627
Views
32
CrossRef citations to date
0
Altmetric
Review

ATAD2 in cancer: a pharmacologically challenging but tractable target

, , , , , & show all
Pages 85-96 | Received 03 Apr 2017, Accepted 15 Nov 2017, Published online: 23 Nov 2017

References

  • Jeon J, Nim S, Teyra J, et al. A systematic approach to identify novel cancer drug targets using machine learning, inhibitor design and high-throughput screening. Genome Med. 2014;6:57.
  • Vogelstein B, Papadopoulos N, Velculescu VE, et al. Cancer genome landscapes. Science. 2013;339:1546–1558.
  • Zhang Z, Li M, Rayburn ER, et al. Oncogenes as novel targets for cancer therapy (part I): growth factors and protein tyrosine kinases. Am J Pharmacogenomics. 2005;5:173–190.
  • Zhang Z, Li M, Rayburn ER, et al. Oncogenes as novel targets for cancer therapy (part II): intermediate signaling molecules. Am J Pharmacogenomics. 2005;5:247–257.
  • Zhang Z, Li M, Rayburn ER, et al. Oncogenes as novel targets for cancer therapy (part III): transcription factors. Am J Pharmacogenomics. 2005;5:327–338.
  • Zhang Z, Li M, Rayburn ER, et al. Oncogenes as novel targets for cancer therapy (part IV): regulators of the cell cycle and apoptosis. Am J Pharmacogenomics. 2005;5:397–407.
  • Dang CV, Reddy EP, Shokat KM, et al. Drugging the ‘undruggable’ cancer targets. Nat Rev Cancer. 2017;17:502–508.
  • Lazo JS, Sharlow ER. Drugging undruggable molecular cancer targets. Annu Rev Pharmacol Toxicol. 2016;56:23–40.
  • Zhang ZY. Drugging the undruggable: therapeutic potential of targeting protein tyrosine phosphatases. Acc Chem Res. 2017;50:122–129.
  • Zhang JT, Liu JY. Drugging the “undruggable” DNA-binding domain of STAT3. Oncotarget. 2016;7:66324–66325.
  • Boussouar F, Jamshidikia M, Morozumi Y, et al. Malignant genome reprogramming by ATAD2. Biochim Biophys Acta. 2013;1829:1010–1014.
  • Ciro M, Prosperini E, Quarto M, et al. ATAD2 is a novel cofactor for MYC, overexpressed and amplified in aggressive tumors. Cancer Res. 2009;69:8491–8498.
  • Vukovic S, Brennan PE, Huggins DJ. Exploring the role of water in molecular recognition: predicting protein ligandability using a combinatorial search of surface hydration sites. J Phys Condens Matter. 2016;28:344007.
  • Vidler LR, Brown N, Knapp S, et al. Druggability analysis and structural classification of bromodomain acetyl-lysine binding sites. J Med Chem. 2012;55:7346–7359.
  • Bamborough P, Chung CW, Demont EH, et al. A chemical probe for the ATAD2 bromodomain. Angew Chem Int Ed Engl. 2016;55:11382–11386.
  • Bamborough P, Chung CW, Furze RC, et al. Structure-based optimization of naphthyridones into potent ATAD2 bromodomain inhibitors. J Med Chem. 2015;58:6151–6178.
  • Chaikuad A, Petros AM, Fedorov O, et al. Structure-based approaches towards identification of fragments for the low-druggability ATAD2 bromodomain. Med Chem Commun. 2014;5:1843–1848.
  • Cattaneo M, Morozumi Y, Perazza D, et al. Lessons from yeast on emerging roles of the ATAD2 protein family in gene regulation and genome organization. Mol Cells. 2014;37:851–856.
  • Hanson PI, Whiteheart SW. AAA+ proteins: have engine, will work. Nat Rev Mol Cell Biol. 2005;6:519–529.
  • Kedzierska S. [Structure, function and mechanisms of action of ATPases from the AAA superfamily of proteins]. Postepy Biochem. 2006;52:330–338.
  • Snider J, Thibault G, Houry WA. The AAA+ superfamily of functionally diverse proteins. Genome Biol. 2008;9:216.
  • Snider J, Houry WA. AAA+ proteins: diversity in function, similarity in structure. Biochem Soc Trans. 2008;36:72–77.
  • Pollack JR, Sorlie T, Perou CM, et al. Microarray analysis reveals a major direct role of DNA copy number alteration in the transcriptional program of human breast tumors. Proc Natl Acad Sci USA. 2002;99:12963–12968.
