Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
1.
Angew Chem Int Ed Engl ; : e202407469, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38980970

RESUMEN

2-Bromo-1-(3,3-dinitroazetidin-1-yl)ethan-1-one (RRx-001) is a hypoxic cell chemotherapeutics with already demonstrated synergism in combined chemo-radiation therapy. The interaction of the compound with secondary low-energy electrons formed in large amounts during the physico-chemical phase of the irradiation may lead to these synergistic effects. The present study focuses on the first step of RRx-001 interaction with low-energy electrons in which a transient anion is formed and fragmented. Combination of two experiments allows us to disentangle the decay of the RRx-001 anion on different timescales. Sole presence of the electron initiates rapid dissociation of NO2 and HNO2 neutrals while NO2- and Br- anions are produced both directly and via intermediate complexes. Based on our quantum chemical calculations, we propose that bidirectional intersystem crossing between π*(NO2) and σ*(C-Br) states explains the experimental spectra. The fast dynamics monitored will impact the condensed phase chemistry of the anion as well.

2.
Heliyon ; 10(13): e33734, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39050474

RESUMEN

This study investigates the photon interaction mechanism of various small molecule radiosensitizers, including Hydrogen Peroxide, Nimorazole, 5-Fluorouracil, NVX-108, and others, using the MCNP 6.3 Monte Carlo simulation code. The simulations focused on quantifying the linear attenuation coefficients, mean free path, and accumulation factors of these radiosensitizers, as well as their interactions in a simulated spherical water phantom irradiated with a 100 keV mono-energetic X-ray source. Our findings reveal significant variations in deposited energy, collision events, and mean free path among the radiosensitizers, indicating different efficacy levels in enhancing radiation therapy. Notably, NVX-108 demonstrated the highest energy deposition, suggesting its potential as a highly effective radiosensitizer. The study also examined the individual attenuation properties of these radiosensitizers against energetic photons, with NVX-108 showing the highest attenuation coefficient and a shorter mean free path, further supporting its superior potential in effective radiosensitization. It can be concluded that NVX-108 has higher interaction tendency with the energetic photons comparing other small-molecules under investigation.

3.
Adv Sci (Weinh) ; 11(29): e2402039, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38828705

RESUMEN

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.


Asunto(s)
Fármacos Sensibilizantes a Radiaciones , Selenio , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Humanos , Selenio/química , Selenio/farmacología , Femenino , Ferrocianuros/química , Animales , Ratones , Nanopartículas/química , Línea Celular Tumoral , Apoptosis/efectos de los fármacos , Neoplasias del Cuello Uterino , Tolerancia a Radiación/efectos de los fármacos , Modelos Animales de Enfermedad
4.
Molecules ; 29(10)2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38792171

RESUMEN

Azido-modified nucleosides have been extensively explored as substrates for click chemistry and the metabolic labeling of DNA and RNA. These compounds are also of interest as precursors for further synthetic elaboration and as therapeutic agents. This review discusses the chemistry of azidonucleosides related to the generation of nitrogen-centered radicals (NCRs) from the azido groups that are selectively inserted into the nucleoside frame along with the subsequent chemistry and biological implications of NCRs. For instance, the critical role of the sulfinylimine radical generated during inhibition of ribonucleotide reductases by 2'-azido-2'-deoxy pyrimidine nucleotides as well as the NCRs generated from azidonucleosides by radiation-produced (prehydrated and aqueous) electrons are discussed. Regio and stereoselectivity of incorporation of an azido group ("radical arm") into the frame of nucleoside and selective generation of NCRs under reductive conditions, which often produce the same radical species that are observed upon ionization events due to radiation and/or other oxidative conditions that are emphasized. NCRs generated from nucleoside-modified precursors other than azidonucleosides are also discussed but only with the direct relation to the same/similar NCRs derived from azidonucleosides.


