Nanomaterials for Diagnosis and Treatment of Brain Cancer: Recent Updates
Abstract
:1. Introduction
2. Nanomaterials for Treatment of Brain Cancer (Brain Tumor)
2.1. Liposomes
2.2. Nano-Micelles
2.3. Dendrimers
2.4. Carbon Nanotubes and Carbon Dots
2.5. Gold and Silver NPs
2.6. Zinc Oxide NPs
3. Nanomaterials for the Diagnosis and Biosensing of Brain Cancer
3.1. Magnetic NPs
3.2. Metallic NPs
3.3. Quantum Dots
3.4. Polymeric Nano Vehicles
3.5. Multimodal Imaging with Nanomaterials
3.6. Extracellular Vesicles (EVs) and Exosomes
3.7. Mesenchymal Stem Cells Engineered Nano-Based Imaging
3.8. Biomimetic Nanocomposites
4. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Zhang, J. Secrets of the Brain: An introduction to the brain anatomical structure and biological function. arXiv 2019, arXiv:1906.03314. [Google Scholar]
- Schiavi, S.; Ocampo-Pineda, M.; Barakovic, M.; Petit, L.; Descoteaux, M.; Thiran, J.-P.; Daducci, A. A new method for accurate in vivo mapping of human brain connections using microstructural and anatomical information. Sci. Adv. 2020, 6, eaba8245. [Google Scholar] [CrossRef] [PubMed]
- Stiller, C.A.; Gatta, G. Oxford Textbook of Cancer in Children; Oxford University Press: Oxford, UK, 2020. [Google Scholar]
- Wang, X.; Yu, Y.; Zang, L.; Zhang, P.; Ma, J.; Chen, D. Targeting clusterin induces apoptosis, reduces growth ability and invasion and mediates sensitivity to chemotherapy in human osteosarcoma cells. Curr. Pharm. Biotechnol. 2020, 21, 131–139. [Google Scholar] [CrossRef] [PubMed]
- Zumel-Marne, A.; Kundi, M.; Castaño-Vinyals, G.; Alguacil, J.; Petridou, E.T.; Georgakis, M.K.; Morales-Suárez-Varela, M.; Sadetzki, S.; Piro, S.; Nagrani, R. Clinical presentation of young people (10–24 years old) with brain tumors: Results from the international MOBI-Kids study. J. Neuro-Oncol. 2020, 147, 427–440. [Google Scholar] [CrossRef] [Green Version]
- Bhaduri, A.; Di Lullo, E.; Jung, D.; Müller, S.; Crouch, E.E.; Espinosa, C.S.; Ozawa, T.; Alvarado, B.; Spatazza, J.; Cadwell, C.R. Outer radial glia-like cancer stem cells contribute to heterogeneity of glioblastoma. Cell Stem Cell 2020, 26, 48–63.e46. [Google Scholar] [CrossRef] [PubMed]
- Brat, D.J.; Aldape, K.; Colman, H.; Holland, E.C.; Louis, D.N.; Jenkins, R.B.; Kleinschmidt-DeMasters, B.; Perry, A.; Reifenberger, G.; Stupp, R. cIMPACT-NOW update 3: Recommended diagnostic criteria for “Diffuse astrocytic glioma, IDH-wildtype, with molecular features of glioblastoma, WHO grade IV”. Acta Neuropathol. 2018, 136, 805–810. [Google Scholar] [CrossRef] [Green Version]
- Nakamura, K.; Smyth, M.J. Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell. Mol. Immunol. 2020, 17, 1–12. [Google Scholar] [CrossRef]
- Nicolaides, T.; Nazemi, K.J.; Crawford, J.; Kilburn, L.; Minturn, J.; Gajjar, A.; Gauvain, K.; Leary, S.; Dhall, G.; Aboian, M. Phase I study of vemurafenib in children with recurrent or progressive BRAFV600E mutant brain tumors: Pacific Pediatric Neuro-Oncology Consortium study (PNOC-002). Oncotarget 2020, 11, 1942. [Google Scholar] [CrossRef]
- Stewart, C.A.; Gay, C.M.; Xi, Y.; Sivajothi, S.; Sivakamasundari, V.; Fujimoto, J.; Bolisetty, M.; Hartsfield, P.M.; Balasubramaniyan, V.; Chalishazar, M.D. Single-cell analyses reveal increased intratumoral heterogeneity after the onset of therapy resistance in small-cell lung cancer. Nat. Cancer 2020, 1, 423–436. [Google Scholar] [CrossRef]
- 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]
- Wojcicki, A.V.; Kadapakkam, M.; Frymoyer, A.; Lacayo, N.; Chae, H.-D.; Sakamoto, K.M. Repurposing drugs for acute myeloid leukemia: A worthy Cause or a Futile Pursuit? Cancers 2020, 12, 441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Blake, Z.; Marks, D.K.; Gartrell, R.D.; Hart, T.; Horton, P.; Cheng, S.K.; Taback, B.; Horst, B.A.; Saenger, Y.M. Complete intracranial response to talimogene laherparepvec (T-Vec), pembrolizumab and whole brain radiotherapy in a patient with melanoma brain metastases refractory to dual checkpoint-inhibition. J. Immunother. Cancer 2018, 6, 25. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Lovell, J.F.; Yoon, J.; Chen, X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat. Rev. Clin. Oncol. 2020, 17, 657–674. [Google Scholar] [CrossRef] [PubMed]
- Jain, K.K. A critical overview of targeted therapies for glioblastoma. Front. Oncol. 2018, 8, 419. [Google Scholar] [CrossRef] [PubMed]
- El Demerdash, N.; Kedda, J.; Ram, N.; Brem, H.; Tyler, B. Novel therapeutics for brain tumors: Current practice and future prospects. Expert Opin. Drug Deliv. 2020, 17, 9–21. [Google Scholar] [CrossRef]
- Puttemans, J.; Lahoutte, T.; D’Huyvetter, M.; Devoogdt, N. Beyond the Barrier: Targeted Radionuclide Therapy in Brain Tumors and Metastases. Pharmaceutics 2019, 11, 376. [Google Scholar] [CrossRef] [Green Version]
- Yan, W.; Khan, M.K.; Wu, X.; Simone, C.B. Spatially fractionated radiation therapy: History, present and the future. Clin. Transl. Radiat. Oncol. 2020, 20, 30. [Google Scholar] [CrossRef] [Green Version]
- Jena, L.; McErlean, E.; McCarthy, H. Delivery across the blood-brain barrier: Nanomedicine for glioblastoma multiforme. Drug Deliv. Transl. Res. 2020, 10, 304–318. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Y.; Peng, Z.; Seven, E.S.; Leblanc, R.M. Crossing the blood-brain barrier with nanoparticles. J. Control. Release 2018, 270, 290–303. [Google Scholar] [CrossRef]
- Rosenblum, D.; Joshi, N.; Tao, W.; Karp, J.M.; Peer, D. Progress and challenges towards targeted delivery of cancer therapeutics. Nat. Commun. 2018, 9, 1410. [Google Scholar] [CrossRef] [Green Version]
- Shi, D.; Mi, G.; Shen, Y.; Webster, T.J. Glioma-targeted dual functionalized thermosensitive Ferri-liposomes for drug delivery through an in vitro blood–brain barrier. Nanoscale 2019, 11, 15057–15071. [Google Scholar] [CrossRef] [PubMed]
- Ma, F.; Zhang, X.; Yin, K.-J. Micrornas in central nervous system diseases: A prospective role in regulating blood-brain barrier integrity. Exp. Neurol. 2020, 323, 113094. [Google Scholar] [CrossRef]
- Alamu, O.; Rado, M.; Ekpo, O.; Fisher, D. Differential Sensitivity of Two Endothelial Cell Lines to Hydrogen Peroxide Toxicity: Relevance for In Vitro Studies of the Blood–Brain Barrier. Cells 2020, 9, 403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, S.; Khan, A.I.; Cai, X.; Song, Y.; Lyu, Z.; Du, D.; Dutta, P.; Lin, Y. Overcoming blood–brain barrier transport: Advances in nanoparticle-based drug delivery strategies. Mater. Today 2020, 37, 112–125. [Google Scholar] [CrossRef] [PubMed]
- Tandel, G.S.; Biswas, M.; Kakde, O.G.; Tiwari, A.; Suri, H.S.; Turk, M.; Laird, J.R.; Asare, C.K.; Ankrah, A.A.; Khanna, N. A review on a deep learning perspective in brain cancer classification. Cancers 2019, 11, 111. [Google Scholar] [CrossRef] [Green Version]
- Molina, E.S.; Schipmann, S.; Stummer, W. Maximizing safe resections: The roles of 5-aminolevulinic acid and intraoperative MR imaging in glioma surgery—Review of the literature. Neurosurg. Rev. 2019, 42, 197–208. [Google Scholar] [CrossRef] [Green Version]
- Choi, I.Y.; Andronesi, O.C.; Barker, P.; Bogner, W.; Edden, R.A.; Kaiser, L.G.; Lee, P.; Marjańska, M.; Terpstra, M.; de Graaf, R.A. Spectral editing in 1H magnetic resonance spectroscopy: Experts’ consensus recommendations. NMR Biomed. 2020, e4411. [Google Scholar] [CrossRef]
- Sharma, U.; Jagannathan, N.R. Metabolism of prostate cancer by magnetic resonance spectroscopy (MRS). Biophys. Rev. 2020, 12, 1163–1173. [Google Scholar] [CrossRef]
- Atun, R.; Bhakta, N.; Denburg, A.; Frazier, A.L.; Friedrich, P.; Gupta, S.; Lam, C.G.; Ward, Z.J.; Yeh, J.M.; Allemani, C. Sustainable care for children with cancer: A Lancet Oncology Commission. Lancet Oncol. 2020, 21, e185–e224. [Google Scholar] [CrossRef]
- Zottel, A.; Videtič Paska, A.; Jovčevska, I. Nanotechnology meets oncology: Nanomaterials in brain cancer research, diagnosis and therapy. Materials 2019, 12, 1588. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Li, M.; Gao, X.; Chen, Y.; Liu, T. Nanotechnology in cancer diagnosis: Progress, challenges and opportunities. J. Hematol. Oncol. 2019, 12, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martins, J.P.; das Neves, J.; de la Fuente, M.; Celia, C.; Florindo, H.; Günday-Türeli, N.; Popat, A.; Santos, J.L.; Sousa, F.; Schmid, R. The solid progress of nanomedicine. Drug Deliv. Transl. Res. 2020, 10, 726–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mendiratta, S.; Hussein, M.; Nasser, H.A.; Ali, A.A.A. Multidisciplinary role of mesoporous silica nanoparticles in brain regeneration and cancers: From crossing the blood–brain barrier to treatment. Part. Part. Syst. Charact. 2019, 36, 1900195. [Google Scholar] [CrossRef] [Green Version]
- Neves, A.R.; Queiroz, J.F.; Lima, S.A.C.; Reis, S. Apo E-functionalization of solid lipid nanoparticles enhances brain drug delivery: Uptake mechanism and transport pathways. Bioconjug. Chem. 2017, 28, 995–1004. [Google Scholar] [CrossRef] [PubMed]
- Muthuraman, A.; Rishitha, N.; Mehdi, S. Role of nanoparticles in bioimaging, diagnosis and treatment of cancer disorder. In Design of Nanostructures for Theranostics Applications; Elsevier: Amsterdam, The Netherlands, 2018; pp. 529–562. [Google Scholar]
- Nowak, M.; Helgeson, M.E.; Mitragotri, S. Delivery of nanoparticles and macromolecules across the blood–brain barrier. Adv. Ther. 2020, 3, 1900073. [Google Scholar] [CrossRef]
- Gonzalez-Carter, D.; Liu, X.; Tockary, T.A.; Dirisala, A.; Toh, K.; Anraku, Y.; Kataoka, K. Targeting nanoparticles to the brain by exploiting the blood–brain barrier impermeability to selectively label the brain endothelium. Proc. Natl. Acad. Sci. USA 2020, 117, 19141–19150. [Google Scholar] [CrossRef]
- Lopalco, A.; Denora, N. Nanoformulations for Drug delivery: Safety, toxicity, and efficacy. In Computational Toxicology; Springer: Berlin/Heidelberg, Germany, 2018; pp. 347–365. [Google Scholar]
- El-Sawy, H.S.; Al-Abd, A.M.; Ahmed, T.A.; El-Say, K.M.; Torchilin, V.P. Stimuli-responsive nano-architecture drug-delivery systems to solid tumor micromilieu: Past, present, and future perspectives. ACS Nano 2018, 12, 10636–10664. [Google Scholar] [CrossRef]
- Lombardo, D.; Kiselev, M.A.; Caccamo, M.T. Smart nanoparticles for drug delivery application: Development of versatile nanocarrier platforms in biotechnology and nanomedicine. J. Nanomater. 2019, 2019, 3702518. [Google Scholar] [CrossRef]
- Biswas, A.; Shukla, A.; Maiti, P. Biomaterials for interfacing cell imaging and drug delivery: An overview. Langmuir 2019, 35, 12285–12305. [Google Scholar] [CrossRef]
- Wu, X.; Yang, H.; Yang, W.; Chen, X.; Gao, J.; Gong, X.; Wang, H.; Duan, Y.; Wei, D.; Chang, J. Nanoparticle-based diagnostic and therapeutic systems for brain tumors. J. Mater. Chem. B 2019, 7, 4734–4750. [Google Scholar] [CrossRef]
- Andrade, K.N.; Pérez, A.M.P.; Arízaga, G.G.C. Passive and active targeting strategies in hybrid layered double hydroxides nanoparticles for tumor bioimaging and therapy. Appl. Clay Sci. 2019, 181, 105214. [Google Scholar] [CrossRef]
- Wang, S.; Zhou, Z.; Yu, G.; Lu, N.; Liu, Y.; Dai, Y.; Fu, X.; Wang, J.; Chen, X. Gadolinium metallofullerene-polypyrrole nanoparticles for activatable dual-modal imaging-guided photothermal therapy. ACS Appl. Mater. Interfaces 2018, 10, 28382–28389. [Google Scholar] [CrossRef] [PubMed]
- Zeng, Y.; Li, H.; Li, Z.; Luo, Q.; Zhu, H.; Gu, Z.; Zhang, H.; Gong, Q.; Luo, K. Engineered gadolinium-based nanomaterials as cancer imaging agents. Appl. Mater. Today 2020, 20, 100686. [Google Scholar] [CrossRef]
- Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M.; Adeli-sardou, M. Evaluation of Carum-loaded Niosomes on Breast Cancer Cells: Physicochemical Properties, In Vitro Cytotoxicity, Flow Cytometric, DNA Fragmentation and Cell Migration Assay. Sci. Rep. 2019, 9, 7139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M.; Lohrasbi-Nejad, A.; Nematollahi, M.H. A new formulation of hydrophobin-coated niosome as a drug carrier to cancer cells. Mater. Sci. Eng. C 2020, 113, 110975. [Google Scholar] [CrossRef] [PubMed]
- Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M.; Nematollahi, M.H. Lawsone-loaded Niosome and its antitumor activity in MCF-7 breast Cancer cell line: A Nano-herbal treatment for Cancer. DARU J. Pharm. Sci. 2018, 26, 11–17. [Google Scholar] [CrossRef]
- Barani, M.; Nematollahi, M.H.; Zaboli, M.; Mirzaei, M.; Torkzadeh-Mahani, M.; Pardakhty, A.; Karam, G.A. In silico and in vitro study of magnetic niosomes for gene delivery: The effect of ergosterol and cholesterol. Mater. Sci. Eng. C 2019, 94, 234–246. [Google Scholar] [CrossRef]
- Barani, M.; Sabir, F.; Rahdar, A.; Arshad, R.; Z Kyzas, G. Nanotreatment and Nanodiagnosis of Prostate Cancer: Recent Updates. Nanomaterials 2020, 10, 1696. [Google Scholar] [CrossRef]
- Barani, M.; Torkzadeh-Mahani, M.; Mirzaei, M.; Nematollahi, M.H. Comprehensive Evaluation of Gene Expression in Negative and Positive Trigger-based Targeting Niosomes in HEK-293 Cell Line. Iran. J. Pharm. Res. 2020, 19, 166–180. [Google Scholar]
- Bilal, M.; Barani, M.; Sabir, F.; Rahdar, A.; Kyzas, G.Z. Nanomaterials for the treatment and diagnosis of Alzheimer’s disease: An overview. NanoImpact 2020, 20, 100251. [Google Scholar] [CrossRef]
- Das, S.S.; Bharadwaj, P.; Bilal, M.; Barani, M.; Rahdar, A.; Taboada, P.; Bungau, S.; Kyzas, G.Z. Stimuli-Responsive Polymeric Nanocarriers for Drug Delivery, Imaging, and Theragnosis. Polymers 2020, 12, 1397. [Google Scholar] [CrossRef] [PubMed]
- Davarpanah, F.; Yazdi, A.K.; Barani, M.; Mirzaei, M.; Torkzadeh-Mahani, M. Magnetic delivery of antitumor carboplatin by using PEGylated-Niosomes. DARU J. Pharm. Sci. 2018, 26, 57–64. [Google Scholar] [CrossRef] [PubMed]
- Ebrahimi, A.K.; Barani, M.; Sheikhshoaie, I. Fabrication of a new superparamagnetic metal-organic framework with core-shell nanocomposite structures: Characterization, biocompatibility, and drug release study. Mater. Sci. Eng. C 2018, 92, 349–355. [Google Scholar] [CrossRef] [PubMed]
- Hajizadeh, M.R.; Maleki, H.; Barani, M.; Fahmidehkar, M.A.; Mahmoodi, M.; Torkzadeh-Mahani, M. In vitro cytotoxicity assay of D-limonene niosomes: An efficient nano-carrier for enhancing solubility of plant-extracted agents. Res. Pharm. Sci. 2019, 14, 448. [Google Scholar]
- Hajizadeh, M.R.; Parvaz, N.; Barani, M.; Khoshdel, A.; Fahmidehkar, M.A.; Mahmoodi, M.; Torkzadeh-Mahani, M. Diosgenin-loaded niosome as an effective phytochemical nanocarrier: Physicochemical characterization, loading efficiency, and cytotoxicity assay. DARU J. Pharm. Sci. 2019, 27, 329–339. [Google Scholar] [CrossRef]
- Rahdar, A.; Hajinezhad, M.R.; Nasri, S.; Beyzaei, H.; Barani, M.; Trant, J.F. The synthesis of methotrexate-loaded F127 microemulsions and their in vivo toxicity in a rat model. J. Mol. Liq. 2020, 313, 113449. [Google Scholar] [CrossRef]
- Rahdar, A.; Taboada, P.; Hajinezhad, M.R.; Barani, M.; Beyzaei, H. Effect of tocopherol on the properties of Pluronic F127 microemulsions: Physico-chemical characterization and in vivo toxicity. J. Mol. Liq. 2019, 277, 624–630. [Google Scholar] [CrossRef]
- Torkzadeh-Mahani, M.; Zaboli, M.; Barani, M.; Torkzadeh-Mahani, M. A combined theoretical and experimental study to improve the thermal stability of recombinant D-lactate dehydrogenase immobilized on a novel superparamagnetic Fe3O4NPs@ metal–organic framework. Appl. Organomet. Chem. 2020, 34, e5581. [Google Scholar] [CrossRef]
- Rahdar, A.; Aliahmad, M.; Samani, M.; HeidariMajd, M.; Susan, M.A.B.H. Synthesis and characterization of highly efficacious Fe-doped ceria nanoparticles for cytotoxic and antifungal activity. Ceram. Int. 2019, 45, 7950–7955. [Google Scholar] [CrossRef]
- Rahdar, A.; Hajinezhad, M.R.; Sankar, V.S.; Askari, F.; Noura, M.; Kyzas, G.Z. Synthesis, characterization, and intraperitoneal biochemical studies of zinc oxide nanoparticles in Rattus norvegicus. Appl. Phys. A 2020, 126, 347. [Google Scholar] [CrossRef]
- Taimoory, S.M.; Rahdar, A.; Aliahmad, M.; Sadeghfar, F.; Hajinezhad, M.R.; Jahantigh, M.; Shahbazi, P.; Trant, J.F. The synthesis and characterization of a magnetite nanoparticle with potent antibacterial activity and low mammalian toxicity. J. Mol. Liq. 2018, 265, 96–104. [Google Scholar] [CrossRef]
- Nikazar, S.; Sivasankarapillai, V.S.; Rahdar, A.; Gasmi, S.; Anumol, P.; Shanavas, M.S. Revisiting the cytotoxicity of quantum dots: An in-depth overview. Biophys. Rev. 2020, 12, 703–718. [Google Scholar] [CrossRef] [PubMed]
- Pillai, A.M.; Sivasankarapillai, V.S.; Rahdar, A.; Joseph, J.; Sadeghfar, F.; Rajesh, K.; Kyzas, G.Z. Green synthesis and characterization of zinc oxide nanoparticles with antibacterial and antifungal activity. J. Mol. Struct. 2020, 1211, 128107. [Google Scholar] [CrossRef]
- Rahdar, A.; Hajinezhad, M.R.; Bilal, M.; Askari, F.; Kyzas, G.Z. Behavioral effects of zinc oxide nanoparticles on the brain of rats. Inorg. Chem. Commun. 2020, 119, 108131. [Google Scholar] [CrossRef]
- Rahdar, A.; Hajinezhad, M.R.; Hamishekar, H.; Ghamkhari, A.; Kyzas, G.Z. Copolymer/graphene oxide nanocomposites as potential anticancer agents. Polym. Bull. 2020, 1–22. [Google Scholar] [CrossRef]
- Saravani, R.; Sargazi, S.; Saravani, R.; Rabbani, M.; Rahdar, A.; Taboada, P. Newly crocin-coated magnetite nanoparticles induce apoptosis and decrease VEGF expression in breast carcinoma cells. J. Drug Deliv. Sci. Technol. 2020, 60, 101987. [Google Scholar] [CrossRef]
- Sivasankarapillai, V.S.; Somakumar, A.K.; Joseph, J.; Nikazar, S.; Rahdar, A.; Kyzas, G.Z. Cancer theranostic applications of MXene nanomaterials: Recent updates. Nano-Struct. Nano-Objects 2020, 22, 100457. [Google Scholar] [CrossRef]
- Sivasankarapillai, V.S.; Pillai, A.M.; Rahdar, A.; Sobha, A.P.; Das, S.S.; Mitropoulos, A.C.; Mokarrar, M.H.; Kyzas, G.Z. On Facing the SARS-CoV-2 (COVID-19) with Combination of Nanomaterials and Medicine: Possible Strategies and First Challenges. Nanomaterials 2020, 10, 852. [Google Scholar] [CrossRef]
- Sayadi, K.; Rahdar, A.; Hajinezhad, M.R.; Nikazar, S.; Susan, M.A.B.H. Atorvastatin-loaded SBA-16 nanostructures: Synthesis, physical characterization, and biochemical alterations in hyperlipidemic rats. J. Mol. Struct. 2020, 1202, 127296. [Google Scholar] [CrossRef]
- Rahdar, A.; Beyzaei, H.; Askari, F.; Kyzas, G.Z. Gum-based cerium oxide nanoparticles for antimicrobial assay. Appl. Phys. A 2020, 126, 324. [Google Scholar] [CrossRef]
- Davarpanah, A.M.; Rahdar, A.; Dastnae, M.A.; Zeybek, O.; Beyzaei, H. (1−x) BaFe12O19/xCoFe2O4 hard/soft magnetic nanocomposites: Synthesis, physical characterization, and antibacterial activities study. J. Mol. Struct. 2019, 1175, 445–449. [Google Scholar] [CrossRef]
- Sengul, A.B.; Asmatulu, E. Toxicity of metal and metal oxide nanoparticles: A review. Environ. Chem. Lett. 2020, 18, 1659–1683. [Google Scholar] [CrossRef]
- Quader, S.; Kataoka, K. Nanomaterial-enabled cancer therapy. Mol. Ther. 2017, 25, 1501–1513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jain, K. Nanobiotechnology-based drug delivery to the central nervous system. Neurodegener. Dis. 2007, 4, 287–291. [Google Scholar] [CrossRef]
- Teleanu, D.M.; Chircov, C.; Grumezescu, A.M.; Volceanov, A.; Teleanu, R.I. Impact of nanoparticles on brain health: An up to date overview. J. Clin. Med. 2018, 7, 490. [Google Scholar] [CrossRef] [Green Version]
- Yasui, T.; Kaji, N.; Baba, Y. Nanobiodevices for biomolecule analysis and imaging. Annu. Rev. Anal. Chem. 2013, 6, 83–96. [Google Scholar] [CrossRef] [PubMed]
- Frank, D.; Tyagi, C.; Tomar, L.; Choonara, Y.E.; du Toit, L.C.; Kumar, P.; Penny, C.; Pillay, V. Overview of the role of nanotechnological innovations in the detection and treatment of solid tumors. Int J Nanomed. 2014, 9, 589–613. [Google Scholar] [CrossRef] [Green Version]
- Gatoo, M.A.; Naseem, S.; Arfat, M.Y.; Dar, A.M.; Qasim, K.; Zubair, S. Physicochemical properties of nanomaterials: Implication in associated toxic manifestations. BioMed Res. Int. 2014, 2014, 498420. [Google Scholar] [CrossRef]
- Malam, Y.; Loizidou, M.; Seifalian, A.M. Liposomes and nanoparticles: Nanosized vehicles for drug delivery in cancer. Trends Pharmacol. Sci. 2009, 30, 592–599. [Google Scholar] [CrossRef] [PubMed]
- Torchilin, V.P. Recent advances with liposomes as pharmaceutical carriers. Nat. Rev. Drug Discov. 2005, 4, 145–160. [Google Scholar] [CrossRef]
- Bozzuto, G.; Molinari, A. Liposomes as nanomedical devices. Int. J. Nanomed. 2015, 10, 975. [Google Scholar] [CrossRef] [Green Version]
- Allen, T.M.; Cullis, P.R. Liposomal drug delivery systems: From concept to clinical applications. Adv. Drug Deliv. Rev. 2013, 65, 36–48. [Google Scholar] [CrossRef]
- Immordino, M.L.; Dosio, F.; Cattel, L. Stealth liposomes: Review of the basic science, rationale, and clinical applications, existing and potential. Int. J. Nanomed. 2006, 1, 297. [Google Scholar]
- Lakkadwala, S.; dos Santos Rodrigues, B.; Sun, C.; Singh, J. Dual functionalized liposomes for efficient co-delivery of anti-cancer chemotherapeutics for the treatment of glioblastoma. J. Control. Release 2019, 307, 247–260. [Google Scholar] [CrossRef] [PubMed]
- Muthu, M.S.; Kulkarni, S.A.; Xiong, J.; Feng, S.-S. Vitamin E TPGS coated liposomes enhanced cellular uptake and cytotoxicity of docetaxel in brain cancer cells. Int. J. Pharm. 2011, 421, 332–340. [Google Scholar] [CrossRef] [PubMed]
