Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.775
Filter
1.
Int J Nanomedicine ; 19: 5227-5243, 2024.
Article in English | MEDLINE | ID: mdl-38855734

ABSTRACT

Purpose: This study aimed to construct targeting drug-loading nanocomposites (FA-FePt/DDP nanoliposomes) to explore their potential in ovarian cancer therapy and molecular magnetic resonance imaging (MMRI). Methods: FA-FePt-NPs were prepared by coupling folate (FA) with polyethylene-glycol (PEG)-coated ferroplatinum nanoparticles and characterized. Then cisplatin (DDP) was encapsulated in FA-FePt-NPs to synthesize FA-PEG-FePt/DDP nanoliposomes by thin film-ultrasonic method and high-speed stirring, of which MMRI potential, magnetothermal effect, and the other involved performance were analyzed. The therapeutic effect of FA-FePt/DDP nanoliposomes combined with magnetic fluid hyperthermia (MFH) on ovarian cancer in vitro and in vivo was evaluated. The expression levels of Bax and epithelial-mesenchymal transition related proteins were detected. The biosafety was also preliminarily observed. Results: The average diameter of FA-FePt-NPs was about 30 nm, FA-FePt/DDP nanoliposomes were about 70 nm in hydrated particle size, with drug slow-release and good cell-specific targeted uptake. In an alternating magnetic field (AMF), FA-FePt/DDP nanoliposomes could rapidly reach the ideal tumor hyperthermia temperature (42~44 °C). MRI scan showed that FA-FePt-NPs and FA-FePt/DDP nanoliposomes both could suppress the T2 signal, indicating a good potential for MMRI. The in vitro and in vivo experiments showed that FA-FePt/DDP-NPs in AMF could effectively inhibit the growth of ovarian cancer by inhibiting cancer cell proliferation, invasion, and migration, and inducing cancer cell apoptosis, much better than that of the other individual therapies; molecularly, E-cadherin and Bax proteins in ovarian cancer cells and tissues were significantly increased, while N-cadherin, Vimentin, and Bcl-2 proteins were inhibited, effectively inhibiting the malignant progression of ovarian cancer. In addition, no significant pathological injury and dysfunction was observed in major visceras. Conclusion: We successfully synthesized FA-FePt/DDP nanoliposomes and confirmed their good thermochemotherapeutic effect in AMF and MMRI potential on ovarian cancer, with no obvious side effects, providing a favorable strategy of integrated targeting therapy and diagnosis for ovarian cancer.


Subject(s)
Antineoplastic Agents , Cisplatin , Folic Acid , Liposomes , Magnetic Resonance Imaging , Ovarian Neoplasms , Polyethylene Glycols , Female , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/therapy , Liposomes/chemistry , Cisplatin/pharmacology , Cisplatin/chemistry , Cisplatin/administration & dosage , Cisplatin/pharmacokinetics , Animals , Folic Acid/chemistry , Folic Acid/pharmacology , Folic Acid/pharmacokinetics , Humans , Magnetic Resonance Imaging/methods , Polyethylene Glycols/chemistry , Cell Line, Tumor , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Mice , Platinum/chemistry , Platinum/pharmacology , Hyperthermia, Induced/methods , Nanocomposites/chemistry , Mice, Nude , Mice, Inbred BALB C , Metal Nanoparticles/chemistry , Magnetic Fields , Particle Size
2.
J Am Chem Soc ; 146(19): 13126-13132, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38696488

ABSTRACT

Cisplatin, a cornerstone in cancer chemotherapy, is known for its DNA-binding capacity and forms lesions that lead to cancer cell death. However, the repair of these lesions compromises cisplatin's effectiveness. This study investigates how phosphorylation of HMGB1, a nuclear protein, modifies its binding to cisplatin-modified DNA (CP-DNA) and thus protects it from repair. Despite numerous methods for detecting protein-DNA interactions, quantitative approaches for understanding their molecular mechanism remain limited. Here, we applied click chemistry-based single-molecule force spectroscopy, achieving high-precision quantification of the interaction between phosphorylated HMGB1 and CP-DNA. This method utilizes a synergy of click chemistry and enzymatic ligation for precise DNA-protein immobilization and interaction in the system. Our results revealed that HMGB1 binds to CP-DNA with a significantly high rupture force of ∼130 pN, stronger than most natural DNA-protein interactions and varying across different DNA sequences. Moreover, Ser14 is identified as the key phosphorylation site, enhancing the interaction's kinetic stability by 35-fold. This increase in stability is attributed to additional hydrogen bonding suggested by molecular dynamics (MD) simulations. Our findings not only reveal the important role of phosphorylated HMGB1 in potentially improving cisplatin's therapeutic efficacy but also provide a precise method for quantifying protein-DNA interactions.


