Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.201
Filter
1.
Mikrochim Acta ; 191(6): 330, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38744738

ABSTRACT

In view of a large number of people infected with Helicobacter pylori (H. pylori) with great harm followed, there is an urgent need to develop a non-invasive, easy-to-operate, and rapid detection method, and to identify effective sterilization strategies. In this study, highly specific nanoprobes with nanozyme activity, Ag@Pt nanoparticles (NPs) with the antibody, were utilized as a novel lateral flow immunoassay (LFIA). The optical label (Ag@Pt NPs) was enhanced by the introduction of the chromogenic substrate 3,3',5,5'-tetramethylbenzidine (TMB) and compared with a gold nanoparticles (Au NPs) optical label. Under the optimal condition, Ag@Pt-LFIA and TMB-enhanced Ag@Pt-LFIA for H. pylori were successfully established, two of which were over twofold and 100-fold more sensitive than conventional visual Au NP-based LFIA, respectively. Furthermore, Ag@Pt NPs with the antibody irradiated with NIR laser (808 nm) at a power intensity of 550 mW/cm2 for 5 min exhibited a remarkable antibacterial effect. The nanoprobes could close to bacteria through effective interactions between antibodies and bacteria, thereby benefiting photothermal sterilization. Overall, Ag@Pt NPs provide promising applications in pathogen detection and therapeutic applications.


Subject(s)
Alloys , Helicobacter pylori , Metal Nanoparticles , Platinum , Silver , Helicobacter pylori/radiation effects , Helicobacter pylori/drug effects , Silver/chemistry , Metal Nanoparticles/chemistry , Platinum/chemistry , Alloys/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Immunoassay/methods , Benzidines/chemistry , Gold/chemistry , Humans , Sterilization/methods , Limit of Detection
2.
PLoS One ; 19(5): e0301358, 2024.
Article in English | MEDLINE | ID: mdl-38771804

ABSTRACT

Drug-resistant bacteria arising from antibiotic abuse infections have always been a serious threat to human health. Killing bacteria with toxic reactive oxygen species (ROS) is an ideal antibacterial method for treating drug-resistant bacterial infections. Here, we prepared Pt-Ru bimetallic nanoclusters (Pt-Ru NCs) with higher peroxidase (POD)-like activity than Pt monometallic nanoclusters. Pt-Ru can easily catalyze the decomposition of H2O2 to produce ·OH, thereby catalyzing the transformation of 3,3',5,5'-tetramethylbiphenylamine (TMB) to blue oxidized TMB (oxTMB). We utilized the POD-like activity of the Pt-Ru NCs for antibacterial therapy. The results showed that at doses of 40 µg/mL and 16 µg/mL, the Pt-Ru NCs exhibited extraordinary antibacterial activity against E. coli and S. aureus, demonstrating the enormous potential of Pt-Ru NCs as antibacterial agents.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Metal Nanoparticles , Platinum , Ruthenium , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Platinum/chemistry , Platinum/pharmacology , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Ruthenium/chemistry , Ruthenium/pharmacology , Metal Nanoparticles/chemistry , Microbial Sensitivity Tests , Peroxidase/metabolism , Hydrogen Peroxide/chemistry , Catalysis , Humans
3.
Anal Chim Acta ; 1309: 342698, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38772661

ABSTRACT

BACKGROUND: The lateral flow immunoassay (LFIA) is widely employed as a point-of-care testing (POCT) technique. However, its limited sensitivity hinders its application in detecting biomarkers with low abundance. Recently, the utilization of nanozymes has been implemented to enhance the sensitivity of LFIA by catalyzing the oxidation of 3,3',5,5'-tetramethylbenzidine (TMB). The catalytic performance of nanozymes plays a crucial role in influencing the sensitivity of LFIA. RESULTS: The Cornus officinalis Sieb. et Zucc-Pd@Pt (CO-Pd@Pt) nanozyme with good peroxidase-like activity was synthesized herein through a facile one-pot method employing Cornus officinalis Sieb. et Zucc extract as a reducing agent. The morphology and composition of the CO-Pd@Pt nanozyme were characterized using TEM, SEM, XRD, and XPS. As a proof of concept, the as-synthesized CO-Pd@Pt nanozyme was utilized in LFIA (CO-Pd@Pt-LFIA) for the detection of human chorionic gonadotropin (hCG). Compared to conventional gold nanoparticles-based LFIA (AuNPs-LFIA), CO-Pd@Pt-LFIA demonstrated a significant enhancement in the limit of detection (LOD, 0.08 mIU/mL), which is approximately 160 times lower than that of AuNPs-LFIA. Furthermore, experiments evaluating accuracy, precision, selectivity, interference, and stability have confirmed the practical applicability of CO-Pd@Pt-LFIA for hCG content determination. SIGNIFICANCE: The present study presents a novel approach for the synthesis of bimetallic nanozymes through environmentally friendly methods, utilizing plant extracts as both protective and reducing agents. Additionally, an easily implementable technique is proposed to enhance signal detection in lateral flow immunoassays.


