300
Views
26
CrossRef citations to date
0
Altmetric
Review

Aptamer Hybrid Nanocomplexes as Targeting Components for Antibiotic/Gene Delivery Systems and Diagnostics: A Review

ORCID Icon, , , , , , , ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 4237-4256 | Published online: 17 Jun 2020

References

  • Stoltenburg R, Reinemann C, Strehlitz B. SELEX—a (r) evolutionary method to generate high-affinity nucleic acid ligands. Biomol Eng. 2007;24(4):381–403. doi:10.1016/j.bioeng.2007.06.00117627883
  • de Vries JW, Schnichels S, Hurst J, et al. DNA nanoparticles for ophthalmic drug delivery. Biomaterials. 2018;157:98–106. doi:10.1016/j.biomaterials.2017.11.04629258013
  • Rabiee N, Yaraki MT, Garakani SM, et al. Recent advances in porphyrin-based nanocomposites for effective targeted imaging and therapy. Biomaterials. 2019;119707.31874428
  • Bahrami S, Baheiraei N, Mohseni M, et al. Three-dimensional graphene foam as a conductive scaffold for cardiac tissue engineering. J Biomater Appl. 2019;34(1):74–85. doi:10.1177/088532821983903730961432
  • Dollins CM, Nair S, Sullenger BA. Aptamers in immunotherapy. Hum Gene Ther. 2008;19(5):443–450. doi:10.1089/hum.2008.04518473674
  • Levy M, Palliser D, Wengerter BC, Almo SC. Aptamer-Targetted Antigen Delivery. Google Patents; 2017.
  • Bayraç C, Eyidoğan F, Öktem HA. DNA aptamer-based colorimetric detection platform for Salmonella enteritidis. Biosens Bioelect. 2017;98:22–28. doi:10.1016/j.bios.2017.06.029
  • Hajebi S, Rabiee N, Bagherzadeh M, et al. Stimulus-responsive polymeric nanogels as smart drug delivery systems. Acta Biomater. 2019;92:1–18. doi:10.1016/j.actbio.2019.05.01831096042
  • Ahmadi S, Rabiee N, Rabiee M. Application of aptamer-based hybrid molecules in early diagnosis and treatment of diabetes mellitus: from the concepts towards the future. Curr Diabetes Rev. 2019;15(4):309–313. doi:10.2174/157339981466618060707555029875005
  • Gooch J, Daniel B, Parkin M, Frascione N. Developing aptasensors for forensic analysis. TrAC Trends Anal Chem. 2017;94:150–160. doi:10.1016/j.trac.2017.07.019
  • Nussbaum O, Oz MB, Tilayov T, Atiya H, Dagan S. A signal amplification probe enhances sensitivity of antibodies and aptamers based Immuno-diagnostic assays. J Immunol Methods. 2017;448:85–90. doi:10.1016/j.jim.2017.06.00128614699
  • Nour S, Baheiraei N, Imani R, et al. Bioactive materials: a comprehensive review on interactions with biological microenvironment based on the immune response. J Bionic Eng. 2019;16(4):563–581. doi:10.1007/s42235-019-0046-z
  • Nour S, Baheiraei N, Imani R, et al. A review of accelerated wound healing approaches: biomaterial-assisted tissue remodeling. J Mater Sci. 2019;30(10):120.
  • Bates PJ, Reyes-Reyes EM, Malik MT, Murphy EM, O’toole MG, Trent JO. G-quadruplex oligonucleotide AS1411 as a cancer-targeting agent: uses and mechanisms. Biochimica Et Biophysica Acta (BBA)-General Subj. 2017;1861(5):1414–1428. doi:10.1016/j.bbagen.2016.12.015
  • Howard PT. Investigating the Mechanism of Novel Anticancer Agent, AS1411: Does Metabolism to Guanine Play a Role? 2017.
  • Feng S, Chen C, Wang W, Que L. An aptamer nanopore-enabled microsensor for detection of theophylline. Biosens Bioelect. 2018;105:36–41. doi:10.1016/j.bios.2018.01.016
  • Pützer BM, Solanki M, Herchenröder O. Advances in cancer stem cell targeting: how to strike the evil at its root. Adv Drug Deliv Rev. 2017;120:89–107. doi:10.1016/j.addr.2017.07.01328736304
  • Tan KX, Danquah MK, Sidhu A, Lau SY, Ongkudon CM. Biophysical characterization of layer‐by‐layer synthesis of aptamer‐drug microparticles for enhanced cell targeting. Biotechnol Prog. 2017.
