DNA Nanostructures for Cancer: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Sadanand Pandey.

The rapid development of multidrug co-delivery and nano-medicines has made spontaneous progress in tumor treatment and diagnosis. DNA is a unique biological molecule that can be tailored and molded into various nanostructures. The addition of ligands or stimuli-responsive elements enables DNA nanostructures to mediate highly targeted drug delivery to the cancer cells. Smart DNA nanostructures, owing to their various shapes, sizes, geometry, sequences, and characteristics, have various modes of cellular internalization and final disposition. On the other hand, functionalized DNA nanocarriers have specific receptor-mediated uptake, and most of these ligand anchored nanostructures able to escape lysosomal degradation.

  • nanotechnology
  • DNA nanostructures
  • stimuli-responsive
  • smart nanocarriers
Please wait, diff process is still running!

References

  1. Aflori, M. Smart Nanomaterials for Biomedical Applications—A Review. Nanomaterials 2021, 11, 396.
  2. Yan, H.; Zhang, X.; Shen, Z.; Seeman, N.C. A robust DNA mechanical device controlled by hybridization topology. Nat. Cell Biol. 2002, 415, 62–65.
  3. Kallenbach, N.R.; Ma, R.-I.; Seeman, N.C. An immobile nucleic acid junction constructed from oligonucleotides. Nat. Cell Biol. 1983, 305, 829–831.
  4. Chao, J.; Liu, H.; Su, S.; Wang, L.; Huang, W.; Fan, C. Structural DNA Nanotechnology for Intelligent Drug Delivery. Small 2014, 10, 4626–4635.
  5. Kim, T.; Nam, K.; Kim, Y.M.; Yang, K.; Roh, Y.H. DNA-Assisted Smart Nanocarriers: Progress, Challenges, and Opportunities. ACS Nano 2021, 15, 1942–1951.
  6. Stuart, M.A.C.; Huck, W.T.S.; Genzer, J.; Müller, M.; Ober, C.; Stamm, M.; Sukhorukov, G.B.; Szleifer, I.; Tsukruk, V.V.; Urban, M.; et al. Emerging applications of stimuli-responsive polymer materials. Nat. Mater. 2010, 9, 101–113.
  7. Li, L.; Yang, Z.; Chen, X. Recent Advances in Stimuli-Responsive Platforms for Cancer Immunotherapy. Accounts Chem. Res. 2020, 53, 2044–2054.
  8. Ruiz-Hernández, E.; Baeza, A.; Vallet-Regí, M. Smart Drug Delivery through DNA/Magnetic Nanoparticle Gates. ACS Nano 2011, 5, 1259–1266.
  9. Chen, Y.-X.; Huang, K.-J.; He, L.-L.; Wang, Y.-H. Tetrahedral DNA probe coupling with hybridization chain reaction for competitive thrombin aptasensor. Biosens. Bioelectron. 2018, 100, 274–281.
  10. Li, H.; Fan, J.; Buhl, E.M.; Huo, S.; Loznik, M.; Göstl, R.; Herrmann, A. DNA hybridization as a general method to enhance the cellular uptake of nanostructures. Nanoscale 2020, 12, 21299–21305.
  11. Juul, S.; Iacovelli, F.; Falconi, M.; Kragh, S.L.; Christensen, B.; Frøhlich, R.; Franch, O.; Kristoffersen, E.L.; Stougaard, M.; Leong, K.W.; et al. Temperature-Controlled Encapsulation and Release of an Active Enzyme in the Cavity of a Self-Assembled DNA Nanocage. ACS Nano 2013, 7, 9724–9734.
  12. Ouyang, X.; Chang, Y.-N.; Yang, K.-W.; Wang, W.-M.; Bai, J.-J.; Wang, J.-W.; Zhang, Y.-J.; Wang, S.-Y.; Xie, B.-B.; Wang, L.-L. A DNA nanoribbon as a potent inhibitor of metallo-β-lactamases. Chem. Commun. 2017, 53, 8878–8881.
  13. Ouyang, X.; Wang, M.-F.; Guo, L.-J.; Cui, C.-J.; Liu, T.; Ren, Y.-A.; Zhao, Y.; Ge, Z.-L.; Guo, X.-Q.; Xie, G.; et al. DNA Nanoribbon-Templated Self-Assembly of Ultrasmall Fluorescent Copper Nanoclusters with Enhanced Luminescence. Angew. Chem. 2020, 132, 11934–11942.
