Biorobotic Drug Delivery for Biomedical Applications
Abstract
:1. Introduction
2. Design of Biorobots
2.1. Materials for Biorobots
2.2. Biorobot Shapes
2.3. Biorobot Engines
3. Cargo for Robotic Delivery
3.1. Chemical Drugs
3.2. Genes and Nucleic Acids
3.3. Therapeutic Proteins
3.4. Cells
4. Target Diseases
4.1. Cancer
4.2. Gastrointestinal Disease
4.3. Ocular Disease
4.4. Brain Disease
5. Robotic Drug Delivery System in the Context of Internet of Things
6. Challenges and Future Perspectives
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Shim, N.; Cho, H.; Jeon, S.I.; Kim, K. Recent developments in chemodrug-loaded nanomedicines and their application in combination cancer immunotherapy. J. Pharm. Investig. 2024, 54, 13–36. [Google Scholar] [CrossRef]
- Lee, J.; Lee, W.T.; Le, X.T.; Youn, Y.S. Cancer cell membrane-decorated hybrid liposomes for treating metastatic breast cancer based on enhanced cancer immunotherapy. J. Pharm. Investig. 2024, 54, 453–465. [Google Scholar] [CrossRef]
- Cho, H.-J. Recent progresses in the development of hyaluronic acid-based nanosystems for tumor-targeted drug delivery and cancer imaging. J. Pharm. Investig. 2019, 50, 115–129. [Google Scholar] [CrossRef]
- Al-Azzawi, S.; Masheta, D. Designing a drug delivery system for improved tumor treatment and targeting by functionalization of a cell-penetrating peptide. J. Pharm. Investig. 2019, 49, 643–654. [Google Scholar] [CrossRef]
- Shim, G.; Ko, S.; Kim, D.; Le, Q.V.; Park, G.T.; Lee, J.; Kwon, T.; Choi, H.G.; Kim, Y.B.; Oh, Y.K. Light-switchable systems for remotely controlled drug delivery. J. Control. Release 2017, 267, 67–79. [Google Scholar] [CrossRef] [PubMed]
- Crommelin, D.J.A.; van Hoogevest, P.; Storm, G. The role of liposomes in clinical nanomedicine development. What now? Now what? J. Control. Release 2020, 318, 256–263. [Google Scholar] [CrossRef]
- Leong, H.S.; Butler, K.S.; Brinker, C.J.; Azzawi, M.; Conlan, S.; Dufés, C.; Owen, A.; Rannard, S.; Scott, C.; Chen, C.; et al. On the issue of transparency and reproducibility in nanomedicine. Nat. Nanotechnol. 2019, 14, 629–635. [Google Scholar] [CrossRef]
- Shamsi, M.; Mohammadi, A.; Manshadi, M.K.D.; Sanati-Nezhad, A. Mathematical and computational modeling of nano-engineered drug delivery systems. J. Control. Release 2019, 307, 150–165. [Google Scholar] [CrossRef] [PubMed]
- Seynhaeve, A.L.B.; Amin, M.; Haemmerich, D.; van Rhoon, G.C.; Ten Hagen, T.L.M. Hyperthermia and smart drug delivery systems for solid tumor therapy. Adv. Drug Deliv. Rev. 2020, 163–164, 125–144. [Google Scholar] [CrossRef]
- Felfoul, O.; Mohammadi, M.; Taherkhani, S.; de Lanauze, D.; Zhong Xu, Y.Z.; Loghin, D.; Essa, S.; Jancik, S.; Houle, D.; Lafleur, M.; et al. Magneto-aerotactic bacteria deliver drug-containing nanoliposomes to tumour hypoxic regions. Nat. Nanotechnol. 2016, 11, 941–947. [Google Scholar] [CrossRef]
- Luo, M.; Feng, Y.; Wang, T.; Guan, J. Micro-/nanorobots at work in active drug delivery. Adv. Funct. Mater. 2018, 28, 1706100. [Google Scholar] [CrossRef]
- Chen, Z.; Boyken, S.E.; Jia, M.; Busch, F.; Flores-Solis, D.; Bick, M.J.; Lu, P.; VanAernum, Z.L.; Sahasrabuddhe, A.; Langan, R.A.; et al. Programmable design of orthogonal protein heterodimers. Nature 2019, 565, 106–111. [Google Scholar] [CrossRef] [PubMed]
