Next Article in Journal
Rational Design of Non-Toxic Multidrug Combinations Demonstrates Durable and Hypoxia-Enhanced Efficacy Against Renal Cell Carcinoma
Previous Article in Journal
Physiologically Based Pharmacokinetic Modeling and Simulations in Lieu of Clinical Pharmacology Studies to Support the New Drug Application of Asciminib
Previous Article in Special Issue
Recent Advances in Nanomedicine: Cutting-Edge Research on Nano-PROTAC Delivery Systems for Cancer Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Latest Achievements in the Development of Nanoparticle-Based Drug Delivery Systems of Pt Drugs and Prodrugs in Cancer Therapy

1
Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy Bucharest, 020021 Bucharest, Romania
2
Department of Dental Medicine, Faculty of Medicine and Pharmacy, University of Oradea, 10 1st Decembrie Street, 410073 Oradea, Romania
3
Department of Medical Semiology, “Sf. Ioan” Emergency Clinical Hospital, “Carol Davila” University of Medicine and Pharmacy, 042122 Bucharest, Romania
4
Department of Surgical Specialties, Faculty of Medicine and Pharmacy, University of Oradea, 10 1st Decembrie Street, 410073 Oradea, Romania
5
ICU II Toxicology, Clinical Emergency Hospital, 014461 Bucharest, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceutics 2025, 17(10), 1267; https://doi.org/10.3390/pharmaceutics17101267
Submission received: 14 August 2025 / Revised: 11 September 2025 / Accepted: 25 September 2025 / Published: 26 September 2025
(This article belongs to the Special Issue Prodrug Strategies for Enhancing Drug Stability and Pharmacokinetics)

Abstract

Even though Pt(II)-based drugs represent the standard in cancer therapy, their use is seriously limited by severe side-effects (renal toxicity, allergic reactions, gastrointestinal disorders, hemorrhage and hearing loss), drug resistance and a grim prognosis. This review presents the results of multiple studies showing different nanoparticle-based platforms as delivery agents in order to overcome these drawbacks. The approach of using nanoparticle-based drug delivery systems of Pt drugs and prodrugs is promising due to key advantages like specific targeting and thereby reduced toxicity to healthy cells; increased stability in the bloodstream; multiple mechanisms of action such as stimulating anti-tumor immunity, responding to environmental stimuli (light, pH, etc.), or penetrating deeper into tissues; enhanced efficacy by their combination with other therapies (chemotherapy, gene therapy) to amplify the anti-tumor effect. However, certain challenges need to be overcome before these solutions can be widely applied in clinics. These include issues related to biocompatibility, large-scale production, and regulatory approvals. In conclusion, using nanoparticles to deliver Pt-based drugs represents an advanced and highly promising strategy to make chemotherapy more effective and less toxic. Nonetheless, further studies are required for the better understanding of intracellular mechanisms of action, toxicity and the pharmacokinetics of nanoparticles, and physical–chemical standardization.

Graphical Abstract

1. Introduction

Transition metal complex-based medicine represents a promising approach for different therapies due to their wide range of actions (anti-inflammatory, antibacterial, antifungal, anticancer), as they can interact with various biological targets, such as deoxyribonucleic acid (DNA) or enzymes, and can induce the formation of reactive oxygen species (ROS) [1]. As a result, researchers seek to discover new metal-based anticancer drugs like Pd(II)-based ones, with lower side-effects. However, Pt(II)-based drugs continue to represent the standard in cancer therapy [1]. The synthesis of cisplatin, cis-[Pt(II)(NH3)2Cl2] ([PtCl2(NH3)2] or CDDP), a Pt(II) complex, was an important discovery representing proof of Pt(II)-containing compounds’ potential as anticancer drugs and encouraged the study of other transition metal-containing complexes [2,3]. However, their use in different schemes is seriously limited by severe side-effects, drug resistance and a grim prognosis [4]. Consequently, the research subject moved to Pt(IV)-based prodrugs, which can exert their anticancer effects after reduction to their Pt(II) counterparts, having fewer side-effects. Therefore, several Pt(IV) compounds, like ormaplatin or tetraplatin, iproplatin and oxoplatin, are the subject of current research [5,6,7]. To overcome conventional Pt-based drug impediments, the usage of supramolecular chemistry was proposed to produce novel delivery platforms integrating the biological properties of Pt compounds and new supramolecular structures, such as nanoparticle (NP)-based delivery systems [1,8]. Besides NP-based delivery and prodrug methods, ligand modification strategies were employed to increase the selectivity of anticancer agents and reduce their side-effects [1]. NP-based drug delivery platforms seem to deliver Pt(IV) prodrugs more selectively and safely to cancer cells, but clinical outcomes were nevertheless unpredictable because of insufficient in vivo pharmacokinetics studies [9].
In this work, we aimed to present some of the most recent research in the field of nanoparticle-based drug delivery systems of Pt anticancer drugs, focusing on their structure, pharmacokinetics, mechanism of action and alternatives of reducing the severe side-effects of conventional cancer therapy.

2. Conventional Pt(II)-Based Cancer Therapy and Its Limitations

Ever since cisplatin (Figure 1) received approval from the U.S. Food and Drug Administration (FDA) for cancer therapy in the 1970s, Pt-based anticancer drugs have become the standard of chemotherapy treatments. However, their wider use is highly limited due to severe side-effects and drug resistance [10].
Cisplatin has been used for the therapy of an array of human tumors like bladder, head and neck, lung, ovarian, and testicular cancers, and against various types of cancers, including carcinomas, germ cell tumors, lymphomas, and sarcomas [11]. It can induce cancer cell death by crosslinking with the purine bases on the DNA, interfering with DNA repair processes, eventually leading to DNA damage [11].
Even though cisplatin has been used for the treatment of numerous human cancers and is effective against various types of cancers, its use is limited by drug resistance and numerous undesirable side-effects such as severe kidney problems, allergic reactions, decreased immunity to infections, gastrointestinal disorders, hemorrhage, and hearing loss especially in younger patients.
The mechanisms of their production and the associated complications and symptoms are depicted in Table 1.
Developing drug resistance is common in cisplatin therapy, and it can occur due to high thiol-containing species (especially glutathione (GSH)) levels within the cancer cell and adenosine triphosphate (ATP)-dependent GSH S-conjugate pumps [20]. Several processes are considered in explaining the influence of GSH on cisplatin transport: GSH may function as a cofactor in the ATP-dependent MRP2-mediated efflux of Pt(GS)2—a conjugate formed between cisplatin and elevated intracellular GSH—thereby contributing to cisplatin resistance in cancer cells; GSH acts as a cytoprotective agent—highly elevated in cancer cells due to the enhanced activity of γ-glutamylcysteine synthetase (the rate-limiting enzyme in GSH synthesis) and therefore being able to regulate the intracellular redox equilibrium, which contributes to cisplatin resistance; high levels of GSH, by chelating Cu, decreasing Cu levels within cells, and can therefore induce the overexpression of the high-affinity Cu transporter (hCtr1) which also functions as a cisplatin transporter—by mediating cisplatin uptake, its sensitivity to this drug is increased (Figure 2) [21,22].
The importance of each of these mechanisms might depend on the specific type of cancer cell and its physiological state. While ATP-dependent MRP2-mediated efflux of cisplatin requires GSH, Pt(GS)2 formation as an important step for cisplatin elimination remains somewhat controversial as different studies show different results [21]. Additionally, cisplatin can induce the upregulation of γ-GCSh and MRP, thus enhancing the cisplatin efflux [21]. Nevertheless, transfection of recombinant DNA coding only for the γ-GCSh subunit leads to an increase in GSH levels in the transfected cells, with a surprising hypersensitivity instead of resistance to cisplatin toxicity, which was proved to be linked with the enhanced uptake of cisplatin in these transfected cells through the upregulation of hCtr1 [21]. Future investigations are needed to clearly demonstrate the role of GSH in regulating the cisplatin sensitivity of cancer cells.

3. Pt(IV) Anticancer Prodrugs

Pt(IV) compounds were synthesized and preclinically (mostly only in vitro) tested to counteract the severe side-effects and drug resistance associated with classical Pt(II) compounds [23]. The octahedral Pt(IV) compounds, that are kinetically inactive, were developed using different synthesis techniques such as conjugation with lipid chains to increase lipophilicity; combination with small peptide segments or NPs to increase drug delivery efficacy; incorporation of bioactive ligands to the axial positions of Pt(IV) compounds for dual-function effects, such as histone deacetylase inhibitors, p53 agonists, alkylating agents, and nonsteroidal anti-inflammatory molecules—molecules that are released after the reduction reaction of the prodrugs to their corresponding Pt(II) active counterparts [24,25,26,27]. This reduction process occurs within the cancer cell due to the redox disequilibrium at the high metabolic rate, increased mitochondrial dysfunction, highly activated cell signaling pathways and fast peroxisomal activities [26,27].

4. Nanoparticle-Based Drug Delivery Systems of Pt Drugs in Cancer Therapy

There are many research directions regarding the next generation Pt chemotherapy and the NP-based delivery platforms of Pt complexes (such as various Pt-polymer complexes, micelles, dendrimers and liposomes) providing promising preclinical and clinical results [28]. Several properties and mechanisms of action of NPs encapsulating Pt drugs were studied (Table 2).
There are many biological models used to assess the performance of NPs delivering Pt-based drugs or prodrugs, such as cancer cell lines (for example, MCF-7, HeLa and A549), mouse xenograft models, syngeneic mouse models or patient-derived xenografts [29,30,31]. For the approval of Pt-based nanoformulations in clinical practice, further research and clinical validation are required to confirm the promising results of preclinical studies. Different platforms, such as lipid-based NPs, polymeric NPs, inorganic NPs, and extracellular vesicles were proved to provide increased drug targeting, bioavailability, reduced systemic toxicity in preclinical and clinical studies, the capacity to reverse drug resistance and the potential to be used in combination therapy. However, problems linked to biocompatibility, scalability, and regulatory approval need to be solved for widespread clinical adoption [32].

4.1. Strategies Used for a More Efficient Cancer Treatment

4.1.1. Mitochondria Targeting

It was thought and further demonstrated that anticancer drugs targeting mitochondria could potentially overcome or reverse cisplatin resistance [33]. Mitochondria have a central place in the study of tumoral resistance to cisplatin for two reasons. Firstly, the reprogramming and variability of tumor metabolism contributes to cisplatin resistance in cancer cells by enabling a switch between mitochondrial oxidative phosphorylation and glycolysis. Secondly, mitochondrial DNA is more prone to being damaged by Pt-based drugs, and this results in affected mitochondrial oxidative phosphorylation [33].
The latest achievements in mitochondria-targeting are pointing to a self-assembled drug delivery nanoplatform formed by lonidamine (LND)-S-S-Pt-triphenylphosphine (TPP)/β-cyclodextrin-grafted hyaluronic acid (HA-CD) synthesized to destroy cisplatin-resistant lung cancer cells, with every component comprising the nanomolecule bearing an important role: HA-CD can find the CD44 receptor on the tumor cell membrane; TPP may target the mitochondria, where the disulfide bond linking LND and the Pt(IV) prodrug is broken by the high levels of GSH, leading to the release of LND that can inhibit glycolysis and damage these organelles; the Pt(IV) molecule is reduced to cisplatin by GSH and further causes mitochondrial damage by targeting the mitochondrial DNA (mtDNA). By decreasing the intracellular level of GSH, this nanoplatform overcame GSH-mediated chemotherapy resistance [33].
Another recent method for targeting mitochondria is represented by the linkage of oxoplatin with lithocholic acid that self-assemble in water to spherical-shaped NPs. Their mechanisms of action might require the reduction in the Pt(IV) core to Pt(II) and simultaneous release of lithocholic acid, both being important for the anticancer effect. It was showed that the complex with the greatest potency was the one bearing a heptanoate rest linked to the Pt(IV) core at the axial trans position to the lithocholic acid rest [34]. It can affect the cancer cell by various mechanisms: halting the cell cycle in the S and G2 phases, affecting DNA, disrupting mitochondrial membrane potential, increasing ROS production, affecting mitochondrial bioenergetics in PC3 cells associated with human prostate cancer, upregulating pro-apoptotic proteins and reducing anti-apoptotic ones from the B-cell lymphoma 2 (BCl-2) family [34,35,36] (Table 3).
The general mitochondrial effects of these compounds are depicted in Figure 3.

4.1.2. Increased Blood Stability

The study of pharmacokinetics is pivotal for the understanding of drug therapeutic potential. Optimizing absorption, delivery and elimination of Pt-based anticancer drugs is essential to increase antitumor efficacy, decrease side-effects and enhance inhibition of the growing of tumors, and a type of NPs, designated PEG-OXA NPs, appear to exhibit these characteristics [9]. PEG-OXA NPs are formed from a new Pt(IV) complex derived from oxaliplatin (OXA), polyethylene glycol (PEG)-OXA, respectively, that has two hydrophobic lipid rests and the hydrophilic PEG in axial positions and can further self-assemble in micellar NPs in an aqueous environment [9]. These NPs have raised the interest of researchers for they seem to facilitate a rapid release of bioactive OXA in tumor cells and manifest a high stability in blood in vitro and an increased half-life in vivo, thus providing an emerging solution for anticancer drug-targeted delivery [9] (Figure 4).
Besides the novel proposal of PEG-OXA NPs, other recent approaches including nanocrystals, self-assembled PtNPs with protein coronas, nano-hydrogels and mesoporous silica nanoparticles (MSN) are depicted in Table 4.

