Metallic Nanoparticles Applications and Therapies: Comparison
Please note this is a comparison between Version 2 by Rita Xu and Version 1 by André F. Moreira.

Metallic-based nanoparticles present a unique set of physicochemical properties that support their application in different fields, such as electronics, medical diagnostics, and therapeutics. Particularly, in cancer therapy, the plasmonic resonance, magnetic behavior, X-ray attenuation, and radical oxygen species generation capacity displayed by metallic nanoparticles make them highly promising theragnostic solutions.

  • metallic nanoparticles
  • metal-polymer nanoconjugates
  • cancer

1. Introduction

The manipulation of matter at the nanoscale led to the development of various nanomaterials, based on polymers, lipids, ceramics, or metals, that exhibit exciting properties for improving the performance of electronics, medical diagnostics, and therapeutics [1,2][1][2]. Particularly, the application of nanoparticles for cancer therapy led to the development of new alternatives from therapeutics (i.e., drug delivery, photothermal and photodynamic effects) to diagnostic and imaging, leveraging the innate ability of these materials to accumulate in the tumor tissue [3,4,5][3][4][5].
In this field, the application of metallic-based nanomaterials (e.g., gold, iron, silver, and copper) has been capturing more attention in recent years due to their characteristic physical (e.g., magnetic behavior, plasmonic resonance, and imaging capacity) and chemical (e.g., radical oxygen species (ROS) generation, and catalytic activity enhancement) properties, which render them as platforms suitable for creating multifunctional cancer therapeutics [2,6,7,8,9][2][6][7][8][9]. Moreover, there are several methodologies described for the synthesis of metallic nanoparticles, such as physical or chemical vapor deposition, sol-gel methods, chemical reduction, hydrothermal methods, solvothermal method, laser ablation, and green synthesis processes, allowing the selection of the process most compatible with the available laboratory/industrial conditions and desired physicochemical properties [10,11,12,13][10][11][12][13]. Additionally, the surface functionalization of metallic nanomaterials, a strategy often used to refine the therapeutics pharmacokinetics, can be performed by well-known and defined methodologies [14]. Thus, the metallic nanomaterials’ theranostic potential provides an all-in-one solution for cancer diagnosis, therapy, and real-time monitoring, which ultimately can improve the therapeutic outcome of anticancer therapy [15,16][15][16]. This is the rationale behind various metallic-based nanomaterials under clinical trials, such as Aurolase®, Nanotherm®, and Magnablate®.
Nevertheless, despite the wide number of publications showing the appealing features of metallic-based nanomaterials, their translation into the clinic is still very limited [17]. Such is widely associated with some toxicological issues, lack of colloidal stability, and the establishment of off-target interactions [18]. Additionally, when subjected to high-energy radiations, the metallic nanoparticles often undergo reshaping processes or are even degraded leading to the loss of their therapeutic potential [19]. Therefore, researchers have been exploring the combination of metallic nanoparticles with other materials, inorganic (e.g., silica) and/or organic (e.g., polymers). Particularly, the combination of the metallic nanoparticles’ physicochemical properties with the superior biological performance of synthetic or natural polymers emerges as a valuable and straightforward approach to develop more effective anti-cancer therapeutics [16,20][16][20].