Subject(s)
Cisplatin , Click Chemistry , DNA , HMGB1 Protein , Molecular Dynamics Simulation , HMGB1 Protein/metabolism , HMGB1 Protein/chemistry , Cisplatin/chemistry , Cisplatin/pharmacology , Cisplatin/metabolism , Phosphorylation , DNA/chemistry , DNA/metabolism , Humans , Protein Binding , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
3.
Int J Nanomedicine ; 19: 4719-4733, 2024.
Article in English | MEDLINE | ID: mdl-38813391

ABSTRACT

Introduction: Lung cancer's high incidence and dismal prognosis with traditional treatments like surgery and radiotherapy necessitate innovative approaches. Despite advancements in nanotherapy, the limitations of single-treatment modalities and significant side effects persist. To tackle lung cancer effectively, we devised a temperature-sensitive hydrogel-based local injection system with near-infrared triggered drug release. Utilizing 2D MXene nanosheets as carriers loaded with R837 and cisplatin (DDP), encapsulated within a temperature-sensitive hydrogel-forming PEG-MXene@DDP@R837@SHDS (MDR@SHDS), we administered in situ injections of MDR@SHDS into tumor tissues combined with photothermal therapy (PTT). The immune adjuvant R837 enhances dendritic cell (DC) maturation and tumor cell phagocytosis, while PTT induces tumor cell apoptosis and necrosis by converting light energy into heat energy. Methods: Material characterization employed transmission electron microscopy, X-ray photoelectron spectroscopy, phase transition temperature, and near-infrared thermography. In vitro experiments assessed Lewis cell proliferation and apoptosis using CCK-8, Edu, and TUNEL assays. In vivo experiments on C57 mouse Lewis transplant tumors evaluated the photothermal effect via near-infrared thermography and assessed DC maturation and CD4+/CD8+ T cell ratios using flow cytometry. The in vivo anti-tumor efficacy of MDR@SHDS was confirmed by tumor growth curve recording and HE and TUNEL staining of tumor sections. Results: The hydrogel exhibited excellent temperature sensitivity, controlled release properties, and high biocompatibility. In vitro experiments revealed that MDR@SHDS combined with PTT had a greater inhibitory effect on tumor cell proliferation compared to MDR@SHD alone. Combining local immunotherapy, chemotherapy, and PTT yielded superior anti-tumor effects than individual treatments. Conclusion: MDR@SHDS, with its simplicity, biocompatibility, and enhanced anti-tumor effects in combination with PTT, presents a promising therapeutic approach for lung cancer treatment, offering potential clinical utility.


Subject(s)
Cisplatin , Imiquimod , Lung Neoplasms , Mice, Inbred C57BL , Animals , Cisplatin/pharmacology , Cisplatin/chemistry , Cisplatin/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Imiquimod/chemistry , Imiquimod/administration & dosage , Imiquimod/pharmacology , Hydrogels/chemistry , Apoptosis/drug effects , Nanostructures/chemistry , Photothermal Therapy/methods , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Delivery Systems/methods , Humans , Temperature , Dendritic Cells/drug effects , Drug Carriers/chemistry , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/pathology
4.
J Mol Model ; 30(6): 187, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38801468

ABSTRACT

CONTEXT: A systematic study of hydrogen bonds in base pairs and the interaction of cisplatin with DNA fragments was carried out. Structure, binding energies, and electron density were analyzed. xTB has proven to be an accurate method for obtaining structures and binding energies in DNA structures. Our xTB values for DNA base binding energy were in the same order and in some cases better than CAM-B3LYP values compared to experimental values. Double-stranded DNA-cisplatin structures have been calculated and the hydrogen bonds of water molecules are a decisive factor contributing to the preference for the cisplatin-Guanine interaction. Higher values of the water hydrogen bonding energies were obtained in cisplatin-Guanine structures. Furthermore, the electrostatic potential was used to investigate and improve the analysis of DNA-cisplatin structures. METHODS: We applied the xTB method and the CAM-B3LYP functional combined with def2-SVP basis set to perform and analyze of the bonding energies of the cisplatin interaction and the effects of the hydrogen bonds. Results were calculated employing the xTB and the ORCA software.


