMed  Google Scholar 

  162. Cameron EE, Bachman KE, Myohanen S, Herman JG, Baylin SB (1999) Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet 21(1):103–107

    Article  CAS  PubMed  Google Scholar 

  163. Zhang J, Lu A, Li L, Yue J, Lu Y (2010) p16 Modulates VEGF expression via its interaction with HIF-1alpha in breast cancer cells. Cancer Invest 28(6):588–597

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Miki K, Shimizu E, Yano S, Tani K, Sone S (2001) Demethylation by 5-aza-2′-deoxycytidine (5-azadC) of p16INK4A gene results in downregulation of vascular endothelial growth factor expression in human lung cancer cell lines. Oncol Res 12(8):335–342

    CAS  PubMed  Google Scholar 

  165. Anand-Apte B, Bao L, Smith R, Iwata K, Olsen BR, Zetter B, Apte SS (1996) A review of tissue inhibitor of metalloproteinases-3 (TIMP-3) and experimental analysis of its effect on primary tumor growth. Biochem Cell Biol 74(6):853–862

    Article  CAS  PubMed  Google Scholar 

  166. Qi JH, Ebrahem Q, Moore N, Murphy G, Claesson-Welsh L, Bond M, Baker A, Anand-Apte B (2003) A novel function for tissue inhibitor of metalloproteinases-3 (TIMP3): inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2. Nat Med 9(4):407–415

    Article  CAS  PubMed  Google Scholar 

  167. Suzuki M, Shinohara F, Rikiishi H (2008) Zebularine-induced reduction in VEGF secretion by HIF-1α degradation in oral squamous cell carcinoma. Mol Med Rep 1(4):465–471

    CAS  PubMed  Google Scholar 

  168. Cecconi D, Donadelli M, Pozza ED, Rinalducci S, Zolla L, Scupoli MT, Righetti PG, Scarpa A, Palmieri M (2009) Synergistic effect of trichostatin A and 5-aza-2′-deoxycytidine on growth inhibition of pancreatic endocrine tumour cell lines: a proteomic study. Proteomics 9(7):1952–1966

    Article  CAS  PubMed  Google Scholar 

  169. Shaker S, Bernstein M, Momparler LF, Momparler RL (2003) Preclinical evaluation of antineoplastic activity of inhibitors of DNA methylation (5-aza-2′-deoxycytidine) and histone deacetylation (trichostatin A, depsipeptide) in combination against myeloid leukemic cells. Leuk Res 27(5):437–444

    Article  CAS  PubMed  Google Scholar 

  170. Griffiths EA, Gore SD (2008) DNA methyltransferase and histone deacetylase inhibitors in the treatment of myelodysplastic syndromes. Semin Hematol 45(1):23–30

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Kirschbaum MH, Foon KA, Frankel P, Ruel C, Pulone B, Tuscano JM, Newman EM (2014) A phase 2 study of belinostat (PXD101) in patients with relapsed or refractory acute myeloid leukemia or patients over the age of 60 with newly diagnosed acute myeloid leukemia: a California Cancer Consortium Study. Leuk Lymphoma 55(10):2301–2304

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Gore SD et al (2006) Combined DNA methyltransferase and histone deacetylase inhibition in the treatment of myeloid neoplasms. Cancer Res 66(12):6361–6369

    Article  CAS  PubMed  Google Scholar 

  173. Voso MT et al (2009) Valproic acid at therapeutic plasma levels may increase 5-azacytidine efficacy in higher risk myelodysplastic syndromes. Clin Cancer Res 15(15):5002–5007

    Article  CAS  PubMed  Google Scholar 

  174. Fandy TE et al (2009) Early epigenetic changes and DNA damage do not predict clinical response in an overlapping schedule of 5-azacytidine and entinostat in patients with myeloid malignancies. Blood 114(13):2764–2773

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Gryder BE, Sodji QH, Oyelere Adegboyega K (2012) Targeted cancer therapy: giving histone deacetylase inhibitors all they need to succeed. Futur Med Chem 4(4):505–524

