Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.355
Filter
1.
Cell Physiol Biochem ; 58(5): 459-476, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39248186

ABSTRACT

BACKGROUND/AIMS: One of the treatments for breast cancer is surgical resection of the tumour and prevention of recurrence with postoperative radiotherapy. Unfortunately, radiotherapy is not always effective enough due to the low sensitivity of cancer cells to ionising radiation. This study aimed to evaluate the radiosensitising properties of resveratrol, piceatannol and polydatin on breast cancer cells, which differ in the presence of hormonal receptors on their surface. METHODS: The experimental part was carried out on triple-negative breast cancer cells (HCC38) and hormone-dependent cells (MCF7). The study assessed the level of cell death, changes in the expression of genes (BAX, BCL-2) and proteins related to the apoptosis process (CASPASE 3, 8 and P53), changes in the expression of antioxidant enzymes (CATALASE, SOD, GPx1/2) and NRF-2. Additionally, the expression level of RAD51 protein and histone H2AX, which are involved in DNA repair processes, was assessed. Statistical significance was evaluated by a two-way analysis of variance (ANOVA) followed by Tukey's post hoc test (p < 0.05). RESULTS: Ionising radiation in combination with resveratrol or piceatannol activates the apoptosis process by internal and external pathways. Greater sensitivity of MCF7 cells compared to HCC38 cells to ionising radiation in combination with resveratrol is associated with a weaker antioxidant response of cells and reduced intensity of DNA damage repair. DNA repair induced by ionising radiation occurs more effectively in HCC38 cells than in MCF7 cells. CONCLUSION: Resveratrol has the highest radiosensitising potential among the tested stilbene for cells of both lines. The effectiveness of ionizing radiation in combination with resveratrol (to a lesser extent with piceatannol) is more significant in MCF7 than in HCC38 cells.


Subject(s)
Apoptosis , Radiation, Ionizing , Radiation-Sensitizing Agents , Resveratrol , Stilbenes , Humans , Stilbenes/pharmacology , Resveratrol/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Female , Radiation-Sensitizing Agents/pharmacology , Cell Line, Tumor , MCF-7 Cells , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Breast Neoplasms/drug therapy , Histones/metabolism , DNA Repair/drug effects , DNA Repair/radiation effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Rad51 Recombinase/metabolism , Caspase 3/metabolism , Glucosides
2.
Chem Biol Drug Des ; 104(2): e14611, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39152534

ABSTRACT

Radiation resistance is a crucial factor influencing therapeutic outcomes in colorectal cancer (CRC). Baicalein (BE), primarily derived from Scutellaria baicalensis, has demonstrated anti-CRC properties. However, the impact of BE on the radiosensitivity of CRC remains unclear. This study aimed to evaluate the radiosensitization effects of BE and elucidate its mechanism in CRC radiotherapy. We established an in vitro radioresistant cell model (CT26-R) using parental CRC cells (CT26) subjected to ionizing radiation (IR). CT26-R cells were pretreated with or without BE, followed by transfection with pcDNA-NC and pcDNA-JAK2. The proliferation of CT26-R cells treated with BE and IR was assessed using a colony formation assay. A CRC animal model was developed in BALB/c mice via CT26-R cell transplantation. The radiosensitizing effect of BE on CRC was evaluated in vivo. TUNEL assay was conducted to detect apoptosis in tumor tissue. The expression levels of p-STAT3, JAK2, PD-L1, and SOCS3 in vitro and in vivo were measured by western blotting. Our results demonstrated that BE significantly increased radiosensitivity in vitro and in vivo and enhanced apoptosis in tumor tissues. Additionally, BE significantly downregulated the expression of p-STAT3, JAK2, and PD-L1, and significantly upregulated SOCS3 expression. These in vivo effects were reversed by pcDNA-JAK2. In summary, our data suggest that BE enhances CRC radiosensitivity by inhibiting the JAK2/STAT3 pathway.


Subject(s)
Apoptosis , Colorectal Neoplasms , Flavanones , Janus Kinase 2 , Mice, Inbred BALB C , Radiation Tolerance , STAT3 Transcription Factor , Signal Transduction , Janus Kinase 2/metabolism , Flavanones/pharmacology , Flavanones/chemistry , Flavanones/therapeutic use , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/radiotherapy , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Mice , Radiation Tolerance/drug effects , Cell Line, Tumor , Signal Transduction/drug effects , Apoptosis/drug effects , Humans , Cell Proliferation/drug effects , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , Radiation-Sensitizing Agents/chemistry
3.
Clin Nucl Med ; 49(10): 966-967, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39093035

ABSTRACT

ABSTRACT: A patient with widespread intensely prostate-specific membrane antigen-expressing, BRCA gene mutation-positive bone metastases at the time of prostate cancer diagnosis had progressed on multiple lines of standard therapy. He received 177 Lu-prostate-specific membrane antigen 8.5 GBq augmented by a short course of olaparib radiosensitization and achieved 90% decrease in serum PSA level after a single treatment. His tumor response was much better than expected by predictive dosimetry. However, his marrow radiotoxicity was worse than anticipated and required hospitalization. This suggests radiosensitizing agents to be a double-edged sword that must be carefully considered and balanced during activity prescription.