  • Wang Y, Ma T, Ma S, et al. Fluorometric determination of the antibiotic kanamycin by aptamer-induced FRET quenching and recovery between MoS 2 nanosheets and carbon dots. Microchimica Acta. 2017;184(1):203–210. doi:10.1007/s00604-016-2011-4
  • Jeong S, Rhee Paeng I. Sensitivity and selectivity on aptamer-based assay: the determination of tetracycline residue in bovine milk. ScientificWorldJournal. 2012;159456(10):1. doi:10.1100/2012/159456
  • Hori SI, Herrera A, Rossi JJ, Zhou J. Current advances in aptamers for cancer diagnosis and therapy. Cancers. 2018;10:1. doi:10.3390/cancers10010009
  • Li F, Wang Q, Zhang H, et al. Characterization of a DNA aptamer for ovarian cancer clinical tissue recognition and in vivo imaging. Cell Physiol Biochem. 2018;51(6):2564–2574. doi:10.1159/00049592530562733
  • Chen C, Zhou S, Cai Y, Tang F. Nucleic acid aptamer application in diagnosis and therapy of colorectal cancer based on cell-SELEX technology. NPJ Prec Oncol. 2017;1(1):37. doi:10.1038/s41698-017-0041-y
  • Wu X, Chen J, Wu M, Zhao JX. Aptamers: active targeting ligands for cancer diagnosis and therapy. Theranostics. 2015;5(4):322. doi:10.7150/thno.1025725699094
  • 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.
  • Wang Y, Li Z, Hu D, Lin C-T, Li J, Lin Y. Aptamer/graphene oxide nanocomplex for in situ molecular probing in living cells. J Am Chem Soc. 2010;132(27):9274–9276. doi:10.1021/ja103169v20565095
  • Beals N, Thiagarajan PS, Soehnlen E, et al. Five-part pentameric nanocomplex shows improved efficacy of doxorubicin in CD44+ cancer cells. ACS Omega. 2017;2(11):7702–7713. doi:10.1021/acsomega.7b0116830023561
  • Nitsche A, Kurth A, Dunkhorst A, et al. One-step selection of Vaccinia virus-binding DNA aptamers by MonoLEX. BMC Biotechnol. 2007;7(1):48. doi:10.1186/1472-6750-7-4817697378
  • Darmostuk M, Rimpelova S, Gbelcova H, Ruml T. Current approaches in SELEX: an update to aptamer selection technology. Biotechnol Adv. 2015;33(6):1141–1161. doi:10.1016/j.biotechadv.2015.02.00825708387
  • Han K, Liang Z, Zhou N. Design strategies for aptamer-based biosensors. Sensors. 2010;10(5):4541–4557. doi:10.3390/s10050454122399891
  • Jin B, Wang S, Lin M, et al. Upconversion nanoparticles based FRET aptasensor for rapid and ultrasensitive bacteria detection. Biosens Bioelect. 2017;90:525–533. doi:10.1016/j.bios.2016.10.029
  • Triebenbach AN, Vogl SJ, Leda Lotspeich-Cole DS, Sikes GM, Happ HK. Detection of Francisella tularensis in Alaskan Mosquitoes (Diptera: culicidae) and assessment of a laboratory model for transmission. J Med Entomol. 2010;47(4):639–648. doi:10.1093/jmedent/47.4.63920695280
  • Vivekananda J, Kiel JL. Methods and Compositions for Aptamers Against Anthrax. Google Patents; 2003.
  • Duan N, Wu S, Chen X, Huang Y, Wang Z. Selection and identification of a DNA aptamer targeted to Vibrio parahemolyticus. J Agric Food Chem. 2012;60(16):4034–4038. doi:10.1021/jf300395z22480209
  • Li P, Yu Q, Zhou L, et al. Probing and characterizing the high specific sequences of ssDNA aptamer against SGIV-infected cells. Virus Res. 2018.