2. Metallic Nanoparticles Applications and Therapies

Nanosized metals present optical and electrical properties that differentiate them from other nanomaterials, supporting their application in biomedicine, such as the development of biosensors (e.g., diagnosis of viruses using colloidal gold nanoparticles), bioimaging agents (e.g., iron oxide-based contrast agents), catalysts, mechanical reinforcement, and drug delivery system or other therapeutics. Moreover, these unique characteristics can be explored to create more effective antitumoral nanomedicines. The high density and X-ray attenuation capacity of metallic nanoparticles allow them the intrinsic capacity to be applied as contrast agents for bioimaging applications [21]. Up to now, several studies available in the literature have already shown that metallic nanoparticles provide a higher contrast enhancement in X-ray computed tomography (CT) imaging than the iodine-based contrast agents conventionally used in the clinic [22,23,24,25,26][22][23][24][25][26]. On the other hand, nanosized metals, such as gold, silver, and copper, show a pronounced plasmonic resonance phenomenon, i.e., the collective oscillation of the conduction band electrons in metal-based nanomaterials in response to the incident photons [27]. This interaction can lead to light absorption or scattering and is dependent on the size, morphology, distance, and dielectric constant of the metallic nanoparticles and surrounding medium [28,29,30][28][29][30]. In turn, the excited surface electrons can decay to the ground state via different processes (e.g., electron-to-photon, electron-to-electron, and electron-to-phonon energy conversion), the two most common events being the release of the absorbed energy in the form of light or heat [31]. The former is often explored to enhance the quantum efficiency and photostability of fluorophores, allowing the detection of lower quantities of biomarkers used in biosensing or bioimaging [32]. The latter is the foundation stone for the application of metallic nanoparticles in cancer hyperthermia/photothermal therapy [33]. However, it is essential to tailor the nanomaterials to interact specifically with near-infrared (NIR) radiation, a region of the spectra where the major biological components (e.g., collagen, hemoglobin, and water) have the lowest or insignificant absorption [34]. This will reduce the off-target interactions and guarantee a site-specific activation of the metallic nanomaterials. Then, the heat generated by the light-nanoparticles interaction can mediate the destruction of the cancer cells [35,36][35][36]. The elevation of the tumor temperature to values superior to 45 °C provokes irreversible damage to cancer cells (e.g., DNA degradation, cell membrane disruption, and protein denaturation), leading to cell death (i.e., tumor ablation). Otherwise, if mild temperature increases are achieved (i.e., between 40 and 45 °C), the cell damage is less pronounced and often reversed by the cell repair mechanisms [5,37,38][5][37][38]. Nevertheless, this creates a time window during which the cancer cells are more sensitive to the action of other therapeutic modalities such as chemotherapy [39]. Furthermore, metallic nanomaterials, such as those composed of iron, nickel, and cobalt, can also present magnetic properties, also allowing their application as contrast agents (magnetic resonance imaging (MRI)) and in tumor magnetic hyperthermia [40]. This capacity to be magnetically manipulated by external magnetic fields is also explored to guide these metallic nanomaterials in the human body and promote a tumor-specific accumulation [41,42][41][42]. At the tumor site, the utilization of alternating magnetic fields will promote the nanoparticles’ vibration and consequently a localized temperature increase will be obtained [43]. Metallic nanomaterials have also shown the capacity to mediate the formation of ROS [18]. This oxidative stress can influence several cellular processes/structures, e.g., intracellular calcium concentrations, activate transcription factors, induce DNA damage, and lipid peroxidation (cell membrane disruption), and increased amounts of ROS are highly cytotoxic [18]. The mechanism of ROS generation by metallic nanomaterials is influenced by their physicochemical properties (e.g., size, chemical structure, surface area, and charge) [44]. Generally, metallic nanomaterials act as the reactant or catalyst for the reduction of molecular oxygen to water, which yields the production of ROS, such as superoxide radicals and hydroxyl radicals [45]. The ROS generation of metallic nanomaterials can be further boosted by light absorption [46]. During this process, the electrons transit to higher energy bands, facilitating the reaction with water or molecular oxygen and consequently the ROS generation, a process denominated by photodynamic effect [47,48][47][48]. Despite the imaging and therapeutic potential of metallic nanoparticles, the in vivo application and translation to the clinic are severely hindered by their low colloidal stability, high reactivity, the formation of the protein corona, and high cytotoxicity [49,50,51][49][50][51]. Therefore, to overcome these limitations, researchers have been combining the superior physicochemical features of metallic nanoparticles with the increased biological properties (e.g., biocompatibility, enhanced blood circulation time, targeting capacity) of synthetic and natural polymers, often referred to as metal-polymer nanocomplexes or nanohybrids.

References

  1. Aghebati-Maleki, A.; Dolati, S.; Ahmadi, M.; Baghbanzhadeh, A.; Asadi, M.; Fotouhi, A.; Yousefi, M.; Aghebati-Maleki, L. Nanoparticles and cancer therapy: Perspectives for application of nanoparticles in the treatment of cancers. J. Cell. Physiol. 2020, 235, 1962–1972.
  2. Goncalves, A.S.C.; Rodrigues, C.F.; Moreira, A.F.; Correia, I.J. Strategies to improve the photothermal capacity of gold-based nanomedicines. Acta Biomater. 2020, 116, 105–137.
  3. Guimaraes, R.S.; Rodrigues, C.F.; Moreira, A.F.; Correia, I.J. Overview of stimuli-responsive mesoporous organosilica nanocarriers for drug delivery. Pharmacol. Res. 2020, 155, 104742.