  • Leachman NT, Brellier F, Ferralli J, et al. ATAD2B is a phylogenetically conserved nuclear protein expressed during neuronal differentiation and tumorigenesis. Dev Growth Differ. 2010;52:747–755.
  • Altintas DM, Shukla MS, Goutte-Gattat D, et al. Direct cooperation between androgen receptor and E2F1 reveals a common regulation mechanism for androgen-responsive genes in prostate cells. Mol Endocrinol. 2012;26:1531–1541.
  • Zou JX, Guo L, Revenko AS, et al. Androgen-induced coactivator ANCCA mediates specific androgen receptor signaling in prostate cancer. Cancer Res. 2009;69:3339–3346.
  • Caron C, Lestrat C, Marsal S, et al. Functional characterization of ATAD2 as a new cancer/testis factor and a predictor of poor prognosis in breast and lung cancers. Oncogene. 2010;29:5171–5181.
  • Dhalluin C, Carlson JE, Zeng L, et al. Structure and ligand of a histone acetyltransferase bromodomain. Nature. 1999;399:491–496.
  • Owen DJ, Ornaghi P, Yang JC, et al. The structural basis for the recognition of acetylated histone H4 by the bromodomain of histone acetyltransferase gcn5p. Embo J. 2000;19:6141–6149.
  • Revenko AS, Kalashnikova EV, Gemo AT, et al. Chromatin loading of E2F-MLL complex by cancer-associated coregulator ANCCA via reading a specific histone mark. Mol Cell Biol. 2010;30:5260–5272.
  • Morozumi Y, Boussouar F, Tan M, et al. Atad2 is a generalist facilitator of chromatin dynamics in embryonic stem cells. J Mol Cell Biol. 2016;8:349–362.
  • Mjelle R, Hegre SA, Aas PA, et al. Cell cycle regulation of human DNA repair and chromatin remodeling genes. DNA Repair (Amst). 2015;30:53–67.
  • Liu J, Lee W, Jiang Z, et al. Genome and transcriptome sequencing of lung cancers reveal diverse mutational and splicing events. Genome Res. 2012;22:2315–2327.
  • Wrzeszczynski KO, Varadan VByrnes J, et al. Identification of tumor suppressors and oncogenes from genomic and epigenetic features in ovarian cancer. Plos One. 2011;6:e28503.
  • Hsia EY, Kalashnikova EV, Revenko AS, et al. Deregulated E2F and the AAA+ coregulator ANCCA drive proto-oncogene ACTR/AIB1 overexpression in breast cancer. Mol Cancer Res. 2010;8:183–193.
  • Zou JX, Revenko AS, Li LB, et al. ANCCA, an estrogen-regulated AAA+ ATPase coactivator for ERalpha, is required for coregulator occupancy and chromatin modification. Proc Natl Acad Sci USA. 2007;104:18067–18072.
  • Duan Z, Zou JX, Yang P, et al. Developmental and androgenic regulation of chromatin regulators EZH2 and ANCCA/ATAD2 in the prostate via MLL histone methylase complex. Prostate. 2013;73:455–466.
  • Kalashnikova EV, Revenko AS, Gemo AT, et al. ANCCA/ATAD2 overexpression identifies breast cancer patients with poor prognosis, acting to drive proliferation and survival of triple-negative cells through control of B-MYB and EZH2. Cancer Res. 2010;70:9402–9412.
  • Krakstad C, Tangen IL, Hoivik EA, et al. ATAD2 overexpression links to enrichment of B-MYB-translational signatures and development of aggressive endometrial carcinoma. Oncotarget. 2015;6:28440–28452.
  • Shang P, Meng F, Liu Y, et al. Overexpression of ANCCA/ATAD2 in endometrial carcinoma and its correlation with tumor progression and poor prognosis. Tumour Biol. 2015;36:4479–4485.
  • Raeder MB, Birkeland E, Trovik J, et al. Integrated genomic analysis of the 8q24 amplification in endometrial cancers identifies ATAD2 as essential to MYC-dependent cancers. PLoS One. 2013;8:e54873.
  • Wu G, Lu X, Wang Y, et al. Epigenetic high regulation of ATAD2 regulates the Hh pathway in human hepatocellular carcinoma. Int J Oncol. 2014;45:351–361.
  • Koo SJ, Fernandez-Montalvan AE, Badock V, et al. ATAD2 is an epigenetic reader of newly synthesized histone marks during DNA replication. Oncotarget. 2016. DOI:10.18632/oncotarget.11855
  • Lu WJ, Chua MS, So SK. Suppression of ATAD2 inhibits hepatocellular carcinoma progression through activation of p53- and p38-mediated apoptotic signaling. Oncotarget. 2015;6:41722–41735.
  • Huang Q, Lin B, Liu H, et al. RNA-Seq analyses generate comprehensive transcriptomic landscape and reveal complex transcript patterns in hepatocellular carcinoma. PLoS One. 2011;6:e26168.
  • Hong S, Bi M, Yan Z, et al. Silencing of ATPase family AAA domain-containing protein 2 inhibits migration and invasion of colorectal cancer cells. Neoplasma. 2016;63:846–855.
  • Wang Y, Klijn JG, Zhang Y, et al. Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 2005;365:671–679.
  • De Angelis PM, Svendsrud DH, Kravik KL, et al. Cellular response to 5-fluorouracil (5-FU) in 5-FU-resistant colon cancer cell lines during treatment and recovery. Mol Cancer. 2006;5:20.
  • Murakami H, Ito S, Tanaka H, et al. Establishment of new intraperitoneal paclitaxel-resistant gastric cancer cell lines and comprehensive gene expression analysis. Anticancer Res. 2013;33:4299–4307.
  • Zou JX, Duan Z, Wang J, et al. Kinesin family deregulation coordinated by bromodomain protein ANCCA and histone methyltransferase MLL for breast cancer cell growth, survival, and tamoxifen resistance. Mol Cancer Res. 2014;12:539–549.
  • Salvesen HB, Carter SL, Mannelqvist M, et al. Integrated genomic profiling of endometrial carcinoma associates aggressive tumors with indicators of PI3 kinase activation. Proc Natl Acad Sci USA. 2009;106:4834–4839.
  • van ‘T Veer LJ, Dai H, van de Vijver MJ, et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415:530–536.
  • Vidalin O, Muslmani M, Estienne C, et al. In vivo target validation using gene invalidation, RNA interference and protein functional knockout models: it is the time to combine. Curr Opin Pharmacol. 2009;9:669–676.
  • Colombo R, Moll J. Target validation to biomarker development: focus on RNA interference. Mol Diagn Ther. 2008;12:63–70.
  • Natt F. siRNAs in drug discovery: target validation and beyond. Curr Opin Mol Ther. 2007;9:242–247.
  • Luo Y, Ye GY, Qin SL, et al. ATAD2 overexpression identifies colorectal cancer patients with poor prognosis and drives proliferation of cancer cells. Gastroenterol Res Pract. 2015;2015:936564.
  • Wu G, Liu H, He H, et al. miR-372 down-regulates the oncogene ATAD2 to influence hepatocellular carcinoma proliferation and metastasis. BMC Cancer. 2014;14:107.
  • Wan WN, Zhang YX, Wang XM, et al. ATAD2 is highly expressed in ovarian carcinomas and indicates poor prognosis. Asian Pac J Cancer Prev. 2014;15:2777–2783.
  • Zheng L, Li T, Zhang Y, et al. Oncogene ATAD2 promotes cell proliferation, invasion and migration in cervical cancer. Oncol Rep. 2015;33:2337–2344.
  • Gashaw I, Ellinghaus P, Sommer A, et al. What makes a good drug target? Drug Discov Today. 2011;16:1037–1043.
  • Gashaw I, Ellinghaus P, Sommer A, et al. What makes a good drug target? Drug Discov Today. 2012;17 Suppl:S2–S30.
  • Henrich S, Salo-Ahen OM, Huang B, et al. Computational approaches to identifying and characterizing protein binding sites for ligand design. J Mol Recognit. 2010;23:209–219.
  • Firestone AJ, Weinger JS, Maldonado M, et al. Small-molecule inhibitors of the AAA+ ATPase motor cytoplasmic dynein. Nature. 2012;484:125–129.
  • Muller S, Filippakopoulos P, Knapp S. Bromodomains as therapeutic targets. Expert Rev Mol Med. 2011;13:e29.
  • Poncet-Montange G, Zhan Y, Bardenhagen JP, et al. Observed bromodomain flexibility reveals histone peptide- and small molecule ligand-compatible forms of ATAD2. Biochem J. 2015;466:337–346.
  • Filippakopoulos P, Picaud S, Mangos M, et al. Histone recognition and large-scale structural analysis of the human bromodomain family. Cell. 2012;149:214–231.
  • Filippakopoulos P, Qi J, Picaud S, et al. Selective inhibition of BET bromodomains. Nature. 2010;468:1067–1073.
  • Nakagawa A, Adams CE, Huang Y, et al. Selective and reversible suppression of intestinal stem cell differentiation by pharmacological inhibition of BET bromodomains. Sci Rep. 2016;6:20390.
  • Nicodeme E, Jeffrey KL, Schaefer U, et al. Suppression of inflammation by a synthetic histone mimic. Nature. 2010;468:1119–1123.
  • Xu M, Unzue A, Dong J, et al. Discovery of CREBBP bromodomain inhibitors by high-throughput docking and hit optimization guided by molecular dynamics. J Med Chem. 2016;59:1340–1349.
  • Harner MJ, Chauder BA, Phan J, et al. Fragment-based screening of the bromodomain of ATAD2. J Med Chem. 2014;57:9687–9692.
  • Hajduk PJ, Huth JR, Fesik SW. Druggability indices for protein targets derived from NMR-based screening data. J Med Chem. 2005;48:2518–2525.
  • Demont EH, Chung CW, Furze RC, et al. Fragment-based discovery of low-micromolar ATAD2 bromodomain inhibitors. J Med Chem. 2015;58:5649–5673.
  • Elkaim J, Castroviejo M, Bennani D, et al. First identification of small-molecule inhibitors of Pontin by combining virtual screening and enzymatic assay. Biochem J. 2012;443:549–559.
  • Chou TF, Brown SJ, Minond D, et al. Reversible inhibitor of p97, DBeQ, impairs both ubiquitin-dependent and autophagic protein clearance pathways. Proc Natl Acad Sci USA. 2011;108:4834–4839.
  • Goller T, Seibold UK, Kremmer E, et al. Atad3 function is essential for early post-implantation development in the mouse. PLoS One. 2013;8:e54799.
  • Issop L, Fan J, Lee S, et al. Mitochondria-associated membrane formation in hormone-stimulated Leydig cell steroidogenesis: role of ATAD3. Endocrinology. 2015;156:334–345.
  • Li S, Yao Y, Xu R, et al. ATAD3 is a limiting factor in mitochondrial biogenesis and adipogenesis of white adipocyte-like 3T3-L1 cells. Mol Cell Biol. 2014. DOI:10.1128/MCB.01170-13
  • Li S, Rousseau D. [ATAD3, a vital membrane-bound mitochondrial ATPase involved in tumor progression]. Med Sci (Paris). 2011;27:1089–1095.
  • Li S, Rousseau D. ATAD3, a vital membrane bound mitochondrial ATPase involved in tumor progression. J Bioenerg Biomembr. 2012;44:189–197.
  • Maleva Kostovska I, Wang J, Bogdanova N, et al. Rare ATAD5 missense variants in breast and ovarian cancer patients. Cancer Lett. 2016;376:173–177.
  • Ghosh A, Ghosh S, Dasgupta D, et al. Hepatitis B virus X protein upregulates hELG1/ATAD5 expression through E2F1 in hepatocellular carcinoma. Int J Biol Sci. 2016;12:30–41.
  • Bell DW, Sikdar N, Lee KY, et al. Predisposition to cancer caused by genetic and functional defects of mammalian Atad5. PLoS Genet. 2011;7:e1002245.
  • Kubota T, Myung K, Donaldson AD. Is PCNA unloading the central function of the Elg1/ATAD5 replication factor C-like complex? Cell Cycle. 2013;12:2570–2579.
  • Lee KY, Fu H, Aladjem MI, et al. ATAD5 regulates the lifespan of DNA replication factories by modulating PCNA level on the chromatin. J Cell Biol. 2013;200:31–44.
  • La Thangue NB, Kerr DJ. Predictive biomarkers: a paradigm shift towards personalized cancer medicine. Nat Rev Clin Oncol. 2011;8:587–596.
  • Contasta I, Berghella AM, Lattanzio R, et al. Novel prognostic biomarkers for personalized cancer treatment. Cancer Immunol. 2015; Springer Berlin Heidelberg; 2014:35–54.
  • Rossi A, Torri V, Garassino MC, et al. The impact of personalized medicine on survival: comparisons of results in metastatic breast, colorectal and non-small-cell lung cancers. Cancer Treat Rev. 2014;40:485–494.
  • Welch DR, Rinker-Schaeffer CW. What defines a useful marker of metastasis in human cancer? J Natl Cancer Inst. 1999;91:1351–1353.
  • Wu SY, Lopez-Berestein G, Calin GA, et al. RNAi therapies: drugging the undruggable. Sci Transl Med. 2014;6:240ps7.
  • Yan R, Hallam A, Stockley PG, et al. Oncogene dependency and the potential of targeted RNAi-based anti-cancer therapy. Biochem J. 2014;461:1–13.
  • Yang WQ, Zhang Y. RNAi-mediated gene silencing in cancer therapy. Expert Opin Biol Ther. 2012;12:1495–1504.

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.