Subject(s)
Lutetium , Phthalazines , Piperazines , Prostatic Neoplasms, Castration-Resistant , Radiation-Sensitizing Agents , Humans , Male , Phthalazines/pharmacology , Piperazines/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Prostatic Neoplasms, Castration-Resistant/pathology , Radiation-Sensitizing Agents/pharmacology , Lutetium/pharmacology , Radioisotopes/pharmacology , Neoplasm Metastasis , Antigens, Surface , Glutamate Carboxypeptidase II/metabolism
4.
J Cancer Res Ther ; 20(4): 1284-1292, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39206990

ABSTRACT

BACKGROUND AND PURPOSE: Radiation therapy is a crucial treatment for nonsmall cell lung cancer (NSCLC), but its effectiveness is limited by the resistance of tumor cells to radiation. This study aimed to evaluate the effect of epicatechin (EC) on radiosensitivity in NSCLC and to determine its relationships with matrix metalloproteinase (MMP)-9. METHODS: MMP-9 expression was detected by Western blotting, and the expression of the DNA damage marker protein was detected by immunofluorescence. Cell viability was assessed using the CCK-8 assay, and cell proliferation was evaluated using the clonogenesis assay. Flow cytometry was used to determine the cell apoptosis, whereas cell migration and invasion were detected using the transwell assays. The cells were treated with ionizing radiation (IR) and EC to verify the sensitizing effect of EC on radiation therapy. RESULTS: MMP-9 expression was elevated in the NSCLC cells and tissues. DNA damage and cell apoptosis were increased, whereas cell vigor, proliferation, migration, and invasion were significantly decreased after IR. MMP-9 knockdown strengthened the impact of IR on the biological behaviors of the cells. EC + IR had the best effect on promoting DNA damage and the biological behaviors of the NSCLC cells; alternatively, the overexpression of MMP-9 weakened the role of EC. CONCLUSIONS: This study shows that EC can downregulate MMP-9 expression, promote DNA damage, reduce cell viability, proliferation, migration, and invasion, and facilitate cell apoptosis, thus, showing potential as a radiosensitizer for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cell Movement , Cell Proliferation , Lung Neoplasms , Matrix Metalloproteinase 9 , Radiation Tolerance , Humans , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Catechin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology
5.
Phytother Res ; 38(8): 4151-4167, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39136618

ABSTRACT

Despite active clinical trials on the use of Oleandrin alone or in combination with other drugs for the treatment of solid tumors, the potential synergistic effect of Oleandrin with radiotherapy remains unknown. This study reveals a new mechanism by which Oleandrin targets ATM and ATR kinase-mediated radiosensitization in lung cancer. Various assays, including clonogenic, Comet, immunofluorescence staining, apoptosis and Cell cycle assays, were conducted to evaluate the impact of oleandrin on radiation-induced double-strand break repair and cell cycle distribution. Western blot analysis was utilized to investigate alterations in signal transduction pathways related to double-strand break repair. The efficacy and toxicity of the combined therapy were assessed in a preclinical xenotransplantation model. Functionally, Oleandrin weakens the DNA damage repair ability and enhances the radiation sensitivity of lung cells. Mechanistically, Oleandrin inhibits ATM and ATR kinase activities, blocking the transmission of ATM-CHK2 and ATR-CHK1 cell cycle checkpoint signaling axes. This accelerates the passage of tumor cells through the G2 phase after radiotherapy, substantially facilitating the rapid entry of large numbers of inadequately repaired cells into mitosis and ultimately triggering mitotic catastrophe. The combined treatment of Oleandrin and radiotherapy demonstrated superior inhibition of tumor proliferation compared to either treatment alone. Our findings highlight Oleandrin as a novel and effective inhibitor of ATM and ATR kinase, offering new possibilities for the development of clinical radiosensitizing adjuvants.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Cardenolides , DNA Damage , Lung Neoplasms , Ataxia Telangiectasia Mutated Proteins/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Animals , Cardenolides/pharmacology , DNA Damage/drug effects , Cell Line, Tumor , Mice , Radiation Tolerance/drug effects , Signal Transduction/drug effects , Apoptosis/drug effects , Radiation-Sensitizing Agents/pharmacology , Mice, Nude , Xenograft Model Antitumor Assays , DNA Repair/drug effects , Cell Proliferation/drug effects , A549 Cells
6.
IET Nanobiotechnol ; 2024: 3786627, 2024.
Article in English | MEDLINE | ID: mdl-39144410