  • Kong C, Wang Y, Fodjo EK, Yang G-X, Han F, Shen X-S. Loop-mediated isothermal amplification for visual detection of Vibrio parahaemolyticus using gold nanoparticles. Microchimica Acta. 2018;185(1):35. doi:10.1007/s00604-017-2594-4
  • Ramlal S, Mondal B, Lavu PS, Bhavanashri N, Kingston J. Capture and detection of Staphylococcus aureus with dual labeled aptamers to cell surface components. Int J Food Microbiol. 2018;265:74–83. doi:10.1016/j.ijfoodmicro.2017.11.00229132030
  • Liu X, Gao T, Gao X, et al. An aptamer based sulfadimethoxine assay that uses magnetized upconversion nanoparticles. Microchimica Acta. 2017;184(9):3557–3563. doi:10.1007/s00604-017-2378-x
  • Luzzago C, Locatelli C, Franco A, et al. Clonal diversity, virulence-associated genes and antimicrobial resistance profile of Staphylococcus aureus isolates from nasal cavities and soft tissue infections in wild ruminants in Italian Alps. Vet Microbiol. 2014;170(1–2):157–161. doi:10.1016/j.vetmic.2014.01.01624565474
  • Wu S, Duan N, Shi Z, Fang C, Wang Z. Simultaneous aptasensor for multiplex pathogenic bacteria detection based on multicolor upconversion nanoparticles labels. Anal Chem. 2014;86(6):3100–3107. doi:10.1021/ac404205c24568625
  • Medley CD, Bamrungsap S, Tan W, Smith JE. Aptamer-conjugated nanoparticles for cancer cell detection. Anal Chem. 2011;83(3):727–734. doi:10.1021/ac102263v21218774
  • Niu W, Teng I-T, Chen X, Tan W, Veige AS. Aptamer-mediated selective delivery of a cytotoxic cationic NHC-Au (I) complex to cancer cells. Dalton Trans. 2018;47(1):120–126. doi:10.1039/C7DT02616A
  • Cheng D, Yu M, Fu F, et al. Dual recognition strategy for specific and sensitive detection of bacteria using aptamer-coated magnetic beads and antibiotic-capped gold nanoclusters. Anal Chem. 2015;88(1):820–825. doi:10.1021/acs.analchem.5b0332026641108
  • Charoenphol P, Bermudez H. Aptamer-targeted DNA nanostructures for therapeutic delivery. Mol Pharm. 2014;11(5):1721–1725. doi:10.1021/mp500047b24739136
  • Sun T-M, Du J-Z, Yao Y-D, et al. Simultaneous delivery of siRNA and paclitaxel via a “Two-in-one” micelleplex promotes synergistic tumor suppression. ACS Nano. 2011;5(2):1483–1494. doi:10.1021/nn103349h21204585
  • Farokhzad OC, Jon S, Khademhosseini A, Tran T-NT, LaVan DA, Langer R. Nanoparticle-aptamer bioconjugates: a new approach for targeting prostate cancer cells. Cancer Res. 2004;64(21):7668–7672. doi:10.1158/0008-5472.CAN-04-255015520166
  • Pang X, Cui C, Wan S, et al. Bioapplications of Cell-SELEX-generated aptamers in cancer diagnostics, therapeutics, theranostics and biomarker discovery: a comprehensive review. Cancers. 2018;10(2):47. doi:10.3390/cancers10020047
  • Bamrungsap S, Chen T, Shukoor MI, et al. Pattern recognition of cancer cells using aptamer-conjugated magnetic nanoparticles. ACS Nano. 2012;6(5):3974–3981. doi:10.1021/nn300232822424140
  • Bidard F-C, Fehm T, Ignatiadis M, et al. Clinical application of circulating tumor cells in breast cancer: overview of the current interventional trials. Cancer Metastasis Rev. 2013;32(1–2):179–188. doi:10.1007/s10555-012-9398-023129208
  • DeSantis C, Ma J, Bryan L, Jemal A. Breast cancer statistics, 2013. CA Cancer J Clin. 2014;64(1):52–62. doi:10.3322/caac.2120324114568
  • Riethdorf S, O’Flaherty L, Hille C, Pantel K. Clinical applications of the CellSearch platform in cancer patients. Adv Drug Deliv Rev. 2018;125:102–121. doi:10.1016/j.addr.2018.01.01129355669
  • Jo H, Her J, Ban C. Dual aptamer-functionalized silica nanoparticles for the highly sensitive detection of breast cancer. Biosens Bioelect. 2015;71:129–136. doi:10.1016/j.bios.2015.04.030
  • Mittal S, Kaur H, Gautam N, Mantha AK. Biosensors for breast cancer diagnosis: a review of bioreceptors, biotransducers and signal amplification strategies. Biosens Bioelect. 2017;88:217–231. doi:10.1016/j.bios.2016.08.028
  • Bernaus A, Gaona X, Esbrí JM, Higueras P, Falkenberg G, Valiente M. Microprobe techniques for speciation analysis and geochemical characterization of mine environments: the mercury district of Almadén in Spain. Environ Sci Technol. 2006;40(13):4090–4095. doi:10.1021/es052392l16856721
  • Tanaka Y, Oda S, Yamaguchi H, Kondo Y, Kojima C, Ono A. 15N− 15N J-coupling across HgII: direct observation of HgII-mediated T− T base pairs in a DNA duplex. J Am Chem Soc. 2007;129(2):244–245. doi:10.1021/ja065552h17212382
  • Zhao Y, Xie X. A novel electrochemical aptamer biosensor based on DNAzyme decorated Au@ Ag core-shell nanoparticles for Hg2+ determination. J Braz Chem Soc. 2018;29(2):232–239.
