Advances and Challenges of Stimuli-Responsive Nucleic Acids Delivery System in Gene Therapy
Abstract
:1. Introduction
2. Classification of Nucleic Acids
3. Multiple Barriers of Gene Delivery In Vivo
4. Stimulus-Responsive Nanocarriers
4.1. pH-Responsive Nanocarriers
4.1.1. pH-Responsive Detachment of Outlayer Coating
4.1.2. pH-Responsive Gene Release
4.1.3. pH-Responsive Charge Reversal
4.2. ROS-Responsive Nanocarriers
4.3. Enzyme-Responsive Nanocarriers
4.4. ATP-Responsive Nanocarriers
4.5. External Stimulus-Responsive Nanocarriers
4.6. Magnetic Field-Controlled Gene Delivery System
4.7. Light-Responsive Gene Delivery System
4.8. Ultrasound-Targeted Gene Delivery System
4.9. Multi-Stimulus Responsive Gene Delivery System
5. Conclusions and Outlooks
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Wirth, T.; Parker, N.; Ylä-Herttuala, S. History of gene therapy. Gene 2013, 525, 162–169. [Google Scholar] [CrossRef] [PubMed]
- Piotrowski-Daspit, A.S.; Kauffman, A.C.; Bracaglia, L.G.; Saltzman, W.M. Polymeric vehicles for nucleic acid delivery. Adv. Drug Deliv. Rev. 2020, 156, 119–132. [Google Scholar] [CrossRef] [PubMed]
- Takakusa, H.; Iwazaki, N.; Nishikawa, M.; Yoshida, T.; Obika, S.; Inoue, T. Drug metabolism and pharmacokinetics of antisense oligonucleotide therapeutics: Typical profiles, evaluation approaches, and points to consider compared with small molecule drugs. Nucleic Acid Ther. 2023, 33, 83–94. [Google Scholar] [CrossRef] [PubMed]
- Rupaimoole, R.; Slack, F.J. MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat. Rev. Drug Discov. 2017, 16, 203–222. [Google Scholar] [CrossRef]
- Noé, V.; Aubets, E.; Félix, A.J.; Ciudad, C.J. Nucleic acids therapeutics using PolyPurine Reverse Hoogsteen hairpins. Biochem. Pharm. 2020, 189, 114371. [Google Scholar] [CrossRef]
- Wang, J.; Lu, Z.; Wientjes, M.G.; Au, J.L. Delivery of siRNA therapeutics: Barriers and carriers. AAPS J. 2010, 12, 492–503. [Google Scholar] [CrossRef]
- Kawabata, K.; Takakura, Y.; Hashida, M. The fate of plasmid DNA after intravenous injection in mice: Involvement of scavenger receptors in its hepatic uptake. Pharm. Res. 1995, 12, 825–830. [Google Scholar] [CrossRef]
- Yu, C.; Li, L.; Hu, P.; Yang, Y.; Wei, W.; Deng, X.; Wang, L.; Tay, F.R.; Ma, J. Recent Advances in Stimulus-Responsive Nanocarriers for Gene Therapy. Adv. Sci. 2021, 8, 2100540. [Google Scholar] [CrossRef]
- Samaridou, E.; Heyes, J.; Lutwyche, P. Lipid nanoparticles for nucleic acid delivery: Current perspectives. Adv. Drug Deliv. Rev. 2020, 154–155, 37–63. [Google Scholar] [CrossRef]
- Santiago-Ortiz, J.L.; Schaffer, D.V. Adeno-associated virus (AAV) vectors in cancer gene therapy. J. Control. Release 2016, 240, 287–301. [Google Scholar] [CrossRef]
- Suda, T.; Liu, D. Hydrodynamic gene delivery: Its principles and applications. Mol. Ther. 2007, 15, 2063–2069. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Zhang, L.; Liu, H.; Cheng, K. Delivery strategies of the CRISPR-Cas9 gene-editing system for therapeutic applications. J. Control. Release 2017, 266, 17–26. [Google Scholar] [CrossRef]
- Bessis, N.; GarciaCozar, F.J.; Boissier, M.C. Immune responses to gene therapy vectors: Influence on vector function and effector mechanisms. Gene Ther. 2004, 11 (Suppl. S1), S10–S17. [Google Scholar] [CrossRef] [PubMed]
- Kotterman, M.A.; Schaffer, D.V. Engineering adeno-associated viruses for clinical gene therapy. Nat. Rev. Genet. 2014, 15, 445–451. [Google Scholar] [CrossRef]
- Wang, M.; Liu, H.; Li, L.; Cheng, Y. A fluorinated dendrimer achieves excellent gene transfection efficacy at extremely low nitrogen to phosphorus ratios. Nat. Commun. 2014, 5, 3053. [Google Scholar] [CrossRef] [PubMed]
- Yan, H.J.; Oommen, O.P.; Yu, D.; Hilborn, J.; Qian, H.; Varghese, O.P. Chondroitin Sulfate-Coated DNA-Nanoplexes Enhance Transfection Efficiency by Controlling Plasmid Release from Endosomes: A New Insight into Modulating Nonviral Gene Transfection. Adv. Funct. Mater. 2015, 25, 3907–3915. [Google Scholar] [CrossRef]
- Chen, D.J.; Majors, B.S.; Zelikin, A.; Putnam, D. Structure-function relationships of gene delivery vectors in a limited polycation library. J. Control. Release 2005, 103, 273–283. [Google Scholar] [CrossRef]
- Shim, M.S.; Kwon, Y.J. Stimuli-responsive polymers and nanomaterials for gene delivery and imaging applications. Adv. Drug Deliv. Rev. 2012, 64, 1046–1059. [Google Scholar] [CrossRef]
- Wang, X.; Zhang, W. The Janus of Protein Corona on nanoparticles for tumor targeting, immunotherapy and diagnosis. J. Control. Release 2022, 345, 832–850. [Google Scholar] [CrossRef]
- Horst, A.K.; Kumashie, K.G.; Neumann, K.; Diehl, L.; Tiegs, G. Antigen presentation, autoantibody production, and therapeutic targets in autoimmune liver disease. Cell. Mol. Immunol. 2021, 18, 92–111. [Google Scholar] [CrossRef]
- Dominguez-Medina, S.; Kisley, L.; Tauzin, L.J.; Hoggard, A.; Shuang, B.; Indrasekara, A.S.; Chen, S.; Wang, L.Y.; Derry, P.J.; Liopo, A.; et al. Adsorption and Unfolding of a Single Protein Triggers Nanoparticle Aggregation. ACS Nano 2016, 10, 2103–2112. [Google Scholar] [CrossRef] [PubMed]
- Fornaguera, C.; Guerra-Rebollo, M.; Lázaro, M.; Cascante, A.; Rubio, N.; Blanco, J.; Borrós, S. In Vivo Retargeting of Poly(beta aminoester) (OM-PBAE) Nanoparticles is Influenced by Protein Corona. Adv. Healthc. Mater. 2019, 8, e1900849. [Google Scholar] [CrossRef] [PubMed]
- Dash, P.; Piras, A.M.; Dash, M. Cell membrane coated nanocarriers-an efficient biomimetic platform for targeted therapy. J. Control. Release 2020, 327, 546–570. [Google Scholar] [CrossRef] [PubMed]
- Fang, R.H.; Kroll, A.V.; Gao, W.; Zhang, L. Cell Membrane Coating Nanotechnology. Adv. Mater. 2018, 30, e1706759. [Google Scholar] [CrossRef]
- Rao, L.; Cai, B.; Bu, L.L.; Liao, Q.Q.; Guo, S.S.; Zhao, X.Z.; Dong, W.F.; Liu, W. Microfluidic Electroporation-Facilitated Synthesis of Erythrocyte Membrane-Coated Magnetic Nanoparticles for Enhanced Imaging-Guided Cancer Therapy. ACS Nano 2017, 11, 3496–3505. [Google Scholar] [CrossRef]