ABSTRACT

Background: Naringenin has shown great promise in the realm of cancer therapeutics, demonstrating excellent cytotoxic action toward cancer cells and the enhanced effects of radiation therapy in vitro. However, the medicinal value of naringenin is severely limited clinically by poor bioavailability. Thus, multiple drug-delivery strategies for overcoming this limitation have been developed, of which liposomes are considered the most suitable due to their amphiphilic, modifiable, and biocompatible characteristics. In this study, we investigated the role of naringenin and liposomal-delivered naringenin as adjuncts to radiotherapy in the MDA-MB-231 triple-negative breast cancer cell line in vitro. Materials and Methods: Liposomal-naringenin was synthesized by thin-film hydration and extrusion and was characterized by spectrophotometry, dynamic light scattering, and zeta potential. The effects of free-from naringenin and liposomal-naringenin were evaluated toward MDA-MB-231 cell viability when combined with varying doses of radiation. Additionally, cell growth patterns, morphology, and colony formation were evaluated. Results: The analysis demonstrated IC50 values of 387.5 and 546.6 µg/ml for naringenin and liposomal-naringenin, respectively. Naringenin and liposomal-naringenin significantly lowered cell viability, proliferation, and colony formation dose-dependently, as compared to radiation in isolation. Conclusion: The findings presented herein concur with previous accounts of the radiosensitizing potential of naringenin and further highlight the considerable biomedical application of liposomal-naringenin within the realm of radiotherapy.


Subject(s)
Cell Survival , Flavanones , Liposomes , Radiation-Sensitizing Agents , Triple Negative Breast Neoplasms , Flavanones/chemistry , Flavanones/pharmacology , Humans , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/drug therapy , Liposomes/chemistry , Cell Line, Tumor , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Cell Survival/drug effects , Female , Cell Proliferation/drug effects , MDA-MB-231 Cells
7.
Molecules ; 29(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39203014

ABSTRACT

Boron-enhanced proton therapy has recently appeared as a promising approach to increase the efficiency of proton therapy on tumor cells, and this modality can further be improved by the use of boron nanoparticles (B NPs) as local sensitizers to achieve enhanced and targeted therapeutic outcomes. However, the mechanisms of tumor cell elimination under boron-enhanced proton therapy still require clarification. Here, we explore possible molecular mechanisms responsible for the enhancement of therapeutic outcomes under boron NP-enhanced proton therapy. Spherical B NPs with a mode size of 25 nm were prepared by methods of pulsed laser ablation in water, followed by their coating by polyethylene glycol to improve their colloidal stability in buffers. Then, we assessed the efficiency of B NPs as sensitizers of cancer cell killing under irradiation with a 160.5 MeV proton beam. Our experiments showed that the combined effect of B NPs and proton irradiation induces an increased level of superoxide anion radical generation, which leads to the depolarization of mitochondria, a drop in their membrane mitochondrial potential, and the development of apoptosis. A comprehensive gene expression analysis (via RT-PCR) confirmed increased overexpression of 52 genes (out of 87 studied) involved in the cell redox status and oxidative stress, compared to 12 genes in the cells irradiated without B NPs. Other possible mechanisms responsible for the B NPs-induced radiosensitizing effect, including one related to the generation of alpha particles, are discussed. The obtained results give a better insight into the processes involved in the boron-induced enhancement of proton therapy and enable one to optimize parameters of proton therapy in order to maximize therapeutic outcomes.


Subject(s)
Apoptosis , Boron , Nanoparticles , Proton Therapy , Humans , Boron/chemistry , Boron/pharmacology , Nanoparticles/chemistry , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Neoplasms/radiotherapy , Neoplasms/drug therapy , Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Oxidative Stress/drug effects
8.
Chem Biol Interact ; 399: 111149, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39032852

ABSTRACT

Rhabdomyosarcoma (RMS) represents one of the most lethal soft-tissue sarcomas in children. The toxic trace element arsenic has been reported to function as a radiosensitizer in sarcomas. To investigate the role of arsenic sulfide (As4S4) in enhancing radiation sensitization in RMS, this study was conducted to elucidate its underlying mechanism in radiotherapy. The combination of As4S4 and radiotherapy showed significant inhibition in RMS cells, as demonstrated by the cell counting kit-8 (CCK-8) assay and flow cytometry. Subsequently, we demonstrated for the first time that As4S4, as well as the knockdown of NFATc3 led to double-strand break (DSB) through increased expression of RAG1. In vivo experiment confirmed that co-treatment efficiently inhibited RMS growth. Furthermore, survival analysis of a clinical cohort consisting of 59 patients revealed a correlation between NFATc3 and RAG1 expression and overall survival (OS). Cox regression analysis also confirmed the independent prognostic significance of NFATc3 and RAG1.Taken together, As4S4 enhances radiosensitivity in RMS via activating NFATc3-RAG1 mediated DSB. NFATc3 and RAG1 are potential therapeutic targets. As4S4 will hopefully serve as a prospective radio-sensitizing agent for RMS.