  • Tan D, He Y, Xing X, Zhao Y, Tang H, Pang D. Aptamer functionalized gold nanoparticles based fluorescent probe for the detection of mercury (II) ion in aqueous solution. Talanta. 2013;113:26–30. doi:10.1016/j.talanta.2013.03.05523708619
  • Farzin L, Shamsipur M, Sheibani S. A review: aptamer-based analytical strategies using the nanomaterials for environmental and human monitoring of toxic heavy metals. Talanta. 2017;174:619–627. doi:10.1016/j.talanta.2017.06.06628738631
  • Lan L, Yao Y, Ping J, Ying Y. Recent progress in nanomaterial-based optical aptamer assay for the detection of food chemical contaminants. ACS Appl Mater Interfaces. 2017;9(28):23287–23301. doi:10.1021/acsami.7b0393728632380
  • Tian J. Aptamer-based colorimetric detection of various targets based on catalytic Au NPs/Graphene nanohybrids. Sens Bio-Sensing Res. 2019;22:100258. doi:10.1016/j.sbsr.2019.100258
  • Giorgi-Coll S, Marín MJ, Sule O, Hutchinson PJ, Carpenter KLH. Aptamer-modified gold nanoparticles for rapid aggregation-based detection of inflammation: an optical assay for interleukin-6. Microchimica Acta. 2019;187(1):13. doi:10.1007/s00604-019-3975-731802241
  • Srivastava M, Nirala NR, Srivastava SK, Prakash R. A comparative study of aptasensor vs immunosensor for label-free PSA cancer detection on GQDs-AuNRs modified screen-printed electrodes. Sci Rep. 2018;8(1):1923. doi:10.1038/s41598-018-19733-z29386538
  • Shen H, Wang J, Liu H, et al. Rapid and selective detection of pathogenic bacteria in bloodstream infections with aptamer-based recognition. ACS Appl Mater Interfaces. 2016;8(30):19371–19378. doi:10.1021/acsami.6b0667127411775
  • Park JY, Jeong HY, Kim MI. Colorimetric detection system for salmonella typhimurium based on peroxidase-like activity of magnetic nanoparticles with DNA aptamers. J Nanomater. 2015;2015.
  • Ozyurt C, Bora B, Ugurlu O, Evran S. Chapter 18 - Pathogen-specific nucleic acid aptamers as targeting components of antibiotic and gene delivery systems In: Andronescu E, Grumezescu AM, editors. Nanostructures for Drug Delivery. Elsevier; 2017:551–577.
  • Lee B, Park J, Ryu M, et al. Antimicrobial peptide-loaded gold nanoparticle-DNA aptamer conjugates as highly effective antibacterial therapeutics against Vibrio vulnificus. Sci Rep. 2017;7(1):13572. doi:10.1038/s41598-017-14127-z29051620
  • Aslam B, Wang W, Arshad MI, et al. Antibiotic resistance: a rundown of a global crisis. Infect Drug Resist. 2018;11:1645–1658.30349322
  • Zhu C-L, Wang X-W, Lin -Z-Z, Xie Z-H, Wang X-R. Cell microenvironment stimuli-responsive controlled-release delivery systems based on mesoporous silica nanoparticles. J Food Drug Anal. 2014;22(1):18–28. doi:10.1016/j.jfda.2014.01.00224673901
  • Ladju RB, Pascut D, Massi MN, Tiribelli C, Sukowati CH. Aptamer: a potential oligonucleotide nanomedicine in the diagnosis and treatment of hepatocellular carcinoma. Oncotarget. 2018;9(2):2951.29416827
  • Shigdar S. What potential do aptamers hold in therapeutic delivery? Future Sci. 2017.
  • Kavruk M, Celikbicak O, Ozalp V, et al. Antibiotic loaded nanocapsules functionalized with aptamer gates for targeted destruction of pathogens. Chem Commun. 2015;51(40):8492–8495. doi:10.1039/C5CC01869B
  • Sancenón F, Yu E, Aznar E, Marcos MD, Martínez-Máñez R. Gated porous materials for biomedical applications. Drug Del Syst. 2017;1:113.
