Review Article
Platinum Based Nanoparticles: Potential Implications and Bio-Medicinal Applications
Pinki and Chaudhary A
Corresponding Author: Dr. Ashu Chaudhary, Assistant Professor, Department of Chemistry, Kurukshetra University, Kurukshetra, Haryana, India 136119
Received: April 22, 2020; Revised: May 06, 2020; Accepted: May 04, 2020
Citation: Chaudhary A & Pinki. (2020) Platinum Based Nanoparticles: Potential Implications and Bio-Medicinal Applications. Adv Nanomed Nanotechnol Res, 2(2): 136-148.
Copyrights: ©2020 Chaudhary A & Pinki. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Share :
  • 1050

    Views & Citations
  • 50

    Likes & Shares

Platinum nanoparticles (PtNPs) are remarkable scientific implement that are being investigated in different biotechnological, nano-medicinal and pharmacological fields. Among the different metallic nanoparticles, platinum nanoparticles (PtNPs) have more favourable rewards and applications, particularly in the biomedical fields. This article predominantly describes the various strategies for PtNPs synthesis, for example, chemical, physical and biological approaches. Moreover, the biomedical applications are extravagantly talked about. The content described herein will be incredibly valuable for specialists in clinical fields and industrial researchers in biologics, empowering them to discover new instants of knowledge into their individual fields.

 

Keywords: Platinum nanoparticles, Cancer therapy, Radiotherapy, Photothermal therapy, Nano-diagnostics, Combination therapy

Abbreviations: PtNPs: platinum nanoparticles; DNA: Deoxyribo nucleic acid; RNA: Ribonucleic acid; TPNs: Trifolium platinum nanoparticles; ROS: Reactive oxygen species; PTT: Photo thermal treatment; ATP: Adenosine tri-phosphate; FePtNPs: Iron-platinum nanoparticles; FDA: Food and drug administration; PVA: Capped PtNP- Polyvinyl capped platinum nanoparticle; NCL: Nanotechnology Characterization Laboratory; PVP: PtNPs- Polyvinylpyrrolidone platinum nanoparticles; N7: Nitrogen-7 atom of guanine base; HT-29: Human colorectal adenocarcinoma cell line; MCF-7: Michigan Cancer Foundation-7; LHRH: Luteinizing Hormone Release Hormone; HRP: Horseradish peroxidase; TMB: 3,3’,5,5’-Tetramethylbenzidine; CAT: Chloramphenicol Acetyl transferase; SOD: Superoxide dismutase; RBCs: Red blood corpuscles; HAS-PtNPs- Human serum albumin- platinum nanoparticles

INTRODUCTION
Nano-chemistry is a blossoming field and is generally applied to biomedical building and nanomedicine. The guarantee of nanoparticle innovation lies in the possibility that the synthesis and physical size of the material. Nanoparticle research has progressed significantly toward that over the most recent two decades. Various advances are associated with the creation of nanomaterials from different sources, for example, physical, chemical, and biological materials and various techniques are utilized to amplify the creation of nanomaterials, for example, the utilization of various crude materials, temperature, and pH. The worldwide market has an appeal for nanoparticles (NPs), and it is expected that this curiosity will arrive at 98 billion dollars by 2025. This exploration is clinically significant the same number of clinical preliminaries concentrated on gene therapy are progressing or even completed. In any case, the most vigorously looked into nanoparticle field is drug delivery. There are a huge number of endorsed details that are monetarily accessible now and no doubt many more in transit. The inspirations for this exploration are improved pharmacokinetics, targeting and stability. The results are the absolute, generally modern and well characterized nanoparticle innovations in play today [1-9].
Generally, physical strategies devour colossal vitality and disseminate radiation, while chemical techniques utilize a few toxic chemical compounds, which are hazardous and unsafe to living creatures, and discharge of harmful chemicals into the environment. Consequently, scientists have investigated cleaner, greener, adaptable, financially adorable and environmentally benign methodologies that can maintain a strategic distance from dangerous chemical substances. Ecologically favorable conditions require different formats, including microorganisms, algae, fungi and plants, and small particles that can go about as alternatives for physical and chemical strategies/methods. The shape and size of the nanoparticles can be constrained by approaches, for example, utilizing various convergences of reducing agent/capping agent, concentrations of precursors, pH and temperature. Reactant nanomaterials like platinum, palladium, and cerium show supreme physicochemical properties and high surface region and have tremendous potential for application in different fields.
Platinum NPs (PtNPs) have prompted another insurgency in the field of nanotechnology including the chemical industry, automobile sector, and biomedical applications, and therapeutic agents for the treatment of various diseases. They are utilized in a similar way in innumerable biomedical fields incorporating diagnostics with various agents for imaging, clinical implants, drug delivery, anticancer activity, antibacterial activity and photothermal treatment as shown in Figure 1 [10-12]. Platinum nanoparticles (Pt-NPs) are thought to fill in as a store house for platinum particles that have the ability to enter cells and incite DNA impairment [13]. Pt-NPs are biocompatible and have enzymatic and synergist property, proposing their potential as anticancer remedial operators [14]. In particular, Pt-NPs have exhibited clinical adequacy in the treatment of malignant growth cells with procured multidrug resistance [15,16], and have been appeared to assume helpful roles in the treatment of malignant cells growth by giving better targeting, gene silencing and drug delivery mechanisms [17]. These properties propose Pt-NPs as a sheltered and elective treatment with insignificant side effects, compared and normally utilized chemotherapeutics. What's more, they can be utilized as a demonstrative diagnostic for the imaging of malignant cells growth [18].
Pt-NPs adopt significant roles in the treatment of malignant cells growth, including their capacity to repress the initiation, propagation and recovery of tumor cells due to their great pharmacokinetic properties. Pelka et al. have revealed that Pt-NPs (size 20 to 100 nm) brought about DNA impairment in human colon carcinoma cell line HT29, in a concentration and time dependent manner [19].

An increment in toxicity of cell was connected with a reduction in Pt-NPs diameter. The harmful impacts didn't have all the earmarks of being because of reactive oxygen species development, prompting the conclusion that Pt ions from Pt-NPs might be utilized as anticancer treatments, with a comparable methodology to cisplatin. So also, Asharani et al. considered the uptake and bioactivity (e.g., cytotoxicity, genotoxicity and protein expression) of Pt-NPs (5–8 nm) in human cells [20]. They saw that Pt-NPs capped with polyvinyl liquor entered the

 
cells through dispersion, and localized inside the cytoplasm, causing DNA impairment. This examination has prescribed Pt-NP-based anticancer specialists’ viability by appropriate surface alterations to build their natural anticancer activity.
The utilizations of nanoparticles essentially rely on their size, shape, morphology and scatterings [21-24]. The interest for this material in innovative applications and the extravagance part is expanding, while the presence of Platinum (Pt) in the Earth's crust (around 0.01 ppm) is being vulnerable. The present Pt yields, such broad use and the expected demand for its future applications have significantly affected its expanding worth and cost. Recently, both the exploration and industrial fields have indicated extensive enthusiasm for the synthesis of synergist PtNPs for use in therapeutic applications due to their low toxicity, high strength, and less harmful effects.
BIOMEDICAL APPLICATIONS OF PLATNUM NANOPARTICLES
Platinum nanoparticles in cancer therapy
 