- Suk, J.S.; Xu, Q.; Kim, N.; Hanes, J.; Ensign, L.M. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv. Drug Deliv. Rev. 2016, 99, 28–51. [Google Scholar] [CrossRef]
- Guan, X.; Guo, Z.; Wang, T.; Lin, L.; Chen, J.; Tian, H.; Chen, X. A pH-Responsive Detachable PEG Shielding Strategy for Gene Delivery System in Cancer Therapy. Biomacromolecules 2017, 18, 1342–1349. [Google Scholar] [CrossRef]
- Liu, S.; Zhang, Y.; Zhao, X.; Wang, J.; Di, C.; Zhao, Y.; Ji, T.; Cheng, K.; Wang, Y.; Chen, L.; et al. Tumor-Specific Silencing of Tissue Factor Suppresses Metastasis and Prevents Cancer-Associated Hypercoagulability. Nano Lett. 2019, 19, 4721–4730. [Google Scholar] [CrossRef]
- Karimi, M.; Ghasemi, A.; Sahandi Zangabad, P.; Rahighi, R.; Moosavi Basri, S.M.; Mirshekari, H.; Amiri, M.; Shafaei Pishabad, Z.; Aslani, A.; Bozorgomid, M.; et al. Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chem. Soc. Rev. 2016, 45, 1457–1501. [Google Scholar] [CrossRef]
- Sun, M.; Wang, K.; Oupický, D. Advances in Stimulus-Responsive Polymeric Materials for Systemic Delivery of Nucleic Acids. Adv. Healthc. Mater. 2018, 7, 1701070. [Google Scholar] [CrossRef]
- Rychak, J.J.; Klibanov, A.L. Nucleic acid delivery with microbubbles and ultrasound. Adv. Drug Deliv. Rev. 2014, 72, 82–93. [Google Scholar] [CrossRef] [PubMed]
- Gupta, A.; Andresen, J.L.; Manan, R.S.; Langer, R. Nucleic acid delivery for therapeutic applications. Adv. Drug Deliv. Rev. 2021, 178, 113834. [Google Scholar] [CrossRef] [PubMed]
- Miele, E.; Spinelli, G.P.; Miele, E.; Di Fabrizio, E.; Ferretti, E.; Tomao, S.; Gulino, A. Nanoparticle-based delivery of small interfering RNA: Challenges for cancer therapy. Int. J. Nanomed. 2012, 7, 3637–3657. [Google Scholar] [CrossRef]
- Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef] [PubMed]
- Kowalski, P.S.; Rudra, A.; Miao, L.; Anderson, D.G. Delivering the Messenger: Advances in Technologies for Therapeutic mRNA Delivery. Mol. Ther. 2019, 27, 710–728. [Google Scholar] [CrossRef]
- Ding, N.; Lee, S.; Lieber-Kotz, M.; Yang, J.; Gao, X. Advances in genome editing for genetic hearing loss. Adv. Drug Deliv. Rev. 2021, 168, 118–133. [Google Scholar] [CrossRef]
- Xu, C.F.; Chen, G.J.; Luo, Y.L.; Zhang, Y.; Zhao, G.; Lu, Z.D.; Czarna, A.; Gu, Z.; Wang, J. Rational designs of in vivo CRISPR-Cas delivery systems. Adv. Drug Deliv. Rev. 2021, 168, 3–29. [Google Scholar] [CrossRef]
- Cong, L.; Ran, F.A.; Cox, D.; Lin, S.; Barretto, R.; Habib, N.; Hsu, P.D.; Wu, X.; Jiang, W.; Marraffini, L.A.; et al. Multiplex genome engineering using CRISPR/Cas systems. Science 2013, 339, 819–823. [Google Scholar] [CrossRef]
- Mout, R.; Ray, M.; Lee, Y.W.; Scaletti, F.; Rotello, V.M. In vivo delivery of CRISPR/Cas9 for therapeutic gene editing: Progress and challenges. Bioconjug. Chem. 2017, 28, 880–884. [Google Scholar] [CrossRef]
- Gao, X.; Jin, Z.; Tan, X.; Zhang, C.; Zou, C.; Zhang, W.; Ding, J.; Das, B.C.; Severinov, K.; Hitzeroth, I.I.; et al. Hyperbranched poly(β-amino ester) based polyplex nanopaticles for delivery of CRISPR/Cas9 system and treatment of HPV infection associated cervical cancer. J. Control. Release 2020, 321, 654–668. [Google Scholar] [CrossRef]
- Li, L.; Song, L.; Liu, X.; Yang, X.; Li, X.; He, T.; Wang, N.; Yang, S.; Yu, C.; Yin, T.; et al. Artificial virus delivers CRISPR-Cas9 system for genome editing of cells in mice. ACS Nano 2017, 11, 95–111. [Google Scholar] [CrossRef] [PubMed]
- Wang, P.; Zhang, L.; Xie, Y.; Wang, N.; Tang, R.; Zheng, W.; Jiang, X. Genome Editing for Cancer Therapy: Delivery of Cas9 Protein/sgRNA Plasmid via a Gold Nanocluster/Lipid Core-Shell Nanocarrier. Adv. Sci. 2017, 4, 1700175. [Google Scholar] [CrossRef] [PubMed]
- Wang, P.; Zhang, L.; Zheng, W.; Cong, L.; Guo, Z.; Xie, Y.; Wang, L.; Tang, R.; Feng, Q.; Hamada, Y.; et al. Thermo-triggered Release of CRISPR-Cas9 System by Lipid-Encapsulated Gold Nanoparticles for Tumor Therapy. Angew. Chem. Int. Ed. Engl. 2018, 57, 1491–1496. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.; Fu, Y.; Huang, C.; Hu, D.; Zhou, K.; Hao, Y.; Chu, B.; Yang, Y.; Qian, Z. Chlorin e6 and CRISPR-Cas9 dual-loading system with deep penetration for a synergistic tumoral photodynamic-immunotherapy. Biomaterials 2020, 255, 120194. [Google Scholar] [CrossRef] [PubMed]
- Dong, Y.; Siegwart, D.J.; Anderson, D.G. Strategies, design, and chemistry in siRNA delivery systems. Adv. Drug Deliv. Rev. 2019, 144, 133–147. [Google Scholar] [CrossRef]
- Dassie, J.P.; Liu, X.Y.; Thomas, G.S.; Whitaker, R.M.; Thiel, K.W.; Stockdale, K.R.; Meyerholz, D.K.; McCaffrey, A.P.; McNamara, J.O., II; Giangrande, P.H. Systemic administration of optimized aptamer-siRNA chimeras promotes regression of PSMA-expressing tumors. Nat. Biotechnol. 2009, 27, 839–849. [Google Scholar] [CrossRef]
- Kruspe, S.; Giangrande, P.H. Aptamer-siRNA Chimeras: Discovery, Progress, and Future Prospects. Biomedicines 2017, 5, 45. [Google Scholar] [CrossRef]
- Zhou, J.; Lazar, D.; Li, H.; Xia, X.; Satheesan, S.; Charlins, P.; O’Mealy, D.; Akkina, R.; Saayman, S.; Weinberg, M.S.; et al. Receptor-targeted aptamer-siRNA conjugate-directed transcriptional regulation of HIV-1. Theranostics 2018, 8, 1575–1590. [Google Scholar] [CrossRef]
- Sonntag, F.; Schmidt, K.; Kleinschmidt, J.A. A viral assembly factor promotes AAV2 capsid formation in the nucleolus. Proc. Natl. Acad. Sci. USA 2010, 107, 10220–10225. [Google Scholar] [CrossRef]
- Wu, Z.; Yang, H.; Colosi, P. Effect of genome size on AAV vector packaging. Mol. Ther. 2010, 18, 80–86. [Google Scholar] [CrossRef]
- Walkey, C.D.; Olsen, J.B.; Guo, H.; Emili, A.; Chan, W.C. Nanoparticle size and surface chemistry determine serum protein adsorption and macrophage uptake. J. Am. Chem. Soc. 2012, 134, 2139–2147. [Google Scholar] [CrossRef] [PubMed]
