318
Views
25
CrossRef citations to date
0
Altmetric
Reviews

Targeting histone deacetylase in thyroid cancer

, , , , , & show all
Pages 179-193 | Published online: 13 Dec 2012

Bibliography

  • SEER Stat Fact Sheets: thyroid. Available from: http://seer.cancer.gov/statfacts/html/thyro.html [Last accessed 19 July 2012]
  • Eheman C, Henley SJ, Ballard-Barbash R, Annual Report to the Nation on the status of cancer, 1975-2008, featuring cancers associated with excess weight and lack of sufficient physical activity. Cancer 2012;118:2338-66
  • Hall SF, Walker H, Siemens R, Increasing detection and increasing incidence in thyroid cancer. World J Surg 2009;33:2567-71
  • Enewold L, Zhu K, Ron E, Rising thyroid cancer incidence in the United States by demographic and tumor characteristics, 1980-2005. Cancer Epidemiol Biomarkers Prev 2009;18:784-91
  • Cooper DS, Doherty GM, Haugen BR, Revised American Thyroid Association management guidelines for patients with thyroid nodules and differentiated thyroid cancer. Thyroid 2009;19:1167-214
  • Durante C, Haddy N, Baudin E, Long-term outcome of 444 patients with distant metastases from papillary and follicular thyroid carcinoma: benefits and limits of radioiodine therapy. J Clin Endocrinol Metab 2006;91:2892-9
  • Schlumberger M, Lacroix L, Russo D, Defects in iodide metabolism in thyroid cancer and implications for the follow-up and treatment of patients. Nat Clin Pract Endocrinol Metab 2007;3:260-9
  • Arturi F, Russo D, Giuffrida D, Sodium-Iodide symporter (NIS) gene expression in lymph node metastases of papillary thyroid carcinomas. Eur J Endocrinol 2000;143:623-7
  • Schlumberger M, Sherman SI. Approach to the patient with advanced differentiated thyroid cancer. Eur J Endocrinol 2012;166:5-11
  • Puxeddu E, Romagnoli S, Dottorini ME. Targeted therapies for advanced thyroid cancer. Curr Opin Oncol 2011;23(1):13-21
  • Ellis LM, Hicklin DJ. Resistance to targeted therapies: refining anticancer therapy in the era of molecular oncology. Clin Cancer Res 2009;15:7471-8
  • Pasquali D, Santoro A, Bufo P, Upregulation of endocrine gland-derived vascular endothelial growth factor in papillary thyroid cancers displaying infiltrative patterns, lymph node metastases, and BRAF mutation. Thyroid 2011;21:391-9
  • Russo D, Damante G, Puxeddu E, Epigenetics of thyroid cancer and novel therapeutic targets. J Mol Endocrinol 2011;46:R73-81
  • Feinberg AP, Ohlsson R, Henikoff S. The epigenetic progenitor origin of human cancer. Nat Rev Genet 2006;7(1):21-33
  • Kouzarides T. Chromatin modifications and their function. Cell 2007;128:693-705
  • Chi P, Allis CD, Wang GG. Covalent histone modifications-miswritten, misinterpreted and mis-erased in human cancers. Nat Rev Cancer 2010;10:457-9
  • Berger SL. The complex language of chromatin regulation during transcription. Nature 2007;447(7143):407-12
  • Yang XJ. The diverse superfamily of lysine acetyltransferases and their roles in leukemia and other diseases. Nucleic Acids Res 2004;32:959-76
  • Carew JS, Giles FJ, Nawrocki ST. Histone deacetylase inhibitors: mechanisms of cell death and promise in combination cancer therapy. Cancer Lett 2008;269:7-17
  • Glozak MA, Sengupta N, Zhang X, Seto E. Acetylation and deacetylation of non-histone proteins. Gene 2005;363:15-23
  • Turner BM. Histone acetylation and an epigenetic code. Bioassays 2000;22:836-45
  • Spiegel S, Milstien S, Grant S. Endogenous modulators and pharmacological inhibitors of histone deacetylases in cancer therapy. Oncogene 2012;31(5):537-51
  • Rajendran P, Williams DE, Ho E, Dashwood RH. Metabolism as a key to histone deacetylase inhibition. Crit Rev Biochem Mol Biol 2011;46(3):181-99
  • McKenna NJ, Xu J, Nawaz Z, Nuclear receptor coactivators: multiple enzymes, multiple complexes, multiple functions. J Steroid Biochem Mol Biol 1999;69(1-6):3-12