- Hu, W.; Lum, G.Z.; Mastrangeli, M.; Sitti, M. Small-scale soft-bodied robot with multimodal locomotion. Nature 2018, 554, 81–85. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Angsantikul, P.; Liu, W.; Esteban-Fernández de Ávila, B.; Thamphiwatana, S.; Xu, M.; Sandraz, E.; Wang, X.; Delezuk, J.; Gao, W.; et al. Micromotors spontaneously neutralize gastric acid for pH-responsive payload release. Angew. Chem. Int. Ed. Engl. 2017, 56, 2156–2161. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Peyer, K.E.; Nelson, B.J. Artificial bacterial flagella for micromanipulation. Lab. Chip 2010, 10, 2203–2215. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Duan, W.; Ahmed, S.; Mallouk, T.E.; Sen, A. Small power: Autonomous nano- and micromotors propelled by self-generated gradients. Nano Today 2013, 8, 531–554. [Google Scholar] [CrossRef]
- Mhanna, R.; Qiu, F.; Zhang, L.; Ding, Y.; Sugihara, K.; Zenobi-Wong, M.; Nelson, B.J. Artificial bacterial flagella for remote-controlled targeted single-cell drug delivery. Small 2014, 10, 1953–1957. [Google Scholar] [CrossRef] [PubMed]
- Wu, Z.; Troll, J.; Jeong, H.H.; Wei, Q.; Stang, M.; Ziemssen, F.; Wang, Z.; Dong, M.; Schnichels, S.; Qiu, T.; et al. A swarm of slippery micropropellers penetrates the vitreous body of the eye. Sci. Adv. 2018, 4, eaat4388. [Google Scholar] [CrossRef]
- Jin, Z.; Nguyen, K.T.; Go, G.; Kang, B.; Min, H.K.; Kim, S.J.; Kim, Y.; Li, H.; Kim, C.S.; Lee, S.; et al. Multifunctional nanorobot system for active therapeutic delivery and synergistic chemo-photothermal therapy. Nano Lett. 2019, 19, 8550–8564. [Google Scholar] [CrossRef] [PubMed]
- Park, J.; Jin, C.; Lee, S.; Kim, J.Y.; Choi, H. Magnetically actuated degradable microrobots for actively controlled drug release and hyperthermia therapy. Adv. Healthc. Mater. 2019, 8, e1900213. [Google Scholar] [CrossRef]
- Madhanagopal, B.R.; Zhang, S.; Demirel, E.; Wady, H.; Chandrasekaran, A.R. DNA nanocarriers: Programmed to deliver. Trends Biochem. Sci. 2018, 43, 997–1013. [Google Scholar] [CrossRef] [PubMed]
- Ramezani, H.; Dietz, H. Building machines with DNA molecules. Nat. Rev. Genet. 2020, 21, 5–26. [Google Scholar] [CrossRef] [PubMed]
- Seeman, N.C.; Sleiman, H.F. DNA nanotechnology. Nat. Rev. Mater. 2017, 3, 17068. [Google Scholar] [CrossRef]
- Li, S.; Jiang, Q.; Liu, S.; Zhang, Y.; Tian, Y.; Song, C.; Wang, J.; Zou, Y.; Anderson, G.J.; Han, J.Y.; et al. A DNA nanorobot functions as a cancer therapeutic in response to a molecular trigger in vivo. Nat. Biotechnol. 2018, 36, 258–264. [Google Scholar] [CrossRef]
- Pena-Francesch, A.; Giltinan, J.; Sitti, M. Multifunctional and biodegradable self-propelled protein motors. Nat. Commun. 2019, 10, 3188. [Google Scholar] [CrossRef] [PubMed]
- Tottori, S.; Zhang, L.; Qiu, F.; Krawczyk, K.K.; Franco-Obregón, A.; Nelson, B.J. Magnetic helical micromachines: Fabrication, controlled swimming, and cargo transport. Adv. Mater. 2012, 24, 811–816. [Google Scholar] [CrossRef] [PubMed]
- Xin, C.; Yang, L.; Li, J.; Hu, Y.; Qian, D.; Fan, S.; Hu, K.; Cai, Z.; Wu, H.; Wang, D.; et al. Conical hollow microhelices with superior swimming capabilities for targeted cargo delivery. Adv. Mater. 2019, 31, e1808226. [Google Scholar] [CrossRef] [PubMed]
- Xing, J.F.; Zheng, M.L.; Duan, X.M. Two-photon polymerization microfabrication of hydrogels: An advanced 3D printing technology for tissue engineering and drug delivery. Chem. Soc. Rev. 2015, 44, 5031–5039. [Google Scholar] [CrossRef] [PubMed]