4.1.3. Increased Anti-Tumoral Immunity

One study described a novel NP-based delivery platform for the targeted delivery of Pt(IV) containing a trisulfide bond, which exhibited increased in vivo anticancer properties and reduced liver damage (compared to cisplatin), namely NP(3S)s [41]. Due to high GSH levels within tumor cells, these compounds might release active Pt(II) and hydrogen sulfide (H2S) that synergistically act together by impacting DNA (the classical mechanism of Pt(II)); activating the stimulator of interferon gene (STING) pathway; activating T cells and subsequently increasing antitumor immunity; and activating the OS mechanisms [41].
Another efficient combination of Pt-based chemotherapy and immunotherapy was shown for osteosarcoma, a type of cancer where most immune checkpoint blockades were not proved to have good results. For instance, a type of NP designated as NP-Pt-IDOi presented a great antitumor effect [42]. NP-Pt-IDOi was synthesized from a ROS-responsive amphiphilic polymer containing thiol-ketal bonds within its structure by co-encapsulating both a Pt(IV) prodrug and an indoleamine-(2/3)-dioxygenase inhibitor (IDOi) [42]. After entering an environment with high levels of ROS, such as the one in cancer cells, the NP released both encapsulated molecules. The Pt(IV) prodrug was reduced to its Pt(II) counterpart, thus damaging DNA and inducing the STING pathway, with a subsequent increase in antitumoral immunity. The concomitantly released IDOi inhibited the metabolism of tryptophan and, consequently, further activated the cytotoxic CD8+ T cells and enhanced the anticancer effects of the nanostructure [42]. Moreover, another way to increase therapy outcome for osteosarcoma by increasing the targeting capacity of the drug and decreasing or reversing the tumor-associated immunosuppression was proved to be achieved by the self-assembly of an OXA-based Pt(IV) prodrug amphiphile, namely Lipo-OXA-ALN, in which alendronate (ALN) acts as a targeting agent for osteosarcoma cells [43]. The effects of the alleged NPs were an enhanced intracellular uptake of OXA, inhibition of cancer cell activity, increased targeting capacity (to spare the healthy bone), increased antitumoral immunity, increased M1/M2 macrophage ratio and modification in the tumoral microenvironment [43].
A further effect of Pt-based drugs, like OXA, is that they can induce immunogenic cell death (ICD) in cancer cells by activating the immune system, and, as a result, strategies to enhance these ICD effects were studied [44]. As a result, it was proved that an NP-encapsulating OXA and a near-infrared-II (NIR-II) photothermal agent IR1061 could achieve this enhancement of ICD in tumors after irradiation with NIR-II, leading to a mild increase in temperature, but with important effects. Increased Pt-DNA binding, higher DNA damage and apoptosis, with subsequent stronger ICD were reported [44]. Such a strategy remarkably increased the therapy results for triple-negative breast cancer 4T1 in comparison with either OXA or NIR-II-based photothermal therapies alone (Figure 5) [44].
Current research focuses on Pt-conjugated iron NPs, hyperthermia sensitive Pt NPs and immunogenic bifunctional nanoparticles, as well (Table 5).

4.1.4. Multistimuli-Responsive Drug Delivery Systems

To overcome the drawbacks of conventional Pt(II) chemotherapy, stimuli-responsive nanoplatform-based delivery systems were considered due to their great potential for exact spatiotemporal drug deliverance [48]. Multistimuli-responsive drug delivery systems are employed to specifically target the site of a drug’s action and release the therapeutic agent by responding to multiple internal and external signals, such as electrical and magnetic fields, acidity, temperature, light irradiation, and redox stimuli [49].
pH-mediated drug delivery methods are on the rise as promising anticancer therapeutic approaches [50]. Liposomes formed from lipid-encapsulated CaCO3 (representing the acidic pH-responsive element of the liposomes) carrying a Pt(IV) prodrug and biotin were proved efficient against thyroid cancer cells, posing high stability and rapid pH-mediated degradation, which, consequently, may direct the pH-responsive delivery of both anticancer molecules [50]. The intracellular effects within the cancer cells of these NPs are increased OS, mitochondrial damage, and DNA damage, ultimately leading to cancer cell death [50].
Another stimuli-responsive approach relies on activation upon irradiation and reduction [48]. Some upconversion NP-based nanoplatforms can incorporate, through hydrophobic interactions, an octahedral Pt(IV) prodrug with an octadecyl aliphatic chain and phenylbutyric acid (i.e., a histone deacetylase inhibitor, with additional anticancer effects) as axial ligands. By further modifications through the binding of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine polyethylene glycol (PEG) 2000 and the peptide arginine-glycine-aspartic, an additional increase in tumor specificity and reduction in systemic toxic effects occur. After upconversion luminescence irradiation and GSH reduction, the NPs release the Pt(II) active counterparts and phenylbutyric acid in the cancer cell cytoplasm, leading to tumor cell death [48].
Synthesis of NPs with both photo- and pH-sensitivity as an encouraging anticancer therapeutic approach was attempted [51]. One such NP is formed from a Pt(IV) prodrug, which can be activated to its Pt(II) active counterpart by ultraviolet A (UVA) irradiation, and demethylcantharidin. Being an inhibitor of protein phosphatase 2A, demethylcantharidin released from the NP in an acidic medium increases the formation of hyper-phosphorylated protein kinase B (pAkt) to halt the DNA repair mechanisms [51,52]. Additionally, a metallo-nano prodrug was developed by attaching a photosensitizer-conjugated Pt(IV) complex to a polymeric core and chelating it with iron. In an acidic microenvironment, iron released from the NP allows the reduction in Pt(IV) to Pt(II) after light irradiation, leading to chemotherapy and photodynamic therapy [40]. Besides these effects, the NP is supposed to cause ferroptosis and tumor-associated macrophage polarization with additional anticancer consequences [53].
A recent study described a nanozyme-based photocatalytic method for Pt(IV) prodrug activation [54]. The authors reported that an AuNP covered by thiol ligands with 1,4,7-triazacyclononane headgroups encapsulating riboflavin-5′-phosphate (FMN) promoted the photocatalytic reduction in a Pt(IV) prodrug to cisplatin in the presence of a reducing agent [54]. Another current approach employs micellar NPs delivering Pt(IV) prodrugs with photosensitivity which, upon UVA irradiation, show enhanced cytotoxicity towards cancer cells, with reduced side-effects and an increased circulation time [55]. Moreover, two recent studies have brought attention to the development of compounds that can be selectively activated upon irradiation and provide spatial and temporal control over the treatment [56]. In this regard Pt(IV) prodrug NPs that could be activated by deeply penetrating ultrasound radiation are considered as a promising alternative to the common activation of Pt(IV) prodrugs through ultraviolet or blue light irradiation that can reach only the surface, being ineffective against deep or very thick tumors [56] A recent study showed that self-assembled P-Rf/cisPt(IV) NPs transporting a Pt(IV) prodrug and riboflavin, displayed enhanced anti-tumoral properties through an increased activation of the prodrug upon low-intensity ultrasound radiation through superoxide anions, whose formation was increased by riboflavin [57].
Stimuli-responsive techniques might also be employed to overcome resistance to conventional chemotherapy [58]. NPs containing both a Pt(IV) prodrug and ursolic acid known for a higher circulation time, tumor accumulation and antitumor activity but without the side-effects associated with conventional therapy, were proved to reverse cisplatin resistance in ovarian cancer in an intracellular reductive and acidic microenvironment through both molecules released [58]. Self-assembled dual-drug polymer micellar NPs and Pt-coordinated dual-responsive nanogels have gained interest as potential anticancer drugs as well (Table 6).
The principles of multistimuli-responsive drug delivery systems are presented in Figure 6.

4.1.5. Combination Chemotherapy

Nanotechnology-facilitated combinational delivery of anticancer molecules represents a new promising strategy in tumor therapy [60]. Several compounds co-delivered with Pt-based drugs and prodrugs through nanoplatforms were proposed.
Adding Fe compounds to induce the intracellular cascade reaction and generate sufficient HO for ferroptosis therapy is of great interest [4,61,62,63,64]. H2O2 depletion-mediated tumor anti-angiogenesis, apoptosis and ferroptosis were found to be achieved by a Pt(IV) prodrug-delivery nanoplatform coated by ferric oxide, which protects the NP in circulation, enhancing delivery to tumor cells, with a selenium core [61]. Several cytotoxic mechanisms on cancer cells were described: ferroptosis caused by HO accumulation from H2O2 induced by Fe(II) in acidic cancerous medium; inactivation of glutathione peroxidase 4 (GPX4); OS augmentation and enhancement of ferroptosis as additional effects of Pt(IV)-reduction to Pt(II) complexes; inhibition of angiogenesis resulting from lower levels of vascular endothelial growth factor-A (VEGF-A); mitochondrial damage and affected angiogenesis caused by the exposed selenium core [61]. Polypeptide vehicles carrying cisplatin and Fe3O4 NPs were synthesized. They were employed as theranostic agents for combination therapy guided by T2-weighted magnetic resonance imaging (MRI). The ferroptosis, caused by the HO mechanism mentioned above, was implied with the difference that Fe3O4 NPs can also be used for the T2-weighted MRI of the tumor [62]. Another study proposed the synthesis of a GSH-responsive NP containing a disulfide bond-based amphiphilic polyphenol, a dopamine-modified cisplatin prodrug and Fe(III) integrated through coordination reactions between Fe(III) and phenols. Increased OS led to ICD, promoting the maturation of dendritic cells and finally enhancing the antitumor immune response [4].
One promising way for targeted drug delivery might be the conjugation of monoclonal antibodies to different nanosystems. As such, cetuximab was used to decorate a Pt-based drug delivery nanoplatform activated upon NIR irradiation. Cetuximab attachment resulted in a nanosystem capable of specific delivery of drugs to epidermal growth factor receptor (EGFR)-hyperexpressing cancer cells in epidermoid carcinoma [65].
In addition, nanoformulation-based drug delivery systems of Pt(IV) prodrugs, like OXA, functionalized with axial ligands represented by other chemotherapeutic agents, like gemcitabine or capecitabine, were proposed. The additional drugs were released upon reduction within cancerous cells with synergistic antitumoral effects and were proved effective in a representative colorectal cancer cell model, with the complexes using gemcitabine being the most active [66].
Multiple studies proposed the association of Pt-based chemotherapy with phototherapy and radiotherapy [20,67,68,69]. The first obvious additional effect besides the therapeutic one is the possibility of simultaneously monitoring the delivery of the Pt-prodrugs by bioimaging [67,68,70]. When synthesis of NPs containing both a Pt(IV) prodrug and a mitochondria-targeting NIR photosensitizer, namely IR780, was developed, it was found that the NIR laser irradiation led to the inhibition of hyperactive energy-generating processes of mitochondria through both photothermal and photodynamic mechanisms, leading to a hindrance of processes requiring energy in the form of ATP, like drug efflux out of the cancer cells or repair of damaged nuclear acids [67,71]. As a result, decrease in pivotal proteins of the nucleotide excision repair pathway activity was induced, thereby enhancing the effects of the Pt-based drugs on the cancer cell’s nucleic acids [67]. Moreover, these NPs allow drug delivery and treatment monitoring through NIR fluorescence and photoacoustic imaging [67]. In another study, Pt(IV)-based polymeric prodrug PVPt with amphiphilic properties was used to encapsulate a theranostic agent, the modified cyanine dye 1-(2-hydroxyethyl)-2-((E)-2-((E)-3-((E)-2-(1-(2-hydroxyethyl)-3,3-dimethylindolin-2-ylidene)ethylidene)-2-chlorocyclohex-1-en-1-ly)vinyl)-3,3-dimethyl-3H-indol-1-ium bromide (HOCyOH or Cy), respectively, through hydrophobic interactions, resulting in NPs formed by self-assembly [68]. These NPs could undergo disassembly and activation under acidic, reductive conditions and NIR laser irradiation, being accompanied by photothermal conversion and increased OS [68]. Regarding the association with radiotherapy, researchers are concentrating on new, safe, and redox-responsive NPs with a higher disulfide density and a better ability to load Pt(IV) prodrugs. These NPs would be useful in reversing cisplatin resistance and improving anticancer outcome by directing more cytotoxic compounds towards tumor cells, scavenging GSH, and causing mitochondrial damage, which would increase Pt-DNA cross-linking and make these cancer cells more sensitive to X-ray radiation. As a result, they are considered promising candidates for anticancer chemoradiotherapy, particularly for cervical cancers [20].
Breaking the redox balance is a promising way to maximize the efficacy of conventional Pt-based cancer therapy because the anticancer activity and drug resistance of chemotherapy are related to the redox state of tumor cells [72]. Therefore, NPs formed from polymers containing a diselenium bond in the main chain surrounding a Pt(IV) prodrug were synthesized. They depleted GSH while increasing the level of OS at the same time, thereby disrupting intracellular redox balance and increasing the antitumoral effect of conventional cisplatin therapy [72].
Additionally, the association of Pt(IV) prodrugs and a ubiquitin-specific protease 1 inhibitor (USP1i) within NPs was found to have an enhanced antitumoral effect in comparison with conventional therapy. USP1i seems to affect the DNA damage repair processes by targeting USP1 to increase the cytotoxic effects of cisplatin against cancer cells [31].
It is widely known that doxorubicin, along with other agents, causes hypoxia-induced multi-drug resistance, resulting in poor therapy results [73]. This process may be reversed and stopped by NPs formed as a co-self-assembly of a PEG-Pt(IV) prodrug and doxorubicin which, under light irradiation, can lead to the generation of oxygen to reverse tumor hypoxia and release active Pt(II) and doxorubicin. Increased OS and enhanced cytotoxicity effects compared to both therapies alone were reported [73].
Another approach combines in NPs an OXA-derived Pt(IV) prodrug and a peptide that targets mitochondria aiming to impact the molecular processes that need energy in the form of ATP [71]. These NPs are supposed to become activated by the acidic tumor microenvironment and increased concentration of GSH within cancer cells, leading to the accumulation of high intracellular levels of Pt-drugs and increased anti-tumoral effects, respectively [71].
Molecules to assess the therapeutic efficacy of the treatment were added besides anticancer drugs and drug tracers [74]. Such NPs were developed from a Pt(IV) compound, a NIR-II fluorophore tracer and a peptide that may be split by caspase-3 and serve as an apoptosis indicator. The combination is considered a promising strategy to determine both the drug pharmacokinetics and its therapeutic success [74].
One strategy potentially effective against cisplatin-resistant tumors might be the synthesis of NPs delivering not only the Pt drug, but also curcumin [75]. This co-delivery platform enhances stability, as well as solubility of curcumin and, additionally, release of the Pt drug and the antioxidant occur in reductive environments, such as those from tumors, with consequent synergistic effects [75] (Table 7).
The possibilities of combination therapy using Pt encapsulated by NPs as delivery systems are presented in Figure 7.

4.1.6. Bioorthogonal Reactions Catalyzed by PtNPs

Pt complexes, in addition to their classical role as cytotoxics, were recently proved to perform bioorthogonal reactions in living organisms [76,77]. Even though some methods based on this approach do not directly employ Pt-based prodrugs, PtNPs serve as catalytic centers for a variety of reactions [76]. For instance, the synthesis of catalytic nanoreactors, represented by PEGylated Pt NPs with special properties, such as a dendritic structure and surface shielding by Pt-S-bonded PEG allow the compound to act within the complex intracellular microenvironment of cancer cells, thus enhancing the in situ biorthogonal activation of anticancer prodrugs [76]. Additionally, a combination between biorthogonal chemistry and an inhalation technique was employed to reverse cisplatin resistance in nonsmall cell lung carcinoma (NSCLC). The effect was achieved by co-delivery of ethacraplatin (i.e., a Pt(IV) prodrug) and nitric oxide (NO) by micelles, specifically targeting cancer cells after inhalation [77]. After arriving in the acidic tumor microenvironment, the Pt(IV) prodrug is released, inhibits GSH S-transferase and reduces the concentration of GSH, leading to increased sensitivity to cisplatin, with additional advantages associated with the release of NO (Figure 8) [77].
A previous study reported the formation of self-assembled coordinative NP BDCNs, which deliver both a Pt(IV) prodrug and a NO prodrug and that accumulate in the tumor. After reduction in the Pt(IV) prodrug to the active Pt(II), activation of NO prodrug occurs by a depropargylation reaction, leading to the release of NO, with synergistic effects [78]. The results of this study show an enhanced the efficiency of bioorthogonal reactions by overcoming the problems posed by the separate administration of the Pt(IV) compound and NO prodrug, such as pharmacokinetic issues [78] (Table 8).