    Article  CAS  Google Scholar 

  176. Kato Y, Yoshimura K, Shin T, Verheul H, Hammers H, Sanni TB, Salumbides BC, Van Erp K, Schulick R, Pili R (2007) Synergistic in vivo antitumor effect of the histone deacetylase inhibitor MS-275 in combination with interleukin 2 in a murine model of renal cell carcinoma. Clin Cancer Res 13(15):4538–4546

    Article  CAS  PubMed  Google Scholar 

  177. Shen L, Ciesielski M, Ramakrishnan S, Miles KM, Ellis L, Sotomayor P, Shrikant P, Fenstermaker R, Pili R (2012) Class I histone deacetylase inhibitor entinostat suppresses regulatory T cells and enhances immunotherapies in renal and prostate cancer models. PLoS ONE 7(1):e30815

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Pili R, Salumbides B, Zhao M, Altiok S, Qian D, Zwiebel J, Carducci MA, Rudek MA (2012) Phase I study of the histone deacetylase inhibitor entinostat in combination with 13-cis retinoic acid in patients with solid tumours. Br J Cancer 106(1):77–84

    Article  CAS  PubMed  Google Scholar 

  179. Smits KM, Melotte V, Niessen HEC, Dubois L, Oberije C, Troost EGC, Starmans MHW, Boutros PC, Vooijs M, van Engeland M, Lambin P (2014) Epigenetics in radiotherapy: where are we heading? Radiother Oncol 111(2):168–177

    Article  PubMed  Google Scholar 

  180. Wachowska M, Muchowicz A, Golab J (2015) Targeting epigenetic processes in photodynamic therapy-induced anticancer immunity. Front Oncol 5:176

    Article  PubMed  PubMed Central  Google Scholar 

  181. Wang Z et al (2015) Broad targeting of angiogenesis for cancer prevention and therapy. Semin Cancer Biol 35(Suppl):S224–S243

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  182. Kuusk T, Grivas N, Bex A (2017) Antiangiogenic therapy combined with immune checkpoint blockade in renal cancer: a review. Angiogenesis. doi:10.1007/s10456-017-9550-0

  183. Ramjiawan RR, Griffioen AW, Duda DG (2017) Anti-angiogenesis for cancer revisited: Is there a role for combinations with immunotherapy? Angiogenesis. doi:10.1007/s10456-017-9552-y

  184. Hamming LC, Slotman BJ, Verheul HMW, Thijssen V (2017) The clinical application of angiostatic therapy in combination with radiotherapy: past, present, future. Angiogenesis. doi:10.1007/s10456-017-9546-9

  185. Dameron KM, Volpert OV, Tainsky MA, Bouck N (1994) Control of angiogenesis in fibroblasts by p53 regulation of thrombospondin-1. Science 265(5178):1582–1584

    Article  CAS  PubMed  Google Scholar 

  186. Volpert OV, Tolsma SS, Pellerin S, Feige JJ, Chen H, Mosher DF, Bouck N (1995) Inhibition of angiogenesis by thrombospondin-2. Biochem Biophys Res Commun 217:326–332

    Article  CAS  PubMed  Google Scholar 

  187. Benjamin D, Colombi M, Moroni C, Hall MN (2011) Rapamycin passes the torch: a new generation of mTOR inhibitors. Nat Rev Drug Discov 10(11):868–880

    Article  CAS  PubMed  Google Scholar 

  188. Heffeter P, Atil B, Kryeziu K, Groza D, Koellensperger G, Korner W, Jungwirth U, Mohr T, Keppler BK, Berger W (2013) The ruthenium compound KP1339 potentiates the anticancer activity of sorafenib in vitro and in vivo. Eur J Cancer 49:3366–3375

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Kassouf E, Tehfe MA, Florescu M, Soulieres D, Lemieux B, Ayoub J-PM, Charpentier D, Yelle L, Daigneault L, Colin P, Momparler RL, Plante I, Lassonde G, Charbonneau MR, Raynal NJ-M, Blais N (2015) Phase I and II studies of the decitabine–genistein drug combination in advanced solid tumors. J Clin Oncol 33(15_suppl):e13556

