Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (42)

Search Parameters:
Keywords = cisplatin prodrug

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
29 pages, 3081 KiB  
Review
“Non-Classical” Platinum Complexes: A Concise Review
by Adriana Bakalova, Nina Ruseva and Emiliya Cherneva
Int. J. Mol. Sci. 2025, 26(13), 6270; https://doi.org/10.3390/ijms26136270 - 28 Jun 2025
Viewed by 474
Abstract
The utilization of platinum complexes in medicine continues to be a prevalent treatment modality for diverse tumour types. However, it should be noted that certain platinum complexes are characterized by a high degree of toxicity. In recent years, there has been a focus [...] Read more.
The utilization of platinum complexes in medicine continues to be a prevalent treatment modality for diverse tumour types. However, it should be noted that certain platinum complexes are characterized by a high degree of toxicity. In recent years, there has been a focus among scientists on synthesizing “non-classic” platinum complexes, such as those with a trans-configuration, Pt(IV) complexes, and mixed ammine/amine platinum complexes, with the aim of reducing the toxic side effects of certain platinum complexes, including cisplatin. For instance, newly synthesized platinum complexes with a trans-configuration exhibited substantial cytotoxic activity which was comparable to that of the corresponding cis-isomers and cisplatin. This finding challenged the prevailing cis-geometry paradigm and prompted a re-evaluation of the structural activity relationships (SARs) of antitumour platinum complexes. It is widely accepted that Pt(IV) complexes act as prodrugs and release the active Pt(II) species. This property renders them promising candidates as anticancer drugs. Furthermore, it has been established that mixed ammine/amine platinum complexes are less toxic than cisplatin. In addition, compared to cisplatin, they have been observed to have equivalent or greater cytotoxic activity. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

15 pages, 2799 KiB  
Article
PEGylation Effects on Amphiphilic Platinum(IV) Complexes: Influence on Uptake, Activation, and Cytotoxicity
by Arpit Sharma, Md Al Amin, Man B. Kshetri, Suha Alqarni, Wjdan Jogadi, Jordan Solmen, Zexin Lin, Shirin Akter and Yao-Rong Zheng
Pharmaceutics 2025, 17(4), 440; https://doi.org/10.3390/pharmaceutics17040440 - 29 Mar 2025
Viewed by 864
Abstract
Background/Objectives: The utilization of amphiphilic Pt(IV) complexes as prodrugs offers a promising strategy to revolutionize Pt-based cancer therapy by enhancing drug delivery and activation. While PEGylation is widely used to optimize drug properties, its impact on the biological behavior of amphiphilic Pt(IV) complexes [...] Read more.
Background/Objectives: The utilization of amphiphilic Pt(IV) complexes as prodrugs offers a promising strategy to revolutionize Pt-based cancer therapy by enhancing drug delivery and activation. While PEGylation is widely used to optimize drug properties, its impact on the biological behavior of amphiphilic Pt(IV) complexes remains unclear. This study systematically investigates how the PEGylation of varying molecular weights influences their cytotoxicity, cellular uptake, and activation. Methods: Pt(IV) complexes were synthesized with PEG chains of different molecular weights using HATU-catalyzed amide bond formation and copper-free click chemistry. Their biological properties were assessed through cell-based analyses, focusing on cytotoxicity, cellular uptake, and activation by biological reductants. Results: Small PEG modifications retained the potent cytotoxicity of amphiphilic Pt(IV) prodrugs, whereas large PEG chains significantly reduced efficacy. The decrease in potency was linked to impaired cellular uptake and mitochondrial accumulation. Additionally, large PEG modifications slowed the reduction and activation of Pt(IV) prodrugs by biological reductants, further limiting their anticancer activities. Conclusions: These findings underscore the critical role of PEGylation in metallodrug design and provide key insights into optimizing PEGylation strategies for enhancing platinum–based cancer therapies. Full article
Show Figures

