Nanomedicine, which is blazing new trails in modern healthcare, exploits the distinctive attributes of nanoparticles (NPs) to reimagine diagnosis and therapy. With imaging, this “magic ammunition” enhances the capability of MRI, a noninvasive, high-resolution technology that can pinpoint cancers with astonishing precision. Unlike traditional treatments, which muddle the distinction between diseased and healthy tissue, gold nanoparticles (AuNPs) directly target tumors, using increased permeability and retention (EPR) effects for crystal-clear, early-stage detection. While gadolinium (Gd (III)) T1 agents are the most widely used in clinical practice, iron oxide T2 agents have fallen out of favor due to failing to perform. Beyond imaging, NPs offer a “one-two punch” in the treatment of complicated disorders by enabling photothermal (PTT), photodynamic (PDT), and multimodal therapies (MMT), as well as nano drug delivery systems (NDDS). This nanotechnology-based technique personalizes medicines for diseases such as tuberculosis (TB) and cardiovascular disease (CVD), focusing on damaged tissues to improve therapeutic response. In tuberculosis, NPs circumvent medication resistance by delivering medicines directly to infected cells, thus avoiding biological barriers that impede standard treatments. Similarly, in CVD, NPs target arterial plaques to improve treatment accuracy while reducing systemic adverse effects. Furthermore, the genomic “blueprint” is evolving parallel to nanotechnology, with India's “Genome India” initiative mapping 10,000 genomes, lighting the path for genetic chips suited for specific disorders and moment tests. The convergence of nanotechnology and genomes is an arcade modification, bringing accuracy and personalization to the forefront of cancer and tuberculosis treatment.
Citation: Dinesh Bhatia, Tania Acharjee, Shruti Shukla, Monika Bhatia. Nano-technological advancements in multimodal diagnosis and treatment[J]. AIMS Biophysics, 2024, 11(4): 464-507. doi: 10.3934/biophy.2024026
Nanomedicine, which is blazing new trails in modern healthcare, exploits the distinctive attributes of nanoparticles (NPs) to reimagine diagnosis and therapy. With imaging, this “magic ammunition” enhances the capability of MRI, a noninvasive, high-resolution technology that can pinpoint cancers with astonishing precision. Unlike traditional treatments, which muddle the distinction between diseased and healthy tissue, gold nanoparticles (AuNPs) directly target tumors, using increased permeability and retention (EPR) effects for crystal-clear, early-stage detection. While gadolinium (Gd (III)) T1 agents are the most widely used in clinical practice, iron oxide T2 agents have fallen out of favor due to failing to perform. Beyond imaging, NPs offer a “one-two punch” in the treatment of complicated disorders by enabling photothermal (PTT), photodynamic (PDT), and multimodal therapies (MMT), as well as nano drug delivery systems (NDDS). This nanotechnology-based technique personalizes medicines for diseases such as tuberculosis (TB) and cardiovascular disease (CVD), focusing on damaged tissues to improve therapeutic response. In tuberculosis, NPs circumvent medication resistance by delivering medicines directly to infected cells, thus avoiding biological barriers that impede standard treatments. Similarly, in CVD, NPs target arterial plaques to improve treatment accuracy while reducing systemic adverse effects. Furthermore, the genomic “blueprint” is evolving parallel to nanotechnology, with India's “Genome India” initiative mapping 10,000 genomes, lighting the path for genetic chips suited for specific disorders and moment tests. The convergence of nanotechnology and genomes is an arcade modification, bringing accuracy and personalization to the forefront of cancer and tuberculosis treatment.