Subject(s)
Cisplatin , DNA , Hydrogen Bonding , Cisplatin/chemistry , DNA/chemistry , Static Electricity , Density Functional Theory , Models, Molecular , Thermodynamics , Water/chemistry , Antineoplastic Agents/chemistry , Base Pairing
5.
ACS Appl Bio Mater ; 7(5): 3431-3440, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38697834

ABSTRACT

Light-induced release of cisplatin from Pt(IV) prodrugs represents a promising approach for precise control over the antiproliferative activity of Pt-based chemotherapeutic drugs. This method has the potential to overcome crucial drawbacks of conventional cisplatin therapy, such as high general toxicity toward healthy organs and tissues. Herein, we report two Pt(IV) prodrugs with BODIPY-based photoactive ligands Pt-1 and Pt-2, which were designed using carbamate and triazole linkers, respectively. Both prodrugs demonstrated the ability to release cisplatin under blue light irradiation without the requirement of an external reducing agent. Dicarboxylated Pt-2 prodrug turned out to be more stable in the dark and more sensitive to light than its monocarbamate Pt-1 counterpart; these observations were explained using DFT calculations. The investigation of the photoreduction mechanism of Pt-1 and Pt-2 prodrugs using DFT modeling and ΔG0 PET estimation suggests that the photoinduced electron transfer from the singlet excited state of the BODIPY axial ligand to the Pt(IV) center is the key step in the light-induced release of cisplatin from the complexes. Cytotoxicity studies demonstrated that both prodrugs were nontoxic in the dark and toxic to MCF-7 cells under low-dose irradiation with blue light, and the observed effect was solely due to the cisplatin release from the Pt(IV) prodrugs. Our research presents an elegant synthetic approach to light-activated Pt(IV) prodrugs and presents findings that may contribute to the future rational design of photoactivatable Pt(IV) prodrugs.


Subject(s)
Antineoplastic Agents , Drug Screening Assays, Antitumor , Light , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Molecular Structure , Materials Testing , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/chemical synthesis , Cell Survival/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , Cisplatin/chemistry , Particle Size , Boron Compounds/chemistry , Boron Compounds/pharmacology , Boron Compounds/chemical synthesis , Photochemical Processes , Density Functional Theory
6.
ACS Nano ; 18(21): 13683-13695, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38749906

ABSTRACT

Tumor metastases and reoccurrence are considered the leading causes of cancer-associated deaths. As an emerging therapeutic method, increasing research efforts have been devoted to immunogenic cell death (ICD)-inducing compounds to solve the challenge. The clinically approved chemotherapeutic Pt complexes are not or are only poorly able to trigger ICD. Herein, the axial functionalization of the Pt(II) complex cisplatin with perfluorocarbon chains into ICD-inducing Pt(IV) prodrugs is reported. Strikingly, while the Pt(II) complex as well as the perfluorocarbon ligands did not induce ICD, the Pt(IV) prodrug demonstrated unexpectantly the induction of ICD through accumulation in the endoplasmic reticulum and generation of reactive oxygen species in this organelle. To enhance the pharmacological properties, the compound was encapsulated with human serum albumin into nanoparticles. While selectively accumulating in the tumorous tissue, the nanoparticles demonstrated a strong tumor growth inhibitory effect against osteosarcoma inside a mouse model. In vivo tumor vaccine analysis also demonstrated the ability of Pt(IV) to be an ideal ICD inducer. Overall, this study reports on axially perfluorocarbon chain-modified Pt(IV) complexes for ICD induction and chemoimmunotherapy in osteosarcoma.


Subject(s)
Antineoplastic Agents , Fluorocarbons , Immunotherapy , Serum Albumin, Human , Fluorocarbons/chemistry , Fluorocarbons/pharmacology , Humans , Animals , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Serum Albumin, Human/chemistry , Cisplatin/pharmacology , Cisplatin/chemistry , Cell Line, Tumor , Nanoparticles/chemistry , Prodrugs/chemistry , Prodrugs/pharmacology , Cell Proliferation/drug effects , Platinum/chemistry , Platinum/pharmacology , Mice, Inbred BALB C , Immunogenic Cell Death/drug effects
7.
Int J Biol Macromol ; 269(Pt 1): 132074, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705320