Asunto(s)
Azidas , Nucleósidos , Nucleósidos/química , Azidas/química , Nitrógeno/química , Radicales Libres/química , Química Clic
5.
Dokl Biochem Biophys ; 516(1): 111-114, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38795244

RESUMEN

Proton therapy can treat tumors located in radiation-sensitive tissues. This article demonstrates the possibility of enhancing the proton therapy with targeted gold nanoparticles that selectively recognize tumor cells. Au-PEG nanoparticles at concentrations above 25 mg/L and 4 Gy proton dose caused complete death of EMT6/P cells in vitro. Binary proton therapy using targeted Au-PEG-FA nanoparticles caused an 80% tumor growth inhibition effect in vivo. The use of targeted gold nanoparticles is promising for enhancing the proton irradiation effect on tumor cells and requires further research to increase the therapeutic index of the approach.


Asunto(s)
Carcinoma de Ehrlich , Oro , Nanopartículas del Metal , Terapia de Protones , Oro/química , Oro/farmacología , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Terapia de Protones/métodos , Animales , Carcinoma de Ehrlich/radioterapia , Carcinoma de Ehrlich/tratamiento farmacológico , Carcinoma de Ehrlich/patología , Ratones , Línea Celular Tumoral , Polietilenglicoles/química
6.
Bull Exp Biol Med ; 176(5): 626-630, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38730109

RESUMEN

We studied the antitumor activity of the combined use of local proton irradiation in two modes (10 and 31 Gy) with preliminary intra-tumoral injection of two types of bismuth nanoparticles differing in surface coating: coated with the amphiphilic molecule Pluronic-F127 or Silane-PEG (5 kDa)-COOH polymer. Nanoparticles were used in doses of 0.75 and 1.5 mg/mouse. In two independent series on experimental tumor model (solid Ehrlich carcinoma), bismuth nanoparticles of both modifications injected directly into the tumor enhanced the antitumor effects of proton therapy. Moreover, the radiosensitizing effect of bismuth nanoparticles administered via this route increased with the increasing the doses of nanoparticles and the doses of radiation exposure. In our opinion, these promising data obtained for the first time extend the possibilities of treating malignant neoplasms.


Asunto(s)
Bismuto , Carcinoma de Ehrlich , Poloxámero , Terapia de Protones , Carcinoma de Ehrlich/radioterapia , Carcinoma de Ehrlich/tratamiento farmacológico , Carcinoma de Ehrlich/patología , Animales , Bismuto/uso terapéutico , Bismuto/química , Ratones , Terapia de Protones/métodos , Poloxámero/química , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/farmacología , Polietilenglicoles/química , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Nanopartículas/química , Femenino
7.
Small ; 20(35): e2400954, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38676336

RESUMEN

In the progression of X-ray-based radiotherapy for the treatment of cancer, the incorporation of nanoparticles (NPs) has a transformative impact. This study investigates the potential of NPs, particularly those comprised of high atomic number elements, as radiosensitizers. This aims to optimize localized radiation doses within tumors, thereby maximizing therapeutic efficacy while preserving surrounding tissues. The multifaceted applications of NPs in radiotherapy encompass collaborative interactions with chemotherapeutic, immunotherapeutic, and targeted pharmaceuticals, along with contributions to photodynamic/photothermal therapy, imaging enhancement, and the integration of artificial intelligence technology. Despite promising preclinical outcomes, the paper acknowledges challenges in the clinical translation of these findings. The conclusion maintains an optimistic stance, emphasizing ongoing trials and technological advancements that bolster personalized treatment approaches. The paper advocates for continuous research and clinical validation, envisioning the integration of NPs as a revolutionary paradigm in cancer therapy, ultimately enhancing patient outcomes.


