AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (632.7 KB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

Research progress of microrobots in tumor drug delivery

Jin Wang1Zhi-Xin Liao2( )
Department of Nutrition, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
College of Chemistry and Chemical Engineering, Southeast University, Nanjing 210000, China
Show Author Information

Highlights

(1) The arrival of the intelligent era and the iteration of robotics technology have focused medical means from the macro to the micro and nano scale.

(2) Tumor drug therapy mainly relies on micro-nano robots to realize drug load encapsulation and path editing. It creates a new chemotherapy mode of "identification - penetration - reversal - elimination".

(3) The relevant researches are mostly in the mechanism basis and in vitro or animal test stage, there are still challenges in the clinical transformation and practical application of micro-nanorobots.

Graphical Abstract

With its cellular/subcellular size, microrobots expand a new vision for exploring drug delivery. It has unique advantages in achieving targeted delivery and precise recruitment of chemotherapy drugs. This paper reviews the relevant research at home and abroad in recent years. It searches the relevant literature in the past 10 years with the keywords "tumor drugs" and "micro-nano robots". The results obtained were described according to "research progress of tumor therapeutic drugs", "nano drug delivery system" and "microrobots drug delivery". Then focused the main discussion on "microrobots and cancer drug delivery". Through this research, its micro-size and multi-source drive provides microrobots technical basis. As the result, they can achieve in-vivo motion efficiency, targeted path planning, biological barrier penetration and efficient drug delivery. It might be beneficial for microrobots to earn better therapeutic of tumor drugs, while decreasing side-effects to other tissues and organs in the future.

Abstract

With its cellular/subcellular size, microrobots expand a new vision for exploring drug delivery. It has unique advantages in achieving targeted delivery and precise recruitment of chemotherapy drugs. This paper reviews the relevant research at home and abroad in recent years. It searches the relevant literature in the past 10 years with the keywords “tumor drugs” and “micro-nano robots”. The results obtained were described according to “research progress of tumor therapeutic drugs”, “nano drug delivery system” and “microrobots drug delivery”. Then focused the main discussion on “microrobots and cancer drug delivery”. Through this research, its micro-size and multi-source drive provides microrobots technical basis. As the result, they can achieve in-vivo motion efficiency, targeted path planning, biological barrier penetration and efficient drug delivery. It might be beneficial for microrobots to earn better therapeutic of tumor drugs, while decreasing side-effects to other tissues and organs in the future.

References

[1]

Li, M. Y., Yang, J., Jiao, N. D., et al. Review of the latest research progress of micro and nano robots. Robot, 2022, 6: 732–749. https://doi.org/10.13973/j.cnki.robot.210377

[2]

Wang, J. J., Dong, R. F., Wu, H. Y., et al. A review on artificial micro/nanomotors for cancer-targeted delivery, diagnosis, and therapy. Nano-Micro Letters, 2019, 12: 1–19. https://doi.org/10.1007/s40820-019-0350-5

[3]

Luo, M., Feng, Y. Z., Wang, T. W., et al. Micro-/nanorobots at work in active drug delivery. Advanced Functional Materials, 2018, 28: 1706100. https://doi.org/10.1002/adfm.20176100

[4]

Liu, M. H., Tu, B. B., Liu, L., et al. Application progress of self-driven micro/nanomotor in active drug delivery. Journal of Southern Medical University, 2020, 40: 445–452. https://doi.org/10.12122/j.issn.1673-4254.2020.03.25

[5]

Ding, J., Venkatesan, R., Zhai, Z. H., et al. Micro- and nanoparticles-based immunoregulation of macrophages for tissue repair and regeneration. Colloids and Surfaces B: Biointerfaces, 2020, 192: 111075. https://doi.org/10.1016/j.colsurfb.2020.111075

[6]

Wei, F. N., Zhong, T. L., Zhan, Z. H., et al. Self-assembled micronanorobots: from assembly mechanisms to applications. Chemnanomat, 2021, 7: 238–252. https://doi.org/10.1002/cnma.202000608

[7]

Jurado-Sanchez, B., Wang, J. Micromotors for environmental applications: a review. Environmental Science: Nano, 2018, 5: 1530–1544. https://doi.org/10.1039/c8en00299a

[8]

Thirunavukkarasu, A., Nithya, R., Sivashankar, R. A review on the role of nanomaterials in the removal of organic pollutants from wastewater. Reviews in Environmental Science and Bio/Technology, 2020, 19: 751–778. https://doi.org/10.1007/s11157-020-09548-8

[9]

Zhang, L., Abbott, J. J., Dong, L. X., et al. Artificial bacterial flagella: fabrication and magnetic control. Applied Physics Letters, 2009, 94: 64107. https://doi.org/10.1063/1.3079655

[10]

Li, F., Fu, X., Huo, Q., et al. Research progress on the nano-delivery systems of antitumor drugs. Nano Life, 2020, 10: 7. https://doi.org/10.1142/S1793984420400061

[11]

Zhu, B., Chen, S. Y., Song, M. M. Research progress of nano-drug delivery system in chemotherapy-based combination therapy. Life Science, 2023, 35: 437–446. https://doi.org/10.13376/j.cbls/2023052

[12]

Zhong, H. Y., Li, L., Liu, C. M., et al. Cancer-related anorexia and its related factors in patients on chemotherapy. Chinese Journal of Nursing, 2023, 38: 1–4.

[13]

Zhong, Y., Zhang, J., Zhang, J., et al. Tumor microenvironment-activatable nanoenzymes for mechanical remodeling of extracellular matrix and enhanced tumor chemotherapy. Advanced Functional Materials, 2020, 31: 2007544. https://doi.org/10.1002/adfm.202007544

[14]

Miller, A. L., Garcia, P. L., Yoon, K. J. Developing effective combination therapy for pancreatic cancer: an overview. Pharmacological Research, 2020, 155: 104740. https://doi.org/10.1016/j.phrs.2020.104740

[15]

Bholakant, R., Dong, B., Zhou, X., et al. Multi-functional polymeric micelles for chemotherapy-based combined cancer therapy. Journal of Materials Chemistry. B, 2021, 9: 8718–8338. https://doi.org/10.1039/D1TB01771C

[16]

Aboeleneen, S. B., Scully, M. A., Harris, J. C., et al. Membrane-wrapped nanoparticles for photothermal cancer therapy. Nano Convergence, 2022, 9: 37. https://doi.org/10.1186/s40580-022-00328-4

[17]

Zhou, Y., Ren, X., Hou, Z., et al. Engineering a photosensitizer nanoplatform for amplified photodynamic immunotherapy via tumor microenvironment modulation. Nanoscale Horizons, 2021, 6: 120–131. https://doi.org/10.1039/D0NH00480D

[18]

Salvador-Morales, C., Grodzinski, P. Nanotechnology tools enabling biological discovery. ACS Nano, 2022, 16: 5062–5084. https://doi.org/10.1021/acsnano.1c10635

[19]

Zheng, N., Wang, Q., Zhang, S., et al. Recent advances in nanotechnology mediated mitochondria-targeted imaging. Journal of Materials Chemistry. B., 2022, 10: 7450–7459. https://doi.org/10.1039/d2t00935h

[20]

Mitchell, M. J., Billingsley, M. M., Haley, R. M., et al. Engineering precision nanoparticles for drug delivery. Nature Reviews Drug Discovery, 2021, 20: 101–124. https://doi.org/10.1038/s41573-020-0090-8

[21]

Jain, P., Kathuria, H., Momin, M. Clinical therapies and nano drug delivery systems for urinary bladder cancer. Pharmacology Therapeutics, 2021, 226: 107871. https://doi.org/10.1016/j.pharmthera.2021.107871

[22]

Prasher, P., Sharma, M., Singh, S. P. Drug encapsulating polysaccharide-loaded metal nanoparticles: a perspective drug delivery system. Drug Development Research, 2020, 82: 145–148. https://doi.org/10.1002/ddr.21754

[23]

Zhang, R. X., Wong, H. L., Xue, H. Y., et al. Nanomedicine of synergistic drug combinations for cancer therapy-strategies and perspectives. Journal of Controlled Release: Official Journal of Controlled Release Society, 2016, 240: 489–503. https://doi.org/10.1016/j.jconrel.2016.06.012

[24]

Ali, E. S., Sharker, S. M., Islam, M. T., et al. Targeting cancer cells with nanotherapeutics and nanodiagnostics: current status and future perspectives. Seminars in Cancer Biology, 2021, 69: 52–68. https://doi.org/10.1016/j.semcancer.2020.01.011

[25]

Cui, G., Su, W., Tan, M. Formation and biological effects of protein corona for food-related nanoparticles. Comprehensive Reviews in Food Science and Food Safety, 2022, 21: 2002–2031. https://doi.org/10.1111/1541-4337.12838

[26]

Pang, S. Y., Yan, X. H. Research progress of micro/nano robots in tumor therapy. Micro/Nano Electronics, 2023, 60: 811–819. https://doi.org/10.13250/j.cnki.wndz.2023.06.001

[27]

Liu, G. Y., Hou, S. L., Tong, P. H., et al. Liposomes: preparation, characteristics, and application strategies in analytical chemistry. Critical Reviews in Analytical Chemistry, 2022, 52: 392–412. https://doi.org/10.1080/10408347.2020.1805293

[28]

Francian, A., Widmer, A., Olsson, T., et al. Delivery of toll like receptor agonists by complement C3-targeted liposomes activates immune cells and reduces tumor growth. Journal of Drug Targeting, 2021, 29: 754–760. https://doi.org/10.1080/1061186X.2021.1878364

[29]

Park, S. J. Protein-nanoparticle interaction: corona formation and conformational changes in proteins on nanoparticles. International Journal of Nanomedicine, 2020, 15: 5783–5802. https://doi.org/10.2147/IJN.S254808

[30]

Yu, B., Goel, S., Ni, D., et al. Reassembly of 89Zr-labeled cancer cell membranes into multicompartment membrane-derived liposomes for PET-trackable tumor-targeted theragnostic. Advanced Materials, 2018, 30: 1704934. https://doi.org/10.1002/adma.201704934

[31]

Paris, A., Tardif, N., Galibert, M. D., et al. AhR and cancer: from gene profiling to targeted therapy. International Journal of Molecular Sciences, 2021, 22: 752. https://doi.org/10.3390/ijms22020752

[32]

Ogueri, K. S., Ogueri, K. S., Ude, C. C., et al. Biomedical applications of polyphosphazenes. Medical Devices & Sensors, 2020, 3: e10113. https://doi.org/10.1002/mds3.10113

[33]

Xia, W., Tao, Z., Zhu, B., et al. Targeted delivery of drugs and genes using polymer nanocarriers for cancer therapy. Multidisciplinary Digital Publishing Institute, 2021, 22: 9118. https://doi.org/10.3390/IJMS22179118

[34]

Yang, Y., He, J., Li, Q., et al. Self-healing of electrical damage in polymers using superparamagnetic nanoparticles. Nature Nanotechnology, 2019, 14: 151–155. https://doi.org/10.1038/s41565-018-0327-4

[35]

Zhang, H. Y., Li, Z. S., Gao, C. Y., et al. Dual-responsive biohybrid neutrobots for active target delivery. Science Robotics, 2021, 6: eaaz9519. https://doi.org/10.1126/scirobotics.aaz9519

[36]

Liu, Y., Shen, J., Shi, J., et al. Functional polymeric core–shell hybrid nanoparticles overcome intestinal barriers and inhibit breast cancer metastasis. Chemical Engineering Journal, 2022, 427: 131742. https://doi.org/10.1016/j.cej.2021.131742

[37]

Huang, Z., Wang, Y., Yao, D., et al. Nanoscale coordination polymers induce immunogenic cell death by amplifying radiation therapy mediated oxidative stress. Nature Communications, 2021, 12: 145. https://doi.org/10.1038/s41467-020-20243-8

[38]

Chen, H. Y., Deng, J., Wang, Y., et al. Hybrid cell membranecoated nanoparticles: a multifunctional biomimetic platform for cancer diagnosis and therapy. Acta Biomaterialia, 2020, 112: 1–13. https://doi.org/10.1016/j.actbio.2020.05.028

[39]

Oroojalian, F., Beygi, M., Baradaran, B., et al. Immune cell membrane-coated biomimetic nanoparticles for targeted cancer therapy. Small, 2021, 17: e2006484. https://doi.org/10.1002/smll.202006484

[40]

Zhao, P., Wang, M., Chen, M., et al. Programming cell pyroptosis with biomimetic nanoparticles for solid tumor immunotherapy. Biomaterials, 2020, 254: 120142. https://doi.org/10.1016/j.biomaterials.2020.120142

[41]

Zhang, M., Cheng, S., Jin, Y., et al. Membrane engineering of cell membrane biomimetic nanoparticles for nanoscale therapeutics. Clinical and Translational Medicine, 2021, 11: e292. https://doi.org/ 10.1002/ctm2.292

[42]

Chen, Y. D., Pan, R. J., Wang, Y., et al. Carbon helical nanorobots capable of cell membrane penetration for single cell targeted SERS bio-sensing and photothermal cancer therapy. Advanced Functional Materials, 2022, 32: 1–10. https://doi.org/10.1002/adfm.202200600

[43]

Xiao, Z. Y., Duan, S. F., Xu, P. Z., et al. Synergistic speed enhancement of an electric photochemical hybrid micromotor by tilt rectification. ACS Nano, 2020, 14: 8658–8667. https://doi.org/10.1021/acsnano.0c03022

[44]

Avila, D., Esteban-Fernandez, B., Angell, C., et al. Acoustically propelled nanomotors for intracellular siRNA delivery. ACS Nano, 2016, 10: 4997–5005. https://doi.org/10.1021/acsnano.6b01415

[45]

Xu, T., Soto, F., Gao, W., et al. Ultrasound-modulated bubble propulsion of chemically powered microengines. Journal of the American Chemical Society, 2014, 136: 8552–8555. https://doi.org/10.1021/ja504150e

[46]

Zou, X. B., Zheng, Q., Wu, D., et al. Controllable cellular micromotors based on optical tweezers. Advanced Functional Materials, 2020, 30: 1–10. https://doi.org/10.1002/adfm.202070181

[47]

Meng, F. L., Matsunaga, D., Golestanian, R. Clustering of magnetic swimmers in a poiseuille flow. Physical Review Letters, 2018, 120: 188101. https://doi.org/10.1103/PhysRevLett.120.188101

[48]
Oulmas, A., Andreff, N., Regnier, S. 3D closed-loop swimming at low Reynolds numbers. International Journal of Robotics Research, 2018 , 37: 1359–1375. https://doi.org/10.1177/0278364918801502
[49]

Qiu, F., Fujita, S., Mhanna, R., et al. Magnetic helical micro-swimmers functionalized with lipoplexes for targeted gene delivery. Advanced Functional Materials, 2015, 25: 1666–1671. https://doi.org/10.1002/adfm.201403891

[50]

Su, Y., Qiu, T., Song, W., et al. Melt electrospinning writing of magnetic microrobots. Advanced Science, 2021, 8: 2003177. https://doi.org/10.1002/advs.202003177

[51]

Yu, J., Wang, B., Du, X., et al. Ultra-extensible ribbon-like magnetic micro-swarm. Nature Communications, 2018, 9: 3260. https://doi.org/10.1038/s41467-018-05749-6

[52]

Vilfan, M., Osterman, N., Vilfan, A. Magnetically driven omnidirectional artificial microswimmers. Soft Matter, 2018, 14: 3415–3422. https://doi.org/10.1039/C8SM00230D

[53]

Ghosh, S., Ghosh, A. Mobile nanotweezers for active colloidal manipulation. Science Robotics, 2018, 3: eaaq0076. https://doi.org/10.1126/sciroboticsaaq0076

[54]

Patil, G., Ghosh, A. Anomalous behavior of highly active helical swimmers. Frontiers in Physics, 2021, 8: 628276. https://doi.org/10.3389/fphy.2020.628276

[55]

Wang, J., Toebes, B. J., Plachokova, A. S., et al. Self-propelled PLGA micromotor with chemotactic response to inflammation. Advanced Healthcare Materials, 2020, 9: e1901710. https://doi.org/10.1002/adhm.201901710

[56]

Schwarz, L., Karnaushenko, D. D., Hebenstreit, F., et al. A rotating spiral micromotor for noninvasive zygote transfer. Advanced Science, 2020, 7: 2000843. https://doi.org/10.1002/advs.202000843

[57]
Ahmed, S., Wang, W., Bai, L., et al. Density and shape effects in the acoustic propulsion of bimetallic nanorod motors. ACS Nano, 2016, 10: 4763–4769. https://doi.org/10.1021/acsnano.6b01344
[58]

Alsaadawi, Y., Eichler-Volf, A., Heigl, M., et al. Control over self-assembled Janus clusters by the strength of magnetic field in H2O2. The European Physical Journal E, 2021, 44: 1–8. https://doi.org/10.1140/epje/s10189-021-00010-3

[59]

Qiu, F. Magnetic helical micro- and nanorobots: toward their biomedical applications. Engineering, 2015, 1: 21–26. https://doi.org/10.15302/J-ENG-2015005

[60]

Yang, L., Chen, X., Wang, L., et al. Targeted single-cell therapeutics with magnetic tubular micromotor by one-step exposure of structured femtosecond optical vortices. Advanced Functional Materials, 2019, 29: 1905745. https://doi.org/10.1002/adfm.201905745

[61]

Alapan, Y., Bozuyuk, U., Erkoc, P., et al. Multifunctional surface micro-rollers for targeted cargo delivery in physiological blood flow. Science Robotics, 2020, 5: eaba5726. https://doi.org/10.1126/scirobotics.aba5726

[62]

Zhong, D., Li, W., Qi, Y., et al. Photosynthetic biohybrid nano-swimmers system to alleviate tumor hypoxia for FL/PA/MR imaging-guided enhanced radio-photodynamic synergetic therapy. Advanced Functional Materials, 2020, 30: 1910395. https://doi.org/10.1002/adfm.201910395

[63]

Yan, X., Zhou, Q., Vincent, M., et al. Multifunctional biohybrid magnetite microrobots for imaging-guided therapy. Science Robotics, 2017, 2: eaaq1155. https://doi.org/10.1126/scirobotics.aaq1155

[64]

Yu, J., Jin, D., Chan, K., et al. Active generation and magnetic actuation of microrobotic swarms in bio-fluids. Nature Communications, 2019, 10: 5631. https://doi.org/10.1038/s41467-019-13576-6

[65]

Yu, J., Xu, T., Lu, Z., et al. On-demand disassembly of paramagnetic nanoparticle chains for micro-robotic cargo delivery. IEEE Transactions on Robotics, 2017, 33: 1213–1225. https://doi.org/10.1109/TRO.2017.2693999

[66]

Ji, F., Li, T., Yu, S., et al. Propulsion gait analysis and fluidic trapping of swinging flexible nanomotors. ACS Nano, 2021, 15: 5118–5128. https://doi.org/10.1021/acsnano.0c10269

[67]

Gao, W., Sattayasamitsathit, S., Orozco, J., et al. Highly efficient catalytic microengines: template electrosynthesis of polyaniline/platinum microtubes. Journal of the American Chemical Society, 2011, 133: 11862–11864. https://doi.org/10.1021/ ja203773g

[68]

Kline, T. R., Paxton, W. F., Mallouk, T. E., et al. Catalytic nanomotors: autonomous movement of striped nanorods. Cheminform, 2005, 117: 744–746. https://doi.org/10.1002/ange.200461890

[69]

Zhang, Y., Hess, H. Chemically-powered swimming and diffusion in the microscopic world. Nature Reviews Chemistry, 2021, 5: 500–510. https://doi.org/10.1038/s41570-021-00281-6

[70]

Naeem, S., Naeem, F., Mujtaba, J., et al. Oxygen generation using catalytic nano/micromotors. Micromachines, 2021, 12: 1251. https://doi.org/10.3390/mi12101251

[71]

Ma, X. F., Shi, H. J., Gu, J. L., et al. Temperature effects on low-cycle fatigue behavior of nickel-based single crystalline superalloy. Acta Mechanica Solida Sinica, 2008, 21: 289–297. https://doi.org/10.1007/s10338-008-0833-2

[72]

Rao, D. V., Reddy, N., Fransaer, J., et al. Self-propulsion of bent bimetallic Janus rods. Journal of Physics D: Applied Physics, 2019, 52: aae6f6. https://doi.org/10.1088/1361-6463/aae6f6

[73]

Zhou, C., Gao, C. Y., Lin, Z. H., et al. Autonomous motion of bubble-powered carbonaceous nanoflask motors. Langmuir, 2020, 36: 7039–7045. https://doi.org/10.1021/acs.langmuir.9b03398

[74]

Ye, H., Ma, G., Kang, J., et al. PT-free microengines at extremely low peroxide levels. Chemical Communications, 2018, 54: 4653–4656. https://doi.org/10.1039/C8CC01548A

[75]

Wei, G., Ren, F. D., Soracha, T., et al. Artificial micromotors in the mouse’s stomach: a step toward in vivo use of synthetic motors. ACS Nano, 2015, 9: 117–123. https://doi.org/10.1021/nn507097k

[76]

Chen, W., Huang, T., Shi, K., et al. Chemotaxis-based self-accumulation of surface-engineered mitochondria for cancer therapeutic improvement. Science Direct, 2020, 35: 100966. https://doi.org/10.1016/J.NANTOD.2020.100966

[77]

Zhang, Y. H., Qiu, W. X., Zhang, M. K., et al. MnO2 motor: a prospective cancer-starving therapy promoter. ACS Applied Materials & Interfaces, 2018, 10: 15030–15039. https://doi.org/10.1021/acsami.8b01818

[78]

Luo, M., Li, S., Wan, J., et al. Enhanced propulsion of urease-powered micromotors by multilayered assembly of ureases on Janus magnetic microparticles. Langmuir, 2020, 36: 7005–7013. https://doi.org/10.1021/acs.langmuir.9b03315

[79]

Hortelão, C. A., García-Jimeno, S., Cano-Sarabia, M., et al. LipoBots: using liposomal vesicles as protective shell of urease-based nanomotors. Advanced Functional Materials, 2020, 30: 2002767. https://doi.org/10.1002/adfm.202002767

[80]

Arqué, X., Andrés, X., Mestre, R., et al. Ionic species affect the self-propulsion of urease-powered micromotors. Research, 2020, 2020: 1–14. https://doi.org/10.34133/2020/2424972

[81]

Maria-Hormigos, R., Jurado-Sánchez, B., Escarpa, A. Surfactant-free β-galactosidase micromotors for “on-the-move” lactose hydrolysis. Advanced Functional Materials, 2018, 28: 1704256. https://doi.org/10.1002/adfm.201704256

[82]

Wang, L., Hortelao, A. C., Huang, X., et al. Lipase-powered mesoporous silica nanomotors for triglyceride degradation. Angewandte Chemie, 2019, 58: 7992–7996. https://doi.org/10.1002/anie.201900697

[83]

Schwarz, L., Medina-Sanchez, M., Schmidt, O. G. Sperm-hybrid micromotors: on-board assistance for nature’s bustling swimmers. Reproduction, 2019, 159: R83–R96. https://doi.org/10.1530/REP-19-0096

[84]

Li, Q. L., Chen, H. T., Feng, X. Y., et al. Nanoparticle-regulated semiartificial magnetotactic bacteria with tunable magnetic moment and magnetic sensitivity. Small, 2019, 15: 1970077. https://doi.org/10.1002/smll.201900427

[85]

Chen, C., Song, T., Yang, C., et al. Dynamic analysis of magnetotaxis bacteria and construction of bacterial robot. Robot, 2015, 5: 40–46. https://doi.org/10.13973/j.cnki.robot.2015.0588

[86]

Akolpoglu, M. B., Alapan, Y., Dogan, N. O., et al. Magnetically steerable bacterial microrobots moving in 3D biological matrices for stimuli-responsive cargo delivery. Science Advances, 2022, 8: 1–14. https://doi.org/10.1126/sciadv.abo6163

[87]

Buss, N., Yasa, O., Alapan, Y., et al. Nano-erythrosome functionalized biohybrid micro swimmers. APL Bioengineering, 2020, 4: 1–7. https://doi.org/10.1063/1.5130670

[88]

Shchelik, I. S., Sieber, S., Gademann, K. Green algae as a drug delivery system for the controlled release of antibiotics. Chemistry–A European Journal, 2020, 26: 16644–16648. https://doi.org/10.1002/chem.202003821

[89]

Akolpoglu, M. B., Dogan, N. O., Bozuyuk, U., et al. High-yield production of biohybrid microalgae for on-demand cargo delivery. Advanced Science, 2020, 7: 1–10. https://doi.org/10.1002/advs.202001256

[90]

Yoon, J., Eyster, T. W., Mishra, A. C., et al. Cardiomyocyte-driven actuation in biohybrid microcylinders. Advanced Materials, 2015, 27: 4509–4515. https://doi.org/10.1002/adma.201501284

[91]

Park, B. W., Zhuang, J., Yasa, O., et al. Multifunctional bacteria-driven micro-swimmers for targeted active drug delivery. ACS Nano, 2017, 11: 8910–8923. https://doi.org/10.1021/acsnano.7b03207

[92]

Vincenti, B., Ramos, G., Cordero, M. L., et al. Magnetotactic bacteria in a droplet self-assemble into a rotary motor. Nature Communications, 2019, 10: 5082. https://doi.org/10.1038/s41467-019-13031-6

[93]

Xu, H. F., Medina-Sánchez, M., Zhang, W. H., et al. Human spermbots for patient-representative 3D ovarian cancer cell treatment. Nanoscale, 2020, 12: 20467–20481. https://doi.org/10.1039/D0NR04488A

[94]

Gu, H., Hanedan, E., Boehler, Q., et al. Artificial microtubules for rapid and collective transport of magnetic microcargoes. Nature Machine Intelligence, 2022, 4: 678–684. https://doi.org/10.1038/s42256-022-00510-7

[95]

Meng, X., Xu, Y. R., Lu, Q. B., et al. Ultrasound-responsive alkaline nanorobots for the treatment of lactic acidosis-mediated doxorubicin resistance. Nanoscale, 2020, 12: 13801–13810. https://doi.org/10.1039/D0NR03726E

[96]

Chen, W., Jiang, R., Sun, X., et al. Self-fueled Janus nanomotors as active drug carriers for propulsion behavior-reinforced permeability and accumulation at the tumor site. Chemistry of Materials: A Publication of the American Chemistry Society, 2022, 34: 7543–7552. https://doi.org/10.1021/acs.chemmater.2c01671

[97]

Gwisai, T., Mirkhani, N., Christiansen, M. G., et al. Magnetic torque-driven living microrobots for increased tumor infiltration. Science Robotics, 2022, 7: eabo0665. https://doi.org/10.1126/scirobotics.abo0665

[98]

Park, J., Jin, C., Lee, S., et al. Magnetically actuated degradable microrobots for actively controlled drug release and hyperthermia therapy. Advanced Healthcare Materials, 2019, 8: 1900213. https://doi.org/10.1002/adhm.201900213

[99]

Wu, H. L., Shi, X. T. Application progress of micro/nano robots in biomedicine. Integration Technology, 2022, 10: 78–92. https://doi.org/10.12146/j.issn.2095-3135.20210310002

[100]

Li, X., Lovell, J., Yoon, J., et al. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nature Reviews Clinical Oncology, 2020, 17: 657–674. https://doi.org/10.1038/s41571-020-0410-2

[101]

Wan, M. M., Chen, H., Wang, Z. D., et al. Nitric oxide-driven nanomotor for deep tissue penetration and multidrug resistance reversal in cancer therap. Advanced Science, 2021, 8: 1–12. https://doi.org/10.1002/advs.202002525

[102]

Xu, L., Zhang, K., Ma, X., et al. Boosting cisplatin chemotherapy by nanomotor-enhanced tumor penetration and DNA adducts formation. Journal of Nanobiotechnology, 2022, 20: 1–18. https://doi.org/10.1186/s12951-022-01622-3

Food & Medicine Homology
Article number: 9420025
Cite this article:
Wang J, Liao Z-X. Research progress of microrobots in tumor drug delivery. Food & Medicine Homology, 2024, 1(2): 9420025. https://doi.org/10.26599/FMH.2024.9420025

394

Views

82

Downloads

0

Crossref

Altmetrics

Received: 13 May 2024
Revised: 10 July 2024
Accepted: 10 July 2024
Published: 17 August 2024
© National R & D Center for Edible Fungus Processing Technology 2024. Published by Tsinghua University Press.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return