Subject(s)
Palladium , Platinum , Palladium/chemistry , Platinum/chemistry , Immunoassay/methods , Humans , Metal Nanoparticles/chemistry , Limit of Detection , Peroxidase/chemistry , Peroxidase/metabolism , Benzidines/chemistry , Catalysis , Oxidation-Reduction
4.
Biosens Bioelectron ; 258: 116354, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38723331

ABSTRACT

Real-time monitoring of biological markers in sweat is a valuable tool for health assessment. In this study, we have developed an innovative wearable biosensor for precise analysis of glucose in sweat during physical activities. The sensor is based on a single-atom catalyst of platinum (Pt) uniformly dispersed on tricobalt tetroxide (Co3O4) nanorods and reduced graphene oxide (rGO), featuring a unique three-dimensional nanostructure and excellent glucose electrocatalytic performance with a wide detection range of 1-800 µM. Additionally, density functional theory calculations have revealed the synergetic role of Pt active sites in the Pt single-atom catalyst (Co3O4/rGO/Pt) in glucose adsorption and electron transfer, thereby enhancing sensor performance. To enable application in wearable devices, we designed an S-shaped microfluidic chip and a point-of-care testing (POCT) device, both of which were validated for effectiveness through actual use by volunteers. This research provides valuable insights and innovative approaches for analyzing sweat glucose using wearable devices, contributing to the advancement of personalized healthcare.


Subject(s)
Biosensing Techniques , Glucose , Graphite , Platinum , Sweat , Wearable Electronic Devices , Biosensing Techniques/instrumentation , Sweat/chemistry , Platinum/chemistry , Humans , Catalysis , Glucose/analysis , Graphite/chemistry , Electrochemical Techniques/instrumentation , Nanotubes/chemistry , Limit of Detection , Equipment Design , Oxides/chemistry
5.
Technol Cancer Res Treat ; 23: 15330338241249692, 2024.
Article in English | MEDLINE | ID: mdl-38706262

ABSTRACT

PURPOSE: PIWI-interacting RNAs (piRNAs) are a type of noncoding small RNA that can interact with PIWI-like RNA-mediated gene silencing (PIWIL) proteins to affect biological processes such as transposon silencing through epigenetic effects. Recent studies have found that piRNAs are widely dysregulated in tumors and associated with tumor progression and a poor prognosis. Therefore, this study aimed to investigate the effect of piR-1919609 on the proliferation, apoptosis, and drug resistance of ovarian cancer cells. METHODS: In this study, we used small RNA sequencing to screen and identify differentially expressed piRNAs in primary ovarian cancer, recurrent ovarian cancer, and normal ovaries. A large-scale verification study was performed to verify the expression of piR-1919609 in different types of ovarian tissue, including ovarian cancer tissue and normal ovaries, by RT-PCR and to analyze its association with the clinical prognosis of ovarian cancer. The expression of PIWILs in ovarian cancer was verified by RT-PCR, Western blotting and immunofluorescence. The effects of piR-1919609 on ovarian cancer cell proliferation, apoptosis and drug resistance were studied through in vitro and in vivo models. RESULTS: (1) piR-1919609 was highly expressed in platinum-resistant ovarian cancer tissues (p < 0.05), and this upregulation was significantly associated with a poor prognosis and a shorter recurrence time in ovarian cancer patients (p < 0.05). (2) PIWIL2 was strongly expressed in ovarian cancer tissues (p < 0.05). It was expressed both in the cytoplasm and nucleus of ovarian cancer cells. (3) Overexpression of piR-1919609 promoted ovarian cancer cell proliferation, inhibited apoptosis, and promoted tumor growth in nude mice. (4) Inhibition of piR-1919609 effectively reversed ovarian cancer drug resistance. CONCLUSION: In summary, we showed that piR-1919609 is involved in the regulation of drug resistance in ovarian cancer cells and might be an ideal potential target for reversing platinum resistance in ovarian cancer.


Subject(s)
Apoptosis , Cell Proliferation , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms , RNA, Small Interfering , Xenograft Model Antitumor Assays , Female , Humans , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Animals , Mice , Cell Line, Tumor , RNA, Small Interfering/genetics , Prognosis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Disease Models, Animal , Platinum/therapeutic use , Platinum/pharmacology
6.
Sci Rep ; 14(1): 11025, 2024 05 14.
Article in English | MEDLINE | ID: mdl-38744861