  • Wu Y, Koenig RJ. Gene regulation by thyroid hormone. Trends Endocrinol Metab 2000;11(6):207-11
  • Miremadi A, Oestergaard MZ, Pharoah PD, Caldas C. Cancer genetics of epigenetic genes. Hum Mol Genet 2007;16(Spec No 1):R28-49
  • Ozdag H, Teschendorff AE, Ahmed AA, Differential expression of selected histone modifier genes in human solid cancers. BMC Genomics 2006;7:90
  • Seligson DB, Horvath S, Shi T, Global histone modification patterns predict risk of prostate cancer recurrence. Nature 2005;435:1262-6
  • Mosashvilli D, Kahl P, Mertens C, Global histone acetylation levels: prognostic relevance in patients with renal cell carcinoma. Cancer Sci 2010;101:2664-9
  • Manzo F, Tambaro FP, Mai A, Altucci L. Histone acetyltransferase inhibitors and preclinical studies. Exp Opin Ther Pat 2009;19(6):761-74
  • Dickinson M, Johnstone RW, Prince HM. Histone deacetylase inhibitors: potential targets responsible for their anti-cancer effect. Invest New Drugs 2010;28(Suppl 1):S3-20
  • Santo L, Hideshima T, Kung AL, Preclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective HDAC6 inhibitor, ACY-1215, in combination with bortezomib in multiple myeloma. Blood 2012;119(11):2579-89
  • Hubbert C, Guardiola A, Shao R, HDAC6 is a microtubule-associated deacetylase. Nature 2002;417:455-8
  • Li G, Jiang H, Chang M, HDAC6 alpha-tubulin deacetylase: a potential therapeutic target in neurodegenerative diseases. J Neurol Sci 2011;304(1-2):1-8
  • Aldana-Masangkay GI, Sakamoto KM. The role of HDAC6 in cancer. J Biomed Biotechnol 2011;2011:875824
  • www.clinicaltrials.gov
  • Wong ST. Emerging treatment combinations: integrating therapy into clinical practice. Am J Health Syst Pharm 2009;66(23 Suppl 6):S9-S14
  • Jagannath S, Dimopoulos MA, Lonial S. Combined proteasome and histone deacetylase inhibition: a promising synergy for patients with relapsed/refractory multiple myeloma. Leuk Res 2010;34:1111-18
  • Prince HM, Bishton MJ, Harrison SJ. Clinical studies of histone deacetylase inhibitors. Clin Cancer Res 2009;15:3958-69
  • Dovey OM, Foster CT, Cowley SM. Emphasizing the positive: a role for histone deacetylases in transcriptional activation. Cell Cycle 2010;9:2700-1
  • Shen F, Kirmani KZ, Xiao Z, Nuclear protein isoforms: implications for cancer diagnosis and therapy. J Cell Biochem 2011;112:756-60
  • Gunderson FQ, Johnson TL. Acetylation by the transcriptional coactivator Gcn5 plays a novel role in co-transcriptional spliceosome assembly. PLoS Genet 2009;5:e1000682
  • Scroggins BT, Robzyk K, Wang D, An acetylation site in the middle domain of Hsp90 regulates chaperone function. Mol Cell 2007;25:151-9
  • Yang MH, Nickerson S, Kim ET, Regulation of RAS oncogenicity by acetylation. Proc Natl Acad Sci USA 2012;109(27):10843-8
  • Nakata S, Yoshida T, Horinaka M, Histone deacetylase inhibitors upregulate death receptor 5/TRAIL-R2 and sensitize apoptosis induced by TRAIL/APO2-L in human malignant tumor cells. Oncogene 2004;23:6261-71
  • Nebbioso A, Clarke N, Voltz E, Tumor-selective action of HDAC inhibitors involves TRAIL induction in acute myeloid leukemia cells. Nat Med 2005;11:77-84
  • Zhang XD, Gillespie SK, Borrow JM, Hersey P. The histone deacetylase inhibitor suberic bishydroxamate regulates the expression of multiple apoptotic mediators and induces mitochondria-dependent apoptosis of melanoma cells. Mol Cancer Ther 2004;3:425-35
  • Ocker M, Schneider-Stock R. Histone deacetylase inhibitors: signalling towards p21cip1/waf1. Int J Biochem Cell Biol 2007;39:1367-74
  • Sandor V, Senderowicz A, Mertins S, P21-dependent g(1)arrest with downregulation of cyclin D1 and upregulation of cyclin E by the histone deacetylase inhibitor FR901228. Br J Cancer 2000;83:817-25