  14. Roh, Y.H.; Lee, J.B.; Kiatwuthinon, P.; Hartman, M.R.; Cha, J.J.; Um, S.H.; Muller, D.; Luo, D. DNAsomes: Multifunctional DNA-Based Nanocarriers. Small 2010, 7, 74–78.
  15. Liang, H.F.; Hong, M.H.; Ho, R.M.; Chung, C.K.; Lin, Y.H.; Chen, C.H.; Sung, H.W. Novel Method Using a Temperature-Sensitive Polymer (Methylcellulose) to Thermally Gel Aqueous Alginate as a pH-Sensitive Hydrogel. Biomacromolecules 2004, 5, 1917–1925.
  16. Chu, T.C. Aptamer mediated siRNA delivery. Nucleic Acids Res. 2006, 34, e73.
  17. Li, W.; Yang, X.; He, L.; Wang, K.; Wang, Q.; Huang, J.; Liu, J.; Wu, B.; Xu, C. Self-assembled DNA nanocentipede as multivalent drug carrier for targeted delivery. ACS Appl. Mater. Interfaces 2016, 8, 25733–25740.
  18. Wang, T.; Chen, C.; Larcher, L.; Barrero, R.; Veedu, R.N. Three decades of nucleic acid aptamer technologies: Lessons learned, progress and opportunities on aptamer development. Biotechnol. Adv. 2019, 37, 28–50.
  19. Yuan, Y.; Gu, Z.; Yao, C.; Luo, D.; Yang, D. Nucleic Acid–Based Functional Nanomaterials as Advanced Cancer Therapeutics. Small 2019, 15, e1900172.
  20. Yang, K.; Chang, Y.; Wen, J.; Lu, Y.; Pei, Y.; Cao, S.; Wang, F.; Pei, Z. Supramolecular Vesicles Based on Complex of Trp-Modified Pillararene and Galactose Derivative for Synergistic and Targeted Drug Delivery. Chem. Mater. 2016, 28, 1990–1993.
  21. Li, F.; Tang, J.; Geng, J.; Luo, D.; Yang, D. Polymeric DNA hydrogel: Design, synthesis and applications. Prog. Polym. Sci. 2019, 98, 101163.
  22. Shi, L.; Mu, C.; Gao, T.; Chen, T.; Hei, S.; Yang, J.; Li, G. DNA nanoflower blooms in nanochannels: A new strategy for miRNA detection. Chem. Commun. 2018, 54, 11391–11394.
  23. Hu, R.; Zhang, X.; Zhao, Z.; Zhu, G.; Chen, T.; Fu, T.; Tan, W. DNA Nanoflowers for Multiplexed Cellular Imaging and Traceable Targeted Drug Delivery. Angew. Chem. 2014, 126, 5931–5936.
  24. Cao, M.; Sun, Y.; Xiao, M.; Li, L.; Liu, X.; Jin, H.; Pei, H. Multivalent Aptamer-modified DNA Origami as Drug Delivery System for Targeted Cancer Therapy. Chem. Res. Chin. Univ. 2019, 36, 1–7.
  25. Hu, Q.; Wang, S.; Wang, L.; Gu, H.; Fan, C. DNA Nanostructure-Based Systems for Intelligent Delivery of Therapeutic Oligonucleotides. Adv. Healthc. Mater. 2018, 7, 1701153.
  26. Pan, Q.; Nie, C.; Hu, Y.; Yi, J.; Liu, C.; Zhang, J.; He, M.; He, M.; Chen, T.-T.; Chu, X. Aptamer-Functionalized DNA Origami for Targeted Codelivery of Antisense Oligonucleotides and Doxorubicin to Enhance Therapy in Drug-Resistant Cancer Cells. ACS Appl. Mater. Interfaces 2019, 12, 400–409.
  27. Tapeinos, C.; Battaglini, M.; Ciofani, G. Advances in the design of solid lipid nanoparticles and nanostructured lipid carriers for targeting brain diseases. J. Control. Release 2017, 264, 306–332.
  28. Wu, D.; Wang, L.; Li, W.; Xu, X.; Jiang, W. DNA nanostructure-based drug delivery nanosystems in cancer therapy. Int. J. Pharm. 2017, 533, 169–178.