- de Ávila, B.E.F.; Angsantikul, P.; Li, J.; Angel Lopez-Ramirez, M.A.; Ramírez-Herrera, D.E.; Thamphiwatana, S.; Chen, C.; Delezuk, J.; Samakapiruk, R.; Ramez, V.; et al. Micromotor-enabled active drug delivery for in vivo treatment of stomach infection. Nat. Commun. 2017, 8, 272. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Thamphiwatana, S.; Liu, W.; Esteban-Fernández de Ávila, B.; Angsantikul, P.; Sandraz, E.; Wang, J.; Xu, T.; Soto, F.; Ramez, V.; et al. Enteric micromotor can selectively position and spontaneously propel in the gastrointestinal tract. ACS Nano 2016, 10, 9536–9542. [Google Scholar] [CrossRef]
- Esteban-Fernández de Ávila, B.; Angell, C.; Soto, F.; Lopez-Ramirez, M.A.; Báez, D.F.; Xie, S.; Wang, J.; Chen, Y. Acoustically propelled nanomotors for intracellular siRNA delivery. ACS Nano 2016, 10, 4997–5005. [Google Scholar] [CrossRef] [PubMed]
- Huang, H.W.; Tibbitt, M.W.; Huang, T.Y.; Nelson, B.J. Matryoshka-inspired micro-origami capsules to enhance loading, encapsulation, and transport of drugs. Soft Robot. 2019, 6, 150–159. [Google Scholar] [CrossRef] [PubMed]
- Hortelão, A.C.; Patiño, T.; Perez-Jiménez, A.; Blanco, À.; Sánchez, S. Enzyme-powered nanobots enhance anticancer drug delivery. Adv. Funct. Mater. 2017, 28, 1705086. [Google Scholar] [CrossRef]
- Alapan, Y.; Yasa, O.; Schauer, O.; Giltinan, J.; Tabak, A.F.; Sourjik, V.; Sitti, M. Soft erythrocyte-based bacterial microswimmers for cargo delivery. Sci. Robot. 2018, 3, eaar4423. [Google Scholar] [CrossRef] [PubMed]
- Yan, X.; Zhou, Q.; Vincent, M.; Deng, Y.; Yu, J.; Xu, J.; Xu, T.; Tang, T.; Bian, L.; Wang, Y.J.; et al. Multifunctional biohybrid magnetite microrobots for imaging-guided therapy. Sci. Robot. 2017, 2, eaaq1155. [Google Scholar] [CrossRef] [PubMed]
- Xu, B.; Han, X.; Hu, Y.; Luo, Y.; Chen, C.H.; Chen, Z.; Shi, P. A remotely controlled transformable soft robot based on engineered cardiac tissue construct. Small 2019, 15, e1900006. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Pan, H.; Chen, Z.; Yin, T.; Zheng, M.; Cai, L. Twin-bioengine self-adaptive micro/nanorobots using enzyme actuation and macrophage relay for gastrointestinal inflammation therapy. Sci. Adv. 2023, 9, eadc8978. [Google Scholar] [CrossRef] [PubMed]
- Simó, C.; Serra-Casablancas, M.; Hortelao, A.C.; Di Carlo, V.; Guallar-Garrido, S.; Plaza-García, S.; Rabanal, R.M.; Ramos-Cabrer, P.; Yagüe, B.; Aguado, L.; et al. Urease-powered nanobots for radionuclide bladder cancer therapy. Nat. Nanotechnol. 2024, 19, 554–564. [Google Scholar] [CrossRef] [PubMed]
- Esteban-Fernández de Ávila, B.; Ramírez-Herrera, D.E.; Campuzano, S.; Angsantikul, P.; Zhang, L.; Wang, J. Nanomotor-enabled pH-responsive intracellular delivery of caspase-3: Toward rapid cell apoptosis. ACS Nano 2017, 11, 5367–5374. [Google Scholar] [CrossRef]
- Wei, X.; Beltrán-Gastélum, M.; Karshalev, E.; Esteban-Fernández de Ávila, B.; Zhou, J.; Ran, D.; Angsantikul, P.; Fang, R.H.; Wang, J.; Zhang, L. Biomimetic micromotor enables active delivery of antigens for oral vaccination. Nano Lett. 2019, 19, 1914–1921. [Google Scholar] [CrossRef]
- Sun, H.; Dai, Y.; Zhang, J.; Xu, J.; Jia, L.; Wang, C.; Wang, L.; Li, C.; Bai, X.; Chen, B.; et al. Magnetically Controlled Cell Robots with Immune-Enhancing Potential. In Proceedings of the 2023 IEEE/RSJ International Conference on Intelligent Robots and Systems (IROS), Detroit, MI, USA, 1–5 October 2023; IEEE Publications: Piscataway, NJ, USA, 2023; pp. 8084–8089. [Google Scholar] [CrossRef]
- Qiu, F.; 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]
- Zhao, Y.; Ye, Z.; Song, D.; Wich, D.; Gao, S.; Khirallah, J.; Xu, Q. Nanomechanical action opens endo-lysosomal compartments. Nat. Commun. 2023, 14, 6645. [Google Scholar] [CrossRef] [PubMed]