Subject(s)
Arsenicals , DNA Breaks, Double-Stranded , NFATC Transcription Factors , Radiation Tolerance , Rhabdomyosarcoma , Sulfides , Humans , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , Sulfides/pharmacology , Sulfides/therapeutic use , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/radiotherapy , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/genetics , Cell Line, Tumor , Male , Female , Arsenicals/pharmacology , Arsenicals/therapeutic use , Animals , Radiation Tolerance/drug effects , NFATC Transcription Factors/metabolism , Mice , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Mice, Nude , Child , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , Mice, Inbred BALB C
9.
J Med Chem ; 67(14): 12366-12385, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39007759

ABSTRACT

The dominant role of non-homologous end-joining in the repair of radiation-induced double-strand breaks identifies DNA-dependent protein kinase (DNA-PK) as an excellent target for the development of radiosensitizers. We report the discovery of a new class of imidazo[4,5-c]pyridine-2-one DNA-PK inhibitors. Structure-activity studies culminated in the identification of 78 as a nM DNA-PK inhibitor with excellent selectivity for DNA-PK compared to related phosphoinositide 3-kinase (PI3K) and PI3K-like kinase (PIKK) families and the broader kinome, and displayed DNA-PK-dependent radiosensitization of HAP1 cells. Compound 78 demonstrated robust radiosensitization of a broad range of cancer cells in vitro, displayed high oral bioavailability, and sensitized colorectal carcinoma (HCT116/54C) and head and neck squamous cell carcinoma (UT-SCC-74B) tumor xenografts to radiation. Compound 78 also provided substantial tumor growth inhibition of HCT116/54C tumor xenografts in combination with radiation. Compound 78 represents a new, potent, and selective class of DNA-PK inhibitors with significant potential as radiosensitizers for cancer treatment.


Subject(s)
DNA-Activated Protein Kinase , Protein Kinase Inhibitors , Radiation-Sensitizing Agents , Humans , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , Animals , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/chemical synthesis , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacokinetics , Radiation-Sensitizing Agents/chemical synthesis , Structure-Activity Relationship , Mice , Cell Line, Tumor , Imidazoles/pharmacology , Imidazoles/chemistry , Imidazoles/chemical synthesis , Imidazoles/pharmacokinetics , Pyridones/pharmacology , Pyridones/chemistry , Pyridones/chemical synthesis , Pyridones/pharmacokinetics , Xenograft Model Antitumor Assays , Mice, Nude , Rats
10.
Anticancer Res ; 44(8): 3587-3591, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060048

ABSTRACT

BACKGROUND/AIM: Acetyl glucose adducts (UTX-114, -115, and -116) were prepared from gefitinib, and their characteristics (e.g., anticancer activity, structural property) were analyzed. MATERIALS AND METHODS: Cytotoxicity and radiosensitizing properties of the UTX-114 family were examined using A431 cells. Supramolecular associations between the UTX-114 family compounds and the tyrosine kinase domain of epidermal growth factor receptor (EGFR-tyk) were also examined. The interactive analyses of the UTX-114 family compounds with EGFR-tyk were performed using docking simulation technique. RESULTS: The UTX-114 family showed a similar cytotoxicity as gefitinib, yielding IC50 values of 31.2 µM (gefitinib), 34.3 µM (UTX-114), 36.8 µM (UTX-115), and 39.4 µM (UTX-116). The EGFR-tyk inhibition ratios (IR) of UTX-114, -115, and -116 to gefitinib were 1.515, 0.983, and 0.551, respectively. The EGFR-tyk inhibitory activity of UTX-114 was higher than that of gefitinib. UTX-114 also showed the highest radiosensitizing activity among the tested compounds. UTX-114 expressed 1841 conformers (-8.989~15.718 kcal/mol) with the solvation free energy (dGW) of UTX-114 decreasing with increasing conformational energy, ranging between -354.955~ -260.815 kJ/mol. Interactive energies of gefitinib, UTX-114, -115, and -116 with EGFR-tyk were -123.640, -144.053, -120.830, and -124.658 kcal/mol, respectively. CONCLUSION: UTX-114 yielded the lowest interaction energy with EGFR-tyk among tested compounds. Given the association behavior between UTX-114 and EGFR-tyk, along with its other observed properties, UTX-114 appears to be a viable therapeutic possibility.


Subject(s)
ErbB Receptors , Gefitinib , Molecular Docking Simulation , Gefitinib/pharmacology , Humans , ErbB Receptors/metabolism , ErbB Receptors/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/chemistry , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Glycosylation , Protein Kinase Inhibitors/pharmacology , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry
11.
Anticancer Res ; 44(8): 3295-3306, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060064

