We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

The combination of histone deacetylase inhibitors and radiotherapy: a promising novel approach for cancer treatment

    Ekta Shirbhate

    Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur-495 009, Chhattisgarh, India

    ,
    Preeti Patel

    Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur-495 009, Chhattisgarh, India

    ,
    Vijay K Patel

    Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur-495 009, Chhattisgarh, India

    ,
    Ravichandran Veerasamy

    Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia

    ,
    Prabodh C Sharma

    Institute of Pharmaceutical Sciences, Kurukshetra University, Kurukshetra-136 119, Haryana, India

    &
    Harish Rajak

    *Author for correspondence:

    E-mail Address: harishdops@yahoo.co.in

    Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur-495 009, Chhattisgarh, India

    Published Online:https://doi.org/10.2217/fon-2020-0385

    HDAC inhibitors (HDACi) play an essential role in various cellular processes, such as differentiation and transcriptional regulation of key genes and cytostatic factors, cell cycle arrest and apoptosis that facilitates the targeting of epigenome of eukaryotic cells. In the majority of cancers, only a handful of patients receive optimal benefit from chemotherapeutics. Additionally, there is emerging interest in the use of HDACi to modulate the effects of ionizing radiations. The use of HDACi with radiotherapy, with the goal of reaching dissimilar, often distinct pathways or multiple biological targets, with the expectation of synergistic effects, reduced toxicity and diminished intrinsic and acquired resistance, conveys an approach of increasing interest. In this review, the clinical potential of HDACi in combination with radiotherapy is described as an efficient synergy for cancer treatment will be overviewed.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Park SY, Kim JS. A short guide to histone deacetylases including recent progress on class II enzymes. Exp. & Mol. Med. 52(2), 204–212 (2020). • This paper highlighted the acetylation and deacetylation mechanism of histone proteins in gene expression and translational modifications.
    • 2. Gennaro ED, Bruzzese F, Caraglia M, Abruzzese A, Budillon A. Acetylation of proteins as novel target for antitumor therapy: review article. Amino Acids 26(4), 435–441 (2004).
    • 3. Vahid F, Zand H, Mirshekarlou EN, Najafi R, Hekmatdoost A. The role dietary of bioactive compounds on the regulation of histone acetylases and deacetylases: a review. Gene 562(1), 8–15 (2015).
    • 4. Rajak H, Singh A, Raghuwanshi K et al. A structural insight into hydroxamic acid based histone deacetylase inhibitors for the presence of anticancer activity. Curr. Med. Chem. 21(23), 2642–2664 (2014).
    • 5. Minucci S, Pelicci PG. Histone deacetylase inhibitors and the promise of an epigenetic (and more) treatments for cancer. Nature Rev. Cancer 6(1), 38–51 (2006).
    • 6. Taunton J, Hassig CA, Schreiber SL. A mammalian histone deacetylase related to the yeast transcriptional regulator Rpd3p. Science 272(5260), 408–409 (1996).
    • 7. Mahlknecht U, Hoelzer D. Histone acetylation modifiers in the pathogenesis of malignant disease. Mol. Med. 6(8), 623–624 (2000). • This paper emphasized brief classification of HDAC enzymes and inhibitors and their role in cancer treatment.