ABSTRACT

Platinum-resistant phenomena in ovarian cancer is very dangerous for women suffering from this disease, because reduces the chances of complete recovery. Unfortunately, until now there are no methods to verify whether a woman with ovarian cancer is platinum-resistant. Importantly, histopathology images also were not shown differences in the ovarian cancer between platinum-resistant and platinum-sensitive tissues. Therefore, in this study, Fourier Transform InfraRed (FTIR) and FT-Raman spectroscopy techniques were used to find chemical differences between platinum-resistant and platinum-sensitive ovarian cancer tissues. Furthermore, Principal Component Analysis (PCA) and machine learning methods were performed to show if it possible to differentiate these two kind of tissues as well as to propose spectroscopy marker of platinum-resistant. Indeed, obtained results showed, that in platinum-resistant ovarian cancer tissues higher amount of phospholipids, proteins and lipids were visible, however when the ratio between intensities of peaks at 1637 cm-1 (FTIR) and at 2944 cm-1 (Raman) and every peaks in spectra was calculated, difference between groups of samples were not noticed. Moreover, structural changes visible as a shift of peaks were noticed for C-O-C, C-H bending and amide II bonds. PCA clearly showed, that PC1 can be used to differentiate platinum-resistant and platinum-sensitive ovarian cancer tissues, while two-trace two-dimensional correlation spectra (2T2D-COS) showed, that only in amide II, amide I and asymmetric CH lipids vibrations correlation between two analyzed types of tissues were noticed. Finally, machine learning algorithms showed, that values of accuracy, sensitivity and specificity were near to 100% for FTIR and around 95% for FT-Raman spectroscopy. Using decision tree peaks at 1777 cm-1, 2974 cm-1 (FTIR) and 1714 cm-1, 2817 cm-1 (FT-Raman) were proposed as spectroscopy marker of platinum-resistant.


Subject(s)
Drug Resistance, Neoplasm , Ovarian Neoplasms , Principal Component Analysis , Spectrum Analysis, Raman , Female , Humans , Spectrum Analysis, Raman/methods , Spectroscopy, Fourier Transform Infrared/methods , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Middle Aged , Platinum , Biomarkers, Tumor , Machine Learning , Aged
7.
Cell Death Dis ; 15(5): 329, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740757

ABSTRACT

Iron is crucial for cell DNA synthesis and repair, but an excess of free iron can lead to oxidative stress and subsequent cell death. Although several studies suggest that cancer cells display characteristics of 'Iron addiction', an ongoing debate surrounds the question of whether iron can influence the malignant properties of ovarian cancer. In the current study, we initially found iron levels increase during spheroid formation. Furthermore, iron supplementation can promote cancer cell survival, cancer spheroid growth, and migration; vice versa, iron chelators inhibit this process. Notably, iron reduces the sensitivity of ovarian cancer cells to platinum as well. Mechanistically, iron downregulates DNA homologous recombination (HR) inhibitor polymerase theta (POLQ) and relieves its antagonism against the HR repair enzyme RAD51, thereby promoting DNA damage repair to resist chemotherapy-induced damage. Additionally, iron tightly regulated by ferritin (FTH1/FTL) which is indispensable for iron-triggered DNA repair. Finally, we discovered that iron chelators combined with platinum exhibit a synergistic inhibitory effect on ovarian cancer in vitro and in vivo. Our findings affirm the pro-cancer role of iron in ovarian cancer and reveal that iron advances platinum resistance by promoting DNA damage repair through FTH1/FTL/POLQ/RAD51 pathway. Our findings highlight the significance of iron depletion therapy, revealing a promising avenue for advancing ovarian cancer treatment.


Subject(s)
DNA Repair , Drug Resistance, Neoplasm , Iron , Ovarian Neoplasms , Rad51 Recombinase , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Drug Resistance, Neoplasm/drug effects , DNA Repair/drug effects , Iron/metabolism , Cell Line, Tumor , Rad51 Recombinase/metabolism , Animals , Ferritins/metabolism , Mice , Platinum/pharmacology , Platinum/therapeutic use , Mice, Nude , Oxidoreductases/metabolism
8.
Nat Commun ; 15(1): 4253, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762636

ABSTRACT

Platinum-based chemotherapy is the cornerstone treatment for female high-grade serous ovarian carcinoma (HGSOC), but choosing an appropriate treatment for patients hinges on their responsiveness to it. Currently, no available biomarkers can promptly predict responses to platinum-based treatment. Therefore, we developed the Pathologic Risk Classifier for HGSOC (PathoRiCH), a histopathologic image-based classifier. PathoRiCH was trained on an in-house cohort (n = 394) and validated on two independent external cohorts (n = 284 and n = 136). The PathoRiCH-predicted favorable and poor response groups show significantly different platinum-free intervals in all three cohorts. Combining PathoRiCH with molecular biomarkers provides an even more powerful tool for the risk stratification of patients. The decisions of PathoRiCH are explained through visualization and a transcriptomic analysis, which bolster the reliability of our model's decisions. PathoRiCH exhibits better predictive performance than current molecular biomarkers. PathoRiCH will provide a solid foundation for developing an innovative tool to transform the current diagnostic pipeline for HGSOC.