  29. Shcharbin, D.; Halets-Bui, I.; Abashkin, V.; Dzmitruk, V.; Loznikova, S.; Odabaşı, M.; Acet, Ö.; Önal, B.; Özdemir, N.; Shcharbina, N.; et al. Hybrid metal-organic nanoflowers and their application in biotechnology and medicine. Colloids Surf. B Biointerfaces 2019, 182, 110354.
  30. Rai, A.; Ferreira, L. Biomedical applications of the peptide decorated gold nanoparticles. Crit. Rev. Biotechnol. 2021, 41, 186–215.
  31. Bamrungsap, S. DNA-Conjugated Magnetic Nanoparticles for Bio-Analytical and Biomedical Applications; University of Florida: Gainesville, FL, USA, 2011.
  32. Zhang, F.; Jiang, S.; Wu, S.; Li, Y.; Mao, C.; Liu, Y.; Yan, H. Complex wireframe DNA origami nanostructures with multi-arm junction vertices. Nat. Nanotechnol. 2015, 10, 779–784.
  33. Hong, F.; Zhang, F.; Liu, Y.; Yan, H. DNA Origami: Scaffolds for Creating Higher Order Structures. Chem. Rev. 2017, 117, 12584–12640.
  34. Meng, H.-M.; Fu, T.; Zhang, X.-B.; Tan, W. Cell-SELEX-based aptamer-conjugated nanomaterials for cancer diagnosis and therapy. Natl. Sci. Rev. 2015, 2, 71–84.
  35. Seaberg, J.; Montazerian, H.; Hossen, N.; Bhattacharya, R.; Khademhosseini, A.; Mukherjee, P. Hybrid Nanosystems for Biomedical Applications. ACS Nano 2021, 15, 2099–2142.
  36. Wilner, O.I.; Willner, I. Functionalized DNA Nanostructures. Chem. Rev. 2012, 112, 2528–2556.
  37. Iinuma, R.; Ke, Y.; Jungmann, R.; Schlichthaerle, T.; Woehrstein, J.B.; Yin, P. Polyhedra Self-Assembled from DNA Tripods and Characterized with 3D DNA-PAINT. Science 2014, 344, 65–69.
  38. Hu, Q.; Li, H.; Wang, L.; Gu, H.; Fan, C. DNA Nanotechnology-Enabled Drug Delivery Systems. Chem. Rev. 2018, 119, 6459–6506.
  39. Meng, H.-M.; Liu, H.; Kuai, H.; Peng, R.; Mo, L.; Zhang, X.-B. Aptamer-integrated DNA nanostructures for biosensing, bioimaging and cancer therapy. Chem. Soc. Rev. 2016, 45, 2583–2602.
  40. Calvo, P.; Gouritin, B.; Brigger, I.; Lasmezas, C.; Deslys, J.-P.; Williams, A.; Andreux, J.P.; Dormont, D.; Couvreur, P. PEGylated polycyanoacrylate nanoparticles as vector for drug delivery in prion diseases. J. Neurosci. Methods 2001, 111, 151–155.
  41. Baig, M.M.F.A.; Zou, T.; Neelakantan, P.; Zhang, C. Development and functionalization of DNA nanostructures for biomedical applications. J. Chin. Chem. Soc. 2021, 68, 228–238.
  42. Zhang, C.; Yang, C.; Whitham, S.A.; Hill, J.H. Development and Use of an Efficient DNA-Based Viral Gene Silencing Vector for Soybean. Mol. Plant Microbe Interact. 2009, 22, 123–131.
  43. Raza, A.; Rasheed, T.; Nabeel, F.; Hayat, U.; Bilal, M.; Iqbal, H.M.N. Endogenous and Exogenous Stimuli-Responsive Drug Delivery Systems for Programmed Site-Specific Release. Molecules 2019, 24, 1117.
  44. Wang, P.; Yin, T.; Li, J.; Zheng, B.; Wang, X.; Wang, Y.; Zheng, J.; Zheng, R.; Shuai, X. Ultrasound-responsive microbubbles for sonography-guided siRNA delivery. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 1139–1149.
  45. Papa, A.-L.; Korin, N.; Kanapathipillai, M.; Mammoto, A.; Mammoto, T.; Jiang, A.; Mannix, R.; Uzun, O.; Johnson, C.; Bhatta, D.; et al. Ultrasound-sensitive nanoparticle aggregates for targeted drug delivery. Biomaterials 2017, 139, 187–194.