4.1.7. Increased Lipophilicity

Novel nanosystems with enhanced hydrophobicity and passive diffusion capacity, increasing cancer cell accumulation, while combating the drawbacks of conventional therapy and improving the simultaneous chemo-/radiotherapy are emerging. In their development the following objectives are pursued: the deliverance of Pt-based drugs and prodrugs, the pass through the blood–brain barrier, and specifically, the capacity to target malignant cells [79].
It was proved that Pt(IV) prodrugs with increased tumor selectivity formed using biotin and naproxen or stearate in axial position had enhanced anticancer effects. These improved anticancer properties were due to lipophilicity rather than the expression of biotin receptors [80].
Analyzing the importance of lipophilicity of Pt(IV)-based compounds, in an in vitro study, it was suggested that for the most hydrophobic compound tested (namely, diamminedichloridodioctanoatoplatinum(IV)) the formation of nanoaggregates resulted in higher cellular uptake by both passive diffusion and endocytosis than by passive diffusion alone [81]. However, since this compound is active at nanomolar concentrations at which the aggregation in culture media is almost inexistent, this phenomenon should not significantly impact its antiproliferative activity [81]. Another study studied a type of polymeric NP, namely [Pt(DACH)(OAc)(OPal)(ox)] incorporated PLGA NPs, delivering a lypophilic Pt(IV) complex, that showed increased anticancer properties even against cisplatin-resistant cells and reduced systemic toxicity [82] (Table 9).
The advantages of nanoformulations with increased lipophilicity are presented in Figure 9.

4.1.8. Targeting Both Malignant and Non-Malignant Cells

One more promising efficient cancer therapy in comparison with the conventional approaches is to target both tumoral and non-tumoral cells at the same time. For this purpose, NP-based codelivery of a Pt(IV) prodrug and Sotuletininb (BLZ-945) showed, after irradiation with light having a 660 nm wavelength, a contraction to small Pt(IV) prodrug-containing NPs and drug deliverance deep inside the tumor, while releasing BLZ-945 around the tumor-associated blood vessels, targeting and damaging tumor-associated macrophages [83].
Even though novel approaches were created for cancer therapy by developing various nanoformulation-based drug delivery systems, most of the studies are limited only to in vivo and in vitro research and the number of approved nanodrugs did not significantly increase throughout the years [84,85]. There are also some drawbacks of the usage of NP-based treatments, such as associated-immunotoxicity, long term toxicity, neural side-effects and costly synthesis [84,86] (Table 10).

4.2. Materials Used in Producing NPs for a More Efficient Cancer Treatment

4.2.1. siRNA Technology

The siRNA technology also appears to play a crucial role in regulating Pt-drug sensitivity.
For instance, epigallocatechin gallate-based NPs carrying both Pt(IV) prodrugs and anti-casein kinase 2 alpha 1 (CSNK2A1) siRNA exerted many beneficial effects by extending the circulation time for Pt-based drugs, increasing the accumulation of the drug within the cancerous cells, decreasing the Pt-drug’s renal side-effects and targeting CSNK2A1, by all of these increasing the tumor’s sensitivity to cisplatin [13].
Another study underlined the potential of utilizing the nuclear-targeting lipid Pt(IV) prodrug with amphiphilic and nuclear-targeting properties to increase Pt-DNA cross-linking formation and using siXkr8 (i.e., small interfering RNA targeting XK-related protein 8) to decrease immunosuppression. This RNA would downregulate the exposed phosphatidylserine (i.e., a phospholipid expressed on cancer cell membrane after Pt-based therapy that binds to corresponding receptors on immune cells, leading to lower antitumoral immunity), leading to improved chemoimmunotherapy. The new compound can thereby amplify the anticancer effects of conventional Pt-based drugs and suppress cancer recurrence [87]. Previous studies showed that self-assembled lipid NP LNP co-delivering XPF-targeted siRNA together with a cisplatin prodrug, can enhance cisplatin-associated cytotoxicity by silencing this nucleotide excision repair (NER)-related gene [88]. Additionally, self-assembled PLGA-PEG/G0-C14 NPs, co-delivering REV1/REV3L-specific siRNA and a cisplatin prodrug, can reverse cisplatin resistance by suppressing the genes involved in the error-prone translesion DNA synthesis [89] (Table 11).
The roles of siRNA technology in the NP-based drug delivery systems of Pt compounds are depicted in Figure 10.

4.2.2. Human Serum Albumin-Based NPs

In addition to the results stated above, precise drug delivery with reduced drug demand and lower side-effects could be achieved using human serum albumin (Figure 11).
Human serum albumin was chosen because studies proved that utilizing this protein is effective for active tumor targeting [90]. The reduction-responsive human serum albumin NPs conjugated with OXA demonstrated improved cytotoxicity and lower rates of resistance compared to the free drug utilization in the treatment of triple-negative breast cancer [90]. The same outcomes were reported for AbPlatin(IV) NPs, using human serum albumin as carrier for a lipophilic Pt(IV) prodrug, whose mechanisms of action were analyzed through multi-omics analysis. Modifications of the malignant cell membrane by alterations of glycerophospholipids and sphingolipids and modifications of purine metabolism were discovered [91]. Another study developed human serum albumin–Pt compound complex NPs, HSA-His242-Pt-Dp44mT NPs, which tend to specifically accumulate within cancer cells through the binding of human serum albumin to the secreted protein acidic and are rich in cysteine (SPARC) protein, highly expressed by cancer cells [92]. As a result of this specific binding, enhanced toxicity towards cancer cells and lower side-effects occur [92] (Table 12).

5. Conclusions

NP-based drug delivery systems represent a promising approach in overcoming the disadvantages of conventional Pt-based cancer therapy, namely chemoresistance and severe side-effects, that make the drug’s long-term administration difficult. Passive and active targeting cancer cells through specific drug delivery systems could lead to lowered drug demand and toxicity.
They use different strategies to achieve an improved anticancer effect, such as mitochondria targeting, increased stability in the circulation, increased anti-tumoral immunity, multistimuli-responsivity, combination chemotherapy, catalyzed bioorthogonal reactions, increased lipophilicity and the capacity to target both malignant and non-malignant cells. In addition, other materials or techniques used in combination with these NPs can improve their efficacy, such as the use of siRNAs, laser irradiation, deeply penetrating ultrasound radiation, and human serum albumin-based NPs. Due to their numerous mechanisms of action, NP-based drug delivery systems could enlarge therapeutic utilization for multiple cancers. Nevertheless, there are some challenges regarding the use of different types of NPs in the clinic, such as problems linked to biocompatibility, scalability, and regulatory approval.
Several future directions emerge for this type of nanomedicine. Further studies are still needed to better understand the molecular mechanisms of NPs inside different malignant cells in different patients. Additionally, for wide clinical administration, more studies are required to better analyze toxicity and pharmacokinetics, along with physical–chemical standardization.

Author Contributions

Conceptualization, V.I., G.M.I., S.V., A.T.T., I.S. and M.S.T.; methodology, V.I. and I.S.; writing—original draft preparation, V.I., G.M.I., S.V., A.T.T., I.S. and M.S.T.; writing—review and editing, V.I., G.M.I., S.V., A.T.T., I.S. and M.S.T.; supervision, M.S.T. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by the University of Oradea, Oradea, Romania.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

No new data was created in this study.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
DNAdeoxyribonucleic acid
ROSreactive oxygen species
OSoxidative stress
Ptplatinum
NPnanoparticle
GSHglutathione
ATPadenosine triphosphate
MRP2multidrug resistance protein 2
Cucopper
LNDlonidamine
TPPtriphenylphosphine
HA-CDβ-cyclodextrin-grafted hyaluronic acid
mtDNAmitochondrial deoxyribonucleic acid
Bcl-2B-cell lymphoma 2
PEGpolyethylene glycol
OXAoxaliplatin
STINGstimulator of interferon genes
IDOiindoleamine-(2/3)-dioxygenase inhibitor
ALNalendronate
ICDimmunogenic cell death
NIR-IInear-infrared-II
UVAultraviolet A
pAktphosphorylated protein kinase B
siRNAsmall interfering RNA
CSNK2A1casein kinase 2 alpha 1
siXkr8small interfering RNA targeting XK-related protein 8
Augold
FMNriboflavin-5′-phosphate
Feiron
OHhydroxyl radical
H2O2hydrogen peroxide
GPX4glutathione peroxidase 4
VEGF-Avascular endothelial growth factor-A
MRImagnetic resonance imaging
EGFRepidermal growth factor receptor
Cy1-(2-hydroxyethyl)-2-((E)-2-((E)-3-((E)-2-(1-(2-hydroxyethyl)-3,3-dimethylindolin-2-ylidene)ethylidene)-2-chlorocyclohex-1-en-1-ly)vinyl)-3,3-dimethyl-3H-indol-1-ium bromide
USP1iubiquitin-specific protease 1 inhibitor
Ssulfur
NSCLCnonsmall cell lung carcinoma
NOnitric oxide
BLZ-945Sotuletininb