    • 8. Watson PJ, Fairall L, Santos GM, Schwabe JW. Structure of HDAC3 bound to co-repressor and inositol tetraphosphate. Nature 481, 335–340 (2012).
    • 9. Muslin AJ, Xing H. 14-3-3 proteins: regulation of subcellular localization by molecular interference. Cell Signal 12, 703–709 (2000).
    • 10. Gao L, Cueto MA, Asselbergs F, Atadja P. Cloning and functional characterization of HDAC11, a novel member of the human histone deacetylasefamily. J. Biol. Chem. 277, 25748–25755 (2002).
    • 11. Patel P, Singh A, Patel VK et al. Pharmacophore based 3D-QSAR, virtual screening and docking studies on novel series of HDAC inhibitors with thiophene linker as anticancer agents. Comb. Chem. & High Throughput Screen. 19(9), 735–751 (2016).
    • 12. Brachmann CB, Sherman JM, Devine SE, Cameron EE, Pillus L, Boeke JD. The SIR2 gene family, conserved from bacteria to humans, functions in silencing, cell cycle progression, and chromosome stability. Genes Dev. 9(23), 2888–2902 (1995).
    • 13. National Cancer Institute. Types of cancer treatment. www.cancer.gov/about-cancer/treatment/types
    • 14. Yang F, Zhao N, Ge D, Chen Y. Next-generation of selective histone deacetylase inhibitors. RSC Adv. 9, 19571–19583 (2019).
    • 15. Wagner FF, Weaver M, Lewis MC, Holson EB. Small molecules inhibitors of zinc-dependent histone deacetylases. Neurotherapeutics 10, 589–604 (2013).
    • 16. Cappellacci L, Perinelli DR, Maggi F, Grifantini M, Petrelli R. Recent progress in histone deacetylase inhibitors as anticancer agents. Curr. Med. Chem. 27(15), 2449 –2493 (2020).
    • 17. Day JA, Cohen SM. Investigating the selectivity of metalloenzyme inhibitors. J. Med. Chem. 56, 7997–8007 (2013).
    • 18. Eckschlager T, Plch J, Stiborova M, Hrabeta J. Histone deacetylase inhibitors as anticancer drugs. Int. J. Mol. Sci. 18(7), 1414 (2017).
    • 19. Mann BS, Johnson JR, Cohen MH, Justice R, Pazdur R. FDA approval summary: Vorinostat for treatment of advanced primary cutaneous T-cell lymphoma. Oncologist 12(10), 1247–1252 (2007).
    • 20. Grant C, Rahman F, Piekarz R et al. Romidepsin: A new therapy for cutaneous T-cell lymphoma and a potential therapy for solid tumors. Expert Rev. Anticancer Ther. 11, 1622 (2011).
    • 21. Chien W, Lee DH, Zheng Y et al. Growth inhibition of pancreatic cancer cells by histone deacetylase inhibitor Belinostat through suppression of multiple pathways including HIF, NFkB, and mTOR signaling in vitro and in vivo. Mol. Carcinog. 53, 722–735 (2014).
    • 22. Sivaraj D, Green MM, Gasparetto C. Panobinostat for the management of multiple myeloma. Future Oncol. 13, 477–488 (2017).
    • 23. Arrowsmith CH, Bountra C, Fish PV, Lee K, Schapira M. Epigenetic protein families: a new frontier for drug discovery. Nat. Rev. Drug Discovery 11, 384–400 (2012).
    • 24. Shirbhate E, Sahu D, Patel P et al. Searching for potential HDAC2 inhibitors: structure-activity relationship studies on indole-based hydroxamic acids as an anticancer agent. Lett. Drug Des. Discov. 17(7), 905–917 (2020).
    • 25. Carafa V, Miceli M, Altucci L, Nebbioso A. Histone deacetylase inhibitors: a patent review (2009–2011). Expert Opin. Ther. Pat. 23, 1–17 (2013).
    • 26. Laubach JP, San-Miguel JF, Hungria V et al. Deacetylase inhibitors: An advance in myeloma therapy? Expert Rev. Hematol. 10(3), 229–237 (2017).