Subject(s)
Cystadenocarcinoma, Serous , Deep Learning , Ovarian Neoplasms , Platinum , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/genetics , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/diagnostic imaging , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/genetics , Platinum/therapeutic use , Middle Aged , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Treatment Outcome , Neoplasm Grading , Cohort Studies , Adult , Reproducibility of Results
9.
J Biomed Sci ; 31(1): 50, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741159

ABSTRACT

BACKGROUND: G-quadruplex DNA (G4) is a non-canonical structure forming in guanine-rich regions, which play a vital role in cancer biology and are now being acknowledged in both nuclear and mitochondrial (mt) genome. However, the impact of G4-based targeted therapy on both nuclear and mt genome, affecting mt function and its underlying mechanisms remain largely unexplored. METHODS: The mechanisms of action and therapeutic effects of a G4-binding platinum(II) complex, Pt-ttpy, on mitochondria were conducted through a comprehensive approaches with in vitro and in vivo models, including ICP-MS for platinum measurement, PCR-based genetic analysis, western blotting (WB), confocal microscope for mt morphology study, extracellular flux analyzer, JC1 and Annexin V apoptosis assay, flow cytometry and high content microscope screening with single-cell quantification of both ROS and mt specific ROS, as well as click-chemistry for IF study of mt translation. Decipher Pt-ttpy effects on nuclear-encoded mt related genes expression were undertaken via RNA-seq, Chip-seq and CUT-RUN assays. RESULTS: Pt-ttpy, shows a highest accumulation in the mitochondria of A2780 cancer cells as compared with two other platinum(II) complexes with no/weak G4-binding properties, Pt-tpy and cisplatin. Pt-ttpy induces mtDNA deletion, copy reduction and transcription inhibition, hindering mt protein translation. Functional analysis reveals potent mt dysfunction without reactive oxygen species (ROS) induction. Mechanistic study provided first evidence that most of mt ribosome genes are highly enriched in G4 structures in their promoter regions, notably, Pt-ttpy impairs most nuclear-encoded mt ribosome genes' transcription through dampening the recruiting of transcription initiation and elongation factors of NELFB and TAF1 to their promoter with G4-enriched sequences. In vivo studies show Pt-ttpy's efficient anti-tumor effects, disrupting mt genome function with fewer side effects than cisplatin. CONCLUSION: This study underscores Pt-ttpy as a G4-binding platinum(II) complex, effectively targeting cancer mitochondria through dual action on mt and nuclear G4-enriched genomes without inducing ROS, offering promise for safer and effective platinum-based G4-targeted cancer therapy.


Subject(s)
G-Quadruplexes , Mitochondria , G-Quadruplexes/drug effects , Humans , Mitochondria/metabolism , Mitochondria/drug effects , Cell Line, Tumor , Genome, Mitochondrial , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Platinum/pharmacology , Animals
10.
J Ovarian Res ; 17(1): 70, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561819

ABSTRACT

OBJECTIVES: This retrospective study aims to evaluating the subsequent management and outcomes after first-line PARPi progression in Chinese ovarian cancer population. METHODS: Clinical and pathologic variables, treatment modalities, and outcomes were assessed. We investigated the subsequent management and outcomes after first-line PARPi progression. The objective response rate (ORR) and disease control rate (DCR) parameters were evaluated to determine the response to subsequent chemotherapy. For the survival analyses, progression-free survival 1 (PFS1), PFS2, overall survival (OS) and PFS2 - PFS1 were analysed. RESULTS: A total of 124 patients received PARPi maintenance treatment after first-line chemotherapy during the study period in our center. 44 of them (35.5%) experienced a recurrence. The median duration of PARPi in these patients was 11.1 months (range: 1.2-75.1 months). A total of 40 patients (40/44, 90.9%) received subsequent chemotherapy with 35 (35/44, 79.5%) and 5 (5/44, 11.4%) patients received platinum-based and non-platinum-based chemotherapy in our center. 2 patients (4.5%) received target therapy and other 2 patients (4.5%) received best supportive care. 27.3% (12/44) patients received secondary cytoreduction surgery (SCS). After subsequent chemotherapy, 14 patients received PARPi retreatment as maintenance therapy. In patients who received platinum-based regimens (n = 35), 23 of 35 patients (65.7%) had complete/partial response (CR/PR), 8 of 35 (22.9%) had stable disease (SD), and 4 of 35 (12.1%) had progressive disease (PD). The ORR and DCR of patients who received subsequent chemotherapy was 65.7% and 88.6%, respectively. 15 patients (57.7%, 15/26) were reported to be platinum resistant with a platinum-free interval (PFI) of < 6 months in patients whose platinum sensitivity of the second line platinum-based regimens was evaluable. Patients who received SCS after PARPi resistant associated with a borderline better PFS2 (median PFS2: 41.9 vs. 29.2 months, P = 0.051) and a non-significantly increased PFS2-PFS1 (median PFS2-PFS1: 12.2 vs. 9.8 months, P = 0.551). Patients with a PFI ≥ 12 months had a significantly better PFS2 (median PFS2: 37.0 vs. 25.3 months, P < 0.001) and a tendency towards a better PFS2-PFS1 than those with a PFI < 12 months (median PFS2-PFS1: 11.2 vs. 8.5 months, P = 0.334). A better PFS2 was observed in patients who received second PARPi maintenance therapy (median PFS2 of 35.4 vs. 28.8 months); however, the difference was not statistically significant (P = 0.200). A better PFS2-PFS1 was observed in patients who received second PARPi maintenance therapy (median PFS2-PFS1: 13.6 vs. 8.9 months, P = 0.002) than those without. CONCLUSIONS: In summary, some degree of resistance to standard subsequent platinum and non-platinum chemotherapy is noted in the entire cohort. A trend towards higher benefit from subsequent chemotherapy after first-line PARP inhibitors progression was observed in the PFI ≥ 12 months subgroup than those with PFI < 12 months. PARPi retreatment as maintenance therapy and SCS can be offered to some patients with PARPi resistance.