  46. Muñoz de Escalona, M.; Sáez-Fernández, E.; Prados, J.C.; Melguizo, C.; Arias, J.L. Magnetic solid lipid nanoparticles in hyperthermia against colon cancer. Int. J. Pharm. 2016, 504, 11–19.
  47. Wu, P.; Gao, W.; Su, M.; Nice, E.C.; Zhang, W.; Lin, J.; Xie, N. Adaptive mechanisms of tumor therapy resistance driven by tumor microenvironment. Front. Cell Dev. Biol. 2021, 9, 357–362.
  48. Schafer, F.Q.; Buettner, G.R. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic. Biol. Med. 2001, 30, 1191–1212.
  49. Ganta, S.; Devalapally, H.; Shahiwala, A.; Amiji, M. A review of stimuli-responsive nanocarriers for drug and gene delivery. J. Control. Release 2008, 126, 187–204.
  50. An, H.; Xu, K.; Chang, L.; Wang, Y.; Qin, J.; Li, W. Thermo-responsive self-healable hydrogels with extremely mild base degradability and bio-compatibility. Polymer 2018, 147, 38–47.
  51. Luckanagul, J.A.; Pitakchatwong, C.; Na Bhuket, P.R.; Muangnoi, C.; Rojsitthisak, P.; Chirachanchai, S.; Wang, Q.; Rojsitthisak, P. Chitosan-based polymer hybrids for thermo-responsive nanogel delivery of curcumin. Carbohydr. Polym. 2018, 181, 1119–1127.
  52. Yildirim, T.; Yildirim, I.; Yañez-Macias, R.; Stumpf, S.; Fritzsche, C.; Hoeppener, S.; Guerrero-Sanchez, C.; Schubert, S.; Schubert, U.S. Dual pH and ultrasound responsive nanoparticles with pH triggered surface charge-conversional properties. Polym. Chem. 2017, 8, 1328–1340.
  53. Di Ianni, T.; Bose, R.J.; Sukumar, U.K.; Bachawal, S.; Wang, H.; Telichko, A.; Herickhoff, C.; Robinson, E.; Baker, S.; Vilches-Moure, J.; et al. Ultrasound/microbubble-mediated targeted delivery of anticancer microRNA-loaded nanoparticles to deep tissues in pigs. J. Control. Release 2019, 309, 1–10.
  54. Karimi, M.; Ghasemi, A.; Zangabad, P.S.; Rahighi, R.; Basri, S.M.M.; Mirshekari, H.; Amiri, M.; Pishabad, Z.S.; Aslani, A.; Bozorgomid, M.; et al. Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chem. Soc. Rev. 2016, 45, 1457–1501.
  55. Pitt, W.G.; Husseini, G.A.; Roeder, B.L.; Dickinson, D.J.; Warden, D.R.; Hartley, J.M.; Jones, P.W. Preliminary Results of Combining Low Frequency Low Intensity Ultrasound and Liposomal Drug Delivery to Treat Tumors in Rats. J. Nanosci. Nanotechnol. 2011, 11, 1866–1870.
  56. Gaspar, V.M.; Moreira, A.F.; de Melo-Diogo, D.; Costa, E.C.; Queiroz, J.A.; Sousa, F.; Pichon, C.; Correia, I.J. Chapter 6—Multifunctional nanocarriers for codelivery of nucleic acids and chemotherapeutics to cancer cells. In Nanobiomaterials in Medical Imaging; Grumezescu, A.M., Ed.; William Andrew Publishing: Norwich, NY, USA, 2016; pp. 163–207.
  57. Eisenbrey, J.; Burstein, O.M.; Kambhampati, R.; Forsberg, F.; Liu, J.-B.; Wheatley, M. Development and optimization of a doxorubicin loaded poly(lactic acid) contrast agent for ultrasound directed drug delivery. J. Control. Release 2010, 143, 38–44.
  58. Bhattacharya, S.; Eckert, F.; Boyko, V.; Pich, A. Temperature-, pH-, and Magnetic-Field-Sensitive Hybrid Microgels. Small 2007, 3, 650–657.
  59. Yu, S.; Wu, G.; Gu, X.; Wang, J.; Wang, Y.; Gao, H.; Ma, J. Magnetic and pH-sensitive nanoparticles for antitumor drug delivery. Colloids Surf. B Biointerfaces 2013, 103, 15–22.
  60. Dobson, J. Magnetic micro- and nano-particle-based targeting for drug and gene delivery. Nanomedicine 2006, 1, 31–37.