- Ogawara, K.; Furumoto, K.; Nagayama, S.; Minato, K.; Higaki, K.; Kai, T.; Kimura, T. Pre-coating with serum albumin reduces receptor-mediated hepatic disposition of polystyrene nanosphere: Implications for rational design of nanoparticles. J. Control. Release 2004, 100, 451–455. [Google Scholar] [CrossRef] [PubMed]
- Ritz, S.; Schöttler, S.; Kotman, N.; Baier, G.; Musyanovych, A.; Kuharev, J.; Landfester, K.; Schild, H.; Jahn, O.; Tenzer, S.; et al. Protein corona of nanoparticles: Distinct proteins regulate the cellular uptake. Biomacromolecules 2015, 16, 1311–1321. [Google Scholar] [CrossRef] [PubMed]
- Nelson, C.E.; Kintzing, J.R.; Hanna, A.; Shannon, J.M.; Gupta, M.K.; Duvall, C.L. Balancing cationic and hydrophobic content of PEGylated siRNA polyplexes enhances endosome escape, stability, blood circulation time, and bioactivity in vivo. ACS Nano 2013, 7, 8870–8880. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Zhang, Q.; Chang, H.; Cheng, Y. Surface-engineered dendrimers in gene delivery. Chem. Rev. 2015, 115, 5274–5300. [Google Scholar] [CrossRef]
- Wang, L.H.; Wu, D.C.; Xu, H.X.; You, Y.Z. High DNA-Binding Affinity and Gene-Transfection Efficacy of Bioreducible Cationic Nanomicelles with a Fluorinated Core. Angew. Chem. Int. Ed. Engl. 2016, 55, 755–759. [Google Scholar] [CrossRef]
- Zhou, Z.; Li, C.; Zhang, M.; Zhang, Q.; Qian, C.; Oupicky, D.; Sun, M. Charge and Assembly Reversible Micelles Fueled by Intracellular ATP for Improved siRNA Transfection. ACS Appl. Mater. Interfaces 2018, 10, 32026–32037. [Google Scholar] [CrossRef]
- Qiu, C.; Han, H.H.; Sun, J.; Zhang, H.T.; Wei, W.; Cui, S.H.; Chen, X.; Wang, J.C.; Zhang, Q. Regulating intracellular fate of siRNA by endoplasmic reticulum membrane-decorated hybrid nanoplexes. Nat. Commun. 2019, 10, 2702. [Google Scholar] [CrossRef]
- Zhang, L.; Wang, P.; Feng, Q.; Wang, N.; Chen, Z.; Huang, Y.; Zheng, W.; Jiang, X. Lipid nanoparticle-mediated efficient delivery of CRISPR/Cas9 for tumor therapy. NPG Asia Mater. 2017, 9, e441. [Google Scholar] [CrossRef]
- Liu, Y.; Chen, J.; Tang, Y.; Li, S.; Dou, Y.; Zheng, J. Synthesis and Characterization of Quaternized Poly(β-amino ester) for Highly Efficient Delivery of Small Interfering RNA. Mol. Pharm. 2018, 15, 4558–4567. [Google Scholar] [CrossRef]
- Rui, Y.; Wilson, D.R.; Sanders, K.; Green, J.J. Reducible Branched Ester-Amine Quadpolymers (rBEAQs) Codelivering Plasmid DNA and RNA Oligonucleotides Enable CRISPR/Cas9 Genome Editing. ACS Appl. Mater. Interfaces 2019, 11, 10472–10480. [Google Scholar] [CrossRef]
- Wang, Y.; Wang, M.; Chen, H.; Liu, H.; Zhang, Q.; Cheng, Y. Fluorinated dendrimer for TRAIL gene therapy in cancer treatment. J. Mater. Chem. B 2016, 4, 1354–1360. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Wang, Q.; Liu, Q.; Zheng, Y.; Zheng, C.; Yi, K.; Zhao, Y.; Gu, Y.; Wang, Y.; Wang, C.; et al. Dual-Locking Nanoparticles Disrupt the PD-1/PD-L1 Pathway for Efficient Cancer Immunotherapy. Adv. Mater. 2019, 31, e1905751. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Chen, Y.; Xin, H.; Wan, T.; Ping, Y. Near-infrared optogenetic engineering of photothermal nanoCRISPR for programmable genome editing. Proc. Natl. Acad. Sci. USA 2020, 117, 2395–2405. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Xu, M.; Zhao, Y.; Chen, X.; Zhu, X.; Wei, C.; Zhao, S.; Liu, J.; Qin, X. Flower-like gold nanoparticles for enhanced photothermal anticancer therapy by the delivery of pooled siRNA to inhibit heat shock stress response. J. Mater. Chem. B 2019, 7, 586–597. [Google Scholar] [CrossRef] [PubMed]
- Tang, H.; Xu, X.; Chen, Y.; Xin, H.; Wan, T.; Li, B.; Pan, H.; Li, D.; Ping, Y. Reprogramming the tumor microenvironment through second-near-infrared-window photothermal genome editing of PD-L1 mediated by supramolecular gold nanorods for enhanced cancer immunotherapy. Adv. Mater. 2021, 33, e2006003. [Google Scholar] [CrossRef]
- He, X.W.; Liu, T.; Chen, Y.X.; Cheng, D.J.; Li, X.R.; Xiao, Y.; Feng, Y.L. Calcium carbonate nanoparticle delivering vascular endothelial growth factor-C siRNA effectively inhibits lymphangiogenesis and growth of gastric cancer in vivo. Cancer Gene Ther. 2008, 15, 193–202. [Google Scholar] [CrossRef]
- Wang, C.Q.; Gong, M.Q.; Wu, J.L.; Zhuo, R.X.; Cheng, S.X. Dual-functionalized calcium carbonate based gene delivery system for efficient gene delivery. RSC Adv. 2014, 4, 38623–38629. [Google Scholar] [CrossRef]
- Zhao, H.; Li, T.; Yao, C.; Gu, Z.; Liu, C.; Li, J.; Yang, D. Dual Roles of Metal-Organic Frameworks as Nanocarriers for miRNA Delivery and Adjuvants for Chemodynamic Therapy. ACS Appl. Mater. Interfaces 2021, 13, 6034–6042. [Google Scholar] [CrossRef]
- Zhuang, J.; Gong, H.; Zhou, J.; Zhang, Q.; Gao, W.; Fang, R.H.; Zhang, L. Targeted gene silencing in vivo by platelet membrane-coated metal-organic framework nanoparticles. Sci. Adv. 2020, 6, eaaz6108. [Google Scholar] [CrossRef]
- Lin, Y.; Wu, J.; Gu, W.; Huang, Y.; Tong, Z.; Huang, L.; Tan, J. Exosome-Liposome Hybrid Nanoparticles Deliver CRISPR/Cas9 System in MSCs. Adv. Sci. 2018, 5, 1700611. [Google Scholar] [CrossRef] [PubMed]
- Munagala, R.; Aqil, F.; Jeyabalan, J.; Kandimalla, R.; Wallen, M.; Tyagi, N.; Wilcher, S.; Yan, J.; Schultz, D.J.; Spencer, W.; et al. Exosome-mediated delivery of RNA and DNA for gene therapy. Cancer Lett. 2021, 505, 58–72. [Google Scholar] [CrossRef] [PubMed]
- Zhao, L.; Gu, C.; Gan, Y.; Shao, L.; Chen, H.; Zhu, H. Exosome-mediated siRNA delivery to suppress postoperative breast cancer metastasis. J. Control. Release 2020, 318, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Lo, Y.L.; Chang, C.H.; Wang, C.S.; Yang, M.H.; Lin, A.M.; Hong, C.J.; Tseng, W.H. PEG-coated nanoparticles detachable in acidic microenvironments for the tumor-directed delivery of chemo- and gene therapies for head and neck cancer. Theranostics 2020, 10, 6695–6714. [Google Scholar] [CrossRef] [PubMed]