Figure 1

18 pages, 2189 KiB  
Review
Noble Metal Complexes in Cancer Therapy: Unlocking Redox Potential for Next-Gen Treatments
by Alina Stefanache, Alina Monica Miftode, Marcu Constantin, Roxana Elena Bogdan Goroftei, Iulia Olaru, Cristian Gutu, Alexandra Vornicu and Ionut Iulian Lungu
Inorganics 2025, 13(2), 64; https://doi.org/10.3390/inorganics13020064 - 19 Feb 2025
Cited by 1 | Viewed by 1139
Abstract
(1) Context: Cancer is still a major problem worldwide, and traditional therapies like radiation and chemotherapy often fail to alleviate symptoms because of side effects, systemic toxicity, and mechanisms of resistance. Beneficial anticancer effects that spare healthy tissues are made possible by [...] Read more.
(1) Context: Cancer is still a major problem worldwide, and traditional therapies like radiation and chemotherapy often fail to alleviate symptoms because of side effects, systemic toxicity, and mechanisms of resistance. Beneficial anticancer effects that spare healthy tissues are made possible by the distinctive redox characteristics of noble metal complexes, especially those containing palladium, gold, silver, and platinum. (2) Methods: The redox processes, molecular targets, and therapeutic uses of noble metal complexes in cancer have been the subject of much study over the last 20 years; novel approaches to ligand design, functionalization of nanoparticles, and tumor-specific drug delivery systems are highlighted. (3) Results: Recent developments include Pt(IV) prodrugs and terpyridine-modified Pt complexes for enhanced selectivity and decreased toxicity; platinum complexes, like cisplatin, trigger reactive oxygen species (ROS) production and DNA damage. Functionalized gold nanoparticles (AuNPs) improve targeted delivery and theranostic capabilities, while gold complexes, particularly Au(I) and Au(III), inhibit redox-sensitive processes such as thioredoxin reductase (TrxR). (4) Conclusions: Ag(I)-based compounds and nanoparticles (AgNPs) induce DNA damage and mitochondrial dysfunction by taking advantage of oxidative stress. As redox-based anticancer medicines, noble metal complexes have the ability to transform by taking advantage of certain biochemical features to treat cancer more effectively and selectively. Full article
(This article belongs to the Special Issue Biological Activity of Metal Complexes)
Show Figures

Graphical abstract

16 pages, 2698 KiB  
Article
Exploring the Anticancer Potential of NO-Donor Oxadiazole Assemblies Against Malignant Pleural Mesothelioma
by Irina A. Stebletsova, Alexander A. Larin, Egor M. Matnurov, Ivan V. Ananyev, Maria V. Babak and Leonid L. Fershtat
Pharmaceutics 2025, 17(2), 230; https://doi.org/10.3390/pharmaceutics17020230 - 10 Feb 2025
Viewed by 1295
Abstract
Background: Nitric oxide (NO) has been linked to the pathogenesis of asbestos-related pleural diseases, including an extremely aggressive cancer called malignant pleural mesothelioma (MPM). Given that MPM cells are characterized by a higher expression of NO synthases and elevated NO production relative to [...] Read more.
Background: Nitric oxide (NO) has been linked to the pathogenesis of asbestos-related pleural diseases, including an extremely aggressive cancer called malignant pleural mesothelioma (MPM). Given that MPM cells are characterized by a higher expression of NO synthases and elevated NO production relative to normal cells, the use of NO-donor compounds could potentially saturate the cancerous cells with NO, triggering their death. Methods: We developed a novel class of NO prodrugs by merging two NO-releasing components, 1,2,5-oxadiazole 2-oxides (furoxans) and 1,2,4-oxadiazoles, and studied their NO-releasing characteristics in a time-dependent manner using the Griess assay. The cytotoxicity against two human MPM cell lines and non-cancerous lung fibroblasts was evaluated using a colorimetric MTT assay. Results: All compounds exhibited excellent NO-donating properties, surpassing the capacity of two reference NO donor compounds, 3-carbamoyl-4-(hydroxymethyl)furoxan (CAS-1609) and 4-ethoxy-3-phenylsulphonylfuroxan (CHF-2363), by at least 1.5–3 times. All oxadiazole hybrids demonstrated high cytotoxicity against MPM cell lines in a low micromolar range, comparable or higher than the cytotoxicity of the standard-of-care drug cisplatin. Conclusions: Notably, the novel compounds displayed a markedly greater selectivity towards cancerous cells than cisplatin when compared with non-cancerous lung fibroblasts, aligning with the intended design. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Graphical abstract

16 pages, 2416 KiB  
Article
Sustainable Nanomedicine: Enhancement of Asplatin’s Cytotoxicity In Vitro and In Vivo Using Green-Synthesized Zinc Oxide Nanoparticles Formed via Microwave-Assisted and Gambogic Acid-Mediated Processes
by Hatem A. F. M. Hassan, Nada K. Sedky, Mohamed S. Nafie, Noha Khalil Mahdy, Iten M. Fawzy, Toka Waleed Fayed, Eduard Preis, Udo Bakowsky and Sherif Ashraf Fahmy
Molecules 2024, 29(22), 5327; https://doi.org/10.3390/molecules29225327 - 12 Nov 2024
Cited by 1 | Viewed by 2449
Abstract
Chemoresistance encountered using conventional chemotherapy demands novel treatment approaches. Asplatin (Asp), a novel platinum (IV) prodrug designed to release cisplatin and aspirin in a reductive environment, has demonstrated high cytotoxicity at reduced drug resistance. Herein, we investigated the ability of green-synthesized nanocarriers to [...] Read more.
Chemoresistance encountered using conventional chemotherapy demands novel treatment approaches. Asplatin (Asp), a novel platinum (IV) prodrug designed to release cisplatin and aspirin in a reductive environment, has demonstrated high cytotoxicity at reduced drug resistance. Herein, we investigated the ability of green-synthesized nanocarriers to enhance Asp’s efficacy. Zinc oxide nanoparticles (ZnO-NPs) were synthesized using a green microwave-assisted method with the reducing and capping agent gambogic acid (GA). These nanoparticles were then loaded with Asp, yielding Asp@ZnO-NPs. Transmission electron microscopy was utilized to study the morphological features of ZnO-NPs. Cell viability studies conducted on MDA-MB-231 breast cancer cells demonstrated the ability of the Asp@ZnO-NPs treatment to significantly decrease Asp’s half-maximal inhibitory concentration (IC50) (5 ± 1 µg/mL). This was further demonstrated using flow cytometric analysis that revealed the capacity of Asp@ZnO-NPs treatment to significantly increase late apoptotic fractions. Furthermore, in vivo studies carried out using solid Ehrlich carcinoma-bearing mice showed significant tumor volume reduction with the Asp@ZnO-NPs treatment (156.3 ± 7.6 mm3), compared to Asp alone (202.3 ± 8.4 mm3) and untreated controls (342.6 ± 10.3 mm3). The histopathological analysis further demonstrated the increased necrosis in Asp@ZnO-NPs-treated group. This study revealed that Asp@ZnO-NPs, synthesized using an eco-friendly approach, significantly enhanced Asp’s anticancer activity, offering a sustainable solution for potent anticancer formulations. Full article
Show Figures

Figure 1

11 pages, 4932 KiB  
Article
Engineering Novel Amphiphilic Platinum(IV) Complexes to Co-Deliver Cisplatin and Doxorubicin
by Wjdan Jogadi, Man B. Kshetri, Suha Alqarni, Arpit Sharma, May Cheline, Md Al Amin, Cynthia Sheets, Angele Nsoure-Engohang and Yao-Rong Zheng
Molecules 2024, 29(17), 4095; https://doi.org/10.3390/molecules29174095 - 29 Aug 2024
Cited by 2 | Viewed by 1288
Abstract
In this study, we report a novel platinum–doxorubicin conjugate that demonstrates superior therapeutic indices to cisplatin, doxorubicin, or their combination, which are commonly used in cancer treatment. This new molecular structure (1) was formed by conjugating an amphiphilic Pt(IV) prodrug of [...] Read more.
In this study, we report a novel platinum–doxorubicin conjugate that demonstrates superior therapeutic indices to cisplatin, doxorubicin, or their combination, which are commonly used in cancer treatment. This new molecular structure (1) was formed by conjugating an amphiphilic Pt(IV) prodrug of cisplatin with doxorubicin. Due to its amphiphilic nature, the Pt(IV)–doxorubicin conjugate effectively penetrates cell membranes, delivering both cisplatin and doxorubicin payloads intracellularly. The intracellular accumulation of these payloads was assessed using graphite furnace atomic absorption spectrometry and fluorescence imaging. Since the therapeutic effects of cisplatin and doxorubicin stem from their ability to target nuclear DNA, we hypothesized that the amphiphilic Pt(IV)–doxorubicin conjugate (1) would effectively induce nuclear DNA damage toward killing cancer cells. To test this hypothesis, we used flow the cytometric analysis of phosphorylated H2AX (γH2AX), a biomarker of nuclear DNA damage. The Pt(IV)–doxorubicin conjugate (1) markedly induced γH2AX in treated MDA-MB-231 breast cancer cells, showing higher levels than cells treated with either cisplatin or doxorubicin alone. Furthermore, MTT cell viability assays revealed that the enhanced DNA-damaging capability of complex 1 resulted in superior cytotoxicity and selectivity against human cancer cells compared to cisplatin, doxorubicin, or their combination. Overall, the development of this amphiphilic Pt(IV)–doxorubicin conjugate represents a new form of combination therapy with improved therapeutic efficacy. Full article
(This article belongs to the Special Issue Exclusive Feature Papers on Molecular Structure)
Show Figures