    Google Scholar 

  190. Puduvalli VK, Wu J, Yuan Y, Armstrong TS, Groves MD, Raizer JJ, Giglio P, Colman H, Peereboom DM, Walbert T, Avgeropoulos NG, Iwamoto FM, Chamberlain MC, Paleologos N, Gilbert MR (2015). Brain tumor trials collaborative bayesian adaptive Adaptive randomized phase II trial of bevacizumab plus vorinostat versus bevacizumab alone in adults with recurrent glioblastoma (BTTC-1102). J Clin Oncol 33:1

    Google Scholar 

  191. Stimson L, La Thangue NB (2009) Biomarkers for predicting clinical responses to HDAC inhibitors. Cancer Lett 280(2):177–183

    Article  CAS  PubMed  Google Scholar 

  192. Wheler JJ, Janku F, Falchook GS, Jackson TL, Fu S, Naing A, Tsimberidou AM, Moulder SL, Hong DS, Yang H, Piha-Paul SA, Atkins JT, Garcia-Manero G, Kurzrock R (2014) Phase I study of anti-VEGF monoclonal antibody bevacizumab and histone deacetylase inhibitor valproic acid in patients with advanced cancers. Cancer Chemother Pharmacol 73(3):495–501

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Plumb JA, Strathdee G, Sludden J, Kaye SB, Brown R (2000) Reversal of drug resistance in human tumor xenografts by 2′-deoxy-5-azacytidine-induced demethylation of the hMLH1 gene promoter. Cancer Res 60(21):6039–6044

    CAS  PubMed  Google Scholar 

  194. Wang L, Li H, Ren Y, Zou S, Fang W, Jiang X, Jia L, Li M, Liu X, Yuan X, Chen G, Yang J, Wu C (2016) Targeting HDAC with a novel inhibitor effectively reverses paclitaxel resistance in non-small cell lung cancer via multiple mechanisms. Cell Death Dis. doi:10.1038/cddis.2015.328

    Google Scholar 

  195. Berndsen RH, Weiss A, Abdul UK, Wong TJ, Meraldi P, Griffioen AW, Dyson PJ, Nowak-Sliwinska P (2017) Combination of ruthenium(II)-arene complex [Ru(eta6-p-cymene)Cl2(pta)] (RAPTA-C) and the epidermal growth factor receptor inhibitor erlotinib results in efficient angiostatic and antitumor activity. Sci Rep. doi:10.1038/srep43005

    Google Scholar 

  196. Yu C, Friday BB, Lai J-P, McCollum A, Atadja P, Roberts LR, Adjei AA (2007) Abrogation of MAPK and Akt signaling by AEE788 synergistically potentiates histone deacetylase inhibitor-induced apoptosis through reactive oxygen species generation. Clin Cancer Res 13(4):1140–1148

    Article  CAS  PubMed  Google Scholar 

  197. Witta SE, Gemmill RM, Hirsch FR, Coldren CD, Hedman K, Ravdel L, Helfrich B, Dziadziuszko R, Chan DC, Sugita M, Chan Z, Baron A, Franklin W, Drabkin HA, Girard L, Gazdar AF, Minna JD, Bunn PA (2006) Restoring E-cadherin expression increases sensitivity to epidermal growth factor receptor inhibitors in lung cancer cell lines. Cancer Res 66(2):944–950

    Article  CAS  PubMed  Google Scholar 

  198. Derksen PWB, Liu X, Saridin F, van der Gulden H, Zevenhoven J, Evers B, van Beijnum JR, Griffioen AW, Vink J, Krimpenfort P, Peterse JL, Cardiff RD, Berns A, Jonkers J (2006) Somatic inactivation of E-cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. Cancer Cell 10(5):437–449

    Article  CAS  PubMed  Google Scholar 

  199. Peinado H, Marin F, Cubillo E, Stark H-J, Fusenig N, Nieto MA, Cano A (2004) Snail and E47 repressors of E-cadherin induce distinct invasive and angiogenic properties in vivo. J Cell Sci 117(Pt 13):2827–2839