- Hansen-Bruhn, M.; de Ávila, B.E.F.; Beltrán-Gastélum, M.; Zhao, J.; Ramírez-Herrera, D.E.; Angsantikul, P.; Vesterager Gothelf, K.; Zhang, L.; Wang, J. Active intracellular delivery of a Cas9/sgRNA complex using ultrasound-propelled nanomotors. Angew. Chem. Int. Ed. Engl. 2018, 57, 2657–2661. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Li, X.; Luo, T.; Wang, R.; Liu, C.; Chen, S.; Li, D.; Yue, J.; Cheng, S.H.; Sun, D. Development of a magnetic microrobot for carrying and delivering targeted cells. Sci. Robot. 2018, 3, eaat8829. [Google Scholar] [CrossRef]
- Go, G.; Jeong, S.G.; Yoo, A.; Han, J.; Kang, B.; Kim, S.; Nguyen, K.T.; Jin, Z.; Kim, C.S.; Seo, Y.R.; et al. Human adipose-derived mesenchymal stem cell-based medical microrobot system for knee cartilage regeneration in vivo. Sci. Robot. 2020, 5, eaay6626. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.; Kim, S.; Kim, S.; Kim, J.Y.; Moon, C.; Nelson, B.J.; Choi, H. A capsule-type microrobot with pick-and-drop motion for targeted drug and cell delivery. Adv. Healthc. Mater. 2018, 7, e1700985. [Google Scholar] [CrossRef] [PubMed]
- Tang, X.; Yang, Y.; Zheng, M.; Yin, T.; Huang, G.; Lai, Z.; Zhang, B.; Chen, Z.; Xu, T.; Ma, T.; et al. Magnetic-acoustic sequentially actuated CAR T cell microrobots for precision navigation and in situ antitumor immunoactivation. Adv. Mater. 2023, 35, e2211509. [Google Scholar] [CrossRef] [PubMed]
- Sun, T.M.; Du, J.Z.; Yan, L.F.; Mao, H.Q.; Wang, J. Self-assembled biodegradable micellar nanoparticles of amphiphilic and cationic block copolymer for siRNA delivery. Biomaterials 2008, 29, 4348–4355. [Google Scholar] [CrossRef] [PubMed]
- Yu, J.; Zhou, B.; Zhang, S.; Yin, H.; Sun, L.; Pu, Y.; Zhou, B.; Sun, Y.; Li, X.; Fang, Y.; et al. Design of a self-driven probiotic-CRISPR/Cas9 nanosystem for sono-immunometabolic cancer therapy. Nat. Commun. 2022, 13, 7903. [Google Scholar] [CrossRef] [PubMed]
- Tang, J.; Guha, C.; Tomé, W.A. Biological effects induced by non-thermal ultrasound and implications for cancer therapy: A review of the current literature. Technol. Cancer Res. Treat. 2015, 14, 221–235. [Google Scholar] [CrossRef] [PubMed]
- Kubota, R.; Yamashita, Y.; Kenmotsu, T.; Yoshikawa, Y.; Yoshida, K.; Watanabe, Y.; Imanaka, T.; Yoshikawa, K. Double-strand breaks in genome-sized DNA caused by ultrasound. ChemPhysChem 2017, 18, 959–964. [Google Scholar] [CrossRef] [PubMed]
- Pfeffer, C.M.; Singh, A.T.K. Apoptosis: A target for anticancer therapy. Int. J. Mol. Sci. 2018, 19, 448. [Google Scholar] [CrossRef] [PubMed]
- Cancela, S.; Canclini, L.; Mourglia-Ettlin, G.; Hernández, P.; Merlino, A. Neuroprotective effects of novel nitrones: In vitro and in silico studies. Eur. J. Pharmacol. 2020, 871, 172926. [Google Scholar] [CrossRef] [PubMed]
- Mboge, M.Y.; Mahon, B.P.; McKenna, R.; Frost, S.C. Carbonic anhydrases: Role in pH control and cancer. Metabolites 2018, 8, 19. [Google Scholar] [CrossRef] [PubMed]
- Sutoo, S.; Maeda, T.; Suzuki, A.; Kato, Y. Adaptation to chronic acidic extracellular pH elicits a sustained increase in lung cancer cell invasion and metastasis. Clin. Exp. Metastasis 2020, 37, 133–144. [Google Scholar] [CrossRef] [PubMed]
- Martinière, A.; Gibrat, R.; Sentenac, H.; Dumont, X.; Gaillard, I.; Paris, N. Uncovering pH at both sides of the root plasma membrane interface using noninvasive imaging. Proc. Natl. Acad. Sci. USA 2018, 115, 6488–6493. [Google Scholar] [CrossRef] [PubMed]