  4. Huang, N.; Liu, Y.; Fang, Y.; Zheng, S.; Wu, J.; Wang, M.; Zhong, W.; Shi, M.; Xing, M.; Liao, W. Gold Nanoparticles Induce Tumor Vessel Normalization and Impair Metastasis by Inhibiting Endothelial Smad2/3 Signaling. ACS Nano 2020, 14, 7940–7958.
  5. Rodrigues, C.F.; Alves, C.G.; Lima-Sousa, R.; Moreira, A.F.; de Melo-Diogo, D.; Correia, I.J. Inorganic-based drug delivery systems for cancer therapy. In Advances and Avenues in the Development of Novel Carriers for Bioactives and Biological Agents; Academic Press: Cambridge, MA, USA, 2020; pp. 283–316.
  6. Liu, X.; Yan, B.; Li, Y.; Ma, X.; Jiao, W.; Shi, K.; Zhang, T.; Chen, S.; He, Y.; Liang, X.; et al. Graphene Oxide-Grafted Magnetic Nanorings Mediated Magnetothermodynamic Therapy Favoring Reactive Oxygen Species-Related Immune Response for Enhanced Antitumor Efficacy. ACS Nano 2020, 14, 1936–1950.
  7. Li, G.; Zhong, X.; Wang, X.; Gong, F.; Lei, H.; Zhou, Y.; Li, C.; Xiao, Z.; Ren, G.; Zhang, L. Titanium carbide nanosheets with defect structure for photothermal-enhanced sonodynamic therapy. Bioact. Mater. 2022, 8, 409–419.
  8. Loh, X.J.; Lee, T.C.; Dou, Q.; Deen, G.R. Utilising inorganic nanocarriers for gene delivery. Biomater. Sci. 2016, 4, 70–86.
  9. Liou, G.Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496.
  10. Tricoli, A.; Righettoni, M.; Teleki, A. Semiconductor gas sensors: Dry synthesis and application. Angew. Chem. Int. Ed. 2010, 49, 7632–7659.
  11. Huynh, K.H.; Pham, X.H.; Kim, J.; Lee, S.H.; Chang, H.; Rho, W.Y.; Jun, B.H. Synthesis, Properties, and Biological Applications of Metallic Alloy Nanoparticles. Int. J. Mol. Sci. 2020, 21, 5174.
  12. Amendola, V.; Meneghetti, M. Laser ablation synthesis in solution and size manipulation of noble metal nanoparticles. Phys. Chem. Chem. Phys. 2009, 11, 3805–3821.
  13. Annamalai, J.; Murugan, P.; Ganapathy, D.; Nallaswamy, D.; Atchudan, R.; Arya, S.; Khosla, A.; Barathi, S.; Sundramoorthy, A.K. Synthesis of various dimensional metal organic frameworks (MOFs) and their hybrid composites for emerging applications—A review. Chemosphere 2022, 298, 134184.
  14. Hu, P.; Chen, L.; Kang, X.; Chen, S. Surface Functionalization of Metal Nanoparticles by Conjugated Metal-Ligand Interfacial Bonds: Impacts on Intraparticle Charge Transfer. Acc. Chem. Res. 2016, 49, 2251–2260.
  15. Neha, D.; Momin, M.; Khan, T.; Gharat, S.; Ningthoujam, R.S.; Omri, A. Metallic nanoparticles as drug delivery system for the treatment of cancer. Expert Opin. Drug Deliv. 2021, 18, 1261–1290.
  16. Xu, J.J.; Zhang, W.C.; Guo, Y.W.; Chen, X.Y.; Zhang, Y.N. Metal nanoparticles as a promising technology in targeted cancer treatment. Drug Deliv. 2022, 29, 664–678.
  17. Anselmo, A.C.; Mitragotri, S. A Review of Clinical Translation of Inorganic Nanoparticles. AAPS J. 2015, 17, 1041–1054.
  18. Canaparo, R.; Foglietta, F.; Limongi, T.; Serpe, L. Biomedical Applications of Reactive Oxygen Species Generation by Metal Nanoparticles. Materials 2020, 14, 53.
  19. Gonzalez-Rubio, G.; Guerrero-Martinez, A.; Liz-Marzan, L.M. Reshaping, Fragmentation, and Assembly of Gold Nanoparticles Assisted by Pulse Lasers. Acc. Chem. Res. 2016, 49, 678–686.