    Article  CAS  PubMed  Google Scholar 

  200. Chen M-C, Chen C-H, Wang J-C, Tsai A-C, Liou J-P, Pan S-L, Teng C-M (2013) The HDAC inhibitor, MPT0E028, enhances erlotinib-induced cell death in EGFR-TKI-resistant NSCLC cells. Cell Death Dis 4:e810

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Lee T-G, Jeong E-H, Kim SY, Kim H-R, Kim CH (2015) The combination of irreversible EGFR TKIs and SAHA induces apoptosis and autophagy-mediated cell death to overcome acquired resistance in EGFR T790M-mutated lung cancer. Int J Cancer 136(11):2717–2729

    Article  CAS  PubMed  Google Scholar 

  202. Witta SE, Jotte RM, Konduri K, Neubauer MA, Spira AI, Ruxer RL, Varella-Garcia M, Bunn PA, Hirsch FR (2012) Randomized phase II trial of erlotinib with and without entinostat in patients with advanced non-small-cell lung cancer who progressed on prior chemotherapy. J Clin Oncol 30(18):2248–2255

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Gray JE, Haura E, Chiappori A, Tanvetyanon T, Williams CC, Pinder-Schenck M, Kish JA, Kreahling J, Lush R, Neuger A, Tetteh L, Akar A, Zhao X, Schell MJ, Bepler G, Altiok S (2014) A phase i, pharmacokinetic, and pharmacodynamic study of panobinostat, an HDAC inhibitor, combined with erlotinib in patients with advanced aerodigestive tract tumors. Clin Cancer Res 20(6):1644–1655

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Loges S, Schmidt T, Carmeliet P (2010) Mechanisms of resistance to anti-angiogenic therapy and development of third-generation anti-angiogenic drug candidates. Genes Cancer 1(1):12–25

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Wang Z et al (2015) Broad targeting of angiogenesis for cancer prevention and therapy. Semin Cancer Biol 35(Suppl):S224–S243

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  206. Ding X, Liu W, Weiss A, Li Y, Wong I, Griffioen AW, van den Bergh H, Xu H, Nowak-Sliwinska P, Ho C-M (2014) Discovery of a low order drug-cell response surface for applications in personalized medicine. Phys Biol 11(6):65003

    Article  Google Scholar 

  207. Treps L, Conradi L-C, Harjes U, Carmeliet P (2016) Manipulating angiogenesis by targeting endothelial metabolism: hitting the engine rather than the drivers—a new perspective? Pharmacol Rev 68(3):872–887

    Article  PubMed  Google Scholar 

  208. Weiss A, Berndsen RH, Ding X, Ho C-M, Dyson PJ, van den Bergh H, Griffioen AW, Nowak-Sliwinska P (2015) A streamlined search technology for identification of synergistic drug combinations. Sci Rep 5:14508

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Yang Q, Tian Y, Liu S, Zeine R, Chlenski A, Salwen HR, Henkin J, Cohn SL (2007) Thrombospondin-1 peptide ABT-510 combined with valproic acid is an effective antiangiogenesis strategy in neuroblastoma. Cancer Res 67(4):1716–1724

    Article  CAS  PubMed  Google Scholar 

  210. Pili R, Kruszewski MP, Hager BW, Lantz J, Carducci MA (2001) Combination of phenylbutyrate and 13-cis retinoic acid inhibits prostate tumor growth and angiogenesis. Cancer Res 61(4):1477–1485

    CAS  PubMed  Google Scholar 

  211. Wang H, Zhou H, Zou Y, Liu Q, Guo C, Gao G, Shao C, Gong Y (2010) Resveratrol modulates angiogenesis through the GSK3β/β-catenin/TCF-dependent pathway in human endothelial cells. Biochem Pharmacol 80(9):1386–1395

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the European Research Council (ERC-StG-2015-680209 to PNS) and the Dutch Cancer Society (VU2014-7234 to AWG and PNS) for financial support.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Patrycja Nowak-Sliwinska.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Berndsen, R.H., Abdul, U.K., Weiss, A. et al. Epigenetic approach for angiostatic therapy: promising combinations for cancer treatment. Angiogenesis 20, 245–267 (2017). https://doi.org/10.1007/s10456-017-9551-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10456-017-9551-z

Keywords

Navigation