Figure 1

19 pages, 8754 KiB  
Article
Unraveling Novel Strategies in Mesothelioma Treatments Using a Newly Synthetized Platinum(IV) Compound
by Cristina Favaron, Ludovica Gaiaschi, Claudio Casali, Fabrizio De Luca, Federica Gola, Margherita Cavallo, Valeria Ramundo, Elisabetta Aldieri, Gloria Milanesi, Silvia Damiana Visonà, Mauro Ravera and Maria Grazia Bottone
Pharmaceutics 2024, 16(8), 1015; https://doi.org/10.3390/pharmaceutics16081015 - 31 Jul 2024
Cited by 1 | Viewed by 1747
Abstract
Malignant mesothelioma is a rare tumor associated with asbestos exposure. Mesothelioma carcinogenesis is related to enhanced reactive oxygen species (ROS) production and iron overload. Despite the recent advances in biomedical sciences, to date the only available treatments include surgery in a small fraction [...] Read more.
Malignant mesothelioma is a rare tumor associated with asbestos exposure. Mesothelioma carcinogenesis is related to enhanced reactive oxygen species (ROS) production and iron overload. Despite the recent advances in biomedical sciences, to date the only available treatments include surgery in a small fraction of patients and platinum-based chemotherapy in combination with pemetrexed. In this view, the purpose of this study was to evaluate the therapeutic potential of the newly synthetized platinum prodrug Pt(IV)Ac-POA compared to cisplatin (CDDP) on human biphasic mesothelioma cell line MSTO-211H using different complementary techniques, such as flow-cytometry, transmission electron microscopy (TEM), and immunocytochemistry. Healthy mesothelial cell lines Met-5A were also employed to assess the cytotoxicity of the above-mentioned compounds. Our in vitro results showed that Pt(IV)Ac-POA significantly interfere with iron metabolisms and more importantly is able to trigger cell death, through different pathways, including ferroptosis, necroptosis, and apoptosis, in neoplastic cells. On the other hand, CDDP triggers mainly apoptotic and necrotic cell death. In conclusion, Pt(IV)Ac-POA may represent a new promising pharmacological agent in the treatment of malignant mesothelioma. Full article
Show Figures

Figure 1

38 pages, 23775 KiB  
Article
Chemotherapeutic Potential of Chlorambucil-Platinum(IV) Prodrugs against Cisplatin-Resistant Colorectal Cancer Cells
by Maria George Elias, Angelico D. Aputen, Shadma Fatima, Timothy J. Mann, Shawan Karan, Meena Mikhael, Paul de Souza, Christopher P. Gordon, Kieran F. Scott and Janice R. Aldrich-Wright
Int. J. Mol. Sci. 2024, 25(15), 8252; https://doi.org/10.3390/ijms25158252 - 28 Jul 2024
Cited by 2 | Viewed by 3208
Abstract
Chlorambucil-platinum(IV) prodrugs exhibit multi-mechanistic chemotherapeutic activity with promising anticancer potential. The platinum(II) precursors of the prodrugs have been previously found to induce changes in the microtubule cytoskeleton, specifically actin and tubulin of HT29 colon cells, while chlorambucil alkylates the DNA. These prodrugs demonstrate [...] Read more.
Chlorambucil-platinum(IV) prodrugs exhibit multi-mechanistic chemotherapeutic activity with promising anticancer potential. The platinum(II) precursors of the prodrugs have been previously found to induce changes in the microtubule cytoskeleton, specifically actin and tubulin of HT29 colon cells, while chlorambucil alkylates the DNA. These prodrugs demonstrate significant anticancer activity in 2D cell and 3D spheroid viability assays. A notable production of reactive oxygen species has been observed in HT29 cells 72 h post treatment with prodrugs of this type, while the mitochondrial membrane potential was substantially reduced. The cellular uptake of the chlorambucil-platinum(IV) prodrugs, assessed by ICP-MS, confirmed that active transport was the primary uptake mechanism, with platinum localisation identified primarily in the cytoskeletal fraction. Apoptosis and necrosis were observed at 72 h of treatment as demonstrated by Annexin V-FITC/PI assay using flow cytometry. Immunofluorescence measured via confocal microscopy showed significant changes in actin and tubulin intensity and in architecture. Western blot analysis of intrinsic and extrinsic pathway apoptotic markers, microtubule cytoskeleton markers, cell proliferation markers, as well as autophagy markers were studied post 72 h of treatment. The proteomic profile was also studied with a total of 1859 HT29 proteins quantified by mass spectroscopy, with several dysregulated proteins. Network analysis revealed dysregulation in transcription, MAPK markers, microtubule-associated proteins and mitochondrial transport dysfunction. This study confirms that chlorambucil-platinum(IV) prodrugs are candidates with promising anticancer potential that act as multi-mechanistic chemotherapeutics. Full article
(This article belongs to the Special Issue Novel Biological Molecules for Cancer Treatments 2.0)
Show Figures