Pt-based compounds with a characterized geometrical conveyance of the ligands around the Pt particles are among the most significant medications presently accessible to treat a few types of malignant growths. The agent worth referencing right now cisplatin, which utilizes its cytotoxic impact by specifically holding with N7 atom of purine bases in DNA [25] with the origin of a DNA–platinum adduct that turns the structure of the DNA duplex, harming its replication and translation. Such a toxicity mechanism was demonstrated to be actually constrained by ligand geometry (i.e., the cis conformation), and solid attempts have been committed to discovering agents like cisplatin with less side reactions and better viability [26, 27]. Right now, look into, a few reports uncover that Pt nanomaterials are identified with cisplatin and studied as potential options for anticancer treatment [28, 29]. To improve the lethal presentation of PtNPs, some researchers have presented a subsequent material, convincing basic changes in the physicochemical properties of NPs to raise their termination inside cells. On account of bimetallic NPs, the arrival of Pt particles may get significant, as the strength of the Pt–Pt bond might be weakened by the existence of another metallic particle, preferring the acidic conditions of the material [30, 31]. The utilization of PtNPs for the applications in intraperitoneal chemotherapy was analyzed by in-situ cross linkable hyaluronic acid gel. The presence of hyaluronic acid carboxyl groups complexed with Pt decreased the arrival of PtNPs in the tumor site. Following 3 days, the researchers recommended that all Pt ions were discharged from the PtNP hybrid framework, demonstrating cytotoxic impacts, most likely because of the combined degradation of hyaluronic acid and Pt ion proclamation [32]. The magnetic nanoparticles with platinum acts as chemotherapeutic agents in the treatment of tumor cells and shows synergistic effect as shown in Figure 2.
NP functionalization can balance their disguise and encourage the obstruction of intracellular systems by the NPs. In numerous reports, folic acid was utilized as a NP capping and cell-disguise specialist [33-35] as a few kinds of malignant growth cells overexpress the folate receptor [36,37]. The proximity of folic acid on PtNPs of 10–15 nm causes higher cytotoxicity than PVP-PtNPs on MCF7 breast malignancy cells, most likely because of expanded receptor mediated endocytosis [34]. The conceivable utilization of 2-3 nm PtNPs balanced out with folic acid in cancer treatment conventions was anticipated, considering their higher explicitness for keratinocytes and mammary breast  malignant cells [33]. In any case, the component of cytotoxicity has never been demonstrated, regardless of the theory about Pt ion release and Pt–DNA adduct formation. This is a significant point, as clearly shown in many papers, for example by electron microscopy, the PtNP confinement in endo-lysosomal vesicles. To additionally demonstrate the key role of the covering in malignant growth treatment, the functionalization of bimetallic NPs was battered by Xu et al. [38] to build up another anticancer drug, in view of cubic FePtNPs covered with a LHRH peptide.
Antibacterial applications of platinum nanoparticles
 
The improvement of new bactericidal operators is at present probably the greatest challenges, because of rising uncertainties about bacterial protection from anti-infection agents. Metallic NPs could assume a profession right now. A few NPs, similar to Ag, Pd, Au, Cu, ZnO and TiO2, have demonstrated promising outcomes [39], yet their therapeutic use is fragmented as a result of undesired side effects in vivo. The antimicrobial activities of Pt particles on Escherichia coli have been portrayed since the time 1965 [40,41], however the antibacterial activity of PtNPs has been ineffectively investigated. In any case, their enzyme mimetic action may be used to initiate intracellular hyper production of ATP, causing bacterio-toxic impacts, growth inhibition and DNA impairment [42]. Surely, the antibacterial impact of PtNPs is because of their ability to upsurge ATP levels, causing the overexpression of a kinase liable for the bacterial evolution arrest. Up to this point, some publications have indicated PtNP antibacterial properties [43,44], which are accounted for to rely upon size and surface chemistry. Antibacterial activity of platinum nanoparticles is shown in Figure 3.

The arrangement of PtNP misusing the in-vitro case was facilitated with reactive oxygen species (ROS) overproduction and bacterial membrane interruption. The antibacterial activity of PtNPs in vivo was exhibited utilizing a grown-up zebrafish creature model. At the concentration used, PtNPs were seen as nontoxic to the creature, while demonstrating the capacity to hinder bacterial expansion and totally rescue the bacterial contamination. Overall, this investigation exhibited that PtNPs are protected to zebrafish cells, and have antimicrobial properties able to kill bacterial infection [45].

In spite of this, the antibacterial properties and mechanism of activity of PtNPs are still issues of conversation. Exploiting the wide assortment of conceivable surface functionalization, the development of PtNP-bacterial vehicles was proposed as a promising framework able to transport drugs to explicit targets in the body.

Platinum nanoparticles in photothermal therapy and radiotherapy

Because of the toxic reactions of anticancer chemotherapies, clinical analysts are growing increasingly successful and site-specific medicines against malignant tumors. Among them, photothermal treatment (PTT) is a non-obtrusive treatment dependent on the utilization of the NP plasmonic impact to locally increase the cellular temperature upon irradiation, causing DNA and RNA impairment, membrane rupture and protein denaturation, lastly prompting malignant growth cell death [46]. The ideal PtNP measurements to be utilized in PTT exhibited that PVP-PtNP phototoxicity was identified with the particle and found that 5–6 nm PtNPs have minor or nontoxic impacts themselves, yet they can cause cell death once illuminated by near-IR laser. The optical properties of bimetallic FePtNPs have additionally been misused to perform PTT of solid tumors. It was accounted for that the 12 nm folate-functionalized 3-mercaptopropionic acid FePtNPs with a cubic shape, when energized by a NIR laser, inspired intracellular harm relative to the NP number, causing necrosis of malignant growth cells like Au nanorods [47]. These outcomes were detected despite the fact that the absorption intensity at 800 nm of FePtNPs was five-fold lower than that of AuNPs. Recently, the utilization of biocompatible 13nm trifolium-like PtNPs (TPNs) has been explored as a potential new photothermal operator. In general, PtNPs have demonstrated to be

acceptable possibility for PTT and radiotherapy, as they can incite cellular damage in particular region following laser irradiation or radiation acquintance.
Platinum nanoparticles in nano-diagnostics
Different properties of PtNPs have attracted attention as of late for biomedical applications. For example, fluorescent Pt nanoclusters have been effectively combined as novel biocompatible bioimaging tests for analytic purposes [48-52]. Moreover, a truly intriguing methodology depends on the utilization of Pt nanomaterials as a part of catalytic nanomotors to develop molecular machines and movement-based recognition techniques. For example, the motion of chemically power-driven nanomotors dependent on bi-segment Au–Pt nanowires has been as of late misused to recognize silver ions, DNA and ribosomal RNA, opening the path for new ideas in diagnostics [53-61].
PtNPs emerged as perfect applicants as enzyme substitutes in diagnostic evaluates [62-64]. PtNPs present numerous focal points, including simple and practical creation and decontamination, stability, resistance to proteases, high synergist action even at extreme pH and temperature, and fondness to HRP substrates. For example, it has been accounted for that the liking of DNA-balanced out PtNPs to TMB is multiple times higher contrasted with that of regular HRP catalyst [66]. Afterward, numerous PtNP-based colorimetric tests have been created [64,65] including the location of DNA, [67] malignant growth cells, [62] tumor markers, [63] metal ions, [68] penicillin anti-toxins, [69] drugs, [70] hydrogen peroxide, [71] glucose, [72] cholesterol, [73] L-cysteine, [74] choline and acetylcholine, [75] proteins, [76] infections, [77] microorganisms [78] and antibodies [79]. The action of a diagnosis on a tumor cell can be represented in Figure 4.
 