  • Zhu C-L., Lu C-H., Song X-Y., Yang H-H., Wang X-R.. Bioresponsive controlled release using mesoporous silica nanoparticles capped with aptamer-based molecular gate. J Am Chem Soc. 2011;133(5):1278–1281. doi:10.1021/ja110094g21214180
  • Elsabahy M, Nazarali A, Foldvari M. Non-viral nucleic acid delivery: key challenges and future directions. Curr Drug Deliv. 2011;8(3):235–244. doi:10.2174/15672011179525617421291381
  • Sadeqzadeh E, Rahbarizadeh F, Ahmadvand D, Rasaee MJ, Parhamifar L, Moghimi SM. Combined MUC1-specific nanobody-tagged PEG-polyethylenimine polyplex targeting and transcriptional targeting of tBid transgene for directed killing of MUC1 over-expressing tumour cells. J Controlled Release. 2011;156(1):85–91. doi:10.1016/j.jconrel.2011.06.022
  • Khaleghi S, Rahbarizadeh F, Ahmadvand D, Hosseini HRM. Anti-HER2 VHH targeted magnetoliposome for intelligent magnetic resonance imaging of breast cancer cells. Cell Mol Bioeng. 2017;10(3):263–272.31719864
  • Yin L, Yuvienco C, Montclare JK. Protein based therapeutic delivery agents: contemporary developments and challenges. Biomaterials. 2017;134:91–116. doi:10.1016/j.biomaterials.2017.04.03628458031
  • Lam P, Steinmetz NF. Plant viral and bacteriophage delivery of nucleic acid therapeutics. Wiley Interdiscipl Rev. 2018;10:1.
  • Pan J, Ruan W, Qin M, et al. Intradermal delivery of STAT3 siRNA to treat melanoma via dissolving microneedles. Sci Rep. 2018;8(1):1117. doi:10.1038/s41598-018-19463-229348670
  • Li L, Hou J, Liu X, et al. Nucleolin-targeting liposomes guided by aptamer AS1411 for the delivery of siRNA for the treatment of malignant melanomas. Biomaterials. 2014;35(12):3840–3850. doi:10.1016/j.biomaterials.2014.01.01924486214
  • Wu S, Li J, Liang H, et al. Aptamer-based self-assembled supramolecular vesicles for pH-responsive targeted drug delivery. Sci China Chem. 2017;60(5):628–634. doi:10.1007/s11426-016-0351-5
  • Bagalkot V, Gao X. siRNA-Aptamer Chimeras on Nanoparticles: Preserving Targeting Functionality for Effective Gene Silencing. ACS Nano. 2011;5(10):8131–8139. doi:10.1021/nn202772p21936502
  • Zintchenko A, Philipp A, Dehshahri A, Wagner E. Simple modifications of branched PEI lead to highly efficient siRNA carriers with low toxicity. Bioconjug Chem. 2008;19(7):1448–1455. doi:10.1021/bc800065f18553894
  • Nimesh S, Aggarwal A, Kumar P, Singh Y, Gupta K, Chandra R. Influence of acyl chain length on transfection mediated by acylated PEI nanoparticles. Int J Pharm. 2007;337(1–2):265–274. doi:10.1016/j.ijpharm.2006.12.03217254724
  • Mazur J, Roy K, Kanwar JR. Recent advances in nanomedicine and survivin targeting in brain cancers. Nanomedicine. 2018;13(1):105–137. doi:10.2217/nnm-2017-028629161215
  • Urmann K, Modrejewski J, Scheper T, Walter J-G. Aptamer-modified nanomaterials: principles and applications. BioNanoMaterials. 2017;18:1–2.
  • Kurosaki T, Higuchi N, Kawakami S, et al. Self-assemble gene delivery system for molecular targeting using nucleic acid aptamer. Gene. 2012;491(2):205–209. doi:10.1016/j.gene.2011.09.02122001405
  • Demeneix B, Behr JP. Polyethylenimine (PEI). Adv Genet. 2005;53:215–230.