References

  1. AlAli, A.; Alkanad, M.; Alkanad, K.; Venkatappa, A.; Sirawase, N.; Warad, I.; Khanum, S.A. A comprehensive review on anti-inflammatory, antibacterial, anticancer and antifungal properties of several bivalent transition metal complexes. Bioorganic Chem. 2025, 160, 108422. [Google Scholar] [CrossRef]
  2. Florea, A.M.; Büsselberg, D. Cisplatin as an Anti-Tumor Drug: Cellular Mechanisms of Activity, Drug Resistance and Induced Side Effects. Cancers 2011, 3, 1351–1371. [Google Scholar] [CrossRef]
  3. Spiro, T.G. Nucleic Acid-Metal Ion Interactions, 1st ed.; John Wiley & Sons, Inc.: New York, NY, USA, 1980; Volume 1, pp. 1–29. [Google Scholar]
  4. Zhang, X.; Zong, Q.; Lin, T.; Ullah, I.; Jiang, M.; Chen, S.; Tang, W.; Guo, Y.; Yuan, Y.; Du, J. Self-assembled metal-phenolic network nanoparticles for delivery of a cisplatin prodrug for synergistic chemo-immunotherapy. Biomater. Sci. 2024, 12, 3649–3658. [Google Scholar] [CrossRef] [PubMed]
  5. Rahman, A.; Roh, J.K.; Wolpert-DeFilippes, M.K.; Goldin, A.; Venditti, J.M.; Woolley, P.V. Therapeutic and Pharmacological Studies of Tetrachloro(d,l-trans)1,2-diaminocyclohexane Platinum (IV) (Tetraplatin), a New Platinum Analogue. Cancer Res. 1988, 48, 1745–1752. [Google Scholar] [PubMed]
  6. Pendyala, L.; Cowens, J.W.; Chheda, G.B.; Dutta, S.P.; Creaven, P.J. Identification of cis-dichloro-bis-isopropylamine platinum(II) as a major metabolite of iproplatin in humans. Cancer Res. 1988, 48, 3533–3536. [Google Scholar] [PubMed]
  7. Blatter, E.E.; Vollano, J.F.; Krishnan, B.S.; Dabrowiak, J.C. Interaction of the antitumor agents cis, cis, trans-Pt (IV)(NH3) 2Cl2 (OH)2 and cis, cis, trans-Pt (IV)[(CH3) 2CHNH2] 2Cl2 (OH)2 and their reduction products with PM2 DNA. Biochemistry 1984, 23, 4817–4820. [Google Scholar] [CrossRef]
  8. Jogadi, W.; Zheng, Y.R. Supramolecular platinum complexes for cancer therapy. Curr. Opin. Chem. Biol. 2023, 73, 102276. [Google Scholar] [CrossRef]
  9. Fu, Y.; Kong, Y.; Li, X.; Cheng, D.; Hou, Y.; Li, Y.; Li, T.; Xiao, Y.; Zhang, Q.; Rong, R. Novel Pt(IV) prodrug self-assembled nanoparticles with enhanced blood circulation stability and improved antitumor capacity of oxaliplatin for cancer therapy. Drug Deliv. 2023, 30, 2171158. [Google Scholar] [CrossRef]
  10. Zutphen, S.v.; Reedijk, J. Targeting platinum anti-tumour drugs: Overview of strategies employed to reduce systemic toxicity. Coord. Chem. Rev. 2005, 249, 2845–2853. [Google Scholar] [CrossRef]
  11. Dasari, S.; Tchounwou, P.B. Cisplatin in cancer therapy: Molecular mechanisms of action. Eur. J. Pharmacol. 2014, 740, 364–378. [Google Scholar] [CrossRef]
  12. Li, L.; Feng, C.; Zhang, W.; Qi, L.; Liu, B.; Wang, H.; Li, C.; Li, Z.; Tu, C.; Zhou, W. Mitigation of Cisplatin-Induced Nephrotoxicity and Augmentation of Anticancer Potency via Tea Polyphenol Nanoparticles’ Codelivery of siRNA from CRISPR/Cas9 Screened Targets. ACS Appl. Mater. Interfaces 2024, 16, 59721–59737. [Google Scholar] [CrossRef]
  13. Miller, R.P.; Tadagavadi, R.K.; Ramesh, G.; Reeves, W.B. Mechanisms of Cisplatin nephrotoxicity. Toxins 2010, 2, 2490–2518. [Google Scholar] [CrossRef] [PubMed]
  14. Makrilia, N.; Syrigou, E.; Kaklamanos, I.; Manolopoulos, L.; Saif, M.W. Hypersensitivity reactions associated with platinum antineoplastic agents: A systematic review. Met. Based Drugs 2010, 2010, 207084. [Google Scholar] [CrossRef] [PubMed]
  15. Abu-Sbeih, H.; Mallepally, N.; Goldstein, R.; Chen, E.; Tang, T.; Dike, U.K.; Al-Asadi, M.; Westin, S.; Halperin, D.; Wang, Y. Gastrointestinal toxic effects in patients with cancer receiving platinum-based therapy. J. Cancer 2020, 11, 3144–3150. [Google Scholar] [CrossRef]
  16. Elmorsy, E.A.; Saber, S.; Hamad, R.S.; Abdel-Reheim, M.A.; El-Kott, A.F.; AlShehri, M.A.; Morsy, K.; Salama, S.A.; Youssef, M.E. Advances in understanding cisplatin-induced toxicity: Molecular mechanisms and protective strategies. Eur. J. Pharm. Sci. 2024, 203, 106939. [Google Scholar] [CrossRef]
  17. Zhang, W.; Zhao, L.; Liu, J.; Du, J.; Wang, Z.; Ruan, C.; Dai, K. Cisplatin induces platelet apoptosis through the ERK signaling pathway. Thromb. Res. 2012, 130, 81–91. [Google Scholar] [CrossRef]
  18. Gao, A.; Zhang, L.; Zhong, D. Chemotherapy-induced thrombocytopenia: Literature review. Discov. Oncol. 2023, 14, 10. [Google Scholar] [CrossRef]
  19. Chattaraj, A.; Syed, M.P.; Low, C.A.; Owonikoko, T.K. Cisplatin-Induced Ototoxicity: A Concise Review of the Burden, Prevention, and Interception Strategies. JCO Oncol. Pract. 2023, 19, 278–283. [Google Scholar] [CrossRef]
  20. Luo, K.; Guo, W.; Yu, Y.; Xu, S.; Zhou, M.; Xiang, K.; Niu, K.; Zhu, X.; Zhu, G.; An, Z.; et al. Reduction-sensitive platinum (IV)-prodrug nano-sensitizer with an ultra-high drug loading for efficient chemo-radiotherapy of Pt-resistant cervical cancer in vivo. J. Control. Release 2020, 326, 25–37. [Google Scholar] [CrossRef]
  21. Chen, H.H.; Kuo, M.T. Role of glutathione in the regulation of Cisplatin resistance in cancer chemotherapy. Met. Based Drugs 2010, 2010, 430939. [Google Scholar] [CrossRef]
  22. Ishikawa, T.; Ali-Osman, F. Glutathione-associated cis-diamminedichloroplatinum(II) metabolism and ATP-dependent efflux from leukemia cells. Molecular characterization of glutathione-platinum complex and its biological significance. J. Biol. Chem. 1993, 268, 20116–20125. [Google Scholar]
  23. Ravera, M.; Gabano, E.; McGlinchey, M.J.; Osella, D. Pt(IV) antitumor prodrugs: Dogmas, paradigms, and realities. Dalton Trans. 2022, 51, 2121–2134. [Google Scholar] [CrossRef] [PubMed]
  24. Li, X.; Liu, Y.; Tian, H. Current Developments in Pt(IV) Prodrugs Conjugated with Bioactive Ligands. Bioinorg. Chem. Appl. 2018, 2018, 8276139. [Google Scholar] [CrossRef] [PubMed]
  25. Marotta, C.; Giorgi, E.; Binacchi, F.; Cirri, D.; Gabbiani, C.; Pratesi, A. An overview of recent advancements in anticancer Pt (IV) prodrugs: New smart drug combinations, activation and delivery strategies. Inorganica Chim. Acta 2023, 548, 121388. [Google Scholar] [CrossRef]
  26. Canil, G.; Braccini, S.; Marzo, T.; Marchetti, L.; Pratesi, A.; Biver, T.; Gabbiani, C. Photocytotoxic Pt (iv) complexes as prospective anticancer agents. Dalton Trans. 2019, 48, 10933–10944. [Google Scholar] [CrossRef]
  27. Iova, V.; Tincu, R.C.; Scrobota, I.; Tudosie, M.S. Pt(IV) Complexes as Anticancer Drugs and Their Relationship with Oxidative Stress. Biomedicines 2025, 13, 981. [Google Scholar] [CrossRef]
  28. Oberoi, H.S.; Nukolova, N.V.; Kabanov, A.V.; Bronich, T.K. Nanocarriers for delivery of platinum anticancer drugs. Adv. Drug Deliv. Rev. 2013, 65, 1667–1685. [Google Scholar] [CrossRef]
  29. Faderin, E.; Iorkula, T.H.; Aworinde, O.R.; Awoyemi, R.F.; Awoyemi, C.T.; Acheampong, E.; Chukwu, J.U.; Agyemang, P.; Onaiwu, G.E.; Ifijen, I.H. Platinum nanoparticles in cancer therapy: Chemotherapeutic enhancement and ROS generation. Med. Oncol. 2025, 42, 42. [Google Scholar] [CrossRef]
  30. Gao, X.; Lei, G.; Wang, B.; Deng, Z.; Karges, J.; Xiao, H.; Tan, D. Encapsulation of Platinum Prodrugs into PC7A Polymeric Nanoparticles Combined with Immune Checkpoint Inhibitors for Therapeutically Enhanced Multimodal Chemotherapy and Immunotherapy by Activation of the STING Pathway. Adv. Sci. 2023, 10, e2205241. [Google Scholar] [CrossRef]
  31. Shang, K.; Zhang, L.; Yu, Y.; Xiao, H.; Gao, Y.; Yang, L.; Huang, J.; Song, H.; Han, H. Disulfide-containing polymer delivery of C527 and a Platinum(IV) prodrug selectively inhibited protein ubiquitination and tumor growth on cisplatin resistant and patient-derived liver cancer models. Mater. Today Bio 2023, 18, 100548. [Google Scholar] [CrossRef]
  32. Morgovan, A.I.; Boia, E.R.; Motofelea, A.C.; Orasan, A.; Negru, M.C.; Guran, K.; Para, D.M.; Sandu, D.; Ciocani, S.; Sitaru, A.M.; et al. Advances in Nanotechnology-Based Cisplatin Delivery for ORL Cancers: A Comprehensive Review. Int. J. Mol. Sci. 2025, 26, 5261. [Google Scholar] [CrossRef]
  33. Lu, H.; Tong, W.; Jiang, M.; Liu, H.; Meng, C.; Wang, K.; Mu, X. Mitochondria-Targeted Multifunctional Nanoprodrugs by Inhibiting Metabolic Reprogramming for Combating Cisplatin-Resistant Lung Cancer. ACS Nano 2024, 18, 21156–21170. [Google Scholar] [CrossRef] [PubMed]
  34. Nafees, M.; Hanif, M.; Muhammad Asif Khan, R.; Faiz, F.; Yang, P. A Dual Action Platinum(IV) Complex with Self-assembly Property Inhibits Prostate Cancer through Mitochondrial Stress Pathway. ChemMedChem 2024, 19, e202400289. [Google Scholar] [CrossRef] [PubMed]
  35. Ma, Z.; Zhang, Y.; Zhang, J.; Zhang, W.; Foda, M.F.; Dai, X.; Han, H. Ultrasmall Peptide-Coated Platinum Nanoparticles for Precise NIR-II Photothermal Therapy by Mitochondrial Targeting. ACS Appl. Mater. Interfaces 2020, 12, 39434–39443. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, Q.; Lin, J.; Li, J.; Zhou, Y.; Bi, Z.; Yang, H.; Lu, W.; Lu, T.; Qian, R.; Yang, X.; et al. Mitochondrial-Targeted Multifunctional Platinum-Based Nano “Terminal-Sensitive Projectile” for Enhanced Cancer Chemotherapy Efficacy. ACS Nano 2025, 19, 8711–8726. [Google Scholar] [CrossRef]
  37. Wang, N.; Deng, Z.; Zhu, Q.; Zhao, J.; Xie, K.; Shi, P.; Wang, Z.; Chen, X.; Wang, F.; Shi, J.; et al. An erythrocyte-delivered photoactivatable oxaliplatin nanoprodrug for enhanced antitumor efficacy and immune response. Chem. Sci. 2021, 12, 14353–14362. [Google Scholar] [CrossRef]
  38. Zeng, X.; Sun, J.; Li, S.; Shi, J.; Gao, H.; Leong, W.S.; Wu, Y.; Li, M.; Liu, C.; Li, P.; et al. Blood-triggered generation of platinum nanoparticle functions as an anti-cancer agent. Nat. Commun. 2020, 11, 567. [Google Scholar] [CrossRef]
  39. Entezar-Almahdi, E.; Heidari, R.; Ghasemi, S.; Mohammadi-Samani, S.; Farjadian, F. Integrin receptor mediated pH-responsive nano-hydrogel based on histidine-modified poly(aminoethyl methacrylamide) as targeted cisplatin delivery system. J. Drug Deliv. Sci. Technol. 2021, 62, 102402. [Google Scholar] [CrossRef]
  40. Zarkesh, K.; Heidari, R.; Iranpour, P.; Azarpira, N.; Ahmadi, F.; Mohammadi-Samani, S.; Farjadian, F. Theranostic Hyaluronan Coated EDTA Modified Magnetic Mesoporous Silica Nanoparticles for Targeted Delivery of Cisplatin. J. Drug Deliv. Sci. Technol. 2022, 77, 103903. [Google Scholar] [CrossRef]
  41. Li, X.; Cai, J.; Zhang, H.; Sun, S.; Zhao, S.; Wang, Z.; Nie, X.; Xu, C.; Zhang, Y.; Xiao, H. A Trisulfide Bond Containing Biodegradable Polymer Delivering Pt(IV) Prodrugs to Deplete Glutathione and Donate H2S to Boost Chemotherapy and Antitumor Immunity. ACS Nano 2024, 18, 7852–7867. [Google Scholar] [CrossRef]
  42. Xiang, D.; Han, X.; Li, J.; Zhang, J.; Xiao, H.; Li, T.; Zhao, X.; Xiong, H.; Xu, M.; Bi, W. Combination of IDO inhibitors and platinum(IV) prodrugs reverses low immune responses to enhance cancer chemotherapy and immunotherapy for osteosarcoma. Mater. Today Bio 2023, 20, 100675. [Google Scholar] [CrossRef]
  43. Yan, J.; Wei, D.; Zhao, Z.; Sun, K.; Sun, Y. Osteosarcoma-targeting PtIV prodrug amphiphile for enhanced chemo-immunotherapy via Ca2+ trapping. Acta Biomater. 2025, 193, 474–483. [Google Scholar] [CrossRef]
  44. Huang, Y.; Wei, D.; Wang, B.; Tang, D.; Cheng, A.; Xiao, S.; Yu, Y.; Huang, W. NIR-II light evokes DNA cross-linking for chemotherapy and immunogenic cell death. Acta Biomater. 2023, 160, 198–210. [Google Scholar] [CrossRef] [PubMed]
  45. Hernández, Á.-P.; Iglesias-Anciones, L.; Vaquero-González, J.J.; Piñol, R.; Criado, J.J.; Rodriguez, E.; Juanes-Velasco, P.; García-Vaquero, M.L.; Arias-Hidalgo, C.; Orfao, A.; et al. Enhancement of Tumor Cell Immunogenicity and Antitumor Properties Derived from Platinum-Conjugated Iron Nanoparticles. Cancers 2023, 15, 3204. [Google Scholar] [CrossRef] [PubMed]
  46. Yu, J.; Liu, S.; Wang, Y.P.; He, X.D.; Zhang, Q.F.; Qi, Y.X.; Zhou, D.F.; Xie, Z.G.; Li, X.Y.; Huang, Y.B. Synergistic enhancement of immunological responses triggered by hyperthermia sensitive Pt NPs via NIR laser to inhibit cancer relapse and metastasis. Bioact. Mater. 2022, 7, 389–400. [Google Scholar] [CrossRef] [PubMed]
  47. Liu, J.; Jiang, X.; Li, Y.; Yang, K.; Weichselbaum, R.R.; Lin, W. Immunogenic Bifunctional Nanoparticle Suppresses Programmed Cell Death-Ligand 1 in Cancer and Dendritic Cells to Enhance Adaptive Immunity and Chemo-Immunotherapy. ACS Nano 2024, 18, 5152–5166. [Google Scholar] [CrossRef]
  48. Liu, X.M.; Zhu, Z.Z.; He, X.R.; Zou, Y.H.; Chen, Q.; Wang, X.Y.; Liu, H.M.; Qiao, X.; Wang, X.; Xu, J.Y. NIR Light and GSH Dual-Responsive Upconversion Nanoparticles Loaded with Multifunctional Platinum(IV) Prodrug and RGD Peptide for Precise Cancer Therapy. ACS Appl. Mater. Interfaces 2024, 16, 40753–40766. [Google Scholar] [CrossRef]
  49. Manisha; Pal, P.; Das, P.; Upadhyay, V.R. Stimuli-Responsive Drug Delivery Systems: From Concept to Clinical Translation. In Next-Generation Drug Delivery Systems. Methods in Pharmacology and Toxicology, 1st ed.; Pathak, A., Singh, S.P., Eds.; Humana: New York, NY, USA, 2025; pp. 133–157. [Google Scholar]
  50. Cheng, Q.; Liu, G.; Yin, X. Facile construction of drugs loaded lipid-coated calcium carbonate as a promising pH-Dependent drug delivery system for thyroid cancer treatment. Heliyon 2023, 9, e18413. [Google Scholar] [CrossRef]
  51. Lu, H.; He, S.; Zhang, Q.; Li, X.; Xie, Z.; Wang, Z.; Qi, Y.; Huang, Y. Dual-sensitive dual-prodrug nanoparticles with light-controlled endo/lysosomal escape for synergistic photoactivated chemotherapy. Biomater. Sci. 2021, 9, 7115–7123. [Google Scholar] [CrossRef]
  52. Kuang, G.; Ding, J.; Xie, W.; Ye, Z.; Zhang, Q. Stimuli-Responsive Nodal Dual-Drug Polymer Nanoparticles for Cancer Therapy. Int. J. Nanomed. 2025, 20, 5181–5192. [Google Scholar] [CrossRef]