ABSTRACT

Treatment for triple negative breast cancer (TNBC) remains a huge challenge due to the lack of targeted therapeutics and tumor heterogenicity. Cisplatin (Cis) have demonstrated favorable therapeutic response in TNBC and thus is used together with various kinase inhibitors to fight the heterogenicity of TNBC. The combination of Cis with SRC inhibitor dasatinib (DAS) has shown encouraging anti-TNBC efficacy although the additive toxicity was commonly observed. To overcome the severe side effects of this Cis involved therapy, here we co-encapsulated Cis and DAS into a self-assembled hyaluronan (HA) nanogel (designated as HA/Cis/DAS (HCD) nanogel) to afford the TNBC targeted delivery by using the 4T1 mouse model. The acquired HCD nanogel was around 181 nm in aqueous solution, demonstrating the pharmacological activities of both Cis and DAS. Taking advantages of HA's targeting capability towards CD44 that is overexpressed on many TNBC cells, the HCD could well maintain the anticancer efficacy of the Cis and DAS combination, significantly increase the maximum tolerated dose and relieve the renal toxicity in vivo. The current HCD nanogel provides a potent strategy to improve the therapeutic outcome of Cis and DAS combination and thus representing a new targeted treatment option for TNBC.


Subject(s)
Cisplatin , Dasatinib , Hyaluronic Acid , Nanogels , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Hyaluronic Acid/chemistry , Animals , Dasatinib/pharmacology , Dasatinib/chemistry , Mice , Cisplatin/pharmacology , Cisplatin/chemistry , Female , Nanogels/chemistry , Cell Line, Tumor , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Polyethyleneimine/chemistry , Mice, Inbred BALB C , Hyaluronan Receptors/metabolism
8.
Int J Pharm ; 656: 124093, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38583822

ABSTRACT

A multifunctional nanoplatform was constructed in this work, with the goal of ameliorating the challenges faced with traditional cancer chemotherapy. Cisplatin (CP) was loaded into mesoporous polydopamine (mPDA) nanoparticles (NPs) with a drug loading of 15.8 ± 0.1 %, and MnO2 used as pore sealing agent. Finally, the NPs were wrapped with platelet membrane (PLTM). P-selectin on the PLTM can bind to CD44, which is highly expressed on the tumor cell membrane, so as to improve the targeting performance of the NPs. In addition, the CD47 on the PLTM can prevent the NPs from being phagocytosed by macrophages, which is conducive to immune escape. The final PLTM-CP@mPDA/MnO2 NPs were found to have a particle size of approximately 198 nm. MnO2 is degraded into Mn2+ in the tumor microenvironment, leading to CP release from the pores in the mPDA. CP both acts as a chemotherapy agent and can also increase the concentration of H2O2 in cells. Mn2+ can catalyze the conversion of H2O2 to OH, resulting in oxidative damage and chemodynamic therapy. In addition, Mn2+ can be used as a contrast agent in magnetic resonance imaging (MRI). In vitro and in vivo experiments were performed to explore the therapeutic effect of the NPs. When the concentration of CP is 30 µg/mL, the NPs cause approximately 50 % cell death. It was found that the PLTM-CP@mPDA/MnO2 NPs are targeted to cancerous cells, and in the tumor site cause extensive apoptosis. Tumor growth is thereby repressed. No negative off-target side effects were noted. MRI could be used to confirm the presence of the NPs in the tumor site. Overall, the nano-platform developed here provides cooperative chemotherapy and chemodynamic therapy, and can potentially be used for effective cancer treatment which could be monitored by MRI.


Subject(s)
Antineoplastic Agents , Blood Platelets , Cisplatin , Indoles , Manganese Compounds , Nanoparticles , Oxides , Polymers , Manganese Compounds/chemistry , Cisplatin/administration & dosage , Cisplatin/pharmacology , Cisplatin/chemistry , Polymers/chemistry , Indoles/chemistry , Indoles/administration & dosage , Animals , Oxides/chemistry , Nanoparticles/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Mice , Blood Platelets/drug effects , Blood Platelets/metabolism , Drug Liberation , Porosity , Mice, Inbred BALB C , Magnetic Resonance Imaging , Drug Carriers/chemistry , Female , Hydrogen Peroxide , Particle Size , Mice, Nude
9.
ACS Nano ; 18(17): 11217-11233, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38627234

ABSTRACT

Due to its intrinsic tumor-targeting attribute, limited immunogenicity, and cage architecture, ferritin emerges as a highly promising nanocarrier for targeted drug delivery. In the effort to develop ferritin cage-encapsulated cisplatin (CDDP) as a therapeutic agent, we found unexpectedly that the encapsulation led to inactivation of the drug. Guided by the structural information, we deciphered the interactions between ferritin cages and CDDP, and we proposed a potential mechanism responsible for attenuating the antitumor efficacy of CDDP encapsulated within the cage. Six platinum prodrugs were then designed to avoid the inactivation. The antitumor activities of these ferritin-platinum prodrug complexes were then evaluated in cells of esophageal squamous cell carcinoma (ESCC). Compared with free CDDP, the complexes were more effective in delivering and retaining platinum in the cells, leading to increased DNA damage and enhanced cytotoxic action. They also exhibited improved pharmacokinetics and stronger antitumor activities in mice bearing ESCC cell-derived xenografts as well as patient-derived xenografts. The successful encapsulation also illustrates the critical significance of comprehending the interactions between small molecular drugs and ferritin cages for the development of precision-engineered nanocarriers.