Subject(s)
Ovarian Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Female , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Retrospective Studies , Ovarian Neoplasms/pathology , Progression-Free Survival , Survival Analysis , Platinum/pharmacology , Platinum/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local/drug therapy
11.
Int J Mol Sci ; 25(7)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38612653

ABSTRACT

To understand chemoresistance in the context of cancer stem cells (CSC), a cisplatin resistance model was developed using a high-grade serous ovarian cancer patient-derived, cisplatin-sensitive sample, PDX4. As a molecular subtype-specific stem-like cell line, PDX4 was selected for its representative features, including its histopathological and BRCA2 mutation status, and exposed to cisplatin in vitro. In the cisplatin-resistant cells, transcriptomics were carried out, and cell morphology, protein expression, and functional status were characterized. Additionally, potential signaling pathways involved in cisplatin resistance were explored. Our findings reveal the presence of distinct molecular signatures and phenotypic changes in cisplatin-resistant PDX4 compared to their sensitive counterparts. Surprisingly, we observed that chemoresistance was not inherently linked with increased stemness. In fact, although resistant cells expressed a combination of EMT and stemness markers, functional assays revealed that they were less proliferative, migratory, and clonogenic-features indicative of an underlying complex mechanism for cell survival. Furthermore, DNA damage tolerance and cellular stress management pathways were enriched. This novel, syngeneic model provides a valuable platform for investigating the underlying mechanisms of cisplatin resistance in a clinically relevant context, contributing to the development of targeted therapies tailored to combat resistance in stem-like ovarian cancer.


Subject(s)
Ovarian Neoplasms , Platinum , Humans , Female , Platinum/pharmacology , Cisplatin/pharmacology , Cisplatin/therapeutic use , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Carcinoma, Ovarian Epithelial
12.
Front Immunol ; 15: 1354613, 2024.
Article in English | MEDLINE | ID: mdl-38617840

ABSTRACT

Metastatic colon cancer remains an incurable disease, and it is difficult for existing treatments to achieve the desired clinical outcome, especially for colon cancer patients who have received first-line treatment. Although immune checkpoint inhibitors (ICIs) have demonstrated durable clinical efficacy in a variety of solid tumors, their response requires an inflammatory tumor microenvironment. However, microsatellite-stable (MSS) colon cancer, which accounts for the majority of colorectal cancers, is a cold tumor that does not respond well to ICIs. Combination regimens open the door to the utility of ICIs in cold tumors. Although combination therapies have shown their advantage even for MSS colon cancer, it remains unclear whether combination therapies show their advantage in patients with pretreated metastatic colon cancer. We report a patient who has achieved complete remission and good tolerance with sintilimab plus bevacizumab and platinum-based chemotherapy after postoperative recurrence. The patient had KRAS mutation and MSS-type colon cancer, and his PD-1+CD8+ and CD3-CD19-CD14+CD16-HLA-DR were both positive. He has achieved a progression-free survival of 43 months and is still being followed up at our center. The above results suggest that this therapeutic regimen is a promising treatment modality for the management of pretreated, MSS-type and KRAS-mutated metastatic colorectal cancer although its application to the general public still needs to be validated in clinical trials.


Subject(s)
Antibodies, Monoclonal, Humanized , Colonic Neoplasms , Proto-Oncogene Proteins p21(ras) , Male , Humans , Bevacizumab/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , Pathologic Complete Response , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Platinum , Microsatellite Repeats , Tumor Microenvironment
13.
J Cancer Res Clin Oncol ; 150(4): 195, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38625410

ABSTRACT

PURPOSE: The objective of the study was to assess the effectiveness and toxicity of platinum-based adjuvant chemoradiotherapy (POCRT) in comparison to postoperative radiotherapy (PORT) in patients with head and neck adenoid cystic carcinoma (HNACC). MATERIALS AND METHODS: This retrospective study analyzed patients diagnosed with HNACC at our center between January 2010 and April 2020. A 1:1 propensity score matching method was used to create a matched cohort. RESULTS: In this study, 206 patients were analyzed, with 147 patients (71.4%) receiving postoperative radiotherapy (PORT) and 59 patients (28.6%) receiving POCRT. Twenty-one patients experienced local-regional failure. The 3-, 5-, and 10-yr local-regional control (LRC) rate for the cohort were 92.0%, 90.6%, and 86.9%, respectively. In both the entire cohort and the matched cohort, the POCRT group exhibited superior LRC compared to the PORT group (Gray's test, all P < 0.05*). Multivariate analysis identified adjuvant concurrent chemotherapy as an independent prognostic factor for LRC (Competing risks regression, HR = 0.144, 95% CI 0.026-0.802, P = 0.027*). In addition, the POCRT group had higher incidences of upper gastrointestinal toxicity and hematologic toxicities, including leukopenia, neutropenia, and anemia (all P < 0.05*). CONCLUSION: In terms of reducing locoregional failures in HNACC patients, POCRT may potentially offer a more effective therapeutic approach than using PORT alone, although it also entails an augmented burden of treatment-related toxicity.