  53. Li, J.; Zhang, Q.; Yang, H.; Lu, W.; Fu, Y.; Xiong, Y.; Wang, X.; Lu, T.; Xin, Y.; Xie, Z.; et al. Sequential dual-locking strategy using photoactivated Pt(IV)-based metallo-nano prodrug for enhanced chemotherapy and photodynamic efficacy by triggering ferroptosis and macrophage polarization. Acta Pharm. Sin. B 2024, 14, 3251–3265. [Google Scholar] [CrossRef] [PubMed]
  54. Mazzei, L.F.; Martínez, Á.; Trevisan, L.; Rosa-Gastaldo, D.; Cortajarena, A.L.; Mancin, F.; Salassa, L. Toward supramolecular nanozymes for the photocatalytic activation of Pt(IV) anticancer prodrugs. Chem. Commun. 2020, 56, 10461–10464. [Google Scholar] [CrossRef] [PubMed]
  55. Xiao, H.; Noble, G.T.; Stefanick, J.F.; Qi, R.; Kiziltepe, T.; Jing, X.; Bilgicer, B. Photosensitive Pt(IV)-azide prodrug-loaded nanoparticles exhibit controlled drug release and enhanced efficacy in vivo. J. Control Release 2014, 173, 11–17. [Google Scholar] [CrossRef] [PubMed]
  56. Liang, G.; Sadhukhan, T.; Banerjee, S.; Tang, D.; Zhang, H.; Cui, M.; Montesdeoca, N.; Karges, J.; Xiao, H. Reduction of Platinum(IV) Prodrug Hemoglobin Nanoparticles with Deeply Penetrating Ultrasound Radiation for Tumor-Targeted Therapeutically Enhanced Anticancer Therapy. Angew. Chem. Int. Ed. Engl. 2023, 62, e202301074. [Google Scholar] [CrossRef]
  57. Kong, C.; Li, Y.; Xu, Y.; Luo, C.; Shen, N.; Tang, Z.; Chen, X. Ultrasound irradiation-induced superoxide anion radical mediates the reduction of tetravalent platinum prodrug for anti-tumor therapy. CCS Chem. 2025, 7, 1142–1156. [Google Scholar] [CrossRef]
  58. Wang, Y.; Luo, Z.; Zhou, D.; Wang, X.; Chen, J.; Gong, S.; Yu, Z. Nano-assembly of ursolic acid with platinum prodrug overcomes multiple deactivation pathways in platinum-resistant ovarian cancer. Biomater. Sci. 2021, 9, 4110–4119. [Google Scholar] [CrossRef]
  59. Duan, Q.Y.; Zhu, Y.X.; Jia, H.R.; Guo, Y.; Zhang, X.; Gu, R.; Li, C.; Wu, F.G. Platinum-Coordinated Dual-Responsive Nanogels for Universal Drug Delivery and Combination Cancer Therapy. Small 2022, 18, 2203260. [Google Scholar] [CrossRef]
  60. Chen, W.; Liu, C.; Ji, X.; Joseph, J.; Tang, Z.; Ouyang, J.; Xiao, Y.; Kong, N.; Joshi, N.; Farokhzad, O.C.; et al. Stanene-Based Nanosheets for β-Elemene Delivery and Ultrasound-Mediated Combination Cancer Therapy. Angew. Chem. Int. Ed. Engl. 2021, 60, 7155–7164. [Google Scholar] [CrossRef]
  61. Xu, Z.; Li, Q.; Zhang, C.; Wang, P.; Xu, X.; Ran, L.; Zhang, L.; Tian, G.; Zhang, G. Amorphous ferric oxide-coating selenium core-shell nanoparticles: A self-preservation Pt(IV) platform for multi-modal cancer therapies through hydrogen peroxide depletion-mediated anti-angiogenesis, apoptosis and ferroptosis. Nanoscale 2022, 14, 11600–11611. [Google Scholar] [CrossRef]
  62. Gao, Z.; He, T.; Zhang, P.; Li, X.; Zhang, Y.; Lin, J.; Hao, J.; Huang, P.; Cui, J. Polypeptide-Based Theranostics with Tumor-Microenvironment-Activatable Cascade Reaction for Chemo-ferroptosis Combination Therapy. ACS Appl. Mater. Interfaces 2020, 12, 20271–20280. [Google Scholar] [CrossRef]
  63. Ren, Z.; Sun, S.; Sun, R.; Cui, G.; Hong, L.; Rao, B.; Li, A.; Yu, Z.; Kan, Q.; Mao, Z. A Metal-Polyphenol-Coordinated Nanomedicine for Synergistic Cascade Cancer Chemotherapy and Chemodynamic Therapy. Adv. Mater. 2020, 32, e1906024. [Google Scholar] [CrossRef] [PubMed]
  64. Mao, X.; Calero-Pérez, P.; Montpeyó, D.; Bruna, J.; Yuste, V.J.; Candiota, A.P.; Lorenzo, J.; Novio, F.; Ruiz-Molina, D. Intranasal Administration of Catechol-Based Pt(IV) Coordination Polymer Nanoparticles for Glioblastoma Therapy. Nanomaterials 2022, 12, 1221. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, Y.; Zhang, X.M.; Sun, Y.; Chen, H.L.; Zhou, L.Y. Cetuximab-decorated and NIR-activated Nanoparticles Based on Platinum(IV)-prodrug: Preparation, Characterization and In-vitro Anticancer Activity in Epidermoid Carcinoma Cells. IJPR 2021, 20, 371–383. [Google Scholar] [PubMed]
  66. Marotta, C.; Cirri, D.; Kanavos, I.; Ronga, L.; Lobinski, R.; Funaioli, T.; Giacomelli, C.; Barresi, E.; Trincavelli, M.L.; Marzo, T.; et al. Oxaliplatin(IV) Prodrugs Functionalized with Gemcitabine and Capecitabine Induce Blockage of Colorectal Cancer Cell Growth-An Investigation of the Activation Mechanism and Their Nanoformulation. Pharmaceutics 2024, 16, 278. [Google Scholar] [CrossRef]
  67. Zhang, W.; Du, X.F.; Liu, B.; Li, C.; Long, J.; Zhao, M.X.; Yao, Z.; Liang, X.J.; Lai, Y. Engineering Supramolecular Nanomedicine for Targeted Near Infrared-triggered Mitochondrial Dysfunction to Potentiate Cisplatin for Efficient Chemophototherapy. ACS Nano 2022, 16, 1421–1435. [Google Scholar] [CrossRef]
  68. Li, S.; Wu, Y.; Xue, X.; Liu, S. NIR and Reduction Dual-Sensitive Polymeric Prodrug Nanoparticles for Bioimaging and Combined Chemo-Phototherapy. Polymers 2022, 14, 287. [Google Scholar] [CrossRef]
  69. Zhang, J.; Zhao, B.; Chen, S.; Wang, Y.; Zhang, Y.; Wang, Y.; Wei, D.; Zhang, L.; Rong, G.; Weng, Y.; et al. Near-Infrared Light Irradiation Induced Mild Hyperthermia Enhances Glutathione Depletion and DNA Interstrand Cross-Link Formation for Efficient Chemotherapy. ACS Nano 2020, 14, 14831–14845. [Google Scholar] [CrossRef]
  70. Kulkarni, B.; Malhotra, M.; Jayakannan, M. Fluorescent ABC-Triblock Polymer Nanocarrier for Cisplatin Delivery to Cancer Cells. Chem. Asian J. 2022, 17, e202101337. [Google Scholar] [CrossRef]
  71. Yang, J.; Yang, Z.; Wang, H.; Chang, Y.; Xu, J.F.; Zhang, X. A Polymeric Nanoparticle to Co-Deliver Mitochondria-Targeting Peptides and Pt(IV) Prodrug: Toward High Loading Efficiency and Combination Efficacy. Angew. Chem. Int. Ed. Engl. 2024, 63, e202402291. [Google Scholar] [CrossRef]
  72. Wei, D.; Yu, Y.; Zhang, X.; Wang, Y.; Chen, H.; Zhao, Y.; Wang, F.; Rong, G.; Wang, W.; Kang, X.; et al. Breaking the Intracellular Redox Balance with Diselenium Nanoparticles for Maximizing Chemotherapy Efficacy on Patient-Derived Xenograft Models. ACS Nano 2020, 14, 16984–16996. [Google Scholar] [CrossRef]
  73. Chen, T.; Xu, S.; Huang, W.; Yan, D. Light-responsive nanodrugs co-self-assembled from a PEG-Pt(IV) prodrug and doxorubicin for reversing multidrug resistance in the chemotherapy process of hypoxic solid tumors. Biomater. Sci. 2022, 10, 3901–3910. [Google Scholar] [CrossRef] [PubMed]
  74. Yu, Y.; Wei, D.; Bing, T.; Wang, Y.; Liu, C.; Xiao, H. A Polyplatin with Hands-Holding Near-Infrared-II Fluorophores and Prodrugs at a Precise Ratio for Tracking Drug Fate with Realtime Readout and Treatment Feedback. Adv. Mater. 2024, 36, e2402452. [Google Scholar] [CrossRef] [PubMed]
  75. Zhang, H.; Wu, Y.; Xu, X.; Chen, C.; Xue, X.; Xu, B.; Li, T.; Chen, Z. Synthesis Characterization of Platinum (IV) Complex Curcumin Backboned Polyprodrugs: In Vitro Drug Release Anticancer Activity. Polymers 2020, 13, 67. [Google Scholar] [CrossRef] [PubMed]
  76. Garcia-Peiro, J.I.; Ortega-Liebana, M.C.; Adam, C.; Lorente-Macías, Á.; Travnickova, J.; Patton, E.E.; Guerrero-López, P.; Garcia-Aznar, J.M.; Hueso, J.L.; Santamaria, J.; et al. Dendritic Platinum Nanoparticles Shielded by Pt-S PEGylation as Intracellular Reactors for Bioorthogonal Uncaging Chemistry. Angew. Chem. Int. Ed. Engl. 2025, 64, e202424037. [Google Scholar] [CrossRef]
  77. Tang, L.; Yin, Y.; Cao, Y.; Liu, H.; Qing, G.; Fu, C.; Li, Z.; Zhu, Y.; Shu, W.; He, S.; et al. Bioorthogonal Chemistry-Guided Inhalable Nanoprodrug to Circumvent Cisplatin Resistance in Orthotopic Nonsmall Cell Lung Cancer. ACS Nano 2024, 18, 32103–32117. [Google Scholar] [CrossRef]
  78. Wu, J.; Hu, Y.; Ye, H.; Zhang, S.; Zhu, J.; Ji, D.; Zhang, Y.; Ding, Y.; Huang, Z. One Stone Two Birds: Redox-Sensitive Colocalized Delivery of Cisplatin and Nitric Oxide Through Cascade Reactions. JACS Au 2022, 2, 2339–2351. [Google Scholar] [CrossRef]
  79. Alfonso-Triguero, P.; Lorenzo, J.; Candiota, A.P.; Arús, C.; Ruiz-Molina, D.; Novio, F. Platinum-Based Nanoformulations for Glioblastoma Treatment: The Resurgence of Platinum Drugs? Nanomaterials 2023, 13, 1619. [Google Scholar] [CrossRef]
  80. Spector, D.; Erofeev, A.; Gorelkin, P.; Skvortsov, D.; Trigub, A.; Markova, A.; Nikitina, V.; Ul’yanovskiy, N.; Shtil’, A.; Semkina, A.; et al. Biotinylated Pt(IV) prodrugs with elevated lipophilicity and cytotoxicity. Dalton Trans. 2023, 52, 866–871. [Google Scholar] [CrossRef]
  81. Ravera, M.; Gabano, E.; Perin, E.; Rangone, B.; Bonzani, D.; Osella, D. Can the Self-Assembling of Dicarboxylate Pt(IV) Prodrugs Influence Their Cell Uptake? Bioinorg. Chem. Appl. 2021, 2021, 9489926. [Google Scholar] [CrossRef]
  82. Abu Ammar, A.; Raveendran, R.; Gibson, D.; Nassar, T.; Benita, S. A Lipophilic Pt(IV) Oxaliplatin Derivative Enhances Antitumor Activity. J. Med. Chem. 2016, 59, 9035–9046. [Google Scholar] [CrossRef]
  83. Wang, J.; Shen, S.; Li, J.; Cao, Z.; Yang, X. Precise Depletion of Tumor Seed and Growing Soil with Shrinkable Nanocarrier for Potentiated Cancer Chemoimmunotherapy. ACS Nano 2021, 15, 4636–4646. [Google Scholar] [CrossRef] [PubMed]
  84. Gavas, S.; Quazi, S.; Karpiński, T.M. Nanoparticles for Cancer Therapy: Current Progress and Challenges. Nanoscale Res. Lett. 2021, 16, 173. [Google Scholar] [CrossRef] [PubMed]
  85. Lan, X.; Zhu, W.; Huang, X.; Yu, Y.; Xiao, H.; Jin, L.; Pu, J.J.; Xie, X.; She, J.; Lui, V.W.Y.; et al. Microneedles Loaded with Anti-PD-1–Cisplatin Nanoparticles for Synergistic Cancer Immuno-Chemotherapy. Nanoscale 2020, 12, 18885–18898. [Google Scholar] [CrossRef] [PubMed]
  86. Hafeez, M.N.; Celia, C.; Petrikaite, V. Challenges towards Targeted Drug Delivery in Cancer Nanomedicines. Processes 2021, 9, 1527. [Google Scholar] [CrossRef]
  87. Wei, D.; Fan, J.; Yan, J.; Liu, C.; Cao, J.; Xu, C.; Sun, Y.; Xiao, H. Nuclear-Targeting Lipid PtIV Prodrug Amphiphile Cooperates with siRNA for Enhanced Cancer Immunochemotherapy by Amplifying Pt-DNA Adducts and Reducing Phosphatidylserine Exposure. J. Am. Chem. Soc. 2024, 146, 1185–1195. [Google Scholar] [CrossRef]
  88. Li, C.; Li, T.; Huang, L.; Yang, M.; Zhu, G. Self-assembled Lipid Nanoparticles for Ratiometric Codelivery of Cisplatin and siRNA Targeting XPF to Combat Drug Resistance in Lung Cancer. Chem. Asian J. 2019, 14, 1570–1576. [Google Scholar] [CrossRef]
  89. Xu, X.; Xie, K.; Zhang, X.Q.; Pridgen, E.M.; Park, G.Y.; Cui, D.S.; Shi, J.; Wu, J.; Kantoff, P.W.; Lippard, S.J.; et al. Enhancing tumor cell response to chemotherapy through nanoparticle-mediated codelivery of siRNA and cisplatin prodrug. Proc. Natl. Acad. Sci. USA 2013, 110, 18638–18643. [Google Scholar] [CrossRef]
  90. Paul, M.; Ghosh, B.; Biswas, S. Human Serum Albumin-Oxaliplatin (Pt(IV)) prodrug nanoparticles with dual reduction sensitivity as effective nanomedicine for triple-negative breast cancer. Int. J. Biol. Macromol. 2024, 256, 128281. [Google Scholar] [CrossRef]
  91. Li, X.; Zhang, L.; Li, T.; Li, S.; Wu, W.; Zhao, L.; Xie, P.; Yang, J.; Li, P.; Zhang, Y.; et al. Abplatin(IV) inhibited tumor growth on a patient derived cancer model of hepatocellular carcinoma and its comparative multi-omics study with cisplatin. J. Nanobiotechnology 2022, 20, 258. [Google Scholar] [CrossRef]
  92. Xu, S.; Luo, W.; Zhu, M.; Zhao, L.; Gao, L.; Liang, H.; Zhang, Z.; Yang, F. Human Serum Albumin-Platinum(II) Agent Nanoparticles Inhibit Tumor Growth Through Multimodal Action Against the Tumor Microenvironment. Mol. Pharm. 2024, 21, 346–357. [Google Scholar] [CrossRef]
Figure 1. The structure of cisplatin.
Figure 1. The structure of cisplatin.
Pharmaceutics 17 01267 g001
Figure 2. Mechanisms of GSH-regulated cisplatin sensitivity of tumor cells. The increased GSH level inside the cancer cell can, on one hand, enhance the MRP2-mediated efflux of cisplatin and regulate the intracellular redox equilibrium, leading to cisplatin resistance, and, on the other hand, lower the level of free Cu within the cell, upregulate the expression of hCtr1, increase the hCtr1-mediated cisplatin influx and lead to sensitization to cisplatin. (Created in BioRender. Iova, V. (2025) https://BioRender.com/wys8gdv, accessed on 23 July 2025).
Figure 2. Mechanisms of GSH-regulated cisplatin sensitivity of tumor cells. The increased GSH level inside the cancer cell can, on one hand, enhance the MRP2-mediated efflux of cisplatin and regulate the intracellular redox equilibrium, leading to cisplatin resistance, and, on the other hand, lower the level of free Cu within the cell, upregulate the expression of hCtr1, increase the hCtr1-mediated cisplatin influx and lead to sensitization to cisplatin. (Created in BioRender. Iova, V. (2025) https://BioRender.com/wys8gdv, accessed on 23 July 2025).
Pharmaceutics 17 01267 g002
Figure 3. The general effects on mitochondria of Pt-encapsulated NPs. After entering the cancer cell, the NPs target and damage the mitochondria, leading to a blockage of energy supply, causing cell death (Created with BioRender https://biorender.com/0hj2h8q, accessed on 1 august 2025).
Figure 3. The general effects on mitochondria of Pt-encapsulated NPs. After entering the cancer cell, the NPs target and damage the mitochondria, leading to a blockage of energy supply, causing cell death (Created with BioRender https://biorender.com/0hj2h8q, accessed on 1 august 2025).
Pharmaceutics 17 01267 g003
Figure 4. The effects of PEG-OXA NPs. These NPs have a high stability in the blood, with an enhanced circulation time, lower systemic toxicity and increased anti-tumor efficacy. (Created with BioRender https://BioRender.com/s4pnyfr, accessed on 1 august 2025).
Figure 4. The effects of PEG-OXA NPs. These NPs have a high stability in the blood, with an enhanced circulation time, lower systemic toxicity and increased anti-tumor efficacy. (Created with BioRender https://BioRender.com/s4pnyfr, accessed on 1 august 2025).
Pharmaceutics 17 01267 g004
Figure 5. Mechanisms of NP-induced increased anti-tumoral immunity. These NPs activate immune cells, increase cytokine secretion, increase the M1/M2 macrophage ratio and can enhance ICD. (Created with BioRender https://BioRender.com/4ffd2r8, accessed on 1 august 2025).