- Ma, D.; Liu, H.; Zhao, P.; Ye, L.; Zou, H.; Zhao, X.; Dai, H.; Kong, X.; Liu, P. Programing Assembling/Releasing Multifunctional miRNA Nanomedicine to Treat Prostate Cancer. ACS Appl. Mater. Interfaces 2020, 12, 9032–9040. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.Z.; Niu, J.; Ma, H.J.; Dad, H.A.; Shao, H.T.; Yuan, T.J.; Peng, L.H. Transdermal siRNA delivery by pH-switchable micelles with targeting effect suppress skin melanoma progression. J. Control. Release 2020, 322, 95–107. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Tian, Y.; Liu, L.; Chen, C.; Zhang, W.; Wang, L.; Guo, Q.; Ding, L.; Fu, H.; Song, H.; et al. Precise Targeting Therapy of Orthotopic Gastric Carcinoma by siRNA and Chemotherapeutic Drug Codelivered in pH-Sensitive Nano Platform. Adv. Healthc. Mater. 2021, 10, e2100966. [Google Scholar] [CrossRef]
- Liu, Y.; Yu, B.; Dai, X.; Zhao, N.; Xu, F.J. Biomineralized calcium carbonate nanohybrids for mild photothermal heating-enhanced gene therapy. Biomaterials 2021, 274, 120885. [Google Scholar] [CrossRef]
- Liu, Y.; Zhao, N.; Xu, F.J. pH-Responsive Degradable Dextran-Quantum Dot Nanohybrids for Enhanced Gene Delivery. ACS Appl. Mater. Interfaces 2019, 11, 34707–34716. [Google Scholar] [CrossRef]
- Chen, L.; Chen, C.; Chen, W.; Li, K.; Chen, X.; Tang, X.; Xie, G.; Luo, X.; Wang, X.; Liang, H.; et al. Biodegradable Black Phosphorus Nanosheets Mediate Specific Delivery of hTERT siRNA for Synergistic Cancer Therapy. ACS Appl. Mater. Interfaces 2018, 10, 21137–21148. [Google Scholar] [CrossRef]
- Saw, P.E.; Yao, H.; Lin, C.; Tao, W.; Farokhzad, O.C.; Xu, X. Stimuli-Responsive Polymer-Prodrug Hybrid Nanoplatform for Multistage siRNA Delivery and Combination Cancer Therapy. Nano Lett. 2019, 19, 5967–5974. [Google Scholar] [CrossRef] [PubMed]
- Nie, J.J.; Qiao, B.K.; Duan, S.; Xu, C.; Chen, B.Y.; Hao, W.J.; Yu, B.R.; Li, Y.L.; Du, J.; Xu, F.J. Unlockable Nanocomplexes with Self-Accelerating Nucleic Acid Release for Effective Staged Gene Therapy of Cardiovascular Diseases. Adv. Mater. 2018, 30, 1801570. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Shen, C.N.; Zhao, N.; Xu, F.J. Redox-Responsive and Drug-Embedded Silica Nanoparticles with Unique Self-Destruction Features for Efficient Gene/Drug Codelivery. Adv. Funct. Mater. 2017, 27, 1606229. [Google Scholar] [CrossRef]
- Qiu, M.; Li, Y.; Bloomer, H.; Xu, Q. Developing Biodegradable Lipid Nanoparticles for Intracellular mRNA Delivery and Genome Editing. Acc. Chem. Res. 2021, 54, 4001–4011. [Google Scholar] [CrossRef]
- Liu, S.; Liu, J.; Li, H.; Mao, K.; Wang, H.; Meng, X.; Wang, J.; Wu, C.; Chen, H.; Wang, X.; et al. An optimized ionizable cationic lipid for brain tumor-targeted siRNA delivery and glioblastoma immunotherapy. Biomaterials 2022, 287, 121645. [Google Scholar] [CrossRef]
- Hafez, I.M.; Maurer, N.; Cullis, P.R. On the mechanism whereby cationic lipids promote intracellular delivery of polynucleic acids. Gene Ther. 2001, 8, 1188–1196. [Google Scholar] [CrossRef]
- Wittrup, A.; Ai, A.; Liu, X.; Hamar, P.; Trifonova, R.; Charisse, K.; Manoharan, M.; Kirchhausen, T.; Lieberman, J. Visualizing lipid-formulated siRNA release from endosomes and target gene knockdown. Nat. Biotechnol. 2015, 33, 870–876. [Google Scholar] [CrossRef]
- Wang, Y.; Qi, H.; Liu, Y.; Duan, C.; Liu, X.; Xia, T.; Chen, D.; Piao, H.L.; Liu, H.X. The double-edged roles of ROS in cancer prevention and therapy. Theranostics 2021, 11, 4839–4857. [Google Scholar] [CrossRef]
- Ruan, C.; Liu, L.; Wang, Q.; Chen, X.; Chen, Q.; Lu, Y.; Zhang, Y.; He, X.; Zhang, Y.; Guo, Q.; et al. Reactive Oxygen Species-Biodegradable Gene Carrier for the Targeting Therapy of Breast Cancer. ACS Appl. Mater. Interfaces 2018, 10, 10398–10408. [Google Scholar] [CrossRef]
- Trachootham, D.; Alexandre, J.; Huang, P. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach? Nat. Rev. Drug Discov. 2009, 8, 579–591. [Google Scholar] [CrossRef]
- Zhou, Z.; Li, H.; Wang, K.; Guo, Q.; Li, C.; Jiang, H.; Hu, Y.; Oupicky, D.; Sun, M. Bioreducible Cross-Linked Hyaluronic Acid/Calcium Phosphate Hybrid Nanoparticles for Specific Delivery of siRNA in Melanoma Tumor Therapy. ACS Appl. Mater. Interfaces 2017, 9, 14576–14589. [Google Scholar] [CrossRef] [PubMed]
- Wang, G.; Zhu, D.; Zhou, Z.; Piao, Y.; Tang, J.; Shen, Y. Glutathione-Specific and Intracellularly Labile Polymeric Nanocarrier for Efficient and Safe Cancer Gene Delivery. ACS Appl. Mater. Interfaces 2020, 12, 14825–14838. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Song, L.; Liu, Q.; Tian, R.; Shang, Y.; Liu, F.; Liu, S.; Zhao, S.; Han, Z.; Sun, J.; et al. A Tubular DNA Nanodevice as a siRNA/Chemo-Drug Co-delivery Vehicle for Combined Cancer Therapy. Angew. Chem. Int. Ed. Engl. 2021, 60, 2594–2598. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.; Zhong, D.; Li, Y.; Wu, H.; Zhang, H.; Mao, H.; Yang, J.; Luo, K.; Gong, Q.; Gu, Z. A tumor-activatable peptide supramolecular nanoplatform for the delivery of dual-gene targeted siRNAs for drug-resistant cancer treatment. Nanoscale 2021, 13, 4887–4898. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Hong, K.; Sun, Q.; Zhu, Y.; Du, J. Bioreducible, arginine-rich polydisulfide-based siRNA nanocomplexes with excellent tumor penetration for efficient gene silencing. Biomater. Sci. 2021, 9, 5275–5292. [Google Scholar] [CrossRef]
- Nie, J.J.; Liu, Y.; Qi, Y.; Zhang, N.; Yu, B.; Chen, D.F.; Yang, M.; Xu, F.J. Charge-reversal nanocomolexes-based CRISPR/Cas9 delivery system for loss-of-function oncogene editing in hepatocellular carcinoma. J. Control. Release 2021, 333, 362–373. [Google Scholar] [CrossRef]
- Liu, X.; Xiang, J.; Zhu, D.; Jiang, L.; Zhou, Z.; Tang, J.; Liu, X.; Huang, Y.; Shen, Y. Fusogenic Reactive Oxygen Species Triggered Charge-Reversal Vector for Effective Gene Delivery. Adv. Mater. 2016, 28, 1743–1752. [Google Scholar] [CrossRef]
- Xiang, J.; Liu, X.; Zhou, Z.; Zhu, D.; Zhou, Q.; Piao, Y.; Jiang, L.; Tang, J.; Liu, X.; Shen, Y. Reactive Oxygen Species (ROS)-Responsive Charge-Switchable Nanocarriers for Gene Therapy of Metastatic Cancer. ACS Appl. Mater. Interfaces 2018, 10, 43352–43362. [Google Scholar] [CrossRef]
- Wang, Q.Y.; Xu, Y.S.; Zhang, N.X.; Dong, Z.P.; Zhao, B.N.; Liu, L.C.; Lu, T.; Wang, Y. Phenylboronic ester-modified anionic micelles for ROS-stimuli response in HeLa cell. Drug Deliv. 2020, 27, 681–690. [Google Scholar] [CrossRef]
- Zheng, N.; Luo, X.; Zhang, Z.; Wang, A.; Song, W. Cationic Polyporphyrins as siRNA Delivery Vectors for Photodynamic and Gene Synergistic Anticancer Therapy. ACS Appl. Mater. Interfaces 2021, 13, 27513–27521. [Google Scholar] [CrossRef]
- Wilson, D.S.; Dalmasso, G.; Wang, L.; Sitaraman, S.V.; Merlin, D.; Murthy, N. Orally delivered thioketal nanoparticles loaded with TNF-α-siRNA target inflammation and inhibit gene expression in the intestines. Nat. Mater. 2010, 9, 923–928. [Google Scholar] [CrossRef] [PubMed]
- Qiu, N.; Gao, J.; Liu, Q.; Wang, J.; Shen, Y. Enzyme-Responsive Charge-Reversal Polymer-Mediated Effective Gene Therapy for Intraperitoneal Tumors. Biomacromolecules 2018, 19, 2308–2319. [Google Scholar] [CrossRef] [PubMed]
- Ren, F.; Tang, R.; Zhang, X.; Madushi, W.M.; Luo, D.; Dang, Y.; Li, Z.; Wei, K.; Chen, G. Overexpression of MMP Family Members Functions as Prognostic Biomarker for Breast Cancer Patients: A Systematic Review and Meta-Analysis. PLoS ONE 2015, 10, e0135544. [Google Scholar] [CrossRef] [PubMed]
- Klassen, L.M.B.; Chequin, A.; Manica, G.C.M.; Biembengut, I.V.; Toledo, M.B.; Baura, V.A.; de Pedrosa, F.O.; Ramos, E.A.S.; Costa, F.F.; de Souza, E.M.; et al. MMP9 gene expression regulation by intragenic epigenetic modifications in breast cancer. Gene 2018, 642, 461–466. [Google Scholar] [CrossRef]
- Malik, R.; Lelkes, P.I.; Cukierman, E. Biomechanical and biochemical remodeling of stromal extracellular matrix in cancer. Trends Biotechnol. 2015, 33, 230–236. [Google Scholar] [CrossRef]
- Paduch, R.; Kandefer-Szerszeń, M.; Szuster-Ciesielska, A.; Plewka, K. Transforming growth factor-beta1 modulates metalloproteinase-2 and -9, nitric oxide, RhoA and alpha-smooth muscle actin expression in colon adenocarcinoma cells. Cell Biol. Int. 2010, 34, 213–223. [Google Scholar] [CrossRef]
- Boehnke, N.; Correa, S.; Hao, L.; Wang, W.; Straehla, J.P.; Bhatia, S.N.; Hammond, P.T. Theranostic Layer-by-Layer Nanoparticles for Simultaneous Tumor Detection and Gene Silencing. Angew. Chem. Int. Ed. Engl. 2020, 59, 2776–2783. [Google Scholar] [CrossRef]
- Komin, A.; Russell, L.M.; Hristova, K.A.; Searson, P.C. Peptide-based strategies for enhanced cell uptake, transcellular transport, and circulation: Mechanisms and challenges. Adv. Drug. Deliv. Rev. 2017, 110–111, 52–64. [Google Scholar] [CrossRef]
- Shi, N.Q.; Qi, X.R.; Xiang, B.; Zhang, Y. A survey on “Trojan Horse” peptides: Opportunities, issues and controlled entry to “Troy”. J. Control. Release 2014, 194, 53–70. [Google Scholar] [CrossRef]
- Ma, D.X.; Shi, N.Q.; Qi, X.R. Distinct transduction modes of arginine-rich cell-penetrating peptides for cargo delivery into tumor cells. Int. J. Pharm. 2011, 419, 200–208. [Google Scholar] [CrossRef]
- Gao, W.; Meng, T.; Shi, N.; Zhuang, H.; Yang, Z.; Qi, X. Targeting and microenvironment-responsive lipid nanocarrier for the enhancement of tumor cell recognition and therapeutic efficiency. Adv. Healthc. Mater. 2015, 4, 748–759. [Google Scholar] [CrossRef] [PubMed]
- Gao, W.; Xiang, B.; Meng, T.T.; Liu, F.; Qi, X.R. Chemotherapeutic drug delivery to cancer cells using a combination of folate targeting and tumor microenvironment-sensitive polypeptides. Biomaterials 2013, 34, 4137–4149. [Google Scholar] [CrossRef] [PubMed]
- Han, Q.J.; Lan, X.T.; Wen, Y.; Zhang, C.Z.; Cleary, M.; Sayyed, Y.; Huang, G.; Tuo, X.; Yi, L.; Xi, Z.; et al. Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier to Enhance Endocytosis as Efficient Targeted Delivery System for Cancer Diagnosis and Therapy. Adv. Healthc. Mater. 2021, 10, e2002143. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Sun, A.; Liu, Y.; Zhang, W.; Pang, N.; Cheng, S.; Qi, X. Amphiphilic dendrimer engineered nanocarrier systems for co-delivery of siRNA and paclitaxel to matrix metalloproteinase-rich tumors for synergistic therapy. NPG Asia Mater. 2018, 10, 238–254. [Google Scholar] [CrossRef]
- Lin, M.; Yang, Z.; Yang, Y.; Peng, Y.; Li, J.; Du, Y.; Sun, Q.; Gao, D.; Yuan, Q.; Zhou, Y.; et al. CRISPR-based in situ engineering tumor cells to reprogram macrophages for effective cancer immunotherapy. Nano Today 2022, 42, 101359. [Google Scholar] [CrossRef]
- Lv, J.; Chang, H.; Wang, Y.; Wang, M.; Xiao, J.; Zhang, Q.; Cheng, Y. Fluorination on polyethylenimine allows efficient 2D and 3D cell culture gene delivery. J. Mater. Chem. B 2015, 3, 642–650. [Google Scholar] [CrossRef]
- Wang, H.; Wang, Y.; Wang, Y.; Hu, J.; Li, T.; Liu, H.; Zhang, Q.; Cheng, Y. Self-Assembled Fluorodendrimers Combine the Features of Lipid and Polymeric Vectors in Gene Delivery. Angew. Chem. Int. Ed. Engl. 2015, 54, 11647–11651. [Google Scholar] [CrossRef]
- Feng, G.; Zha, Z.; Huang, Y.; Li, J.; Wang, Y.; Ke, W.; Chen, H.; Liu, L.; Song, Y.; Ge, Z. Sustained and Bioresponsive Two-Stage Delivery of Therapeutic miRNA via Polyplex Micelle-Loaded Injectable Hydrogels for Inhibition of Intervertebral Disc Fibrosis. Adv. Healthc. Mater. 2018, 7, e1800623. [Google Scholar] [CrossRef]
- Girish, K.S.; Kemparaju, K. The magic glue hyaluronan and its eraser hyaluronidase: A biological overview. Life Sci. 2007, 80, 1921–1943. [Google Scholar] [CrossRef]
- McAtee, C.O.; Barycki, J.J.; Simpson, M.A. Emerging roles for hyaluronidase in cancer metastasis and therapy. Adv. Cancer Res. 2014, 123, 1–34. [Google Scholar] [CrossRef]
- Bertrand, P.; Girard, N.; Duval, C.; d’Anjou, J.; Chauzy, C.; Ménard, J.F.; Delpech, B. Increased hyaluronidase levels in breast tumor metastases. Int. J. Cancer 1997, 73, 327–331. [Google Scholar] [CrossRef]
- Lokeshwar, V.B.; Rubinowicz, D.; Schroeder, G.L.; Forgacs, E.; Minna, J.D.; Block, N.L.; Nadji, M.; Lokeshwar, B.L. Stromal and epithelial expression of tumor markers hyaluronic acid and HYAL1 hyaluronidase in prostate cancer. J. Biol. Chem. 2001, 276, 11922–11932. [Google Scholar] [CrossRef] [PubMed]
- He, Y.; Zhou, J.; Ma, S.; Nie, Y.; Yue, D.; Jiang, Q.; Wali, A.R.; Tang, J.Z.; Gu, Z. Multi-Responsive “Turn-On” Nanocarriers for Efficient Site-Specific Gene Delivery In Vitro and In Vivo. Adv. Healthc. Mater. 2016, 5, 2799–2812. [Google Scholar] [CrossRef] [PubMed]
- Rajendrakumar, S.K.; Venu, A.; Revuri, V.; George Thomas, R.; Thirunavukkarasu, G.K.; Zhang, J.; Vijayan, V.; Choi, S.Y.; Lee, J.Y.; Lee, Y.K.; et al. Hyaluronan-Stabilized Redox-Sensitive Nanoassembly for Chemo-Gene Therapy and Dual T1/T2 MR Imaging in Drug-Resistant Breast Cancer Cells. Mol. Pharm. 2019, 16, 2226–2234. [Google Scholar] [CrossRef]
- Sun, Y.; Liu, L.; Zhou, L.; Yu, S.; Lan, Y.; Liang, Q.; Liu, J.; Cao, A.; Liu, Y. Tumor Microenvironment-Triggered Charge Reversal Polymetformin-Based Nanosystem Co-Delivered Doxorubicin and IL-12 Cytokine Gene for Chemo-Gene Combination Therapy on Metastatic Breast Cancer. ACS Appl. Mater. Interfaces 2020, 12, 45873–45890. [Google Scholar] [CrossRef]
- Zhao, Y.; He, Z.; Gao, H.; Tang, H.; He, J.; Guo, Q.; Zhang, W.; Liu, J. Fine Tuning of Core-Shell Structure of Hyaluronic Acid/Cell-Penetrating Peptides/siRNA Nanoparticles for Enhanced Gene Delivery to Macrophages in Antiatherosclerotic Therapy. Biomacromolecules 2018, 19, 2944–2956. [Google Scholar] [CrossRef]
- Choi, K.Y.; Silvestre, O.F.; Huang, X.; Min, K.H.; Howard, G.P.; Hida, N.; Jin, A.J.; Carvajal, N.; Lee, S.W.; Hong, J.I.; et al. Versatile RNA interference nanoplatform for systemic delivery of RNAs. ACS Nano 2014, 8, 4559–4570. [Google Scholar] [CrossRef]
- Jiang, C.; Chen, J.; Li, Z.; Wang, Z.; Zhang, W.; Liu, J. Recent advances in the development of polyethylenimine-based gene vectors for safe and efficient gene delivery. Expert Opin. Drug Deliv. 2019, 16, 363–376. [Google Scholar] [CrossRef]
- Naito, M.; Ishii, T.; Matsumoto, A.; Miyata, K.; Miyahara, Y.; Kataoka, K. A phenylboronate-functionalized polyion complex micelle for ATP-triggered release of siRNA. Angew. Chem. Int. Ed. Engl. 2012, 51, 10751–10755. [Google Scholar] [CrossRef]
- Zhang, J.; Wang, Y.; Chen, J.; Liang, X.; Han, H.; Yang, Y.; Li, Q.; Wang, Y. Inhibition of cell proliferation through an ATP-responsive co-delivery system of doxorubicin and Bcl-2 siRNA. Int. J. Nanomed. 2017, 12, 4721–4732. [Google Scholar] [CrossRef]
- Yoshinaga, N.; Ishii, T.; Naito, M.; Endo, T.; Uchida, S.; Cabral, H.; Osada, K.; Kataoka, K. Polyplex Micelles with Phenylboronate/Gluconamide Cross-Linking in the Core Exerting Promoted Gene Transfection through Spatiotemporal Responsivity to Intracellular pH and ATP Concentration. J. Am. Chem. Soc. 2017, 139, 18567–18575. [Google Scholar] [CrossRef] [PubMed]
- Fan, X.; Zhao, X.; Su, W.; Tang, X. Triton X-100-Modified Adenosine Triphosphate-Responsive siRNA Delivery Agent for Antitumor Therapy. Mol. Pharm. 2020, 17, 3696–3708. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.; Zhang, Q.; Zhang, M.; Li, H.; Chen, G.; Qian, C.; Oupicky, D.; Sun, M. ATP-activated decrosslinking and charge-reversal vectors for siRNA delivery and cancer therapy. Theranostics 2018, 8, 4604–4619. [Google Scholar] [CrossRef] [PubMed]
- Li, F.; Yu, W.; Zhang, J.; Dong, Y.; Ding, X.; Ruan, X.; Gu, Z.; Yang, D. Spatiotemporally programmable cascade hybridization of hairpin DNA in polymeric nanoframework for precise siRNA delivery. Nat. Commun. 2021, 12, 1138. [Google Scholar] [CrossRef] [PubMed]
- Bauer, L.M.; Situ, S.F.; Griswold, M.A.; Samia, A.C. High-performance iron oxide nanoparticles for magnetic particle imaging-guided hyperthermia (hMPI). Nanoscale 2016, 8, 12162–12169. [Google Scholar] [CrossRef] [PubMed]
- Cole, A.J.; David, A.E.; Wang, J.; Galbán, C.J.; Yang, V.C. Magnetic brain tumor targeting and biodistribution of long-circulating PEG-modified, cross-linked starch-coated iron oxide nanoparticles. Biomaterials 2011, 32, 6291–6301. [Google Scholar] [CrossRef]
- Dwivedi, P.; Kiran, S.; Han, S.; Dwivedi, M.; Khatik, R.; Fan, R.; Mangrio, F.A.; Du, K.; Zhu, Z.; Yang, C.; et al. Magnetic Targeting and Ultrasound Activation of Liposome-Microbubble Conjugate for Enhanced Delivery of Anticancer Therapies. ACS Appl. Mater. Interfaces 2020, 12, 23737–23751. [Google Scholar] [CrossRef]
- Wang, Z.; Chang, Z.; Lu, M.; Shao, D.; Yue, J.; Yang, D.; Zheng, X.; Li, M.; He, K.; Zhang, M.; et al. Shape-controlled magnetic mesoporous silica nanoparticles for magnetically-mediated suicide gene therapy of hepatocellular carcinoma. Biomaterials 2018, 154, 147–157. [Google Scholar] [CrossRef]
- Zhou, Z.; Sun, Y.; Shen, J.; Wei, J.; Yu, C.; Kong, B.; Liu, W.; Yang, H.; Yang, S.; Wang, W. Iron/iron oxide core/shell nanoparticles for magnetic targeting MRI and near-infrared photothermal therapy. Biomaterials 2014, 35, 7470–7478. [Google Scholar] [CrossRef]
- Wang, J.; Chen, P.; Dong, Y.; Xie, H.; Wang, Y.; Soto, F.; Ma, P.; Feng, X.; Du, W.; Liu, B.F. Designer exosomes enabling tumor targeted efficient chemo/gene/photothermal therapy. Biomaterials 2021, 276, 121056. [Google Scholar] [CrossRef]
- Qiu, F.M.; Fujita, S.; Mhanna, R.; Zhang, L.; Simona, B.R.; Nelson, B.J. Magnetic Helical Microswimmers Functionalized with Lipoplexes for Targeted Gene Delivery. Adv. Funct. Mater. 2015, 25, 1666–1671. [Google Scholar] [CrossRef]
- Sun, Q.; Yang, Z.; Lin, M.; Peng, Y.; Wang, R.; Du, Y.; Zhou, Y.; Li, J.; Qi, X. Phototherapy and anti-GITR antibody-based therapy synergistically reinvigorate immunogenic cell death and reject established cancers. Biomaterials 2021, 269, 120648. [Google Scholar] [CrossRef] [PubMed]
- Tang, H.; Wang, J.; Yu, L.; Zhang, S.; Yang, H.; Li, X.; Brash, J.L.; Wang, L.; Chen, H. Ultrahigh Efficiency and Minimalist Intracellular Delivery of Macromolecules Mediated by Latent-Photothermal Surfaces. ACS Appl. Mater. Interfaces 2021, 13, 12594–12602. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Du, Y.; Sun, Q.; Peng, Y.; Wang, R.; Zhou, Y.; Wang, Y.; Zhang, C.; Qi, X. Albumin-Based Nanotheranostic Probe with Hypoxia Alleviating Potentiates Synchronous Multimodal Imaging and Phototherapy for Glioma. ACS Nano 2020, 14, 6191–6212. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Miao, W.; Wang, F.; Cheng, Y. A Self-Assembled Coumarin-Anchored Dendrimer for Efficient Gene Delivery and Light-Responsive Drug Delivery. Biomacromolecules 2018, 19, 2194–2201. [Google Scholar] [CrossRef]
- Lin, X.; Wu, M.; Li, M.; Cai, Z.; Sun, H.; Tan, X.; Li, J.; Zeng, Y.; Liu, X.; Liu, J. Photo-responsive hollow silica nanoparticles for light-triggered genetic and photodynamic synergistic therapy. Acta Biomater. 2018, 76, 178–192. [Google Scholar] [CrossRef]
- Zhang, Q.; Kuang, G.; He, S.; Lu, H.; Cheng, Y.; Zhou, D.; Huang, Y. Photoactivatable Prodrug-Backboned Polymeric Nanoparticles for Efficient Light-Controlled Gene Delivery and Synergistic Treatment of Platinum-Resistant Ovarian Cancer. Nano Lett. 2020, 20, 3039–3049. [Google Scholar] [CrossRef]
- Kuang, G.; Lu, H.; He, S.; Xiong, H.; Yu, J.; Zhang, Q.; Huang, Y. Near-Infrared Light-Triggered Polyprodrug/siRNA Loaded Upconversion Nanoparticles for Multi-Modality Imaging and Synergistic Cancer Therapy. Adv. Healthc. Mater. 2021, 10, e2100938. [Google Scholar] [CrossRef]
- Mallidi, S.; Anbil, S.; Bulin, A.L.; Obaid, G.; Ichikawa, M.; Hasan, T. Beyond the Barriers of Light Penetration: Strategies, Perspectives and Possibilities for Photodynamic Therapy. Theranostics 2016, 6, 2458–2487. [Google Scholar] [CrossRef]
- Wang, J.; He, H.; Xu, X.; Wang, X.; Chen, Y.; Yin, L. Far-red light-mediated programmable anti-cancer gene delivery in cooperation with photodynamic therapy. Biomaterials 2018, 171, 72–82. [Google Scholar] [CrossRef]
- Chen, L.; Li, G.; Wang, X.; Li, J.; Zhang, Y. Spherical Nucleic Acids for Near-Infrared Light-Responsive Self-Delivery of Small-Interfering RNA and Antisense Oligonucleotide. ACS Nano 2021, 15, 11929–11939. [Google Scholar] [CrossRef] [PubMed]
- Høgset, A.; Prasmickaite, L.; Selbo, P.K.; Hellum, M.; Engesaeter, B.; Bonsted, A.; Berg, K. Photochemical internalisation in drug and gene delivery. Adv. Drug Deliv. Rev. 2004, 56, 95–115. [Google Scholar] [CrossRef] [PubMed]
- Park, S.J.; Park, W.; Na, K. Tumor Intracellular-Environment Responsive Materials Shielded Nano-Complexes for Highly Efficient Light-Triggered Gene Delivery without Cargo Gene Damage. Adv. Funct. Mater. 2015, 25, 3472–3482. [Google Scholar] [CrossRef]
- Selbo, P.K.; Weyergang, A.; Høgset, A.; Norum, O.J.; Berstad, M.B.; Vikdal, M.; Berg, K. Photochemical internalization provides time- and space-controlled endolysosomal escape of therapeutic molecules. J. Control. Release 2010, 148, 2–12. [Google Scholar] [CrossRef] [PubMed]
- Zhao, L.; Luo, Y.; Huang, Q.; Cao, Z.; Yang, X. Photo-Enhanced CRISPR/Cas9 System Enables Robust PD-L1 Gene Disruption in Cancer Cells and Cancer Stem-Like Cells for Efficient Cancer Immunotherapy. Small 2020, 16, 2004879. [Google Scholar] [CrossRef] [PubMed]
- Defelippis, M.R.; Faraggi, M.; Klapper, M.H. Redox Potentials Of The Azide And Dithiocyanate Radicals. J. Phys. Chem. 1990, 94, 2420–2424. [Google Scholar] [CrossRef]
- Ma, Z.; Du, Z.; Chen, X.; Wang, X.; Yang, Y. Fine tuning the LightOn light-switchable transgene expression system. Biochem. Biophys. Res. Commun. 2013, 440, 419–423. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Chen, X.; Yang, Y. Spatiotemporal control of gene expression by a light-switchable transgene system. Nat. Methods 2012, 9, 266–269. [Google Scholar] [CrossRef]
- He, M.; Wang, Y.; Chen, X.; Zhao, Y.; Lou, K.; Wang, Y.; Huang, L.; Hou, X.; Xu, J.; Cai, X.; et al. Spatiotemporally controllable diphtheria toxin expression using a light-switchable transgene system combining multifunctional nanoparticle delivery system for targeted melanoma therapy. J. Control. Release 2020, 319, 1–14. [Google Scholar] [CrossRef]
- Sirsi, S.R.; Borden, M.A. Advances in ultrasound mediated gene therapy using microbubble contrast agents. Theranostics 2012, 2, 1208–1222. [Google Scholar] [CrossRef]
- Wu, J.; Li, R.K. Ultrasound-targeted microbubble destruction in gene therapy: A new tool to cure human diseases. Genes Dis. 2017, 4, 64–74. [Google Scholar] [CrossRef]
- Walsh, A.P.G.; Gordon, H.N.; Peter, K.; Wang, X. Ultrasonic particles: An approach for targeted gene delivery. Adv. Drug Deliv. Rev. 2021, 179, 113998. [Google Scholar] [CrossRef]
- Un, K.; Kawakami, S.; Suzuki, R.; Maruyama, K.; Yamashita, F.; Hashida, M. Suppression of melanoma growth and metastasis by DNA vaccination using an ultrasound-responsive and mannose-modified gene carrier. Mol. Pharm. 2011, 8, 543–554. [Google Scholar] [CrossRef]
- Tayier, B.; Deng, Z.; Wang, Y.; Wang, W.; Mu, Y.; Yan, F. Biosynthetic nanobubbles for targeted gene delivery by focused ultrasound. Nanoscale 2019, 11, 14757–14768. [Google Scholar] [CrossRef]
- Shapiro, M.G.; Goodwill, P.W.; Neogy, A.; Yin, M.; Foster, F.S.; Schaffer, D.V.; Conolly, S.M. Biogenic gas nanostructures as ultrasonic molecular reporters. Nat. Nanotechnol. 2014, 9, 311–316. [Google Scholar] [CrossRef]
- Zhang, C.; Chen, J.; Song, Y.; Luo, J.; Jin, P.; Wang, X.; Xin, L.; Qiu, F.; Yao, J.; Wang, G.; et al. Ultrasound-Enhanced Reactive Oxygen Species Responsive Charge-Reversal Polymeric Nanocarriers for Efficient Pancreatic Cancer Gene Delivery. ACS Appl. Mater. Interfaces 2022, 14, 2587–2596. [Google Scholar] [CrossRef]
- Gao, Y.; Jia, L.; Wang, Q.; Hu, H.; Zhao, X.; Chen, D.; Qiao, M. pH/Redox Dual-Responsive Polyplex with Effective Endosomal Escape for Codelivery of siRNA and Doxorubicin against Drug-Resistant Cancer Cells. ACS Appl. Mater. Interfaces 2019, 11, 16296–16310. [Google Scholar] [CrossRef]
- Dadfar, S.M.; Roemhild, K.; Drude, N.I.; von Stillfried, S.; Knüchel, R.; Kiessling, F.; Lammers, T. Iron oxide nanoparticles: Diagnostic, therapeutic and theranostic applications. Adv. Drug Deliv. Rev. 2019, 138, 302–325. [Google Scholar] [CrossRef]
- Dong, X.; Yin, W.; Zhang, X.; Zhu, S.; He, X.; Yu, J.; Xie, J.; Guo, Z.; Yan, L.; Liu, X.; et al. Intelligent MoS2 Nanotheranostic for Targeted and Enzyme-/pH-/NIR-Responsive Drug Delivery To Overcome Cancer Chemotherapy Resistance Guided by PET Imaging. ACS Appl. Mater. Interfaces 2018, 10, 4271–4284. [Google Scholar] [CrossRef]
- Fu, S.; Cai, Z.; Ai, H. Stimulus-Responsive Nanoparticle Magnetic Resonance Imaging Contrast Agents: Design Considerations and Applications. Adv. Healthc. Mater. 2021, 10, e2001091. [Google Scholar] [CrossRef]
- Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef] [PubMed]
- Loughrey, D.; Dahlman, J.E. Non-liver mRNA Delivery. Acc. Chem. Res. 2022, 55, 13–23. [Google Scholar] [CrossRef] [PubMed]
- Poon, W.; Zhang, Y.N.; Ouyang, B.; Kingston, B.R.; Wu, J.L.Y.; Wilhelm, S.; Chan, W.C.W. Elimination Pathways of Nanoparticles. ACS Nano 2019, 13, 5785–5798. [Google Scholar] [CrossRef] [PubMed]
Stimuli | Delivery Systems | Sensitive Linkers/Materials | Drugs | References |
---|---|---|---|---|
pH | PEI | Schiff base bond | Luciferase plasmid | [27] |
Peptide nanoparticles (PNPs) | Peptide | Tissue factor (TF) siRNA | [28] | |
Metal–organic framework (MOF) nanoparticle ZIF-8 | ZIF-8 | miR-34a mimic | [69] | |
Solid lipid nanoparticles | Imine bond | miR-200 and irinotecan | [74] | |
PEI−PBA/miR146a/PEI−DMA-C225, PMPC polyplexes | Ester bond | miR146a | [75] | |
CaP-phospholipid complexes | CaP | As2O3 and HER2 siRNA | [77] | |
Calcium carbonate nanohybrids | Calcium carbonate | p53 plasmid | [78] | |
Dextran-quantum dot nanohybrids | Schiff base bond | p53 plasmid | [79] | |
Black phosphorus nanosheets | Black phosphorus nanosheets | HTERT siRNA | [80] | |
Lipid nanoparticles (LNP) | Ionizable lipids | CD47 siRNA and PD-L1 siRNA | [85] | |
ROS | Heparin nanoparticles | Disulfide bond | miR-499 | [82] |
Substance P (SP)-crosslinked BPEI | Boric acid ester bond | Plk1 siRNA | [89] | |
DNA nanostructures | Disulfide bond | Doxorubicin, Bcl2 siRNA and P-gp siRNA | [93] | |
Arginine-modified poly(disulfide amine)/siRNA nanocomplexes | Disulfide bond | KRAS siRNA | [95] | |
Heparin nanoparticles | Disulfide bond | CRISPR/Cas 9 plasmid targeting survivin | [96] | |
Fusogenic lipidic polyplexes | Boric acid ester bond | TRAIL plasmid | [97] | |
Poly[(2-acryloyl)ethyl(p-boronic acid benzyl) diethylammonium Bromide] (B-PDEAEA) | Boric acid ester bond | TRAIL plasmid | [98] | |
Polyphotosensitizers (pPSs) | Thioketal | HIF-1α siRNA | [100] | |
Enzyme | Cationic polymer PQDEA | Acetyloxybenzyl ester | TRAIL plasmid | [102] |
Liposome-based layer-by-layer nanoparticles | MMP9-sensitive peptide | Luciferase plasmid | [107] | |
Amphiphilic Dendrimer engineered nanocarrier system (ADENS) | MMP2/9-sensitive peptide | Paclitaxel and VEGF siRNA | [114] | |
PEI-based nanoparticles HPT-PF | MMP2/9-sensitive peptide | IL-12 plasmid and CRISPR/Cas 9 plasmid targeting CD47 | [115] | |
Polyelectrolyte complexes | HA | Luciferase plasmid | [123] | |
Hyaluronic acid (HA) coated nanoassembly | HA | Bcl-2 shRNA encoded plasmid and doxorubicin | [124] | |
Polymetformin (pmet)-based nanosystem | HA | IL-12 plasmid | [125] | |
Peptide | HA | LOX-1 siRNA | [126] | |
ATP | Polyplex Micelles | Phenylboronic acid (PBA) group | Luciferase plasmid | [131] |
4-carboxyphenylboronic acid (PBA) and dopamine Grafted vitamin E (VEDA) | Phenylboronic acid (PBA) group | Eg5 siRNA and EGFR siRNA | [132] | |
PEI-PBA | Phenylboronic acid (PBA) group | Bcl2 siRNA | [133] | |
DNA nanostructures | ATP aptamer | Plk1 siRNA | [134] | |
Magnetic field | Magnetic mesoporous silica nanoparticles (M-MSNs) | Iron oxide nanoparticles | The herpes simplex virus thymidine kinase/ganciclovir (HSV-TK/GCV) plasmid | [138] |
Exosome-based platforms | Iron oxide nanoparticles | DOX and molecular beacon targeting the miR-21 | [140] | |
Lipoplexes functionalized artificial bacterial flagella | Artificial bacterial flagella | pDNA encoding yellow–green fluorescent Venus protein | [141] | |
Light | Gold Nanorod | HSP70 promoter | CRISPR/Cas 9 plasmid targeting AAVS1 | [64] |
Gold Nanorod | HSP70 promoter | CRISPR/Cas 9 plasmid targeting PD-L1 | [66] | |
Coumarin modified PAMAM | Coumarin | TRAIL plasmid | [145] | |
Prodrug-backboned polymeric nanoparticle system | Pt(IV) prodrug | Pt(IV) prodrug and c-fos siRNA | [147] | |
TK-PEI/HAP/p53 NCs | Pheophytin a | p53 plasmid | [150] | |
Spherical nucleic acid | Pheophorbide a | HIF-1α siRNA and Bcl2 siRNA | [151] | |
Ultrasound | Mannose-modified bubble lipoplexes | Bubble lipoplexes | Plasmid co-expressing ubiquitylated gp100 and TRP-2 | [163] |
Cationic biosynthetic Nanobubble (CBNB) | Nanobubble | The pEGFP and pCMV-Luc reporter plasmids | [164] | |
B-PDEAEA loaded liposome | IR780 | TRAIL plasmid | [166] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lin, M.; Qi, X. Advances and Challenges of Stimuli-Responsive Nucleic Acids Delivery System in Gene Therapy. Pharmaceutics 2023, 15, 1450. https://doi.org/10.3390/pharmaceutics15051450
Lin M, Qi X. Advances and Challenges of Stimuli-Responsive Nucleic Acids Delivery System in Gene Therapy. Pharmaceutics. 2023; 15(5):1450. https://doi.org/10.3390/pharmaceutics15051450
Chicago/Turabian StyleLin, Meng, and Xianrong Qi. 2023. "Advances and Challenges of Stimuli-Responsive Nucleic Acids Delivery System in Gene Therapy" Pharmaceutics 15, no. 5: 1450. https://doi.org/10.3390/pharmaceutics15051450
APA StyleLin, M., & Qi, X. (2023). Advances and Challenges of Stimuli-Responsive Nucleic Acids Delivery System in Gene Therapy. Pharmaceutics, 15(5), 1450. https://doi.org/10.3390/pharmaceutics15051450