- Zhan, C.; Gu, B.; Xie, C.; Li, J.; Liu, Y.; Lu, W. Cyclic RGD conjugated poly (ethylene glycol)-co-poly (lactic acid) micelle enhances paclitaxel anti-glioblastoma effect. J. Control. Release 2010, 143, 136–142. [Google Scholar] [CrossRef] [PubMed]
- Postma, T.; Heimans, J.; Luykx, S.; Van Groeningen, C.; Beenen, L.; Hoekstra, O.; Taphoorn, M.; Zonnenberg, B.; Klein, M.; Vermorken, J. A phase II study of paclitaxel in chemonaive patients with recurrent high-grade glioma. Ann. Oncol. 2000, 11, 409–413. [Google Scholar] [CrossRef] [PubMed]
- Anfosso, L.; Efferth, T.; Albini, A.; Pfeffer, U. Microarray expression profiles of angiogenesis-related genes predict tumor cell response to artemisinins. Pharm. J. 2006, 6, 269–278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qiao, Y.; Wan, J.; Zhou, L.; Ma, W.; Yang, Y.; Luo, W.; Yu, Z.; Wang, H. Stimuli-responsive nanotherapeutics for precision drug delivery and cancer therapy. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2019, 11, e1527. [Google Scholar] [CrossRef] [Green Version]
- Pantshwa, J.M.; Kondiah, P.P.; Choonara, Y.E.; Marimuthu, T.; Pillay, V. Nanodrug Delivery Systems for the Treatment of Ovarian Cancer. Cancers 2020, 12, 213. [Google Scholar] [CrossRef] [Green Version]
- Yu, F.; Jiang, F.; Tang, X.; Wang, B. N-octyl-N-arginine-chitosan micelles for gambogic acid intravenous delivery: Characterization, cell uptake, pharmacokinetics, and biodistribution. Drug Dev. Ind. Pharm. 2018, 44, 615–623. [Google Scholar] [CrossRef] [PubMed]
- Fathi, M.; Majidi, S.; Zangabad, P.S.; Barar, J.; Erfan-Niya, H.; Omidi, Y. Chitosan-based multifunctional nanomedicines and theranostics for targeted therapy of cancer. Med. Res. Rev. 2018, 38, 2110–2136. [Google Scholar] [CrossRef] [PubMed]
- Feng, S.-T.; Li, J.; Luo, Y.; Yin, T.; Cai, H.; Wang, Y.; Dong, Z.; Shuai, X.; Li, Z.-P. pH-sensitive nanomicelles for controlled and efficient drug delivery to human colorectal carcinoma LoVo cells. PLoS ONE 2014, 9, e100732. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Shi, Y.; Kim, J.Y.; Park, K.; Cheng, J.-X. Overcoming the barriers in micellar drug delivery: Loading efficiency, in vivo stability, and micelle–cell interaction. Expert Opin. Drug Deliv. 2010, 7, 49–62. [Google Scholar] [CrossRef]
- Agrawal, P.; Singh, R.P.; Sharma, G.; Mehata, A.K.; Singh, S.; Rajesh, C.V.; Pandey, B.L.; Koch, B.; Muthu, M.S. Bioadhesive micelles of d-α-tocopherol polyethylene glycol succinate 1000: Synergism of chitosan and transferrin in targeted drug delivery. Colloids Surf. B Biointerfaces 2017, 152, 277–288. [Google Scholar] [CrossRef]
- Sonali; Agrawal, P.; Singh, R.P.; Rajesh, C.V.; Singh, S.; Vijayakumar, M.R.; Pandey, B.L.; Muthu, M.S. Transferrin receptor-targeted vitamin E TPGS micelles for brain cancer therapy: Preparation, characterization and brain distribution in rats. Drug Deliv. 2016, 23, 1788–1798. [Google Scholar] [CrossRef] [Green Version]
- Mittal, S.; Ashhar, M.U.; Qizilbash, F.F.; Qamar, Z.; Narang, J.K.; Kumar, S.; Ali, J.; Baboota, S. Ligand Conjugated Targeted Nanotherapeutics for Treatment of Neurological Disorders. Curr. Pharm. Des. 2020, 26, 2291–2305. [Google Scholar] [CrossRef]
- Niu, J.; Wang, L.; Yuan, M.; Zhang, J.; Chen, H.; Zhang, Y. Dual-targeting nanocarrier based on glucose and folic acid functionalized pluronic P105 polymeric micelles for enhanced brain distribution. J. Drug Deliv. Sci. Technol. 2020, 57, 101343. [Google Scholar] [CrossRef]
- Nanjwade, B.K.; Bechra, H.M.; Derkar, G.K.; Manvi, F.; Nanjwade, V.K. Dendrimers: Emerging polymers for drug-delivery systems. Eur. J. Pharm. Sci. 2009, 38, 185–196. [Google Scholar] [CrossRef]
- Noriega-Luna, B.; Godínez, L.A.; Rodríguez, F.J.; Rodríguez, A.; Zaldívar-Lelo de Larrea, G.; Sosa-Ferreyra, C.; Mercado-Curiel, R.; Manríquez, J.; Bustos, E. Applications of dendrimers in drug delivery agents, diagnosis, therapy, and detection. J. Nanomater. 2014, 2014, 507273. [Google Scholar] [CrossRef] [Green Version]
- Mishra, V.; Kesharwani, P. Dendrimer technologies for brain tumor. Drug Discov. Today 2016, 21, 766–778. [Google Scholar] [CrossRef]
- Ambekar, R.S.; Choudhary, M.; Kandasubramanian, B. Recent advances in dendrimer-based nanoplatform for cancer treatment: A review. Eur. Polym. J. 2020, 126, 109546. [Google Scholar] [CrossRef]
- He, H.; Li, Y.; Jia, X.-R.; Du, J.; Ying, X.; Lu, W.-L.; Lou, J.-N.; Wei, Y. PEGylated Poly (amidoamine) dendrimer-based dual-targeting carrier for treating brain tumors. Biomaterials 2011, 32, 478–487. [Google Scholar] [CrossRef] [PubMed]
- Yan, H.; Wang, L.; Wang, J.; Weng, X.; Lei, H.; Wang, X.; Jiang, L.; Zhu, J.; Lu, W.; Wei, X. Two-order targeted brain tumor imaging by using an optical/paramagnetic nanoprobe across the blood brain barrier. Acs Nano 2012, 6, 410–420. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Zhao, Z.; Gao, H.; Rostami, I.; You, Q.; Jia, X.; Wang, C.; Zhu, L.; Yang, Y. Enhanced blood-brain-barrier penetrability and tumor-targeting efficiency by peptide-functionalized poly (amidoamine) dendrimer for the therapy of gliomas. Nanotheranostics 2019, 3, 311. [Google Scholar] [CrossRef] [PubMed]
- Muniswamy, V.J.; Raval, N.; Gondaliya, P.; Tambe, V.; Kalia, K.; Tekade, R.K. ‘Dendrimer-Cationized-Albumin’encrusted polymeric nanoparticle improves BBB penetration and anticancer activity of doxorubicin. Int. J. Pharm. 2019, 555, 77–99. [Google Scholar] [CrossRef]
- Mahajan, S.; Patharkar, A.; Kuche, K.; Maheshwari, R.; Deb, P.K.; Kalia, K.; Tekade, R.K. Functionalized carbon nanotubes as emerging delivery system for the treatment of cancer. Int. J. Pharm. 2018, 548, 540–558. [Google Scholar] [CrossRef]
- Harsha, P.; Thotakura, N.; Kumar, M.; Sharma, S.; Mittal, A.; Khurana, R.K.; Singh, B.; Negi, P.; Raza, K. A novel PEGylated carbon nanotube conjugated mangiferin: An explorative nanomedicine for brain cancer cells. J. Drug Deliv. Sci. Technol. 2019, 53, 101186. [Google Scholar] [CrossRef]
- You, Y.; Wang, N.; He, L.; Shi, C.; Zhang, D.; Liu, Y.; Luo, L.; Chen, T. Designing dual-functionalized carbon nanotubes with high blood–brain-barrier permeability for precise orthotopic glioma therapy. Dalton Trans. 2019, 48, 1569–1573. [Google Scholar] [CrossRef]
- Mishra, V.; Patil, A.; Thakur, S.; Kesharwani, P. Carbon dots: Emerging theranostic nanoarchitectures. Drug Discov. Today 2018, 23, 1219–1232. [Google Scholar] [CrossRef]
- Zuo, J.; Jiang, T.; Zhao, X.; Xiong, X.; Xiao, S.; Zhu, Z. Preparation and application of fluorescent carbon dots. J. Nanomater. 2015. [Google Scholar] [CrossRef] [Green Version]
- Zheng, M.; Liu, S.; Li, J.; Qu, D.; Zhao, H.; Guan, X.; Hu, X.; Xie, Z.; Jing, X.; Sun, Z. Integrating oxaliplatin with highly luminescent carbon dots: An unprecedented theranostic agent for personalized medicine. Adv. Mater. 2014, 26, 3554–3560. [Google Scholar] [CrossRef] [PubMed]
- Hettiarachchi, S.D.; Graham, R.M.; Mintz, K.J.; Zhou, Y.; Vanni, S.; Peng, Z.; Leblanc, R.M. Triple conjugated carbon dots as a nano-drug delivery model for glioblastoma brain tumors. Nanoscale 2019, 11, 6192–6205. [Google Scholar] [CrossRef]