ABSTRACT

BACKGROUND/AIM: Despite the established antitumor effectiveness and synergistic interactions of melatonin with photon irradiation, its role in carbon-ion radiotherapy remains uncertain. This study aimed to elucidate the mechanisms and potential clinical advantages of combining exogenous melatonin therapy with carbon-ion radiotherapy. MATERIALS AND METHODS: The investigation assessed the impact of combining exogenous melatonin with photon or carbon-ion irradiation on cell-cycle modulation and DNA-repair capability using the melanoma cell line B16F10. RNA sequencing and bioinformatics analysis were conducted to explore mechanisms and evaluate potential clinical benefits, with validation performed on the osteosarcoma cell line LM8. RESULTS: Pre-treatment with melatonin reduced the survival fraction of B16F10 and LM8 cells upon exposure to photon and carbon-ion radiation. Mechanistically, melatonin was found to inhibit G2/M arrest, preserve DNA damage, and suppress key genes involved in DNA double-strand break repair after 8 Gy carbon-ion radiation. Furthermore, RNA sequencing and bioinformatics analysis revealed favorable changes in genes associated with survival and metastasis, highlighting potential clinical significance. LM8 cells treated with melatonin exhibited increased radiosensitivity and suppression of DNA-repair proteins. CONCLUSION: The combination of exogenous melatonin not only heightened radiosensitivity and modulated hallmark tumor gene sets in vitro but also markedly suppressed the efficiency of DNA double-strand break-repair pathway, thus enhancing the cytotoxicity of carbon-ion radiotherapy.


Subject(s)
DNA Repair , Heavy Ion Radiotherapy , Melatonin , Radiation Tolerance , Radiation-Sensitizing Agents , Melatonin/pharmacology , Cell Line, Tumor , Radiation Tolerance/drug effects , Mice , Animals , Humans , DNA Repair/drug effects , DNA Repair/radiation effects , Radiation-Sensitizing Agents/pharmacology , Cell Survival/drug effects , Cell Survival/radiation effects , Melanoma, Experimental/radiotherapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects
12.
Cancer Med ; 13(13): e7332, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38967145

ABSTRACT

BACKGROUND: Radiotherapy (RT) is a widely utilized tumor treatment approach, while a significant obstacle in this treatment modality is the radioresistance exhibited by tumor cells. To enhance the effectiveness of RT, scientists have explored radiosensitization approaches, including the use of radiosensitizers and physical stimuli. Nevertheless, several approaches have exhibited disappointing results including adverse effects and limited efficacy. A safer and more effective method of radiosensitization involves low-intensity ultrasound (LIUS), which selectively targets tumor tissue and enhances the efficacy of radiation therapy. METHODS: This review summarized the tumor radioresistance reasons and explored LIUS potential radiosensitization mechanisms. Moreover, it covered diverse LIUS application strategies in radiosensitization, including the use of LIUS alone, ultrasound-targeted intravascular microbubble destruction, ultrasound-mediated targeted radiosensitizers delivery, and sonodynamic therapy. Lastly, the review presented the limitations and prospects of employing LIUS-RT combined therapy in clinical settings, emphasizing the need to connect research findings with practical applications. RESULTS AND CONCLUSION: LIUS employs cost-effective equipment to foster tumor radiosensitization, curtail radiation exposure, and elevate the quality of life for patients. This efficacy is attributed to LIUS's ability to utilize thermal, cavitation, and mechanical effects to overcome tumor cell resistance to RT. Multiple experimental analyses have underscored the effectiveness of LIUS in inducing tumor radiosensitization using diverse strategies. While initial studies have shown promising results, conducting more comprehensive clinical trials is crucial to confirm its safety and effectiveness in real-world situations.


Subject(s)
Neoplasms , Radiation-Sensitizing Agents , Ultrasonic Therapy , Humans , Neoplasms/radiotherapy , Neoplasms/therapy , Radiation-Sensitizing Agents/therapeutic use , Radiation-Sensitizing Agents/pharmacology , Ultrasonic Therapy/methods , Combined Modality Therapy , Animals , Radiation Tolerance , Ultrasonic Waves
13.
Int J Nanomedicine ; 19: 6499-6513, 2024.
Article in English | MEDLINE | ID: mdl-38946887

ABSTRACT

Purpose: To address the problem of suboptimal reactive oxygen species (ROS) production in Radiation therapy (RT) which was resulted from exacerbated tumor hypoxia and the heterogeneous distribution of radiation sensitizers. Materials and Methods: In this work, a novel nanomedicine, designated as PLGA@IR780-Bi-DTPA (PIBD), was engineered by loading the radiation sensitizer Bi-DTPA and the photothermal agent IR780 onto poly(lactic-co-glycolic acid) (PLGA). This design leverages the tumor-targeting ability of IR780 to ensure selective accumulation of the nanoparticles in tumor cells, particularly within the mitochondria. The effect of the photothermal therapy-enhanced radiation therapy was also examined to assess the alleviation of hypoxia and the enhancement of radiation sensitivity. Results: The PIBD nanoparticles exhibited strong capacity in mitochondrial targeting and selective tumor accumulation. Upon activation by 808 nm laser irradiation, the nanoparticles effectively alleviated local hypoxia by photothermal effect enhanced blood supplying to improve oxygen content, thereby enhancing the ROS production for effective RT. Comparative studies revealed that PIBD-induced RT significantly outperformed conventional RT in treating hypoxic tumors. Conclusion: This design of tumor-targeting photothermal therapy-enhanced radiation therapy nanomedicine would advance the development of targeted drug delivery system for effective RT regardless of hypoxic microenvironment.


Subject(s)
Nanoparticles , Photothermal Therapy , Polylactic Acid-Polyglycolic Acid Copolymer , Reactive Oxygen Species , Animals , Photothermal Therapy/methods , Reactive Oxygen Species/metabolism , Nanoparticles/chemistry , Cell Line, Tumor , Humans , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Mice , Indoles/pharmacology , Indoles/chemistry , Tumor Hypoxia/drug effects , Tumor Hypoxia/radiation effects , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/radiotherapy , Neoplasms/therapy , Neoplasms/metabolism , Nanomedicine
14.
Adv Sci (Weinh) ; 11(29): e2402039, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38828705

ABSTRACT

Exploring efficient and low-toxicity radiosensitizers to break through the bottleneck of radiation tolerance, immunosuppression and poor prognosis remains one of the critical developmental challenges in radiotherapy. Nanoheterojunctions, due to their unique physicochemical properties, have demonstrated excellent radiosensitization effects in radiation energy deposition and in lifting tumor radiotherapy inhibition. Herein, they doped selenium (Se) into prussian blue (PB) to construct a nano-heterojunction (Se@PB), which could promote the increase of Fe2+/Fe3+ ratio and conversion of Se to a high valence state with Se introduction. The Fe2+-Se-Fe3+ electron transfer chain accelerates the rate of electron transfer on the surface of the nanoparticles, which in turn endows it with efficient X-ray energy transfer and electron transport capability, and enhances radiotherapy physical sensitivity. Furthermore, Se@PB induces glutathione (GSH) depletion and Fe2+ accumulation through pro-Fenton reaction, thereby disturbs the redox balance in tumor cells and enhances biochemical sensitivity of radiotherapy. As an excellent radiosensitizer, Se@PB effectively enhances X-ray induced mitochondrial dysfunction and DNA damage, thereby promotes cell apoptosis and synergistic cervical cancer radiotherapy. This study elucidates the radiosensitization mechanism of Se-doped nanoheterojunction from the perspective of the electron transfer chain and biochemistry reaction, which provides an efficient and low-toxic strategy in radiotherapy.


Subject(s)
Radiation-Sensitizing Agents , Selenium , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology , Humans , Selenium/chemistry , Selenium/pharmacology , Female , Ferrocyanides/chemistry , Animals , Mice , Nanoparticles/chemistry , Cell Line, Tumor , Apoptosis/drug effects , Uterine Cervical Neoplasms , Radiation Tolerance/drug effects , Disease Models, Animal
15.
Toxicol In Vitro ; 99: 105878, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906201

ABSTRACT

Copper-Cysteamine nanoparticles (Cu-Cy NPs) have emerged as promising radiosensitizers in cancer treatment. This study aims to investigate the combined therapeutic effect of these nanoparticles and cisplatin using a clinical linear accelerator to enhance the efficacy of chemoradiation therapy for cervical cancer. Following successful synthesis and characterization of Cu-Cy NPs, the cytotoxicity effect of these nanoparticles and cisplatin in various concentrations was evaluated on HeLa cancer cells, individually and in combination. Additionally, the radiobiological effects of these agents were investigated under a 6MV linear accelerator. At a concentration of 25 mg/L, Cu-Cy NPs displayed no significant cytotoxicity toward HeLa cancer cells. However, when combined with 2Gy X-ray irradiation at this concentration, the nanoparticles demonstrated a potent radiosensitizing effect. Notably, cell viability and migration rate in the combination group (Cu-Cy NPs + cisplatin + radiation) were significantly reduced compared to the radiation-alone group. Additionally, the combination treatment induced a significantly higher rate of apoptosis compared to the radiation-alone group. Overall, Cu-Cy NPs exhibited a significant dose-dependent synergistic enhancement of radiation efficacy when combined with cisplatin under X-ray exposure, and may provide a promising approach to improve the therapeutic effect of conventional radiation therapy.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Survival , Chemoradiotherapy , Cisplatin , Copper , Radiation-Sensitizing Agents , Cisplatin/pharmacology , Humans , Copper/chemistry , Copper/pharmacology , HeLa Cells , Cell Survival/drug effects , Antineoplastic Agents/pharmacology , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/chemistry , Apoptosis/drug effects , Nanoparticles/chemistry , Cell Movement/drug effects , X-Rays
16.
Mol Pharm ; 21(7): 3218-3232, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38885477