Subject(s)
Carcinoma, Adenoid Cystic , Carcinoma , Head and Neck Neoplasms , Leukopenia , Humans , Chemoradiotherapy, Adjuvant/adverse effects , Radiotherapy, Adjuvant , Carcinoma, Adenoid Cystic/therapy , Retrospective Studies , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/radiotherapy , Platinum/therapeutic use
14.
J Phys Chem B ; 128(19): 4809-4820, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38646680

ABSTRACT

We present a novel bionanocatalyst fabricated by the adsorption-reduction of metal ions on a polyurethane/S-layer protein biotemplate. The bioinspired support was obtained by the adsorption of S-layer proteins (isolated from Lentilactobacillus kefiri) on polyurethane particles. Silver and platinum nanoparticles were well-loaded on the surface of the support after the combination with metallic salts and reduction with H2 at room temperature. Transmission electron microscopy analysis revealed the strawberry-like morphology of the bionanocatalysts with a particle size, dn, of 2.39 nm for platinum and 9.60 nm for silver. Both systems catalyzed the hydrogenation of p-nitrophenol to p-aminophenol with high efficiency in water at mild conditions in the presence of NaBH4. Three different amounts of bionanocatalyst were tested, and in all cases, conversions between 97 and 99% were observed. The catalysts displayed excellent recyclability over ten cycles, and no extensive damage in their nanostructure was noted after them. The bionanocatalysts were stable during their production, storage, and use, thanks to the fact that the biosupport provides an effective driving force in the formation and stabilization of the metallic nanoparticles. The successful bioinspired production strategy and the good catalytic ability of the systems are encouraging in the search for nontoxic, simple, clean, and eco-friendly procedures for the synthesis and exploitation of nanostructures.


Subject(s)
Metal Nanoparticles , Platinum , Silver , Metal Nanoparticles/chemistry , Catalysis , Platinum/chemistry , Silver/chemistry , Oxidation-Reduction , Polyurethanes/chemistry , Nitrophenols/chemistry , Particle Size , Aminophenols/chemistry
15.
Colloids Surf B Biointerfaces ; 238: 113910, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38640797

ABSTRACT

This study represents an innovative approach to construct multi-functional nanoplatforms for cancer diagnosis and therapy by combining hyaluronic acid (HA) with iron-platinum nanoparticles (FePt NPs). These HA-coated FePt NPs, referred to as FePt@HA NPs, demonstrated remarkable biocompatibility, high absorption, and excellent light-to-heat conversion properties in the near-infrared (NIR) region, making them ideal candidates for photothermal therapy (PTT). In vitro studies revealed their effective cancer cell eradication under NIR laser irradiation, while in vivo experiments on mice showcased their superior heating capabilities. Moreover, FePt@HA NPs exhibited a distinct and strong photoacoustic (PA) signal, facilitating enhanced and precise intra-tumoral PA imaging. Our results highlight the potential of FePt@HA NPs as promising photothermal agents for future PTT applications. They offer high selectivity, precision, and minimal side effects in cancer treatment, along with their valuable PA imaging application for tumor localization and characterization.


Subject(s)
Hyaluronic Acid , Iron , Metal Nanoparticles , Photoacoustic Techniques , Photothermal Therapy , Platinum , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Photoacoustic Techniques/methods , Platinum/chemistry , Platinum/pharmacology , Animals , Mice , Iron/chemistry , Humans , Metal Nanoparticles/chemistry , Cell Survival/drug effects , Mice, Inbred BALB C , Particle Size , Surface Properties , Cell Line, Tumor
16.
J Inorg Biochem ; 256: 112573, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38678913