Graphical abstract

33 pages, 47187 KiB  
Article
A Phyto-mycotherapeutic Supplement, Namely Ganostile, as Effective Adjuvant in Brain Cancer Management: An In Vitro Study Using U251 Human Glioblastoma Cell Line
by Ludovica Gaiaschi, Fabrizio De Luca, Elisa Roda, Beatrice Ferrari, Claudio Casali, Chiara Rita Inguscio, Federica Gola, Enrico Pelloni, Elena Savino, Mauro Ravera, Paola Rossi and Maria Grazia Bottone
Int. J. Mol. Sci. 2024, 25(11), 6204; https://doi.org/10.3390/ijms25116204 - 5 Jun 2024
Cited by 7 | Viewed by 2206
Abstract
The current standard oncotherapy for glioblastoma is limited by several adverse side effects, leading to a short-term patient survival rate paralleled by a worsening quality of life (QoL). Recently, Complementary and Integrative Medicine’s (CIM) innovative approaches have shown positive impacts in terms of [...] Read more.
The current standard oncotherapy for glioblastoma is limited by several adverse side effects, leading to a short-term patient survival rate paralleled by a worsening quality of life (QoL). Recently, Complementary and Integrative Medicine’s (CIM) innovative approaches have shown positive impacts in terms of better response to treatment, side effect reduction, and QoL improvement. In particular, promising potential in cancer therapy has been found in compounds coming from phyto- and mycotherapy. The objective of this study was to demonstrate the beneficial effects of a new phyto-mycotherapy supplement, named Ganostile, in the human glioblastoma cell line U251, in combination with chemotherapeutic agents, i.e., Cisplatin and a new platinum-based prodrug. Choosing a supplement dosage that mimicked oral supplementation in humans (about 1 g/day), through in vitro assays, microscopy, and cytometric analysis, it has emerged that the cells, after 48hr continuous exposure to Ganostile in combination with the chemical compounds, showed a higher mortality and a lower proliferation rate than the samples subjected to the different treatments administered individually. In conclusion, our data support the use of Ganostile in integrative oncology protocols as a promising adjuvant able to amplify conventional and new drug effects and also reducing resistance mechanisms often observed in brain tumors. Full article
Show Figures

Figure 1

20 pages, 2267 KiB  
Article
Oxaliplatin(IV) Prodrugs Functionalized with Gemcitabine and Capecitabine Induce Blockage of Colorectal Cancer Cell Growth—An Investigation of the Activation Mechanism and Their Nanoformulation
by Carlo Marotta, Damiano Cirri, Ioannis Kanavos, Luisa Ronga, Ryszard Lobinski, Tiziana Funaioli, Chiara Giacomelli, Elisabetta Barresi, Maria Letizia Trincavelli, Tiziano Marzo and Alessandro Pratesi
Pharmaceutics 2024, 16(2), 278; https://doi.org/10.3390/pharmaceutics16020278 - 16 Feb 2024
Cited by 3 | Viewed by 2442
Abstract
The use of platinum-based anticancer drugs, such as cisplatin, oxaliplatin, and carboplatin, is a common frontline option in cancer management, but they have debilitating side effects and can lead to drug resistance. Combination therapy with other chemotherapeutic agents, such as capecitabine and gemcitabine, [...] Read more.
The use of platinum-based anticancer drugs, such as cisplatin, oxaliplatin, and carboplatin, is a common frontline option in cancer management, but they have debilitating side effects and can lead to drug resistance. Combination therapy with other chemotherapeutic agents, such as capecitabine and gemcitabine, has been explored. One approach to overcome these limitations is the modification of traditional Pt(II) drugs to obtain new molecules with an improved pharmacological profile, such as Pt(IV) prodrugs. The design, synthesis, and characterization of two novel Pt(IV) prodrugs based on oxaliplatin bearing the anticancer drugs gemcitabine or capecitabine in the axial positions have been reported. These complexes were able to dissociate into their constituents to promote cell death and induce apoptosis and cell cycle blockade in a representative colorectal cancer cell model. Specifically, the complex bearing gemcitabine resulted in being the most active on the HCT116 colorectal cancer cell line with an IC50 value of 0.49 ± 0.04. A pilot study on the encapsulation of these complexes in biocompatible PLGA-PEG nanoparticles is also included to confirm the retention of the pharmacological properties and cellular drug uptake, opening up to the possible delivery of the studied complexes through their nanoformulation. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Figure 1