- Vela Ramirez, J.E.; Sharpe, L.A.; Peppas, N.A. Current state and challenges in developing oral vaccines. Adv. Drug Deliv. Rev. 2017, 114, 116–131. [Google Scholar] [CrossRef] [PubMed]
- Kwon, S.G.; Kwon, Y.W.; Lee, T.W.; Park, G.T.; Kim, J.H. Recent advances in stem cell therapeutics and tissue engineering strategies. Biomater. Res. 2018, 22, 36. [Google Scholar] [CrossRef] [PubMed]
- Parmar, M.; Grealish, S.; Henchcliffe, C. The future of stem cell therapies for Parkinson disease. Nat. Rev. Neurosci. 2020, 21, 103–115. [Google Scholar] [CrossRef]
- Aguado, B.A.; Mulyasasmita, W.; Su, J.; Lampe, K.J.; Heilshorn, S.C. Improving viability of stem cells during syringe needle flow through the design of hydrogel cell carriers. Tissue Eng. Part A 2012, 18, 806–815. [Google Scholar] [CrossRef]
- Moldogazieva, N.T.; Lutsenko, S.V.; Terentiev, A.A. Reactive oxygen and nitrogen species-induced protein modifications: Implication in carcinogenesis and anticancer therapy. Cancer Res. 2018, 78, 6040–6047. [Google Scholar] [CrossRef] [PubMed]
- Weinberg, F.; Ramnath, N.; Nagrath, D. Reactive oxygen species in the tumor microenvironment: An overview. Cancers 2019, 11, 1191. [Google Scholar] [CrossRef] [PubMed]
- Hinshaw, D.C.; Shevde, L.A. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef] [PubMed]
- Le, Q.V.; Suh, J.; Oh, Y.K. Nanomaterial-based modulation of tumor microenvironments for enhancing Chemo/Immunotherapy. AAPS J. 2019, 21, 64. [Google Scholar] [CrossRef] [PubMed]
- Wu, Z.; Li, T.; Gao, W.; Xu, T.; Jurado-Sánchez, B.; Li, J.; Gao, W.; He, Q.; Zhang, L.; Wang, J. Cell-membrane-coated synthetic nanomotors for effective biodetoxification. Adv. Funct. Mater. 2015, 25, 3881–3887. [Google Scholar] [CrossRef]
- Abdelmohsen, L.K.E.A.; Nijemeisland, M.; Pawar, G.M.; Janssen, G.J.A.; Nolte, R.J.M.; van Hest, J.C.M.; Wilson, D.A. Dynamic loading and unloading of proteins in polymeric stomatocytes: Formation of an enzyme-loaded supramolecular nanomotor. ACS Nano 2016, 10, 2652–2660. [Google Scholar] [CrossRef] [PubMed]
- Villa, K.; Krejčová, L.; Novotný, F.; Heger, Z.; Sofer, Z.; Pumera, M. Cooperative multifunctional self-propelled paramagnetic microrobots with chemical handles for cell manipulation and drug delivery. Adv. Funct. Mater. 2018, 28, 1804343. [Google Scholar] [CrossRef]
- Sun, J.; Mathesh, M.; Li, W.; Wilson, D.A. Enzyme-powered nanomotors with controlled size for biomedical applications. ACS Nano 2019, 13, 10191–10200. [Google Scholar] [CrossRef]
- Wan, M.; Chen, H.; Wang, Q.; Niu, Q.; Xu, P.; Yu, Y.; Zhu, T.; Mao, C.; Shen, J. Bio-inspired nitric-oxide-driven nanomotor. Nat. Commun. 2019, 10, 966. [Google Scholar] [CrossRef] [PubMed]
- Wu, Z.; Li, L.; Yang, Y.; Hu, P.; Li, Y.; Yang, S.Y.; Wang, L.V.; Gao, W. A microrobotic system guided by photoacoustic computed tomography for targeted navigation in intestines in vivo. Sci. Robot. 2019, 4, eaax0613. [Google Scholar] [CrossRef] [PubMed]
- Schamel, D.; Mark, A.G.; Gibbs, J.G.; Miksch, C.; Morozov, K.I.; Leshansky, A.M.; Fischer, P. Nanopropellers and their actuation in complex viscoelastic media. ACS Nano 2014, 8, 8794–8801. [Google Scholar] [CrossRef] [PubMed]
- Mikuła-Pietrasik, J.; Witucka, A.; Pakuła, M.; Uruski, P.; Begier-Krasińska, B.; Niklas, A.; Tykarski, A.; Książek, K. Comprehensive review on how platinum- and taxane-based chemotherapy of ovarian cancer affects biology of normal cells. Cell Mol. Life Sci. 2019, 76, 681–697. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Medina-Sánchez, M.; Magdanz, V.; Schwarz, L.; Hebenstreit, F.; Schmidt, O.G. Sperm-hybrid micromotor for targeted drug delivery. ACS Nano 2018, 12, 327–337. [Google Scholar] [CrossRef] [PubMed]
- Shim, G.; Le, Q.V.; Suh, J.; Choi, S.; Kim, G.; Choi, H.G.; Kim, Y.B.; Macgregor, R.B., Jr.; Oh, Y.K. Sequential activation of anticancer therapy triggered by tumor microenvironment-selective imaging. J. Control. Release 2019, 298, 110–119. [Google Scholar] [CrossRef] [PubMed]
- Isaacson, K.J.; Martin Jensen, M.M.; Subrahmanyam, N.B.; Ghandehari, H. Matrix-metalloproteinases as targets for controlled delivery in cancer: An analysis of upregulation and expression. J. Control. Release 2017, 259, 62–75. [Google Scholar] [CrossRef] [PubMed]
- Ceylan, H.; Yasa, I.C.; Yasa, O.; Tabak, A.F.; Giltinan, J.; Sitti, M. 3D-printed biodegradable microswimmer for theranostic cargo delivery and release. ACS Nano 2019, 13, 3353–3362. [Google Scholar] [CrossRef] [PubMed]
- Narayanan, M.; Reddy, K.M.; Marsicano, E. Peptic Ulcer Disease and Helicobacter pylori infection. Mo. Med. 2018, 115, 219–224. [Google Scholar]
- Xu, Q.; Boylan, N.J.; Suk, J.S.; Wang, Y.Y.; Nance, E.A.; Yang, J.C.; McDonnell, P.J.; Cone, R.A.; Duh, E.J.; Hanes, J. Nanoparticle diffusion in, and microrheology of, the bovine vitreous ex vivo. J. Control. Release 2013, 167, 76–84. [Google Scholar] [CrossRef]
- Chen, J.; Howell, C.; Haller, C.A.; Patel, M.S.; Ayala, P.; Moravec, K.A.; Dai, E.; Liu, L.; Sotiri, I.; Aizenberg, M.; et al. An immobilized liquid interface prevents device associated bacterial infection in vivo. Biomaterials 2017, 113, 80–92. [Google Scholar] [CrossRef]
- Koo, H.; Moon, H.; Han, H.; Na, J.H.; Huh, M.S.; Park, J.H.; Woo, S.J.; Park, K.H.; Kwon, I.C.; Kim, K.; et al. The movement of self-assembled amphiphilic polymeric nanoparticles in the vitreous and retina after intravitreal injection. Biomaterials 2012, 33, 3485–3493. [Google Scholar] [CrossRef]
- Pardridge, W.M. Blood-brain barrier delivery. Drug Discov. Today 2007, 12, 54–61. [Google Scholar] [CrossRef] [PubMed]
- Chai, Z.; Hu, X.; Wei, X.; Zhan, C.; Lu, L.; Jiang, K.; Su, B.; Ruan, H.; Ran, D.; Fang, R.H.; et al. A facile approach to functionalizing cell membrane-coated nanoparticles with neurotoxin-derived peptide for brain-targeted drug delivery. J. Control. Release 2017, 264, 102–111. [Google Scholar] [CrossRef] [PubMed]
- Chai, Z.; Ran, D.; Lu, L.; Zhan, C.; Ruan, H.; Hu, X.; Xie, C.; Jiang, K.; Li, J.; Zhou, J.; et al. Ligand-modified cell membrane enables the targeted delivery of drug nanocrystals to glioma. ACS Nano 2019, 13, 5591–5601. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.L.; Jiang, G.; Song, Q.X.; Gu, X.; Hu, M.; Wang, X.L.; Song, H.H.; Chen, L.P.; Lin, Y.Y.; Jiang, D.; et al. Lipoprotein-biomimetic nanostructure enables efficient targeting delivery of siRNA to Ras-activated glioblastoma cells via macropinocytosis. Nat. Commun. 2017, 8, 15144. [Google Scholar] [CrossRef] [PubMed]
- Yang, T.; Fogarty, B.; LaForge, B.; Aziz, S.; Pham, T.; Lai, L.; Bai, S. Delivery of small interfering RNA to inhibit vascular endothelial growth factor in zebrafish using natural brain endothelia cell-secreted exosome nanovesicles for the treatment of brain cancer. AAPS J. 2017, 19, 475–486. [Google Scholar] [CrossRef] [PubMed]