ABSTRACT

This paper describes the synthesis, structural analysis, as well as the magnetic and spectroscopic characterizations of three new dicopper(II) complexes with dinucleating phenol-based ligands containing different thioether donor substituents: aromatic (1), aliphatic (2) or thiophene (3). Temperature-dependent magnetometry reveals the presence of antiferromagnetic coupling for 1 and 3 (J = -2.27 cm-1 and -5.01 cm-1, respectively, H = -2JS1S2) and ferromagnetic coupling for 2 (J = 5.72 cm-1). Broken symmetry DFT calculations attribute this behavior to a major contribution from the dz2 orbitals for 1 and 3, and from the dx2-y2 orbitals for 2, along with the p orbitals of the oxygens. The bioinspired catalytic activities of these complexes related to catechol oxidase were studied using 3,5-di-tert-butylcatechol as substrate. The order of catalytic rates for the substrate oxidation follows the trend 1 > 2 > 3 with kcat of (90.79 ± 2.90) × 10-3 for 1, (64.21 ± 0.99) × 10-3 for 2 and (14.20 ± 0.32) × 10-3 s-1 for 3. The complexes also cleave DNA through an oxidative mechanism with minor-groove preference, as indicated by experimental and molecular docking assays. Antimicrobial potential of these highly active complexes has shown that 3 inhibits both Staphylococcus aureus bacterium and Epidermophyton floccosum fungus. Notably, the complexes were found to be nontoxic to normal cells but exhibited cytotoxicity against epidermoid carcinoma cells, surpassing the activity of the metallodrug cisplatin. This research shows the multifaceted properties of these complexes, making them promising candidates for various applications in catalysis, nucleic acids research, and antimicrobial activities.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Oxidation-Reduction , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Ligands , Sulfides/chemistry , Sulfides/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Platinum/chemistry , Platinum/pharmacology , Cell Line, Tumor
17.
Mikrochim Acta ; 191(5): 289, 2024 04 29.
Article in English | MEDLINE | ID: mdl-38683210

ABSTRACT

As a common chlorinated nicotinic pesticide with high insecticidal activity, acetamiprid has been widely used for pest control. However, the irrational use of acetamiprid will pollute the environment and thus affect human health. Therefore, it is crucial to develop a simple, highly sensitive, and rapid method for acetamiprid residue detection. In this study, the capture probe (Fe3O4@Pt-Aptamer) was connected with the signal probe (Au@DTNB@Ag CS-cDNA) to form an assembly with multiple SERS-enhanced effects. Combined with magnetic separation technology, a SERS sensor with high sensitivity and stability was constructed to detect acetamiprid residue. Based on the optimal conditions, the SERS intensity measured at 1333 cm-1 is in relation to the concentration of acetamiprid in the range 2.25 × 10-9-2.25 × 10-5 M, and the calculated limit of detection (LOD) was 2.87 × 10-10 M. There was no cross-reactivity with thiacloprid, clothianidin, nitenpyram, imidacloprid, and chlorpyrifos, indicating that this method has good sensitivity and specificity. Finally, the method was applied to the detection of acetamiprid in cucumber samples, and the average recoveries were 94.19-103.58%, with RSD < 2.32%. The sensor can be used to analyse real samples with fast detection speed, high sensitivity, and high selectivity.


Subject(s)
Aptamers, Nucleotide , Gold , Limit of Detection , Metal Nanoparticles , Neonicotinoids , Silver , Spectrum Analysis, Raman , Neonicotinoids/analysis , Aptamers, Nucleotide/chemistry , Gold/chemistry , Silver/chemistry , Metal Nanoparticles/chemistry , Spectrum Analysis, Raman/methods , Platinum/chemistry , Insecticides/analysis , Cucumis sativus/chemistry
18.
Cancer Immunol Immunother ; 73(6): 106, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38634928

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors have changed previous treatment paradigm of advanced urothelial carcinoma (UC). The ARON-2 study (NCT05290038) aimed to assess the real-world effectiveness of pembrolizumab in patients recurred or progressed after platinum-based chemotherapy. PATIENTS AND METHODS: Medical records of patients with documented metastatic UC treated by pembrolizumab as second-line therapy were retrospectively collected from 88 institutions in 23 countries. Patients were assessed for overall survival (OS), progression-free survival (PFS) and overall response rate (ORR). Cox proportional hazards models were adopted to explore the presence of prognostic factors. RESULTS: In total, 836 patients were included: 544 patients (65%) received pembrolizumab after progression to first-line platinum-based chemotherapy in the metastatic setting (cohort A) and 292 (35%) after recurring within < 12 months since the completion of adjuvant or neoadjuvant chemotherapy (cohort B). The median follow-up time was 15.3 months. The median OS and the ORR were 10.5 months and 31% in the overall study population, 9.1 months and 29% in cohort A and 14.6 months and 37% in cohort B. At multivariate analysis, ECOG-PS ≥ 2, bone metastases, liver metastases and pembrolizumab setting (cohort A vs B) proved to be significantly associated with worst OS and PFS. Stratified by the presence of 0, 1-2 or 3-4 prognostic factors, the median OS was 29.4, 12.5 and 4.1 months (p < 0.001), while the median PFS was 12.2, 6.4 and 2.8 months, respectively (p < 0.001). CONCLUSIONS: Our study confirms that pembrolizumab is effective in the advanced UC real-world context, showing outcome differences between patients recurred or progressed after platinum-based chemotherapy.