ABSTRACT

Squamous cell carcinoma (SCC) is a common nonmelanoma skin cancer. Radiotherapy plays an integral role in treating SCC due to its characteristics, such as diminished intercellular adhesion, heightened cell migration and invasion capabilities, and immune evasion. These problems lead to inaccurate tumor boundary positioning and radiotherapy tolerance in SCC treatment. Thus, accurate localization and enhanced radiotherapy sensitivity are imperative for effective SCC treatment. To address the existing limitations in SCC therapy, we developed monoglyceride solid lipid nanoparticles (MG SLNs) and enveloped them with the A431 cell membrane (A431 CM) to create A431@MG. The characterization results showed that A431@MG was spherical. Furthermore, A431@MG had specific targeting for A431 cells. In A431 tumor-bearing mice, A431@MG demonstrated prolonged accumulation within tumors, ensuring precise boundary localization of SCC. We further advanced the approach by preparing MG SLNs encapsulating 5-aminolevulinic acid methyl ester (MLA) and desferrioxamine (DFO) with an A431 CM coating to yield A431@MG-MLA/DFO. Several studies have revealed that DFO effectively reduced iron content, impeding protoporphyrin IX (PpIX) biotransformation and promoting PpIX accumulation. Simultaneously, MLA was metabolized into PpIX upon cellular entry. During radiotherapy, the heightened PpIX levels enhanced reactive oxygen species (ROS) generation, inducing DNA and mitochondrial damage and leading to cell apoptosis. In A431 tumor-bearing mice, the A431@MG-MLA/DFO group exhibited notable radiotherapy sensitization, displaying superior tumor growth inhibition. Combining A431@MG-MLA/DFO with radiotherapy significantly improved anticancer efficacy, highlighting its potential to serve as an integrated diagnostic and therapeutic strategy for SCC.


Subject(s)
Carcinoma, Squamous Cell , Cell Membrane , Nanoparticles , Radiation-Sensitizing Agents , Skin Neoplasms , Animals , Mice , Nanoparticles/chemistry , Humans , Cell Line, Tumor , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/administration & dosage , Cell Membrane/metabolism , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/administration & dosage , Lipids/chemistry , Xenograft Model Antitumor Assays , Deferoxamine/chemistry , Deferoxamine/pharmacology , Mice, Nude , Female , Mice, Inbred BALB C , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Liposomes
17.
ACS Appl Mater Interfaces ; 16(27): 34450-34466, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38941284

ABSTRACT

Radiosensitizers play a pivotal role in enhancing radiotherapy (RT). One of the challenges in RT is the limited accumulation of nanoradiosensitizers and the difficulty in activating antitumor immunity. Herein, a smart strategy was used to achieve in situ aggregation of nanomanganese adjuvants (MnAuNP-C&B) to enhance RT-induced antitumor immunity. The aggregated MnAuNP-C&B system overcomes the shortcomings of small-sized nanoparticles that easily flow back into blood vessels and diffuse into surrounding tissues, and it also prolongs the retention time of nanomanganese within cancer cells and tumors. The MnAuNP-C&B system significantly enhances the radiosensitization effect in RT. Additionally, the pH-responsive disassembly of MnAuNP-C&B triggers the release of Mn2+, further promoting RT-induced activation of the STING pathway and eliciting robust antitumor immunity. Overall, our study presents a smart strategy wherein in situ aggregation of nanomanganese effectively inhibits tumor growth through radiosensitization and the activation of antitumor immunity.


Subject(s)
Radiation-Sensitizing Agents , Animals , Mice , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacology , Humans , Manganese/chemistry , Cell Line, Tumor , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Female , Mice, Inbred BALB C , Neoplasms/radiotherapy , Neoplasms/immunology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology
18.
Int J Mol Sci ; 25(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38891817

ABSTRACT

(1) Head and neck squamous cell carcinoma (HNSCC) is common, while treatment is difficult, and mortality is high. Kinase inhibitors are promising to enhance the effects of radiotherapy. We compared the effects of the PARP inhibitors talazoparib and niraparib and that of the DNA-PKcs inhibitor AZD7648, combined with ionizing radiation. (2) Seven HNSCC cell lines, including Cal33, CLS-354, Detroit 562, HSC4, RPMI2650 (HPV-negative), UD-SCC-2 and UM-SCC-47 (HPV-positive), and two healthy fibroblast cell lines, SBLF8 and SBLF9, were studied. Flow cytometry was used to analyze apoptosis and necrosis induction (AnnexinV/7AAD) and cell cycle distribution (Hoechst). Cell inactivation was studied by the colony-forming assay. (3) AZD7648 had the strongest effects, radiosensitizing all HNSCC cell lines, almost always in a supra-additive manner. Talazoparib and niraparib were effective in both HPV-positive cell lines but only consistently in one and two HPV-negative cell lines, respectively. Healthy fibroblasts were not affected by any combined treatment in apoptosis and necrosis induction or G2/M-phase arrest. AZD7648 alone was not toxic to healthy fibroblasts, while the combination with ionizing radiation reduced clonogenicity. (4) In conclusion, talazoparib, niraparib and, most potently, AZD7648 could improve radiation therapy in HNSCC. Healthy fibroblasts tolerated AZD7648 alone extremely well, but irradiation-induced effects might occur. Our results justify in vivo studies.