Asunto(s)
Nanopartículas Multifuncionales , Humanos , Rayos X , Nanopartículas Multifuncionales/química , Neoplasias/terapia , Neoplasias/diagnóstico por imagen , Animales , Fármacos Sensibilizantes a Radiaciones/química , Fármacos Sensibilizantes a Radiaciones/uso terapéutico
8.
J Drug Target ; 32(5): 544-556, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38469874

RESUMEN

Radio-photothermal therapy is an effective modality for cancer treatment. To overcome the radio-resistance in the hypoxic microenvironment and improve the sensitivity of radiotherapy, metal nanoparticles, and radio-photothermal therapy are widely used in the research of improving the curative effect and reducing the side effects of radiotherapy. Here, we developed red blood membrane camouflaging bismuth nanoflowers (RBCM-BNF) with outstanding physiological stability and biodegradability for lung tumours. In vitro data proved that the RBCM-BNF had the greatest cancer cell-killing ability combined with X-ray irradiation and photo-thermal treatment. Meanwhile, in vivo studies revealed that RBCM-BNF can alleviate the hypoxic microenvironment and promote tumour cell apoptosis by inhibiting HIF-1α expression and increasing caspase-3 expression. Therefore, RBCM-BNF had a good radio-sensitising effect and might be a promising biomimetic nanoplatform as a therapeutic target for cancer.


Asunto(s)
Bismuto , Membrana Eritrocítica , Neoplasias Pulmonares , Terapia Fototérmica , Bismuto/química , Neoplasias Pulmonares/terapia , Animales , Humanos , Terapia Fototérmica/métodos , Ratones , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ratones Endogámicos BALB C , Nanopartículas/química , Ratones Desnudos , Microambiente Tumoral , Células A549 , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
9.
Cancer Biol Ther ; 25(1): 2317999, 2024 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-38445632

RESUMEN

Rectal cancer accounts for the second highest cancer-related mortality, which is predominant in Western civilizations. The treatment for rectal cancers includes surgery, radiotherapy, chemotherapy, and immunotherapy. Radiotherapy, specifically external beam radiation therapy, is the most common way to treat rectal cancer because radiation not only limits cancer progression but also significantly reduces the risk of local recurrence. However, therapeutic radiation-induced radioresistance to rectal cancer cells and toxicity to normal tissues are major drawbacks. Therefore, understanding the mechanistic basis of developing radioresistance during and after radiation therapy would provide crucial insight to improve clinical outcomes of radiation therapy for rectal cancer patients. Studies by various groups have shown that radiotherapy-mediated changes in the tumor microenvironment play a crucial role in developing radioresistance. Therapeutic radiation-induced hypoxia and functional alterations in the stromal cells, specifically tumor-associated macrophage (TAM) and cancer-associated fibroblasts (CAF), play a crucial role in developing radioresistance. In addition, signaling pathways, such as - the PI3K/AKT pathway, Wnt/ß-catenin signaling, and the hippo pathway, modulate the radiation responsiveness of cancer cells. Different radiosensitizers, such as small molecules, microRNA, nanomaterials, and natural and chemical sensitizers, are being used to increase the effectiveness of radiotherapy. This review highlights the mechanism responsible for developing radioresistance of rectal cancer following radiotherapy and potential strategies to enhance the effectiveness of radiotherapy for better management of rectal cancer.


Asunto(s)
Fibroblastos Asociados al Cáncer , MicroARNs , Neoplasias Primarias Secundarias , Neoplasias del Recto , Humanos , Fosfatidilinositol 3-Quinasas , Neoplasias del Recto/radioterapia , Inmunoterapia , Microambiente Tumoral
10.
ACS Nano ; 18(11): 8325-8336, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38447099

RESUMEN

Radiotherapy is a mainstay treatment used in clinics for locoregional therapy, although it still represents a great challenge to improve the sensitivity and accuracy of radiotherapy for tumors. Here, we report the conjugated polymer, polydiiododiacetylene (PIDA), with an iodine content of 84 wt %, as a highly effective computed tomography (CT) contrast agent and tumor microenvironment-responsive radiosensitizer. PIDA exhibited several key properties that contribute to the improvement of precision radiotherapy. The integrated PIDA nanofibers confined within the tumor envelope demonstrated amplified CT intensity and prolonged retention, providing an accurate calculation of dose distribution and precise radiation delivery for CT image-guided radiotherapy. Therefore, our strategy pioneers PIDA nanofibers as a bridge to cleverly connect a fiducial marker to guide accurate radiotherapy and a radiosensitizer to improve tumor sensitivity, thereby minimizing potential damage to surrounding tissues and facilitating on-demand therapeutic intervention in tumors.