Figure 5. Mechanisms of NP-induced increased anti-tumoral immunity. These NPs activate immune cells, increase cytokine secretion, increase the M1/M2 macrophage ratio and can enhance ICD. (Created with BioRender https://BioRender.com/4ffd2r8, accessed on 1 august 2025).
Pharmaceutics 17 01267 g005
Figure 6. The principles of multistimuli-responsive drug delivery systems of Pt drugs. Because of the special characteristics of the tumor microenvironment (low pH and an increased level of reductive species) and of the NPs (photosensitivity), precise spatiotemporal delivery of anticancer molecules can occur. (Created with BioRender https://BioRender.com/62axkhb, accessed on 1 august 2025).
Figure 6. The principles of multistimuli-responsive drug delivery systems of Pt drugs. Because of the special characteristics of the tumor microenvironment (low pH and an increased level of reductive species) and of the NPs (photosensitivity), precise spatiotemporal delivery of anticancer molecules can occur. (Created with BioRender https://BioRender.com/62axkhb, accessed on 1 august 2025).
Pharmaceutics 17 01267 g006
Figure 7. The association of Pt drugs/prodrugs with other compounds or techniques in NP-based combination therapy. These Pt compounds can be associated with other elements (Fe compounds or selenium) monoclonal antibodies (cetuximab), other anti-tumor therapies (chemotherapeutic agents—capecitabine, gemcitabine-phototherapy and radiotherapy) mitochondria targeting agents, USP1i and antioxidants (curcumin). These combinations result in an improved anticancer effect, lower systemic toxicity and the possibility to reverse the resistance of tumors to drugs. (Created with BioRender https://BioRender.com/xyg5ul1, accessed on 1 august 2025).
Figure 7. The association of Pt drugs/prodrugs with other compounds or techniques in NP-based combination therapy. These Pt compounds can be associated with other elements (Fe compounds or selenium) monoclonal antibodies (cetuximab), other anti-tumor therapies (chemotherapeutic agents—capecitabine, gemcitabine-phototherapy and radiotherapy) mitochondria targeting agents, USP1i and antioxidants (curcumin). These combinations result in an improved anticancer effect, lower systemic toxicity and the possibility to reverse the resistance of tumors to drugs. (Created with BioRender https://BioRender.com/xyg5ul1, accessed on 1 august 2025).
Pharmaceutics 17 01267 g007
Figure 8. Bioortoghonal reactions performed by Pt compounds in cancer therapy. PtNPs can act within the complex intracellular microenvironment of cancer cells, with in situ activation of anticancer prodrugs into active molecules. Additionally, micelles delivering ethacraplatin and NO can reduce the intracellular GSH and block GSH S-transferase, thus enhancing the sensitivity towards cisplatin. (Created with BioRender https://BioRender.com/52dh5hc, accessed on 1 august 2025).
Figure 8. Bioortoghonal reactions performed by Pt compounds in cancer therapy. PtNPs can act within the complex intracellular microenvironment of cancer cells, with in situ activation of anticancer prodrugs into active molecules. Additionally, micelles delivering ethacraplatin and NO can reduce the intracellular GSH and block GSH S-transferase, thus enhancing the sensitivity towards cisplatin. (Created with BioRender https://BioRender.com/52dh5hc, accessed on 1 august 2025).
Pharmaceutics 17 01267 g008
Figure 9. The advantages of NPs with increased lipophilicity in delivering Pt drugs/prodrugs. The increased lipophilicity can help in the penetration of the blood–brain barrier, and the obtained NPs can reduce the systemic toxicity associated with chemotherapy, optimize the chemo-/radiotherapy and increase cancer cell uptake by both passive diffusion and endocytosis. (Created with BioRender https://BioRender.com/efub7kc, accessed on 1 august 2025).
Figure 9. The advantages of NPs with increased lipophilicity in delivering Pt drugs/prodrugs. The increased lipophilicity can help in the penetration of the blood–brain barrier, and the obtained NPs can reduce the systemic toxicity associated with chemotherapy, optimize the chemo-/radiotherapy and increase cancer cell uptake by both passive diffusion and endocytosis. (Created with BioRender https://BioRender.com/efub7kc, accessed on 1 august 2025).
Pharmaceutics 17 01267 g009
Figure 10. The effects of Pt drugs-siRNA NP-based codelivery systems. The use of siRNAs can reverse cisplatin resistance and reduce treatment-associated immunosuppression. (Created with BioRender https://BioRender.com/x1edlli, accessed on 1 August 2025).
Figure 10. The effects of Pt drugs-siRNA NP-based codelivery systems. The use of siRNAs can reverse cisplatin resistance and reduce treatment-associated immunosuppression. (Created with BioRender https://BioRender.com/x1edlli, accessed on 1 August 2025).
Pharmaceutics 17 01267 g010
Figure 11. The advantages of Pt complexes delivered by human serum albumin NPs. These NPs can help in localized drug delivery, reduce drug demand, lower the intensity of side-effects, reverse resistance to cisplatin and improve the overall anti-tumor effect. (Created with BioRender https://BioRender.com/yolk07y, accessed on 1 august 2025).
Figure 11. The advantages of Pt complexes delivered by human serum albumin NPs. These NPs can help in localized drug delivery, reduce drug demand, lower the intensity of side-effects, reverse resistance to cisplatin and improve the overall anti-tumor effect. (Created with BioRender https://BioRender.com/yolk07y, accessed on 1 august 2025).
Pharmaceutics 17 01267 g011
Table 1. The side-effects induced by cisplatin therapy.
Table 1. The side-effects induced by cisplatin therapy.
Cisplatin-Induced Side-EffectsComplications and SymptomsMechanisms
Renal Toxicityacute kidney injurypyroptosis
hypomagnesemiaoxidative damage
Fanconi-like syndromeinflammation [12]
distal renal tubular acidosis
hypocalcemia
wasting renal salt
hyperuricemia [13]
Allergic Reactionsskin rashtype I allergic reactions [14]
flushing
abdominal cramping
itchy palms
back pain [14]
Gastrointestinal Disorderscolitisdirect epithelial damage
diarrhea [15]inflammation
disruption of the normal integrity of the gastrointestinal mucosa
microbiome alteration [16]
Hemorrhagereduced chemotherapy dosagesapoptosis through the ERK signaling pathway [17,18]
postponed treatment
bleeding
unfavorable oncological outcomes [17,18]
Hearing Losstinnitusdirect damage to mitochondrial and nuclear DNA
high-frequency hearing lossapoptosis caused by cell cycle arrest and activation of p53
decreased ability to hear normal conversation [19]generation of ROS by the activation of NADPH oxidase 3 and xanthine oxidase [19]
Table 2. Nanoparticle properties and functional mechanisms for platinum drug delivery.
Table 2. Nanoparticle properties and functional mechanisms for platinum drug delivery.
Nanoparticle properties and functional mechanisms for platinum drug deliverymithocondria targeting
increased stability in the circulation
increased anti-tumoral immunity
multistimuli-responsive drug delivery systems
siRNA technology
nanozyme-based photocatalytic conversion of Pt(IV) to Pt(II)
combination chemotherapy
deeply penetrating ultrasound radiation activation
bioorthogonal reactions catalyzed by PtNPs
increased lipophilicity
human serum albumin-based NPs
targeting both malignant and non-malignant cells
Table 3. Latest Pt nanoparticle-mediated drug delivery systems and formulation targeting mitochondria.
Table 3. Latest Pt nanoparticle-mediated drug delivery systems and formulation targeting mitochondria.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
LND-S-S-Pt-TPP/HA-CDSelf-assembled nanotargeted drug delivery systemHA-CD
TPP
LND
cisplatin prodrug (Pt(IV))
HA-CD targets membrane CD44Synergistic destruction of cis-platin-resistant lung cancer cells by disruption of mitochondria[33]
TPP targets mitochondria
LND damages mitochondria and inhibits glycolysis
GSH-mediated reduction in Pt(IV) with mtDNA damage
PL-IIISelf-assembled, spherical shaped NPsOxoplatin
Lithocholic acid
Heptanoate
Halting cell cycle in the S and G2 phases10-fold higher cytotoxicity compared to cisplatin in PC3 cells[34]
Damaging DNA
Disruption of mitochondrial membrane potential
Increased ROS productionMechanisms of action requiring reduction in the Pt(IV) core to Pt(II) and simultaneous release of lithocholic acid, with unique cytotoxicity
Alteration of mitochondrial bioenergetics
Upregulation of pro-apoptotic proteins and reduction in anti-apoptotic ones from BCl-2 family
TPP-PtUltrasmall peptide-coated platinum nanoparticlesTPP peptides
Ultrasmall PtNPs
MonodispersityDeliverance of TPP-Pt to the thermally susceptible tumor mitochondria, with minimal side-effects[35]
High stability
Biosafety
Enhanced uptake of cancer cells
Priority of mitochondria
NTSBPt-based nanoprojectileDSPE-PEG2K-IR780
TSB (FFa-Pt(IV) prodrug-TPP)
DSPE-PEG2K-IR780 enhances cellular internalization and equips the carrier with PDT and PTT capabilitiesEnhanced sensitivity of tumor cells to Pt-based chemotherapeutic drugs[36]
TSB targets cellular mitochondria releasing (OXA) (which attacks the mtDNA) and FFa (which disrupts the electron transport chain)
Table 4. Latest Pt nanoparticle-mediated drug delivery systems and formulation increasing blood stability.
Table 4. Latest Pt nanoparticle-mediated drug delivery systems and formulation increasing blood stability.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
PEG-OXA NPsMicellar NPsOXA
PEG
long lipid chains
Optimized absorption, delivery and eliminationFacilitation of rapid bioactive OXA release in tumor cells
Manifestation
of high stability in blood in vitro
Increased half-life in vivo
[9]
Increased antitumor efficacy
Decreased side-effects
Enhanced inhibition of growing tumors
Stability in whole blood or plasma due to the low critical micelle concentration
ERY1-PEG3k/PPA-Pt(IV)-NCsNanocrystalsA small-molecule Pt(IV) prodrug [Pt(DACH)(PPA)(OCOCH2CH2COOH)(ox)]
dielectric nanocrystals
PEG3k
ERY1
Enhanced stability due to the ERY1-mediated binding to erythrocytes in the bloodstreamOvercoming of shortcomings (i.e., reduced circulation, failure to accumulate in the tumor, and dose-limiting toxicity) of traditional Pt-based chemotherapy through an erythrocyte-delivered and NIR photoactivatable Pt(IV) nanoprodrug[37]
Prolonged circulation in the blood
Minimized side-effects
Increased Pt accumulation in tumor through erythrocyte delivery approach
Enhanced immunopotentiation
Oxaliplatin release in a controllable manner upon irradiation
PtNPs with protein coronasSelf-assembled PtNPs with protein coronasCisplatin
HAS and other blood proteins
Rapidly generated in vivo in human blood upon treatment with cisplatinFunctioning as a biocompatible drug delivery platform for chemotherapy-resistant tumor treatment[38]
Accumulation in tumors
Persistence in the body for an extended period of time by NPs’ interaction with HSA coating them
Consumption of intracellular GSH
Activation of apoptosis
Capacity to reverse tumor resistance to daunorubicin
Cisplatin-Tetrac-His-P(AEMA-co-PEGDMA)Nano-hydrogelAminoethyl methacrylamide
PEG dimethacrylate
L-histidine
Cisplatin
tetraiodothyroacetic acid
Suitable nano sizeSynthesis of a novel nano-hydrogel (NH) and depicted its application in the process of targeted delivery of cisplatin[39]
Relatively good hemo-compatibility
pH-responsive drug release pattern
Active targeting via integrin receptors
Improved pharmacokinetic parameters
Cisplatin-EDTA-MMSN @ HAMSNMagnetic mesoporous silica nanoparticles (MMSNs)
EDTA
Hyaluronic acid
cisplatin
pH-responsive behaviorSynthesis of a novel multifunctional pH-responsive, biocompatible and biodegradable nanoplatform for efficient drug delivery and magnetic resonance imaging[40]
Improved internalization in cancer cells overexpressing CD44 receptor compared to normal cells
Improved pharmacokinetic parameters
High adsorption capacity of cisplatin
Reduced side-effects
Theranostic nature
Table 5. Latest Pt nanoparticle-mediated drug delivery systems and formulations increasing anti-tumoral immunity.
Table 5. Latest Pt nanoparticle-mediated drug delivery systems and formulations increasing anti-tumoral immunity.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
NP(3S)sReduction-responsive NPs formed though self-assemblyPolymers containing trisulfide bonds, using bis(2-hydroxyethyl) trisulfide, 1,2,4,5-cyclohexanetetracarboxylic dianhydride and mPEG-5k
CisPt(IV) prodrug
DNA damageNP(3S)s holding great promise for clinical translation due to low toxicity profile and potent anticancer activity, by enhancing antitumor immunity and OS pathways[41]
Activation of STING pathway
Activation of T cells
Activation of OS mechanisms
Reduction to and release of Pt(II) and H2S by GSH
NP-Pt-IDOiSelf-assembled polymeric NPsPHPM
Pt(IV)-C12
NLG919
DNA damageSuperior anticancer activity in vitro and in vivo in mouse models of osteosarcoma
Efficient combination of chemotherapy and immunotherapy
[42]
Induction of STING pathway
Increased anti-tumor immunity
Activation of cytotoxic CD8+ T cells by tryptophan metabolism inhibition
ALN-OXA NPsSelf-assembled lipid NPsLipo-OXA-ALNTargeting osteosarcoma cellsT enhancing the therapeutic effects against osteosarcoma by osteosarcoma targeting capacity, increasing chemotherapy sensitivity and improving the immune microenvironment.[43]
Enhanced 189 intracellular uptake of OXA
Inhibition of cancer cell activity
Increased antitumoral immunity
Increased M1/M2 macro-phage ratio
Modifying the tumoral microenvironment
NP3Self-assembled NPsP1 polymer obtained from DSB, 1,2,4,5-Cyclohexanetetracarboxylic Dianhydride and mPEG5k-OH