The CAT-like action of PtNPs was additionally exploited in combination with a V-chip stage to identify cancer biomarkers on the cell surface and in serum [62]. Besides, folic acid functionalized PtNPs on graphene oxide nanosheets have been proposed as an unaided eye colorimetric test for the location of malignant tumor cells [79]. Overall, the building of PtNPs and the itemized investigation of their mechanism of interactions with biological frameworks may have huge effect on the advancement of novel and basic purpose of care systems for the discovery of ecological contaminants or biomarkers in complex infections.
Platinum nanoparticles in nanomedicine
PtNPs are promising applicants as nanozymes for the treatment of oxidative-stress related ailments, because of their capacity to act as false CAT, HRP and SOD catalysts. Recent outcomes with fullerene and cerium oxide NPs on models of immune system and neurodegenerative maladies, diabetes, endometriosis, and ischemia have pushed the exploration towards the acknowledgment of effective nanozymes [81,82]. At present, PtNPs are demonstrating safe applications to a few human pathologies, as exhibited in recently distributed works [83, 84]. Not quite the same as other metal nanoparticles [85], PtNPs have high stability in acidic cell vesicle conditions, estimating cytocompatibility and resilience in vivo.
In vitro enzyme like properties of PtNPs propose a wide scope of uses in nanomedicine, in any event, estimating the utilization of PtNPs as a preventive treatment for certain kinds of malignancy and cardiovascular diseases [86].Several investigations exhibited that the scavenging capacities of PtNPs are kept up in a cellular atmosphere [87,88] since PtNPs can shield cells from ROS-initiated death after introduction to UV-A [89], X-rays [90] or ultrasound [91]. In contrast with cerium oxide NPs, the cancer prevention agent movement of PtNPs was exhibited to be increasingly productive, by assessing apoptosis prevention in HT-1080 human breast fibrosarcoma cells presented to 200 mM H2O2. This outcome is likely due to PtNP chemical stability and protection from accumulation, in spite of their lower SOD activity in vitro [92].
In like manner, it was accounted for in vivo that 1-2 nm PVP-PtNPs can expand the life expectancy of the short-lived mutant nematode Caenorhabditis elegans, influenced by significant levels of oxidative stress. The impact of the nanomaterial was more noticeable than that got with EUK-8, a recognised SOD/CAT mimetic, utilized in a similar scope of focuses [93]. A significant translational medication use of PtNPs may be the assurance of keratinocytes from ROS-initiated apoptosis, upon UV illumination. The topical utilization of PtNP-based gel secured the model mice of photosensitivity dermatitis against UVA-initiated skin damage [94]. The improvement of PtNP-based dermal definitions could greatly affect the clinical and cosmetic market. A potential novel utilization of PtNPs as elective treatment for osteoporosis has additionally been proposed in the ovariectomy-induced bone misfortune, depending on the intragastric administration of the NPs [95].
The high oxygen affinity and antioxidant action appeared by HSA–PtNP complexes let us predict new improvements for oxygen transportation in blood. This has been portrayed as elective material to red blood corpuscles (RBCs) for transfusion in some clinical pathologies [86]. Recently, the utilization of citrate-capped PtNPs as radical scavenging materials was portrayed in a cell model of Cerebral Cavernous Malformation, an uncommon cerebrovascular oxidative stress-related malady. Low concentrations of PtNPs were shown to entirely re-establish the cell physiological homeostasis in 48 h of treatment [96] giving the expectation for new ways to deal with uncommon infections.
Platinum nanoparticles in combination therapy
PtNPs are potential medicines for combination therapy due to their notable highlights of accumulation of ROS and ROS scavenging properties in the treatment of complex ailments, for example, malignant growth of cells and neurodegeneration. Combination therapy procedures have been utilized to advance synergetic adequacy and overwhelmed the resistance of platinum drugs. For example, the blend of platinum suppositories and imaging agents permits the broadcasting of drug loaded NPs inside the body and the tumor. Combination therapy has displayed diminished fundamental poisonous quality in contrast with either photothermal treatment or chemotherapy alone. Further, PtNPs can be exploited appropriate functionalization distinctive reducing agents. For the most part, platinum-based drugs are utilized in the treatment of cancer, including ovarian, head and neck, and lung malignant growth. Platinum-based drugs yield positively charged, receptive aquatic species that therefore can form stable DNA-adducts and in the long run cause cell death [97]. Nano-capsules of cisplatin enter the cells more proficiently than free compounds. Expanded degrees of platinum gathering cause cisplatin-DNA-adduct formation in IGROV-1 cells [98]. A combination of PtNPs with illumination by fast moving ions viably improves the strong lethal maltreatment to DNA [99].
Antifungal activity of platinum nanoparticle
Commercial antifungal operators lead to side effects, for example, liver infection, sickness, renal disappointment, increment of internal heat level, anddiarrhoea. At present, elective treatment is required for recovery from contagious ailment. Already, Gardea-Torresdey et al. [100] revealed silver NPs as having potential antifungal action against spore-creating organisms. Recently, an examination looked at the antifungal action of PtNPs and industrially accessible antifungal agents. The bio-fabricated PtNPs indicated potential antifungal movement against different pathogenic parasites, for example, C. acutatum, C. fulvum, P. drechsleri, D. bryoniae, and P. capsici [101]. The industriousness of the biopolymer intervened synthesized platinum nanocomposites (GKPtNPs) was evaluated to investigate the antifungal activity against fungal strains, for example, A. parasiticus and A. flavus. They detected antifungal action of the nanocomposite instigated the morphology of the mycelia, membrane damage, expanded the degree of ROS, eventually prompting DNA impairment and cell death [102].
Toxicology of platinum nanoparticles
These days, the request of nanoPt in biomedicine is still discussed, because of its muddled toxicological characterization. Regardless of the announcement of FDA about the security of Pt in the zero-oxidation state [103] and the endorsed utilization of consumer products in Japan, distributed outcomes on PtNP cytotoxicity are as yet clashing and the conceivable harmful mechanisms are not completely comprehended. The lethal impact of different sorts of NPs is as often as possible described by the induction of oxidative stress, DNA impairment, and cell cycle apprehension, [85,104-108] prompting explicit organ failure [103,109]. Be that as it may, a few information exhibits that a critical role in cell function impairment is regularly played by various contaminants present, similar to endotoxins, harmful coatings, or NP synthesis reaction side-products [110-112]. In spite of the fact that it has been demonstrated that diverse metallic NPs discharge particles once inside the cell, there are no authoritative information showing that cell damage observed after PtNP administration could be comparable because of the release of Pt constituent part [113-117].
Examinations concentrated on the role of PtNP size in cytotoxicity [45, 118-121] demonstrate that it could represent a significant parameter influencing molecular mechanisms inside the cell, despite the fact that with contradictory outcomes. While 8 nm NPs didn't show unfavorable impacts, administration of 1 nm PtNPs to renal cells in culture incited cytotoxicity in a dose dependent way in a similar scope of concentrations [119]. Manikandan et al. tried NP sizes going from 1 to 21 nm on a Neuro 2Å cell line, perceiving that PtNPs of 5–6 nmwas completely cytocompatible, while PtNPs of different size-initiated cell impairment [118]. Then again, Konieczny et al. seen that PVP-PtNPs of 6 nm induced an abatement of metabolic activity and genotoxic impacts, despite the fact that they didn't adjust the morphology, feasibility and migration capability of essential keratinocytes [120].
 
A few examinations concentrated on the role of various coatings just as the generation of Pt ions as an outcome of NP endocytosis in the acidic endosome/lysosome compartments, trailed by the degradation of the particles [122-124,113-115]. For example, high concentrations (80 and 160 mg/mL) of 5-8 nm PtNPs secured by PVA cause mild toxicity following 72 h [114]. Polymer coated-PtNPs were realized in the cytoplasm and in cell vesicles, and their disguise was related with reactive oxygen species (ROS) overproduction, DNA impairment, and cell division termination. The researchers proposed a potential combined impact of NPs and Pt2+ions in ROS overproduction and DNA impairment (Figure 5). Nonetheless, the destiny of Pt ions discharged inside the cells from potential NP disintegration was not affirmed and the effect of the distinctive covering was not explained. The cytotoxicity mechanism of coated PtNPs ought to be profoundly examined considering all the segments of the particles and their own particular cell interaction mechanism.
Taking into account PtNP foundational organization, PVA-capped PtNP bio-interactions were additionally tried with human red blood corpuscles [125] revealing a decent level of biocompatibility, in spite of the fact that with some modification in membrane topography. Different investigations [126,127] including polymer-PtNPs were performed with very small, dendrimer-epitomized PtNPs of 1 nm, indicating toxicity just upon oxidation or maturing steps. The clarification of such conduct isn't totally clear, in spite of the fact that the oxidation of the Pt atoms on the NP surface after their introduction to air was viewed as liable for the release of toxic ions.
Numerous other in-vivo reports portrayed the security profile of PtNPs as they are applied in nanomedicine as cell reinforcements. Conversely, extraordinary in-vivo investigations indicated that intratracheal instillation of 21 nm PtNPs may incite inflammatory reactions related with time-dependent decrease of antioxidant molecules [128] underlining the troubles in assessing the immune responses of entire living beings. Mice treated with 1 nm PtNPs endured intense and chronic nephrotoxicity, yet no indication of toxicity was seen in the other inspected tissues (heart, lungs, spleen, liver). Much the same as in vitro information, 8 nm PtNPs didn't show negative impacts on any tissue, indicating a protected biocompatibility profile [118]. As indicated by the general rules of the National Cancer Institute's Nanotechnology Characterization Laboratory (NCL), [129] we might want to prescribe the accompanying procedures to accomplish safe and biocompatible PtNP arrangements:
1.        Complete batch-to-batch physicochemical assessment of NP properties, specifically, in complex conditions like cell culture media (size distribution, stability, zeta potential, accumulation state, protein corona formation).
2.        Extensive refinement methodology, essential to minimizing the presence of pathogenic spores or bioactive molecules, as bacterial poisons, that can without much of a stretch beat the helpful impacts of the unadulterated material and the green coatings.
3.        Assessment of the toxicological impact of every reagent, including the solvent of the PtNPs (e.g., after NP precipitation).
4.        Control of the nonappearance of endotoxin and bacterial contamination (for example by the LAL test).
5.        Evaluation of the toxicity of the covering materials as such.
CONCLUSION AND FUTURE PERSPECTIVES
Pt-based nanomaterials are key players in numerous domains of science and innovation. Specifically, as examined in the previous areas, they are promising applicants in biomedical applications, coordinating the functions of nanocarriers and nanozymes. In purpose of-care diagnostic technology, PtNPs can be utilized as counterfeit compounds to replace expensive and sensitive HRP and CAT in new colorimetric and fluorometric biosensors, and to create novel unaided eye diagnostic approaches. This is an especially fascinating field, since the high catalytic effectiveness of nanoPt combined with their stability in a wide scope of conditions (counting pH and temperature) can prompt the advancement of ultrasensitive, minimal effort, and compact tests, which can be stored for quite a long time at room temperature and performed outside specific research centers, with no temperature control or instrumental prerequisites.
In nanomedicine, PtNPs can be valuable for combination therapy in the treatment of complex diseases brought about by the accumulation of intracellular ROS. By exploiting NP versatile surface functionalization with their inherent antioxidant properties, PtNPs can be utilized to create multifunctional nano formulations with ROS scavenging properties. Also, it might be imagined that PtNPs could be additionally designed to replace damaged proteins in defective molecular pathways prompting maladies. Curiously, a few reports have indicated the higher capability of PtNPs compared to different nanozymes, for example, ceria and fullerenes, for the treatment of a few human pathologies. To completely disclose the capability of PtNPs in biomedicine, be that as it may, a detailed picture of their various properties is as yet required, including a precise examination of the underlying antioxidant mechanism and their toxicological aspects.
Our examination of the toxicological information about PtNPs proposes that the material in essence doesn't cause recognizable toxicity, rather it can essentially counteract oxidative stress damage, because of its strong catalytic properties. Concerning cisplatin-like toxic impacts by PtNPs, further examinations are expected to explain such conceivable conduct, since the anticancer mechanism of this medication depends on DNA intercalation of Pt, provided a cis geometry of the ligands around the Pt ion. The properties of PtNPs and their combinations with different materials (e.g., metals, polymers, targeting agents, and so on.) could likewise propose different applications, for example, photothermal-and radiotherapies, or the control of bacterial growth. Further research is required in the last fields, as just few outcomes by a restricted group of specialists have been introduced on the different fascinating roles and components of PtNPs. Overall, the rising field of PtNP applications in nanomedicine is developing fast, and it is conceivable to predict various and promising results for these nanomaterials in the next future.
ACKNOWLEDGEMENT
 
One of the author Ms. Pinki wish to express gratitude to the Council of Scientific and Industrial Research (CSIR) (Ref. No.-16/06/2019(i) EU-V (CSIR-UGC NET JUNE, 2019)), New Delhi, India for financial assistance in the form of JRF.

1.      Patnaik S, Gupta KC (2013) Novel polyethylenimine-derived nanoparticles for in vivo gene delivery. Expert Opin Drug Deliv 10: 215-228.

2.      Ensign LM, Schneider C, Suk JS, Cone R, Hanes J (2012) Mucus penetrating nanoparticles: Biophysical tool and method of drug and gene delivery. Adv Mater 24: 3887-3894.

3.      Duceppe N, Tabrizian M (2011) Advances in using chitosan-based nanoparticles for in vitro and in vivo drug and gene delivery. Expert Opin Drug Deliv 7: 1191-1207.

4.      Pissuwan D, Niidome T, Cortie MB (2011) The forthcoming applications of gold nanoparticles in drug and gene delivery systems. J Controlled Release 149: 65-71.

5.      Kiriyama A, Iga K, Shibata N (2013) Availability of polymeric nanoparticles for specific enhanced and targeted drug delivery. Ther Deliv 4: 1261-1278.

6.      Sridhar R, Ramakrishna S (2013) Electrosprayed nanoparticles for drug delivery and pharmaceutical applications. Biomatter 3: 024281.

7.      Tosi G, Bortot B, Ruozi B, Dolcetta D, Vandelli MA, et al. (2013) Potential use of polymeric nanoparticles for drug delivery across the blood-brain barrier. Curr Med Chem 20: 2212-2225.

8.      Cheng R, Meng F, Deng C, Klok HA, Zhong Z (2013) Dual and multi-stimuli responsive polymeric nanoparticles for programmed site-specific drug delivery. Biomaterials 34:3647-3657.

9.      Prow TW, Grice JE, Lin LL, Faye R, Butler M, et al. (2011) Nanoparticles and microparticles for skin drug delivery. Adv Drug Deliv Rev 63: 470-491.

10.   Johnstone TC, Suntharalingam K, Lippard SJ (2016) The next generation of platinum drugs: Targeted Pt(II) agents, nanoparticle delivery, and Pt(IV) prodrugs. Chem Rev 116: 3436-3486.

11.   Wang Z, Chen L, Huang C, Huang Y, Jia N (2017) Albumin-mediated platinum nanocrystals for in vivo enhanced computed tomography imaging. J Mater Chem B 5: 3498-3510.

12.   Doherty RE, Sazanovich IV, McKenzie LK, Stasheuski AS, Coyle R, et al. (2016) Photodynamic killing of cancer cells by a Platinum (II) complex with cyclometallating ligand. Sci Rep 6: 22668.

13.   Nejdl L, Kudr J, Moulick A, Hegerova D, Ruttkay-Nedecky B, et al. (2017) Platinum nanoparticles induce damage to DNA and inhibit DNA replication. PLoS ONE 12: 00180798.

14.   Pedone D, Moglianetti M, De Luca E, Bardi G, Pompa PP (2017) Platinum nanoparticles in nanobiomedicine. Chem Soc Rev 46: 4951-4975.

15.   Kang S, Kang K, Chae A, Kim YK, Jang H, et al. (2019) Fucoidan-coated coral-like Pt nanoparticles for computed tomography-guided highly enhanced synergistic anticancer effect against drug-resistant breast cancer cells. Nanoscale 11: 15173-15183.

16.   Kankala RK, Liu CG, Yang DY, Wang SB, Chen AZ (2020) Ultrasmall platinum nanoparticles enable deep tumor penetration and synergistic therapeutic abilities through free radical species-assisted catalysis to combat cancer multidrug resistance. Chem Eng J 383: 123138.

17.   Manikandan M, Hasan N, Wu HF (2013) Platinum nanoparticles for the photothermal treatment of Neuro 2A cancer cells. Biomaterials 34: 5833-5842.

18.   Gu T, Wang Y, Lu Y, Cheng L, Feng L, et al. (2019) Platinum nanoparticles to enable electrodynamic therapy for effective cancer treatment. Adv Mater 31: 1806803.

19.   Pelka J, Gehrke H, Esselen M, Turk M, Crone M, et al. (2009) Cellular uptake of platinum nanoparticles in human colon carcinoma cells and their impact on cellular redox systems and DNA integrity. Chem Res Toxicol 22: 649-659.

20.   Asharani PVN, Xinyi N, Hande MP, Valiyaveettil S (2010) DNA damage and p53-mediated growth arrest in human cells treated with platinum nanoparticles. Nanomedicine (Lond.) 5: 51-64.

21.   Coe S, Woo WK, Bawendi M, Bulović V (2002) Electroluminescence from single monolayers of nanocrystals in molecular organic devices. Nature 420: 800-803.

22.   Kelly KL, Coronado E, Zhao LL, Schatz GC (2003) The optical properties of metal nanoparticles: The influence of size, shape and dielectric environment. J Phys Chem B 107: 668-677.

23.   Abilash G, Podila R, Ramakrishna M, Karnam L, Janardhana C, et al. (2011) Catalytic reduction of 4-nitrophenol using biogenic gold and silver nanoparticles derived from Breynia rhamnoides. Langmuir 27: 15268-15274.

24.   Leong GJ, Schulze MC, Strand MB, Maloney D, Frisco SL, et al. (2014) Shape-directed platinum nanoparticle synthesis: Nanoscale design of novel catalysts. Appl Organomet Chem 28: 1-17.

25.   Comenge J, Sotelo C, Romero F, Gallego O, Barnadas A, et al. (2012) Detoxifying antitumoral drugs via nanoconjugation: The case of gold nanoparticles and cisplatin. PloS One 7: 047562.

26.   Johnstone TC, Suntharalingam K, Lippard SJ (2016) The next generation of platinum drugs: targeted Pt (II) agents, nanoparticle delivery, and Pt (IV) prodrugs. Chem Rev 116: 3436-3486.

27.   Teng B, Yu C, Zhang X, Feng Q, Wen L, et al. (2017) Up conversion nanoparticles loaded with eIF4E siRNA and platinum (IV) prodrug to sensitize platinum-based chemotherapy for laryngeal cancer and bioimaging. J Mater Chem B 5: 307-317.

28.   Estrela-Llopis VR, Chevichalova AV, Trigubova NA, Ryzhuk EV (2014) Heterocoagulation of polysaccharide-coated platinum nanoparticles with ovarian-cancer cells. Colloid J 76: 609-621.

29.   Tian Y, Zi W, Li X, Yanji L, Bian K, et al. (2017) Biologically inspired self-assembly of bacitracin-based platinum nanoparticles with anti-tumor effects. New J Chem 41: 2941-2948.

30.   López T, Alvarez M, González RD, Uddin MJ, Bustos J, et al. (2011) Synthesis, characterization and in vitro cytotoxicity of Pt-TiO2 nanoparticles. Adsorption 17: 573-581.

31.   Morozkin E, Zaporozhchenko I, Kharkova M, Cherepanova A, Laktionov P, et al. (2013) Cytotoxic and immunomodulating properties of silver and platinum nanocomposites. Chem Sustain Develop 21: 147-154.

32.   Cho EJ, Sun B, Doh KO, Wilson EM, Torregrosa-AllenS, et al. (2015) Intraperitoneal delivery of platinum with in-situ crosslinkable hyaluronic acid gel for local therapy of ovarian cancer. Biomater 37: 312-319.

33.   Mironava T, Simon M, Rafailovich MH, Rigas B (2013) Platinum folate nanoparticles toxicity: cancer vs. normal cell. Toxicol in Vitro 27: 882-889.

34.   Teow Y, Valiyaveettil S (2010) Active targeting of cancer cells using folic acid-conjugated platinum nanoparticles. Nanoscale 2: 2607-2613.

35.   Pedone D, Moglianetti M, De Luca E, Bardi G, Pompa PP  (2017) Platinum nanoparticles in nanobiomedicine. Chem Soc Rev 46: 4951-4975.

36.   Elnakat H, Ratnam M (2004) Distribution, functionality and gene regulation of folate receptor isoforms: implications in targeted therapy. Adv Drug Delivery Rev 56: 1067-1084.

37.   Parker N, Turk MJ, Westrick E, Lewis JD, Low P, et al. (2005) Folate receptor expression in carcinomas and normal tissues determined by a quantitative radioligand binding assay. Anal Biochem 338: 284-293.

38.   Xu C, Yuan Z, Kohler N, Kim J, Chung MA, et al. (2009) FePt Nanoparticles as an Fe Reservoir for Controlled Fe Release and Tumor Inhibition. J Am Chem Soc1 31: 15346-15351.

39.   Beyth N, Houri-Haddad Y, Domb A, Khan W, Hazan R (2015) Alternative antimicrobial approach: nano-antimicrobial materials. Evid Based Complement Alternat Med 246012.

40.   Rosenberg B, Van Camp L, Grimley EB, Thomson AJ (1967) The inhibition of growth or cell division in Escherichia coli by different ionic species of platinum (IV) complexes. J Biol Chem 242: 1347-1352.

41.   Rosenberg B, Vancamp L, Trosko JE, Mansour VH (1969) Platinum compounds: A new class of potent antitumour agents. Nature 222: 385-386.

42.   Zhao Y, Ye C, Liu W, Chen R, Jiang X (2014) Tuning the composition of AuPt bimetallic nanoparticles for antibacterial application. Angew Chem Int Ed 53: 8127-8131.

43.   Chwalibog A, Sawosz E, Hotowy A, Szeliga J, Mitura S, et al. (2010) Visualization of interaction between inorganic nanoparticles and bacteria or fungi. Int J Nanomed 5:1085.

44.   Kebede MA, Imae T, Wu CM, Cheng KB (2017) Cellulose fibers functionalized by metal nanoparticles stabilized in dendrimer for formaldehyde decomposition and antimicrobial activity. Chem Eng J 311: 340-347.

45.   Manikandan M, Hasan N, Wu HF (2013) Platinum nanoparticles for the photothermal treatment of Neuro 2A cancer cells. Biomaterials 34: 5833-5842.

46.   Au L, Zheng D, Zhou F, Li ZY, Li X, et al. (2008) A quantitative study on the photothermal effect of immuno gold nanocages targeted to breast cancer cells. ACS Nano 2: 1645-1652.

47.   Chen CL, KuoLR, LeeSY, Hwu YK, Chou SW, et al. (2013) Photothermal cancer therapy via femtosecond-laser-excited FePt nanoparticles. Biomater 34: 1128-1134.

48.   Tanaka SI, Miyazaki J, Tiwari DK, Jin T, Inouye Y (2011) Fluorescent Platinum Nanoclusters: Synthesis, Purification, Characterization, and Application to Bioimaging. Angew Chem Int Ed 50: 431-435.

49.   Le Guevel X, Trouillet V, Spies C, Jung G, Schneider M (2012) Synthesis of Yellow-Emitting Platinum Nanoclusters by Ligand Etching. J Phys Chem C 116: 6047-6051.

50.   Yuan X, Luo Z, Zhang Q, Zhang X, Zheng Y, et al. (2011) Synthesis of Highly Fluorescent Metal (Ag, Au, Pt, and Cu) Nanoclusters by Electrostatically Induced Reversible Phase Transfer. ACS Nano 5: 8800-8808.

51.   Kawasaki H, Yamamoto H, Fujimori H, Arakawa R, Inada M, et al. (2010) Surfactant-free solution synthesis of fluorescent platinum sub-nanoclusters. Chem Commun 46: 3759-3761.

52.   Lim H, Ju Y, Kim J (2016) Tailoring Catalytic Activity of Pt Nanoparticles Encapsulated Inside Dendrimers by Tuning Nanoparticle Sizes with Sub-nanometer Accuracy for Sensitive Chemiluminescence-based Analyses. Anal Chem 88: 4751-4758.

53.   Wu J, Balasubramanian S, Kagan D, Manesh KM, Campuzano S, et al. (2010) Motion-based DNA detection using catalytic nanomotors. Nat Commun1:36.

54.   Kagan D, Calvo-Marzal P, Balasubramanian S, Sattayasamitsathit S, Manesh KM, et al. (2009) Chemical Sensing Based on Catalytic Nanomotors: Motion-Based Detection of Trace Silver. J Am Chem Soc 131: 12082-12083.

55.   Bandodkar AJ, Wang J (2016) Wearable Biofuel Cells: A Review. Electroanalysis 28: 1188-1200.

56.   Chng EL, Zhao G, Pumera M (2014) Towards biocompatible nano/microscale machines: self-propelled catalytic nanomotors not exhibiting acute toxicity. Nanoscale 6: 2119-2124.

57.   Li J, Rozen I, Wang J (2016) Rocket Science at the Nanoscale. ACS Nano10: 5619-5634.

58.   Peng F, Tu Y, Wilson DA (2017) Micro/nanomotors towards in vivo application: cell, tissue and biofluid. Chem Soc Rev 46: 5289-5310.

59.   Wilson DA, Nolte RJ, van Hest JC (2012) Autonomous movement of platinum-loaded stomatocytes. Nat Chem 4: 268-274.

60.   Tu Y, Peng F, White PB, Wilson DA (2017) Angew. Chem Int Ed 26: 1521-3773.

61.   Paxton WF, Kistler KC, Olmeda CC, Sen A, St Angelo SK, et al. (2004) Catalytic nanomotors: autonomous movement of striped nanorods. J Am Chem Soc 126: 13424-13431.

62.   Kim MI, Kim MS, Woo MA, Ye Y, Kang KS, et al. (2014) Highly efficient colorimetric detection of target cancer cells utilizing superior catalytic activity of graphene oxide–magnetic-platinum nanohybrids. Nanoscale 6: 1529-1536.

63.   Song Y, Xia X, Wu X, Wang P, Qin L (2014) Integration of Platinum Nanoparticles with a Volumetric Bar-Chart Chip for Biomarker Assays. Angew Chem 126: 12659-12663.

64.   Sun Y, Wang J, Li W, Zhang J, Zhang Y, et al. (2015) DNA-stabilized bimetallic nanozyme and its application on colorimetric assay of biothiols. Biosens Bioelectron 74: 1038-1046.

65.   Ju Y, Kim J (2015) Dendrimer-encapsulated Pt nanoparticles with peroxidase-mimetic activity as biocatalytic labels for sensitive colorimetric analyses. Chem Commun 51: 13752-13755.

66.   Fu Y, Zhao X, Zhang J, Li W (2014) DNA-Based Platinum Nanozymes for Peroxidase Mimetics. J Phys Chem C 118: 18116-18125.

67.   Chen W, Fang X, Li H, Cao H, Kong J (2017) DNA-mediated inhibition of peroxidase-like activities on platinum nanoparticles for simple and rapid colorimetric detection of nucleic acids. Biosens Bioelectron 94: 169-175.

68.   Chen W, Fang X, Li H, Cao H, Kong J (2016) A Simple Paper-Based Colorimetric Device for Rapid Mercury (II) Assay Sci Rep 6: 31948.

69.   Kwon D, Lee W, Kim W, Yoo H, Shin HC, et al. (2015) Colorimetric detection of penicillin antibiotic residues in pork using hybrid magnetic nanoparticles and penicillin class-selective, antibody-functionalized platinum nanoparticles. Anal Methods 7: 7639-7645.

70.   You JG, Liu YW, Lu CY, Tseng WL, Yu CJ (2017) Colorimetric assay of heparin in plasma based on the inhibition of oxidase-like activity of citrate-capped platinum nanoparticles. Biosens Bioelectron 92: 442-448.

71.   Xing L, Rong Q, Ma Z (2015) Non-enzymatic electrochemical sensing of hydrogen peroxide based on polypyrrole/platinum nanocomposites. Sens Actuators B 221: 242-247.

72.   Ji X, Lau HY, Ren X, Peng B, Zhai P, et al. (2016) Highly Sensitive Metabolite Biosensor Based on Organic Electrochemical Transistor Integrated with Microfluidic Channel and Poly(N‐vinyl‐2‐pyrrolidone) ‐Capped Platinum Nanoparticles. Adv Mater Technol 1: 1600042.

73.   Shi W, Fan H, Ai S, Zhu L (2015) Honeycomb-like nitrogen-doped porous carbon supporting Pt nanoparticles as enzyme mimic for colorimetric detection of cholesterol. Sens Actuators B 221: 1515-1522.

74.   Pan N, Li-Ying W, Wu LL, Peng CF, Xie ZJ (2017) Colorimetric determination of cysteine by exploiting its inhibitory action on the peroxidase-like activity of Au@Pt core-shell nanohybrids. Microchim Acta 184: 65-72.

75.   He SB, Wu GW, Deng HH, Liu AL, Lin XH, et al. (2014) Choline and acetylcholine detection based on peroxidase-like activity and protein antifouling property of platinum nanoparticles in bovine serum albumin scaffold. Biosens Bioelectron 62: 331-336.

76.   Gao F, Du L, Zhang Y, Zhou F, Tang D (2016) A sensitive sandwich-type electrochemical aptasensor for thrombin detection based on platinum nanoparticles decorated carbon nanocages as signal labels. Biosens Bioelectron 86: 185-193.

77.   Yang ZH, Zhuo Y, Yuan R, Chai YQ (2016) A nanohybrid of platinum nanoparticles-porous ZnO–hemin with electrocatalytic activity to construct an amplified immunosensor for detection of influenza. Biosens Bioelectron 78: 321-327.

78.   Dutta G, Nagarajan S, Lapidus LJ, Lillehoj PB (2017) Enzyme-free electrochemical immunosensor based on methylene blue and the electro-oxidation of hydrazine on Pt nanoparticles. Biosens Bioelectron 92: 372-377.

79.   Gao Z, Xu M, Hou L, Chen G, Tang D (2013) Irregular-shaped platinum nanoparticles as peroxidase mimics for highly efficient colorimetric immunoassay. Anal Chim Acta 776: 79-86.

80.   Zhang LN, Deng HH, Lin FL, Xu XW, Weng SH, et al. (2014) In Situ Growth of Porous Platinum Nanoparticles on Graphene Oxide for Colorimetric Detection of Cancer Cells. Anal Chem 86: 2711-2718.

81.   De Marzi L, Monaco A, De Lapuente J, Ramos D, Borras M, et al. (2013) Cytotoxicity and Genotoxicity of Ceria Nanoparticles on Different Cell Lines in Vitro. Int J Mol Sci 14: 3065.

82.   Gamucci O, Bardi G (2014) Cerium dioxide nanoparticles selectively up-regulate C-C chemokine receptor 2 and CD16 expression on human monocytes. EURO Nano Tox Lett 5: 1-16.

83.   Kim YJ, Kim DB, Lee YH, Choi SY, Park JS, et al. (2009) Effects of nanoparticulate saponin-platinum conjugates on 2,4-dinitrofluorobenzene-induced macrophage inflammatory protein-2 gene expression via reactive oxygen species production in RAW 264.7 cells. BMB Rep 42: 304-309.

84.   Tsuji G, Hashimoto-Hachiya A, Takemura M, Kanemaru T, Ichihashi M, et al. (2017) Palladium and Platinum Nanoparticles Activate AHR and NRF2 in Human Keratinocytes-Implications in Vitiligo Therapy. J Invest Dermatol 137: 1582-1586.

85.   De Matteis V, Malvindi MA, Galeone A, Brunetti V, De Luca E, et al. (2015) Negligible particle-specific toxicity mechanism of silver nanoparticles: The role of Ag+ ion release in the cytosol. Nanomedicine 11: 731-739.

86.   Hosaka H, Haruki R, Yamada K, Bottcher C, Komatsu T (2014) Hemoglobin–Albumin Cluster Incorporating a Pt Nanoparticle: Artificial O2 Carrier with Antioxidant Activities. PLoS One 9: 0110541.

87.   Nakanishi H, Hamasaki T, Kinjo T, Teruya K, Kabayama S, et al. (2013) Size-dependent antioxidative activity of platinum nanoparticles. BMC Proc 7: 1-2.

88.   Lee JW, Son J, Yoo KM, Lo YM, Moon B (2014) Characterization of the antioxidant activity of gold@platinum nanoparticles. RSC Adv 4: 19824-19830.

89.   Hamasaki T, Kashiwagi T, Imada T, Nakamichi N, Aramaki S, et al. (2008) Kinetic Analysis of Superoxide Anion Radical-Scavenging and Hydroxyl Radical-Scavenging Activities of Platinum Nanoparticles. Langmuir 24: 7354-7364.

90.   Jawaid P, Rehman MU, Yoshihisa Y, Li P, Zhao QI, et al. (2014) Effects of SOD/catalase mimetic platinum nanoparticles on radiation-induced apoptosis in human lymphoma U937 cells. Apoptosis 19: 1006-1016.

91.   Jawaid P, Rehman MU, Hassan MA, Zhao QL, Li P et al. (2016) Effect of platinum nanoparticles on cell death induced by ultrasound in human lymphoma U937 cells. Ultrason Sonochem 31: 206-215.

92.   Clark A, Zhu A, Sun K, Petty HR (2011) Cerium oxide and platinum nanoparticles protect cells from oxidant-mediated apoptosis. J Nanopart Res 13: 5547-5555.

93.   Yan H, Kinjo T, Tian H, Hamasaki T, Teruya K, et al. (2011) Mechanism of the lifespan extension of caenorhabditis elegans by electrolyzed reduced water-participation of Pt Nanoparticles. Bio Sci Biotechnol Biochem 75: 1295-1299.

94.   Yoshihisa Y, Honda A, Zhao QL, Makino T, Abe R, et al. (2010) Protective effects of platinum nanoparticles against UV-light-induced epidermal inflammation. Exp Dermatol 19: 1000-1006.

95.   Kim WK, Kim JC, Park HJ, Sul OJ, Lee MH, et al. (2012) Platinum nanoparticles reduce ovariectomy-induced bone loss by decreasing osteoclastogenesis. Exp Mol Med 44: 432-439.

96.   Moglianetti M, De Luca E, Pedone D, Marotta R, Catelani T, et al. (2016) Platinum nanozymes recover cellular ROS homeostasis in an oxidative stress-mediated disease model. Nanoscale 8: 3739-3752.

97.   Ho YP,Au-YeungSC, To KK (2003) Platinum-based anticancer agents: Innovative design strategies and biological perspectives. Med Res Rev 23: 633-655.