Asunto(s)
Nanofibras , Neoplasias , Polímero Poliacetilénico , Fármacos Sensibilizantes a Radiaciones , Radioterapia Guiada por Imagen , Humanos , Carbono , Microambiente Tumoral , Fármacos Sensibilizantes a Radiaciones/farmacología , Fármacos Sensibilizantes a Radiaciones/uso terapéutico
11.
Small Methods ; 8(1): e2301131, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37906050

RESUMEN

Radiotherapy (RT) has been a classical therapeutic method of cancer for several decades. It attracts tremendous attention for the precise and efficient treatment of local tumors with stimuli-responsive nanomaterials, which enhance RT. However, there are few systematic reviews summarizing the newly emerging stimuli-responsive mechanisms and strategies used for tumor radio-sensitization. Hence, this review provides a comprehensive overview of recently reported studies on stimuli-responsive nanomaterials for radio-sensitization. It includes four different approaches for sensitized RT, namely endogenous response, exogenous response, dual stimuli-response, and multi stimuli-response. Endogenous response involves various stimuli such as pH, hypoxia, GSH, and reactive oxygen species (ROS), and enzymes. On the other hand, exogenous response encompasses X-ray, light, and ultrasound. Dual stimuli-response combines pH/enzyme, pH/ultrasound, and ROS/light. Lastly, multi stimuli-response involves the combination of pH/ROS/GSH and X-ray/ROS/GSH. By elaborating on these responsive mechanisms and applying them to clinical RT diagnosis and treatment, these methods can enhance radiosensitive efficiency and minimize damage to surrounding normal tissues. Finally, this review discusses the additional challenges and perspectives related to stimuli-responsive nanomaterials for tumor radio-sensitization.


Asunto(s)
Nanoestructuras , Neoplasias , Humanos , Especies Reactivas de Oxígeno , Neoplasias/radioterapia , Neoplasias/tratamiento farmacológico , Nanoestructuras/uso terapéutico
12.
Adv Sci (Weinh) ; 11(7): e2307858, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38063844

RESUMEN

Hypoxia-associated radioresistance in rectal cancer (RC) has severely hampered the response to radioimmunotherapy (iRT), necessitating innovative strategies to enhance RC radiosensitivity and improve iRT efficacy. Here, a catalytic radiosensitizer, DMPtNPS, and a STING agonist, cGAMP, are integrated to overcome RC radioresistance and enhance iRT. DMPtNPS promotes efficient X-ray energy transfer to generate reactive oxygen species, while alleviating hypoxia within tumors, thereby increasing radiosensitivity. Mechanistically, the transcriptomic and immunoassay analysis reveal that the combination of DMPtNPS and RT provokes bidirectional regulatory effects on the immune response, which may potentially reduce the antitumor efficacy. To mitigate this, cGAMP is loaded into DMPtNPS to reverse the negative impact of DMPtNPS and RT on the tumor immune microenvironment (TiME) through the type I interferon-dependent pathway, which promotes cancer immunotherapy. In a bilateral tumor model, the combination treatment of RT, DMPtNPS@cGAMP, and αPD-1 demonstrates a durable complete response at the primary site and enhanced abscopal effect at the distant site. This study highlights the critical role of incorporating catalytic radiosensitizers and STING agonists into the iRT approach for RC.