Subject(s)
Antibodies, Monoclonal, Humanized , Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Humans , Adjuvants, Immunologic , Platinum , Retrospective Studies
19.
Int J Mol Sci ; 25(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38673932

ABSTRACT

Platinum-containing chemotherapeutic drugs are efficacious in many forms of cancer but are dose-restricted by serious side effects, of which peripheral neuropathy induced by oxidative-nitrosative-stress-mediated chain reactions is most disturbing. Recently, hope has been raised regarding the catalytic antioxidants mangafodipir (MnDPDP) and calmangafodipir [Ca4Mn(DPDP)5; PledOx®], which by mimicking mitochondrial manganese superoxide dismutase (MnSOD) may be expected to overcome oxaliplatin-associated chemotherapy-induced peripheral neuropathy (CIPN). Unfortunately, two recent phase III studies (POLAR A and M trials) applying Ca4Mn(DPDP)5 in colorectal cancer (CRC) patients receiving multiple cycles of FOLFOX6 (5-FU + oxaliplatin) failed to demonstrate efficacy. Instead of an anticipated 50% reduction in the incidence of CIPN in patients co-treated with Ca4Mn(DPDP)5, a statistically significant increase of about 50% was seen. The current article deals with confusing differences between early and positive findings with MnDPDP in comparison to the recent findings with Ca4Mn(DPDP)5. The POLAR failure may also reveal important mechanisms behind oxaliplatin-associated CIPN itself. Thus, exacerbated neurotoxicity in patients receiving Ca4Mn(DPDP)5 may be explained by redox interactions between Pt2+ and Mn2+ and subtle oxidative-nitrosative chain reactions. In peripheral sensory nerves, Pt2+ presumably leads to oxidation of the Mn2+ from Ca4Mn(DPDP)5 as well as from Mn2+ in MnSOD and other endogenous sources. Thereafter, Mn3+ may be oxidized by peroxynitrite (ONOO-) into Mn4+, which drives site-specific nitration of tyrosine (Tyr) 34 in the MnSOD enzyme. Conformational changes of MnSOD then lead to the closure of the superoxide (O2•-) access channel. A similar metal-driven nitration of Tyr74 in cytochrome c will cause an irreversible disruption of electron transport. Altogether, these events may uncover important steps in the mechanism behind Pt2+-associated CIPN. There is little doubt that the efficacy of MnDPDP and its therapeutic improved counterpart Ca4Mn(DPDP)5 mainly depends on their MnSOD-mimetic activity when it comes to their potential use as rescue medicines during, e.g., acute myocardial infarction. However, pharmacokinetic considerations suggest that the efficacy of MnDPDP on Pt2+-associated neurotoxicity depends on another action of this drug. Electron paramagnetic resonance (EPR) studies have demonstrated that Pt2+ outcompetes Mn2+ and endogenous Zn2+ in binding to fodipir (DPDP), hence suggesting that the previously reported protective efficacy of MnDPDP against CIPN is a result of chelation and elimination of Pt2+ by DPDP, which in turn suggests that Mn2+ is unnecessary for efficacy when it comes to oxaliplatin-associated CIPN.


Subject(s)
Antineoplastic Agents , Manganese , Oxaliplatin , Peripheral Nervous System Diseases , Platinum , Humans , Antineoplastic Agents/adverse effects , Edetic Acid/analogs & derivatives , Manganese/adverse effects , Nitrosative Stress/drug effects , Oxaliplatin/adverse effects , Oxaliplatin/pharmacology , Oxidative Stress/drug effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/metabolism , Platinum/adverse effects , Pyridoxal Phosphate/analogs & derivatives , Pyridoxal Phosphate/pharmacology , Pyridoxal Phosphate/metabolism , Superoxide Dismutase/metabolism , Clinical Trials, Phase III as Topic
20.
Talanta ; 274: 125920, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38574532

ABSTRACT

Herby, the interaction of metallothioneins with commonly used Pt-based anticancer drugs - cisplatin, carboplatin, and oxaliplatin - was investigated using the combined power of elemental (i.e. LA-ICP-MS, CE-ICP-MS) and molecular (i.e. MALDI-TOF-MS) analytical techniques providing not only required information about the interaction, but also the benefit of low sample consumption. The amount of Cd and Pt incorporated within the protein was determined for protein monomers and dimer/oligomers formed by non-oxidative dimerization. Moreover, fluorescence spectrometry using Zn2+-selective fluorescent indicator - FluoZin3 - was employed to monitor the ability of Pt drugs to release natively occurring Zn from the protein molecule. The investigation was carried out using two protein isoforms (i.e. MT2, MT3), and significant differences in behaviour of these two isoforms were observed. The main attention was paid to elucidating whether the protein dimerization/oligomerization may be the reason for the potential failure of the anticancer therapy based on these drugs. Based on the results, it was demonstrated that the interaction of MT2 (both monomers and dimers) interacted with Pt drugs significantly less compared to MT3 (both monomers and dimers). Also, a significant difference between monomeric and dimeric forms (both MT2 and MT3) was not observed. This may suggest that dimer formation is not the key factor leading to the inactivation of Pt drugs.


Subject(s)
Metallothionein , Spectrometry, Fluorescence , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Metallothionein/metabolism , Metallothionein/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Spectrometry, Fluorescence/methods , Carboplatin/pharmacology , Oxaliplatin/pharmacology , Cisplatin/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/chemistry , Platinum/chemistry , Metallothionein 3 , Cytostatic Agents/pharmacology , Cytostatic Agents/chemistry , Mass Spectrometry/methods , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...