  • Qu J, Yu S, Zheng Y, Zheng Y, Yang H, Zhang J. Aptamer and its applications in neurodegenerative diseases. Cell Mol Life Sci. 2017;74(4):683–695. doi:10.1007/s00018-016-2345-427563707
  • Landi A, Babiuk LA, van Drunen Littel-van den Hurk S. High transfection efficiency, gene expression, and viability of monocyte-derived human dendritic cells after nonviral gene transfer. J Leukoc Biol. 2007;82(4):849–860. doi:10.1189/jlb.090656117626798
  • Shahidi‐Hamedani N, Shier WT, Moghadam Ariaee F, Abnous K, Ramezani M. Targeted gene delivery with noncovalent electrostatic conjugates of sgc‐8c aptamer and polyethylenimine. J Gene Med. 2013;15(6–7):261–269.23794147
  • Askarian S, Abnous K, Ayatollahi S, Farzad SA, Oskuee RK, Ramezani M. PAMAM-pullulan conjugates as targeted gene carriers for liver cell. Carbohydr Polym. 2017;157:929–937. doi:10.1016/j.carbpol.2016.10.03027988010
  • Ozyurt C, Bora B, Ugurlu O, Evran S. Pathogen-specific nucleic acid aptamers as targeting components of antibiotic and gene delivery systems. Nanostruct Drug Del. 2017;551–577.
  • Cheng J, Teply BA, Sherifi I, et al. Formulation of functionalized PLGA–PEG nanoparticles for in vivo targeted drug delivery. Biomaterials. 2007;28(5):869–876. doi:10.1016/j.biomaterials.2006.09.04717055572
  • Elminger MW, Bell M, Schüett BS, Langkamp M, Kutoh E, Ranke MB. Transactivation of the IGFBP-2 promoter in human tumor cell lines. Mol Cell Endocrinol. 2001;175(1–2):211–218. doi:10.1016/S0303-7207(00)00454-811325531
  • Reslan L, Mestas J-L, Herveau S, Béra J-C, Dumontet C. Transfection of cells in suspension by ultrasound cavitation. J Controlled Release. 2010;142(2):251–258. doi:10.1016/j.jconrel.2009.10.029
  • Emami J, Maghzi P, Hasanzadeh F, Sadeghi H, Mirian M, Rostami M. PLGA-PEG-RA-based polymeric micelles for tumor targeted delivery of irinotecan. Pharm Dev Technol. 2018;23(1):41–54. doi:10.1080/10837450.2017.134095028608760
  • Subramanian N, Kanwar JR, Athalya P, et al. EpCAM aptamer mediated cancer cell specific delivery of EpCAM siRNA using polymeric nanocomplex. J Biomed Sci. 2015;22(1):4. doi:10.1186/s12929-014-0108-925576037
  • Sarrach S, Huang Y, Niedermeyer S, et al. Spatiotemporal patterning of EpCAM is important for murine embryonic endo-and mesodermal differentiation. Sci Rep. 2018;8(1):1801. doi:10.1038/s41598-018-20131-829379062
  • Castilla C, Congregado B, Chinchón D, Torrubia FJ, Japón MA, Sáez C. Bcl-xL is overexpressed in hormone-resistant prostate cancer and promotes survival of LNCaP cells via interaction with proapoptotic Bak. Endocrinology. 2006;147(10):4960–4967. doi:10.1210/en.2006-050216794010
  • Beck-Broichsitter M, Merkel OM, Kissel T. Controlled pulmonary drug and gene delivery using polymeric nano-carriers. J Controlled Release. 2012;161(2):214–224. doi:10.1016/j.jconrel.2011.12.004
  • Wang L, Zhou Y, Wu M, et al. Functional nanocarrier for drug and gene delivery via local administration in mucosal tissues. Nanomedicine. 2018;13(1):69–88. doi:10.2217/nnm-2017-014329173025
  • Yamazoe H. Multifunctional protein microparticles for medical applications. Biomaterials. 2018;155:1–12. doi:10.1016/j.biomaterials.2017.10.04529154040
  • Li X, Chen Y, Wang M, Ma Y, Xia W, Gu H. A mesoporous silica nanoparticle–PEI–fusogenic peptide system for siRNA delivery in cancer therapy. Biomaterials. 2013;34(4):1391–1401. doi:10.1016/j.biomaterials.2012.10.07223164421