  20. Bhatia, S. Natural Polymers vs Synthetic Polymer. In Natural Polymer Drug Delivery Systems; Springer: Cham, Switzerland, 2016; pp. 95–118.
  21. Aslan, N.; Ceylan, B.; Koç, M.M.; Findik, F. Metallic nanoparticles as X-ray computed tomography (CT) contrast agents: A review. J. Mol. Struct. 2020, 1219, 128599.
  22. Liu, Y.; Ai, K.; Lu, L. Nanoparticulate X-ray computed tomography contrast agents: From design validation to in vivo applications. Acc. Chem. Res. 2012, 45, 1817–1827.
  23. Cheheltani, R.; Ezzibdeh, R.M.; Chhour, P.; Pulaparthi, K.; Kim, J.; Jurcova, M.; Hsu, J.; Blundell, C.; Litt, H.; Ferrari, V.A.; et al. Tunable, biodegradable gold nanoparticles as contrast agents for computed tomography and photoacoustic imaging. Biomaterials 2016, 102, 87–97.
  24. De La Vega, J.C.; Esquinas, P.L.; Gill, J.K.; Jessa, S.; Gill, B.; Thakur, Y.; Saatchi, K.; Hafeli, U.O. Comparison of Rhenium and Iodine as Contrast Agents in X-ray Imaging. Contrast Media Mol. Imaging 2021, 2021, 1250360.
  25. Berger, M.; Bauser, M.; Frenzel, T.; Hilger, C.S.; Jost, G.; Lauria, S.; Morgenstern, B.; Neis, C.; Pietsch, H.; Sülzle, D.; et al. Hafnium-Based Contrast Agents for X-ray Computed Tomography. Inorg. Chem. 2017, 56, 5757–5761.
  26. Bae, K.T.; McDermott, R.; Gierada, D.S.; Heiken, J.P.; Nolte, M.A.; Takahashi, N.; Hong, C. Gadolinium-enhanced computed tomography angiography in multi-detector row computed tomography. Acad. Radiol. 2004, 11, 61–68.
  27. Werts, M.H.V.; Allix, F.; Francais, O.; Frochot, C.; Griscom, L.; Le Pioufle, B.; Loumaigne, M.; Midelet, J. Manipulation and Optical Detection of Colloidal Functional Plasmonic Nanostructures in Microfluidic Systems. IEEE J. Sel. Top. Quantum Electron. 2014, 20, 102–114.
  28. Wang, L.; Hasanzadeh Kafshgari, M.; Meunier, M. Optical Properties and Applications of Plasmonic-Metal Nanoparticles. Adv. Funct. Mater. 2020, 30, 2005400.
  29. Noguez, C. Surface Plasmons on Metal Nanoparticles: The Influence of Shape and Physical Environment. J. Phys. Chem. C 2007, 111, 3806–3819.
  30. Liz-Marzán, L.M. Tailoring Surface Plasmons through the Morphology and Assembly of Metal Nanoparticles. Langmuir 2006, 22, 32–41.
  31. Liang, J.; Liu, H.; Yu, J.; Zhou, L.; Zhu, J. Plasmon-enhanced solar vapor generation. Nanophotonics 2019, 8, 771–786.
  32. Jeong, Y.; Kook, Y.M.; Lee, K.; Koh, W.G. Metal enhanced fluorescence (MEF) for biosensors: General approaches and a review of recent developments. Biosens. Bioelectron. 2018, 111, 102–116.
  33. Lin, Q.; Jia, M.; Fu, Y.; Li, B.; Dong, Z.; Niu, X.; You, Z. Upper-Critical-Solution-Temperature Polymer Modified Gold Nanorods for Laser Controlled Drug Release and Enhanced Anti-Tumour Therapy. Front. Pharm. 2021, 12, 738630.
  34. de Melo-Diogo, D.; Pais-Silva, C.; Dias, D.R.; Moreira, A.F.; Correia, I.J. Strategies to Improve Cancer Photothermal Therapy Mediated by Nanomaterials. Adv. Healthc. Mater. 2017, 6, 1700073.
  35. Bettaieb, A.; Wrzal, K.P.; Averill-Bates, D.A. Hyperthermia: Cancer Treatment and Beyond. Cancer Treat. Conv. Innov. Approaches 2013, 257–283.
  36. Zhang, Y.; Zhan, X.; Xiong, J.; Peng, S.; Huang, W.; Joshi, R.; Cai, Y.; Liu, Y.; Li, R.; Yuan, K.; et al. Temperature-dependent cell death patterns induced by functionalized gold nanoparticle photothermal therapy in melanoma cells. Sci. Rep. 2018, 8, 8720.
  37. Fernandes, N.; Rodrigues, C.F.; Moreira, A.F.; Correia, I.J. Overview of the application of inorganic nanomaterials in cancer photothermal therapy. Biomater. Sci. 2020, 8, 2990–3020.
  38. Xing, Y.; Cai, Z.; Xu, M.; Ju, W.; Luo, X.; Hu, Y.; Liu, X.; Kang, T.; Wu, P.; Cai, C.; et al. Raman observation of a molecular signaling pathway of apoptotic cells induced by photothermal therapy. Chem. Sci. 2019, 10, 10900–10910.
  39. Jiang, Z.; Li, T.; Cheng, H.; Zhang, F.; Yang, X.; Wang, S.; Zhou, J.; Ding, Y. Nanomedicine potentiates mild photothermal therapy for tumor ablation. Asian J. Pharm. Sci. 2021, 16, 738–761.
  40. Rudakov, G.A.; Tsiberkin, K.B.; Ponomarev, R.S.; Henner, V.K.; Ziolkowska, D.A.; Jasinski, J.B.; Sumanasekera, G. Magnetic properties of transition metal nanoparticles enclosed in carbon nanocages. J. Magn. Magn. Mater. 2019, 472, 34–39.
  41. Soheilian, R.; Choi, Y.S.; David, A.E.; Abdi, H.; Maloney, C.E.; Erb, R.M. Toward Accumulation of Magnetic Nanoparticles into Tissues of Small Porosity. Langmuir 2015, 31, 8267–8274.
  42. Guo, X.; Li, W.; Luo, L.; Wang, Z.; Li, Q.; Kong, F.; Zhang, H.; Yang, J.; Zhu, C.; Du, Y.; et al. External Magnetic Field-Enhanced Chemo-Photothermal Combination Tumor Therapy via Iron Oxide Nanoparticles. ACS Appl. Mater. Interfaces 2017, 9, 16581–16593.
  43. Farzin, A.; Etesami, S.A.; Quint, J.; Memic, A.; Tamayol, A. Magnetic Nanoparticles in Cancer Therapy and Diagnosis. Adv. Healthc. Mater. 2020, 9, e1901058.
  44. Sengul, A.B.; Asmatulu, E. Toxicity of metal and metal oxide nanoparticles: A review. Environ. Chem. Lett. 2020, 18, 1659–1683.
  45. Wu, H.; Yin, J.J.; Wamer, W.G.; Zeng, M.; Lo, Y.M. Reactive oxygen species-related activities of nano-iron metal and nano-iron oxides. J. Food Drug Anal. 2014, 22, 86–94.
  46. Yuan, P.; Ding, X.; Yang, Y.Y.; Xu, Q.-H. Metal Nanoparticles for Diagnosis and Therapy of Bacterial Infection. Adv. Healthc. Mater. 2018, 7, 1701392.
  47. Juarranz, Á.; Jaén, P.; Sanz-Rodríguez, F.; Cuevas, J.; González, S. Photodynamic therapy of cancer. Basic principles and applications. Clin. Transl. Oncol. 2008, 10, 148–154.
  48. Wilson, B.C. Photodynamic Therapy for Cancer: Principles. Can. J. Gastroenterol. 2002, 16, 743109.
  49. Vinković Vrček, I.; Pavičić, I.; Crnković, T.; Jurašin, D.; Babič, M.; Horák, D.; Lovric, M.; Ferhatovic, L.; Curlin, M.; Gajovic, S. Does surface coating of metallic nanoparticles modulate their interference with in vitro assays? RSC Adv. 2015, 5, 70787–70807.
  50. Rajendran, K.; Pujari, L.; Krishnamoorthy, M.; Sen, S.; Dharmaraj, D.; Karuppiah, K.; Ethiraj, K. Toxicological evaluation of biosynthesised hematite nanoparticles in vivo. Colloids Surf. B Biointerfaces 2021, 198, 111475.
  51. García-Álvarez, R.; Hadjidemetriou, M.; Sánchez-Iglesias, A.; Liz-Marzán, L.M.; Kostarelos, K. In vivo formation of protein corona on gold nanoparticles. The effect of their size and shape. Nanoscale 2018, 10, 1256–1264.
More
ScholarVision Creations