    • 27. Micelli C, Rastelli G. Histone deacetylases: structural determinants of inhibitor selectivity. Drug Discov. Today 20, 718–735 (2015).
    • 28. Juliandi B, Abematsu M, tsukasa S, Tsujimura K, Smith A, Nakashima K. Induction of superficial cortical layer neurons from mouse embryonic stem cells by valproic acid. Neurosci. Res. 72(1), 23–31 (2012).
    • 29. Zhan P, Itoh Y, Suzuki T, Liu X. Strategies for the discovery of target-specific or isoform-selective modulators. J. Med. Chem. 58, 7611–7633 (2015). •• The paper revealed currently available rational design tactics for acquiring class- and isoform-selective inhibitors.
    • 30. Pommier Y, Sordet O, Antony S, Hayward RL, Kohn KW. Apoptosis defects and chemotherapy resistance: molecular interaction maps and networks. Oncogene 23(16), 2934–2949 (2004).
    • 31. Powis G, Mustacich D, Coon A. The role of the redox protein thioredoxin in cell growth and cancer. Free Radic. Biol. Med. 29(3), 312–322 (2000).
    • 32. Chung YM, Yoo YD, Park JK, Kim YT, Kim HJ. Increased expression of peroxiredoxin II confers resistance to cisplatin. Anticancer Res. 21(2A), 1129–1133 (2001).
    • 33. Robey RW, Chakraborty AR, Basseville A et al. Histone deacetylase inhibitors: emerging mechanisms of resistance. Mol. Pharm. 8(6), 2021–2031 (2011).
    • 34. Fantin VR, Richon VM. Mechanisms of resistance to histone deacetylase inhibitors and their therapeutic implications. Clin. Cancer Res. 13(24), 7237–7242 (2007).
    • 35. Nolan L, Johnson PW, Ganesan A, Packham G, Crabb SJ. Will histone deacetylase inhibitors require combination with other agents to fulfil their therapeutic potential? Br. J. Cancer 99(5), 689–694 (2008).
    • 36. Lindemann RK, Gabrielli B, Johnstone RW. Histone-deacetylase inhibitors for the treatment of cancer. Cell Cycle 3(6), 777–786 (2004).
    • 37. Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat. Rev. Drug Discov. 5(9), 769–784 (2006).
    • 38. Suraweera A, O'Byrne KJ, Richard DJ. Combination therapy with histone deacetylase inhibitors (HDACi) for the treatment of cancer: achieving the full therapeutic potential of HDACi. Front. Oncol. 8, 1–15 (2018).
    • 39. Chinnaiyan P, Vallabhaneni G, Armstrong E, Huang SM, Harari PM. Modulation of radiation response by histone deacetylase inhibition. Int. J. Radiat. Oncology Biol. Phys. 62(1), 223–229 (2005).
    • 40. Delaney G, Jacob S, Featherstone C, Barton M. The role of radiotherapy in cancer treatment:estimating optimal utilization from a review of evidence-based clinical guidelines. Cancer 104, 1129–1137 (2005).
    • 41. O'Sullivan B, Waldron J. Principles in radiation oncology. Wiley, NY, USA (2004).
    • 42. Lyubimova NV, Coultas PG, Yuen K, Martin RF. In vivo radioprotection of mouse brainendothelial cells by Hoechst 33342. Br. J. Radiol. 74, 77–82 (2001).
    • 43. Camphausen K, Tofilon PJ. Inhibition of histone deacetylation: a strategy for tumor radiosensitization. J. Clin. Oncol. 25, 4051–4060 (2007).
    • 44. Camphausen K, Cerna D, Scott T et al. Enhancement of in vitro and in vivo tumor cell radiosensitivity by valproic acid. Int. J. Cancer 114(3), 380–386 (2005).
    • 45. Camphausen K, Burgan W, Cerra M et al. Enhanced radiation-induced cell killing and prolongation of gamma H2AX foci expression by the histone deacetylase inhibitor MS-275. Cancer Res. 64(1), 316–321 (2004).