Subject(s)
Antineoplastic Agents , Cisplatin , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Ferritins , Prodrugs , Prodrugs/chemistry , Prodrugs/pharmacology , Humans , Ferritins/chemistry , Ferritins/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Mice , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Cisplatin/pharmacology , Cisplatin/chemistry , Drug Design , Platinum/chemistry , Platinum/pharmacology , Mice, Nude , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Drug Delivery Systems
10.
J Control Release ; 370: 14-42, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615892

ABSTRACT

Cisplatin (CDDP), as a broad-spectrum anticancer drug, is able to bind to DNA and inhibit cell division. Despite the widespread use of cisplatin since its discovery, cisplatin resistance developed during prolonged chemotherapy, similar to other small molecule chemotherapeutic agents, severely limits its clinical application. Cisplatin resistance in cancer cells is mainly caused by three reasons: DNA repair, decreased cisplatin uptake/increased efflux, and cisplatin inactivation. In earlier combination therapies, the emergence of multidrug resistance (MDR) in cancer cells prevented the achievement of the desired therapeutic effect even with the accurate combination of two chemotherapeutic drugs. Therefore, combination therapy using nanocarriers for co-delivery of drugs is considered to be ideal for alleviating cisplatin resistance and reducing cisplatin-related toxicity in cancer cells. This article provides an overview of the design of cisplatin nano-drugs used to combat cancer cell resistance, elucidates the mechanisms of action of cisplatin and the pathways through which cancer cells develop resistance, and finally discusses the design of drugs and related carriers that can synergistically reduce cancer resistance when combined with cisplatin.


Subject(s)
Antineoplastic Agents , Cisplatin , Drug Resistance, Neoplasm , Neoplasms , Cisplatin/administration & dosage , Cisplatin/pharmacology , Cisplatin/chemistry , Humans , Drug Resistance, Neoplasm/drug effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Animals , Neoplasms/drug therapy , Nanoparticles/chemistry , Drug Carriers/chemistry , Drug Delivery Systems
11.
Biochem Biophys Res Commun ; 712-713: 149936, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38640736

ABSTRACT

As cisplatin is one of the most broadly used chemotherapeutics, it is widely tested in vitro & in vivo assays, involving attempts to better understand its mechanism of action, develop strategies to mitigate its toxicity, or develop new drug combinations. Presently, for in vitro assays, dissolving cisplatin in dimethyl sulfoxide (DMSO) is discouraged due to its significant reduction in drug activity, Alternatively, inorganic solvents like normal saline (NS) are recommended. However, this approach is still problematic, including 1) instability of cisplatin in NS, 2) limited solubility, 3) the need to avoid long-term storage at -80 °C (or -20 °C) after dissolving, and 4) complications when combining with other DMSO-solubilized compounds. Here, we report a DMSO-HCl mixture as an alternative solvent to address these challenges. Cisplatin in DMSO-HCl not only retains comparable drug activity to cisplatin in NS but also exhibits increased stability over an extended period. Our brief report sheds light on cisplatin action, providing insights to aid in cancer research in vitro.


Subject(s)
Antineoplastic Agents , Cisplatin , Dimethyl Sulfoxide , Solvents , Cisplatin/pharmacology , Cisplatin/chemistry , Solvents/chemistry , Dimethyl Sulfoxide/pharmacology , Dimethyl Sulfoxide/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Humans , Solubility , Drug Stability , Cell Line, Tumor , Hydrogen-Ion Concentration
12.
J Inorg Biochem ; 254: 112518, 2024 05.
Article in English | MEDLINE | ID: mdl-38460483

ABSTRACT

Cisplatin is widely used as anticancer drugs, and DNA is considered as the main target. Considering its high affinity towards cysteines and the important role of cystine containing proteins, we applied a competitive activity-based protein profiling strategy to identify protein cysteines that bind with cisplatin in HeLa cells. Living cells were treated with cisplatin at cytotoxic concentrations, then the protein was extracted. After labeling with desthiobiotin iodoacetamide (DBIA) probe, protein was precipitated, digested and isotopically labeled, subsequently the peptides were combined, and the biotinylated cysteine-containing peptides were enriched and quantified by LC-MS/MS. A total of 3571 peptides which originated from 1871 proteins were identified using the DBIA probe. Among them, 46 proteins were screened as targets, including proteins that have been identified as binding proteins by previous study. A novel cisplatin target, calpain-1 (CAPN1), was identified and validated as binding with cisplatin in vitro.