- Sonali, M.K.V.; Singh, R.P.; Agrawal, P.; Mehata, A.K.; Datta Maroti Pawde, N.; Sonkar, R.; Muthu, M.S. Nanotheranostics: Emerging strategies for early diagnosis and therapy of brain cancer. Nanotheranostics 2018, 2, 70. [Google Scholar] [CrossRef]
- Fan, Z.; Fu, P.P.; Yu, H.; Ray, P.C. Theranostic nanomedicine for cancer detection and treatment. J. Food Drug Anal. 2014, 22, 3–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Norden, A.D.; Drappatz, J.; Wen, P.Y. Novel anti-angiogenic therapies for malignant gliomas. Lancet Neurol. 2008, 7, 1152–1160. [Google Scholar] [CrossRef]
- Pandey, S.; Oza, G.; Mewada, A.; Shah, R.; Thakur, M.; Sharon, M. Folic acid mediated synaphic delivery of doxorubicin using biogenic gold nanoparticles anchored to biological linkers. J. Mater. Chem. B 2013, 1, 1361–1370. [Google Scholar] [CrossRef]
- Alle, M.; Kim, T.H.; Park, S.H.; Lee, S.-H.; Kim, J.-C. Doxorubicin-carboxymethyl xanthan gum capped gold nanoparticles: Microwave synthesis, characterization, and anti-cancer activity. Carbohydr. Polym. 2020, 229, 115511. [Google Scholar] [CrossRef]
- Mohamed, M.S.; Veeranarayanan, S.; Poulose, A.C.; Nagaoka, Y.; Minegishi, H.; Yoshida, Y.; Maekawa, T.; Kumar, D.S. Type 1 ribotoxin-curcin conjugated biogenic gold nanoparticles for a multimodal therapeutic approach towards brain cancer. Biochim. Biophys. Acta (BBA)—Gen. Subj. 2014, 1840, 1657–1669. [Google Scholar] [CrossRef]
- Le Ouay, B.; Stellacci, F. Antibacterial activity of silver nanoparticles: A surface science insight. Nano Today 2015, 10, 339–354. [Google Scholar] [CrossRef] [Green Version]
- Abdal Dayem, A.; Hossain, M.K.; Lee, S.B.; Kim, K.; Saha, S.K.; Yang, G.-M.; Choi, H.Y.; Cho, S.-G. The role of reactive oxygen species (ROS) in the biological activities of metallic nanoparticles. Int. J. Mol. Sci. 2017, 18, 120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ebrahimzadeh, Z.; Salehzadeh, A.; Naeemi, A.; Jalali, A. Silver nanoparticles biosynthesized by Anabaena flos-aquae enhance the apoptosis in breast cancer cell line. Bull. Mater. Sci. 2020, 43, 1–7. [Google Scholar] [CrossRef]
- Salazar-García, S.; García-Rodrigo, J.F.; Martínez-Castañón, G.A.; Ruiz-Rodríguez, V.M.; Portales-Pérez, D.P.; Gonzalez, C. Silver nanoparticles (AgNPs) and zinc chloride (ZnCl2) exposure order determines the toxicity in C6 rat glioma cells. J. Nanopart. Res. 2020, 22, 253. [Google Scholar] [CrossRef]
- Locatelli, E.; Naddaka, M.; Uboldi, C.; Loudos, G.; Fragogeorgi, E.; Molinari, V.; Pucci, A.; Tsotakos, T.; Psimadas, D.; Ponti, J. Targeted delivery of silver nanoparticles and alisertib: In vitro and in vivo synergistic effect against glioblastoma. Nanomedicine 2014, 9, 839–849. [Google Scholar] [CrossRef] [Green Version]
- Jin, T.; Sun, D.; Su, J.; Zhang, H.; Sue, H.J. Antimicrobial efficacy of zinc oxide quantum dots against Listeria monocytogenes, Salmonella enteritidis, and Escherichia coli O157:H7. J. Food Sci. 2009, 74, M46–M52. [Google Scholar] [CrossRef]
- Rasmussen, J.W.; Martinez, E.; Louka, P.; Wingett, D.G. Zinc oxide nanoparticles for selective destruction of tumor cells and potential for drug delivery applications. Expert Opin. Drug Deliv. 2010, 7, 1063–1077. [Google Scholar] [CrossRef] [Green Version]
- Jin, B.; Bae, S.; Lee, S.; Im, S. Effects of native defects on optical and electrical properties of ZnO prepared by pulsed laser deposition. Mater. Sci. Eng. B 2000, 71, 301–305. [Google Scholar] [CrossRef]
- Wang, Z.L. Nanostructures of zinc oxide. Mater. Today 2004, 7, 26–33. [Google Scholar] [CrossRef]
- Lao, J.; Huang, J.; Wang, D.; Ren, Z. ZnO nanobridges and nanonails. Nano Lett. 2003, 3, 235–238. [Google Scholar] [CrossRef]
- Zhang, H.; Yang, D.; Li, D.; Ma, X.; Li, S.; Que, D. Controllable growth of ZnO microcrystals by a capping-molecule-assisted hydrothermal process. Cryst. Growth Des. 2005, 5, 547–550. [Google Scholar] [CrossRef]
- Ostrovsky, S.; Kazimirsky, G.; Gedanken, A.; Brodie, C. Selective cytotoxic effect of ZnO nanoparticles on glioma cells. Nano Res. 2009, 2, 882–890. [Google Scholar] [CrossRef] [Green Version]
- Shim, K.H.; Hulme, J.; Maeng, E.H.; Kim, M.-K.; An, S.S.A. Analysis of zinc oxide nanoparticles binding proteins in rat blood and brain homogenate. Int. J. Nanomed. 2014, 9, 217. [Google Scholar]
- Wei, L.; Wang, J.; Chen, A.; Liu, J.; Feng, X.; Shao, L. Involvement of PINK1/parkin-mediated mitophagy in ZnO nanoparticle-induced toxicity in BV-2 cells. Int. J. Nanomed. 2017, 12, 1891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, A.K.; Singh, V.; Gera, R.; Purohit, M.P.; Ghosh, D. Zinc oxide nanoparticle induces microglial death by NADPH-oxidase-independent reactive oxygen species as well as energy depletion. Mol. Neurobiol. 2017, 54, 6273–6286. [Google Scholar] [CrossRef] [PubMed]
- Jeng, H.A.; Swanson, J. Toxicity of metal oxide nanoparticles in mammalian cells. J. Environ. Sci. Health Part A 2006, 41, 2699–2711. [Google Scholar] [CrossRef] [PubMed]
- Wahab, R.; Kaushik, N.K.; Verma, A.K.; Mishra, A.; Hwang, I.; Yang, Y.-B.; Shin, H.-S.; Kim, Y.-S. Fabrication and growth mechanism of ZnO nanostructures and their cytotoxic effect on human brain tumor U87, cervical cancer HeLa, and normal HEK cells. JBIC J. Biol. Inorg. Chem. 2011, 16, 431–442. [Google Scholar] [CrossRef]
- Attia, H.; Nounou, H.; Shalaby, M. Zinc oxide nanoparticles induced oxidative DNA damage, inflammation and apoptosis in rat’s brain after oral exposure. Toxics 2018, 6, 29. [Google Scholar] [CrossRef] [Green Version]
- Reddy, L.H.; Couvreur, P. Nanotechnology for therapy and imaging of liver diseases. J. Hepatol. 2011, 55, 1461–1466. [Google Scholar] [CrossRef] [Green Version]
- Baetke, S.C.; Lammers, T.; Kiessling, F. Applications of nanoparticles for diagnosis and therapy of cancer. Br. J. Radiol. 2015, 88, 20150207. [Google Scholar] [CrossRef]
- Meyers, J.D.; Doane, T.; Burda, C.; Basilion, J.P. Nanoparticles for imaging and treating brain cancer. Nanomedicine 2012, 8, 123–143. [Google Scholar] [CrossRef] [Green Version]
- Weissleder, R.; Nahrendorf, M.; Pittet, M.J. Imaging macrophages with nanoparticles. Nat. Mater. 2014, 13, 125–138. [Google Scholar] [CrossRef] [PubMed]
- Vasefi, F.; MacKinnon, N.; Farkas, D.L.; Kateb, B. Review of the potential of optical technologies for cancer diagnosis in neurosurgery: A step toward intraoperative neurophotonics. Neurophotonics 2016, 4, 011010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, W.; Fan, W.; Lau, J.; Deng, L.; Shen, Z.; Chen, X. Emerging blood–brain-barrier-crossing nanotechnology for brain cancer theranostics. Chem. Soc. Rev. 2019, 48, 2967–3014. [Google Scholar] [CrossRef] [PubMed]
- Huang, R.; Harmsen, S.; Samii, J.M.; Karabeber, H.; Pitter, K.L.; Holland, E.C.; Kircher, M.F. High precision imaging of microscopic spread of glioblastoma with a targeted ultrasensitive SERRS molecular imaging probe. Theranostics 2016, 6, 1075. [Google Scholar] [CrossRef]
- Nicolson, F.; Andreiuk, B.; Andreou, C.; Hsu, H.-T.; Rudder, S.; Kircher, M.F. Non-invasive in vivo imaging of cancer using surface-enhanced spatially offset Raman spectroscopy (SESORS). Theranostics 2019, 9, 5899. [Google Scholar] [CrossRef]