  61. Schenck, J.F. Physical interactions of static magnetic fields with living tissues. Prog. Biophys. Mol. Biol. 2005, 87, 185–204.
  62. Xie, W.; Gao, Q.; Wang, D.; Guo, Z.; Gao, F.; Wang, X.; Cai, Q.; Feng, S.-S.; Fan, H.; Sun, X.; et al. Doxorubicin-loaded Fe3O4@MoS2-PEG-2DG nanocubes as a theranostic platform for magnetic resonance imaging-guided chemo-photothermal therapy of breast cancer. Nano Res. 2018, 11, 2470–2487.
  63. Bordat, A.; Boissenot, T.; Nicolas, J.; Tsapis, N. Thermoresponsive polymer nanocarriers for biomedical applications. Adv. Drug Deliv. Rev. 2019, 138, 167–192.
  64. Pippa, N.; Meristoudi, A.; Pispas, S.; Demetzos, C. Temperature-dependent drug release from DPPC:C12H25-PNIPAM-COOH liposomes: Control of the drug loading/release by modulation of the nanocarriers’ components. Int. J. Pharm. 2015, 485, 374–382.
  65. Liu, D.; Yang, F.; Xiong, F.; Gu, N. The Smart Drug Delivery System and Its Clinical Potential. Theranostics 2016, 6, 1306–1323.
  66. Huda, S.; Alam, A.; Sharma, P.K. Smart nanocarriers-based drug delivery for cancer therapy: An innovative and developing strategy. J. Drug Deliv. Sci. Technol. 2020, 60, 102018.
  67. Schwerdt, A.; Zintchenko, A.; Concia, M.; Roesen, N.; Fisher, K.; Lindner, L.H.; Issels, R.; Wagner, E.; Ogris, M. Hyperthermia-Induced Targeting of Thermosensitive Gene Carriers to Tumors. Hum. Gene Ther. 2008, 19, 1283–1292.
  68. Hooshmand, S.; Hayat, S.M.; Ghorbani, A.; Khatami, M.; Pakravanan, K.; Darroudi, M. Preparation and Applications of Superparamagnetic Iron Oxide Nanoparticles in Novel Drug Delivery Systems: An Overview Article. Curr. Med. Chem. 2020, 1, 1–12.
  69. Chen, K.-J.; Liang, H.-F.; Chen, H.-L.; Wang, Y.; Cheng, P.-Y.; Liu, H.-L.; Xia, Y.; Sung, H.-W. A Thermoresponsive Bubble-Generating Liposomal System for Triggering Localized Extracellular Drug Delivery. ACS Nano 2013, 7, 438–446.
  70. Karimi, M.; Zangabad, P.S.; Ghasemi, A.; Amiri, M.; Bahrami, M.; Malekzad, H.; Asl, H.G.; Mahdieh, Z.; Bozorgomid, M.; Ghasemi, A.; et al. Temperature-Responsive Smart Nanocarriers for Delivery of Therapeutic Agents: Applications and Recent Advances. ACS Appl. Mater. Interfaces 2016, 8, 21107–21133.
  71. Alsehli, M. Polymeric nanocarriers as stimuli-responsive systems for targeted tumor (cancer) therapy: Recent advances in drug delivery. Saudi Pharm. J. 2020, 28, 255–265.
  72. Ward, M.A.; Georgiou, T.K. Thermoresponsive Polymers for Biomedical Applications. Polymers 2011, 3, 1215–1242.
  73. Bergueiro, J.; Calderón, M. Thermoresponsive Nanodevices in Biomedical Applications. Macromol. Biosci. 2014, 15, 183–199.
  74. Le, P.N.; Huynh, K.; Tran, N.Q. Advances in thermosensitive polymer-grafted platforms for biomedical applications. Mater. Sci. Eng. C 2018, 92, 1016–1030.
  75. Chen, Q.; Li, C.; Yang, X.; Huang, J.; Liu, S.; Liu, W. Self-assembled DNA nanowires as quantitative dual-drug nanocarriers for antitumor chemophotodynamic combination therapy. J. Mater. Chem. B 2017, 5, 7529–7537.
  76. Fu, G.; Soboyejo, W. Swelling and diffusion characteristics of modified poly (N-isopropylacrylamide) hydrogels. Mater. Sci. Eng. C 2010, 30, 8–13.