Subject(s)
Antineoplastic Agents , Cisplatin , Humans , Cisplatin/pharmacology , Cisplatin/chemistry , Chromatography, Liquid , HeLa Cells , Tandem Mass Spectrometry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Proteins , Cysteine/chemistry , Peptides
13.
Molecules ; 29(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38474631

ABSTRACT

A wide range of platinum(0)-η2-(E)-1,2-ditosylethene complexes bearing isocyanide, phosphine and N-heterocyclic carbene ancillary ligands have been prepared with high yields and selectivity. All the novel products underwent thorough characterization using spectroscopic techniques, including NMR and FT-IR analyses. Additionally, for some compounds, the solid-state structures were elucidated through X-ray diffractometry. The synthesized complexes were successively evaluated for their potential as anticancer agents against two ovarian cancer cell lines (A2780 and A2780cis) and one breast cancer cell line (MDA-MB-231). The majority of the compounds displayed promising cytotoxicity within the micromolar range against A2780 and MDA-MB-231 cells, with IC50 values comparable to or even surpassing those of cisplatin. However, only a subset of compounds was cytotoxic against cisplatin-resistant cancer cells (A2780cis). Furthermore, the assessment of antiproliferative activity on MRC-5 normal cells revealed certain compounds to exhibit in vitro selectivity. Notably, complexes 3d, 6a and 6b showed low cytotoxicity towards normal cells (IC50 > 100 µM) while concurrently displaying potent cytotoxicity against cancer cells.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Coordination Complexes , Methane/analogs & derivatives , Ovarian Neoplasms , Phosphines , Female , Humans , Cisplatin/chemistry , Platinum/chemistry , Cell Line, Tumor , Cyanides , Spectroscopy, Fourier Transform Infrared , Coordination Complexes/chemistry , Antineoplastic Agents/chemistry , Ligands
14.
J Mater Chem B ; 12(20): 4843-4853, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38444277

ABSTRACT

Metallic nanomaterials have gained significant attention in cancer therapy as potential nanocarriers due to their unique properties at the nanoscale. However, nanomaterials face several drawbacks, including biocompatibility, stability, and cellular uptake. Hematite (α-Fe2O3) nanoparticles are emerging as promising nano-carriers to reduce adverse outcomes of conventional chemotherapeutics. However, the shape-mediated drug carrier mechanics of hematite nanomaterials are not raveled. In this study, we tailored hematite nanoparticles in ellipsoidal (EHNP) and spherical (SHNP) shapes with excellent biocompatibility and efficient drug encapsulation and release. We elucidate that EHNP exhibits higher cellular uptake than SHNP. With effective cellular internalization, the cisplatin-loaded EHNP showed excellent cytotoxicity with an IC50 value of 200 nM compared to the cisplatin-loaded SHNP. The flow cytometry cell sorting (FACS) analysis showed a four-fold increase in cell death by arresting the cells at the G0/G1 and G1 phases for cis-EHNP compared to cis-SHNP. The results show that ellipsoidal-shaped hematite nanoparticles can act as attractive nanocarriers with improved therapeutic efficacy in cancer therapy.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Cisplatin , Drug Carriers , Ferric Compounds , Humans , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Drug Carriers/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Cisplatin/chemistry , Female , Particle Size , Drug Screening Assays, Antitumor , Cell Survival/drug effects , Cell Proliferation/drug effects , MCF-7 Cells
15.
Adv Mater ; 36(21): e2308504, 2024 May.
Article in English | MEDLINE | ID: mdl-38546279

ABSTRACT

Anexelekto (AXL) is an attractive molecular target for ovarian cancer therapy because of its important role in ovarian cancer initiation and progression. To date, several AXL inhibitors have entered clinical trials for the treatment of ovarian cancer. However, the disadvantages of low AXL affinity and severe off-target toxicity of these inhibitors limit their further clinical applications. Herein, by rational design of a nonapeptide derivative Nap-Phe-Phe-Glu-Ile-Arg-Leu-Arg-Phe-Lys (Nap-IR), a strategy of in situ nanofiber formation is proposed to suppress ovarian cancer growth. After administration, Nap-IR specifically targets overexpressed AXL on ovarian cancer cell membranes and undergoes a receptor-instructed nanoparticle-to-nanofiber transition. In vivo and in vitro experiments demonstrate that in situ formed Nap-IR nanofibers efficiently induce apoptosis of ovarian cancer cells by blocking AXL activation and disrupting subsequent downstream signaling events. Remarkably, Nap-IR can synergistically enhance the anticancer effect of cisplatin against HO8910 ovarian tumors. It is anticipated that the Nap-IR can be applied in clinical ovarian cancer therapy in the near future.