Pt(IV)-C16
IR1061
Increasing ICD in tumors after irradiation with NIR-IIIncreased therapy results for triple-negative breast cancer 4T1 compared to either OXA or NIR-II-based photothermal therapies alone[44]
Increased Pt-DNA binding
Higher DNA damage and apoptosis
IONP@BCP@[PtCl(GUDCA)en] NPsPt compound conjugated with iron oxide NPsIONPs
BCP
[PtCl(GUDCA)en]
Enhanced Pt-associated cytotoxicity Cisplatin-derived agents together with high value of IONPs as drug delivery systems and immunogenic cell death[45]
Activation of endoplasmic reticulum stress pathways with activation of ICD
Biocompatibility in biological systems
I-Pt NPsHyperthermia sensitive Pt NPsBMS-1
Mal-modified PEG
PtNPs
Improved biocompatibilitySynergistic augmentation of immunological responses and photothermal ablation of tumors
Prevention of cancer relapses and metastasis
[46]
NIR laser irradiation-mediated photothermal conversion and PTT mediated tumor ablation, with BMS-1 release and Mal exposure
Capture of tumor-associated antigens by exposed Mal and their presentation to APCs
BMS-1-mediated inhibition of immunosuppression and stimulation of immune response
OxPt/BPNanoscale coordination polymerOXA
2-bromopalmitic acid
Inhibition of palmitoyl acyltransferase DHHC3
Downregulation of PD-L1 expression in both cancer cells and dendritic cells
Potential of NCPs to simultaneously reprogram cancer cells and DCs for potent cancer treatment[47]
Enhanced DC maturation
Increased intracellular OS
Enhanced cancer ICD
Stimulation of infiltration and activation of cytotoxic T lymphocytes
Reduction in the population of immunosuppressive regulatory T cells
Table 6. Latest Pt nanoparticle multistimuli-responsive drug delivery systems.
Table 6. Latest Pt nanoparticle multistimuli-responsive drug delivery systems.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
BT-Pt (IV)@PEG/CaCO3LiposomesCaCO3 NPs
BT
OXA-DSPE
DSPE-PEG
Cholesterol
DPPC
High StabilityBiocompatible and reliable substrate for establishing pH-mediated drug delivery methods
Promising for possible therapeutic application
[50]
Rapid pH-mediated degradation
Increased OS in cancer cells
Mitochondrial and DNA damage inside cancer cells
UCNP/Pt(IV)-RGDUpconversion NPsNaYF4:Yb,Tm@NaYF4 UCNPs
DSPE-PEG2000
PHB-Pt(IV)-18Cprodrug
c(RGDyK)
High biocompatibilityMultifunctionality
Precision through enhanced therapeutic efficacy, targeted delivery, multimodal imaging
Personalized treatment
[48]
Tumor specificity
Profound cytotoxicity upon UCL irradiation and GSH reduction
Real-time UCL imaging capacity
DDNPsDual-Sensitive Dual-Prodrug NanoparticlesPt(IV) prodrug
DMC
nanoplatform
Photosensitivity, with generation of active Pt(II) from inert Pt(IV) under UVA lightPowerful synergistic anticancer effect in vitro and in vivo, with great potential as a safe and multifunctional drug delivery system for precise nanomedicine in clinical treatments[51]
Acid-sensitivity, with release of DMC and blockage of the DNA repair pathway
Endo/lysosomal escape for better photoactivated chemotherapy
DDPoly NPsSelf-assembled dual-drug polymer micellar NPsDMC
MPEG
Acid- and reduction-sensitivityEnhanced anticancer efficacy against cancer cells compared to SDPoly NPs, highlighting its potential for nanomedicine development[52]
Tumor-specific activation
Blocking DNA repair, with enhancing Pt(II)-induced apoptosis
NPS-G-FeMetallo-nano prodrugPolyaspartamide-PEG
Chlorin e6-Pt(IV)
Fe3+
gallic acid
PEG-CS
Activation by both acidic tumor microenvironment and light, resulting in the activation of both chemotherapy and PDTA versatile platform for the codelivery of therapeutic agents, exhibiting significant potential for synergistic tumor therapy while minimizing adverse side-effects[53]
Metallo-triggered ferroptosis
Polarization of TAM
Pt(IV)-UA NPsSelf-assembly of dual prodrug amphiphile into NPsPt(IV)-UA-PEG dual prodrug amphiphileHigh circulation timeDevelopment of a stimuli-responsive dual prodrug amphiphile nano-assembly providing a new strategy to overcome drug resistance[58]
High tumor accumulation
High antitumor activity
Lack of side-effects associated with conventional therapy
Reversing cisplatin resistance
Drug release in intracellular reductive and acidic environments
Pt-Coordinated Dual-Responsive NanogelsNanogelHA-βCD
PEI
cisplatin
Hyaluronidase and GSH responsiveness, releasing the loaded drugsThis dual-responsive nanogel-based platform can serve as a multifunctional platform capable of specific delivery of desired drugs to treat cancer or other diseases[59]
Small-molecule drug and protein loading and intracellular delivery capacity
Capacity to co-deliver different cargoes to realize combination cancer therapy
FMN@TACN AuNPsSupramolecular nanozymeAuNPs
C11-thiol bearing a 1,4,7-triazacyclononane headgroup
FMN
Photocatalyzation of the reductive activation of the prodrug cis,cis,trans-[Pt(NH3)2(Cl2)(O2CCH2CH2COOH)2] to cisplatin in the presence of an electron donor through an excited-state electron transfer processTACN AuNPs are suitable components to develop supramolecular nanomaterials capable to carry out flavin-mediated catalytic reactions using Pt(IV) prodrugs as substrates[54]
Potential strategy to control spatio-temporally the effects of Pt anticancer drugs via light activation and catalytic amplification
NC1–NC4Micellar NPsPhotosensitive Pt(IV)–azide prodrug complexes
based on cisplatin and oxaliplatin (C1-C4)
Rapid release of biologically active Pt(II) and enhanced cytotoxicity upon UVA irradiationPt(IV) complexes, specifically when formulated into micellar nanoparticles have potential to offer a robust platform for controlled delivery and selective activation of Pt-based anticancer therapeutics[55]
Great stability in the dark
Increased uptake by cancer cells
Enhanced circulation time in the bloodstream
Decreased systemic toxicity
Increased tumor growth inhibition
NPsSelf-assembled NPsPt1 (Pt(IV) prodrug)
Hemoglobin
DSPE-PEG2K
Biocompatibility and sonosensitivity via hemoglobinThe first example of Pt(IV) prodrug NP activation upon exposure to ultrasound radiation for deep tissue penetrating anticancer therapy[56]
Activation upon deeply penetrating ultrasound radiation with tumor eradication via apoptosis
Treatment of deep-seated or large tumors
Selective accumulation inside the tumor
Stability under physiological conditions
P-Rf/cisPt(IV) NPsSelf-assembled NPsmPEG-b-PLG
Ac-cisPt(IV)-OH
Rf-(OH)2
mPEG-NH2
Enhanced activation upon ultrasound radiation via riboflavin, mediated by the superoxide ionsEnhancing activation efficacy of Pt(IV) prodrugs under low-intensity ultrasound conditions
Innovative ultrasonic chemical reaction mechanism
Novel insights into ultrasound-mediated activation of Pt(IV) prodrugs
[57]
Significant antitumor effects even under low-intensity ultrasound radiation
Table 7. Latest achievements in the production of NPs for combination chemotherapy.
Table 7. Latest achievements in the production of NPs for combination chemotherapy.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
PFS-NPSelf-assembled metal–phenolic network NPsA disulfide bond-based amphiphilic polyphenol
A dopamine-modified cisplatin prodrug
Fe3+ ions
GSH-responsivenessInnovative cisplatin prodrug NP approach offering a promising reference for minimizing side-effects and optimizing the therapeutic effects of cisplatin-based drugs, for synergistic chemo-immunotherapy[4]
Consumption of intracellular GSH with disruption of the redox homeostasis
Amplification of the OS
Generation of ICD, with subsequent activation of the anti-tumor immune system
iAIO@NSe-PtAmorphous ferric oxide-coating selenium core–shell NPsFerric oxide shell
AIO on the surface
Pt(IV) prodrug
Se core
Avoidance of the inactivation of the Pt(IV) prodrug in the blood and increasing its accumulation in the tumorExcellent tumor targeting, biocompatibility and anti-tumor efficiency in vitro and in vivo, and proving to be a novel example of a self-preservation Pt(IV) nanoplatform for H2O2 depletion-mediated tumor anti-angiogenesis, apoptosis, and ferroptosis[61]
Leading to cellular H2O2 deficiency and cancer cell ferroptosis
GSH consumption, with increased OS
Effective apoptotic cell death
Inactivation of SOD1, with increased OS
By decreasing H2O2, causing reduction in VEGF-A expression, blocking tumor angiogenesis, disruption of mitochondrial respiration and cancer angiogenesis
Pt&Fe3O4@PPpolypeptide NPsc,c,t-[Pt(NH3)2Cl2(O2CCH2CH2CO2H)2] prodrug
PGA
PLL
PEG-NHS
Theranostic agents for combination therapy guided by T2-weighted MRINew strategy to construct polypeptide-based theranostics with tumor-microenvironment-activatable cascade reactions, promising for cancer treatment application[62]
Pt-based drug and Fe2+/3+ release triggered by the reducing reagent and pH conditions
Ferroptosis induction upon entering tumor microenvironment
Induction of apoptosis in tumor cell
Efficient inhibition of cancer cell growth
No significant systemic toxicity
PTCG NPsMetal–polyphenol-coordinated NPsEGCG
phenolic Pt(IV) prodrug
PEG-b-PPOH
High stability in blood circulationA promising strategy to develop advanced nanomedicine for cascade cancer therapy by efficiently combining chemotherapy and chemodynamic therapy with excellent anticancer efficacy[63]
Strong metal–polyphenol coordination interactions
Efficient drug release after cellular internalization
Increased OS by H2O2 generated by cisplatin and Fe-based Fenton reactions
Avoidance of systemic toxicity
Pt-Fe NCPsNanostructured coordination polymersPt(IV) prodrug bearing bis-catechol groups
Fe(III) ions
Dual pH- and redox-sensitivityOpening a future path for investigation of intranasal Pt nanoderivatives for brain tumor treatment, overcoming the blood–brain barrier permeability challenges, even in high-grade brain tumors such as glioblastom.[64]
Controlled release
Cytotoxic effect comparable to cisplatin
Slower release profile and activation period for the Pt(IV) prodrug activation
Increased accumulation of Pt in tumors in vivo
Complete cure and prolonged survival of the tested cohort in vivo
Intranasal administration
Reduced side-effects
Cetuximab-Pt-INPsUltrasonic emulsification-based self-assembled NPsMPEG-PLA-Pt(IV) prodrug (condensation between c,c,t-[Pt(NH3)2Cl2(OOCCH2CH2COOH)(OH)] and
MPEG-PLA)
Mal-PEG-PLA
Indocyanine green
cetuximab
Specific delivery of drugs to EGFR-A novel kind of cetuximab-decorated and NIR-activated NPs that can be selectively internalized into cancer cells via receptor-mediated endocytosis
Promising potential for targeted and effective therapy against EGFR-hyperexpressing cells of epidermoid carcinoma
[65]
Hyperexpressing cancer cells through cetuximab
Higher cytotoxicity and cancer cell uptake upon NIR irradiation
NPTGPLGA-PEG-based NPsOxaliplatin(IV)(Gem)2 (PTG)
PLGA-PEG-COOH
Reduced side-effectsSynergistic combination of established chemotherapeutic agents within a Pt(IV) scaffold, coupled with the potential benefits of NP delivery systems for more effective and tolerable anticancer treatments[66]
Overcoming drug-resistance mechanisms
Enhanced cancer targeting
Increased growth inhibition
Release of active molecules -Pt(II) and gemcitabine-under tumor-associated reductive medium
SSCV5 NPsNanoprecipitation-based self-assembled NPsPt-VES prodrug
PSSV polymer
GSH scavengingA safe and facile nano-sensitizer and a promising route for the neoadjuvant chemoradiotherapy of cervical cancers.[20]
mitochondrial damage
reverse of the cisplatin resistance by consuming intracellular GSH
DNA damage and apoptosis
Sensitization of cancer cells to X-ray radiation
Accumulation inside and growth inhibition of cisplatin-resistant cancer cells
Reduced side-effects
High Pt(IV) prodrug loading
capacity
IR780@Pt NPsSupramolecular self-assembly-formed NPsPt-CD prodrug
Ad-BH
IR780
NIR-irradiation induced mitochondrial dysfunction of cancer cellsInnovative nanomedicine IR780@Pt NPs mediating targeted induction of mitochondrial dysfunction to potentiate chemotherapy
High significance of combinatorial therapy by multifunctional nanotheranostics for synergistically enhanced cancer therapy
[67]
Downregulation of key proteins of the NER pathway, with enhanced chemotherapeutic effect
NIR fluorescence and photoacoustic imaging capacity
Increased tumor inhibition performance
PVPt@Cy NPsHydrophobic interaction-induced self-assembled NPsc,c,t-[Pt(NH3)2Cl2(OH)(O2CCH2CH2CH2CO2H)]
α-tochoferol
mPEG2K-OH
PMA
Cy
Synchronous chemotherapy, PDT and PTTPromising approach in imaging-guided combined chemo-phototherapy[68]
disaggregation under acidic, reductive conditions and NIR irradiation, with photothermal conversion and OS generation
An enhanced in vitro anticancer efficiency under irradiation
Strong NIR fluorescence and photothermal imaging
Pt–I–IR780 NPsSelf-assembled NPsP1 polymer (Pt(IV) in the main chain and pendant pair-wise iodides)
IR780
NIR irradiation-mediated mild hyperthermiaExternal NIR light to irradiate NPs with photothermal sensitizers to produce mild hyperthermia, which increases the formation of Pt-DNA interstrand cross-links with efficient Pt-based chemotherapy[69]
Increased GSH-mediated reduction in Pt(IV) to Pt(II)
Reduced GSH-mediated detoxification of cisplatin
Increased Pt-DNA interstrand cross-links
ABC triblock Pt-prodrug NPsSelf-assembled NPsPBI-tagged ABC-triblock PCL
Cisplatin
PEG
Excellent shielding against cisplatin detoxification by the intracellular GSH speciesNanoformulation with both enhanced efficiency against cancer cells and capacity to monitor intracellular administration of non-luminescent cisplatin[70]
Extracellular stability and intracellular lysosome-mediated biodegradation of PCL with release of Pt-drug
Enhanced cancer cell growth inhibition
Simultaneous monitoring and delivery aspects of the Pt-prodrug
PDDNEmulsion interfacial polymerization-based NPsPt(IV) prodrug derived from OXA