[1] | Colvin VL (2003) The potential environmental impact of engineered nanomaterials. Nat Biotechnol 21: 1166-1170. https://doi.org/10.1038/nbt875 |
[2] | Boisseau P, Loubaton B (2011) Nanomedicine, nanotechnology in medicine. CR Phys 12: 620-636. https://doi.org/10.1016/j.crhy.2011.06.001 |
[3] | Dessale M, Mengistu G, Mengist HM (2022) Nanotechnology: a promising approach for cancer diagnosis, therapeutics and theragnosis. Int J Nanomed 17: 3735-3749. https://doi.org/10.2147/ijn.s378074 |
[4] | Bi Y, Liu Y, Zhang X, et al. (2019) Ultrathin metal films as the transparent electrode in ITO-free organic optoelectronic devices. Adv Opt Mater 7: 1800778. https://doi.org/10.1002/adom.201800778 |
[5] | Tessonnier J, Rosenthal D, Hansen TW, et al. (2009) Analysis of the structure and chemical properties of some commercial carbon nanostructures. Carbon 47: 1779-1798. https://doi.org/10.1016/j.carbon.2009.02.032 |
[6] | Zhang B, Kwok CT, Cheng FT, et al. (2011) Fabrication of nano-structured HA/CNT coatings on Ti6Al4V by electrophoretic deposition for biomedical applications. J Nanosci Nanotechno 11: 10740-10745. https://doi.org/10.1166/jnn.2011.3939 |
[7] | Scott RWJ, Wilson OM, Crooks RM (2004) Synthesis, characterization, and applications of dendrimer-encapsulated nanoparticles. J Phys Chem B 109: 692-704. https://doi.org/10.1021/jp0469665 |
[8] | Thakur S, Kesharwani P, Tekade RK, et al. (2015) Impact of pegylation on biopharmaceutical properties of dendrimers. Polymer 59: 67-92. https://doi.org/10.1016/j.polymer.2014.12.051 |
[9] | Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al. (2013) Liposome: classification, preparation, and applications. Nanoscale Res Lett 8: 102. https://doi.org/10.1186/1556-276x-8-102 |
[10] | Zong T, Mei L, Gao H, et al. (2014) Synergistic dual-ligand doxorubicin liposomes improve targeting and therapeutic efficacy of brain glioma in animals. Mol Pharmaceutics 11: 2346-2357. https://doi.org/10.1021/mp500057n |
[11] | Sultana A, Zare M, Thomas V, et al. (2022) Nano-based drug delivery systems: conventional drug delivery routes, recent developments and future prospects. Med Drug Discov 15: 100134. https://doi.org/10.1016/j.medidd.2022.100134 |
[12] | Jou AP Biointerfaces based on the Combination of Synthetic Polymers and Biomolecules (2019). https://doi.org/10.5821/dissertation-2117-177241 |
[13] | Kwon GS, Okano T (1996) Polymeric micelles as new drug carriers. Adv Drug Deliver Rev 21: 107-116. https://doi.org/10.1016/s0169-409x(96)00401-2 |
[14] | Lindauer M, Hochhaus A (2018) Dasatinib. Small Molecules in Hematology : 29-68. https://doi.org/10.1007/978-3-319-91439-8_2 |
[15] | Henrique Lima T, Fernandes-Cunha GM, Jensen CE, et al. (2016) Bioactive glass nanoparticles-loaded poly(ɛ-caprolactone) nanofiber as substrate for ARPE-19 cells. J Nanomater 2016: 4360659. https://doi.org/10.1155/2016/4360659 |
[16] | Di Tommaso C., Bourges, et al. (2012) Novel micelle carriers for cyclosporine A topical ocular delivery: in vivo cornea penetration, ocular distribution and efficacy studies. Eur J Pharm Biopharm 81: 257-264. https://doi.org/10.1016/j.ejpb.2012.02.014 |
[17] | Masters BR (2007) Ernst abbe and the foundation of scientific microscopes. Opt Photonics News 18: 18-23. https://doi.org/10.1364/opn.18.2.000018 |
[18] | Morita S, Sugawara YSY, Fukano YFY (1993) Atomic force microscope combined with scanning tunneling microscope [AFM/STM]. Jpn J Appl Phys 32: 2983. https://doi.org/10.1143/jjap.32.2983 |
[19] | Yong YC, Wang YZ, Zhong JJ (2018) Nano-spectroscopic imaging of proteins with near-field scanning optical microscopy (NSOM). Curr Opin Biotechnol 54: 106-113. https://doi.org/10.1016/j.copbio.2018.01.022 |
[20] | Ishikawa T, Aoyagi H, Asaka T, et al. (2012) A compact X-ray free-electron laser emitting in the sub-ångström region. Nat Photonics 6: 540-544. https://doi.org/10.1038/nphoton.2012.141 |