- Tang, Y.; Han, T.; Everts, M.; Zhu, Z.B.; Gillespie, G.Y.; Curiel, D.T.; Wu, H. Directing adenovirus across the blood-brain barrier via melanotransferrin (P97) transcytosis pathway in an in vitro model. Gene Ther. 2007, 14, 523–532. [Google Scholar] [CrossRef] [PubMed]
- Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef] [PubMed]
- van de Haar, H.J.; Burgmans, S.; Jansen, J.F.A.; van Osch, M.J.P.; van Buchem, M.A.; Muller, M.; Hofman, P.A.M.; Verhey, F.R.J.; Backes, W.H. Blood-brain barrier leakage in patients with early Alzheimer disease. Radiology 2016, 281, 527–535. [Google Scholar] [CrossRef]
- Gray, M.T.; Woulfe, J.M. Striatal blood-brain barrier permeability in Parkinson’s disease. J. Cereb. Blood Flow. Metab. 2015, 35, 747–750. [Google Scholar] [CrossRef]
- Joseph, A.; Contini, C.; Cecchin, D.; Nyberg, S.; Ruiz-Perez, L.; Gaitzsch, J.; Fullstone, G.; Tian, X.; Azizi, J.; Preston, J.; et al. Chemotactic synthetic vesicles: Design and applications in blood-brain barrier crossing. Sci. Adv. 2017, 3, e1700362. [Google Scholar] [CrossRef]
- Mergenthaler, P.; Lindauer, U.; Dienel, G.A.; Meisel, A. Sugar for the brain: The role of glucose in physiological and pathological brain function. Trends Neurosci. 2013, 36, 587–597. [Google Scholar] [CrossRef] [PubMed]
- Miraz, M.; Ali, M.; Excell, P.; Picking, R. Internet of nano-things, things and everything: Future growth trends. Future Internet 2018, 10, 68. [Google Scholar] [CrossRef]
- Eo, J.; Cepeda, B.; Kim, J.; Kim, N. A new paradigm of pharmaceutical drug delivery systems (DDS): Challenges for space, time, and shapes. Innov. Pharm. 2018, 9, 1–13. [Google Scholar] [CrossRef] [PubMed]
- Sayeed, M.A.; Mohanty, S.P.; Kougianos, E.; Zaveri, H.P. An IoT-based drug delivery system for refractory epilepsy. In Proceedings of the IEEE International Conference on Consumer Electronics (ICCE), Las Vegas, NV, USA, 11–13 January 2019; IEEE Publications: Piscataway, NJ, USA, 2019; pp. 1–4. [Google Scholar] [CrossRef]
- Garcia-Gradilla, V.; Orozco, J.; Sattayasamitsathit, S.; Soto, F.; Kuralay, F.; Pourazary, A.; Katzenberg, A.; Gao, W.; Shen, Y.; Wang, J. Functionalized ultrasound-propelled magnetically guided nanomotors: Toward practical biomedical applications. ACS Nano 2013, 7, 9232–9240. [Google Scholar] [CrossRef] [PubMed]
- Dang, X.; Bardhan, N.M.; Qi, J.; Gu, L.; Eze, N.A.; Lin, C.W.; Kataria, S.; Hammond, P.T.; Belcher, A.M. Deep-tissue optical imaging of near cellular-sized features. Sci. Rep. 2019, 9, 3873. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, P.B.; Kang, B.; Bappy, D.M.; Choi, E.; Park, S.; Ko, S.Y.; Park, J.O.; Kim, C.S. Real-time microrobot posture recognition via biplane X-ray imaging system for external electromagnetic actuation. Int. J. Comput. Assist. Radiol. Surg. 2018, 13, 1843–1852. [Google Scholar] [CrossRef] [PubMed]
- Lynn, L.A. Artificial intelligence systems for complex decision-making in acute care medicine: A review. Patient Saf. Surg. 2019, 13, 6. [Google Scholar] [CrossRef] [PubMed]
- Davenport, T.; Kalakota, R. The potential for artificial intelligence in healthcare. Future Healthc. J. 2019, 6, 94–98. [Google Scholar] [CrossRef] [PubMed]
- Gao, W.; Dong, R.; Thamphiwatana, S.; Li, J.; Gao, W.; Zhang, L.; Wang, J. Artificial micromotors in the Mouse’s stomach: A step toward in vivo use of synthetic motors. ACS Nano 2015, 9, 117–123. [Google Scholar] [CrossRef]
- Lu, H.; Zhang, M.; Yang, Y.; Huang, Q.; Fukuda, T.; Wang, Z.; Shen, Y. A bioinspired multilegged soft millirobot that functions in both dry and wet conditions. Nat. Commun. 2018, 9, 3944. [Google Scholar] [CrossRef]
- Servant, A.; Qiu, F.; Mazza, M.; Kostarelos, K.; Nelson, B.J. Controlled in vivo swimming of a swarm of bacteria-like microrobotic flagella. Adv. Mater. 2015, 27, 2981–2988. [Google Scholar] [CrossRef] [PubMed]
- Kei Cheang, U.K.; Lee, K.; Julius, A.A.; Kim, M.J. Multiple-robot drug delivery strategy through coordinated teams of microswimmers. Appl. Phys. Lett. 2014, 105, 083705. [Google Scholar] [CrossRef]
- Zhao, Q.; Liang, Y.; Ren, L.; Yu, Z.; Zhang, Z.; Ren, L. Bionic intelligent hydrogel actuators with multimodal deformation and locomotion. Nano Energy 2018, 51, 621–631. [Google Scholar] [CrossRef]
Cargo Type | Therapeutics | Therapeutic Release Mechanism | Robotic DDS Type | Power Source | Targeted Disease/Purpose | Ref |
---|---|---|---|---|---|---|
Chemical drug | DOX | Active release by gas generation | Mesoporous silica nanoparticle | Urease | Cancer | [33] |
DOX | pH response | Erythrocytes-based bacterial microswimmer | Bacterial movement + magnetic field | Cancer | [34] | |
DOX | Thermal response by NIR | Floating-plane microrobot | Cardiomyocyte contraction | Cancer | [36] | |
Fluorouracil | Hyperthermia response | Helical microrobot | Magnetic field | Cancer | [20] | |
Clarithromycin | Active release by gas generation | Self-propelled microsphere | Magnesium catalysis | H. pylori infection | [29] | |
Curcumin, 5-amino salicylic acid | pH response | Self-propelled yeast particle | Glucose oxidase and catalase | Gastric ulcer and colitis | [37] | |
Iodine-131 | - | Mesoporous silica nanoparticle | Urease | Bladder cancer | [38] | |
Protein | Caspase 3 | pH response | Gold nanowire | Ultrasound | Cancer | [39] |
Staphylococcal α-toxin | pH response | Biomimetic Micromotor | Magnesium catalysis | Oral vaccine for S. aureus infection | [40] | |
Thrombin | Cargo unlock by aptamer-subtract binding | DNA origami nanorobot | Aptamer conformation | Cancer | [24] | |
Cytokine/cell | - | Macrophage-bound iron oxide nanoparticle | Magnetic field | Cancer | [41] | |
DNA/RNA | Plasmid DNA | Passive release | Artificial bacterial flagella | Magnetic field | Gene disorder diseases | [42] |
siRNA | Gold nanowire | Ultrasound | Gene disorder diseases | [30] | ||
mRNA | Passive release | Azobenzene lipid nanoparticle | UV/Vis light | Gene transfection | [43] | |
Cas9/sgRNA | Passive release | Gold nanowire | Ultrasound | Knock-down of targeted gene | [44] | |
Cell | MC3T3-E1 fibroblasts, mesenchymal stem cells | Passive release | Magnetic 3D microrobot | Magnetic field | Cell delivery | [45] |
Human adipose–derived mesenchymal stem cell | Passive release | Magnetic 3D microrobot | Magnetic field | Knee cartilage regeneration | [46] | |
Olfactory receptor neuron | Pick and drop | Capsule-type microrobot | Magnetic field | Cell delivery | [47] | |
CAR-T cell | Passive release | Magnetic beaded modified cells | Magnetic field | Cancer | [48] |
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. |
© 2024 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
Le, Q.-V.; Shim, G. Biorobotic Drug Delivery for Biomedical Applications. Molecules 2024, 29, 3663. https://doi.org/10.3390/molecules29153663
Le Q-V, Shim G. Biorobotic Drug Delivery for Biomedical Applications. Molecules. 2024; 29(15):3663. https://doi.org/10.3390/molecules29153663
Chicago/Turabian StyleLe, Quoc-Viet, and Gayong Shim. 2024. "Biorobotic Drug Delivery for Biomedical Applications" Molecules 29, no. 15: 3663. https://doi.org/10.3390/molecules29153663