    • 46. Kim JH, Shin JH, Kim IH. Susceptibility and radiosensitization of human glioblastoma cells to trichostatin A, a histone deacetylase inhibitor. Int. J. Radiat. Oncol. Biol. Phys. 59, 1174–1180 (2004).
    • 47. Zhang Y, Jung M, Dritschilo A, Jung M. Enhancement of radiation sensitivity of human squamous carcinoma cells by histone deacetylase inhibitors. Radiat. Res. 161(6), 667–674 (2004).
    • 48. Geng L, Cuneo KC, Fu A, Tu T, Atadja PW, Hallahan DE. Histone deacetylase (HDAC) inhibitor LBH589 increases duration of gamma-H2AX foci and confines HDAC4 to the cytoplasm in irradiated non-small cell lung cancer. Cancer Res. 66(23), 11298–11304 (2006).
    • 49. Folkvord S, Ree AH, Furre T, Halvorsen T, Flatmark K. Radiosensitization by SAHA in experimental colorectal carcinoma models – in vivo effects and relevance of histone acetylation status. Int. J. Radiat. Oncol. Biol. Phys. 74(2), 546–552 (2009).
    • 50. Chen X, Wong P, Radany E, Wong JYC. HDAC inhibitor, valproic acid, induces p53-dependent radiosensitization of colon cancer cells. Cancer Biother. Radiopharm. 24(6), 689–699 (2009).
    • 51. Camphausen K, Scott T, Sproull M, Tofilon PJ. Enhancement of xenograft tumor radiosensitivity by the histone deacetylase inhibitor MS-275 and correlation with histone hyperacetylation. Clin. Cancer Res. 10(18), 6066–6071 (2004).
    • 52. Munshi A, Kurland JF, Nishikawa T et al. Histone deacetylase inhibitors radiosensitize human melanoma cells by suppressing DNA repair activity. Clin. Cancer Res. 11(13), 4912–4922 (2005).
    • 53. Baschnagel A, Russo A, Burgan WE et al. Vorinostat enhances the radiosensitivity of a breast cancer brain metastatic cell line grown in vitro and as intracranial xenografts. Mol. Cancer Ther. 8(6), 1589–1595 (2009).
    • 54. Dong Q, Sharma S, Liu H et al. HDAC inhibitors reverse acquired radio resistance of KYSE-150R esophageal carcinoma cells by modulating Bmi-1 expression. Toxicology Lett. 224, 121–129 (2014).
    • 55. Moertl S, Payer S, Kell R, Winkler K, Anastasov N, Atkinson MJ. Comparison of radiosensitization by HDAC inhibitors CUDC-1010 and SAHA in pancreatic cancer cells. Int. J. Mol. Sci. 20, 3259 (2019).
    • 56. Rogakou EP, Pilch DR, Orr AH, Ivanova VS, Bonner WM. DNA-double strand breaks induce histone H2AX phosphorylation on serine 139. J. Biol. Chem. 273(10), 5858–5868 (1998).
    • 57. Munshi A, Tanaka T, Hobba ML, Tucker SL, Richon VM, Meyn RE. Vorinostat, a histone deacetylase inhibitor enhances the response of human tumor cells to ionizing radiation through prolongation of gamma-H2AX foci. Mol. Cancer Ther. 5(8), 1967–1974 (2006).
    • 58. Zhang Y, Adachi M, Zhao X, Kawamura R, Imai K. Histone decetylase inhibitors FK228,N-(2-aminophenyl)-4-[N-(pyridine-3-ylmethoxycarbonyl)amino-methyl]benzamide and m-carboxycinnamic acid bishydroxamide augment radiation-induced cell death in gastrointestinal adenocarcinoma cells. Int. J. Cancer 110(2), 301–380 (2004).
    • 59. Shabason JE, Tofilon PJ, Camphausen K. Grand rounds at the National Institutes of Health: HDAC inhibitors as radiation modifiers, from bench to clinic. J. Cell. Mol. Med. 15, 2735–2744 (2011).
    • 60. Blattmann C, Oertel S, Ehemann V et al. Enhancement of radiation response in osteosarcoma and rhabdomyosarcoma cell lines by histone deacetylase inhibition. Int. J. Radiat. Oncol. Biol. Phys. 78(1), 237–245 (2010).
    • 61. Kao GD, McKenna WG, Guenther MG, Muschel RJ, Lazar MA, Yen TJ. Histone deacetylase 4 interacts with 53BP1 to mediate the DNA damage response. J. Cell Biol. 160, 1017–1027 (2003).
    • 62. Storch K, Eke I, Borgmann K et al. Three-dimensional cell growth confers radioresistance by chromatin density modification. Cancer Res. 70(10), 3925–3934 (2010).
    • 63. Groselj B, Sharma NL, Hamdy FC, Kerr M, Kiltie AE. Histone deacetylase inhibitors as radiosensitizers: effects on DNA damage signaling and repair. Br. J. Cancer 108, 748–754 (2013).
    • 64. Ree AH, Dueland S, Folkvord S et al. Vorinostat, a histone deacetylase inhibitor, combined with pelvic palliative radiotherapy for gastrointestinal carcinoma: The Pelvic Radiation and Vorinostat (PRAVO) phase 1 study. Lancet Oncol. 11(5), 459–464 (2010).
    • 65. Thurn KT, Thomas S, Moore A, Munster PN. Rational therapeutic combinations with histone deacetylase inhibitors for the treatment of cancer. Future Oncol. 7(2), 263–283 (2011). •• This paper demonstrated the rationale and clinical progress of various combinations of HDACi with other chemotherapeutic drugs along with future therapeutic uses of HDACi for treatment of cancer.
    • 66. Kuribayashi T, Ohar M, Sora S, Kubota N. Scriptaid, a novel histonedeacetylase inhibitor, enhances the response of human tumor cells to radiation. Int. J. Mol. Med. 25(1), 25–29 (2010).
    • 67. Zhang Y, Manfred J, Dritschiloa A, Junga M. Enhancement of radiation sensitivity of human squamous carcinoma cells by histone deacetylase inhibitors. Rad. Res. 161, 667–674 (2004).
    • 68. Xaio W, Graham PH, Hao J et al. Combination therapy with the histone deacetylase inhibitor LBH589 and radiation is an effective regimen for prostate cancer cells. PLoS One 8(8), 1–14 (2013).
    • 69. Groselj B, Ruan JL, Scott H et al. Radiosensitization in vivo by histone deacetylase inhibition with no increase in early normal tissue radiation toxicity. Mol. Cancer Ther. 17(2), 381–392 (2018).
    • 70. Santini V, Gozzini A, Ferrari G. Histone deacetylase inhibitors: molecular and biological activity as a premise to clinical application. Curr. Drug Met. 8, 383–394 (2007).
    • 71. Banuelos CA, Banath JP, MacPhail SH, Zhao J, Reitsema T, Olive PL. Radiosensitization by the histone deacetylase inhibitor PCI-24781. Clin. Cancer Res. 13(22), 6816–6825 (2007).
    • 72. Chen X, Wong P, Radany E, Wong J. HDAC inhibitor, valproic acid, induces p53-dependent radiosensitization of colon cancer cells. Cancer Biother. Radiopharm. 24(6), 689–699 (2009).
    • 73. Hosein AN, Lim YC, Day B et al. The effect of valproic acid in combination with irradiation and temozolomide on primary human Glioblastoma cells. J. Neurooncol. 122(2), 263–271 (2015).
    • 74. Tofilon PJ, Camphausen KA. HDAC inhibitors in cancer care. Oncol. J. 24, 1–9 (2010).
    • 75. Noguchi H, Yamashita H, Murakami T et al. Successful treatment of anaplastic thyroid carcinoma with a combination of oral valproic acid, chemotherapy, radiation and surgery. Endocrine J. 56(2), 245–249 (2009).
    • 76. Karagiannis TC, El-Osta A. Review: modulation of cellular radiation responses by histone deacetylase inhibitors. Oncogene 25, 3885–3893 (2006).