  77. Hajebi, S.; Rabiee, N.; Bagherzadeh, M.; Ahmadi, S.; Rabiee, M.; Roghani-Mamaqani, H.; Tahriri, M.; Tayebi, L.; Hamblin, M.R. Stimulus-responsive polymeric nanogels as smart drug delivery systems. Acta Biomater. 2019, 92, 1–8.
  78. Hu, Y.; Darcos, V.; Monge, S.; Li, S. Thermo-responsive drug release from self-assembled micelles of brush-like PLA/PEG analogues block copolymers. Int. J. Pharm. 2015, 491, 152–161.
  79. Daniel-Da-Silva, A.L.; Ferreira, L.; Gil, A.; Trindade, T. Synthesis and swelling behavior of temperature responsive κ-carrageenan nanogels. J. Colloid Interface Sci. 2011, 355, 512–517.
  80. Wang, D.; Huang, H.; Zhou, M.; Lu, H.; Chen, J.; Chang, Y.-T.; Gao, J.; Chai, Z.; Hu, Y. A thermoresponsive nanocarrier for mitochondria-targeted drug delivery. Chem. Commun. 2019, 55, 4051–4054.
  81. Ghamkhari, A.; Sarvari, R.; Ghorbani, M.; Hamishehkar, H. Novel thermoresponsive star-liked nanomicelles for targeting of anticancer agent. Eur. Polym. J. 2018, 107, 143–154.
  82. Muhammad, K.; Zhao, J.; Gao, B.; Feng, Y. Polymeric nano-carriers for on-demand delivery of genes via specific responses to stimuli. J. Mater. Chem. B 2020, 8, 9621–9641.
  83. Yan, L.; Li, X. Biodegradable Stimuli-Responsive Polymeric Micelles for Treatment of Malignancy. Curr. Pharm. Biotechnol. 2016, 17, 227–236.
  84. Fomina, N.; Sankaranarayanan, J.; Almutairi, A. Photochemical mechanisms of light-triggered release from nanocarriers. Adv. Drug Deliv. Rev. 2012, 64, 1005–1020.
  85. Fleige, E.; Quadir, M.A.; Haag, R. Stimuli-responsive polymeric nanocarriers for the controlled transport of active compounds: Concepts and applications. Adv. Drug Deliv. Rev. 2012, 64, 866–884.
  86. Xiong, Q.; Lim, Y.; Li, D.; Pu, K.; Liang, L.; Duan, H. Photoactive Nanocarriers for Controlled Delivery. Adv. Funct. Mater. 2020, 30, 1903896.
  87. Ebrahimi, G.; Asadikaram, G.; Akbari, H.; Nematollahi, M.H.; Abolhassani, M.; Shahabinejad, G.; Khodadadnejad, L.; Hashemi, M. Elevated levels of DNA methylation at the OPRM1 promoter region in men with opioid use disorder. Am. J. Drug Alcohol Abus. 2018, 44, 193–199.
  88. Sun, S.; Liang, S.; Xu, W.-C.; Xu, G.; Wu, S. Photoresponsive polymers with multi-azobenzene groups. Polym. Chem. 2019, 10, 4389–4401.
  89. Klajn, R. Spiropyran-based dynamic materials. Chem. Soc. Rev. 2014, 43, 148–184.
  90. Paramonov, S.V.; Lokshin, V.; Fedorova, O.A. Spiropyran, chromene or spirooxazine ligands: Insights into mutual relations between complexing and photochromic properties. J. Photochem. Photobiol. C Photochem. Rev. 2011, 12, 209–236.
  91. Bertrand, O.; Gohy, J.-F. Photo-responsive polymers: Synthesis and applications. Polym. Chem. 2016, 8, 52–73.
  92. Molla, M.R.; Rangadurai, P.; Antony, L.; Swaminathan, S.; De Pablo, J.J.; Thayumanavan, S. Dynamic actuation of glassy polymersomes through isomerization of a single azobenzene unit at the block copolymer interface. Nat. Chem. 2018, 10, 659–666.
  93. Khatami, M.; Heli, H.; Jahani, P.M.; Azizi, H.; Nobre, M.A.L. Copper/copper oxide nanoparticles synthesis using Stachys lavandulifolia and its antibacterial activity. IET Nanobiotechnol. 2017, 11, 709–713.
  94. Bartelds, R.; Nematollahi, M.H.; Pols, T.; Stuart, M.C.A.; Pardakhty, A.; Asadikaram, G.; Poolman, B. Niosomes, an alternative for liposomal delivery. PLoS ONE 2018, 13, e0194179.