[21] | Le Gros MA, Clowney EJ, Magklara A, et al. (2016) Soft X-ray tomography reveals gradual chromatin compaction and reorganization during neurogenesis in vivo. Cell Rep 17: 2125-2136. https://doi.org/10.1016/j.celrep.2016.10.060 |
[22] | Sorrentino A, Nicolás J, Valcárcel R, et al. (2015) MISTRAL: a transmission soft X-ray microscopy beamline for cryo nano-tomography of biological samples and magnetic domains imaging. J Synchrotron Radiat 22: 1112-1117. https://doi.org/10.1107/s1600577515008632 |
[23] | Chaurand P, Schwartz SA, Billheimer D, et al. (2004) Integrating histology and imaging mass spectrometry. Anal Chem 76: 1145-1155. https://doi.org/10.1021/ac0351264 |
[24] | Michalski A, Damoc E, Hauschild JP, et al. (2011) Mass spectrometry-based proteomics using Q exactive, a high-performance benchtop quadrupole orbitrap mass spectrometer. Mol Cell Proteomics 10: M111.011015. https://doi.org/10.1074/mcp.M111.011015 |
[25] | Guerquin-Kern JL, Wu TD, Quintana C, et al. (2005) Progress in analytical imaging of the cell by dynamic secondary ion mass spectrometry (SIMS microscopy). BBA-Gen Subjects 1724: 228-238. https://doi.org/10.1016/j.bbagen.2005.05.013 |
[26] | Spraggins JM, Djambazova KV, Rivera ES, et al. (2019) High-performance molecular imaging with MALDI trapped ion-mobility time-of-flight (timsTOF) mass spectrometry. Anal Chem 91: 14552-14560. https://doi.org/10.1021/acs.analchem.9b03612 |
[27] | Milne JL, Borgnia MJ, Bartesaghi A, et al. (2012) Cryo-electron microscopy—a primer for the non-microscopist. FEBS J 280: 28-45. https://doi.org/10.1111/febs.12078 |
[28] | Wan NY, Chiu NW, Zhou Z (2004) Full contrast transfer function correction in 3D cryo-EM reconstruction. 2004 International Conference on Communications, Circuits and Systems . https://doi.org/10.1109/icccas.2004.1346339 |
[29] | Cryo-electron microscopy: small electrons to visualize large molecules (2020). Available from: https://www.unisr.it/en/news/2020/6/criomicroscopia-elettronica-piccoli-elettroni-per-visualizzare-grandi-molecole |
[30] | Elbaum M, Seifer S, Houben L, et al. (2021) Toward compositional contrast by cryo-STEM. Acc Chem Res 54: 3621-3631. https://doi.org/10.1021/acs.accounts.1c00279 |
[31] | Bhella D (2019) Cryo-electron microscopy: an introduction to the technique, and considerations when working to establish a national facility. Biophys Rev 11: 515-519. https://doi.org/10.1007/s12551-019-00571-w |
[32] | Kowalewska B, Drozdz W, Kowalewski L (2021) Positron emission tomography (PET) and single-photon emission computed tomography (SPECT) in autism research: literature review. Irish J Psychol Med 39: 272-286. https://doi.org/10.1007/s12551-019-00571-w |
[33] | Tsoukalas C, Psimadas D, Kastis GA, et al. (2018) A novel metal-based imaging probe for targeted dual-modality SPECT/MR imaging of angiogenesis. Front Chem 6: 224. https://doi.org/10.3389/fchem.2018.00224 |
[34] | Zhang Y, Hong H, Engle JW, et al. (2011) Positron emission tomography imaging of CD105 expression with a 64Cu-labeled monoclonal antibody: NOTA is superior to DOTA. Plos One 6: e28005. https://doi.org/10.1371/journal.pone.0028005 |
[35] | Chen F, Nayak TR, Goel S, et al. (2014) In vivo tumor vasculature targeted PET/NIRF imaging with TRC105(Fab)-conjugated, dual-labeled mesoporous silica nanoparticles. Mol Pharmaceutics 11: 4007-4014. https://doi.org/10.1021/mp500306k |
[36] | Madhumathy P, Pandey D (2022) Deep learning based photo acoustic imaging for non-invasive imaging. Multimed Tools Appl 81: 7501-7518. https://doi.org/10.1007/s11042-022-11903-6 |
[37] | Liu Y, Zhen W, Wang Y, et al. (2019) One-dimensional Fe2P acts as a Fenton agent in response to NIR II light and ultrasound for deep tumor synergetic theranostics. Angew Chem 58: 2407-2412. https://doi.org/10.1002/anie.201813702 |