  • Yin Q, Gao Y, Zhang Z, Zhang P, Li Y. Bioreducible poly (β-amino esters)/shRNA complex nanoparticles for efficient RNA delivery. J Controlled Release. 2011;151(1):35–44. doi:10.1016/j.jconrel.2010.12.014
  • Parhiz H, Hashemi M, Hatefi A, Shier WT, Farzad SA, Ramezani M. Arginine-rich hydrophobic polyethylenimine: potent agent with simple components for nucleic acid delivery. Int J Biol Macromol. 2013;60:18–27. doi:10.1016/j.ijbiomac.2013.05.00123680600
  • Nia AH, Behnam B, Taghavi S, et al. Evaluation of chemical modification effects on DNA plasmid transfection efficiency of single-walled carbon nanotube–succinate–polyethylenimine conjugates as non-viral gene carriers. MedChemComm. 2017;8(2):364–375. doi:10.1039/C6MD00481D30108752
  • Kesharwani P, Gajbhiye V, Jain NK. A review of nanocarriers for the delivery of small interfering RNA. Biomaterials. 2012;33(29):7138–7150. doi:10.1016/j.biomaterials.2012.06.06822796160
  • Moradian H, Fasehee H, Keshvari H, Faghihi S. Poly (ethyleneimine) functionalized carbon nanotubes as efficient nano-vector for transfecting mesenchymal stem cells. Colloids Surf B. 2014;122:115–125. doi:10.1016/j.colsurfb.2014.06.056
  • Mohammadi M, Salmasi Z, Hashemi M, Mosaffa F, Abnous K, Ramezani M. Single-walled carbon nanotubes functionalized with aptamer and piperazine–polyethylenimine derivative for targeted siRNA delivery into breast cancer cells. Int J Pharm. 2015;485(1):50–60. doi:10.1016/j.ijpharm.2015.02.03125712164
  • Subramanian N, Raghunathan V, Kanwar JR, et al. Target-specific delivery of doxorubicin to retinoblastoma using epithelial cell adhesion molecule aptamer. Mol Vis. 2012;18:2783.23213278
  • Macdonald J, Henri J, Roy K, et al. EpCAM immunotherapy versus specific targeted delivery of drugs. Cancers. 2018;10(1):19. doi:10.3390/cancers10010019
  • Taghavi S, HashemNia A, Mosaffa F, Askarian S, Abnous K, Ramezani M. Preparation and evaluation of polyethylenimine-functionalized carbon nanotubes tagged with 5TR1 aptamer for targeted delivery of Bcl-xL shRNA into breast cancer cells. Colloids Surf B. 2016;140:28–39. doi:10.1016/j.colsurfb.2015.12.021
  • Singh R, Pantarotto D, McCarthy D, et al. Binding and condensation of plasmid DNA onto functionalized carbon nanotubes: toward the construction of nanotube-based gene delivery vectors. J Am Chem Soc. 2005;127(12):4388–4396.15783221
  • Ferreira C, Matthews C, Missailidis S. DNA aptamers that bind to MUC1 tumour marker: design and characterization of MUC1-binding single-stranded DNA aptamers. Tumor Biol. 2006;27(6):289–301. doi:10.1159/000096085
  • Yang B, Chen B, He M, Yin X, Xu C, Hu B. An aptamer-based dual-functional probe for rapid and specific counting and imaging of MCF-7 cells. Anal Chem. 2018.
  • Charbgoo F, Alibolandi M, Taghdisi SM, Abnous K, Soltani F, Ramezani M. MUC1 aptamer-targeted DNA micelles for dual tumor therapy using doxorubicin and KLA peptide. Nanomedicine. 2018;14(3):685–697. doi:10.1016/j.nano.2017.12.01029317345
  • Ebrahimian M, Taghavi S, Mokhtarzadeh A, Ramezani M, Hashemi M. Co-delivery of doxorubicin encapsulated PLGA nanoparticles and Bcl-xL shRNA using alkyl-modified PEI into breast cancer cells. Appl Biochem Biotechnol. 2017;183(1):126–136. doi:10.1007/s12010-017-2434-328236188
  • Afsharzadeh M, Hashemi M, Mokhtarzadeh A, Abnous K, Ramezani M. Recent advances in co-delivery systems based on polymeric nanoparticle for cancer treatment. Artif Cells Nanomed Biotechnol. 2017;1–16.
