Progressive cancer targeting by programmable aptamer-tethered nanostructures

Fatemeh Mohammadi , Hamed Zahraee , Farkhonde Zibadi , Zahra Khoshbin , Mohammad Ramezani , Mona Alibolandi , Khalil Abnous , Seyed Mohammad Taghdisi

MedComm ›› 2024, Vol. 5 ›› Issue (11) : e775

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (11) : e775 DOI: 10.1002/mco2.775
REVIEW

Progressive cancer targeting by programmable aptamer-tethered nanostructures

Author information +
History +
PDF

Abstract

Scientific research in recent decades has affirmed an increase in cancer incidence as a cause of death globally. Cancer can be considered a plurality of various diseases rather than a single disease, which can be a multifaceted problem. Hence, cancer therapy techniques acquired more accelerated and urgent approvals compared to other therapeutic approaches. Radiotherapy, chemotherapy, immunotherapy, and surgery have been widely adopted as routine cancer treatment strategies to suppress disease progression and metastasis. These therapeutic approaches have lengthened the longevity of countless cancer patients. Nonetheless, some inherent limitations have restricted their application, including insignificant therapeutic efficacy, toxicity, negligible targeting, non-specific distribution, and multidrug resistance. The development of therapeutic oligomer nanoconstructs with the advantages of chemical solid-phase synthesis, programmable design, and precise adjustment is crucial for advancing smart targeted drug nanocarriers. This review focuses on the significance of the different aptamer-assembled nanoconstructs as multifunctional nucleic acid oligomeric nanoskeletons in efficient drug delivery. We discuss recent advancements in the design and utilization of aptamer-tethered nanostructures to enhance the efficacy of cancer treatment. Valuably, this comprehensive review highlights self-assembled aptamers as the exceptionally intelligent nano-biomaterials for targeted drug delivery based on their superior stability, high specificity, excellent recoverability, inherent biocompatibility, and versatile functions.

Keywords

aptamer / bioimaging / cancer / DNAnanotechnology / gene therapy / nucleic acid nanostructures / targeted drug delivery

Cite this article

Download citation ▾
Fatemeh Mohammadi, Hamed Zahraee, Farkhonde Zibadi, Zahra Khoshbin, Mohammad Ramezani, Mona Alibolandi, Khalil Abnous, Seyed Mohammad Taghdisi. Progressive cancer targeting by programmable aptamer-tethered nanostructures. MedComm, 2024, 5(11): e775 DOI:10.1002/mco2.775

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

BrownJS, AmendSR, AustinRH, Gatenby RA, HammarlundEU, PientaKJ. Updating the definition of cancer. Mol Cancer Res. 2023; 21(11): 1142-1147.

[2]

LinH-Y, ParkJY. Epidemiology of cancer. Anesthesia for Oncological Surgery. Springer; 2024: 11-16.

[3]

ShindeA, Panchal K, PatraP, et al. QbD enabled development and evaluation of pazopanib loaded nanoliposomes for PDAC treatment. AAPS PharmSciTech. 2024; 25(5): 97.

[4]

KesharwaniP, Chandra J, KarimS, GuptaG, Karwasra R, SharmaA. αvβ3 integrin targeting RGD peptide-based nanoparticles as an effective strategy for selective drug delivery to tumor microenvironment. J Drug Del Sci Tech. 2024; 96: 105663.

[5]

AdelM, Kadhim MM, MuttasharHH, HachimSK, Abdullaha SA, RheimaAM. Two-dimensional silicon carbide monolayer as a promising drug delivery vehicle for hydroxyurea anti-cancer drug. Kor J Chem Eng. 2023; 40(6): 1433-1439.

[6]

EhrlichP. Collected Studies on Immunity. John Wiley & Sons; 1910.

[7]

HeS, DuY, TaoH, DuanH. Advances in aptamer-mediated targeted delivery system for cancer treatment. Int J Biol Macromol. 2023; 238: 124173.

[8]

EzikeTC, OkpalaUS, OnojaUL, et al. Advances in drug delivery systems, challenges and future directions. Heliyon. 2023; 9(6): e17488.

[9]

LiuX, JiangB, ChengA, et al. The effects of doxorubicin loaded aptamer S3-linked DNA tetrahedrons on nasopharyngeal carcinoma. J Otolaryngol Head Neck Surg. 2023; 52(1): 79.

[10]

GuoZ, SongH, TianY, et al. SiRNF8 delivered by DNA framework nucleic acid effectively sensitizes chemotherapy in colon cancer. Int J Nanomed. 2024; 19: 171-188.

[11]

TianT, ZhaoC, LiS, et al. Liver-targeted delivery of small interfering RNA of C–C chemokine receptor 2 with tetrahedral framework nucleic acid attenuates liver cirrhosis. ACS Appl Mater Interfaces. 2023; 15(8): 10492-10505.

[12]

CaoM, SunY, XiaoM, et al. Multivalent aptamer-modified DNA origami as drug delivery system for targeted cancer therapy. Chem Res Chin Univ. 2019; 36(2): 254-260.

[13]

ChiQ, YangZ, XuK, WangC, LiangH. DNA nanostructure as an efficient drug delivery platform for immunotherapy. Front Pharma. 2020; 10: 1585.

[14]

ChengJY, MayesAM, RossCA. Nanostructure engineering by templated self-assembly of block copolymers. Nat Mater. 2004; 3(11): 823-828.

[15]

LacroixA, Sleiman HF. DNA nanostructures: current challenges and opportunities for cellular delivery. ACS Nano. 2021; 15(3): 3631-3645.

[16]

IjasH, ShenB, Heuer-JungemannA, et al. Unraveling the interaction between doxorubicin and DNA origami nanostructures for customizable chemotherapeutic drug release. Nucleic Acids Res. 2021; 49(6): 3048-3062.

[17]

WangW, LinM, ChenYR, et al. Y-shaped backbone-rigidified DNA tiles for the construction of supersized nondeformable tetrahedrons for precise cancer therapies. Anal Chem. 2024; 96(4): 1488-1497.

[18]

ZhouJ, RossiJ. Aptamers as targeted therapeutics: current potential and challenges. Nat Rev Drug Discov. 2017; 16(3): 181-202.

[19]

MeyerC, HahnU, RentmeisterA. Cell-specific aptamers as emerging therapeutics. J Nucl Acids. 2011; 2011: 904750.

[20]

KhoshbinZ, Verdian A, TaghdisiSM, DaneshNM, AbnousK. A novel liquid crystal assay based on aptazyme-assisted bioprobe for ultra-sensitive monitoring of lead ion. Sens Actuators B Chem. 2023; 375: 132926.

[21]

MohammadiF, Khoshbin Z, RamezaniM, et al. A simple label-free aptasensor for acrylamide monitoring using a four-way arm junction structure of DNA strands. Microchem J. 2024; 199: 110215.