[38] | Wang LV (2009) Multiscale photoacoustic microscopy and computed tomography. Nat Photonics 3: 503-509. https://doi.org/10.1038/nphoton.2009.157 |
[39] | Ando T, Xuan W, Xu T, et al. (2011) Comparison of therapeutic effects between pulsed and continuous wave 810-nm wavelength laser irradiation for traumatic brain injury in mice. Plos One 6: e26212. https://doi.org/10.1371/journal.pone.0026212 |
[40] | Yao DK, Maslov K, Shung KK, et al. (2010) In vivo label-free photoacoustic microscopy of cell nuclei by excitation of DNA and RNA. Opt Lett 35: 4139. https://doi.org/10.1364/ol.35.004139 |
[41] | Bücking TM, Van Den Berg PJ, Balabani S (2018) Processing methods for photoacoustic Doppler flowmetry with a clinical ultrasound scanner. J Biomed Opt 23: 026009. https://doi.org/10.1117/1.jbo.23.2.026009 |
[42] | Bêche B, Pelletier N, Gaviot E, et al. (2004) Single-mode TE00–TM00 optical waveguides on SU-8 polymer. Optics Commun 230: 91-94. https://doi.org/10.1016/j.optcom.2003.11.016 |
[43] | Kim J, Park S, Jung Y, et al. (2016) Programmable real-time clinical photoacoustic and ultrasound imaging system. Sci Rep 6: 35137. https://doi.org/10.1038/srep35137 |
[44] | Weber M, Khan TA, Patalag LJ, et al. (2020) Photoactivatable fluorophore for stimulated emission depletion (STED) microscopy and bioconjugation technique for hydrophobic labels. Chemistry 27: 451-458. https://doi.org/10.1002/chem.202004645 |
[45] | Tortarolo G, Sun Y, Teng KW, et al. (2019) Photon-separation to enhance the spatial resolution of pulsed STED microscopy. Nanoscale 11: 1754-1761. https://doi.org/10.1039/c8nr07485b |
[46] | Sednev MV, Belov VN, Hell SW (2015) Fluorescent dyes with large Stokes shifts for super-resolution optical microscopy of biological objects: a review. Methods Appl Fluores 3: 042004. https://doi.org/10.1002/ejoc.202000093 |
[47] | Bachman JL, Pavlich CI, Boley AJ, et al. (2019) Synthesis of carboxy ATTO 647N using redox cycling for xanthone access. Org Lett 22: 381-385. https://doi.org/10.1021/acs.orglett.9b03981 |
[48] | Yazdi AA, Popma A, Wong W, et al. (2016) 3D printing: an emerging tool for novel microfluidics and lab-on-a-chip applications. Microfluid Nanofluid 20: 50. https://doi.org/10.1007/s10404-016-1715-4 |
[49] | Berlanda SF, Breitfeld M, Dietsche CL, et al. (2020) Recent advances in microfluidic technology for bioanalysis and diagnostics. Anal Chem 93: 311-331. https://doi.org/10.1016/j.trac.2022.116894 |
[50] | Walsh DI, Kong DS, Murthy SK, et al. (2017) Enabling microfluidics: from clean rooms to makerspaces. Trends Biotechnol 35: 383-392. https://doi.org/10.1016/j.tibtech.2017.01.001 |
[51] | Wang Y, Zhao J, Zhu Y, et al. (2021) Monolithic integration of nanorod arrays on microfluidic chips for fast and sensitive one-step immunoassays. Microsyst Nanoeng 7: 65. https://doi.org/10.1038/s41378-021-00291-w |
[52] | Gokaltun A, Yarmush ML, Asatekin A, et al. (2017) Recent advances in nonbiofouling PDMS surface modification strategies applicable to microfluidic technology. Technology 5: 1-12. https://doi.org/10.1142/s2339547817300013 |
[53] | Ling Y (2009) Modified plastic surfaces for high performance microfluidic immunoassay devices. Nanyang Technological University, Singapore . https://doi.org/10.32657/10356/15152 |
[54] | Chinnappan R, Mir TA, Alsalameh S, et al. (2023) Emerging biosensing methods to monitor lung cancer biomarkers in biological samples: a comprehensive review. Cancers 15: 3414. https://doi.org/10.3390/cancers15133414 |
[55] | Torino S, Corrado B, Iodice M, et al. (2018) PDMS-based microfluidic devices for cell culture. Inventions 3: 65. https://doi.org/10.1016/bs.mcb.2018.09.007 |
[56] | Peppas NA, Hilt JZ, Khademhosseini A, et al. (2006) Hydrogels in biology and medicine: from molecular principles to bionanotechnology. Adv Mat 18: 1345-1360. https://doi.org/10.1002/adma.200501612 |
[57] | Shen C, Li Y, Wang Y, et al. (2019) Non-swelling hydrogel-based microfluidic chips. Lab Chip 19: 3962-3973. https://doi.org/10.1039/c9lc00564a |
[58] | Maher S, Santos A, Kumeria T, et al. (2017) Multifunctional microspherical magnetic and pH responsive carriers for combination anticancer therapy engineered by droplet-based microfluidics. J Mater Chem B 5: 4097-4109. https://doi.org/10.1039/c7tb00588a |
[59] | Lv D, Hu Z, Lu L, et al. (2017) Three-dimensional cell culture: a powerful tool in tumor research and drug discovery. Oncol Lett 14: 6999-7010. https://doi.org/10.3892/ol.2017.7134 |
[60] | Ortiz-Casas B, Galdámez-Martínez A, Gutiérrez-Flores J, et al. (2021) Bio-acceptable 0D and 1D ZnO nanostructures for cancer diagnostics and treatment. Mater Today 50: 533-569. https://doi.org/10.1016/j.mattod.2021.07.025 |
[61] | Joensson HN, Uhlen M, Svahn HA (2011) Droplet size based separation by deterministic lateral displacement-separating droplets by cell-induced shrinking. Lab Chip 11: 1305-1310. https://doi.org/10.1039/c0lc00688b |
[62] | Arruebo M, Valladares M, González-Fernández F (2009) Antibody-conjugated nanoparticles for biomedical applications. J Nanomater 2009: 1-24. https://doi.org/10.1155/2009/439389 |
[63] | Yang NC, Hu ML (2004) A fluorimetric method using fluorescein di-β-d-galactopyranoside for quantifying the senescence-associated β-galactosidase activity in human foreskin fibroblast Hs68 cells. Anal Biochem 325: 337-343. https://doi.org/10.1016/j.ab.2003.11.012 |
[64] | Leung CM, De Haan P, Ronaldson-Bouchard K, et al. (2022) A guide to the organ-on-a-chip. Nat Rev Method Prime 2: 33. https://doi.org/10.1038/s43586-022-00118-6 |
[65] | Park G., Rim, et al. (2024) Replacing animal testing with stem cell-organoids: advantages and limitations. Stem Cell Rev Rep 20: 1375-1386. https://doi.org/10.1007/s12015-024-10723-5 |
[66] | Mastrangeli M, Millet S, Van Den Eijnden-Van Raaij J (2019) Organ-on-chip in development: Towards a roadmap for organs-on-chip. ALTEX/Altern Anim Ex 36: 650-668. https://doi.org/10.14573/altex.1908271 |
[67] | Djomehri SI, Burman B, Gonzalez ME, et al. (2018) A reproducible scaffold-free 3D organoid model to study neoplastic progression in breast cancer. J Cell Commun Signal 13: 129-143. https://doi.org/10.1007/s12079-018-0498-7 |
[68] | Huh D, Matthews BD, Mammoto A, et al. (2010) Reconstituting organ-level lung functions on a chip. Science 328: 1662-1668. https://doi.org/10.1126/science.1188302 |
[69] | Jain A, Barrile R, Van Der Meer A, et al. (2017) Primary human lung alveolus-on-a-chip model of intravascular thrombosis for assessment of therapeutics. Clin Pharmacol Ther/Clin Pharmacol Ther 103: 332-340. https://doi.org/10.1002/cpt.742 |
[70] | Si L, Bai H, Rodas M, et al. (2020) Human organs-on-chips as tools for repurposing approved drugs as potential influenza and COVID19 therapeutics in viral pandemics. BioRxiv . http://dx.doi.org/10.1101/2020.04.13.039917 |
[71] | Bang S, Jeong S, Choi N, et al. (2019) Brain-on-a-chip: a history of development and future perspective. Biomicrofluidics 13: 051301. https://doi.org/10.1063/1.5120555 |
[72] | Jahromi MAM, Abdoli A, Rahmanian M, et al. (2019) Microfluidic brain-on-a-chip: perspectives for mimicking neural system disorders. Mol Neurobiol 56: 8489-8512. https://doi.org/10.1007/s12035-019-01653-2 |
[73] | Harrison RG, Greenman M, Mall FP, et al. (1907) Observations of the living developing nerve fiber. Anat Rec 1: 116-128. https://doi.org/10.1002/ar.1090010503 |
[74] | Schimek K, Frentzel S, Luettich K, et al. (2020) Human multi-organ chip co-culture of bronchial lung culture and liver spheroids for substance exposure studies. Sci Rep 10: 7865. https://doi.org/10.1038/s41598-020-64219-6 |
[75] | Morelli M, Rodríguez MC, Queiroz K (2024) A high-throughput gut-on-chip platform to study the epithelial responses to enterotoxins. Sci Rep 14: 5797. https://doi.org/10.1038/s41598-024-56520-5 |
