Journal Home > Volume 16 , Issue 2

In recent years, the deep integration of basic research and clinical translational research of nanotechnology and oncology has led to the emergence of a new branch, namely integrated nano-oncology. This is an emerging and important interdisciplinary field, which plays an irreplaceable role in the diagnosis, treatment, early warning, monitoring and prevention of tumors, and has become a new interdisciplinary frontier. Here main advances of integrated nano-oncology was reviewed, mainly included controlled preparation of nanomaterials, ultra-sensitive detection of tumor biomarkers, multi-functional nanoimaging probes and integrated diagnosis and treatment technology, innovative nano drugs and nano drug delivery system, DNA nanotechnology, RNA nanotechnology, nano self-assembly technology, nanosensors, intelligent nanorobots, nanotherapeutic machines. The terms, concepts, trends and challenges are also discussed with the aim of promoting the application of nanotechnology in integrated oncology and solving the scientific and key technical problems in basic and clinical translational research of cancer.


menu
Abstract
Full text
Outline
About this article

Advances and Prospects in Integrated Nano-oncology

Show Author's information Jinlei Jiang1,4Xinyuan Cui2Yixin Huang1,4Dongmei Yan3Bensong Wang1Ziyang Yang1,4Mingrui Chen1Junhao Wang1Yuna Zhang1Guan Liu1Cheng Zhou1,4Shengsheng Cui1,4Jian Ni1,4Fuhua Yang2( )Daxiang Cui1,4,5( )
Institute of Nano Biomedicine and Engineering, School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
Lianyungang Maternal and Child Health Hospital, Lianyungang 222000, China
National Engineering Research Center for Nanotechnology, Shanghai 200241, China
School of Medicine, Henan University, Kaifeng 475004, China

Abstract

In recent years, the deep integration of basic research and clinical translational research of nanotechnology and oncology has led to the emergence of a new branch, namely integrated nano-oncology. This is an emerging and important interdisciplinary field, which plays an irreplaceable role in the diagnosis, treatment, early warning, monitoring and prevention of tumors, and has become a new interdisciplinary frontier. Here main advances of integrated nano-oncology was reviewed, mainly included controlled preparation of nanomaterials, ultra-sensitive detection of tumor biomarkers, multi-functional nanoimaging probes and integrated diagnosis and treatment technology, innovative nano drugs and nano drug delivery system, DNA nanotechnology, RNA nanotechnology, nano self-assembly technology, nanosensors, intelligent nanorobots, nanotherapeutic machines. The terms, concepts, trends and challenges are also discussed with the aim of promoting the application of nanotechnology in integrated oncology and solving the scientific and key technical problems in basic and clinical translational research of cancer.

Keywords: nanomaterials, nanoprobes, molecular imaging, targeted therapy, nano drug delivery system, integrated nano-oncology, nano diagnosis

References(152)

[1]

D.M. Fan. Holistic integrative medicine: Toward a new era of medical advancement. Frontiers of Medicine, 2017, 11(1): 152−159. https://doi.org/10.1007/s11684-017-0499-6

[2]

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

[3]

J. Kudr, Y. Haddad, L. Richtera, et al. Magnetic nanoparticles: From design and synthesis to real world applications. Nanomaterials, 2017, 7(9): 243. https://doi.org/10.3390/nano7090243

[4]

T. Kang, F.Y. Li, S. Baik, et al. Surface design of magnetic nanoparticles for stimuli-responsive cancer imaging and therapy. Biomaterials, 2017, 136: 98−114. https://doi.org/10.1016/j.biomaterials.2017.05.013

[5]

K. Sztandera, M. Gorzkiewicz, B. Klajnert-Maculewicz. Gold nanoparticles in cancer treatment. Molecular Pharmaceutics, 2019, 16(1): 1−23. https://doi.org/10.1021/acs.molpharmaceut.8b00810

[6]

N. Elahi, M. Kamali, M.H. Baghersad. Recent biomedical applications of gold nanoparticles: A review. Talanta, 2018, 184: 537−556. https://doi.org/10.1016/j.talanta.2018.02.088

[7]

Y. Volkov. Quantum dots in nanomedicine: Recent trends, advances and unresolved issues. Biochemical and Biophysical Research Communications, 2015, 468(3): 419−427. https://doi.org/10.1016/j.bbrc.2015.07.039

[8]

K.J. McHugh, L.H. Jing, A.M. Behrens, et al. Biocompatible semiconductor quantum dots as cancer imaging agents. Advanced Materials, 2018, 30(18): e1706356. https://doi.org/10.1002/adma.201706356

[9]

D. Maiti, X.M. Tong, X.Z. Mou, et al. Carbon-based nanomaterials for biomedical applications: A recent study. Frontiers in Pharmacology, 2018, 9: 1401. https://doi.org/10.3389/fphar.2018.01401

[10]

F. Farjadian, A. Roointan, S. Mohammadi-Samani, et al. Mesoporous silica nanoparticles: Synthesis, pharmaceutical applications, biodistribution, and biosafety assessment. Chemical Engineering Journal, 2019, 359: 684−705. https://doi.org/10.1016/j.cej.2018.11.156

[11]

M. Manzano, M. Vallet-Regí. Mesoporous silica nanoparticles for drug delivery. Advanced Functional Materials, 2020, 30(2): 1902634. https://doi.org/10.1002/adfm.201902634

[12]

A. Gao, X.L. Hu, M. Saeed, et al. Overview of recent advances in liposomal nanoparticle-based cancer immunotherapy. Acta Pharmacologica Sinica, 2019, 40(9): 1129−1137. https://doi.org/10.1038/s41401-019-0281-1

[13]

Y. Panahi, M. Farshbaf, M. Mohammadhosseini, et al. Recent advances on liposomal nanoparticles: Synthesis, characterization and biomedical applications. Artificial Cells,Nanomedicine,and Biotechnology, 2017, 45(4): 788−799. https://doi.org/10.1080/21691401.2017.1282496