  95. Wang, B.; Chen, K.; Yang, R.; Yang, F.; Liu, J. Stimulus-responsive polymeric micelles for the light-triggered release of drugs. Carbohydr. Polym. 2014, 103, 510–519.
  96. Jia, S.; Fong, W.-K.; Graham, B.; Boyd, B.J. Photoswitchable Molecules in Long-Wavelength Light-Responsive Drug Delivery: From Molecular Design to Applications. Chem. Mater. 2018, 30, 2873–2887.
  97. Silva, J.M.; Silva, E.; Reis, R.L. Light-triggered release of photocaged therapeutics—Where are we now? J. Control. Release 2019, 298, 154–176.
  98. Olejniczak, J.; Carling, C.-J.; Almutairi, A. Photocontrolled release using one-photon absorption of visible or NIR light. J. Control. Release 2015, 219, 18–30.
  99. Khatami, M.; Sharifi, I.; Nobre, M.A.L.; Zafarnia, N.; Aflatoonian, M.R. Waste-grass-mediated green synthesis of silver nanoparticles and evaluation of their anticancer, antifungal and antibacterial activity. Green Chem. Lett. Rev. 2018, 11, 125–134.
  100. Zhao, W.; Zhao, Y.; Wang, Q.; Liu, T.; Sun, J.; Zhang, R. Remote Light-Responsive Nanocarriers for Controlled Drug Delivery: Advances and Perspectives. Small 2019, 15, e1903060.
  101. Raza, A.; Hayat, U.; Rasheed, T.; Bilal, M.; Iqbal, H.M. “Smart” materials-based near-infrared light-responsive drug delivery systems for cancer treatment: A review. J. Mater. Res. Technol. 2019, 8, 1497–1509.
  102. Mi, P. Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and theranostics. Theranostics 2020, 10, 4557–4588.
  103. Tong, R.; Hemmati, H.D.; Langer, R.; Kohane, D.S. Photoswitchable Nanoparticles for Triggered Tissue Penetration and Drug Delivery. J. Am. Chem. Soc. 2012, 134, 8848–8855.
  104. Yan, B.; Boyer, J.-C.; Branda, N.R.; Zhao, Y. Near-Infrared Light-Triggered Dissociation of Block Copolymer Micelles Using Upconverting Nanoparticles. J. Am. Chem. Soc. 2011, 133, 19714–19717.
  105. Luo, D.; Carter, K.A.; Razi, A.; Geng, J.; Shao, S.; Giraldo, D.; Sunar, U.; Ortega, J.; Lovell, J.F. Doxorubicin encapsulated in stealth liposomes conferred with light-triggered drug release. Biomaterials 2016, 75, 193–202.
  106. Croissant, J.; Chaix, A.; Mongin, O.; Wang, M.; Clément, S.; Raehm, L.; Durand, J.-O.; Hugues, V.; Blanchard-Desce, M.; Maynadier, M.; et al. Two-Photon-Triggered Drug Delivery via Fluorescent Nanovalves. Small 2014, 10, 1752–1755.
  107. Cui, Y.-X.; Sun, Y.-X.; Li, Y.H.; Tang, A.N.; Zhu, L.N.; Kong, D.M. DNA-Based pH-Responsive Core–Shell Drug Nanocarrier for Tumor-Targeted Chemo-Photodynamic Therapy. Adv. Mater. Interfaces 2020, 7, 2000292.
  108. Yu, D.; Li, W.; Zhang, Y.; Zhang, B. Anti-tumor efficiency of paclitaxel and DNA when co-delivered by pH responsive ligand modified nanocarriers for breast cancer treatment. Biomed. Pharmacother. 2016, 83, 1428–1435.
  109. Tian, Q.; Wang, Y.; Deng, R.; Lin, L.; Liu, Y.; Li, J. Carbon nanotube enhanced label-free detection of microRNAs based on hairpin probe triggered solid-phase rolling-circle amplification. Nanoscale 2015, 7, 987–993.
  110. Wang, M.; Hu, H.; Sun, Y.; Qiu, L.; Zhang, J.; Guan, G. A pH-sensitive gene delivery system based on folic acid-PEG-chitosan—PAMAM-plasmid DNA complexes for cancer cell targeting. Biomaterials 2013, 34, 10120–10132.