  • Yu X, Guo ZS, Marcu MG, Modulation of p53, ErbB1, ErbB2, and Raf-1 expression in lung cancer cells by depsipeptide FR901228. J Natl Cancer Inst 2002;94:504-13
  • Oh ET, Park MT, Choi BH, Novel histone deacetylase inhibitor CG200745 induces clonogenic cell death by modulating acetylation of p53 in cancer cells. Invest New Drugs 2012;30:435-42
  • Groenendyk J, Sreenivasaiah PK, Kim do H, Biology of endoplasmic reticulum stress in the heart. Circ Res 2010;107:1185-97
  • Kawaguchi Y, Kovacs JJ, McLaurin A, The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 2003;115:727-38
  • Hideshima T, Bradner JE, Wong J, Small-molecule inhibition of proteasome and aggresome function induces synergistic antitumor activity in multiple myeloma. Proc Natl Acad Sci USA 2005;102:8567-72
  • Rosato RR, Almenara JA, Grant S. The histone deacetylase inhibitor MS-275 promotes differentiation or apoptosis in human leukemia cells through a process regulated by generation of reactive oxygen species and induction of p21CIP1/WAF1. Cancer Res 2003;63:3637-45
  • Robert T, Vanoli F, Chiolo I, HDACs link the DNA damage response, processing of double-strand breaks and autophagy. Nature 2011;471:74-9
  • Kramer OH, Baus D, Knauer SK, Acetylation of Stat1 modulates NF-kappaB activity. Genes Dev 2006;20:473-85
  • Brest P, Lassalle S, Hofman V, MiR-129-5p is required for histone deacetylase inhibitor-induced cell death in thyroid cancer cells. Endocr Rel Cancer 2011;18:711-19
  • Barneda-Zahonero B, Parra M. Histone deacetylases and cancer. Mol Oncol 2012; Epub ahead of print
  • Johnstone RV. Histone-deacetylase inhibitors: novel drugs for the treatment of cancer. Nat Rev Drug Discov 2002;1:287-9
  • Kondo T, Asa SL, Ezzat S. Epigenetic dysregulation in thyroid neoplasia. Endocrinol Metab Clin North Am 2008;37:389-400
  • Puppin C, Passon N, Lavarone E, Levels of histone acetylation in thyroid tumors. Biochem Biophys Res Commun 2011;411:679-83
  • Schweppe RE, Klopper JP, Korch C, Deoxyribonucleic acid profiling analysis of 40 human thyroid cancer cell lines reveals cross-contamination resulting in cell line redundancy and misidentification. J Clin Endocrinol Metab 2008;93:4331-41
  • Zarnegar R, Brunaud L, Kanauchi H, Increasing the effectiveness of radioactive iodine therapy in the treatment of thyroid cancer using Trichostatin A, a histone deacetylase inhibitor. Surgery 2002;132:984-90
  • Yuan GB, Kuang AR, Fan Q, Combined effects of all-trans-retinoic acid and trichostatin A on the induction of differentiation of thyroid carcinoma cells. Chin J Cancer 2010;29:379-84
  • Kitazono M, Bates S, Fok P, The histone deacetylase inhibitor FR901228 (depsipeptide) restores expression and function of pseudo-null p53. Cancer Biol Ther 2002;1:665-8
  • Copland JA, Marlow LA, Williams SF, Molecular diagnosis of a BRAF papillary thyroid carcinoma with multiple chromosome abnormalities and rare adrenal and hypothalamic metastases. Thyroid 2006;16:1293-302
  • Imanishi R, Ohtsuru A, Iwamatsu M, A histone deacetylase inhibitor enhances killing of undifferentiated thyroid carcinoma cells by p53 gene therapy. J Clin Endocrinol Metab 2002;87(10):4821-4
  • Marlow LA, Reynolds LA, Cleland AS, Reactivation of suppressed RhoB is a critical step for the inhibition of anaplastic thyroid cancer growth. Cancer Res 2009;69:1536-44
  • Mitsiades CS, Poulaki V, McMullan C, Novel histone deacetylase inhibitors in the treatment of thyroid cancer. Clin Cancer Res 2005;11:3958-65
  • Clinckspoor I, Verlinden L, Overbergh L, 1,25-dihydroxyvitamin D3 and a superagonistic analog in combination with paclitaxel or suberoylanilide hydroxamic acid have potent antiproliferative effects on anaplastic thyroid cancer. J Steroid Biochem Mol Biol 2011;124:1-9
  • Luong MT, O'Kelly J, Braunstein GD, Antitumor activity of suberoylanilide hydroxamic acid against thyroid cancer cell lines in vitro and in vivo. Clin Cancer Res 2006;12(18):5570-7