Asunto(s)
Interferón Tipo I , Nanopartículas , Neoplasias del Recto , Humanos , Radioinmunoterapia , Neoplasias del Recto/terapia , Nanopartículas/uso terapéutico , Hipoxia , Microambiente Tumoral
13.
Int J Mol Sci ; 24(17)2023 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-37686211

RESUMEN

This paper presents an assessment of nuclear reaction yields of protons, α-particles, and neutrons in human tissue-equivalentmaterial in proton therapy using a simulation with Geant 4. In this study, we also check an enhancement of nuclear reactions due to the presence of Bi, Au, 11B, and 10B radiosensitizer nanoparticles. We demonstrate that a proton beam induces a noticeable amount of nuclear reactions in the tissue. Nevertheless, the enhancement of nuclear reaction products due to radiosensitizer nanoparticles is found to be negligible.


Asunto(s)
Nanopartículas , Neoplasias , Fármacos Sensibilizantes a Radiaciones , Humanos , Protones , Neoplasias/radioterapia , Partículas alfa , Simulación por Computador , Fármacos Sensibilizantes a Radiaciones/uso terapéutico
14.
Mol Imaging Biol ; 2023 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-37721686

RESUMEN

PURPOSE: Tumor hypoxia contributes to aggressive phenotypes and diminished therapeutic responses to radiation therapy (RT) with hypoxic tissue being 3-fold less radiosensitive than normoxic tissue. A major challenge in implementing hypoxic radiosensitizers is the lack of a high-resolution imaging modality that directly quantifies tissue-oxygen. The electron paramagnetic resonance oxygen-imager (EPROI) was used to quantify tumor oxygenation in two murine tumor models: E0771 syngeneic transplant breast cancers and primary p53/MCA soft tissue sarcomas, with the latter autochthonous model better recapitulating the tumor microenvironment in human malignancies. We hypothesized that tumor hypoxia differs between these models. We also aimed to quantify the absolute change in tumor hypoxia induced by the mitochondrial inhibitor papaverine (PPV) and its effect on RT response. PROCEDURES: Tumor oxygenation was characterized in E0771 and primary p53/MCA sarcomas via EPROI, with the former model also being quantified indirectly via diffuse reflectance spectroscopy (DRS). After confirming PPV's effect on hypoxic fraction (via EPROI), we compared the effect of 0 versus 2 mg/kg PPV prior to 20 Gy on tumor growth delay and survival. RESULTS: Hypoxic sarcomas were more radioresistant than normoxic sarcomas (p=0.0057, 2-way ANOVA), and high baseline hypoxic fraction was a significant (p=0.0063, Cox Regression Model) hazard in survivability regardless of treatment. Pre-treatment with PPV before RT did not radiosensitize tumors in the sarcoma or E0771 model. In the sarcoma model, EPROI successfully identified baseline hypoxic tumors. DRS quantification of total hemoglobin, saturated hemoglobin, changes in mitochondrial potential and glucose uptake showed no significant difference in E0771 tumors pre- and post-PPV. CONCLUSION: EPROI provides 3D high-resolution pO2 quantification; EPR is better suited than DRS to characterize tumor hypoxia. PPV did not radiosensitize E0771 tumors nor p53/MCA sarcomas, which may be related to the complex pattern of vasculature in each tumor. Additionally, understanding model-dependent tumor hypoxia will provide a much-needed foundation for future therapeutic studies with hypoxic radiosensitizers.

15.
Int J Nanomedicine ; 18: 4449-4470, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37555189

RESUMEN

Radiopharmaceuticals serve as a major part of nuclear medicine contributing to both diagnosis and treatment of several diseases, especially cancers. Currently, most radiopharmaceuticals are based on small molecules with targeting ability. However, some concerns over their stability or non-specific interactions leading to off-target localization are among the major challenges that need to be overcome. Emulsion technology has great potential for the fabrication of carrier systems for radiopharmaceuticals. It can be used to create particles with different compositions, structures, sizes, and surface characteristics from a wide range of generally recognized as safe (GRAS) materials, which allows their functionality to be tuned for specific applications. In particular, it is possible to carry out surface modifications to introduce targeting and stealth properties, as well as to control the particle dimensions to manipulate diffusion and penetration properties. Moreover, emulsion preparation methods are usually simple, economic, robust, and scalable, which makes them suitable for medical applications. In this review, we highlight the potential of emulsion technology in nuclear medicine for developing targeted radionuclide therapies, for use as radiosensitizers, and for application in radiotracer delivery in gamma imaging techniques.