  • Zhang RX, Ahmed T, Li LY, Li J, Abbasi AZ, Wu XY. Design of nanocarriers for nanoscale drug delivery to enhance cancer treatment using hybrid polymer and lipid building blocks. Nanoscale. 2017;9(4):1334–1355. doi:10.1039/C6NR08486A27973629
  • Ståhl S, Gräslund T, Karlström AE, Frejd FY, Nygren P-Å, Löfblom J. Affibody molecules in biotechnological and medical applications. Trends Biotechnol. 2017;35(8):691–712. doi:10.1016/j.tibtech.2017.04.00728514998
  • Taghavi S, Nia AH, Abnous K, Ramezani M. Polyethylenimine-functionalized carbon nanotubes tagged with AS1411 aptamer for combination gene and drug delivery into human gastric cancer cells. Int J Pharm. 2017;516(1–2):301–312. doi:10.1016/j.ijpharm.2016.11.02727840158
  • Tyagi P, Subramony JA. Nanotherapeutics in oral and parenteral drug delivery: key learnings and future outlooks as we think small. J Controlled Release. 2018;272:159–168. doi:10.1016/j.jconrel.2018.01.009
  • Gou Y, Zhang Z, Li D, et al. HSA-based multi-target combination therapy: regulating drugs’ release from HSA and overcoming single drug resistance in a breast cancer model. Drug Deliv. 2018;25(1):321–329. doi:10.1080/10717544.2018.142824529350051
  • D’ANGELO B, Benedetti E, Cimini A, Giordano A. MicroRNAs: a puzzling tool in cancer diagnostics and therapy. Anticancer Res. 2016;36(11):5571–5575. doi:10.21873/anticanres.1114227793880
  • Duan Y, Xing Z, Yang J, et al. Chondroitin sulfate-functionalized polyamidoamine-mediated miR-34a delivery for inhibiting the proliferation and migration of pancreatic cancer. RSC Adv. 2016;6(75):70870–70876. doi:10.1039/C6RA15716E
  • Wu D, Wang C, Yang J, et al. Improving the intracellular drug concentration in lung cancer treatment through the codelivery of doxorubicin and miR-519c mediated by porous PLGA microparticle. Mol Pharm. 2016;13(11):3925–3933. doi:10.1021/acs.molpharmaceut.6b0070227684197
  • Wang Y, Chen J, Liang X, et al. An ATP-responsive codelivery system of doxorubicin and miR-34a to synergistically inhibit cell proliferation and migration. Mol Pharm. 2017;14(7):2323–2332. doi:10.1021/acs.molpharmaceut.7b0018428591517
  • Tang Q, Ma X, Zhang Y, Cai X, Xue W, Ma D. Self-sensibilized polymeric prodrug co-delivering MMP-9 shRNA plasmid for combined treatment of tumors. Acta Biomater. 2018;69:277–289. doi:10.1016/j.actbio.2018.01.01429369806
  • Yang Y, Xie X, Yang Y, et al. Polymer nanoparticles modified with photo-and pH-dual-responsive polypeptides for enhanced and targeted cancer therapy. Mol Pharm. 2016;13(5):1508–1519. doi:10.1021/acs.molpharmaceut.5b0097727043442
  • Yu S, He C, Lv Q, et al. pH and reduction-sensitive disulfide cross-linked polyurethane micelles for bio-triggered anti-tumor drug delivery. J Controlled Release. 2015;213:e99–e100. doi:10.1016/j.jconrel.2015.05.166
  • Yang S, Ren Z, Chen M, et al. Nucleolin-targeting AS1411-aptamer-modified graft polymeric micelle with Dual pH/redox sensitivity designed to enhance tumor therapy through the codelivery of doxorubicin/TLR4 siRNA and suppression of invasion. Mol Pharm. 2017.
  • Zhang C, Yang S, Zhu W, et al. Distinctive polymer micelle designed for siRNA delivery and reversal of MDR1 gene‐dependent multidrug resistance. J Biomed Mater Res Part B. 2017;105(7):2093–2106. doi:10.1002/jbm.b.33748
  • Yi Y, Wang H, Wang X, et al. Photocontrollable drug release nanosystem for multifunctional synergistic cancer therapy. acs appl mater interfaces. 2017;9(7):5847–5854. doi:10.1021/acsami.6b1541428124556
  • Liu Z, Sun X, Xiao S, et al. Characterization of aptamer-mediated gene delivery system for liver cancer therapy. Oncotarget. 2017;9:6.
  • Shahidi-Hamedani N, Shier WT, Moghadam Ariaee F, Abnous K, Ramezani M. Targeted gene delivery with noncovalent electrostatic conjugates of sgc-8c aptamer and polyethylenimine. J Gene Med. 2013;15(6–7):261–269.23794147
  • Wang G-H, Huang G-L, Zhao Y, et al. ATP triggered drug release and DNA co-delivery systems based on ATP responsive aptamers and polyethylenimine complexes. J Mater Chem B. 2016;4(21):3832–3841. doi:10.1039/C5TB02764K32263321
  • Lee J, Oh J, Lee E-S, Kim Y-P, Lee M. Conjugation of prostate cancer-specific aptamers to polyethylene glycol-grafted polyethylenimine for enhanced gene delivery to prostate cancer cells. J Ind Eng Chem. 2019;73:182–191. doi:10.1016/j.jiec.2019.01.023
  • Wu D, Zhang Y, Xu X, et al. RGD/TAT-functionalized chitosan-graft-PEI-PEG gene nanovector for sustained delivery of NT-3 for potential application in neural regeneration. Acta Biomater. 2018;72:266–277. doi:10.1016/j.actbio.2018.03.03029578088