98.   Hamelers IH, Sta horst RW, Voortman J, de Kruij B, Reedijk J, et al. (2009) High cytotoxicity of cisplatin nanocapsules in ovarian carcinoma cells depends on uptake by caveolae-mediated endocytosis. Clin Cancer Res 15: 2009.

99.   Porcel E, Liehn S, Remita H, Usami N, Koayashi K, et al. (2010) Platinum nanoparticles: A promising material for future cancer therapy? Nanotechnology 21: 085103.

100. Gardea-Torresdey JL, Parsons JG, Gomez E, Peralta-Videa J, Troiani HE, et al. (2002) Formation and growth of Au Nanoparticles inside live Alfalfa plants. Nano Lett 2: 397-401.

101. Sahin B, Aygün A, Gündüz H, Sahin K, Demir E, et al. (2018) Cytotoxic effects of platinum nanoparticles obtained from pomegranate extract by the green synthesis method on the MCF-7 Cell line. Colloids Surf B Biointerfaces 163: 119-124.

102. Deepika G, Sashidhar RB (2018) Biopolymer-mediated synthesis and characterization of platinum nanocomposite and its anti-fungal activity against A. parasiticus and A. flavus. Micro Nano Lett 13: 1491-1496.

103. FDA (2020) Breast implants. Available online at: http:// www.fda.gov/ Medical Devices/ Products and Medical Procedures/ Implants and Prosthetics/ Breast Implants/ UCM064040.

104. Oberdorster G, Oberdorster E, Oberdorster J (2005) Nanotoxicology: An emerging discipline evolving from studies of ultrafine particles. Environ Health Perspect 113: 823-839.

105. Foldbjerg RB, Autrup H (2013) Mechanisms of Silver Nanoparticle Toxicity. Arch Basic Appl Med 1: 5-15.

106. Sabella S, Carney RP, Brunetti V, Malvindi MA, Al-Juffali N, et al. (2014) A general mechanism for intracellular toxicity of metal-containing nanoparticles. Nanoscale 6: 7052-7061.

107. Gliga AR, Skoglund S, Odnevall Wallinder I, Fadeel B, Karlsson HL (2014) Size-dependent cytotoxicity of silver nanoparticles in human lung cells: The role of cellular uptake, agglomeration and Ag release. Part Fibre Toxicol 11: 11.

108. Teow Y, Asharani PV, Hande MP, Valiyaveettil S (2011) Health impact and safety of engineered nanomaterials. Chem Commun 47: 7025-7038.

109. Sharifi S, Behzadi S, Laurent S, Laird Forrest M, Stroeve P, et al. (2012) Toxicity of nanomaterials. Chem Soc Rev 41: 2323-2343.

110. Oostingh GJ, Casals E, Italiani P, Colognato R, Stritzinger R, et al. (2011) Problems and challenges in the development and validation of human cell-based assays to determine nanoparticle-induced immunomodulatory effects. Part Fibre Toxicol 8: 8.

111. Neun BW, Dobrovolskaia MA (2011) Characterization of nanoparticles intended for drug delivery. ed. S. E. McNeil, Humana Press, Totowa, NJ 121–130.

112. Dobrovolskaia MA, Neun BW, Clogston JD, Grossman JH, McNeil SE (2014) Choice of method for endotoxin detection depends on nanoformulation. Nanomedicine 9: 1847-1856.

113. Gao J, Liang G, Zhang B, Kuang Y, Zhang X, et al. (2007) FePt@CoS(2) yolk-shell nanocrystals as a potent agent to kill HeLa cells. J Am Chem Soc 129: 1428-1433.

114. Asharani PV, Xinyi N, Hande MP, Valiyaveettil S (2010) DNA damage and p53-mediated growth arrest in human cells treated with platinum nanoparticles. Nanomedicine 5: 51-64.

115. Kutwin M, Sawosz E, Jaworski S, Hinzmann M, Wierzbicki M, et al. (2016) Investigation of platinum nanoparticle properties against U87 glioblastoma multiforme. Arch Med Sci 13: 1322-1334.

116. Gehrke H, Pelka J, Hartinger C, Blank H, Bleimund F, et al. (2011) Platinum nanoparticles and their cellular uptake and DNA platination at non-cytotoxic concentrations. Arch Toxicol 85: 799-812.

117. Pelka J, Gehrke H, Esselen M, Turk M, Crone M, et al. (2009) Cellular uptake of platinum nanoparticles in human colon carcinoma cells and their impact on cellular redox systems and DNA integrity. Chem Res Toxicol 22: 649-659.

118. Yamagishi Y, Watari A, Hayata Y, Li X, Kondoh M, et al. (2013) Acute and chronic nephrotoxicity of platinum nanoparticles in mice. Nanoscale Res Lett 8: 395.

119. Yamagishi Y, Watari A, Hayata Y, Li X, Kondoh M, et al. (2013) Hepatotoxicity of sub-nanosized platinum particles in mice. Die Pharmazie-An International J of Pharmaceutical Sciences 68: 178-182.

120. Konieczny P, Goralczyk AG, Szmyd R, Skalniak L, Koziel J, et al. (2013) Effects triggered by platinum nanoparticles on primary keratinocytes. Int J Nanomed 8: 3963-3975.

121. Buchtelova H, Dostalova S, Michalek P, Krizkova S, Strmiska V, et al. (2017) Size-related cytotoxicological aspects of polyvinylpyrrolidone-capped platinum nanoparticles. Food Chem Toxicol 105: 337-346.

122. Zheng X, Chen W, Cui P, Wang Z, Zhang W (2014) Design of multifunctional FePt/GO nanocomposites for targeting, dual-modal imaging diagnostic and in situ therapeutic potential theranostic platform. RSC Adv 4: 58489-58494.

123. Chen W, Zheng X, Li S, Zhang W, Wen X, et al. (2015) Nanopart Res 17: 1-10.

124. Ghosh S, Nitnavare R, Dewle A, Tomar GB, Chippalkatti R, et al. (2015) Novel platinum-palladium bimetallic nanoparticles synthesized by Dioscorea bulbifera: Anticancer and antioxidant activities. Int J Nanomed 10: 7477-7490.

125. Asharani PV, Sethu S, Vadukumpully S, Zhong S, Lim CT, et al. (2010) Investigations on the structural damage in human erythrocytes exposed to silver, gold, and platinum nanoparticles. Adv Funct Mater 20: 1233-1242.

126. Cheng HJ, Wu TH, Chien CT, Tu HW, Cha TS, et al. (2016) Corrosion-activated chemotherapeutic function of nanoparticulate platinum as a cisplatin resistance-overcoming prodrug with limited autophagy induction. Small 12: 6124-6133.

127. Chien CT, Yan JY, Chiu WC, Wu TH, Liu CY, et al. (2013) Caged Pt nanoclusters exhibiting corrodibility to exert tumor-inside activation for anticancer chemotherapeutics. Adv Mater 25: 5067-5073.

128. Park EJ, Kim H, Kim Y, Park K (2010) Intratracheal instillation of platinum nanoparticles may induce inflammatory responses in mice. Arch Pharmacal Res 33: 727-735.

 

129. Crist RM, Grossman JH, Patri AK, Stern ST, Dobrovolskaia MA, et al. (2013) Common pitfalls in nanotechnology: Lessons learned from NCI’s nanotechnology characterization laboratory. Integr Biol 5: 66-73.