    • 77. Suraweera A, O'Byrne KJ, Richard DJ. Combination therapy with histone deacetylase inhibitors (HDACi) for the treatment of cancer: achieving the full therapeutic potential of HDACi. Front. Oncol. 8, 92 (2018). •• This paper demonstrated the combination of HDACi with other anticancer agents and their possibilities for conversion into preclinical and clinical investigations.
    • 78. Miller CP, Singh MM, Rivera-Del NV, Manton CA, Chandra J. Therapeutic strategies to enhance the anticancer efficacy of histone deacetylase inhibitors. J. Biomed. Biotech. doi:10.1155/2011/514261 (2011).
    • 79. Karagiannis T, El-Osta A. Clinical potential of histone deacetylase inhibitors as standalone therapeutics and in combination with other chemotherapeutics or radiotherapy for cancer. Epigenetics 1(3), 121–126 (2006).
    • 80. Zhang F, Zhang T, Teng ZH, Zhang R, Wang JB, Mei QB. Sensitization to γ-irradiation induced cell cycle arrest and apoptosis by the histone deacetylase inhibitor trichostatin A in non-small cell lung cancer (NSCLC) cells. Cancer Biol. Ther. 8(9), 823–831 (2009).
    • 81. Choi CYH, Wakelee HA, Neal JW et al. Vorinostat and concurrent stereotactic radiosurgery for non–small cell lung cancer brain metastases: A phase 1 dose escalation trial. Int. J. Radiat. Oncol. Biol. Phys. 99(1), 16–21 (2017).
    • 82. Perona M, Thomasz L, Rossich L et al. Radiosensitivity enhancement of human thyroid carcinoma cells by the inhibitors of histone deacetylase sodium butyrate and valproic acid. Mol. Cell Endrocrinol. 478, 141–150 (2018).
    • 83. Paillas S, Then CK, Kilgas S et al. The histone deacetylase inhibitor Romidepsin spares normal tissues while acting as an effective radiosensitizer in bladder tumors in vivo. Int. J. Radiat. Oncol. Biol. Phys. 107(1), 212–221 (2020).
    • 84. Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Drug Discov. 5, 769–784 (2006).
    • 85. Johnstone RW. Histone-deacetylase inhibitors: Novel drugs for the treatment of cancer. Drug Discov. 1, 287–299 (2002).
    • 86. Rosse G. Novel and selective inhibitors of histone deacetylase. ACS Med. Chem. Lett. 3, 879–880 (2012).
    • 87. Sharda A, Rashid M, Shah SG et al. Elevated HDAC activity and altered histone phosphoacetylation confer acquired radio-resistant phenotype to breast cancer cells. Clin Epigenetics 12(1), 4 (2020).
    • 88. Zhang M, Hu C, Moses N et al. HDAC6 regulates DNA damage response via deacetylating MLH1. J. Biol. Chem. 294(15), 5813–5826 (2019).
    • 89. Sferra R, Pompili S, Festuccia C et al. the possible prognostic role of histone deacetylase and transforming growth factor factor β/Smad signaling in high grade gliomas treated by radio-chemotherapy: a preliminary immunohistochemical study. Eur. J. Histochem. 61(2), 86–95 (2017).
    • 90. Marampon F, Megiomi F, Camero S et al. HDAC4 and HDAC6 sustain DNA double strand break repair and stem-like phenotype by promoting radio resistance in glioblastoma cells. Cancer Lett. 397, 1–11 (2017).
    • 91. Yang W, Liu Yingying, Gao R, Tu H, Sun T. HDAC6 inhibition induces glioma stem cells differentiation and enhances radiation sensitivity through the SHH/Gli1 signaling pathway. Cancer Lett. 415, 164–176 (2018)
    • 92. Oronsky B, Scicinski J, Kim MM et al. Turning on the radio: epigenetic inhibitors as potential radiopromising agents. Biomol. 6, 32 (2016).