mitochondria-targeting cytotoxic peptide
High drug loading efficiencyAppreciable combination efficacy on both cell line-derived and patient-derived xenograft lung cancer model[71]
Combination therapy
Precise drug ratio
Ph-induced drug release
Activation upon GSH-mediated reduction
High biocompatibility and reduced side-effects
Inhibition of drug efflux and DNA damage repair
Appreciable antitumor effects
NP(Se)sMicellar NPsmPEG5000-(CHTA-co-DSB)-mPEG5000 (diselenium bond containing polymer)
phospholipid like morpholinized Pt(IV) prodrug (C16-CisPt-TA)
GSH depletion and ROS generationA promising strategy to break the redox balance for maximizing the efficacy of Pt-based cancer therapy[72]
Significant antitumor effect
Biocompatibility
Decreased drug resistance
NP-Pt-USP1iSelf-assembled NPsPHHM
C527 (USP1i)
Pt (IV)–C12 prodrug
GSH sensitivityUSP1i combined with a Pt(IV) prodrug in NPs could inhibit the growth of liver cancer, with the possibility of a future precise cancer therapy with cisplatin and USP1i to overcome cisplatin resistance in the clinic[31]
Inhibition of the DNA damage repair
Increased sensitivity of tumor cells to cisplatin
Enhanced anticancer effect
PEG-Pt(IV)@DOX NPsCo-self-assembled
NPs
PEG-Pt(IV) prodrug
doxorubicin
Longer blood retention A promising platform for combination chemotherapy of hypoxic solid tumors in the clinic[73]
Enhanced accumulation in tumor sites
Light irradiation-mediated in situ
Generation of O2, with release of active Pt(II) and doxorubicin
Increased ROS
Enhanced cytotoxicity
Alleviation of hypoxia-induced MDR of doxorubicin
Reduced side-effects
NanoplatinDTRSelf-assembled NPsPolyplatinDT
caspase-3 cleavable peptide
Delivery of drugs and fluorophores concomitantly at a precise D/T ratioThe first theranostic nanoplatform with anticancer drugs, drug tracers, and drug efficacy reporters that can work in concert to provide insight into the drug fate and mechanism of action[74]
Tracking of the Pt drugs via NIR-II imaging
Evaluation of the therapeutic efficacy via the apoptosis reporter
PCPt NPsSelf-assembled NPsPCPtFixed drugs loading ratioPCPt NPs as a promising platform for Pt and curcumin co-based combination chemotherapy circumventing mono-chemotherapy limitations[75]
High drugs loading content
Improved solubility and stability of curcumin
Drug release under reductive conditions
Efficient synergistic chemotherapy
Excellent reversal ability of tumor resistance to cisplatin
Effective intracellular uptake
Enhanced cell proliferation inhibition
Table 8. Current progress in bioorthogonal reactions catalyzed by PtNPs.
Table 8. Current progress in bioorthogonal reactions catalyzed by PtNPs.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
PEG-dPt-2Dendritic NPsdPt-2 (formed from H2PtCl6 and ascorbic acid assisted by Pluronic F-127)
PEG-SH
Bioorthogonal catalytic nanoreactors to enable the in situ release of anticancer drugs through depropargylation reactions Developing noble metal-based nanodevices in bioorthogonal catalysis
Offering new opportunities to modulate the optical properties and bioactivity of small molecules in the highly crowded intracellular environment
[76]
High biocompatibility
High surface area-to-volume ratio
Increased catalytic performance of Pt NPs
EA-Pt@MDBCOSelf-assembled polymeric micellesEA-Pt
PEOz-b-PLA-GSNO
DSPE-PEG2000-DBCO
Improved targeting performance toward pulmonary cancerous regions after prelabeling with azide via inhalationInhalable EA-Pt@MDBCO effectively reversed cisplatin resistance in an NSCLC model
New therapeutic option for lethal NSCLC in clinic
[77]
pH-sensitivity
Depletion of intracellular GSH
Inhibition of GSH-S-transferase activity
Improved therapeutic outcome against NSCLC
Reversion of cisplatin resistance
NO release within GSH-enriched cancer cells, with synergistic effects
Increase in the survival rate
In vivo biosafety
BDCNsSelf-assembled coordinative NPsCompound A—Pt(IV) prodrug bearing two terminal carboxyl groups
Compound B—NO prodrug O2-propargyl diazeniumdiolate with two terminal carboxyl groups
Fe3+ ions
Accumulation in tumor through passive targetingOvercoming of current bio-orthogonal chemistry shortcomings, especially the separated administration and targeting ability[78]
Stability in the circulation
Reduction in Pt(IV) to cisplatin in cancer cells
Cisplatin-mediated depropargylation of prodrug B to generate high levels of NO
Flexibly adjusted as needed in proportion to two prodrugs
Cascade reactions specifically initiated at the tumor site with both synergistic anticancer activity and reduced side-effects
Avoidance of pharmacokinetic complexity of separated administration
Enhanced the efficiency of bio-orthogonal reactions
Fe3+-initiated Fenton reaction-synthesized produced hydroxyl radicals with tumoricidal activity
Table 9. Latest advancements in the production of NPs delivering lipophilic Pt-based compounds.
Table 9. Latest advancements in the production of NPs delivering lipophilic Pt-based compounds.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
Dicarboxylate Pt(IV) Prodrugs
Self-assembled NPs
Self-assembled NPsDicarboxylato Pt(IV) prodrugs- Ace, But, Hex, OctFormation of nanoaggregates at the highest concentration tested for Oct and HexThis study underlined the important link between cancer cell uptake and lipophilicity of the prodrug. Lipophilic compounds can form nanoaggregates, especially at high concentrations, which can improve the cellular uptake through both passive diffusion and endocytosis but do not enhance the antiproliferative effect of complexes active at nanomolar concentrations.[81]
Higher cellular uptake by both passive diffusion and endocytosis for Oct
Antiproliferative activity not significantly impacted by aggregation for Oct
Increased lipophilicity
Disaggregation in the complete cell culture media (by virtue of the presence of bovine serum albumin)
Low zeta potentials causing low stability and high dispersity
[Pt(DACH)(OAc)(OPal)(ox)] incorporated PLGA NPsPt(IV) prodrug-incorporated polymeric NPs[Pt(DACH)(OAc)(OPal)(ox)]
PLGA
lipoid E80
OCA
Unique potency against a panel of cancer cells, including cisplatin-resistant tumor cellsModification of OXA into a lipophilic Pt(IV) complex containing both lipophilic and hydrophilic axial ligands improves performance and facilitates incorporation into polymeric NPs[82]
Enhanced in vitro cellular Pt accumulation, DNA platination, and antiproliferative effect compared to OXA
Decreased tumor growth rates compared to control and OXA treatment groups in vivo
Reduced systematic toxicity and side-effects
by incorporating the prodrug in PLGA NPs.
Table 10. Current directions in targeting both malignant and non-malignant cells.
Table 10. Current directions in targeting both malignant and non-malignant cells.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
BLZ@S-NP/Ptcore–shell–corona self-assembled NPsmPEG45-b-PHEP20/Pt
TK-PPE
BLZ-945
Ce6
Differentially targeting tumor cells and TAMsBLZ@S-NP/Pt differentially and precisely delivering agents to TAMs and tumor cells located in different spatial distribution
Synergistic anticancer effects in multiple tumor models
[83]
Shrinkage to small Pt(IV) prodrug-conjugating NPs
Deep tumor penetration under light irradiation
Release of BLZ-945 in the perivascular regions of tumor to deplete TAMs
Inhibition of tumor growth, prevention of metastasis, and increase in survival period under light irradiation
Reversion of the immunosuppressive tumor microenvironment and activation of the T cell-mediated antitumor immune response
Prolonged circulation in vivo
aPD-1/CDDP@NPsLipid-coated NPscis-[Pt(NH3)2(H2O)2]2(NO3)2
DOPA
DOTAP
DSPE-PEG-AA
Cholesterol
aPD-1
Synergistic immuno-chemotherapyDevelopment of a microneedle patch loaded with pH-responsive tumor-targeted lipid NPs which allows local delivery of aPD-1 and cisplatin precisely to cancer tissues for cancer therapy[85]
Effective increase in the immune response
Enhanced tumor regression
Microneedle-induced T-cell response
Lockage of PD-1 in T-cells by aPD-1, with enhanced T-cell activation
Increase in direct cytotoxicity of cisplatin in tumor cells
Increased response rate in the animal model unresponsive to aPD-1 systemic therapy
Improved tumor targeting
Decreased systemic toxicity
Table 11. Latest achievements in the production of NPs co-delivering Pt-based compounds and siRNAs.
Table 11. Latest achievements in the production of NPs co-delivering Pt-based compounds and siRNAs.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
Pt-TPNs/siRNATea Polyphenol NPsEGCG
anti-CSNK2A1 siRNA
cisplatin(IV) prodrug
Extended circulation timePt-TPNs/siRNA not only enhances the anticancer effects but also mitigates cisplatin-induced renal toxicity, achieving efficacy while reducing toxicity[13]
Increased accumulation within the cancer cells
Decreased Pt-drugs-associated renal side-effects
Augmented cisplatin susceptibility of cancer cells
NPt(IV)@siXkr8Self-assembled lipid NPsLipo-Pt(IV)-R8K
siXkr8
Reduction in phosphatidylserine level exposure, with subsequent decrease in immunosuppressionAmplification of conventional Pt-based drug anticancer effects of and suppression of cancer recurrence through improved chemoimmunotherapy[87]
Increased Pt-DNA cross-linking formation
Accumulation in tumors
Cancer cell nucleus targeting
Activation in a reduced microenvironment
LNPSelf-assembled lipid NPscisplatin prodrug
XPF-targeted siRNA
Efficient transport of the molecules into cells A multi-targeted NP system that can specifically silence an NER-related gene to promote apoptosis induced by cisplatin, especially in cisplatin-refractory tumors[88]
DNA damage
Downregulation of both mRNA and levels of XPF, potentiating the Pt drug
Improved cytotoxicity in both cisplatin-sensitive and -resistant human lung cancer cells
PLGA-PEG/G0-C14 NPsSelf-assembled NPs[Pt(NH3)2Cl2(O2C(CH2)8CH3)2] cisplatin prodrug
REV1/REV3L-specific siRNAs
PLGA-PEG block copolymers
G0-C14 cationic lipid
Suppression of REV1 and REV3L involved in the error-prone translesion DNA synthesisCo-delivering a DNA-damaging chemotherapeutic and siRNAs that impair the cell’s ability to repair the DNA damage, which can sensitize cancer cells to chemotherapeutics, and shows superior tumor inhibition compared with monochemotherapy[89]
Sensitization of resistant tumors to chemotherapy
Reduced frequency of acquired drug resistance of relapsed tumors
Table 12. Latest insights in developing human serum albumin-based NPs.
Table 12. Latest insights in developing human serum albumin-based NPs.
CompoundType of NPComponentsCharacteristicsMain FindingReferences
OSHB and OSHG NPsDesolvation technique-based
NPs
OXA-SS-HSA conjugate
BAC (for OSHB)
GLUT (for OSHG)
Small size, uniform surfaces, and a satisfactory encapsulation coefficientRelease of OXA in the tumor milieu via glutathione-sensitive prodrug degradation and NP disassembly
Smart nanomedicine strategy to realize a robust anticancer response with reduced off-target effects in triple-negative cancer therapy
[90]
Active tumor targeting via HSA
Dual reduction sensitivity to GSH (greater for OSHB)
Enhanced cytotoxicity and cell death (greater for OSHB)
Reduced drug resistance
Biocompatibility
Excellent tumor-suppressing efficacy
AbPlatin(IV)Self-assembled NPsCisPt(IV) hydrophobic Pt(IV) prodrug
HSA
Better tumor-targeting effectDevelopment of abplatin(IV) and the use of multi-omics for the mechanism elucidation of prodrug
Progress in clinical translation of the prodrug
[91]
Greater tumor inhibition rate even on cisplatin-resistant cells
Alterations of glycerophospholipids and sphingolipids in malignant cell membranes
Modifications of purine metabolism, with downregulated ATP and up-regulated xanthosine and hypoxanthine
Lower IC50 compared to cisplatin in vitro
Enhanced Pt-DNA adducts formation
Significant upregulation of ABAT and CLDN6 genes, with inhibition of cancer cell proliferation and apoptosis
Increased arginine and decreased carnitine, with apoptosis
HSA-His242-Pt-Dp44mT NPsHSA–Pt compound complex NPs[Pt(Dp44mT)Cl]
HSA
glutaraldehyde
Enhanced inhibition of tumor growthDeveloping a novel generation of Pt-based drugs
Enabling multimodal therapy, and further improving drug delivery
Important insights into the interaction of HSA with metal drugs but also supporting the medical background of HSA vectors
[92]
Reduced toxicity
Destruction of cancer cells by inducing apoptosis, autophagy, and inhibiting angiogenesis
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Iova, V.; Iova, G.M.; Tiron, A.T.; Scrobota, I.; Vlad, S.; Tudosie, M.S. Latest Achievements in the Development of Nanoparticle-Based Drug Delivery Systems of Pt Drugs and Prodrugs in Cancer Therapy. Pharmaceutics 2025, 17, 1267. https://doi.org/10.3390/pharmaceutics17101267

AMA Style

Iova V, Iova GM, Tiron AT, Scrobota I, Vlad S, Tudosie MS. Latest Achievements in the Development of Nanoparticle-Based Drug Delivery Systems of Pt Drugs and Prodrugs in Cancer Therapy. Pharmaceutics. 2025; 17(10):1267. https://doi.org/10.3390/pharmaceutics17101267

Chicago/Turabian Style

Iova, Vlad, Gilda Mihaela Iova, Andreea Taisia Tiron, Ioana Scrobota, Silviu Vlad, and Mihail Silviu Tudosie. 2025. "Latest Achievements in the Development of Nanoparticle-Based Drug Delivery Systems of Pt Drugs and Prodrugs in Cancer Therapy" Pharmaceutics 17, no. 10: 1267. https://doi.org/10.3390/pharmaceutics17101267

APA Style

Iova, V., Iova, G. M., Tiron, A. T., Scrobota, I., Vlad, S., & Tudosie, M. S. (2025). Latest Achievements in the Development of Nanoparticle-Based Drug Delivery Systems of Pt Drugs and Prodrugs in Cancer Therapy. Pharmaceutics, 17(10), 1267. https://doi.org/10.3390/pharmaceutics17101267

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop