113
Views
19
CrossRef citations to date
0
Altmetric
Original Research

Targeting breast cancer stem cells by a self-assembled, aptamer-conjugated DNA nanotrain with preloading doxorubicin

, &
Pages 6831-6842 | Published online: 27 Aug 2019

References

  • Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. Ca-Cancer J Clin. 2015;65(2):87–108. doi:10.3322/caac.2129425651787
  • Haffty BG, Yang Q, Reiss M, et al. Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J Clin Oncol. 2006;24(36):5652–5657. doi:10.1200/JCO.2006.06.566417116942
  • Voduc KD, Cheang MS, Gelmon K, Nielsen TO, Kennecke H. Breast cancer subtypes and the risk of local and regional relapse. J Clin Oncol. 2010;28(10):1684–1691. doi:10.1200/JCO.2009.24.928420194857
  • Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer. 2008;8(10):755–768. doi:10.1038/nrc249918784658
  • Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer. 2005;5(4):275–284. doi:10.1038/nrc159015803154
  • Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. P Natl Acad Sci USA. 2003;100(7):3983–3988. doi:10.1073/pnas.0530291100
  • Somasunderam A, Thiviyanathan V, Tanaka T, et al. Combinatorial selection of DNA thioaptamers targeted to the HA binding domain of human CD44. Biochemistry. 2010;49(42):9106–9112. doi:10.1021/bi100950320843027
  • Sun H, Zhu X, Lu PY, Rosato RR, Tan W, Zu Y. Oligonucleotide aptamers: new tools for targeted cancer therapy. Mol Ther Nucl Acids. 2014;3(8):e182. doi:10.1038/mtna.2014.32
  • Zhang Z, Ali MM, Eckert MA, et al. A polyvalent aptamer system for targeted drug delivery. Biomaterials. 2013;34(37):9728–9735. doi:10.1016/j.biomaterials.2013.08.07924044994
  • Huang YF, Shangguan D, Liu H, et al. Molecular assembly of an aptamer-drug conjugate for targeted drug delivery to tumor cells. Chembiochem. 2009;10(5):862–868. doi:10.1002/cbic.20090017219253922
  • Subramanian N, Raghunathan V, Kanwar JR, et al. Target-specific delivery of doxorubicin to retinoblastoma using epithelial cell adhesion molecule aptamer. Mol Vis. 2012;18(283):2783–2795.23213278
  • Zhu G, Zheng J, Song E, et al. Self-assembled, aptamer-tethered DNA nanotrains for targeted transport of molecular drugs in cancer theranostics. P Natl Acad Sci USA. 2013;110(20):7998–8003. doi:10.1073/pnas.1220817110
  • Dirks RM, Pierce NA. Triggered amplification by hybridization chain reaction. P Natl Acad Sci USA. 2004;101(43):15275–15278. doi:10.1073/pnas.0407024101
  • Bagalkot V, Farokhzad OC, Langer R, Jon S. An aptamer-doxorubicin physical conjugate as a novel targeted drug-delivery platform. Angew Chem Int Edit. 2006;45(48):8149–8152. doi:10.1002/anie.200602251
  • Mcdermott SP, Wicha MS. Targeting breast cancer stem cells. Mol Oncol. 2010;4(5):404–419. doi:10.1016/j.molonc.2010.06.00520599450
  • Bleau AM, Hambardzumyan D, Ozawa T, et al. PTEN/PI3K/AKT pathway regulates the side population phenotype and ABCG2 activity in glioma tumor stem-like cells. Cell Stem Cell. 2009;4(3):226–235. doi:10.1016/j.stem.2009.01.00719265662
  • Gargini R, Cerliani JP, Escoll M, Anton IM, Wandosell F. Cancer stem cell-like phenotype and survival are coordinately regulated by Akt/FoxO/Bim pathway. Stem Cells. 2015;33(3):646–660. doi:10.1002/stem.190425407338
  • Kumar D, Shankar S, Srivastava RK, Srivastava RK. Rottlerin induces autophagy and apoptosis in prostate cancer stem cells via PI3K/AKT/mTOR signaling pathway. Cancer Lett. 2014;343(2):179–189. doi:10.1016/j.canlet.2013.10.00324125861
  • Hiromura M, Okada F, Obata T, et al. Inhibition of Akt kinase activity by a peptide spanning the βA strand of the proto-oncogene TCL1. J Biol Chem. 2004;279(51):53407–53418. doi:10.1074/jbc.M40377520015459205
  • Davoudi Z, Akbarzadeh A, Rahmatiyamchi M, Movassaghpour AA, Alipour M, Nejatikoshki K. Molecular target therapy of AKT and NF-kB signaling pathways and multidrug resistance by specific cell penetrating inhibitor peptides in HL-60 cells. Asian Pac J Cancer P. 2014;15(10):4353–4358. doi:10.7314/APJCP.2014.15.10.4353
  • Xu F, Yang T, Chen Y. Quantification of microRNA by DNA-peptide probe and liquid chromatography-tandem mass spectrometry-based quasi-targeted proteomics. Anal Chem. 2016;88(1):754–763. doi:10.1021/acs.analchem.5b0305626641144
  • Xu J, Liu Y, Yu Y, Ni Q, Chen Y. Subcellular quantification of doxorubicin and its metabolite in cultured human leukemia cells using liquid chromatography-tandem mass spectrometry. Anal Lett. 2012;45(14):1980–1994. doi:10.1080/00032719.2012.680056
  • Yang T, Xu F, Xu J, Fang D, Yu Y, Chen Y. Comparison of liquid chromatography-tandem mass spectrometry-based targeted proteomics and conventional analytical methods for the determination of P-glycoprotein in human breast cancer cells. J Chromatogr B. 2013;936(1):18–24. doi:10.1016/j.jchromb.2013.07.023
  • Buck E, Eyzaguirre A, Brown E, et al. Rapamycin synergizes with the epidermal growth factor receptor inhibitor erlotinib in non-small-cell lung, pancreatic, colon, and breast tumors. Mol Cancer Ther. 2006;5(11):2676–2684. doi:10.1158/1535-7163.MCT-05-023517121914
  • Mohan P, Rapoport N. Doxorubicin as a molecular nanotheranostic agent: effect of doxorubicin encapsulation in micelles or nanoemulsions on the ultrasound-mediated intracellular delivery and nuclear trafficking. Mol Pharmaceut. 2010;7(6):1959–1973. doi:10.1021/mp100269f
  • Guertin AD, Martin MM, Roberts B, et al. Unique functions of CHK1 and WEE1 underlie synergistic anti-tumor activity upon pharmacologic inhibition. Cancer Cell Int. 2012;12(1):45–56. doi:10.1186/1475-2867-12-4523148684
  • Nitin N, Santangelo PJ, Kim G, Nie S, Bao G. Peptide-linked molecular beacons for efficient delivery and rapid mRNA detection in living cells. Nucleic Acids Res. 2004;32(6):e58. doi:10.1093/nar/gnh06315084673
  • Yu F, Yao H, Zhu P, et al. Let-7 regulates self renewal and tumorigenicity of breast cancer cells. Cell. 2007;131(6):1109–1123. doi:10.1016/j.cell.2007.10.05418083101
  • Sun TM, Wang YC, Wang F, et al. Cancer stem cell therapy using doxorubicin conjugated to gold nanoparticles via hydrazone bonds. Biomaterials. 2014;35(2):836–845. doi:10.1016/j.biomaterials.2014.01.02624144908
  • Kyaw T, Winship A, Tay C, et al. Cytotoxic and proinflammatory CD8+ T lymphocytes promote development of vulnerable atherosclerotic plaques in ApoE-deficient mice. Circulation. 2013;127(9):1028–1039. doi:10.1161/CIRCULATIONAHA.112.00134723395974
  • Wu C, Han D, Chen T, et al. Building a multifunctional aptamer-based DNA nanoassembly for targeted cancer therapy. J Am Chem Soc. 2013;135(49):18644–18650. doi:10.1021/ja409461724245521
  • Wu XA, Choi CH, Zhang C, Hao L, Mirkin CA. Intracellular fate of spherical nucleic acid nanoparticle conjugates. J Am Chem Soc. 2014;136(21):7726–7733. doi:10.1021/ja503010a24841494
  • Alibolandi M, Taghdisi SM, Ramezani P, et al. Smart AS1411-aptamer conjugated pegylated PAMAM dendrimer for the superior delivery of camptothecin to colon adenocarcinoma in vitro and in vivo. Int J Pharmaceut. 2017;519(1):352–364. doi:10.1016/j.ijpharm.2017.01.044
  • Hu Y, Fu L. Targeting cancer stem cells: a new therapy to cure cancer patients. Am J Cancer Res. 2012;2(3):340–356.22679565
  • Morrison BJ, Schmidt CW, Lakhani SR, Reynolds BA, Lopez JA. Breast cancer stem cells: implications for therapy of breast cancer. Breast Cancer Res. 2008;10(4):210–223. doi:10.1186/bcr211118671830
  • Eliaz RE Jr, Szoka F. Liposome-encapsulated doxorubicin targeted to CD44: a strategy to kill CD44-overexpressing tumor cells. Cancer Res. 2001;61(6):2592–2601.11289136
  • Peer D, Margalit R. Loading mitomycin C inside long circulating hyaluronan targeted nano-liposomes increases its antitumor activity in three mice tumor models. Int J Cancer. 2004;108(5):780–789. doi:10.1002/ijc.1145914696107
  • Dan P, Margalit R. Tumor-targeted hyaluronan nanoliposomes increase the antitumor activity of liposomal doxorubicin in syngeneic and human xenograft mouse tumor models. Neoplasia. 2004;6(4):343–353. doi:10.1593/neo.0346015256056
  • Ponta H, Sherman L, Herrlich PA. CD44: from adhesion molecules to signalling regulators. Nat Rev Mol Cell Bio. 2003;4(1):33–45. doi:10.1038/nrm100412511867
  • Skotheim RI, Nees M. Alternative splicing in cancer: noise, functional, or systematic? Int J Biochem Cell B. 2007;39(7):1432–1449. doi:10.1016/j.biocel.2007.02.016
  • Jackson DG, Bell JI, Dickinson R, Timans J, Shields J, Whittle N. Proteoglycan forms of the lymphocyte homing receptor CD44 are alternatively spliced variants containing the V3 exon. J Cell Biol. 1995;128(4):673–685. doi:10.1083/jcb.128.4.6737532175
  • Radom F, Jurek PM, Mazurek MP, Otlewski J, Jeleń F. Aptamers: molecules of great potential. Biotechnol Adv. 2013;31(8):1260–1274. doi:10.1016/j.biotechadv.2013.04.00723632375
  • Tan W, Wang H, Chen Y, et al. Molecular aptamers for drug delivery. Trends Biotechnol. 2011;29(12):634–640. doi:10.1016/j.tibtech.2011.06.00921821299
  • Tao W, Gantier MP, Xiang D, et al. EpCAM aptamer-mediated survivin silencing sensitized cancer stem cells to doxorubicin in a breast cancer model. Theranostics. 2015;5(12):1456–1472. doi:10.7150/thno.1169226681989
  • Wang J, Sefah K, Altman MB, et al. Aptamer-conjugated nanorods for targeted photothermal therapy of prostate cancer stem cells. Chem Asian J. 2013;8(10):2417–2422. doi:10.1002/asia.20130037523757285
  • Naor D, Sionov RV, Ish-Shalom D. CD44: structure, function, and association with the malignant process. Adv Cancer Res. 1997;71:241–319.9111868
  • Roa WH, Azarmi S, Al-Hallak MHDK, Finlay WH, Magliocco AM, Löbenberg R. Inhalable nanoparticles, a non-invasive approach to treat lung cancer in a mouse model. J Control Release. 2011;150(1):49–55. doi:10.1016/j.jconrel.2010.10.03521059378
  • Maeda H, Nakamura H, Fang J. The EPR effect for macromolecular drug delivery to solid tumors: improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv Drug Deliver Rev. 2013;65(1):71–79. doi:10.1016/j.addr.2012.10.002
  • Bi S, Yue S, Zhang S. Hybridization chain reaction: a versatile molecular tool for biosensing, bioimaging, and biomedicine. Chem Soc Rev. 2017;46(14):4281–4298. doi:10.1039/C7CS00055C28573275
  • Bachur NR, Gee M. Daunorubicin metabolism by rat tissue preparations. J Pharmacol Exp Ther. 1971;177(3):567–572.4398190
  • Dan P, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2(12):751–760. doi:10.1038/nnano.2007.38718654426
  • Petros RA, Desimone JM. Strategies in the design of nanoparticles for therapeutic applications. Nat Rev Drug Discov. 2010;9(8):615–627. doi:10.1038/nrd259120616808
  • Yu M, Jambhrunkar S, Thorn P, Chen J, Gu W, Yu C. Hyaluronic acid modified mesoporous silica nanoparticles for targeted drug delivery to CD44-overexpressing cancer cells. Nanoscale. 2013;5(1):178–183. doi:10.1039/C2NR32145A23076766
  • Misra S, Heldin P, Hascall VC, et al. Hyaluronan–CD44 interactions as potential targets for cancer therapy. Febs J. 2011;278(9):1429–1443. doi:10.1111/j.1742-4658.2011.08071.x21362138
  • Cun X, Chen J, Ruan S, et al. A novel strategy through combining iRGD peptide with tumor-microenvironment-responsive and multistage nanoparticles for deep tumor penetration. ACS Appl Mater Inter. 2015;7(49):27458–27466. doi:10.1021/acsami.5b09391
  • Onoue S, Yamada S, Chan H. Nanodrugs: pharmacokinetics and safety. Int J Nanomed. 2014;9(1):1025–1037. doi:10.2147/IJN.S38378