  • Borbone E, Berlingieri MT, De Bellis F, Histone deacetylase inhibitors induce thyroid cancer-specific apoptosis through proteasome-dependent inhibition of TRAIL degradation. Oncogene 2010;29:105-16
  • Kelly WK, O'Connor OA, Krug LM, Phase I study of an oral histone deacetylase inhibitor, suberoylanilide hydroxamic acid, in patients with advanced cancer. J Clin Oncol 2005;23:3923-31
  • Woyach JA, Kloos RT, Ringel MD, Lack of therapeutic effect of the histone deacetylase inhibitor vorinostat in patients with metastatic radioiodine-refractory thyroid carcinoma. J Clin Endocrinol Metab 2009;94:164-70
  • Gottlicher M, Minucci S, Zhu P, Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J 2001;20(24):6969-78
  • Xiao X, Ning L, Chen H. Notch1 mediates growth suppression of papillary and follicular thyroid cancer cells by histone deacetylase inhibitors. Mol Cancer Ther 2009;8(2):350-6
  • Shen WT, Wong TS, Chung WY, Valproic acid inhibits growth, induces apoptosis, and modulates apoptosis-regulatory and differentiation gene expression in human thyroid cancer cells. Surgery 2005;138:979-85
  • Catalano MG, Fortunati N, Pugliese M, 2006 Valproic acid, a histone deacetylase inhibitor, enhances sensitivity to doxorubicin in anaplastic thyroid cancer cells. J Endocrinol 2006;191:465-72
  • Kim TH, Yoo YH, Kang DY, Efficacy on anaplastic thyroid carcinoma of valproic acid alone or in combination with doxorubicin, a synthetic chenodeoxycholic acid derivative, or lactacystin. Int J Oncol 2009;34(5):1353-62
  • Zhang Y, Li N, Caron C. HDAC-6 interacts with and deacetylates tubulin and microtubules in vivo. EMBO J 2003;22(5):1168-79
  • Matsuyama A, Shimazu T, Sumida Y, In vivo destabilization of dynamic microtubules by HDAC6-mediated deacetylation. EMBO J 2002;21(24):6820-31
  • Catalano MG, Poli R, Pugliese M, Valproic acid enhances tubulin acetylation and apoptotic activity of paclitaxel on anaplastic thyroid cancer cell lines. Endocr Relat Cancer 2007;14(3):839-45
  • Atadja P. Development of the pan-DAC inhibitor panobinostat (LBH589): successes and challenges. Cancer Lett 2009;280(2):233-41
  • Catalano MG, Pugliese M, Gargantini E, Cytotoxic activity of the histone deacetylase inhibitor panobinostat (LBH589) in anaplastic thyroid cancer in vitro and in vivo. Int J Cancer 2012;130(3):694-704
  • Catalano MG, Fortunati N, Pugliese M, Histone deacetylase inhibition modulates e-cadherin expression and suppresses migration and invasion of anaplastic thyroid cancer cells. J Clin Endocrinol Metab 2012;97(7):E1150-9
  • Altmann A, Eisenhut M, Bauder-Wust U, Therapy of thyroid carcinoma with the histone deacetylase inhibitor MS-275. Eur J Nucl Med Mol Imaging 2010;37(12):2286-97
  • Wischnewski F, Pantel K, Schwarzenbach H. 2006 Promoter demethylation and histone acetylation mediate gene expression of MAGE-A1, -A2, -A3, and -A12 in human cancer cells. Mol Cancer Res 2006;4:339-49
  • Mitmaker EJ, Griff NJ, Grogan RH, Modulation of matrix metalloproteinase activity in human thyroid cancer cell lines using demethylating agents and histone deacetylase inhibitors. Surgery 2011;149:504-11
  • Lazar V, Bidart JM, Caillou B, Expression of the Na+/I- symporter gene in human thyroid tumors: a comparison study with other thyroid-specific genes. J Clin Endocrinol Metab 1999;84:3228-34
  • Gerard AC, Daumerie C, Mestdagh C, Correlation between the loss of thyroglobulin iodination and the expression of thyroid-specific proteins involved in iodine metabolism in thyroid carcinomas. J Clin Endocrinol Metab 2003;88:4977-83
  • Arturi F, Russo D, Bidart JM, Expression pattern of the pendrin and sodium/iodide symporter (NIS) gene in human thyroid carcinoma cell lines and human thyroid tumors. Eur J Endocrinol 2001;145:129-35