Asunto(s)
Medicina Nuclear , Fármacos Sensibilizantes a Radiaciones , Radiofármacos , Emulsiones/química , Tecnología , Radioisótopos
16.
Int J Mol Sci ; 24(15)2023 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-37569898

RESUMEN

Pancreatic cancer stands among the deadliest forms of cancer, and the existing treatments fall short of providing adequate efficacy. Novel and more effective treatment approaches are urgently required to address this critical medical challenge. In this study, we aimed to evaluate the anti-cancer efficacy of gold nanoparticles (GNPs) in combination with radiotherapy (RT). A 3D pancreatic cancer co-culture spheroid model of MIA PaCa-2 cancer cells and patient-derived cancer-associated fibroblasts (CAF-98) was used. The spheroids were treated with GNPs (7.5 µg/mL) and 2 Gy of RT. The spheroids' cell viability was assessed through the CellTiter-Glo 3D assay, and an immunofluorescence assay was used to assess the DNA DSBs via the expression of the DNA damage marker 53BP1. Co-culture samples showed a 10.8% (p < 0.05) increase in proliferation and a 13.0% (p < 0.05) decrease in DNA DSB when compared to monoculture samples, However, they displayed a 175% (p < 0.001) increase in GNPs uptake when compared to monoculture spheroids. Using GNPs/RT, we were able to show a significant reduction of 6.2% (p < 0.05) in spheroid size and an increase of 14.3% (p < 0.05) in DNA DSB damage in co-culture samples. The combination of GNPs with RT demonstrated remarkable radiosensitization effects, representing a promising approach to enhance cancer treatment efficacy. These effects were particularly noteworthy in the more treatment-resistant co-culture spheroid model.


Asunto(s)
Nanopartículas del Metal , Neoplasias Pancreáticas , Fármacos Sensibilizantes a Radiaciones , Humanos , Técnicas de Cocultivo , Oro/farmacología , Estudios Prospectivos , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias Pancreáticas/radioterapia , ADN , Neoplasias Pancreáticas
17.
Int J Mol Sci ; 24(15)2023 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-37569557

RESUMEN

In this study, we present a complete set of electron scattering cross-sections from 1-Methyl-5-Nitroimidazole (1M5NI) molecules for impact energies ranging from 0.1 to 1000 eV. This information is relevant to evaluate the potential role of 1M5NI as a molecular radiosensitizers. The total electron scattering cross-sections (TCS) that we previously measured with a magnetically confined electron transmission apparatus were considered as the reference values for the present analysis. Elastic scattering cross-sections were calculated by means of two different schemes: The Schwinger multichannel (SMC) method for the lower energies (below 15 eV) and the independent atom model-based screening-corrected additivity rule with interferences (IAM-SCARI) for higher energies (above 15 eV). The latter was also applied to calculate the total ionization cross-sections, which were complemented with experimental values of the induced cationic fragmentation by electron impact. Double differential ionization cross-sections were measured with a reaction microscope multi-particle coincidence spectrometer. Using a momentum imaging spectrometer, direct measurements of the anion fragment yields and kinetic energies by the dissociative electron attachment are also presented. Cross-sections for the other inelastic channels were derived with a self-consistent procedure by sampling their values at a given energy to ensure that the sum of the cross-sections of all the scattering processes available at that energy coincides with the corresponding TCS. This cross-section data set is ready to be used for modelling electron-induced radiation damage at the molecular level to biologically relevant media containing 1M5NI as a potential radiosensitizer. Nonetheless, a proper evaluation of its radiosensitizing effects would require further radiobiological experiments.