[14]

Y.Z. Shen, W.T. Li. HA/HSA co-modified erlotinib-albumin nanoparticles for lung cancer treatment. Drug Design,Development and Therapy, 2018, 12: 2285−2292. https://doi.org/10.2147/DDDT.S169734

[15]

R. Solanki, H. Rostamabadi, S. Patel, et al. Anticancer nano-delivery systems based on bovine serum albumin nanoparticles: A critical review. International Journal of Biological Macromolecules, 2021, 193: 528−540. https://doi.org/10.1016/j.ijbiomac.2021.10.040

[16]

S.H. Wen, J.J. Zhou, K.Z. Zheng, et al. Advances in highly doped upconversion nanoparticles. Nature Communications, 2018, 9: 2415. https://doi.org/10.1038/s41467-018-04813-5

[17]

S. Wilhelm. Perspectives for upconverting nanoparticles. ACS Nano, 2017, 11(11): 10644−10653. https://doi.org/10.1021/acsnano.7b07120

[18]

J. Chao, H.L. Zhang, Y.K. Xing, et al. Programming DNA origami assembly for shape-resolved nanomechanical imaging labels. Nature Protocols, 2018, 13(7): 1569−1585. https://doi.org/10.1038/s41596-018-0004-y

[19]

Y.S. Chen, S.L. Cheng, A.M. Zhang, et al. Salivary analysis based on surface enhanced Raman scattering sensors distinguishes early and advanced gastric cancer patients from healthy persons. Journal of Biomedical Nanotechnology, 2018, 14(10): 1773−1784. https://doi.org/10.1166/jbn.2018.2621

[20]

M.A. Aslam, C.L. Xue, K. Wang, et al. SVM based classification and prediction system for gastric cancer using dominant features of saliva. Nano Biomedicine and Engineering, 2019, 12(1): 1−13. https://doi.org/10.5101/nbe.v12i1.p1-13

[21]

D.P. Yang, S.H. Chen, P. Huang, et al. Bacteria-template synthesized silver microspheres with hollow and porous structures as excellent SERS substrate. Green Chemistry, 2010, 12(11): 2038−2042. https://doi.org/10.1039/C0GC00431F

[22]

X.C. Yu, L. He, M. Pentok, et al. An aptamer-based new method for competitive fluorescence detection of exosomes. Nanoscale, 2019, 11(33): 15589−15595. https://doi.org/10.1039/C9NR04050A

[23]

R. Huang, L. He, S. Li, et al. A simple fluorescence aptasensor for gastric cancer exosome detection based on branched rolling circle amplification. Nanoscale, 2020, 12(4): 2445−2451. https://doi.org/10.1039/C9NR08747H

[24]

D. Zhu, W. Liu, W.F. Cao, et al. Multiple amplified electrochemical detection of microRNA-21 using hierarchical flower-like gold nanostructures combined with gold-enriched hybridization chain reaction. Electroanalysis, 2018, 30(7): 1349−1356. https://doi.org/10.1002/elan.201700696

[25]

Y. Zhang, Z.H. Shuai, H. Zhou, et al. Single-molecule analysis of microRNA and logic operations using a smart plasmonic nanobiosensor. Journal of the American Chemical Society, 2018, 140(11): 3988−3993. https://doi.org/10.1021/jacs.7b12772

[26]

J.P. Zhang, Y.L. Liu, X. Zhi, et al. DNA-templated silver nanoclusters locate microRNAs in the nuclei of gastric cancer cells. Nanoscale, 2018, 10(23): 11079−11090. https://doi.org/10.1039/C8NR02634C

[27]

Y.Y. Tian, L. Zhang, L.H. Wang. DNA-functionalized plasmonic nanomaterials for optical biosensing. Biotechnology Journal, 2020, 15(1): e1800741. https://doi.org/10.1002/biot.201800741

[28]

S.J. Lin, X. Zhi, D. Chen, et al. A flyover style microfluidic chip for highly purified magnetic cell separation. Biosensors and Bioelectronics, 2019, 129: 175−181. https://doi.org/10.1016/j.bios.2018.12.058

[29]

H. Tang, J.Q. Niu, X.N. Pan, et al. Topology optimization based deterministic lateral displacement array design for cell separation. Journal of Chromatography A, 2022, 1679: 463384. https://doi.org/10.1016/j.chroma.2022.463384

[30]

X. Zhi, M. Deng, H. Yang, et al. A novel HBV genotypes detecting system combined with microfluidic chip, loop-mediated isothermal amplification and GMR sensors. Biosensors and Bioelectronics, 2014, 54: 372−377. https://doi.org/10.1016/j.bios.2013.11.025

[31]

Y. Zheng, K. Wang, J.J. Zhang, et al. Simultaneous Quantitative Detection of Helicobacter Pylori Based on a Rapid and Sensitive Testing Platform using Quantum Dots-Labeled Immunochromatiographic Test Strips. Nanoscale Research Letters, 2016, 11(1): 62. https://doi.org/10.1186/s11671-016-1254-7

[32]

S. Gao, L. Kang, M. Deng, et al. A giant magnetoimpedance-based microfluidic system for multiplex immunological assay. Nano Biomedicine and Engineering, 2016, 8(4): 240−245. https://doi.org/10.5101/nbe.v8i4.p240-245

[33]

K. Wang, J.C. Yang, H. Xu, et al. Smartphone-imaged multilayered paper-based analytical device for colorimetric analysis of carcinoembryonic antigen. Analytical and Bioanalytical Chemistry, 2020, 412(11): 2517−2528. https://doi.org/10.1007/s00216-020-02475-1

[34]
K. Wang, D.X. Cui. The application of immunochromatographic analysis in early detection of gastric cancer. In: Gastric Cancer Prewarning and Early Diagnosis System. Dordrecht: Springer, 2017: 129−156.
DOI
[35]

W.J. Wu, X.Y. Liu, M.F. Shen, et al. Multicolor quantum dot nanobeads based fluorescence-linked immunosorbent assay for highly sensitive multiplexed detection. Sensors and Actuators B:Chemical, 2021, 338: 129827. https://doi.org/10.1016/j.snb.2021.129827

[36]

J.A. Harrell, R. Kopelman. Biocompatible probes measure intracellular activity. Biophotonics International, 2000, 7: 22−24.