14 pages, 758 KiB  
Review
The Immune Landscape and Immunotherapeutic Strategies in Platinum-Refractory Testicular Germ Cell Tumors
by Konstantinos Evmorfopoulos, Konstantinos Marsitopoulos, Raphael Karachalios, Athanasios Karathanasis, Konstantinos Dimitropoulos, Vassilios Tzortzis, Ioannis Zachos and Panagiotis J. Vlachostergios
Cancers 2024, 16(2), 428; https://doi.org/10.3390/cancers16020428 - 19 Jan 2024
Cited by 2 | Viewed by 2942
Abstract
Testicular germ cell tumors (TGCTs) are cancers with very good prognosis, even in the metastatic setting, with high curative potential mainly attributed to the introduction of cisplatin-based chemotherapy. However, approximately 15% of the patients develop platinum-refractory disease and suffer multiple relapses. Therefore, there [...] Read more.
Testicular germ cell tumors (TGCTs) are cancers with very good prognosis, even in the metastatic setting, with high curative potential mainly attributed to the introduction of cisplatin-based chemotherapy. However, approximately 15% of the patients develop platinum-refractory disease and suffer multiple relapses. Therefore, there is an unmet need for novel therapeutic agents with improved efficacy and minimal long-term side effects. Recent advances in the development of immunotherapeutic agents, particularly immune checkpoint inhibitors (ICIs), have offered an opportunity to test their activity in various tumor types, including GCTs. This review aims to analyze the immune microenvironment of these tumors and present the most recently available data from studies that have tested immunotherapeutic agents against GCTs. The majority of the available knowledge derives from case reports or small cohort studies, particularly those involving ICIs of the PD-1/PD-L1 axis alone or in combination with anti-CTLA-4 monoclonal antibodies. Other immunotherapeutic targeted approaches, including antibody-drug conjugates, antibody prodrugs, vaccines, tyrosine kinase inhibitors, chimeric antigen receptor (CAR) T-cell therapy, have biological rationales and have shown preliminary activity or are currently being tested. Growing evidence on these and other approaches will assist in broadening the currently limited treatment armamentarium against platinum-refractory TGCTs. Full article
(This article belongs to the Special Issue Immune Checkpoint Inhibitors for Urologic Cancers)
Show Figures

Figure 1

17 pages, 1680 KiB  
Article
Pyrenebutyrate Pt(IV) Complexes with Nanomolar Anticancer Activity
by Anife Ahmedova, Rositsa Mihaylova, Silviya Stoykova, Veronika Mihaylova, Nikola Burdzhiev, Viktoria Elincheva, Georgi Momekov and Denitsa Momekova
Pharmaceutics 2023, 15(9), 2310; https://doi.org/10.3390/pharmaceutics15092310 - 13 Sep 2023
Cited by 4 | Viewed by 1865
Abstract
Research on platinum-based anticancer drugs continuously strives to develop new non-classical platinum complexes. Pt(IV) prodrugs are the most promising, and their activation-by-reduction mechanism of action is being explored as a prospect for higher selectivity and efficiency. Herein, we present the anticancer potency and [...] Read more.
Research on platinum-based anticancer drugs continuously strives to develop new non-classical platinum complexes. Pt(IV) prodrugs are the most promising, and their activation-by-reduction mechanism of action is being explored as a prospect for higher selectivity and efficiency. Herein, we present the anticancer potency and chemical reactivity of Pt(IV) complexes formed by linking pyrene butyric acid with cisplatin. The results from cytotoxicity screening on 10 types of cancer cell lines and non-malignant cells (HEK-293) indicated IC50 values as low as 50–70 nM for the monosubstituted Pt(IV) complex against leukemia cell lines (HL-60 and SKW3) and a cisplatin-resistant derivative (HL-60/CDDP). Interestingly, the bis-substituted complex is virtually non-toxic to both healthy and cancerous cells of adherent types. Nevertheless, it shows high cytotoxicity against multidrug-resistant derivatives HL-60/CDDP and HL-60/Dox. The reactivity of the complexes with biological reductants was monitored by the NMR method. Furthermore, the platinum uptake by the treated cells was examined on two types of cellular cultures: adherent and suspension growing, and proteome profiling was conducted to track expression changes of key apoptosis-related proteins in HL-60 cells. The general conclusion points to a possible cytoskeletal entrapment of the bulkier bis-pyrene complex that could be limiting its cytotoxicity to adherent cells, both cancerous and healthy ones. Full article
(This article belongs to the Special Issue Novel Metal-Based Drugs for Anticancer and Antiviral Applications)
Show Figures