[76] | Varala R, Kotra V, Kanuri AK, et al. (2023) Nano drug delivery-benefits, limitations and future perspective. Nano Med Mater 3: 244. https://doi.org/10.59400/nmm.v3i2.244 |
[77] | Park JH, Byun JY, Mun H, et al. (2014) A regeneratable, label-free, localized surface plasmon resonance (LSPR) aptasensor for the detection of ochratoxin A. Biosens Bioelectron 59: 321-327. https://doi.org/10.1016/j.bios.2014.03.059 |
[78] | Zhang F, Cao J, Chen X, et al. (2015) Noninvasive dynamic imaging of tumor early response to nanoparticle-mediated photothermal therapy. Theranostics 5: 1444-1455. https://doi.org/10.7150/thno.13398 |
[79] | Josefsen LB, Boyle RW (2008) Photodynamic therapy and the development of metal-based photosensitisers. Metal-based Drugs 2008: 1-23. https://doi.org/10.1155/2008/276109 |
[80] | Gutiérrez Y, Alcaraz de la Osa R, Ortiz D, et al. (2018) Plasmonics in the ultraviolet with Aluminum, Gallium, Magnesium and Rhodium. Appl Sci 8: 64. https://doi.org/10.3390/app8010064 |
[81] | Li J, Liu X, Tan L, et al. (2019) Zinc-doped Prussian blue enhances photothermal clearance of Staphylococcus aureus and promotes tissue repair in infected wounds. Nat Commun 10: 4490. https://doi.org/10.1038/s41467-019-12429-6 |
[82] | Kim G, Huang SW, Day KC, et al. (2007) Indocyanine-green-embedded PEBBLEs as a contrast agent for photoacoustic imaging. J Biomed Opt 12: 044020. https://doi.org/10.1117/1.2771530 |
[83] | Shafirstein G, Bäumler W, Hennings LJ, et al. (2011) Indocyanine green enhanced near-infrared laser treatment of murine mammary carcinoma. Int J Cancer 130: 1208-1215. https://doi.org/10.1002/ijc.26126 |
[84] | Beumer K, Bhattacharyya G, Bibikova M, et al. (2006) Efficient gene targeting in drosophila with zinc-finger nucleases. Genetics 172: 2391-2403. https://doi.org/10.1534/genetics.105.052829 |
[85] | Sood R, Carrington B, Bishop K, et al. (2013) Efficient methods for targeted mutagenesis in zebrafish using zinc-finger nucleases: data from targeting of nine genes using CompoZr or CoDA ZFNs. Plos One 8: e57239. https://doi.org/10.1371/journal.pone.0057239 |
[86] | Tan J, Zhao Y, Wang B, et al. (2020) Efficient CRISPR/Cas9-based plant genomic fragment deletions by microhomology-mediated end joining. Plant Biotechnol J 18: 2161-2163. https://doi.org/10.1111/pbi.13390 |
[87] | Biochemistry, BiochemistryCRISPR-Cas9-Based Genome Editing of Human Cells—Synthesis-Based BioFusion TechnologyLab.Synthesis-Based BioFusion Technology Lab (2019). Available from: https://biochemistry.khu.ac.kr/lab/?p=2672 |
[88] | Tuszynski MH, Thal L, Pay M, et al. (2005) A phase 1 clinical trial of nerve growth factor gene therapy for Alzheimer disease. Nat Med 11: 551. https://doi.org/10.1038/nm1239 |
[89] | Alvarez XA, Alvarez I, Iglesias O, et al. (2016) Synergistic increase of serum BDNF in Alzheimer patients treated with cerebrolysin and donepezil: association with cognitive improvement in ApoE4 cases. Int J Neuropsychopharmacol 19: pyw024. https://doi.org/10.1093/ijnp/pyw024 |
[90] | Fotinopoulou A, Tsachaki M, Vlavaki M, et al. (2005) BRI2 interacts with amyloid precursor protein (APP) and regulates amyloid β (Aβ) production. J Biol Chem 280: 30768-30772. https://doi.org/10.1074/jbc.c500231200 |
[91] | Mason C, Manzotti E (2010) Regenerative medicine cell therapies: numbers of units manufactured and patients treated between 1988 and 2010. Regen Med 5: 307-313. https://doi.org/10.2217/rme.10.37 |
[92] | Barba Serrahima A Intrinsic Osteoinduction and Osteogenesis of Biomimetic Calcium Phosphate Scaffolds with Different Nano-, Micro- and Macroporosities : ectopic and Orthotopic Implantation in a Canine Model (2023). https://doi.org/10.5821/dissertation-2117-177812 |
[93] | Gregory DA, Tripathi L, Fricker AT, et al. (2021) Bacterial cellulose: a smart biomaterial with diverse applications. Mater Sci Eng R Rep 145: 100623. https://doi.org/10.1016/j.mser.2021.100623 |
[94] | Zhang Y, Liu X, Zeng L, et al. (2019) Polymer fiber scaffolds for bone and cartilage tissue engineering. Adv Funct Mater 29: 1903279. https://doi.org/10.1002/adfm.201903279 |
[95] | Zhang X, Wang Y, Gao Z, et al. (2023) Advances in wound dressing based on electrospinning nanofibers. J Appl Polym Sci 141: e54746. https://doi.org/10.1002/app.54746 |
[96] | Wellstein A, Zugmaier G, Califano JA, et al. (1991) Tumor growth dependent on Kaposi's sarcoma-derived fibroblast growth factor inhibited by pentosan polysulfate. JNCI J Natl Cancer I 83: 716-720. https://doi.org/10.1093/jnci/83.10.716 |
[97] | Biazar E, Khorasani M, Montazeri N, et al. (2010) Types of neural guides and using nanotechnology for peripheral nerve reconstruction. Int J Nanomed 5: 839-852. https://doi.org/10.2147/IJN.S11883 |
[98] | Guaccio A, Guarino V, Perez MA, et al. (2011) Influence of electrospun fiber mesh size on hMSC oxygen metabolism in 3D collagen matrices: experimental and theoretical evidences. Biotechnol Bioeng 108: 1965-1976. https://doi.org/10.1002/bit.23113 |
[99] | Aggarwal S, Pittenger MF (2004) Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 105: 1815-1822. https://doi.org/10.1182/blood-2004-04-1559 |
[100] | Merzendorfer H, Cohen E (2019) Chitin/chitosan: versatile ecological, industrial, and biomedical applications. Extracellular Sugar-Based Biopolymers Matrices : 541-624. https://doi.org/10.1007/978-3-030-12919-4_14 |
[101] | Liu S, Yang S, Ho PC (2017) Intranasal administration of carbamazepine-loaded carboxymethyl chitosan nanoparticles for drug delivery to the brain. Asian J Pharm Sci 13: 72-81. https://doi.org/10.1016/j.ajps.2017.09.001 |
[102] | Jain SK, Puri R (2014) Development, characterization and in vivo localization study of topical 5-fluorouracil gels: a comparative study with conventional formulation. Curr Drug Deliv 11: 401-414. https://doi.org/10.2174/1567201810666131210112150 |
[103] | Patil NH, Devarajan PV (2014) Insulin-loaded alginic acid nanoparticles for sublingual delivery. Drug Deliv 23: 429-436. https://doi.org/10.3109/10717544.2014.916769 |
[104] | Pratap-Singh A, Guo Y, Baldelli A, et al. (2023) Concept for a unidirectional release mucoadhesive buccal tablet for oral delivery of antidiabetic peptide drugs such as insulin, glucagon-like peptide 1 (GLP-1), and their analogs. Pharmaceutics 15: 2265. https://doi.org/10.3390/pharmaceutics15092265 |
[105] | Haque S, Md S, Fazil M, et al. (2012) Venlafaxine loaded chitosan NPs for brain targeting: pharmacokinetic and pharmacodynamic evaluation. Carbohyd Polym 89: 72-79. https://doi.org/10.1016/j.carbpol.2012.02.051 |
[106] | Maity S, Sa B (2014) Development and evaluation of Ca+2 ion cross-linked carboxymethyl xanthan gum tablet prepared by wet granulation technique. AAPS PharmSciTech 15: 920-927. https://doi.org/10.1208/s12249-014-0123-x |
[107] | Laffleur F, Michalek M (2017) Modified xanthan gum for buccal delivery—a promising approach in treating sialorrhea. Inter J Biol Macrom 102: 1250-1256. https://doi.org/10.1016/j.ijbiomac.2017.04.123 |
[108] | Liang P Gold Nanoparticle-Based Colorimetric Sensors for Detection of DNA and Small Molecules (2016). https://doi.org/10.25148/etd.fidc000729 |
[109] | Chen W, Xu X, Jia H, et al. (2013) Therapeutic nanomedicine based on dual-intelligent functionalized gold nanoparticles for cancer imaging and therapy in vivo. Biomaterials 34: 8798-8807. https://doi.org/10.1016/j.biomaterials.2013.07.084 |
[110] | Huang K, Ma H, Liu J, et al. (2012) Size-dependent localization and penetration of ultrasmall gold nanoparticles in cancer cells, multicellular spheroids, and tumors in vivo. ACS Nano 6: 4483-4493. https://doi.org/10.1021/nn301282m |
[111] | Bauckneht M, Ciccarese C, Laudicella R, et al. (2024) Theranostics revolution in prostate cancer: basics, clinical applications, open issues and future perspectives. Cancer Treat Rev 102698. https://doi.org/10.1016/j.ctrv.2024.102698 |
[112] | Krasia-Christoforou T, Georgiou TK (2013) Polymeric theranostics: using polymer-based systems for simultaneous imaging and therapy. J Mater Chem B 1: 3002. https://doi.org/10.1039/c3tb20191k |
[113] | Shetake NG, Das SK, Kumar A, et al. (2024) Nano-inducer of ferroptosis for targeted chemotherapy of human triple negative breast carcinoma. Biomater Adv 161: 213868. https://doi.org/10.1016/j.bioadv.2024.213868 |
[114] | Zhu X, Xie L, Tian J, et al. (2024) A multi-mode Rhein-based nano-platform synergizing ferrotherapy/chemotherapy-induced immunotherapy for enhanced tumor therapy. Acta Biomater 180: 383-393. https://doi.org/10.1016/j.actbio.2024.03.030 |
[115] | Cui L, Xu Q, Lou W, et al. (2024) Chitosan oligosaccharide-functionalized nano-prodrug for cascade chemotherapy through oxidative stress amplification. Int J Biol Macromol 268: 131641. https://doi.org/10.1016/j.ijbiomac.2024.131641 |
[116] | Pei Y, Liu M, He C, et al. (2024) Application of pH and hyaluronidase dual-responsive mesoporous carbon nitride nano-drug delivery system for chemodynamic therapy and chemotherapy combination therapy of non-small cell lung cancer. Appl Mater Today 41: 102469. https://doi.org/10.1016/j.apmt.2024.102469 |
[117] | Radchenko V, Engle JW, Roy C, et al. (2016) 18th European Symposium on Radiopharmacy and Radiopharmaceuticals: Salzburg, Austria. EJNMMI Radiopharmacy Chem 1: 10. https://doi.org/10.1186/s41181-016-0012-6 |
[118] | Gao Y, Wang X, He X, et al. (2019) A dual-functional photosensitizer for ultraefficient photodynamic therapy and synchronous anticancer efficacy monitoring. Adv Funct Mater 29: 1902673. https://doi.org/10.1002/adfm.201902673 |
[119] | Li Y, Tan C, Zhang W, et al. (2014) Phosphorescent iridium(III)-bis-N-heterocyclic carbene complexes as mitochondria-targeted theranostic and photodynamic anticancer agents. Biomaterials 39: 95-104. https://doi.org/10.1016/j.biomaterials.2014.10.070 |
[120] | Flores-Cruz RD, Espinoza-Guillén A, Reséndiz-Acevedo K, et al. (2024) Doble synergetic anticancer activity through a combined chemo-photodynamic therapy and bioimaging of a novel Cas-ZnONPs all-in-one system. J Inorg Biochem 258: 112623. https://doi.org/10.1016/j.jinorgbio.2024.112623 |
[121] | Lapotko D (2009) Plasmonic nanoparticle-generated photothermal bubbles and their biomedical applications. Nanomedicine 4: 813-845. https://doi.org/10.2217/nnm.09.59 |
[122] | Yin X, Zhao H, He Z, et al. (2023) Application of aptamer-functionalized nanomaterials in molecular imaging of tumors. Nanotechnol Rev 12: 20230107. https://doi.org/10.1515/ntrev-2023-0107 |
[123] | Li Z, Cheng L, Xu X, et al. (2024) Cuproptosis-based layer-by-layer silk fibroin nanoplatform-loaded PD-L1 siRNA combining photothermal and chemodynamic therapy against metastatic breast cancer. Mater Today Bio 29: 101298. https://doi.org/10.1016/j.mtbio.2024.101298 |
[124] | Zhang X, Bai Z, Cui Z, et al. (2024) Development of a sequential release nanomaterial for co-delivery of hypoxia-induced tirapazamine and HIF-1α siRNA in cancer therapy. Colloid Surface B 245: 114260. https://doi.org/10.1016/j.colsurfb.2024.114260 |
[125] | Higaki S, Muramatsu M, Matsuda A, et al. (2018) Defensive effect of microRNA-200b/c against amyloid-beta peptide-induced toxicity in Alzheimer's disease models. Plos One 13: e0196929. https://doi.org/10.1371/journal.pone.0196929 |
[126] | Li QQ, Lai KL, Chan PS, et al. (2016) Micellar delivery of dasatinib for the inhibition of pathologic cellular processes of the retinal pigment epithelium. Colloid Surface B 140: 278-286. https://doi.org/10.1016/j.colsurfb.2015.12.053 |