Subject(s)
Apoptosis , Indazoles , Phthalazines , Piperidines , Poly(ADP-ribose) Polymerase Inhibitors , Radiation-Sensitizing Agents , Squamous Cell Carcinoma of Head and Neck , Humans , Phthalazines/pharmacology , Indazoles/pharmacology , Piperidines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Cell Line, Tumor , Radiation-Sensitizing Agents/pharmacology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Apoptosis/drug effects , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/radiotherapy , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism
19.
Cell Death Dis ; 15(6): 426, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890278

ABSTRACT

Radiation therapy (RT) remains a common treatment for cancer patients worldwide, despite the development of targeted biological compounds and immunotherapeutic drugs. The challenge in RT lies in delivering a lethal dose to the cancerous site while sparing the surrounding healthy tissues. Low linear energy transfer (low-LET) and high linear energy transfer (high-LET) radiations have distinct effects on cells. High-LET radiation, such as alpha particles, induces clustered DNA double-strand breaks (DSBs), potentially inducing cell death more effectively. However, due to limited range, alpha-particle therapies have been restricted. In human cancer, mutations in TP53 (encoding for the p53 tumor suppressor) are the most common genetic alteration. It was previously reported that cells carrying wild-type (WT) p53 exhibit accelerated senescence and significant rates of apoptosis in response to RT, whereas cells harboring mutant p53 (mutp53) do not. This study investigated the combination of the alpha-emitting atoms RT based on internal Radium-224 (224Ra) sources and systemic APR-246 (a p53 reactivating compound) to treat tumors with mutant p53. Cellular models of colorectal cancer (CRC) or pancreatic ductal adenocarcinoma (PDAC) harboring mutant p53, were exposed to alpha particles, and tumor xenografts with mutant p53 were treated using 224Ra source and APR-246. Effects on cell survival and tumor growth, were assessed. The spread of alpha emitters in tumors was also evaluated as well as the spatial distribution of apoptosis within the treated tumors. We show that mutant p53 cancer cells exhibit radio-sensitivity to alpha particles in vitro and to alpha-particles-based RT in vivo. APR-246 treatment enhanced sensitivity to alpha radiation, leading to reduced tumor growth and increased rates of tumor eradication. Combining alpha-particles-based RT with p53 restoration via APR-246 triggered cell death, resulting in improved therapeutic outcomes. Further preclinical and clinical studies are needed to provide a promising approach for improving treatment outcomes in patients with mutant p53 tumors.


Subject(s)
Alpha Particles , Radiation-Sensitizing Agents , Tumor Suppressor Protein p53 , Alpha Particles/therapeutic use , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Mice , Radiation-Sensitizing Agents/pharmacology , Mutation , Quinuclidines/pharmacology , Cell Line, Tumor , Mice, Nude , Xenograft Model Antitumor Assays , Apoptosis/drug effects , Apoptosis/radiation effects , Neoplasms/radiotherapy , Neoplasms/genetics , Neoplasms/pathology
20.
In Vivo ; 38(4): 1719-1730, 2024.
Article in English | MEDLINE | ID: mdl-38936939

ABSTRACT

BACKGROUND/AIM: ClFdA is a second-generation antineoplastic agent that has demonstrated significant anticancer activity, particularly against acute lymphoblastic leukemia and has been shown to have radiosensitizing activity. The aim of the study was to explore the genotoxic, cytotoxic and radiosensitizing effects of clofarabine (ClFdA) on bone marrow cells (BMCs), normoblasts and leukocytes of mice in vivo. MATERIALS AND METHODS: Cytotoxicity was determined by the reduction in reticulocytes (RET), and genotoxicity was determined by the induction of micronucleated reticulocytes (MN-RET) in the peripheral blood and by DNA break induction in leukocytes determined by single-cell gel electrophoresis (SCGE). The radiosensitizing capacity of ClFdA was determined in leukocytes and BMCs by SCGE. RESULTS: Two mechanisms of MN-RET induction were identified according to the antecedents, that could be due to inhibition of DNA synthesis and demethylation of G-C regions, and subsequent chromosome fragility. ClFdA cytotoxicity causes two contiguous peaks, an early peak that seems to inhibit MN-RET induction and a second peak that seems to be caused by ribonucleotide reductase (RR) and/or DNA synthesis inhibitions. ClFdA induced early DNA damage in noncycling leukocytes, and also radiosensitizes leukocytes immediately after treatment. ClFdA-ionizing radiation (IR) causes two time-dependent episodes of DNA damage, the latest after 80 min triggers a major breakage of DNA. In terms of the number of damaged cells, leukocytes and BMCs are similarly sensitive to ionizing radiation; BMCs are slightly more sensitive than leukocytes to ClFdA, but BMCs are doubly sensitive to combined treatment. CONCLUSION: ClFdA causes early DNA damage and radiosensitivity in non-proliferating leukocytes, which rules out the most favored hypotheses of the participation of RR and DNA polymerase inhibition.


Subject(s)
Clofarabine , DNA Damage , Leukocytes , Radiation-Sensitizing Agents , Animals , Clofarabine/pharmacology , Mice , Radiation-Sensitizing Agents/pharmacology , Leukocytes/drug effects , Leukocytes/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Arabinonucleosides/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/radiation effects , Bone Marrow Cells/metabolism , Adenine Nucleotides/pharmacology , Male , Reticulocytes/drug effects , Reticulocytes/radiation effects , Antineoplastic Agents/pharmacology , Micronucleus Tests
SELECTION OF CITATIONS
SEARCH DETAIL