  111. Boyacioglu, O.; Stuart, C.H.; Kulik, G.; Gmeiner, W.H. Dimeric DNA Aptamer Complexes for High-capacity–targeted Drug Delivery Using pH-sensitive Covalent Linkages. Mol. Ther. Nucleic Acids 2013, 2, e107.
  112. Sethuraman, V.A.; Na, K.; Bae, Y.H. pH-Responsive Sulfonamide/PEI System for Tumor Specific Gene Delivery: An in Vitro Study. Biomacromolecules 2006, 7, 64–70.
  113. Andersen, E.S.; Dong, M.; Nielsen, M.; Jahn, K.; Subramani, R.; Mamdouh, W.; Golas, M.M.; Sander, B.; Stark, H.; Oliveira, C.; et al. Self-assembly of a nanoscale DNA box with a controllable lid. Nat. Cell Biol. 2009, 459, 73–76.
  114. Li, Y.; Chen, Y.; Pan, W.; Yu, Z.; Yang, L.; Wang, H.; Li, N.; Tang, B. Nanocarriers with multi-locked DNA valves targeting intracellular tumor-related mRNAs for controlled drug release. Nanoscale 2017, 9, 17318–17324.
  115. Shi, J.; Yang, X.; Li, Y.; Wang, D.; Liu, W.; Zhang, Z.; Liu, J.; Zhang, K. MicroRNA-responsive release of Cas9/sgRNA from DNA nanoflower for cytosolic protein delivery and enhanced genome editing. Biomaterials 2020, 256, 120221.
  116. Ye, W.; Chen, X.; Li, X.; Liu, Y.; Jia, F.; Jin, Q.; Ji, J. Structure-Switchable DNA Programmed Disassembly of Nanoparticles for Smart Size Tunability and Cancer-Specific Drug Release. ACS Appl. Mater. Interfaces 2020, 12, 22560–22571.
  117. Zhao, N.; Deng, L.; Luo, D.; Zhang, P. One-step fabrication of biomass-derived hierarchically porous carbon/MnO nanosheets composites for symmetric hybrid supercapacitor. Appl. Surf. Sci. 2020, 526, 146696–146703.
  118. Kuzuya, A.; Ohya, Y. Nanomechanical Molecular Devices made of DNA Origami. Accounts Chem. Res. 2014, 47, 1742–1749.
  119. Zangabad, P.S.; Karimi, M.; Mehdizadeh, F.; Malekzad, H.; Ghasemi, A.; Bahrami, S.; Zare, H.; Moghoofei, M.; Hekmatmanesh, A.; Hamblin, M.R. Nanocaged platforms: Modification, drug delivery and nanotoxicity. Opening synthetic cages to release the tiger. Nanoscale 2017, 9, 1356–1392.
  120. Douglas, S.; Bachelet, I.; Church, G.M. A Logic-Gated Nanorobot for Targeted Transport of Molecular Payloads. Science 2012, 335, 831–834.
  121. Chong, S.C.; Blake, R. Exogenous attention and endogenous attention influence initial dominance in binocular rivalry. Vis. Res. 2006, 46, 1794–1803.
  122. Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mater. 2013, 12, 991–1003.
  123. Zhao, H.; Liu, X.; Yu, L.; Lin, S.; Zhang, C.; Xu, H.; Leng, Z.; Huang, W.; Lei, J.; Li, T.; et al. Comprehensive landscape of epigenetic-dysregulated lncRNAs reveals a profound role of enhancers in carcinogenesis in BC subtypes. Mol. Ther. Nucleic Acids 2021, 23, 667–681.
  124. Pierce, A.P.; De Waal, E.; McManus, L.M.; Shireman, P.; Chaudhuri, A.R. Oxidation and structural perturbation of redox-sensitive enzymes in injured skeletal muscle. Free Radic. Biol. Med. 2007, 43, 1584–1593.
  125. Chen, W.-H.; Liao, W.-C.; Sohn, Y.S.; Fadeev, M.; Cecconello, A.; Nechushtai, R.; Willner, I. Stimuli-Responsive Nucleic Acid-Based Polyacrylamide Hydrogel-Coated Metal-Organic Framework Nanoparticles for Controlled Drug Release. Adv. Funct. Mater. 2017, 28, 1705137.
  126. Wu, X.; Gao, Y.; Dong, C.-M. Polymer/gold hybrid nanoparticles: From synthesis to cancer theranostic applications. RSC Adv. 2015, 5, 13787–13796.
More