Subject(s)
Axl Receptor Tyrosine Kinase , Intercellular Signaling Peptides and Proteins , Nanofibers , Ovarian Neoplasms , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Humans , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Nanofibers/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Cell Line, Tumor , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Oligopeptides/chemistry , Oligopeptides/pharmacology , Mice , Protein Binding , Cisplatin/pharmacology , Cisplatin/chemistry
16.
Article in English | MEDLINE | ID: mdl-38330770

ABSTRACT

Cisplatin is a potent cytotoxic agent used in the treatment of various malignancies and exerts its antitumor effect through malignant cell DNA damage and apoptosis induction. Evaluation of systemic delivery of cisplatin is important in optimization of cisplatin treatment. However, accurate quantification of systemic cisplatin is challenging due to its various forms in circulation. This study aimed to develop a sensitive (LOQ < 0.1 µg/mL) and precise Ultra Performance Liquid Chromatography (UPLC) - Tandem Mass Spectrometry (MS/MS) method for quantifying free cisplatin in microdialysates and plasma. Furthermore the aim was to compare free cisplatin concentrations measured in standard plasma samples with those obtained from intravenous microdialysis catheters in a porcine model. The method developed utilizes dichloro(ethylenediamine)platinum(II) as an internal standard that co-elutes with cisplatin, ensuring precise correction for ion suppression/enhancement effects. The method was validated, demonstrating linearity up to 100 µg/mL and good intermediate precision (CV% < 6 %) in the range of 1.0-100 µg/mL, with an LOQ of 0.03 µg/mL. The pharmacokinetic parameters (AUC0-last, Cmax, T1/2, and Tmax) showed no significant differences between the two sampling methods. This validated LC-MS/MS method provides a reliable tool for quantifying systemic free cisplatin concentrations, facilitating future systemic and local pharmacokinetic evaluations for optimization of cisplatin-based cancer treatments.


Subject(s)
Cisplatin , Tandem Mass Spectrometry , Animals , Swine , Chromatography, Liquid/methods , Cisplatin/analysis , Cisplatin/chemistry , Tandem Mass Spectrometry/methods , Plasma/chemistry , Liquid Chromatography-Mass Spectrometry , Reproducibility of Results , Chromatography, High Pressure Liquid/methods
17.
J Inorg Biochem ; 254: 112505, 2024 05.
Article in English | MEDLINE | ID: mdl-38377623

ABSTRACT

Platinum-based anticancer drugs, while potent, are associated with numerous and severe side effects. Hyperthermia therapy is an effective adjuvant in anticancer treatment, however, clinically used platinum drugs have not been optimised for combination with hyperthermia. The derivatisation of existing anticancer drugs with appropriately chosen thermoresponsive moieties results in drugs being activated only at the heated site. Perfluorinated chains of varying lengths were installed on carboplatin, a clinically approved drug, leading to the successful synthesis of a series of mono- and di- substituted platinum(IV) carboplatin prodrugs. Some of these complexes display relevant thermosensitivity on ovarian cancer cell lines, i.e., being inactive at 37 °C while having comparable activity to carboplatin under mild hyperthermia (42 °C). Nuclear magnetic resonance spectroscopy and mass spectrometry indicated that carboplatin is likely the active platinum(II) anticancer agent upon reduction and cyclic voltammetry revealed that the length of the fluorinated alkyl chain has a strong influence on the rate of carboplatin formation, regulating the subsequent cytotoxicity.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Prodrugs , Female , Humans , Carboplatin/pharmacology , Carboplatin/chemistry , Prodrugs/chemistry , Antineoplastic Agents/chemistry , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Platinum/chemistry , Cisplatin/chemistry
18.
Adv Mater ; 36(19): e2312488, 2024 May.
Article in English | MEDLINE | ID: mdl-38301714

ABSTRACT

Drug resistance significantly hampers the clinical application of existing platinum-based anticancer drugs. New platinum medications that possess distinct mechanisms of action are highly desired for the treatment of Pt-resistant cancers. Herein, a nanoscale trans-platinum(II)-based supramolecular coordination self-assembly (Pt-TCPP-BA) is prepared via using trans-[PtCl2(pyridine)(NH3)] (transpyroplatin), tetracarboxylporphyrin (TCPP), and benzoic acid (BA) as building blocks to combat drug resistance in platinum-based chemotherapy. Mechanistic studies indicate that Pt-TCPP-BA shows a hydrogen-peroxide-responsive dissociation behavior along with the generation of bioactive trans-Pt(II) and TCPP-Pt species. Different from cisplatin, these degradation products interact with DNA via interstrand cross-links and small groove binding, and induce significant upregulation of cell-death-related proteins such as p53, cleaved caspase 3, p21, and phosphorylated H2A histone family member X in cisplatin-resistant cancer cells. As a result, Pt-TCPP-BA exhibits potent killing effects against Pt-resistant tumors both in vitro and in vivo. Overall, this work not only provides a new platinum drug for combating drug-resistant cancer but also offers a new paradigm for the development of platinum-based supramolecular anticancer drugs.


Subject(s)
Antineoplastic Agents , Platinum , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Humans , Cell Line, Tumor , Animals , Platinum/chemistry , Platinum/pharmacology , Drug Resistance, Neoplasm/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Mice , Organoplatinum Compounds/chemistry , Organoplatinum Compounds/pharmacology , Cisplatin/pharmacology , Cisplatin/chemistry , DNA/chemistry , DNA/metabolism , Apoptosis/drug effects
19.
Dalton Trans ; 53(8): 3476-3483, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38270175

ABSTRACT

The reaction of Pt-based anticancer agents with arsenic trioxide affords robust complexes known as arsenoplatins. The prototype of this family of anticancer compounds is arsenoplatin-1 (AP-1) that contains an As(OH)2 fragment linked to a Pt(II) moiety derived from cisplatin. Crystallographic and spectrometric studies of AP-1 binding to a B-DNA double helix dodecamer are presented here, in comparison with cisplatin and transplatin. Results reveal that AP-1, cisplatin and transplatin react differently with the DNA model system. Notably, in the AP-1/DNA systems, the Pt-As bond can break down with time and As-containing fragments can be released. These results have implications for the understanding of the mechanism of action of arsenoplatins.


Subject(s)
Antineoplastic Agents , Arsenic Trioxide/analogs & derivatives , DNA, B-Form , Cisplatin/chemistry , Transcription Factor AP-1/metabolism , Antineoplastic Agents/chemistry , DNA/chemistry
20.
Adv Healthc Mater ; 13(11): e2303667, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38178648

ABSTRACT

Currently, cisplatin resistance has been recognized as a multistep cascade process for its clinical chemotherapy failure. Hitherto, it remains challenging to develop a feasible and promising strategy to overcome the cascade drug resistance (CDR) issue for achieving fundamentally improved chemotherapeutic efficacy. Herein, a novel self-assembled nanoagent is proposed, which is constructed by Pt(IV) prodrug, cyanine dye (cypate), and gadolinium ion (Gd3+), for systematically conquering the cisplatin resistance by employing near-infrared (NIR) light activated mild-temperature hyperthermia in tumor targets. The proposed nanoagents exhibit high photostability, GSH/H+-responsive dissociation, preferable photothermal conversion, and enhanced cellular uptake performance. In particular, upon 785-nm NIR light irradiation, the generated mild temperature of ≈ 43 °C overtly improves the cell membrane permeability and drug uptake, accelerates the disruption of intracellular redox balance, and apparently enhances the formation of Pt-DNA adducts, thereby effectively overcoming the CDR issue and achieves highly improved therapeutic efficacy for cisplatin-resistant tumor ablation.


Subject(s)
Cisplatin , Drug Resistance, Neoplasm , Hyperthermia, Induced , Indoles , Propionates , Cisplatin/pharmacology , Cisplatin/chemistry , Drug Resistance, Neoplasm/drug effects , Humans , Animals , Hyperthermia, Induced/methods , Mice , Cell Line, Tumor , Infrared Rays , Gadolinium/chemistry , Gadolinium/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Prodrugs/chemistry , Prodrugs/pharmacology , Mice, Inbred BALB C , Neoplasms/therapy , Neoplasms/drug therapy , Neoplasms/pathology , Mice, Nude , Carbocyanines/chemistry , Carbocyanines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...