- Richard, S.; Boucher, M.; Lalatonne, Y.; Mériaux, S.; Motte, L. Iron oxide nanoparticle surface decorated with cRGD peptides for magnetic resonance imaging of brain tumors. Biochim. Biophys. Acta (BBA)—Gen. Subj. 2017, 1861, 1515–1520. [Google Scholar] [CrossRef]
- Shen, Z.; Liu, T.; Yang, Z.; Zhou, Z.; Tang, W.; Fan, W.; Liu, Y.; Mu, J.; Li, L.; Bregadze, V.I. Small-sized gadolinium oxide based nanoparticles for high-efficiency theranostics of orthotopic glioblastoma. Biomaterials 2020, 235, 119783. [Google Scholar] [CrossRef]
- Miao, Z.; Hu, D.; Gao, D.; Fan, L.; Ma, Y.; Ma, T.; Liu, X.; Zheng, H.; Zha, Z.; Sheng, Z. Tiny 2D silicon quantum sheets: A brain photonic nanoagent for orthotopic glioma theranostics. Sci. Bull. 2020. [Google Scholar] [CrossRef]
- Liu, C.; Chen, J.; Zhu, Y.; Gong, X.; Zheng, R.; Chen, N.; Chen, D.; Yan, H.; Zhang, P.; Zheng, H. Highly sensitive MoS 2–indocyanine green hybrid for photoacoustic imaging of orthotopic brain glioma at deep site. Nano-Micro Lett. 2018, 10, 48. [Google Scholar] [CrossRef] [Green Version]
- Peng, C.; Gao, X.; Xu, J.; Du, B.; Ning, X.; Tang, S.; Bachoo, R.M.; Yu, M.; Ge, W.-P.; Zheng, J. Targeting orthotopic gliomas with renal-clearable luminescent gold nanoparticles. Nano Res. 2017, 10, 1366–1376. [Google Scholar] [CrossRef]
- Davidi, E.S.; Dreifuss, T.; Motiei, M.; Shai, E.; Bragilovski, D.; Lubimov, L.; Kindler, M.J.J.; Popovtzer, A.; Don, J.; Popovtzer, R. Cisplatin-conjugated gold nanoparticles as a theranostic agent for head and neck cancer. Head Neck 2018, 40, 70–78. [Google Scholar] [CrossRef] [PubMed]
- Feng, Q.; Shen, Y.; Fu, Y.; Muroski, M.E.; Zhang, P.; Wang, Q.; Xu, C.; Lesniak, M.S.; Li, G.; Cheng, Y. Self-assembly of gold nanoparticles shows microenvironment-mediated dynamic switching and enhanced brain tumor targeting. Theranostics 2017, 7, 1875. [Google Scholar] [CrossRef] [PubMed]
- Coluccia, D.; Figueiredo, C.A.; Wu, M.Y.; Riemenschneider, A.N.; Diaz, R.; Luck, A.; Smith, C.; Das, S.; Ackerley, C.; O’Reilly, M. Enhancing glioblastoma treatment using cisplatin-gold-nanoparticle conjugates and targeted delivery with magnetic resonance-guided focused ultrasound. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 1137–1148. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.; Chen, W.; Chen, Y.; Zhang, H.; Liu, C.; Deng, Z.; Sheng, Z.; Chen, J.; Liu, X.; Yan, F. Focused Ultrasound-Augmented Delivery of Biodegradable Multifunctional Nanoplatforms for Imaging-Guided. Adv. Sci. 2018, 5, 1700474. [Google Scholar] [CrossRef] [PubMed]
- Neuschmelting, V.; Harmsen, S.; Beziere, N.; Lockau, H.; Hsu, H.T.; Huang, R.; Razansky, D.; Ntziachristos, V.; Kircher, M.F. Dual-modality surface-enhanced resonance Raman scattering and multispectral Optoacoustic tomography nanoparticle approach for brain tumor delineation. Small 2018, 14, 1800740. [Google Scholar] [CrossRef]
- Gao, X.; Yue, Q.; Liu, Z.; Ke, M.; Zhou, X.; Li, S.; Zhang, J.; Zhang, R.; Chen, L.; Mao, Y. Guiding brain-tumor surgery via blood–brain-barrier-permeable gold nanoprobes with acid-triggered MRI/SERRS signals. Adv. Mater. 2017, 29, 1603917. [Google Scholar] [CrossRef]
- Lu, B.; Huang, Y.; Chen, Z.; Ye, J.; Xu, H.; Chen, W.; Long, X. Niosomal Nanocarriers for Enhanced Skin Delivery of Quercetin with Functions of Anti-Tyrosinase and Antioxidant. Molecules 2019, 24, 2322. [Google Scholar] [CrossRef] [Green Version]
- Garrigue, P.; Tang, J.; Ding, L.; Bouhlel, A.; Tintaru, A.; Laurini, E.; Huang, Y.; Lyu, Z.; Zhang, M.; Fernandez, S. Self-assembling supramolecular dendrimer nanosystem for PET imaging of tumors. Proc. Natl. Acad. Sci. USA 2018, 115, 11454–11459. [Google Scholar] [CrossRef] [Green Version]
- Narendra; Mehata, A.K.; Viswanadh, M.K.; Sonkar, R.; Pawde, D.M.; Priya, V.; Singh, M.; Koch, B.; Muthu, S.M. Formulation and in vitro evaluation of upconversion nanoparticle-loaded liposomes for brain cancer. Ther. Deliv. 2020, 11, 557–571. [Google Scholar] [CrossRef]
- Ren, F.; Liu, H.; Zhang, H.; Jiang, Z.; Xia, B.; Genevois, C.; He, T.; Allix, M.; Sun, Q.; Li, Z. Engineering NIR-IIb fluorescence of Er-based lanthanide nanoparticles for through-skull targeted imaging and imaging-guided surgery of orthotopic glioma. Nano Today 2020, 34, 100905. [Google Scholar] [CrossRef]
- Wang, X.; Tu, M.; Tian, B.; Yi, Y.; Wei, Z.; Wei, F. Synthesis of tumor-targeted folate conjugated fluorescent magnetic albumin nanoparticles for enhanced intracellular dual-modal imaging into human brain tumor cells. Anal. Biochem. 2016, 512, 8–17. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Yang, L.; Zhang, H.; Tian, B.; Li, R.; Hou, X.; Wei, F. Fluorescent magnetic PEI-PLGA nanoparticles loaded with paclitaxel for concurrent cell imaging, enhanced apoptosis and autophagy in human brain cancer. Colloids Surf. B Biointerfaces 2018, 172, 708–717. [Google Scholar] [CrossRef] [PubMed]
- Du, C.; Liu, X.; Hu, H.; Li, H.; Yu, L.; Geng, D.; Chen, Y.; Zhang, J. Dual-targeting and excretable ultrasmall SPIONs for T 1-weighted positive MR imaging of intracranial glioblastoma cells by targeting the lipoprotein receptor-related protein. J. Mater. Chem. B 2020, 8, 2296–2306. [Google Scholar] [CrossRef] [PubMed]
- Bagheri, S.; Yasemi, M.; Safaie-Qamsari, E.; Rashidiani, J.; Abkar, M.; Hassani, M.; Mirhosseini, S.A.; Kooshki, H. Using gold nanoparticles in diagnosis and treatment of melanoma cancer. Artif. Cells Nanomed. Biotechnol. 2018, 46, 462–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perry, H.L.; Botnar, R.M.; Wilton-Ely, J.D. Gold nanomaterials functionalised with gadolinium chelates and their application in multimodal imaging and therapy. Chem. Commun. 2020, 56, 4037–4046. [Google Scholar] [CrossRef]
- Smilowitz, H.M.; Meyers, A.; Rahman, K.; Dyment, N.A.; Sasso, D.; Xue, C.; Oliver, D.L.; Lichtler, A.; Deng, X.; Ridwan, S.M. Intravenously-injected gold nanoparticles (AuNPs) access intracerebral F98 rat gliomas better than AuNPs infused directly into the tumor site by convection enhanced delivery. Int. J. Nanomed. 2018, 13, 3937. [Google Scholar] [CrossRef] [Green Version]
- Zhao, L.; Li, Y.; Zhu, J.; Sun, N.; Song, N.; Xing, Y.; Huang, H.; Zhao, J. Chlorotoxin peptide-functionalized polyethylenimine-entrapped gold nanoparticles for glioma SPECT/CT imaging and radionuclide therapy. J. Nanobiotechnol. 2019, 17, 30. [Google Scholar] [CrossRef] [Green Version]
- Meola, A.; Rao, J.; Chaudhary, N.; Sharma, M.; Chang, S.D. Gold nanoparticles for brain tumor imaging: A systematic review. Front. Neurol. 2018, 9, 328. [Google Scholar] [CrossRef]
- Cho, J.-H.; Kim, A.-R.; Kim, S.-H.; Lee, S.-J.; Chung, H.; Yoon, M.-Y. Development of a novel imaging agent using peptide-coated gold nanoparticles toward brain glioma stem cell marker CD133. Acta Biomater. 2017, 47, 182–192. [Google Scholar] [CrossRef]
- Lee, C.; Kim, G.R.; Yoon, J.; Kim, S.E.; Yoo, J.S.; Piao, Y. In vivo delineation of glioblastoma by targeting tumor-associated macrophages with near-infrared fluorescent silica coated iron oxide nanoparticles in orthotopic xenografts for surgical guidance. Sci. Rep. 2018, 8, 11122. [Google Scholar] [CrossRef]