Figure 1

22 pages, 31839 KiB  
Article
A Photoactivated Ru (II) Polypyridine Complex Induced Oncotic Necrosis of A549 Cells by Activating Oxidative Phosphorylation and Inhibiting DNA Synthesis as Revealed by Quantitative Proteomics
by Li Zhu, Hui Liu, Yang Dou, Qun Luo, Liangzhen Gu, Xingkai Liu, Qianxiong Zhou, Juanjuan Han and Fuyi Wang
Int. J. Mol. Sci. 2023, 24(9), 7756; https://doi.org/10.3390/ijms24097756 - 24 Apr 2023
Cited by 5 | Viewed by 2799
Abstract
The ruthenium polypyridine complex [Ru(dppa)2(pytp)] (PF6)2 (termed as ZQX-1), where dppa = 4,7-diphenyl-1,10-phenanthroline and pytp = 4′-pyrene-2,2′:6′,2′′-terpyridine, has been shown a high and selective cytotoxicity to hypoxic and cisplatin-resistant cancer cells either under irradiation with blue light or [...] Read more.
The ruthenium polypyridine complex [Ru(dppa)2(pytp)] (PF6)2 (termed as ZQX-1), where dppa = 4,7-diphenyl-1,10-phenanthroline and pytp = 4′-pyrene-2,2′:6′,2′′-terpyridine, has been shown a high and selective cytotoxicity to hypoxic and cisplatin-resistant cancer cells either under irradiation with blue light or upon two-photon excitation. The IC50 values of ZQX-1 towards A549 cancer cells and HEK293 health cells are 0.16 ± 0.09 µM and >100 µM under irradiation at 420 nm, respectively. However, the mechanism of action of ZQX-1 remains unclear. In this work, using the quantitative proteomics method we identified 84 differentially expressed proteins (DEPs) with |fold-change| ≥ 1.2 in A549 cancer cells exposed to ZQX-1 under irradiation at 420 nm. Bioinformatics analysis of the DEPs revealed that photoactivated ZQX-1 generated reactive oxygen species (ROS) to activate oxidative phosphorylation signaling to overproduce ATP; it also released ROS and pyrene derivative to damage DNA and arrest A549 cells at S-phase, which synergistically led to oncotic necrosis and apoptosis of A549 cells to deplete excess ATP, evidenced by the elevated level of PRAP1 and cleaved capase-3. Moreover, the DNA damage inhibited the expression of DNA repair-related proteins, such as RBX1 and GPS1, enhancing photocytotoxicity of ZQX-1, which was reflected in the inhibition of integrin signaling and disruption of ribosome assembly. Importantly, the photoactivated ZQX-1 was shown to activate hypoxia-inducible factor 1A (HIF1A) survival signaling, implying that combining use of ZQX-1 with HIF1A signaling inhibitors may further promote the photocytotoxicity of the prodrug. Full article
Show Figures

Figure 1

22 pages, 3648 KiB  
Article
Platinum(IV)-Loaded Degraded Glycol Chitosan as Efficient Platinum(IV) Drug Delivery Platform
by Yvonne Lerchbammer-Kreith, Nadine S. Sommerfeld, Klaudia Cseh, Xian Weng-Jiang, Uchechukwu Odunze, Andreas G. Schätzlein, Ijeoma F. Uchegbu, Mathea S. Galanski, Michael A. Jakupec and Bernhard K. Keppler
Pharmaceutics 2023, 15(4), 1050; https://doi.org/10.3390/pharmaceutics15041050 - 24 Mar 2023
Cited by 5 | Viewed by 3399
Abstract
A new class of anticancer prodrugs was designed by combining the cytotoxicity of platinum(IV) complexes and the drug carrier properties of glycol chitosan polymers: Unsymmetrically carboxylated platinum(IV) analogues of cisplatin, carboplatin and oxaliplatin, namely (OC-6-44)-acetatodiammine(3-carboxypropanoato)dichloridoplatinum(IV), (OC-6-44)-acetaodiammine(3-carboxypropanoato)(cyclobutane-1,1-dicarboxylato)platinum(IV) and (OC-6-44)-acetato(3-carboxypropanoato)(1R,2R-cyclohexane-1,2-diamine)oxalatoplatinum(IV) were synthesised and conjugated via [...] Read more.
A new class of anticancer prodrugs was designed by combining the cytotoxicity of platinum(IV) complexes and the drug carrier properties of glycol chitosan polymers: Unsymmetrically carboxylated platinum(IV) analogues of cisplatin, carboplatin and oxaliplatin, namely (OC-6-44)-acetatodiammine(3-carboxypropanoato)dichloridoplatinum(IV), (OC-6-44)-acetaodiammine(3-carboxypropanoato)(cyclobutane-1,1-dicarboxylato)platinum(IV) and (OC-6-44)-acetato(3-carboxypropanoato)(1R,2R-cyclohexane-1,2-diamine)oxalatoplatinum(IV) were synthesised and conjugated via amide bonding to degraded glycol chitosan (dGC) polymers with different chain lengths (5, 10, 18 kDa). The 15 conjugates were investigated with 1H and 195Pt NMR spectroscopy, and average amounts of platinum(IV) units per dGC polymer molecule with ICP-MS, revealing a range of 1.3–22.8 platinum(IV) units per dGC molecule. Cytotoxicity was tested with MTT assays in the cancer cell lines A549, CH1/PA-1, SW480 (human) and 4T1 (murine). IC50 values in the low micromolar to nanomolar range were obtained, and higher antiproliferative activity (up to 72 times) was detected with dGC-platinum(IV) conjugates in comparison to platinum(IV) counterparts. The highest cytotoxicity (IC50 of 0.036 ± 0.005 µM) was determined in CH1/PA-1 ovarian teratocarcinoma cells with a cisplatin(IV)–dGC conjugate, which is hence 33 times more potent than the corresponding platinum(IV) complex and twice more potent than cisplatin. Biodistribution studies of an oxaliplatin(IV)–dGC conjugate in non-tumour-bearing Balb/C mice showed an increased accumulation in the lung compared to the unloaded oxaliplatin(IV) analogue, arguing for further activity studies. Full article
(This article belongs to the Section Drug Targeting and Design)
Show Figures

Figure 1

19 pages, 2666 KiB  
Article
Development of Novel Pt(IV)-Carbohydrate Derivatives as Targeted Anticancer Agents against Osteosarcoma
by Eoin Moynihan, Silvia Panseri, Giada Bassi, Arianna Rossi, Elisabetta Campodoni, Eithne Dempsey, Monica Montesi, Trinidad Velasco-Torrijos and Diego Montagner
Int. J. Mol. Sci. 2023, 24(7), 6028; https://doi.org/10.3390/ijms24076028 - 23 Mar 2023
Cited by 5 | Viewed by 2840
Abstract
Despite the enormous importance of cisplatin as a chemotherapeutic agent, its application is impacted by dose-limiting side effects and lack of selectivity for cancer cells. Researchers can overcome these issues by taking advantage of the pro-drug nature of the platinum(IV) oxidation state, and [...] Read more.
Despite the enormous importance of cisplatin as a chemotherapeutic agent, its application is impacted by dose-limiting side effects and lack of selectivity for cancer cells. Researchers can overcome these issues by taking advantage of the pro-drug nature of the platinum(IV) oxidation state, and by modifying the coordination sphere of the metal centre with specific vectors whose receptors are overexpressed in tumour cell membranes (e.g., carbohydrates). In this paper we report the synthesis of four novel carbohydrate-modified Pt(IV) pro-drugs, based on the cisplatin scaffold, and their biological activity against osteosarcoma (OS), a malignant tumour which is most common in adolescents and young adults. The carbohydrate-targeting vectors and Pt scaffold are linked using copper-catalysed azide–alkyne cycloaddition (CuAAC) chemistry, which is synonymous with mild and robust reaction conditions. The novel complexes are characterised using multinuclear 1D-2D NMR (1H, 13C and 195Pt), IR, HR-MS, Elem. Analyses, and CV. Cytotoxicity on 2D and 3D and cell morphology studies on OS cell lines, as well as non-cancerous human foetal osteoblasts (hFOBs), are discussed. Full article
(This article belongs to the Special Issue Metal-Based Complexes in Cancer 2.0)
Show Figures

Figure 1

Back to TopTop