[22]

ZahraeeH, Khoshbin Z, RamezaniM, AlibolandiM, AbnousK, TaghdisiSM. A tag-free fluorescent aptasensor for tobramycin detection using a hybridization of three aptamer strands and SYBR Green I dye. Spectrochim Acta A Mol Biomol Spectrosc. 2023; 290: 122305.

[23]

GallinaME, ZhouY, JohnsonCJ, et al. Aptamer-conjugated, fluorescent gold nanorods as potential cancer theradiagnostic agents. Mater Sci Eng C. 2016; 59: 324-332.

[24]

XuanW, PengY, DengZ, et al. A basic insight into aptamer–drug conjugates (ApDCs). Biomaterials. 2018; 182: 216-226.

[25]

MoritaY, LeslieM, KameyamaH, Volk DE, TanakaT. Aptamer therapeutics in cancer: current and future. Cancers (Basel). 2018; 10(3): 80.

[26]

MohammadiF, Zahraee H, Izadpanah KazemiM, et al. Recent advances in aptamer-based platforms for cortisol hormone monitoring. Talanta. 2024; 266(pt 1):125010.

[27]

KhoshbinZ, Zahraee H, ZamanianJ, et al. A label-free liquid crystal-assisted aptasensor for trace level detection of tobramycin in milk and chicken egg samples. Anal Chim Acta. 2022; 1236: 340588.

[28]

XingH, HwangK, LiJ, TorabiSF, LuY. DNA aptamer technology for personalized medicine. Curr Opin Chem Eng. 2014; 4: 79-87.

[29]

FanR, TaoX, ZhaiX, et al. Application of aptamer–drug delivery system in the therapy of breast cancer. Biomed Pharmacother. 2023; 161: 114444.

[30]

BagalkotV, Farokhzad OC, LangerR, JonS. An aptamer–doxorubicin physical conjugate as a novel targeted drug-delivery platform. Angew Chem Int Ed. 2006; 45(48): 8149-8152.

[31]

HuY, DuanJ, ZhanQ, Wang F, LuX, YangXD. Novel MUC1 aptamer selectively delivers cytotoxic agent to cancer cells in vitro. PLoS One. 2012; 7(2): e31970.

[32]

HuangYF, Shangguan D, LiuH, et al. Molecular assembly of an aptamer–drug conjugate for targeted drug delivery to tumor cells. Chembiochem. 2009; 10(5): 862-868.

[33]

ParasharA, PandeyKK, YadavML. Different approaches for aptamer conjugated drugs preparation. Aptamers: Biotechnological Applications of a Next Generation Tool. 2019: 91-100.

[34]

HuR, LiuT, ZhangXB, et al. DLISA: a DNAzyme-based ELISA for protein enzyme-free immunoassay of multiple analytes. Anal Chem. 2015; 87(15): 7746-7753.

[35]

FarokhzadOC, KarpJM, LangerR. Nanoparticle–aptamer bioconjugates for cancer targeting. Expert Opin Drug Deliv. 2006; 3(3): 311-324.

[36]

ZhuY, Chandra P, ShimYB. Ultrasensitive and selective electrochemical diagnosis of breast cancer based on a hydrazine-Au nanoparticle–aptamer bioconjugate. Anal Chem. 2013; 85(2): 1058-1064.

[37]

ShiH, YeX, HeX, et al. Au@ Ag/Au nanoparticles assembled with activatable aptamer probes as smart “nano-doctors” for image-guided cancer thermotherapy. Nanoscale. 2014; 6(15): 8754-8761.

[38]

XiaoZ, Farokhzad OC. Aptamer-functionalized nanoparticles for medical applications: challenges and opportunities. ACS Nano. 2012; 6(5): 3670-3676.

[39]

LiJ, PeiH, ZhuB, et al. Self-assembled multivalent DNA nanostructures for noninvasive intracellular delivery of immunostimulatory CpG oligonucleotides. ACS Nano. 2011; 5(11): 8783-8789.

[40]

MengHM, LiuH, KuaiH, Peng R, MoL, ZhangXB. Aptamer-integrated DNA nanostructures for biosensing, bioimaging and cancer therapy. Chem Soc Rev. 2016; 45(9): 2583-2602.

[41]

LeeDS, QianH, TayCY, Leong DT. Cellular processing and destinies of artificial DNA nanostructures. Chem Soc Rev. 2016; 45(15): 4199-4225.

[42]

LiuQ, JinC, WangY, et al. Aptamer-conjugated nanomaterials for specific cancer cell recognition and targeted cancer therapy. NPG Asia Mater. 2014; 6(4): e95.

[43]

ZhouJ, LiuZ, LiF. Upconversion nanophosphors for small-animal imaging. Chem Soc Rev. 2012; 41(3): 1323-1349.

[44]

WatsonJD, CrickFH. Molecular structure of nucleic acids; a structure for deoxyribose nucleic acid. Nature. 1953; 171(4356): 737-738.

[45]

YouX, HeR, LiuS-Y, Dai Z. Advances in fluorescence imaging of RNAs in living cells based on functional nucleic acid probes. Biomed Anal. 2024; 1(1): 1-27.

[46]

YuH, HanX, WangW, et al. Modified unit-mediated strand displacement reactions for direct detection of single nucleotide variants in active double-stranded DNA. ACS Nano. 2024; 18(19): 12401-12411.

[47]

HuangY, RyssyJ, NguyenMK, Loo J, HallstenS, KuzykA. Measuring the affinities of RNA and DNA aptamers with DNA origami-based chiral plasmonic probes. Anal Chem. 2022; 94(50): 17577-17586.

[48]

WenY, PeiH, WanY, et al. DNA nanostructure-decorated surfaces for enhanced aptamer-target binding and electrochemical cocaine sensors. Anal Chem. 2011; 83(19): 7418-7423.

[49]

SinghN, SinghA, DhankaM, Bhatia D. DNA functionalized programmable hybrid biomaterials for targeted multiplexed applications. J Mater Chem B. 2024; 12(30): 7267-7291.

[50]

LiuB, WangF, ChaoJ. Programmable nanostructures based on framework-DNA for applications in biosensing. Sensors (Basel). 2023; 23(6): 3313.

[51]

SeemanNC. The challenge of structural control on the nanoscale: bottom-up self-assembly of nucleic acids in 3D. Int J Nanotechnol. 2005; 2(4): 348-370.

[52]

FeldkampU, Niemeyer CM. Rational design of DNA nanoarchitectures. Angew Chem Int Ed Engl. 2006; 45(12): 1856-1876.

[53]

BennerSA. Rethinking nucleic acids from their origins to their applications. Philos Trans Roy Soc B: Biol Sci. 2023; 378(1871): 20220027.

[54]

FuJ, LiuM, LiuY, YanH. Spatially-interactive biomolecular networks organized by nucleic acid nanostructures. Acc Chem Res. 2012; 45(8): 1215-1226.

[55]

GolubE, AlbadaHB, LiaoWC, Biniuri Y, WillnerI. Nucleoapzymes: hemin/G-quadruplex DNAzyme-aptamer binding site conjugates with superior enzyme-like catalytic functions. J Am Chem Soc. 2016; 138(1): 164-172.

[56]

LvY, PengR, ZhouY, Zhang X, TanW. Catalytic self-assembly of a DNA dendritic complex for efficient gene silencing. Chem Commun (Camb). 2016; 52(7): 1413-1415.

[57]

CaiR, YinF, ChenH, Tian Y, ZhouN. A fluorescent aptasensor for Staphylococcus aureus based on strand displacement amplification and self-assembled DNA hexagonal structure. Microchim Acta. 2020; 187(5): 304.

[58]

HuangG, XieQ, ChiJ, LinC, LinX, XieZ. Magneto-fluorescent nanobiosensor with tetrahedral framework nucleic acid-confined aptamer for trace adenosine triphosphate detection. Sens Actuators B Chem. 2024; 417: 136129.

[59]

AugspurgerEE, RanaM, YigitMV. Chemical and biological sensing using hybridization chain reaction. ACS Sens. 2018; 3(5): 878-902.

[60]

VenkataramanS, DirksRM, RothemundPW, Winfree E, PierceNA. An autonomous polymerization motor powered by DNA hybridization. Nat Nanotechnol. 2007; 2(8): 490-494.

[61]

OuyangX, LiJ, LiuH, et al. Self-assembly of DNA-based drug delivery nanocarriers with rolling circle amplification. Methods. 2014; 67(2): 198-204.

[62]

CarneiroKMM, Hamblin GD, HänniKD, et al. Stimuli-responsive organization of block copolymers on DNA nanotubes. Chem Sci. 2012; 3(6): 1980-1986.

[63]

CaiR, ZhangS, ChenL, Li M, ZhangY, ZhouN. Self-assembled DNA nanoflowers triggered by a DNA walker for highly sensitive electrochemical detection of Staphylococcus aureus. ACS Appl Mater Interfaces. 2021; 13(4): 4905-4914.

[64]

OuyangX, LiJ, LiuH, et al. Rolling circle amplification-based DNA origami nanostructrures for intracellular delivery of immunostimulatory drugs. Small. 2013; 9(18): 3082-3087.

[65]

YuY, LeiY, DongQ, Li Y. Functionalization of Tile-based DNA nanocages with gold nanoparticles (AuNPs) to form AuNP cluster-DNA cage hybrids. ChemNanoMat. 2020; 6(8): 1175-1178.

[66]

ChaiSQ, LvWY, HeJH, et al. Highly sensitive detection of miR-21 through target-activated catalytic hairpin assembly of X-shaped DNA nanostructures. Anal Chem. 2021; 93(43): 14545-14551.

[67]

ZhangX, Yadavalli VK. Functional self-assembled DNA nanostructures for molecular recognition. Nanoscale. 2012; 4(7): 2439-2446.

[68]

GuoT, XiangY, LuH, et al. Interfacial DNA framework-enhanced background-to-signal transition for ultrasensitive and specific micro-RNA detection. ACS Appl Mater Interfaces. 2022; 14(16): 18209-18218.

[69]

JiangY, ZhouH, ZhaoW, Zhang S. ATP-triggered drug release of self-assembled 3D DNA nanostructures for fluorescence imaging and tumor therapy. Anal Chem. 2022; 94(18): 6771-6780.

[70]

ZhouF, LiuH. Direct nanofabrication using DNA nanostructure. Methods Mol Biol. 2017; 1500: 217-235.

[71]

SuoZ, LiangR, LiuR, et al. A convenient paper-based fluorescent aptasensor for high-throughput detection of Pb2+ in multiple real samples (water–soil–food). Anal Chim Acta. 2023; 1239: 340714.

[72]

KumarA, AhmadA, AnsariMM, et al. Functionalized-DNA Nanostructures as Potential Targeted Drug Delivery Systems for Cancer Therapy. Elsevier; 2022: 54-68.

[73]

StoltenbergRM, Woolley AT. DNA-templated nanowire fabrication. Biomed Microdevices. 2004; 6(2): 105-111.

[74]

WangDX, WangJ, WangYX, et al. DNA nanostructure-based nucleic acid probes: construction and biological applications. Chem Sci. 2021; 12(22): 7602-7622.

[75]

WangH, LuoD, WangH, Wang F, LiuX. Construction of smart stimuli-responsive DNA nanostructures for biomedical applications. Chemistry. 2021; 27(12): 3929-3943.

[76]

RenK, XuY, LiuY, YangM, JuH. A responsive “nano string light” for highly efficient mRNA imaging in living cells via accelerated DNA cascade reaction. ACS Nano. 2017; 12(1): 263-271.

[77]

YangF, ChengY, CaoY, et al. MicroRNA triggered DNA “nano wheel” for visualizing intracellular microRNA via localized DNA cascade reaction. Anal Chem. 2019; 91(15): 9828-9835.

[78]

ChenQ, LiC, YangX, et al. Self-assembled DNA nanowires as quantitative dual-drug nanocarriers for antitumor chemophotodynamic combination therapy. J Mater Chem B. 2017; 5(36): 7529-7537.

[79]

SeemanNC. Nucleic acid junctions and lattices. J Theor Biol. 1982; 99(2): 237-47.

[80]

WinfreeE, LiuF, WenzlerLA, Seeman NC. Design and self-assembly of two-dimensional DNA crystals. Nature. 1998; 394(6693): 539-544.

[81]

ZhouJ, RossiJJ. Cell-specific aptamer-mediated targeted drug delivery. Oligonucleotides. 2011; 21(1): 1-10.

[82]

QiZ, WeiC, ZhangF, Wang Z, ZuoX. Tetrahedral DNA frameworks for biosensing and imaging analysis in living cells. Nano Today. 2024; 54: 102127.

[83]

KimJ, JangD, ParkH, Jung S, KimDH, KimWJ. Functional-DNA-driven dynamic nanoconstructs for biomolecule capture and drug delivery. Adv Mater. 2018; 30(45): e1707351.

[84]

KhoshbinZ, Moeenfard M, ZahraeeH, DavoodianN. A fluorescence imaging-supported aptasensor for sensitive monitoring of cadmium pollutant in diverse samples: a critical role of metal organic frameworks. Talanta. 2022; 246: 123514.

[85]

WeiM, XinL, FengS, Liu Y. Simultaneous electrochemical determination of ochratoxin A and fumonisin B1 with an aptasensor based on the use of a Y-shaped DNA structure on gold nanorods. Mikrochim Acta. 2020; 187(2): 102.

[86]

ZhangK, CaoJ, WuY, et al. A fluorometric aptamer method for kanamycin by applying a dual amplification strategy and using double Y-shaped DNA probes on a gold bar and on magnetite nanoparticles. Mikrochim Acta. 2019; 186(2): 120.

[87]

WangZ, QinW, ZhuangJ, et al. Virus-mimicking cell capture using heterovalency magnetic DNA nanoclaws. ACS Appl Mater Interfaces. 2019; 11(13): 12244-12252.

[88]

HuangJ, WuY, HeH, et al. Acidic microenvironment triggered in situ assembly of activatable three-arm aptamer nanoclaw for contrast-enhanced imaging and tumor growth inhibition in vivo. Theranostics. 2022; 12(7): 3474-3487.

[89]

KadrmasJL, RavinAJ, LeontisNB. Relative stabilities of DNA three-way, four-way and five-way junctions (multi-helix junction loops): unpaired nucleotides can be stabilizing or destabilizing. Nucleic Acids Res. 1995; 23(12): 2212-2222.

[90]

IvensE, Cominetti MMD, SearceyM. Junctions in DNA: underexplored targets for therapeutic intervention. Bioorg Med Chem. 2022; 69: 116897.

[91]

HollidayR. A mechanism for gene conversion in fungi. Genet Res. 2009; 5(2): 282-304.

[92]

DuckettDR, Murchie AI, DiekmannS, von KitzingE, KemperB, LilleyDM. The structure of the Holliday junction, and its resolution. Cell. 1988; 55(1): 79-89.

[93]

KallenbachNR, MaR-I, SeemanNC. An immobile nucleic acid junction constructed from oligonucleotides. Nature. 1983; 305(5937): 829-831.

[94]

FuTJ, SeemanNC. DNA double-crossover molecules. Biochemistry. 1993; 32(13): 3211-3220.

[95]

LaingBM, Juliano RL. DNA three-way junctions stabilized by hydrophobic interactions for creation of functional nanostructures. Chembiochem. 2015; 16(9): 1284-1287.

[96]

YeJ, GaoJ, HuangW, Yuan R, XuW. Bifunctional three-way DNA junction-based strand displacement recycling for amplifiable electrochemical bivariate biosensing. Sens Actuators B Chem. 2023; 390: 133986.

[97]

DuckettDR, LilleyDM. The three-way DNA junction is a Y-shaped molecule in which there is no helix-helix stacking. EMBO J. 1990; 9(5): 1659-1664.

[98]

ZahraeeH, Khoshbin Z, MohammadiF, MashreghiM, AbnousK, TaghdisiSM. Three-way junction skeleton biosensors based on aptamers, DNAzymes, and DNA hybridization probes. TrAC Trend Anal Chem. 2023; 165: 117160.

[99]

MuhuriS, MimuraK, MiyoshiD, Sugimoto N. Stabilization of three-way junctions of DNA under molecular crowding conditions. J Am Chem Soc. 2009; 131(26): 9268-9280.

[100]

LiY, TsengYD, KwonSY, et al. Controlled assembly of dendrimer-like DNA. Nat Mater. 2004; 3(1): 38-42.

[101]

JeongEH, JeongH, JangB, et al. Aptamer-incorporated DNA Holliday junction for the targeted delivery of siRNA. J Indust Eng Chem. 2017; 56: 55-61.

[102]

KhajavianZ, Esmaelpourfarkhani M, RamezaniM, AlibolandiM, AbnousK, TaghdisiSM. A highly sensitive, simple and label-free fluorescent aptasensor for tobramycin sensing based on PicoGreen intercalation into DNA duplex regions of three-way junction origami. Microchem J. 2021; 160: 105657.

[103]

WuC, HanD, ChenT, et al. Building a multifunctional aptamer-based DNA nanoassembly for targeted cancer therapy. J Am Chem Soc. 2013; 135(49): 18644-18650.

[104]

ZhangL, GuoS, ZhuJ, et al. Engineering DNA three-way junction with multifunctional moieties: sensing platform for bioanalysis. Anal Chem. 2015; 87(22): 11295-11300.

[105]

RothemundPW. Folding DNA to create nanoscale shapes and patterns. Nature. 2006; 440(7082): 297-302.

[106]

MeyerT. Engineering a Multi-Functional DNA Origami Nanorod for the Control of Nanoscale Processes. 2019.

[107]

ChenX, JiaB, LuZ, LiaoL, YuH, LiZ. Aptamer-integrated scaffolds for biologically functional DNA origami structures. ACS Appl Mater Interfaces. 2021; 13(33): 39711-39718.

[108]

EklundAS, Comberlato A, ParishIA, JungmannR, Bastings MMC. Quantification of strand accessibility in biostable DNA origami with single-staple resolution. ACS Nano. 2021; 15(11): 17668-17677.

[109]

ZhangK, FanZ, YaoB, et al. Entropy-driven electrochemiluminescence ultra-sensitive detection strategy of NF-kappaB p50 as the regulator of cytokine storm. Biosens Bioelectron. 2021; 176: 112942.

[110]

AroraAA, de Silva C. Beyond the smiley face: applications of structural DNA nanotechnology. Nano Rev Exp. 2018; 9(1): 1430976.

[111]

DouglasSM, DietzH, LiedlT, Högberg B, GrafF, ShihWMJN. Self-assembly of DNA into nanoscale three-dimensional shapes. Nature. 2009; 459(7245): 414-418.

[112]

GodonogaM, LinTY, OshimaA, et al. A DNA aptamer recognising a malaria protein biomarker can function as part of a DNA origami assembly. Sci Rep. 2016; 6(1): 21266.

[113]

XingC, ChenS, LinQ, et al. An aptamer-tethered DNA origami amplifier for sensitive and accurate imaging of intracellular microRNA. Nanoscale. 2022; 14(4): 1327-1332.

[114]

ChenQ, WangX, ChenJ, et al. Multiple-aptamer-integrated DNA-origami-based chemical nose sensors for accurate identification of cancer cells. Anal Chem. 2022; 94(28): 10192-10197.

[115]

GilboaE, McNamara J 2nd, PastorF. Use of oligonucleotide aptamer ligands to modulate the function of immune receptors. Clin Cancer Res. 2013; 19(5): 1054-1062.

[116]

DollinsCM, NairS, BoczkowskiD, et al. Assembling OX40 aptamers on a molecular scaffold to create a receptor-activating aptamer. Chem Biol. 2008; 15(7): 675-682.

[117]

KuaiH, ZhaoZ, MoL, et al. Circular bivalent aptamers enable in vivo stability and recognition. J Am Chem Soc. 2017; 139(27): 9128-9131.

[118]

YangY, SunX, XuJ, et al. Circular bispecific aptamer-mediated artificial intercellular recognition for targeted T cell immunotherapy. ACS Nano. 2020; 14(8): 9562-9571.

[119]

AbnousK, DaneshNM, RamezaniM, Charbgoo F, BahreyniA, TaghdisiSM. Targeted delivery of doxorubicin to cancer cells by a cruciform DNA nanostructure composed of AS1411 and FOXM1 aptamers. Expert Opin Drug Deliv. 2018; 15(11): 1045-1052.

[120]

ShiH, LeiY, GeJ, et al. A simple, pH-activatable fluorescent aptamer probe with ultralow background for bispecific tumor imaging. Anal Chem. 2019; 91(14): 9154-9160.

[121]

FuX, PengF, LeeJ, et al. Aptamer-functionalized DNA nanostructures for biological applications. Top Curr Chem (Cham). 2020; 378(2): 21.

[122]

ZhangXJ, ZhaoZ, WangX, et al. A versatile strategy for convenient circular bivalent functional nucleic acids construction. Natl Sci Rev. 2023; 10(2): nwac107.

[123]

JiaL, HuQ, ZhangT, et al. Engineering biomimetic biosensor using dual-targeting multivalent aptamer regulated 3D DNA walker enables high-performance detection of heterogeneous circulating tumor cells. Small. 2023; 19(38): e2302542.

[124]

LiuX, LiH, ZhaoY, Yu X, XuD. Multivalent aptasensor array and silver aggregated amplification for multiplex detection in microfluidic devices. Talanta. 2018; 188: 417-422.

[125]

DirksRM, PierceNA. Triggered amplification by hybridization chain reaction. Proc Natl Acad Sci U S A. 2004; 101(43): 15275-15278.

[126]

HassanU, BashirR. Research highlights: highlights from the latest articles in nanomedicine. Nanomedicine. 2013; 8(9): 1369-1371.

[127]

LyuY, ChenG, ShangguanD, et al. Generating cell targeting aptamers for nanotheranostics using cell-SELEX. Theranostics. 2016; 6(9): 1440-1452.

[128]

ZhuG, ZhengJ, SongE, et al. Self-assembled, aptamer-tethered DNA nanotrains for targeted transport of molecular drugs in cancer theranostics. Proc Natl Acad Sci U S A. 2013; 110(20): 7998-8003.

[129]

HuangZ, WangD, ZhangQ, Zhang Y, PengR, TanW. Leveraging aptamer-based DNA nanotechnology for bioanalysis and cancer therapeutics. Acc Mater Res. 2024; 5(4): 438-452.

[130]

ChenJH, SeemanNC. Synthesis from DNA of a molecule with the connectivity of a cube. Nature. 1991; 350(6319): 631-633.

[131]

GoodmanRP, BerryRM, TurberfieldAJ. The single-step synthesis of a DNA tetrahedron. Chem Commun (Camb). 2004;(12): 1372-1373.

[132]

YanJ, ZhanX, ZhangZ, et al. Tetrahedral DNA nanostructures for effective treatment of cancer: advances and prospects. J Nanobiotechnology. 2021; 19(1): 412.

[133]

FengQM, GuoYH, XuJJ, ChenHY. Self-assembled DNA tetrahedral scaffolds for the construction of electrochemiluminescence biosensor with programmable DNA cyclic amplification. ACS Appl Mater Interfaces. 2017; 9(20): 17637-17644.

[134]

ShenB, LiL, LiuC, et al. Mesoporous nanozyme-enhanced DNA tetrahedron electrochemiluminescent biosensor with three-dimensional walking nanomotor-mediated CRISPR/Cas12a for ultrasensitive detection of exosomal microRNA. Anal Chem. 2023; 95(9): 4486-4495.

[135]

ShaoX, LinS, PengQ, et al. Tetrahedral DNA nanostructure: a potential promoter for cartilage tissue regeneration via regulating chondrocyte phenotype and proliferation. Small. 2017; 13(12): 1602770.

[136]

PengQ, ShaoXR, XieJ, et al. Understanding the biomedical effects of the self-assembled tetrahedral DNA nanostructure on living cells. ACS Appl Mater Interfaces. 2016; 8(20): 12733-12739.

[137]

CuiW, YangX, DouY, et al. Effects of tetrahedral DNA nanostructures on the treatment of osteoporosis. Cell Prolif. 2024; 57(7): e13625.

[138]

WangD, LiS, ZhaoZ, Zhang X, TanW. Engineering a second-order DNA logic-gated nanorobot to sense and release on live cell membranes for multiplexed diagnosis and synergistic therapy. Angew Chem Int Ed Engl. 2021; 60(29): 15816-15820.

[139]

MeiQ, WeiX, SuF, et al. Stability of DNA origami nanoarrays in cell lysate. Nano Lett. 2011; 11(4): 1477-1482.

[140]

HeY, YeT, SuM, et al. Hierarchical self-assembly of DNA into symmetric supramolecular polyhedra. Nature. 2008; 452(7184): 198-201.

[141]

ChengXS, JinX. The braid index of complicated DNA polyhedral links. PLoS One. 2012; 7(11): e48968.

[142]

ShihWM, QuispeJD, JoyceGF. A 1.7-kilobase single-stranded DNA that folds into a nanoscale octahedron. Nature. 2004; 427(6975): 618-621.

[143]

HanX, JiangY, LiS, et al. Multivalent aptamer-modified tetrahedral DNA nanocage demonstrates high selectivity and safety for anti-tumor therapy. Nanoscale. 2019; 11(1): 339-347.

[144]

SundarayK, BaralB, SubudhiU. DNA polyhedrons cube, prism, and square pyramid protect the catalytic activity of catalase: a thermodynamics and kinetics study. Int J Biol Macromol. 2024; 264(pt 2):130557.

[145]

BanerjeeA, BhatiaD, SaminathanA, Chakraborty S, KarS, KrishnanY. Controlled release of encapsulated cargo from a DNA icosahedron using a chemical trigger. Angew Chem Int Ed Engl. 2013; 52(27): 6854-6857.

[146]

ZhangK, LiuJ, SongQ, et al. DNA nanosponge for adsorption and clearance of intracellular miR-21 and enhanced antitumor chemotherapy. ACS Appl Mater Interfaces. 2019; 11(50): 46604-46613.

[147]

SunW, GuZ. Therapeutics. DNA Nanoclews for Stimuli-Responsive Anticancer Drug Delivery. 2016: 141-150.

[148]

ZhuG, MeiL, VishwasraoHD, et al. Intertwining DNA-RNA nanocapsules loaded with tumor neoantigens as synergistic nanovaccines for cancer immunotherapy. Nat Commun. 2017; 8(1): 1482.

[149]

LuoD, LinX, ZhaoY, et al. A dynamic DNA nanosponge for triggered amplification of gene-photodynamic modulation. Chem Sci. 2022; 13(18): 5155-5163.

[150]

JiangQ, ZhaoS, LiuJ, SongL, WangZG, Ding B. Rationally designed DNA-based nanocarriers. Adv Drug Deliv Rev. 2019; 147: 2-21.

[151]

WangX-y, YanY, ZhiS, BiS. Dumbbell DNA-mediated rolling circle amplification for visual biosensing of intracellular glutathione. Sens Actuators B Chem. 2022; 373: 132745.

[152]

LvZ, ZhuY, LiF. DNA functional nanomaterials for controlled delivery of nucleic acid-based drugs. Front Bioeng Biotechnol. 2021; 9: 720291.

[153]

OrganizationWH. Global battle against cancer won’t be won with treatment alone-effective prevention measures urgently needed to prevent cancer crisis. Cent Eur J Public Health. 2014; 22(1): 23-28.

[154]

Brannon-PeppasL, Blanchette JO. Nanoparticle and targeted systems for cancer therapy. Adv Drug Deliv Rev. 2004; 56(11): 1649-1659.

[155]

MinottiG, MennaP, SalvatorelliE, CairoG, GianniL. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev. 2004; 56(2): 185-229.

[156]

NussbaumerS, Bonnabry P, VeutheyJL, Fleury-SouverainS. Analysis of anticancer drugs: a review. Talanta. 2011; 85(5): 2265-2289.

[157]

GhanghoriaR, Kesharwani P, TekadeRK, JainNK. Targeting luteinizing hormone-releasing hormone: a potential therapeutics to treat gynecological and other cancers. J Controlled Release. 2018; 269: 277-301.

[158]

SeidiK, Neubauer HA, MorigglR, Jahanban-EsfahlanR, Javaheri T. Tumor target amplification: Implications for nano drug delivery systems. J Controlled Release. 2018; 275: 142-161.

[159]

KhanMI, Hossain MI, HossainMK, et al. Recent progress in nanostructured smart drug delivery systems for cancer therapy: a review. ACS Appl Bio Mater. 2022; 5(3): 971-1012.

[160]

XinY, HuangQ, TangJQ, et al. Nanoscale drug delivery for targeted chemotherapy. Cancer Lett. 2016; 379(1): 24-31.

[161]

PandeyMK, Balwani S, SharmaPK, ParmarVS, GhoshB, WattersonAC. Design, synthesis and anti-inflammatory evaluation of PEGylated 4-methyl and 4, 8-dimethylcoumarins. Eur J Pharm Sci. 2010; 39(1-3): 134-40.

[162]

IwamotoT. Clinical application of drug delivery systems in cancer chemotherapy: review of the efficacy and side effects of approved drugs. Biol Pharm Bull. 2013; 36(5): 715-718.

[163]

ZhangG, ZengX, LiP. Nanomaterials in cancer-therapy drug delivery system. J Biomed Nanotechnol. 2013; 9(5): 741-750.

[164]

ChenW, ZhouS, GeL, WuW, JiangX. Translatable high drug loading drug delivery systems based on biocompatible polymer nanocarriers. Biomacromolecules. 2018; 19(6): 1732-1745.

[165]

SamadA, Sultana Y, AqilM. Liposomal drug delivery systems: an update review. Curr Drug Deliv. 2007; 4(4): 297-305.

[166]

LuY, ZhangE, YangJ, Cao Z. Strategies to improve micelle stability for drug delivery. Nano Res. 2018; 11(10): 4985-4998.

[167]

BiancoA, Kostarelos K, PratoM. Applications of carbon nanotubes in drug delivery. Curr Opin Chem Biol. 2005; 9(6): 674-679.

[168]

MishraD, Hubenak JR, MathurAB. Nanoparticle systems as tools to improve drug delivery and therapeutic efficacy. J Biomed Mater Res Part A. 2013; 101(12): 3646-3660.

[169]

KimJ, YungBC, KimWJ, Chen X. Combination of nitric oxide and drug delivery systems: tools for overcoming drug resistance in chemotherapy. J Controlled Release. 2017; 263: 223-230.

[170]

LongL-y, ZhangJ, YangZ, Guo Y, HuX, WangY. Transdermal delivery of peptide and protein drugs: strategies, advantages and disadvantages. J Drug Del Sci Tech. 2020; 60: 102007.

[171]

ChenZ, Kankala RK, YangZ, et al. Antibody-based drug delivery systems for cancer therapy: mechanisms, challenges, and prospects. Theranostics. 2022; 12(8): 3719-3746.

[172]

TorchilinVP, Lukyanov AN. Peptide and protein drug delivery to and into tumors: challenges and solutions. Drug Discov Today. 2003; 8(6): 259-266.

[173]

ChenK, LiuB, YuB, et al. Advances in the development of aptamer drug conjugates for targeted drug delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2017; 9(3): e1438.

[174]

ZhuG, ChenX. Aptamer-based targeted therapy. Adv Drug Deliv Rev. 2018; 134: 65-78.

[175]

XueC, ZhangS, YuX, HuS, LuY, WuZS. Periodically ordered, nuclease-resistant DNA nanowires decorated with cell-specific aptamers as selective theranostic agents. Angew Chem. 2020; 132(40): 17693-17700.

[176]

PeiW, LiuM, WuY, et al. High payload and targeted release of anthracyclines by aptamer-tethered DNA nanotrains—thermodynamic and release kinetic study. Euro J Pharma Sci. 2020; 148: 105319.

[177]

WangD, LiuM, WuY, et al. Self-assembled DNA nanotrains for targeted delivery of mithramycin dimers coordinated by different metal ions: effect of binding affinity on drug loading, release and cytotoxicity. J Mol Liq. 2021; 339: 116722.

[178]

ZhuN, ZhangB, YuQ. Genetic engineering-facilitated coassembly of synthetic bacterial cells and magnetic nanoparticles for efficient heavy metal removal. ACS Appl Mater Interfaces. 2020; 12(20): 22948-22957.

[179]

ChampanhacC, TengIT, CansizS, et al. Development of a panel of DNA aptamers with high affinity for pancreatic ductal adenocarcinoma. Sci Rep. 2015; 5(1): 16788.

[180]

WangY, ZhangY, LiPC, et al. Development of novel aptamer-based targeted chemotherapy for bladder cancer. Cancer Res. 2022; 82(6): 1128-1139.

[181]

ChangM, YangC-S, HuangD-M. Aptamer-conjugated DNA icosahedral nanoparticles as a carrier of doxorubicin for cancer therapy. ACS Nano. 2011; 5(8): 6156-6163.

[182]

MaW, YangY, LiuZ, et al. Self-assembled multivalent aptamer drug conjugates: enhanced targeting and cytotoxicity for HER2-positive gastric cancer. ACS Appl Mater Interfaces. 2023; 15(37): 43359-43373.

[183]

RahimiH, Abdollahzade A, RamezaniM, AlibolandiM, AbnousK, TaghdisiSM. Targeted delivery of doxorubicin to tumor cells using engineered circular bivalent aptamer. J Drug Del Sci Tech. 2022; 75: 103692.

[184]

WangM, ZengJ, WangJ, Wang X, WangY, GanN. Dual-mode aptasensor for simultaneous detection of multiple food-borne pathogenic bacteria based on colorimetry and microfluidic chip using stir bar sorptive extraction. Mikrochim Acta. 2021; 188(8): 244.

[185]

WangJ, WangH, WangH, et al. Nonviolent self-catabolic DNAzyme nanosponges for smart anticancer drug delivery. ACS Nano. 2019; 13(5): 5852-5863.

[186]

MalinaJ, Kostrhunova H, ScottP, BrabecV. Metallohelices stabilize DNA three-way junctions and induce DNA damage in cancer cells. Nucleic Acids Res. 2023; 51(14): 7174-7183.

[187]

KhoshbinZ, Sameiyan E, ZahraeeH, et al. A simple and robust aptasensor assembled on surfactant-mediated liquid crystal interface for ultrasensitive detection of mycotoxin. Anal Chim Acta. 2023; 1270: 341478.

[188]

TaghdisiSM, DaneshNM, RamezaniM, Yazdian-Robati R, AbnousK. A novel AS1411 aptamer-based three-way junction pocket DNA nanostructure loaded with doxorubicin for targeting cancer cells in vitro and in vivo. Mol Pharm. 2018; 15(5): 1972-1978.

[189]

WangY, WeiX, ZhangC, Zhang F, LiangW. Nanoparticle delivery strategies to target doxorubicin to tumor cells and reduce side effects. Ther Deliv. 2010; 1(2): 273-287.

[190]

PloskerGL, DFaulds. Epirubicin: a review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in cancer chemotherapy. Drugs. 1993; 45(5): 788-856.

[191]

MaY, YuS, NiS, et al. Targeting strategies for enhancing paclitaxel specificity in chemotherapy. Front Cell Dev Biol. 2021; 9: 626910.

[192]

PritchardJR, BrunoPM, GilbertLA, Capron KL, LauffenburgerDA, HemannMT. Defining principles of combination drug mechanisms of action. Proc Natl Acad Sci U S A. 2013; 110(2): E170-E179.

[193]

ZhouF, WangP, PengY, et al. Molecular engineering-based aptamer–drug conjugates with accurate tunability of drug ratios for drug combination targeted cancer therapy. Angew Chem Int Ed Engl. 2019; 58(34): 11661-11665.

[194]

FidlerIJ. Critical Determinants of Metastasis. Elsevier; 2002: 89-96.

[195]

InoueK, SlatonJW, KarashimaT, et al. The prognostic value of angiogenesis factor expression for predicting recurrence and metastasis of bladder cancer after neoadjuvant chemotherapy and radical cystectomy. Clin Cancer Res. 2000; 6(12): 4866-4873.

[196]

GreenJA, KirwanJM, TierneyJF, et al. Survival and recurrence after concomitant chemotherapy and radiotherapy for cancer of the uterine cervix: a systematic review and meta-analysis. Lancet. 2001; 358(9284): 781-786.

[197]

FischerR, Breidert M, KeckT, MakowiecF, Lohrmann C, HarderJ. Early recurrence of pancreatic cancer after resection and during adjuvant chemotherapy. Saudi J Gastroenterol. 2012; 18(2): 118-121.

[198]

Fung-Kee-FungM, OliverT, ElitL, Oza A, HirteHW, BrysonP. Optimal chemotherapy treatment for women with recurrent ovarian cancer. Curr Oncol. 2007; 14(5): 195-208.

[199]

CaoJ, ChenZ, ChiJ, SunY, SunY. Recent progress in synergistic chemotherapy and phototherapy by targeted drug delivery systems for cancer treatment. Artif Cells Nanomed Biotechnol. 2018; 46(sup1): 817-830.

[200]

WethFR, Hoggarth GB, WethAF, et al. Unlocking hidden potential: advancements, approaches, and obstacles in repurposing drugs for cancer therapy. Br J Cancer. 2024; 130(5): 703-715.

[201]

LeeSS, WeilCR, BoydLR, DeCesaris C, GaffneyD, SunejaG. Trends in use of radiation therapy, chemotherapy, and combination chemoradiotherapy in advanced uterine cancer before, during, and after GOG 258. Int J Gyneco Cancer. 2023; 33(9): 1408-1418.

[202]

FangX, XuJ, JinK, QianJ. Combining of immunotherapeutic approaches with chemotherapy for treatment of gastric cancer: achievements and limitations. Int Immunopharmacol. 2023; 118: 110062.

[203]

ChenY, YuR, LiuY. Combine radiotherapy and immunotherapy in esophageal squamous cell carcinoma. Crit Rev Oncol Hematol. 2023; 190: 104115.

[204]

LiuY, LinY, XiaoH, et al. mRNA-responsive two-in-one nanodrug for enhanced anti-tumor chemo-gene therapy. J Controlled Release. 2024; 369: 765-774.

[205]

WuY, LiuM, PeiW, et al. Thermodynamics, in vitro release and cytotoxity studies on doxorubicin–toluidine blue O combination drugs co-loaded in aptamer-tethered DNA nanostructures. J Mol Liq. 2020; 320: 114390.

[206]

XuZ, NiR, ChenY. Targeting breast cancer stem cells by a self-assembled, aptamer-conjugated DNA nanotrain with preloading doxorubicin. Int J Nanomed. 2019; 14: 6831-6842.

[207]

HuangQ, LiuX, ZhangP, Wu Z, ZhaoZ. A DNA nano-train carrying a predefined drug combination for cancer therapy. Chem Res Chin Univ. 2022; 38(4): 928-934.

[208]

GuoY, CaoX, ZhengX, Abbas SKJ, LiJ, TanW. Construction of nanocarriers based on nucleic acids and their applications in nanobiology delivery systems. Natl Sci Rev. 2022; 9(5): nwac006.

[209]

ZhangT, TianT, LinY. Functionalizing framework nucleic-acid-based nanostructures for biomedical application. Adv Mater. 2022; 34(46): e2107820.

[210]

YanJ, YuH, TangX, et al. Highly triple-effective synergy based on tetrahedral DNA nanostructure-induced tumor vaccines for cancer therapy. Mater Design. 2023; 226: 111584.

[211]

LiM, YangG, ZhengY, et al. NIR/pH-triggered aptamer-functionalized DNA origami nanovehicle for imaging-guided chemo-phototherapy. J Nanobiotechnol. 2023; 21(1): 186.

[212]

LiQ, WangF, ShiL, et al. Nanotrains of DNA copper nanoclusters that triggered a cascade Fenton-like reaction and glutathione depletion to doubly enhance chemodynamic therapy. ACS Appl Mater Interfaces. 2022; 14(33): 37280-37290.

[213]

AsrorovAM, GuZ, MinKA, Shin MC, HuangY. Advances on tumor-targeting delivery of cytotoxic proteins. ACS Pharmacol Transl Sci. 2020; 3(1): 107-118.

[214]

JiangY, PanX, ChangJ, et al. Supramolecularly engineered circular bivalent aptamer for enhanced functional protein delivery. J Am Chem Soc. 2018; 140(22): 6780-6784.

[215]

NedorezovaDD, Dubovichenko MV, BelyaevaEP, GrigorievaED, Peresadina AV, KolpashchikovDM. Specificity of oligonucleotide gene therapy (OGT) agents. Theranostics. 2022; 12(16): 7132-7157.

[216]

ZhengJ, YangR, ShiM, et al. Rationally designed molecular beacons for bioanalytical and biomedical applications. Chem Soc Rev. 2015; 44(10): 3036-3055.

[217]

RenYJ, ZhangY. An update on RNA interference-mediated gene silencing in cancer therapy. Expert Opin Biol Ther. 2014; 14(11): 1581-1592.

[218]

SandlerAD, Chihara H, KobayashiG, et al. CpG oligonucleotides enhance the tumor antigen-specific immune response of a granulocyte macrophage colony-stimulating factor-based vaccine strategy in neuroblastoma. Cancer Res. 2003; 63(2): 394-399.

[219]

ComaS, NoeV, LavarinoC, et al. Use of siRNAs and antisense oligonucleotides against survivin RNA to inhibit steps leading to tumor angiogenesis. Oligonucleotides. 2004; 14(2): 100-113.

[220]

GaoS, TianH, GuoY, et al. miRNA oligonucleotide and sponge for miRNA-21 inhibition mediated by PEI-PLL in breast cancer therapy. Acta Biomater. 2015; 25: 184-193.

[221]

YildizhanH, BarkanNP, Karahisar TuranS, et al. Treatment strategies in cancer from past to present. Drug Targeting and Stimuli Sensitive Drug Delivery Systems. Elsevier; 2018: 1-37.

[222]

WangG, ReedE, LiQQ. Molecular basis of cellular response to cisplatin chemotherapy in non-small cell lung cancer (review). Oncol Rep. 2004; 12(5): 955-965.

[223]

BeltingM, Sandgren S, WittrupA. Nuclear delivery of macromolecules: barriers and carriers. Adv Drug Deliv Rev. 2005; 57(4): 505-527.

[224]

DuvvuriM, KriseJP. Intracellular drug sequestration events associated with the emergence of multidrug resistance: a mechanistic review. Front Biosci. 2005; 10(2): 1499-1509.

[225]

EckfordPD, SharomFJ. ABC efflux pump-based resistance to chemotherapy drugs. Chem Rev. 2009; 109(7): 2989-3011.

[226]

ChenC, LuL, YanS, et al. Autophagy and doxorubicin resistance in cancer. Anticancer Drugs. 2018; 29(1): 1-9.

[227]

WangLJ, HanQ, QiuJG, Zhang CY. Cooperative in situ assembly of G-quadruplex DNAzyme nanowires for one-step sensing of CpG methylation in human genomes. Nano Lett. 2022; 22(1): 347-354.

[228]

SunH, ZhuX, LuPY, RosatoRR, TanW, ZuY. Oligonucleotide aptamers: new tools for targeted cancer therapy. Mol Ther Nucleic Acids. 2014; 3(8): e182.

[229]

LaoYH, PhuaKK, LeongKW. Aptamer nanomedicine for cancer therapeutics: barriers and potential for translation. ACS Nano. 2015; 9(3): 2235-2254.

[230]

SchneiderCS, XuQ, BoylanNJ, et al. Nanoparticles that do not adhere to mucus provide uniform and long-lasting drug delivery to airways following inhalation. Sci Adv. 2017; 3(4): e1601556.

[231]

FanQ, LiZ, YinJ, et al. Inhalable pH-responsive DNA tetrahedron nanoplatform for boosting anti-tumor immune responses against metastatic lung cancer. Biomaterials. 2023; 301: 122283.

[232]

ZhangZ, XuD, WangJ, et al. Rolling circle amplification-based DNA nano-assembly for targeted drug delivery and gene therapy. Biomacromolecules. 2023; 24(1): 439-448.

[233]

LimDY, HwangBH. Aptamer-modified tetrahedral DNA nanostructure-immobilized liposome for specific gene delivery and potential cancer theragnostic. Biotechnol J. 2024; 19(1): e2300156.

[234]

PangL, ShahH, WangH, Shu D, QianSY, SathishV. EpCAM-targeted 3WJ RNA nanoparticle harboring delta-5-desaturase siRNA inhibited lung tumor formation via DGLA peroxidation. Mol Ther Nucleic Acids. 2020; 22: 222-235.

[235]

ZhongW, HuangL, LinY, XingC, LuC. Endogenous dual miRNA-triggered dynamic assembly of DNA nanostructures for in-situ dual siRNA delivery. Sci China Mater. 2023; 66(7): 1-9.

[236]

SpillerDG, WoodCD, RandDA, White MR. Measurement of single-cell dynamics. Nature. 2010; 465(7299): 736-745.

[237]

ByrneJD, Betancourt T, Brannon-PeppasL. Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev. 2008; 60(15): 1615-1626.

[238]

FarzinL, Shamsipur M, MoassesiME, SheibaniS. Clinical aspects of radiolabeled aptamers in diagnostic nuclear medicine: a new class of targeted radiopharmaceuticals. Bioorg Med Chem. 2019; 27(12): 2282-2291.

[239]

ZengY, NixonRL, LiuW, WangR. The applications of functionalized DNA nanostructures in bioimaging and cancer therapy. Biomaterials. 2021; 268: 120560.

[240]

ChaoJ, LiuH, SuS, WangL, HuangW, Fan C. Structural DNA nanotechnology for intelligent drug delivery. Small. 2014; 10(22): 4626-4635.

[241]

ZhangH, ZhouL, ZhuZ, YangC. Recent progress in aptamer-based functional probes for bioanalysis and biomedicine. Chemistry. 2016; 22(29): 9886-900.

[242]

ZhangD, ZhengA, LiJ, et al. Tumor microenvironment activable self-assembled DNA hybrids for pH and redox dual-responsive chemotherapy/PDT treatment of hepatocellular carcinoma. Adv Sci (Weinh). 2017; 4(4): 1600460.

[243]

MaW, ChenB, JiaR, et al. In situ hand-in-hand DNA tile assembly: a pH-driven and aptamer-targeted DNA nanostructure for TK1 mRNA visualization and synergetic killing of cancer cells. Anal Chem. 2021; 93(30): 10511-10518.

[244]

YangY, ZhuW, ChengL, et al. Tumor microenvironment (TME)-activatable circular aptamer-PEG as an effective hierarchical-targeting molecular medicine for photodynamic therapy. Biomaterials. 2020; 246: 119971.

[245]

EvankoD. Hybridization chain reaction. Nat Methods. 2004; 1(3): 186-186.

[246]

KhuuPA, VothAR, HaysFA, Ho PS. The stacked-X DNA Holliday junction and protein recognition. J Mol Recognit. 2006; 19(3): 234-242.

[247]

WangP, MeyerTA, PanV, DuttaPK, KeY. The beauty and utility of DNA origami. Chem. 2017; 2(3): 359-382.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

190

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/