[37]

R. Weissleder. Molecular imaging: Exploring the next frontier. Radiology, 1999, 212(3): 609−614. https://doi.org/10.1148/radiology.212.3.r99se18609

[38]

C. Wang, C.C. Bao, S.J. Liang, et al. HAI-178 antibody-conjugated fluorescent magnetic nanoparticles for targeted imaging and simultaneous therapy of gastric cancer. Nanoscale Research Letters, 2014, 9(1): 274. https://doi.org/10.1186/1556-276X-9-274

[39]
T. Yin, H.G. Wu, Q. Zhang, et al. In vivo targeted therapy of gastric tumors via the mechanical rotation of a flower-like Fe3O4@Au nanoprobe under an alternating magnetic field. NPG Asia Materials, 2017, 9(7): e408.
DOI
[40]

B.F. Pan, D.X. Cui, Y. Sheng, et al. Dendrimer-modified magnetic nanoparticles enhance efficiency of gene delivery system. Cancer Research, 2007, 67(17): 8156−8163. https://doi.org/10.1158/0008-5472.CAN-06-4762

[41]

Y.F. Kong, J. Chen, F. Gao, et al. A multifunctional ribonuclease-A-conjugated CdTe quantum dot cluster nanosystem for synchronous cancer imaging and therapy. Small, 2010, 6(21): 2367−2373. https://doi.org/10.1002/smll.201001050

[42]

J. Ruan, H. Song, Q.R. Qian, et al. HER2 monoclonal antibody conjugated RNase-A-associated CdTe quantum dots for targeted imaging and therapy of gastric cancer. Biomaterials, 2012, 33(29): 7093−7102. https://doi.org/10.1016/j.biomaterials.2012.06.053

[43]

C. Li, Y. Ji, C. Wang, et al. BRCAA1 antibody- and Her2 antibody-conjugated amphiphilic polymer engineered CdSe/ZnS quantum dots for targeted imaging of gastric cancer. Nanoscale Research Letters, 2014, 9: 244. https://doi.org/10.1186/1556-276X-9-244

[44]

P. Huang, J. Lin, X.S. Wang, et al. Light-triggered theranostics based on photosensitizer-conjugated carbon dots for simultaneous enhanced-fluorescence imaging and photodynamic therapy. Advanced Materials, 2012, 24(37): 5104−5110. https://doi.org/10.1002/adma.201200650

[45]

Z.M. Li, P. Huang, X.J. Zhang, et al. RGD-conjugated dendrimer-modified gold nanorods for in vivo tumor targeting and photothermal therapy. Molecular Pharmaceutics, 2010, 7(1): 94−104. https://doi.org/10.1021/mp9001415

[46]

P. Huang, L. Bao, C.L. Zhang, et al. Folic acid-conjugated Silica-modified gold nanorods for X-ray/CT imaging-guided dual-mode radiation and photo-thermal therapy. Biomaterials, 2011, 32(36): 9796−9809. https://doi.org/10.1016/j.biomaterials.2011.08.086

[47]

S.H. Chen, C.C. Bao, C.L. Zhang, et al. EGFR antibody conjugated bimetallic Au@Ag nanorods for enhanced SERS-based tumor boundary identification, targeted photoacoustic imaging and photothermal therapy. Nano Biomedicine and Engineering, 2016, 8(4): 315−328. https://doi.org/10.5101/nbe.v8i4.p315-328

[48]

C.C. Bao, N. Beziere, P. del Pino, et al. Gold nanoprisms as optoacoustic signal nanoamplifiers for in vivo bioimaging of gastrointestinal cancers. Small, 2013, 9(1): 68−74. https://doi.org/10.1002/smll.201201779

[49]

S.J. Liang, C. Li, C.L. Zhang, et al. CD44v6 monoclonal antibody-conjugated gold nanostars for targeted photoacoustic imaging and plasmonic photothermal therapy of gastric cancer stem-like cells. Theranostics, 2015, 5(9): 970−984. https://doi.org/10.7150/thno.11632

[50]

X. Zhi, Y.L. Liu, L.N. Lin, et al. Oral pH sensitive GNS@ab nanoprobes for targeted therapy of Helicobacter pylori without disturbance gut microbiome. Nanomedicine:Nanotechnology,Biology and Medicine, 2019, 20: 102019. https://doi.org/10.1016/j.nano.2019.102019

[51]

Z.J. Zhou, C.L. Zhang, Q.R. Qian, et al. Folic acid-conjugated silica capped gold nanoclusters for targeted fluorescence/X-ray computed tomography imaging. Journal of Nanobiotechnology, 2013, 11: 17. https://doi.org/10.1186/1477-3155-11-17

[52]

C.L. Zhang, Z.J. Zhou, X. Zhi, et al. Insights into the distinguishing stress-induced cytotoxicity of chiral gold nanoclusters and the relationship with GSTP1. Theranostics, 2015, 5(2): 134−149. https://doi.org/10.7150/thno.10363

[53]

C. Zhou, G.Y. Hao, P. Thomas, et al. Near-infrared emitting radioactive gold nanoparticles with molecular pharmacokinetics. Angewandte Chemie International Edition, 2012, 51(40): 10118−10122. https://doi.org/10.1002/anie.201203031

[54]

M. He, P. Huang, C.L. Zhang, et al. Phase- and size-controllable synthesis of hexagonal upconversion rare-earth fluoride nanocrystals through an oleic acid/ionic liquid two-phase system. Chemistry, 2012, 18(19): 5954−5969. https://doi.org/10.1002/chem.201102419

[55]

J.B. Ma, P. Huang, M. He, et al. Folic acid-conjugated LaF3: Yb, Tm@SiO2 nanoprobes for targeting dual-modality imaging of upconversion luminescence and X-ray computed tomography. The Journal of Physical Chemistry B, 2012, 116(48): 14062−14070. https://doi.org/10.1021/jp309059u

[56]

F. Chen, P. Huang, Y.J. Zhu, et al. The photoluminescence, drug delivery and imaging properties of multifunctional Eu3+/Gd3+ dual-doped hydroxyapatite nanorods. Biomaterials, 2011, 32(34): 9031−9039. https://doi.org/10.1016/j.biomaterials.2011.08.032

[57]

X. Hu, J.H. Sun, F.Y. Li, et al. Renal-clearable hollow bismuth subcarbonate nanotubes for tumor targeted computed tomography imaging and chemoradiotherapy. Nano Letters, 2018, 18(2): 1196−1204. https://doi.org/10.1021/acs.nanolett.7b04741

[58]

C.Y. Wang, Y.P. Xiao, W.W. Zhu, et al. Photosensitizer-modified MnO2 nanoparticles to enhance photodynamic treatment of abscesses and boost immune protection for treated mice. Small, 2020, 16(28): e2000589. https://doi.org/10.1002/smll.202000589

[59]

Y. Ding, Q. Yan, J.W. Ruan, et al. Bone marrow mesenchymal stem cells and electroacupuncture downregulate the inhibitor molecules and promote the axonal regeneration in the transected spinal cord of rats. Cell Transplantation, 2011, 20(4): 475−491. https://doi.org/10.3727/096368910X528102

[60]

J. Ruan, H. Song, C. Li, et al. DiR-labeled Embryonic Stem Cells for Targeted Imaging of in vivo Gastric Cancer Cells. Theranostics, 2012, 2(6): 618−628. https://doi.org/10.7150/thno.4561

[61]

J. Ruan, J. Shen, Z. Wang, et al. Efficient preparation and labeling of human induced pluripotent stem cells by nanotechnology. International Journal of Nanomedicine, 2011, 6: 425−435. https://doi.org/10.2147/IJN.S16498

[62]

D.X. Cui, C.L. Zhang, B. Liu, et al. Regression of gastric cancer by systemic injection of RNA nanoparticles carrying both ligand and siRNA. Scientific Reports, 2015, 5: 10726. https://doi.org/10.1038/srep10726

[63]

C.X. Yue, Y.M. Yang, C.L. Zhang, et al. ROS-responsive mitochondria-targeting blended nanoparticles: Chemo- and photodynamic synergistic therapy for lung cancer with on-demand drug release upon irradiation with a single light source. Theranostics, 2016, 6(13): 2352−2366. https://doi.org/10.7150/thno

[64]

C.X. Yue, C.L. Zhang, G. Alfranca, et al. Near-Infrared Light Triggered ROS-activated Theranostic Platform based on Ce6-CPT-UCNPs for Simultaneous Fluorescence Imaging and Chemo-Photodynamic Combined Therapy. Theranostics, 2016, 6(4): 456−469. https://doi.org/10.7150/thno.14101

[65]

Y.L. Liu, Y.X. Pan, W. Cao, et al. A tumor microenvironment responsive biodegradable CaCO3/MnO2- based nanoplatform for the enhanced photodynamic therapy and improved PD-L1 immunotherapy. Theranostics, 2019, 9(23): 6867−6884. https://doi.org/10.7150/thno.37586

[66]

H. Song, R. He, K. Wang, et al. Anti-HIF-1α antibody-conjugated pluronic triblock copolymers encapsulated with Paclitaxel for tumor targeting therapy. Biomaterials, 2010, 31(8): 2302−2312. https://doi.org/10.1016/j.biomaterials.2009.11.067

[67]

J.L. Huang, G. Jiang, Q.X. Song, et al. Lipoprotein-biomimetic nanostructure enables efficient targeting delivery of siRNA to Ras-activated glioblastoma cells via macropinocytosis. Nature Communications, 2017, 8: 15144. https://doi.org/10.1038/ncomms15144

[68]

Z. Zhang, J. Guan, Z.X. Jiang, et al. Brain-targeted drug delivery by manipulating protein corona functions. Nature Communications, 2019, 10: 3561. https://doi.org/10.1038/s41467-019-11593-z

[69]

G.B. Yang, S.Z.F. Phua, W.Q. Lim, et al. A hypoxia-responsive albumin-based nanosystem for deep tumor penetration and excellent therapeutic efficacy. Advanced Materials, 2019, 31(25): e1901513. https://doi.org/10.1002/adma.201901513

[70]

Z.L. Chai, D.N. Ran, L.W. Lu, et al. Ligand-modified cell membrane enables the targeted delivery of drug nanocrystals to glioma. ACS Nano, 2019, 13(5): 5591−5601. https://doi.org/10.1021/acsnano.9b00661

[71]

Y. Zhang, K.M. Cai, C. Li, et al. Macrophage-membrane-coated nanoparticles for tumor-targeted chemotherapy. Nano Letters, 2018, 18(3): 1908−1915. https://doi.org/10.1021/acs.nanolett.7b05263

[72]

R.H. Fang,W.W. Gao, L.F. Zhang. Targeting drugs to tumours using cell membrane-coated nanoparticles. Nat Rev Clin Oncol, 2023, 20(1): 33−48. https://doi.org/10.1038/s41571-022-00699-x

[73]

G.Z. Li, S.P. Wang, D.S. Deng, et al. Fluorinated chitosan to enhance transmucosal delivery of sonosensitizer-conjugated catalase for sonodynamic bladder cancer treatment post-intravesical instillation. ACS Nano, 2020, 14(2): 1586−1599. https://doi.org/10.1021/acsnano.9b06689

[74]

J. Chen, H.L. Luo, Y. Liu, et al. Oxygen-self-produced nanoplatform for relieving hypoxia and breaking resistance to sonodynamic treatment of pancreatic cancer. ACS Nano, 2017, 11(12): 12849−12862. https://doi.org/10.1021/acsnano.7b08225

[75]

C. Feng, R.Z. Chen, W.W. Fang, et al. Synergistic effect of CD47 blockade in combination with cordycepin treatment against cancer. Frontiers in Pharmacology, 2023, 14: 1144330. https://doi.org/10.3389/fphar.2023.1144330

[76]

P.F. Zhao, W.M. Yin, A.H. Wu, et al. Dual-targeting to cancer cells and M2 macrophages via biomimetic delivery of mannosylated albumin nanoparticles for drug-resistant cancer therapy. Advanced Functional Materials, 2020, 30(16): 1700403. https://doi.org/10.1002/adfm.201700403

[77]

Y. Qian, S. Qiao, Y.F. Dai, et al. Molecular-targeted immunotherapeutic strategy for melanoma via dual-targeting nanoparticles delivering small interfering RNA to tumor-associated macrophages. ACS Nano, 2017, 11(9): 9536−9549. https://doi.org/10.1021/acsnano.7b05465

[78]

C.R. Shi, T. Liu, Z.D. Guo, et al. Reprogramming tumor-associated macrophages by nanoparticle-based reactive oxygen species photogeneration. Nano Letters, 2018, 18(11): 7330−7342. https://doi.org/10.1021/acs.nanolett.8b03568

[79]

W.W. Zhang, L.J. Li, D.G. Li, et al. The first approved gene therapy product for cancer ad-p53 (gendicine): 12 years in the clinic. Human Gene Therapy, 2018, 29(2): 160−179. https://doi.org/10.1089/hum.2017.218

[80]

T.Z. Zhan, N. Rindtorff, J. Betge, et al. CRISPR/Cas9 for cancer research and therapy. Seminars in Cancer Biology, 2019, 55: 106−119. https://doi.org/10.1016/j.semcancer.2018.04.001

[81]

J. Kim, A. Jozic, Y.X. Lin, et al. Engineering lipid nanoparticles for enhanced intracellular delivery of mRNA through inhalation. ACS Nano, 2022, 16(9): 14792−14806. https://doi.org/10.1021/acsnano.2c05647

[82]

F.M. Pi, D.W. Binzel, T.J. Lee, et al. Nanoparticle orientation to control RNA loading and ligand display on extracellular vesicles for cancer regression. Nature Nanotechnology, 2018, 13(1): 82−89. https://doi.org/10.1038/s41565-017-0012-z

[83]

Y.C. Pan, J.J. Yang, X.W. Luan, et al. Near-infrared upconversion–activated CRISPR-Cas9 system: A remote-controlled gene editing platform. Science Advances, 2019, 5(4): eaav7199. https://doi.org/10.1126/sciadv.aav7199

[84]

R.B. Patel, M.Z. Ye, P.M. Carlson, et al. Development of an in situ cancer vaccine via combinational radiation and bacterial-membrane-coated nanoparticles. Advanced Materials, 2019, 31(43): 1902626. https://doi.org/10.1002/adma.201902626

[85]
B. Hou, D.G. Wang, J. Gao, H. Wang, Y.P. Li, H.J. Yu. Advances of microenvironment-activated nanosized drug delivery system for cancer immunotherapy. Acta Pharmaceutica Sinica, 2019, 12, 1802-1809. (in Chinese)
[86]

R. Yang, J. Xu, L.G. Xu, et al. Cancer cell membrane-coated adjuvant nanoparticles with mannose modification for effective anticancer vaccination. ACS Nano, 2018, 12(6): 5121−5129. https://doi.org/10.1021/acsnano.7b09041

[87]

L.Q. Liu, Y. Wang, X. Guo, et al. A biomimetic polymer magnetic nanocarrier polarizing tumor-associated macrophages for potentiating immunotherapy. Small, 2020, 16(38): e2003543. https://doi.org/10.1002/smll.202003543

[88]

F.Y. Zhou, B. Feng, H.J. Yu, et al. Tumor microenvironment-activatable prodrug vesicles for nanoenabled cancer chemoimmunotherapy combining immunogenic cell death induction and CD47 blockade. Advanced Materials, 2019, 31(14): e1805888. https://doi.org/10.1002/adma.201805888

[89]

O. Rixe, J.C. Morris, V.K. Puduvalli, et al. First-in-human, first-in-class phase 1a study of BXQ-350 for solid tumors and gliomas. Journal of Clinical Oncology, 2018, 36(15_suppl): 2517. https://doi.org/10.1200/jco.2018.36.15_suppl.2517

[90]

W. Li, F.L. Wu, S.L. Zhao, et al. Correlation between PD-1/PD-L1 expression and polarization in tumor-associated macrophages: A key player in tumor immunotherapy. Cytokine &Growth Factor Reviews, 2022, 67: 49−57. https://doi.org/10.1016/j.cytogfr.2022.07.004

[91]

W.Q. Wang, Y.L. Jin, X. Liu, et al. Endogenous stimuli-activatable nanomedicine for immune theranostics for cancer. Advanced Functional Materials, 2021, 31(26): 2100386. https://doi.org/10.1002/adfm.202100386

[92]

E. M. Cheng, N. W. Tsarovsky, P. M, Sondel, A. L. Rakhmilevich. Interleukin-12 as an in situ cancer vaccine component: a review. Cancer Immunol Immunother, 2022, 71(9): 2057−2065. https://doi.org/10.1007/s00262-022-03144-1

[93]

L. Zhou, P.C. Zhang, H. Wang, et al. Smart nanosized drug delivery systems inducing immunogenic cell death for combination with cancer immunotherapy. Accounts of Chemical Research, 2020, 53(9): 1761−1772. https://doi.org/10.1021/acs.accounts.0c00254

[94]

Y.Z. Chang, L.Z. He, Z.B. Li, et al. Designing core–shell gold and selenium nanocomposites for cancer radiochemotherapy. ACS Nano, 2017, 11(5): 4848−4858. https://doi.org/10.1021/acsnano.7b01346

[95]

H.J. Song, H. Sun, N.N. He, et al. Gadolinium-based ultra-small nanoparticles augment radiotherapy-induced T-cell response to synergize with checkpoint blockade immunotherapy. Nanoscale, 2022, 14(31): 11429−11442. https://doi.org/10.1039/D2NR02620A

[96]

A. Wicki, D. Witzigmann, V. Balasubramanian, et al. Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications. Journal of Controlled Release, 2015, 200: 138−157. https://doi.org/10.1016/j.jconrel.2014.12.030

[97]

M.L. Etheridge, S.A. Campbell, A.G. Erdman, et al. The big picture on nanomedicine: The state of investigational and approved nanomedicine products. Nanomedicine:Nanotechnology,Biology and Medicine, 2013, 9(1): 1−14. https://doi.org/10.1016/j.nano.2012.05.013

[98]
Chen, H. B., Gu, Z. J., An, H. W., Chen, C. Y., Chen, J., Cui, R., Chen, S. Q., Chen, W. H., Chen, X. S., Chen, X. Y. et al. Precise nanomedicine for intelligent therapy of cancer. Science China Chemistry, 2018, 61(12): 1503–1552.
DOI
[99]

M. Germain, F. Caputo, S. Metcalfe, et al. Delivering the power of nanomedicine to patients today. Journal of Controlled Release, 2020, 326: 164−171. https://doi.org/10.1016/j.jconrel.2020.07.007

[100]
E.K.H. Chow, D. Ho. Cancer nanomedicine: From drug delivery to imaging. Science Translational Medicine, 2013, 5(216): 216rv4
DOI
[101]
Gonzalez-Valdivieso, J., Girotti, A., Schneider, J., Arias, F. J. Advanced nanomedicine and cancer: Challenges and opportunities in clinical translation. International Journal of Pharmaceutics, 2021, 599: 120438.
DOI
[102]

C. von Roemeling, W. Jiang, C.K. Chan, et al. Breaking down the barriers to precision cancer nanomedicine. Trends in Biotechnology, 2017, 35(2): 159−171. https://doi.org/10.1016/j.tibtech.2016.07.006

[103]

J.I. Hare, T. Lammers, M.B. Ashford, et al. Challenges and strategies in anti-cancer nanomedicine development: An industry perspective. Advanced Drug Delivery Reviews, 2017, 108: 25−38. https://doi.org/10.1016/j.addr.2016.04.025

[104]

Z.M. Li, X.T. Shan, Z.D. Chen, et al. Applications of surface modification technologies in nanomedicine for deep tumor penetration. Advanced Science, 2020, 8(1): 2002589. https://doi.org/10.1002/advs.202002589

[105]

S. Kunjachan, J. Ehling, G. Storm, et al. Noninvasive imaging of nanomedicines and nanotheranostics: Principles, progress, and prospects. Chemical Reviews, 2015, 115(19): 10907−10937. https://doi.org/10.1021/cr500314d

[106]

Y.Z. Min, J.M. Caster, M.J. Eblan, et al. Clinical translation of nanomedicine. Chemical Reviews, 2015, 115(19): 11147−11190. https://doi.org/10.1021/acs.chemrev.5b00116

[107]

B. Pelaz, C. Alexiou, R.A. Alvarez-Puebla, et al. Diverse applications of nanomedicine. ACS Nano, 2017, 11(3): 2313−2381. https://doi.org/10.1021/acsnano.6b06040

[108]

J.Y. Ren, N. Andrikopoulos, K. Velonia, et al. Chemical and biophysical signatures of the protein corona in nanomedicine. Journal of the American Chemical Society, 2022, 144(21): 9184−9205. https://doi.org/10.1021/jacs.2c02277

[109]

M. Sousa de Almeida, E. Susnik, B. Drasler, et al. Understanding nanoparticle endocytosis to improve targeting strategies in nanomedicine. Chemical Society Reviews, 2021, 50(9): 5397−5434. https://doi.org/10.1039/D0CS01127D

[110]

C.B. He, D.M. Liu, W.B. Lin. Nanomedicine applications of hybrid nanomaterials built from metal-ligand coordination bonds: Nanoscale metal-organic frameworks and nanoscale coordination polymers. Chemical Reviews, 2015, 115(19): 11079−11108. https://doi.org/10.1021/acs.chemrev.5b00125

[111]

J.H. Lee, J.H. Choi, S.TD. Chueng, et al. Nondestructive characterization of stem cell neurogenesis by a magneto-plasmonic nanomaterial-based exosomal miRNA detection. ACS Nano, 2019, 13(8): 8793−8803. https://doi.org/10.1021/acsnano.9b01875

[112]

S.Y. Xu, B. Liu, J.Y. Fan, et al. Engineered mesenchymal stem cell-derived exosomes with high CXCR4 levels for targeted siRNA gene therapy against cancer. Nanoscale, 2022, 14(11): 4098−4113. https://doi.org/10.1039/D1NR08170E

[113]
J. Czyz, C. Wiese, A. Rolletschek, et al. Potential of embryonic and adult stem cells in vitro. Biological Chemistry, 2003, 384(10–11): 1391–1409.
DOI
[114]

P.C. Chagastelles, N.B. Nardi. Biology of stem cells: An overview. Kidney International Supplements, 2011, 1(3): 63−67. https://doi.org/10.1038/kisup.2011.15

[115]

U.M. Domanska, R.C. Kruizinga, W.B. Nagengast, et al. A review on CXCR4/CXCL12 axis in oncology: No place to hide. European Journal of Cancer, 2013, 49(1): 219−230. https://doi.org/10.1016/j.ejca.2012.05.005

[116]

Z.D. Wang, J. Sun, Y.Q. Feng, et al. Oncogenic roles and drug target of CXCR4/CXCL12 axis in lung cancer and cancer stem cell. Tumor Biology, 2016, 37(7): 8515−8528. https://doi.org/10.1007/s13277-016-5016-z

[117]

D.X. Cui, H. Zhang, Z. Wang, et al. Effects of dendrimer-functionalized multi-walled carbon nanotubes on murine embryonic stem cells. ECS Transactions, 2008, 13(14): 111−116. https://doi.org/10.1149/1.2998536

[118]

J. Ruan, J.J. Ji, H. Song, et al. Fluorescent magnetic nanoparticle-labeled mesenchymal stem cells for targeted imaging and hyperthermia therapy of in vivo gastric cancer. Nanoscale Research Letters, 2012, 7: 309. https://doi.org/10.1186/1556-276X-7-309

[119]

C. Li, J. Ruan, M. Yang, et al. Human induced pluripotent stem cells labeled with fluorescent magnetic nanoparticles for targeted imaging and hyperthermia therapy for gastric cancer. Cancer Biology &Medicine, 2015, 12(3): 163−174. https://doi.org/10.7497/j.issn.2095-3941.2015.0040

[120]

Y.L. Liu, M. Yang, J.P. Zhang, et al. Human induced pluripotent stem cells for tumor targeted delivery of gold nanorods and enhanced photothermal therapy. ACS Nano, 2016, 10(2): 2375−2385. https://doi.org/10.1021/acsnano.5b07172

[121]

J. Conde, C.C. Bao, D.X. Cui, et al. Antibody-drug gold nanoantennas with Raman spectroscopic fingerprints for in vivo tumour theranostics. Journal of Controlled Release, 2014, 183: 87−93. https://doi.org/10.1016/j.jconrel.2014.03.045

[122]
J. Conde, F.R. Tian, Y. Hernández, et al. In vivo tumor targeting via nanoparticle-mediated therapeutic siRNA coupled to inflammatory response in lung cancer mouse models. Biomaterials, 2013, 34(31): 7744–7753.
DOI
[123]

C.C. Bao, J. Conde, F. Pan, et al. Gold nanoprisms as a hybrid in vivo cancer theranostic platform for in situ photoacoustic imaging, angiography, and localized hyperthermia. Nano Research, 2016, 9(4): 1043−1056. https://doi.org/10.1007/s12274-016-0996-y

[124]

J. Conde, J.T. Dias, V. Grazú, et al. Revisiting 30 years of biofunctionalization and surface chemistry of inorganic nanoparticles for nanomedicine. Frontiers in Chemistry, 2014, 2: 48. https://doi.org/10.3389/fchem.2014.00048

[125]
J. Conde, A. Ambrosone, Y. Hernandez, et al. 15 years on siRNA delivery: Beyond the State-of-the-Art on inorganic nanoparticles for RNAi therapeutics. Nano Today, 2015, 10(4): 421–450.
DOI
[126]

A. Kumari, S.K. Yadav, S.C. Yadav. Biodegradable polymeric nanoparticles based drug delivery systems. Colloids and Surfaces B:Biointerfaces, 2010, 75(1): 1−18. https://doi.org/10.1016/j.colsurfb.2009.09.001

[127]

M.M. Roberts, J.L. White, M.G. Grütter, et al. Three-dimensional structure of the adenovirus major coat protein hexon. Science, 1986, 232(4754): 1148−1151. https://doi.org/10.1126/science.3704642

[128]

Y.L. Liu, X. Zhi, M. Yang, et al. Tumor-triggered drug release from calcium carbonate-encapsulated gold nanostars for near-infrared photodynamic/photothermal combination antitumor therapy. Theranostics, 2017, 7(6): 1650−1662. https://doi.org/10.7150/thno.17602

[129]
A.A. Date, J. Hanes, L.M. Ensign, Nanoparticles for oral delivery: Design, evaluation and state-of-the-art. Journal of Controlled Release, 2016, 240: 504–526.
DOI
[130]
P. Singh, S.K. Sahoo. Nano-oncology: Clinical application for cancer therapy and future perspectives. In: Cancer Nanotheranostics. Cham: Springer, 2021: 49−95.
DOI
[131]
Liu, Y., Solomon, M., Achilefu, S. Perspectives and potential applications of nanomedicine in breast and prostate cancer. Medicinal Research Reviews, 2013, 33(1): 3–32.
DOI
[132]

N. Abood, M. Jabir, H. Kadhim. TNF-α; loaded on gold nanoparticles as a good therapeutic agent against breast cancer AMJ13 cells. Nano Biomedicine and Engineering, 2020, 12(3): 262−271. https://doi.org/10.5101/nbe.v12i3.p262-271

[133]

X.X. Han, M.J. Mitchell, G.J. Nie. Nanomaterials for therapeutic RNA delivery. Matter, 2020, 3(6): 1948−1975. https://doi.org/10.1016/j.matt.2020.09.020

[134]

M. Kumar, U. Kumar, A.K. Singh. Therapeutic nanoparticles: Recent developments and their targeted delivery applications. Nano Biomedicine and Engineering, 2022, 14(1): 38−52. https://doi.org/10.5101/nbe.v14i1.p38-52

[135]

Y. Lu, W.J. Sun, Z. Gu. Stimuli-responsive nanomaterials for therapeutic protein delivery. Journal of Controlled Release, 2014, 194: 1−19. https://doi.org/10.1016/j.jconrel.2014.08.015

[136]

A. Alekhya, A.K. Sailaja. Formulation and evaluation of letrozole nanoparticles by salting out technique and determination of anti-cancer activity by MTT assay. Nano Biomedicine and Engineering, 2022, 14(3): 246−253. https://doi.org/10.5101/nbe.v14i3.p246-253

[137]

V. Biju. Chemical modifications and bioconjugate reactions of nanomaterials for sensing, imaging, drug delivery and therapy. Chemical Society Reviews, 2014, 43(3): 744−764. https://doi.org/10.1039/C3CS60273G

[138]

S.A. Rashdan. Chemical detection of the toxicity of nanoparticles of metals and metal oxides. Nano Biomedicine and Engineering, 2021, 13(4): 401−413. https://doi.org/10.5101/nbe.v13i4.p401-413

[139]

B. Elder, R. Neupane, E. Tokita, et al. Nanomaterial patterning in 3D printing. Advanced Materials, 2020, 32(17): e1907142. https://doi.org/10.1002/adma.201907142

[140]

K. Thorkelsson, P. Bai, T. Xu. Self-assembly and applications of anisotropic nanomaterials: A review. Nano Today, 2015, 10(1): 48−66. https://doi.org/10.1016/j.nantod.2014.12.005

[141]

E. Baquedano, R.V. Martinez, J.M. Llorens, et al. Fabrication of silicon nanobelts and nanopillars by soft lithography for hydrophobic and hydrophilic photonic surfaces. Nanomaterials, 2017, 7(5): 109. https://doi.org/10.3390/nano7050109

[142]

D.M. Ju, Y. Zhang, R. Li, et al. Mechanism-independent manipulation of single-wall carbon nanotubes with atomic force microscopy tip. Nanomaterials, 2020, 10(8): 1494. https://doi.org/10.3390/nano10081494

[143]

G. Gonçalves, M. Vila, M.T. Portolés, et al. Nano-graphene oxide: A potential multifunctional platform for cancer therapy. Advanced Healthcare Materials, 2013, 2(8): 1072−1090. https://doi.org/10.1002/adhm.201300023

[144]

A. Sawdon, E. Weydemeyer, C.A. Peng. Tumor photothermolysis: Using carbon nanomaterials for cancer therapy. European Journal of Nanomedicine, 2013, 5(3): 131−140. https://doi.org/10.1515/ejnm-2013-0006

[145]

A.O. Choi, S.J. Cho, J. Desbarats, et al. Quantum dot-induced cell death involves Fas upregulation and lipid peroxidation in human neuroblastoma cells. Journal of Nanobiotechnology, 2007, 5: 1. https://doi.org/10.1186/1477-3155-5-1

[146]

W.J. Chen, Y.T. Xu, D.C. Yang, et al. Preparation of liposomes coated superparamagnetic iron oxide nanoparticles for targeting and imaging brain glioma. Nano Biomedicine and Engineering, 2022, 14(1): 71−80. https://doi.org/10.5101/nbe.v14i1.p71-80

[147]

G.K. Ibadi, A.A. Taha, S.M.H. Al-Jawad. Anticancer activity of copper-chitosan nanocomposite conjugated with folic acid. Nano Biomedicine and Engineering, 2022, 14(4): 317−328. https://doi.org/10.5101/nbe.v14i4.p317-328

[148]

A.M. Tomşa, A.L. Răchişan, A.A. Aldea, et al. Perspectives of gold nanoparticles and their applications in pancreatic cancer (Review). Experimental and Therapeutic Medicine, 2021, 21(3): 258. https://doi.org/10.3892/etm.2021.9689

[149]

G. Barsisa, A. Belay, G. Beyene, et al. Synthesis europium (Eu3+) doped zinc oxide nanoparticles via the co-precipitation method for photocatalytic applications. Nano Biomedicine and Engineering, 2022, 14(1): 58−70. https://doi.org/10.5101/nbe.v14i1.p58-70

[150]

Y.H. Zheng, Y. Wang, M.Y. Xia, et al. The combination of nanotechnology and traditional Chinese medicine (TCM) inspires the modernization of TCM: Review on nanotechnology in TCM-based drug delivery systems. Drug Delivery and Translational Research, 2022, 12(6): 1306−1325. https://doi.org/10.1007/s13346-021-01029-x

[151]

N.C. Seeman, H.F. Sleiman. DNA nanotechnology. Nature Reviews Materials, 2017, 3: 17068. https://doi.org/10.1038/natrevmats.2017.68

[152]

P.X. Guo, F. Haque, B. Hallahan, et al. Uniqueness, advantages, challenges, solutions, and perspectives in therapeutics applying RNA nanotechnology. Nucleic Acid Therapeutics, 2012, 22(4): 226−245. https://doi.org/10.1089/nat.2012.0350

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 29 July 2023
Revised: 30 October 2023
Accepted: 24 November 2023
Published: 05 February 2024
Issue date: June 2024

Copyright

© The Author(s) 2024.

Acknowledgements

Acknowledgements

This work was financially supported by International Cooperation Project of National Natural Science Foundation of China (No. 82020108017), Innovation Group Project of National Natural Science Foundation of China (No. 81921002), the National Key Research and Development Project of China (No. 2017YFA0205301), Standard project of Shanghai Science and Technology Commission (21DZ2203200), and Instrument Project of Shanghai Science and Technology Commission (20142201300), and China Postdoctoral Science Foundation (Grant No. 2020M671130).

Rights and permissions

This is an open-access article distributed under  the  terms  of  the  Creative  Commons  Attribution  4.0 International  License (CC BY) (http://creativecommons.org/licenses/by/4.0/), which  permits  unrestricted  use,  distribution,  and reproduction in any medium, provided the original author and source are credited.

Return