- Tamba, B.; Streinu, V.; Foltea, G.; Neagu, A.; Dodi, G.; Zlei, M.; Tijani, A.; Stefanescu, C. Tailored surface silica nanoparticles for blood-brain barrier penetration: Preparation and in vivo investigation. Arab. J. Chem. 2018, 11, 981–990. [Google Scholar] [CrossRef]
- Ding, F.; Chen, Z.; Kim, W.Y.; Sharma, A.; Li, C.; Ouyang, Q.; Zhu, H.; Yang, G.; Sun, Y.; Kim, J.S. A nano-cocktail of an NIR-II emissive fluorophore and organoplatinum (ii) metallacycle for efficient cancer imaging and therapy. Chem. Sci. 2019, 10, 7023–7028. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McHugh, K.J.; Jing, L.; Behrens, A.M.; Jayawardena, S.; Tang, W.; Gao, M.; Langer, R.; Jaklenec, A. Biocompatible semiconductor quantum dots as cancer imaging agents. Adv. Mater. 2018, 30, 1706356. [Google Scholar] [CrossRef] [PubMed]
- Huang, N.; Cheng, S.; Zhang, X.; Tian, Q.; Pi, J.; Tang, J.; Huang, Q.; Wang, F.; Chen, J.; Xie, Z. Efficacy of NGR peptide-modified PEGylated quantum dots for crossing the blood–brain barrier and targeted fluorescence imaging of glioma and tumor vasculature. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 83–93. [Google Scholar] [CrossRef] [PubMed]
- Chan, M.H.; Liu, R.S.; Hsiao, M. Light/Ultrasound Responsive 750 nm-Emitted Non-toxic Indium Phosphide Quantum Dots Hybrid Nanobubble for Brain Tumor Imaging. FASEB J. 2019, 33, 662–666. [Google Scholar]
- Tang, J.; Huang, N.; Zhang, X.; Zhou, T.; Tan, Y.; Pi, J.; Pi, L.; Cheng, S.; Zheng, H.; Cheng, Y. Aptamer-conjugated PEGylated quantum dots targeting epidermal growth factor receptor variant III for fluorescence imaging of glioma. Int. J. Nanomed. 2017, 12, 3899. [Google Scholar] [CrossRef] [Green Version]
- Madhankumar, A.; Mrowczynski, O.D.; Patel, S.R.; Weston, C.L.; Zacharia, B.E.; Glantz, M.J.; Siedlecki, C.A.; Xu, L.-C.; Connor, J.R. Interleukin-13 conjugated quantum dots for identification of glioma initiating cells and their extracellular vesicles. Acta Biomater. 2017, 58, 205–213. [Google Scholar] [CrossRef]
- Carvalho, I.C.; Mansur, A.A.; Carvalho, S.M.; Florentino, R.M.; Mansur, H.S. L-cysteine and poly-L-arginine grafted carboxymethyl cellulose/Ag-In-S quantum dot fluorescent nanohybrids for in vitro bioimaging of brain cancer cells. Int. J. Biol. Macromol. 2019, 133, 739–753. [Google Scholar] [CrossRef]
- Perini, G.; Palmieri, V.; Ciasca, G.; De Spirito, M.; Papi, M. Unravelling the potential of graphene quantum dots in biomedicine and neuroscience. Int. J. Mol. Sci. 2020, 21, 3712. [Google Scholar] [CrossRef]
- Ge, J.; Lan, M.; Zhou, B.; Liu, W.; Guo, L.; Wang, H.; Jia, Q.; Niu, G.; Huang, X.; Zhou, H.; et al. A graphene quantum dot photodynamic therapy agent with high singlet oxygen generation. Nat. Commun. 2014, 5, 4596. [Google Scholar] [CrossRef] [Green Version]
- Jovanović, S.P.; Syrgiannis, Z.; Marković, Z.M.; Bonasera, A.; Kepić, D.P.; Budimir, M.D.; Milivojević, D.D.; Spasojević, V.D.; Dramićanin, M.D.; Pavlović, V.B.; et al. Modification of structural and luminescence properties of graphene quantum dots by gamma irradiation and their application in a photodynamic therapy. Acs Appl. Mater. Interfaces 2015, 7, 25865–25874. [Google Scholar] [CrossRef] [PubMed]
- Shao, J.; Zhang, J.; Jiang, C.; Lin, J.; Huang, P. Biodegradable titanium nitride MXene quantum dots for cancer phototheranostics in NIR-I/II biowindows. Chem. Eng. J. 2020, 400, 126009. [Google Scholar] [CrossRef]
- Liu, Y.; Liu, J.; Zhang, J.; Li, X.; Lin, F.; Zhou, N.; Yang, B.; Lu, L. Noninvasive Brain Tumor Imaging Using Red Emissive Carbonized Polymer Dots across the Blood–Brain Barrier. ACS Omega 2018, 3, 7888–7896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, B.; Sheng, Z.; Hu, D.; Liu, C.; Zheng, H.; Liu, B. Through scalp and skull NIR-II photothermal therapy of deep orthotopic brain tumors with precise photoacoustic imaging guidance. Adv. Mater. 2018, 30, 1802591. [Google Scholar] [CrossRef]
- Liu, Y.; Liu, J.; Chen, D.; Wang, X.; Zhang, Z.; Yang, Y.; Jiang, L.; Qi, W.; Ye, Z.; He, S. Fluorination Enhances NIR-II Fluorescence of Polymer Dots for Quantitative Brain Tumor Imaging. Angew. Chem. Int. Ed. 2020, 59, 21049–21057. [Google Scholar] [CrossRef]
- Suárez-García, S.; Arias-Ramos, N.; Frias, C.; Candiota, A.P.; Arús, C.; Lorenzo, J.; Ruiz-Molina, D.; Novio, F. Dual T 1/T 2 Nanoscale Coordination Polymers as Novel Contrast Agents for MRI: A Preclinical Study for Brain Tumor. ACS Appl. Mater. Interfaces 2018, 10, 38819–38832. [Google Scholar] [CrossRef]
- Huang, X.; Deng, G.; Liao, L.; Zhang, W.; Guan, G.; Zhou, F.; Xiao, Z.; Zou, R.; Wang, Q.; Hu, J. CuCo 2 S 4 nanocrystals: A new platform for multimodal imaging guided photothermal therapy. Nanoscale 2017, 9, 2626–2632. [Google Scholar] [CrossRef]
- Ho, Y.N.; Shu, L.J.; Yang, Y.L. Imaging mass spectrometry for metabolites: Technical progress, multimodal imaging, and biological interactions. Wiley Interdiscip. Rev. Syst. Biol. Med. 2017, 9, e1387. [Google Scholar] [CrossRef]
- Mathiyazhakan, M.; Wiraja, C.; Xu, C. A concise review of gold nanoparticles-based photo-responsive liposomes for controlled drug delivery. Nano-Micro Lett. 2018, 10, 10. [Google Scholar] [CrossRef] [Green Version]
- Duan, Y.; Hu, D.; Guo, B.; Shi, Q.; Wu, M.; Xu, S.; Liu, X.; Jiang, J.; Sheng, Z.; Zheng, H. Nanostructural Control Enables Optimized Photoacoustic–Fluorescence–Magnetic Resonance Multimodal Imaging and Photothermal Therapy of Brain Tumor. Adv. Funct. Mater. 2020, 30, 1907077. [Google Scholar] [CrossRef]
- Villa, C.; Campione, M.; Santiago-González, B.; Alessandrini, F.; Erratico, S.; Zucca, I.; Bruzzone, M.G.; Forzenigo, L.; Malatesta, P.; Mauri, M. Self-assembled pH-sensitive fluoromagnetic nanotubes as archetype system for multimodal imaging of brain cancer. Adv. Funct. Mater. 2018, 28, 1707582. [Google Scholar] [CrossRef]
- Liu, X.; Du, C.; Li, H.; Jiang, T.; Luo, Z.; Pang, Z.; Geng, D.; Zhang, J. Engineered superparamagnetic iron oxide nanoparticles (SPIONs) for dual-modality imaging of intracranial glioblastoma via EGFRvIII targeting. Beilstein J. Nanotechnol. 2019, 10, 1860–1872. [Google Scholar] [CrossRef] [PubMed]
- Yang, Z.; Du, Y.; Sun, Q.; Peng, Y.; Wang, R.; Zhou, Y.; Wang, Y.; Zhang, C.; Qi, X. Albumin-Based Nanotheranostic Probe with Hypoxia Alleviating Potentiates Synchronous Multimodal Imaging and Phototherapy for Glioma. ACS Nano 2020, 14, 6191–6212. [Google Scholar] [CrossRef] [PubMed]
- Hallal, S.; Ebrahimkhani, S.; Shivalingam, B.; Graeber, M.B.; Kaufman, K.L.; Buckland, M.E. The emerging clinical potential of circulating extracellular vesicles for non-invasive glioma diagnosis and disease monitoring. Brain Tumor Pathol. 2019, 36, 29–39. [Google Scholar] [CrossRef]
- Wang, H.; Jiang, D.; Li, W.; Xiang, X.; Zhao, J.; Yu, B.; Wang, C.; He, Z.; Zhu, L.; Yang, Y. Evaluation of serum extracellular vesicles as noninvasive diagnostic markers of glioma. Theranostics 2019, 9, 5347. [Google Scholar] [CrossRef]
- Dumontel, B.; Susa, F.; Limongi, T.; Canta, M.; Racca, L.; Chiodoni, A.; Garino, N.; Chiabotto, G.; Centomo, M.L.; Pignochino, Y. ZnO nanocrystals shuttled by extracellular vesicles as effective Trojan nano-horses against cancer cells. Nanomedicine 2019, 14, 2815–2833. [Google Scholar] [CrossRef] [Green Version]
- Rufino-Ramos, D.; Albuquerque, P.R.; Carmona, V.; Perfeito, R.; Nobre, R.J.; de Almeida, L.P. Extracellular vesicles: Novel promising delivery systems for therapy of brain diseases. J. Control. Release 2017, 262, 247–258. [Google Scholar] [CrossRef]
- Jia, G.; Han, Y.; An, Y.; Ding, Y.; He, C.; Wang, X.; Tang, Q. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials 2018, 178, 302–316. [Google Scholar] [CrossRef]
- Kim, S.M.; Jeong, C.H.; Woo, J.S.; Ryu, C.H.; Lee, J.-H.; Jeun, S.-S. In vivo near-infrared imaging for the tracking of systemically delivered mesenchymal stem cells: Tropism for brain tumors and biodistribution. Int. J. Nanomed. 2016, 11, 13. [Google Scholar] [CrossRef] [Green Version]
- Kalimuthu, S.; Oh, J.M.; Gangadaran, P.; Zhu, L.; Lee, H.W.; Rajendran, R.L.; Jeon, Y.H.; Jeong, S.Y.; Lee, S.-W.; Lee, J. In vivo tracking of chemokine receptor CXCR4-engineered mesenchymal stem cell migration by optical molecular imaging. Stem Cells Int. 2017, 2017. [Google Scholar] [CrossRef]
- Pavon, L.F.; Sibov, T.T.; de Souza, A.V.; da Cruz, E.F.; Malheiros, S.M.; Cabral, F.R.; de Souza, J.G.; Boufleur, P.; de Oliveira, D.M.; de Toledo, S.R.C. Tropism of mesenchymal stem cell toward CD133+ stem cell of glioblastoma in vitro and promote tumor proliferation in vivo. Stem Cell Res. Ther. 2018, 9, 310. [Google Scholar] [CrossRef] [PubMed]
- Qiao, Y.; Gumin, J.; MacLellan, C.J.; Gao, F.; Bouchard, R.; Lang, F.F.; Stafford, R.J.; Melancon, M.P. Magnetic resonance and photoacoustic imaging of brain tumor mediated by mesenchymal stem cell labeled with multifunctional nanoparticle introduced via carotid artery injection. Nanotechnology 2018, 29, 165101. [Google Scholar] [CrossRef] [PubMed]
- Lim, S.; Yoon, H.Y.; Jang, H.J.; Song, S.; Kim, W.; Park, J.; Lee, K.E.; Jeon, S.; Lee, S.; Lim, D.-K. Dual-Modal Imaging-Guided Precise Tracking of Bioorthogonally Labeled Mesenchymal Stem Cells in Mouse Brain Stroke. ACS Nano 2019, 13, 10991–11007. [Google Scholar] [CrossRef] [PubMed]
- Hao, X.; Xu, B.; Chen, H.; Wang, X.; Zhang, J.; Guo, R.; Shi, X.; Cao, X. Stem cell-mediated delivery of nanogels loaded with ultrasmall iron oxide nanoparticles for enhanced tumor MR imaging. Nanoscale 2019, 11, 4904–4910. [Google Scholar] [CrossRef] [PubMed]
- Wu, S.Q.; Yang, C.X.; Yan, X.P. A Dual-Functional Persistently Luminescent Nanocomposite Enables Engineering of Mesenchymal Stem Cells for Homing and Gene Therapy of Glioblastoma. Adv. Funct. Mater. 2017, 27, 1604992. [Google Scholar] [CrossRef]
- Hsu, F.-T.; Wei, Z.-H.; Hsuan, Y.C.-Y.; Lin, W.; Su, Y.-C.; Liao, C.-H.; Hsieh, C.-L. MRI tracking of polyethylene glycol-coated superparamagnetic iron oxide-labelled placenta-derived mesenchymal stem cells toward glioblastoma stem-like cells in a mouse model. Artif. Cells Nanomed. Biotechnol. 2018, 46, S448–S459. [Google Scholar] [CrossRef]
- Duan, Y.; Wu, M.; Hu, D.; Pan, Y.; Hu, F.; Liu, X.; Thakor, N.; Ng, W.H.; Liu, X.; Sheng, Z. Biomimetic Nanocomposites Cloaked with Bioorthogonally Labeled Glioblastoma Cell Membrane for Targeted Multimodal Imaging of Brain Tumors. Adv. Funct. Mater. 2020, 30, 2004346. [Google Scholar] [CrossRef]
- Wang, C.; Wu, B.; Wu, Y.; Song, X.; Zhang, S.; Liu, Z. Camouflaging Nanoparticles with Brain Metastatic Tumor Cell Membranes: A New Strategy to Traverse Blood–Brain Barrier for Imaging and Therapy of Brain Tumors. Adv. Funct. Mater. 2020, 30, 1909369. [Google Scholar] [CrossRef]
- Jia, Y.; Wang, X.; Hu, D.; Wang, P.; Liu, Q.; Zhang, X.; Jiang, J.; Liu, X.; Sheng, Z.; Liu, B. Phototheranostics: Active targeting of orthotopic glioma using biomimetic proteolipid nanoparticles. ACS Nano 2018, 13, 386–398. [Google Scholar] [CrossRef]
- Grosu, F.; Ungureanu, A.; Bianchi, E.; Moscu, B.; Coldea, L.; Stupariu, A.L.; Pirici, I.; Roman-Filip, C.C. Multifocal and multicentric low-grade oligoastrocytoma in a young patient. Rom. J. Morphol. Embryol. 2017, 58, 207–210. [Google Scholar]
- Wang, X.; Hu, Y.; Wang, R.; Zhao, P.; Gu, W.; Ye, L. Albumin-mediated synthesis of fluoroperovskite KMnF3 nanocrystals for T1-T2 dual-modal magnetic resonance imaging of brain gliomas with improved sensitivity. Chem. Eng. J. 2020, 395, 125066. [Google Scholar] [CrossRef]
Imaging Technique | Selection Parameters Based on Characteristics | Nanostructures Used | Ref. |
---|---|---|---|
Surface-enhanced resonance Raman scattering imaging (SERRS) | High specificity and provides data about the location of biochemical components of cells. | SERRS NPs with 68Ga comprised of gold core and silica shell | [147,148] |
Magnetic resonance imaging (MRI) | Sensitive to changes in the cartilage and bone and provides an elaboration of the anatomical structure of the brain with high soft-tissue contrast. | Iron oxide NPs’ surface decorated with peptides; gadolinium oxide-based NPs | [149,150] |
Photoacoustic (PA) imaging | Acquires molecular data with high resolution in real-time and can be used simultaneously with other imaging techniques. | Silicon quantum sheets, molybdenum di-sulfide nanosheets conjugated with indocyanine green | [151,152] |
Fluorescence (FL) imaging | Non-invasive with low spatial resolution. | Gold NPs | [153,154,155] |
Focused ultrasound (FUS) | Real-time visualization of neural anatomy with 3D contrast-enhanced images. | Cisplatin gold NP conjugates, mesoporous organo-silica NPs | [156,157] |
Multimodal imaging | Possibility to map cell density to understand heterogeneity of the tissues; high sensitivity and specificity. | SERRS-MSOT*-nanostar with gold core embedded in silica coat functionalized with PEG, SERRS-MRI gold nanoprobes | [158,159] |
Positron emission tomography (PET) | Nuclear imaging technique to identify pathophysiological changes in the brain; unlimited penetration. | Alanine modified gado-fullerene NPs, self-assembled amphiphilic dendrimer nano-system | [160,161] |
Computed tomography (CT) | Able to provide electron density differences among tissues to establish diagnosis. | Transferrin conjugated liposome, Lanthanide NPs | [162,163] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mukhtar, M.; Bilal, M.; Rahdar, A.; Barani, M.; Arshad, R.; Behl, T.; Brisc, C.; Banica, F.; Bungau, S. Nanomaterials for Diagnosis and Treatment of Brain Cancer: Recent Updates. Chemosensors 2020, 8, 117. https://doi.org/10.3390/chemosensors8040117
Mukhtar M, Bilal M, Rahdar A, Barani M, Arshad R, Behl T, Brisc C, Banica F, Bungau S. Nanomaterials for Diagnosis and Treatment of Brain Cancer: Recent Updates. Chemosensors. 2020; 8(4):117. https://doi.org/10.3390/chemosensors8040117
Chicago/Turabian StyleMukhtar, Mahwash, Muhammad Bilal, Abbas Rahdar, Mahmood Barani, Rabia Arshad, Tapan Behl, Ciprian Brisc, Florin Banica, and Simona Bungau. 2020. "Nanomaterials for Diagnosis and Treatment of Brain Cancer: Recent Updates" Chemosensors 8, no. 4: 117. https://doi.org/10.3390/chemosensors8040117
APA StyleMukhtar, M., Bilal, M., Rahdar, A., Barani, M., Arshad, R., Behl, T., Brisc, C., Banica, F., & Bungau, S. (2020). Nanomaterials for Diagnosis and Treatment of Brain Cancer: Recent Updates. Chemosensors, 8(4), 117. https://doi.org/10.3390/chemosensors8040117