  • Puxeddu E, Durante C, Avenia N, Clinical implication of BRAF mutation in thyroid carcinoma. Trends Endocrinol Metab 2008;19:138-45
  • Trapasso F, Iuliano R, Chiefari E, Iodide symporter gene expression in normal and transformed rat thyroid cells. Eur J Endocrinol 1999;140:447-51
  • Knauf JA, Fagin JA. Role of MAPK pathway oncoproteins in thyroid cancer pathogenesis and as drug targets. Curr Opin Cell Biol 2009;21:296-303
  • Furuya F, Shimura H, Suzuki H, Histone deacetylase inhibitors restore radioiodide uptake and retention in poorly differentiated and anaplastic thyroid cancer cells by expression of the sodium/iodide symporter thyroperoxidase and thyroglobulin. Endocrinology 2004;145:2865-75
  • Puppin C, D'Aurizio F, D'Elia AV, Effects of histone acetylation on NIS promoter and expression of thyroid-specific transcription factors. Endocrinology 2005;146:3967-74
  • Kitazono M, Robey R, Zhan Z, Low concentrations of the histone deacetylase inhibitor, depsipeptide (FR901228), increase expression of the Na+/I- symporter and iodine accumulation in poorly differentiated thyroid carcinoma cells. J Clin Endocrinol Metab 2001;86:3430-5
  • Fortunati N, Catalano MG, Arena K, Valproic acid induces the expression of the Na+/I- symporter and iodide uptake in poorly differentiated thyroid cancer cells. J Clin Endocrinol Metab 2004;89:1006-9
  • Hou P, Bojdani E, Xing M. Induction of thyroid gene expression and radioiodine uptake in thyroid cancer cells by targeting major signaling pathways. J Clin Endocrinol Metab 2010;95:820-8
  • Liu Z, Xing M. Induction of sodium/iodide symporter (NIS) expression and radioiodine uptake in non-thyroid cancer cells. PLoS One 2012;7(2):e31729
  • Zhang M, Guo R, Xu H, Retinoic acid and tributyrin induce in-vitro radioiodine uptake and inhibition of cell proliferation in a poorly differentiated follicular thyroid carcinoma. Nucl Med Commun 2011;32:605-10
  • Li W, Venkataraman GM, Ain KB. Protein synthesis inhibitors, in synergy with 5-Azacytidine, restore sodium/iodide symporter gene expression in human thyroid adenoma cell line, KAK-1, suggesting trans-active transcriptional repressor. J Clin Endocrinol Metab 2007;92:1080-7
  • Provenzano MJ, Fitzgerald MP, Krager K, Domann FE. Increased iodine uptake in thyroid carcinoma after treatment with sodium butyrate and decitabine (5-Aza-dC). Otolaryngol Head Neck Surg 2007;137:722-8
  • Kramer OH, Gottlicher M, Heinzel T. Histone deacetylase as a therapeutic target. Trends Endocrinol Metab 2001;7:294-300
  • Sherman EJ, Fury MG, Tuttle RM, Phase II study of depsipeptide (DEP) in radioiodine (RAI)-refractory metastatic nonmedullary thyroid carcinoma. J Clin Oncol 2009;27(15S):6059
  • Kapiteijn E, Schneider TC, Morreau H, New treatment modalities in advanced thyroid cancer. Ann Oncol 2012;23:10-18
  • Puppin C, Passon N, Hershman JM, Cooperative effects of SAHA and VPA on expression of NIS mRNA and proliferation of thyroid cancer cells. J Mol Endocrinol 2012;48:217-27
  • Noguchi H, Yamashita H, Murakami T, Successful treatment of anaplastic thyroid carcinoma with combination of oral valproic acid, chemotherapy, radiation and surgery. Endocr J 2009;56:245-9
  • Celano M, Calvagno MG, Bulotta S, Cytotoxic effects of Gemcitabine-loaded liposomes in human anaplastic thyroid carcinoma cells. BMC Cancer 2004;4:63
  • Celano M, Schenone S, Cosco D, Cytotoxic effects of a novel pyrazolopyrimidine derivative entrapped in liposomes in anaplastic thyroid cancer cells in vitro and in xenograft tumors in vivo. Endocr Rel Cancer 2008;15:499-510
  • Paolino D, Cosco D, Racanicchi L, Gemcitabine-loaded PEGylated unilamellar liposomes vs GEMZAR®: biodistribution, pharmacokinetic features and in vivo antitumor activity. J Control Rel 2010;144:144-50

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.