Asunto(s)
Electrones , Transporte de Electrón , Fenómenos Físicos , Movimiento (Física)
18.
Future Med Chem ; 15(13): 1133-1147, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37529897

RESUMEN

Background: Targeting CDK4/6 has advanced breast cancer treatment. Herein, new quinazolinones were synthesized with acetamide linkers as potential anti-breast cancer agents. Methods & results: In vitro cytotoxic evaluation on human breast cancer cell lines (MCF7 and MDA-MB-231) identified 1,3-benzodioxole (5d) to be of the highest potency. It showed good inhibitory activity on CDK4/6. Compound 5d arrested the cell cycle at the G1-phase, caused induction of early and late apoptosis in an Annexin V-FITC assay, led to an increase in the level of caspase-3 and upregulated Bax expression and downregulated Bcl-2 in MCF7 cells. Compound 5d showed good radiosensitizing activity when combined with a single dose of 8-Gy γ-radiation. Conclusion: This study introduces quinazolinone scaffolds as new CDK4/6 inhibitors for breast cancer.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Humanos , Femenino , Proliferación Celular , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Quinazolinonas/farmacología , Ciclo Celular , Apoptosis , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/metabolismo
19.
Int J Mol Sci ; 24(14)2023 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-37511566

RESUMEN

The efficacy of antitumor radiotherapy can be enhanced by utilizing nonradioactive nanoparticles that emit secondary radiation when activated by a primary beam. They consist of small volumes of a radiosensitizing substance embedded within a polymer layer, which is coated with tumor-specific antibodies. The efficiency of nanosensitizers relies on their successful delivery to the tumor, which depends on their size. Increasing their size leads to a higher concentration of active substance; however, it hinders the penetration of nanosensitizers through tumor capillaries, slows down their movement through the tissue, and accelerates their clearance. In this study, we present a mathematical model of tumor growth and radiotherapy with the use of intravenously administered tumor-specific nanosensitizers. Our findings indicate that their optimal size for achieving maximum tumor radiosensitization following a single injection of their fixed total volume depends on the permeability of the tumor capillaries. Considering physiologically plausible spectra of capillary pore radii, with a nanoparticle polymer layer width of 7 nm, the optimal radius of nanoparticles falls within the range of 13-17 nm. The upper value is attained when considering an extreme spectrum of capillary pores.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Modelos Teóricos , Neoplasias/radioterapia , Neoplasias/irrigación sanguínea , Polímeros
20.
Heliyon ; 9(6): e17252, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37389057

RESUMEN

Radiation therapy against cancer frequently fails to attain the desired outcomes because of several restricting aspects. Radiation therapy is not a targeted antitumor treatment, and it poses serious threats to normal tissues as well. In many cases, some inherent features of tumors make them resistant to radiation therapy. Several nanoparticles have shown the capacity to upgrade the viability of radiation treatment because they can directly interact with ionizing radiation to increase cellular radiation sensitivity. Several types of nanomaterials have been investigated as radio-sensitizers, to improve the efficacy of radiotherapy and overcome radio-resistance including, metal-based nanoparticles, quantum dots, silica-based nanoparticles, polymeric nanoparticles, etc. Despite all this research and development, certain challenges associated with the exploitation of nanoparticles to enhance and improve radiation therapy for cancer treatment are encountered. Potential applications of nanoparticles as radiosensitizers is hindered by the difficulties associated with ensuring their production at a large scale with improved characterizations and because of certain biological challenges. By overcoming the shortcomings of nanoparticles like working on the pharmacokinetics, and physical and chemical characterization, the therapy can be improved. It is expected that in the future more knowledge will be available regarding nanoparticles and their clinical efficacy, leading to the successful development of nanotechnology-based radiation therapies for a variety of cancers. This review highlights the limitations of conventional radiotherapy in cancer treatment and explores the potential of nanotechnology, specifically the use of nanomaterials, to overcome these challenges. It discusses the concept of using nanomaterials to enhance the effectiveness of radiation therapy and provides an overview of different types of nanomaterials and their beneficial properties. The review emphasizes the need to address the obstacles and limitations associated with the application of nanotechnology in cancer radiation therapy for successful clinical translation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA