Статті в журналах з теми "Radiation induced bystander effect; non-targeted effect in radiotherapy"

Щоб переглянути інші типи публікацій з цієї теми, перейдіть за посиланням: Radiation induced bystander effect; non-targeted effect in radiotherapy.

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся з топ-36 статей у журналах для дослідження на тему "Radiation induced bystander effect; non-targeted effect in radiotherapy".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Переглядайте статті в журналах для різних дисциплін та оформлюйте правильно вашу бібліографію.

1

Jokar, Safura, Inês A. Marques, Saeedeh Khazaei, Tania Martins-Marques, Henrique Girao, Mafalda Laranjo, and Maria Filomena Botelho. "The Footprint of Exosomes in the Radiation-Induced Bystander Effects." Bioengineering 9, no. 6 (May 31, 2022): 243. http://dx.doi.org/10.3390/bioengineering9060243.

Повний текст джерела
Анотація:
Radiation therapy is widely used as the primary treatment option for several cancer types. However, radiation therapy is a nonspecific method and associated with significant challenges such as radioresistance and non-targeted effects. The radiation-induced non-targeted effects on nonirradiated cells nearby are known as bystander effects, while effects far from the ionising radiation-exposed cells are known as abscopal effects. These effects are presented as a consequence of intercellular communications. Therefore, a better understanding of the involved intercellular signals may bring promising new strategies for radiation risk assessment and potential targets for developing novel radiotherapy strategies. Recent studies indicate that radiation-derived extracellular vesicles, particularly exosomes, play a vital role in intercellular communications and may result in radioresistance and non-targeted effects. This review describes exosome biology, intercellular interactions, and response to different environmental stressors and diseases, and focuses on their role as functional mediators in inducing radiation-induced bystander effect (RIBE).
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Mairs, Robert J., Natasha E. Fullerton, Michael R. Zalutsky, and Marie Boyd. "Targeted Radiotherapy: Microgray Doses and the Bystander Effect." Dose-Response 5, no. 3 (July 1, 2007): dose—response.0. http://dx.doi.org/10.2203/dose-response.07-002.mairs.

Повний текст джерела
Анотація:
Indirect effects may contribute to the efficacy of radiotherapy by sterilizing malignant cells that are not directly irradiated. However, little is known of the influence of indirect effects in targeted radionuclide treatment. We compared γ-radiation-induced bystander effects with those resulting from exposure to three radiohaloanalogues of meta-iodobenzylguanidine (MIBG): [131I]MIBG (low linear energy transfer (LET) β-emitter), [123I]MIBG (high LET Auger electron emitter), and meta-[211At]astatobenzylguanidine ([211At]MABG) (high LET α-emitter). Cells exposed to media from γ-irradiated cells exhibited a dose-dependent reduction in survival fraction at low dosage and a plateau in cell kill at > 2 Gy. Cells treated with media from [131I]MIBG demonstrated a dose-response relationship with respect to clonogenic cell death and no annihilation of this effect at high radiopharmaceutical dosage. In contrast, cells receiving media from cultures treated with [211At]MABG or [123I]MIBG exhibited dose-dependent toxicity at low dose but elimination of cytotoxicity with increasing radiation dose (i.e. U-shaped survival curves). Therefore radionuclides emitting high LET radiation may elicit toxic or protective effects on neighboring untargeted cells at low and high dose respectively. We conclude that radiopharmaceutical-induced bystander effects may depend on LET and be distinct from those elicited by conventional radiotherapy.
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Fernandez-Palomo, Cristian, Zacharenia Nikitaki, Valentin Djonov, Alexandros G. Georgakilas, and Olga A. Martin. "Non-Targeted Effects of Synchrotron Radiation: Lessons from Experiments at the Australian and European Synchrotrons." Applied Sciences 12, no. 4 (February 17, 2022): 2079. http://dx.doi.org/10.3390/app12042079.

Повний текст джерела
Анотація:
Studies have been conducted at synchrotron facilities in Europe and Australia to explore a variety of applications of synchrotron X-rays in medicine and biology. We discuss the major technical aspects of the synchrotron irradiation setups, paying specific attention to the Australian Synchrotron (AS) and the European Synchrotron Radiation Facility (ESRF) as those best configured for a wide range of biomedical research involving animals and future cancer patients. Due to ultra-high dose rates, treatment doses can be delivered within milliseconds, abiding by FLASH radiotherapy principles. In addition, a homogeneous radiation field can be spatially fractionated into a geometric pattern called microbeam radiotherapy (MRT); a coplanar array of thin beams of microscopic dimensions. Both are clinically promising radiotherapy modalities because they trigger a cascade of biological effects that improve tumor control, while increasing normal tissue tolerance compared to conventional radiation. Synchrotrons can deliver high doses to a very small volume with low beam divergence, thus facilitating the study of non-targeted effects of these novel radiation modalities in both in-vitro and in-vivo models. Non-targeted radiation effects studied at the AS and ESRF include monitoring cell–cell communication after partial irradiation of a cell population (radiation-induced bystander effect, RIBE), the response of tissues outside the irradiated field (radiation-induced abscopal effect, RIAE), and the influence of irradiated animals on non-irradiated ones in close proximity (inter-animal RIBE). Here we provide a summary of these experiments and perspectives on their implications for non-targeted effects in biomedical fields.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Trott, Klaus-Rüdiger. "Non-Targeted Radiation Effects in Radiotherapy &Roles of Radiation-Induced Genomic Instability and of the Bystander Effect in Cancer Cure by Radiotherapy." Acta Oncologica 40, no. 8 (January 2001): 976–80. http://dx.doi.org/10.1080/02841860152708260.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Hara, Daiki, Wensi Tao, Ryder M. Schmidt, Yu-Ping Yang, Sylvia Daunert, Nesrin Dogan, John Chetley Ford, Alan Pollack, and Junwei Shi. "Boosted Radiation Bystander Effect of PSMA-Targeted Gold Nanoparticles in Prostate Cancer Radiosensitization." Nanomaterials 12, no. 24 (December 14, 2022): 4440. http://dx.doi.org/10.3390/nano12244440.

Повний текст джерела
Анотація:
Metal nanoparticles are effective radiosensitizers that locally enhance radiation doses in targeted cancer cells. Compared with other metal nanoparticles, gold nanoparticles (GNPs) exhibit high biocompatibility, low toxicity, and they increase secondary electron scatter. Herein, we investigated the effects of active-targeting GNPs on the radiation-induced bystander effect (RIBE) in prostate cancer cells. The impact of GNPs on the RIBE presents implications for secondary cancers or spatially fractionated radiotherapy treatments. Anti-prostate-specific membrane antigen (PSMA) antibodies were conjugated with PEGylated GNPs through EDC–NHS chemistry. The media transfer technique was performed to induce the RIBE on the non-irradiated bystander cells. This study focused on the LNCaP cell line, because it can model a wide range of stages relating to prostate cancer progression, including the transition from androgen dependence to castration resistance and bone metastasis. First, LNCaP cells were pretreated with phosphate buffered saline (PBS) or PSMA-targeted GNPs (PGNPs) for 24 h and irradiated with 160 kVp X-rays (0–8 Gy). Following that, the collected culture media were filtered (sterile 0.45 µm polyethersulfone) in order to acquire PBS- and PGNP- conditioned media (CM). Then, PBS- and PGNP-CM were transferred to the bystander cells that were loaded with/without PGNPs. MTT, γ-H2AX, clonogenic assays and reactive oxygen species assessments were performed to compare RIBE responses under different treatments. Compared with 2 Gy-PBS-CM, 8 Gy-PBS-CM demonstrated a much higher RIBE response, thus validating the dose dependence of RIBE in LNCaP cells. Compared with PBS-CM, PGNP-CM exhibited lower cell viability, higher DNA damage, and a smaller survival fraction. In the presence of PBS-CM, bystander cells loaded with PGNPs showed increased cell death compared with cells that did not have PGNPs. These results demonstrate the PGNP-boosted expression and sensitivity of RIBE in prostate cancer cells.
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Qin, Feng, Guodong Chen, Kwan Yu, Miaomiao Yang, Wei Cao, Peizhong Kong, Shengjie Peng, Mingyu Sun, Lili Nie та Wei Han. "Golgi Phosphoprotein 3 Mediates Radiation-Induced Bystander Effect via ERK/EGR1/TNF-α Signal Axis". Antioxidants 11, № 11 (1 листопада 2022): 2172. http://dx.doi.org/10.3390/antiox11112172.

Повний текст джерела
Анотація:
The radiation-induced bystander effect (RIBE), an important non-targeted effect of radiation, has been proposed to be associated with irradiation-caused secondary cancers and reproductive damage beyond the irradiation-treated area after radiotherapy. However, the mechanisms for RIBE signal(s) regulation and transduction are not well understood. In the present work, we found that a Golgi protein, GOLPH3, was involved in RIBE transduction. Knocking down GOLPH3 in irradiated cells blocked the generation of the RIBE, whereas re-expression of GOLPH3 in knockdown cells rescued the RIBE. Furthermore, TNF-α was identified as an important intercellular signal molecule in the GOLPH3-mediated RIBE. A novel signal axis, GOLPH3/ERK/EGR1, was discovered to modulate the transcription of TNF-α and determine the level of released TNF-α. Our findings provide new insights into the molecular mechanism of the RIBE and a potential target for RIBE modulation.
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Jasmer, Kimberly J., Kristy E. Gilman, Kevin Muñoz Forti, Gary A. Weisman, and Kirsten H. Limesand. "Radiation-Induced Salivary Gland Dysfunction: Mechanisms, Therapeutics and Future Directions." Journal of Clinical Medicine 9, no. 12 (December 18, 2020): 4095. http://dx.doi.org/10.3390/jcm9124095.

Повний текст джерела
Анотація:
Salivary glands sustain collateral damage following radiotherapy (RT) to treat cancers of the head and neck, leading to complications, including mucositis, xerostomia and hyposalivation. Despite salivary gland-sparing techniques and modified dosing strategies, long-term hypofunction remains a significant problem. Current therapeutic interventions provide temporary symptom relief, but do not address irreversible glandular damage. In this review, we summarize the current understanding of mechanisms involved in RT-induced hyposalivation and provide a framework for future mechanistic studies. One glaring gap in published studies investigating RT-induced mechanisms of salivary gland dysfunction concerns the effect of irradiation on adjacent non-irradiated tissue via paracrine, autocrine and direct cell–cell interactions, coined the bystander effect in other models of RT-induced damage. We hypothesize that purinergic receptor signaling involving P2 nucleotide receptors may play a key role in mediating the bystander effect. We also discuss promising new therapeutic approaches to prevent salivary gland damage due to RT.
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Ma, Linlin, Zhujing Ye, Yarui Zhang, Wenyu Shi, Jingdong Wang, and Hongying Yang. "Irradiated microvascular endothelial cells may induce bystander effects in neural stem cells leading to neurogenesis inhibition." Journal of Radiation Research 63, no. 2 (January 21, 2022): 192–201. http://dx.doi.org/10.1093/jrr/rrab125.

Повний текст джерела
Анотація:
Abstract Radiation-induced neurocognitive dysfunction (RIND) has attracted a lot of attention lately due to the significant improvement of the survival of cancer patients after receiving cranial radiotherapy. The detailed mechanisms are not completely understood, but extensive evidence supports an involvement of the inhibition of hippocampal neurogenesis, which may result from radiation-induced depletion of neural stem cells (NSCs) as well as the damage to neurogenic niches. As an important component of neurogenic niches, vascular cells interact with NSCs through different signaling mechanisms, which is similar to the characteristics of radiation-induced bystander effect (RIBE). But whether RIBE is involved in neurogenesis inhibition contributed by the damaged vascular cells is unknown. Thus, the purpose of the present study was to investigate the occurrence of RIBEs in non-irradiated bystander NSCs induced by irradiated bEnd.3 vascular endothelial cells in a co-culture system. The results show that compared with the NSCs cultured alone, the properties of NSCs were significantly affected after co-culture with bEnd.3 cells, and further change was induced without obvious oxidative stress and apoptosis when bEnd.3 cells were irradiated, manifesting as a reduction in the proliferation, neurosphere-forming capability and differentiation potential of NSCs. All these results suggest that the damaged vascular endothelial cells may contribute to neurogenesis inhibition via inducing RIBEs in NSCs, thus leading to RIND.
Стилі APA, Harvard, Vancouver, ISO та ін.
9

Alcaraz, Miguel, Amparo Olivares, Marina Andreu-Gálvez, Daniel Gyingiri Achel, Ana María Mercado, and Miguel Alcaraz-Saura. "Paradoxical Radiosensitizing Effect of Carnosic Acid on B16F10 Metastatic Melanoma Cells: A New Treatment Strategy." Antioxidants 11, no. 11 (October 31, 2022): 2166. http://dx.doi.org/10.3390/antiox11112166.

Повний текст джерела
Анотація:
Carnosic acid (CA) is a phenolic diterpene characterized by its high antioxidant activity; it is used in industrial, cosmetic, and nutritional applications. We evaluated the radioprotective capacity of CA on cells directly exposed to X-rays and non-irradiated cells that received signals from X-ray treated cells (radiation induced bystander effect, RIBE). The genoprotective capacity was studied by in vivo and in vitro micronucleus assays. Radioprotective capacity was evaluated by clonogenic cell survival, MTT, apoptosis and intracellular glutathione assays comparing radiosensitive cells (human prostate epithelium, PNT2) with radioresistant cells (murine metastatic melanoma, B16F10). CA was found to exhibit a genoprotective capacity in cells exposed to radiation (p < 0.001) and in RIBE (p < 0.01). In PNT2 cells, considered as normal cells in our study, CA achieved 97% cell survival after exposure to 20 Gy of X-rays, eliminating 67% of radiation-induced cell death (p < 0.001), decreasing apoptosis (p < 0.001), and increasing the GSH/GSSH ratio (p < 0.01). However, the administration of CA to B16F10 cells decreased cell survival by 32%, increased cell death by 200% (p < 0.001) compared to irradiated cells, and increased cell death by 100% (p < 0.001) in RIBE bystander cells (p < 0.01). Furthermore, it increased apoptosis (p < 0.001) and decreased the GSH/GSSG ratio (p < 0.01), expressing a paradoxical radiosensitizing effect in these cells. Knowing the potential mechanisms of action of substances such as CA could help to create new applications that would protect healthy cells and exclusively damage neoplastic cells, thus presenting a new desirable strategy for cancer patients in need of radiotherapy.
Стилі APA, Harvard, Vancouver, ISO та ін.
10

Javadi, Seyed Mohammadreza, Mohammad Abdolahad, Solmaz Hashemi, Mohammadali Khayamian, Mohammad Salemizadeh Parizi, Shohreh Vanaei, Hamidreza Mirzaei, et al. "Effect of Post IORT Wound Fluid Secretion (PIWFS) on the Behavior of Breast Cancer Cells: Stimulator or Inhibitor; Report of an Experimental Study on Breast Cancer." Archives of Iranian Medicine 25, no. 2 (February 1, 2022): 78–84. http://dx.doi.org/10.34172/aim.2022.13.

Повний текст джерела
Анотація:
Background: Although investigating the probable side effects of post intraoperative radiotherapy wound fluid secretion (PIWFS) is crucial, especially in clinical cases, no report has been published on the effect of PIWFS on the remaining tumor cells (in the vital state) in cavity side margins or surrounding regions. These tumor cells might be directly/indirectly exposed to intraoperative radiation therapy (IORT). Here, for the first time, we investigated the effect of PIWFS on tumor cells of the same patient extracted from the excised tumor in the spheroid form. Methods: We generated 8 human-derived breast tumor spheroids from 4 patient specimens who received to IORT, dissociated and cultured them in microfluidic devices. The spheroids from each sample were treated with the patients’ PIWFS and DMEM medium separately. Two different parameters, called area and number of detached cells (NDCs), were determined and investigated to evaluate the spheroids’ vital and proliferative states. Results: The results showed severe transformation in tumor spheroids’ function into more invasive and proliferative functions after treatment with PIWFS. Conclusion: Although the radiation-induced bystander effect may have a role in this observation, further experiments must be done to better clarify the probable desired or non-desired effects of post-IORT secretion for both the remaining tumor cells and the surrounding immune cells.
Стилі APA, Harvard, Vancouver, ISO та ін.
11

Yu, Kwan Ngok. "Radiation-Induced Rescue Effect: Insights from Microbeam Experiments." Biology 11, no. 11 (October 23, 2022): 1548. http://dx.doi.org/10.3390/biology11111548.

Повний текст джерела
Анотація:
The present paper reviews a non-targeted effect in radiobiology known as the Radiation-Induced Rescue Effect (RIRE) and insights gained from previous microbeam experiments on RIRE. RIRE describes the mitigation of radiobiological effects in targeted irradiated cells after they receive feedback signals from co-cultured non-irradiated bystander cells, or from the medium previously conditioning those co-cultured non-irradiated bystander cells. RIRE has established or has the potential of establishing relationships with other non-traditional new developments in the fields of radiobiology, including Radiation-Induced Bystander Effect (RIBE), Radiation-Induced Field Size Effect (RIFSE) and ultra-high dose rate (FLASH) effect, which are explained. The paper first introduces RIRE, summarizes previous findings, and surveys the mechanisms proposed for observations. Unique opportunities offered by microbeam irradiations for RIRE research and some previous microbeam studies on RIRE are then described. Some thoughts on future priorities and directions of research on RIRE exploiting unique features of microbeam radiations are presented in the last section.
Стилі APA, Harvard, Vancouver, ISO та ін.
12

Cahoon, Paul, Valentina Giacometti, Francis Casey, Emily Russell, Conor McGarry, Kevin M. Prise, and Stephen J. McMahon. "Investigating spatial fractionation and radiation induced bystander effects: a mathematical modelling approach." Physics in Medicine & Biology 66, no. 22 (November 11, 2021): 225007. http://dx.doi.org/10.1088/1361-6560/ac3119.

Повний текст джерела
Анотація:
Abstract Radiation induced bystander effects (RIBEs) have been shown to cause death in cells receiving little or no physical dose. In standard radiotherapy, where uniform fields are delivered and all cells are directly exposed to radiation, this phenomenon can be neglected. However, the role of RIBEs may become more influential when heterogeneous fields are considered. Mathematical modelling can be used to determine how these heterogeneous fields might influence cell survival, but most established techniques account only for the direct effects of radiation. To gain a full appreciation of how non-uniform fields impact cell survival, it is also necessary to consider the indirect effects of radiation. In this work, we utilise a mathematical model that accounts for both the direct effects of radiation on cells and RIBEs. This model is used to investigate how spatially fractionated radiotherapy plans impact cell survival in vitro. These predictions were compared to survival in normal and cancerous cells following exposure to spatially fractionated plans using a clinical linac. The model is also used to explore how spatially fractionated radiotherapy will impact tumour control in vivo. Results suggest that spatially fractionated plans are associated with higher equivalent uniform doses than conventional uniform plans at clinically relevant doses. The model predicted only small changes changes in normal tissue complication probability, compared to the larger protection seen clinically. This contradicts a central paradigm of radiotherapy where uniform fields are assumed to maximise cell kill and may be important for future radiotherapy optimisation.
Стилі APA, Harvard, Vancouver, ISO та ін.
13

Rita, Ghosh, and Hansda Surajit. "Targeted and non-targeted effects of radiation in mammalian cells: An overview." Archives of Biotechnology and Biomedicine 5, no. 1 (April 12, 2021): 013–19. http://dx.doi.org/10.29328/journal.abb.1001023.

Повний текст джерела
Анотація:
Radiation of different wavelengths can kill living organisms, although, the mechanism of interactions differs depending on their energies. Understanding the interaction of radiation with living cells is important to assess their harmful effects and also to identify their therapeutic potential. Temporally, this interaction can be broadly divided in three stages – physical, chemical and biological. While radiation can affect all the important macromolecules of the cells, particularly important is the damage to its genetic material, the DNA. The consequences of irradiation include- DNA damage, mutation, cross-linkages with other molecules, chromosomal aberrations and DNA repair leading to altered gene expression and/or cell death. Mutations in DNA can lead to heritable changes and is important for the induction of cancer. While some of these effects are through direct interaction of radiation with the target, radiation can interact with the surrounding environment to result in its indirect actions. The effects of radiation depend not only on the total dose but also on the dose rate, LET etc. and also on the cell types. However, action of radiation on organisms is not restricted to interactions with irradiated cells, i.e. target cells alone; it also exerts non-targeted effects on neighboring unexposed cells to produce productive responses; this is known as bystander effect. The bystander effects of ionizing radiations are well documented and contribute largely to the relapse of cancer and secondary tumors after radiotherapy. Irradiation of cells with non-ionizing Ultra-Violet light also exhibits bystander responses, but such responses are very distinct from that produced by ionizing radiations.
Стилі APA, Harvard, Vancouver, ISO та ін.
14

Mohd Siam, Fuaada, and Muhamad Hanis Nasir. "Mechanistic Model of Radiation-Induced Bystander Effects to Cells using Structured Population Approach." MATEMATIKA 34, no. 3 (December 31, 2018): 149–65. http://dx.doi.org/10.11113/matematika.v34.n3.1147.

Повний текст джерела
Анотація:
In irradiation process, instead of traverse on the targeted cells, there is side effect happens to non-targeted cells. The targeted cells that had been irradiated with ionizing radiation emits damaging signal molecules to the surrounding and then, damage the bystander cells. The type of damage considered in this work is the number of double-strand breaks (DSBs) of deoxyribonucleic acid (DNA) in cell’s nucleus. By using mathematical approach, a mechanistic model that can describe this phenomenon is developed based on a structured population approach. Then, the accuracy of the model is validated by its ability to match the experimental data. The Particle Swarm (PS) optimization is employed for the data fitting procedure. PS optimization searches the parameter value that minimize the errors between the model simulation data and experimental data. It is obtained that the mathematical modelling proposed in this paper is strongly in line with the experimental data.
Стилі APA, Harvard, Vancouver, ISO та ін.
15

Meade, Aidan D., Orla Howe, Valérie Unterreiner, Ganesh D. Sockalingum, Hugh J. Byrne, and Fiona M. Lyng. "Vibrational spectroscopy in sensing radiobiological effects: analyses of targeted and non-targeted effects in human keratinocytes." Faraday Discussions 187 (2016): 213–34. http://dx.doi.org/10.1039/c5fd00208g.

Повний текст джерела
Анотація:
Modern models of radiobiological effects include mechanisms of damage initiation, sensing and repair, for those cells that directly absorb ionizing radiation as well as those that experience molecular signals from directly irradiated cells. In the former case, the effects are termed targeted effects while, in the latter, non-targeted effects. It has emerged that phenomena occur at low doses below 1 Gy in directly irradiated cells that are associated with cell-cycle-dependent mechanisms of DNA damage sensing and repair. Likewise in non-targeted bystander-irradiated cells the effect saturates at 0.5 Gy. Both effects at these doses challenge the limits of detection of vibrational spectroscopy. In this paper, a study of the sensing of both targeted and non-targeted effects in HaCaT human keratinocytes irradiated with gamma ray photons is conducted with vibrational spectroscopy. In the case of directly irradiated cells, it is shown that the HaCaT cell line does exhibit both hyperradiosensitivity and increased radioresistance at low doses, a transition between the two effects occurring at a dose of 200 mGy, and that cell survival and other physiological effects as a function of dose follow the induced repair model. Both Raman and FTIR signatures are shown to follow a similar model, suggesting that the spectra include signatures of DNA damage sensing and repair. In bystander-irradiated cells, pro- and anti-apoptotic signalling and mechanisms of ROS damage were inhibited in the mitogen-activated protein kinase (MAPK) transduction pathway. It is shown that Raman spectral profiles of bystander-irradiated cells are correlated with markers of bystander signalling and molecular transduction. This work demonstrates for the first time that both targeted and non-targeted effects of ionizing radiation damage are detected by vibrational spectroscopy in vitro.
Стилі APA, Harvard, Vancouver, ISO та ін.
16

Sprung, Carl N., Marian Cholewa, Noriko Usami, Katsumi Kobayashi, and Jeffrey C. Crosbie. "DNA damage and repair kinetics after microbeam radiation therapy emulation in living cells using monoenergetic synchrotron X-ray microbeams." Journal of Synchrotron Radiation 18, no. 4 (May 14, 2011): 630–36. http://dx.doi.org/10.1107/s0909049511011836.

Повний текст джерела
Анотація:
A novel synchrotron-based approach, known as microbeam radiation therapy (MRT), currently shows considerable promise in increased tumour control and reduced normal tissue damage compared with conventional radiotherapy. Different microbeam widths and separations were investigated using a controlled cell culture system and monoenergetic (5.35 keV) synchrotron X-rays in order to gain further insight into the underlying cellular response to MRT. DNA damage and repair was measured using fluorescent antibodies against phosphorylated histone H2AX, which also allowed us to verify the exact location of the microbeam path. Beam dimensions that reproduced promising MRT strategies were used to identify useful methods to study the underpinnings of MRT. These studies include the investigation of different spatial configurations on bystander effects. γH2AX foci number were robustly induced in directly hit cells and considerable DNA double-strand break repair occurred by 12 h post-10 Gy irradiation; however, many cells had some γH2AX foci at the 12 h time point. γH2AX foci at later time points did not directly correspond with the targeted regions suggesting cell movement or bystander effects as a potential mechanism for MRT effectiveness. Partial irradiation of single nuclei was also investigated and in most cases γH2AX foci were not observed outside the field of irradiation within 1 h after irradiation indicating very little chromatin movement in this time frame. These studies contribute to the understanding of the fundamental radiation biology relating to the MRT response, a potential new therapy for cancer patients.
Стилі APA, Harvard, Vancouver, ISO та ін.
17

Slattery, Ciara, Khanh Nguyen, Laura Shields, Isabel Vega-Carrascal, Sean Singleton, Fiona M. Lyng, Brendan McClean, and Aidan D. Meade. "Application of Advanced Non-Linear Spectral Decomposition and Regression Methods for Spectroscopic Analysis of Targeted and Non-Targeted Irradiation Effects in an In-Vitro Model." International Journal of Molecular Sciences 23, no. 21 (October 26, 2022): 12986. http://dx.doi.org/10.3390/ijms232112986.

Повний текст джерела
Анотація:
Irradiation of the tumour site during treatment for cancer with external-beam ionising radiation results in a complex and dynamic series of effects in both the tumour itself and the normal tissue which surrounds it. The development of a spectral model of the effect of each exposure and interaction mode between these tissues would enable label free assessment of the effect of radiotherapeutic treatment in practice. In this study Fourier-transform Infrared microspectroscopic imaging was employed to analyse an in-vitro model of radiotherapeutic treatment for prostate cancer, in which a normal cell line (PNT1A) was exposed to low-dose X-ray radiation from the scattered treatment beam, and also to irradiated cell culture medium (ICCM) from a cancer cell line exposed to a treatment relevant dose (2Gy). Various exposure modes were studied and reference was made to previously acquired data on cellular survival and DNA double strand break damage. Spectral analysis with manifold methods, linear spectral fitting, non-linear classification and non-linear regression approaches were found to accurately segregate spectra on irradiation type and provide a comprehensive set of spectral markers which differentiate on irradiation mode and cell fate. The study demonstrates that high dose irradiation, low-dose scatter irradiation and radiation-induced bystander exposure (RIBE) signalling each produce differential effects on the cell which are observable through spectroscopic analysis.
Стилі APA, Harvard, Vancouver, ISO та ін.
18

Yokoya, Akinari, and Noriko Usami. "Targeting Specific Sites in Biological Systems with Synchrotron X-Ray Microbeams for Radiobiological Studies at the Photon Factory." Quantum Beam Science 4, no. 1 (January 9, 2020): 2. http://dx.doi.org/10.3390/qubs4010002.

Повний текст джерела
Анотація:
X-ray microbeams have been used to explore radiobiological effects induced by targeting a specific site in living systems. Synchrotron radiation from the Photon Factory, Japan, with high brilliance and highly parallel directionality is a source suitable for delivering a particular beam size or shape, which can be changed according to target morphology by using a simple metal slit system (beam size from 5 μm to several millimeters). Studies have examined the non-targeted effects, called bystander cellular responses, which are thought to be fundamental mechanisms of low-dose or low-dose-rate effects in practical radiation risk research. Narrow microbeams several tens of micrometers or less in their size targeted both the cell nucleus and the cytoplasm. Our method combined with live-cell imaging techniques has challenged the traditional radiobiological dogma that DNA damage is the only major cause of radiation-induced genetic alterations and is gradually revealing the role of organelles, such as mitochondria, in these biological effects. Furthermore, three-dimensionally cultured cell systems have been used as microbeam targets to mimic organs. Combining the spatial fractionation of X-ray microbeams and a unique ex vivo testes organ culture technique revealed that the tissue-sparing effect was induced in response to the non-uniform radiation fields. Spatially fractionated X-ray beams may be a promising tool in clinical radiation therapy.
Стилі APA, Harvard, Vancouver, ISO та ін.
19

Najafi, Masoud, Alireza Shirazi, Elahe Motevaseli, Ghazale Geraily, Peyman Amini, Leila Farhadi Tooli, and Dheyauldeen Shabeeb. "Melatonin Modulates Regulation of NOX2 and NOX4 Following Irradiation in the Lung." Current Clinical Pharmacology 14, no. 3 (December 31, 2019): 224–31. http://dx.doi.org/10.2174/1574884714666190502151733.

Повний текст джерела
Анотація:
Background: Exposure to ionizing radiation may lead to chronic upregulation of inflammatory mediators and pro-oxidant enzymes, which give rise to continuous production of reactive oxygen species (ROS). NADPH oxidases are among the most important ROS producing enzymes. Their upregulation is associated with DNA damage and genomic instability. In the present study, we sought to determine the expressions of NADPH oxidases; NOX2 and NOX4, in rat’s lung following whole body or pelvis irradiation. In addition, we evaluated the protective effect of melatonin on the expressions of NOX2 and NOX4, as well as oxidative DNA injury. Materials and Methods: 35 male rats were divided into 7 groups, G1: control; G2: melatonin (100 mg/kg) treatment; G3: whole body irradiation (2 Gy); G4: melatonin plus whole body irradiation; G5: local irradiation to pelvis area; G6: melatonin treatment plus 2 Gy gamma rays to pelvis area; G7: scatter group. All the rats were sacrificed after 24 h. afterwards, the expressions of TGFβR1, Smad2, NF- κB, NOX2 and NOX4 were detected using real-time PCR. Also, the level of 8-OHdG was detected by ELISA, and NOX2 and NOX4 protein levels were detected by western blot. Results: Whole body irradiation led to the upregulation of all genes, while local pelvis irradiation caused upregulation of TGFβR1, NF-κB, NOX2 and NOX4, as well as protein levels of NOX2 and NOX4. Treatment with melatonin reduced the expressions of these genes and also alleviated oxidative injury in both targeted and non-targeted lung tissues. Results also showed no significant reduction for NOX2 and NOX4 in bystander tissues following melatonin treatment. Conclusion: It is possible that upregulation of NOX2 and NOX4 is involved in radiation-induced targeted and non-targeted lung injury. Melatonin may reduce oxidative stress following upregulation of these enzymes in directly irradiated lung tissues but not for bystander.
Стилі APA, Harvard, Vancouver, ISO та ін.
20

Li, Yinuo, Yoshitaka Matsumoto, Lili Chen, Yu Sugawara, Emiho Oe, Nanami Fujisawa, Mitsuhiro Ebara, and Hideyuki Sakurai. "Smart Nanofiber Mesh with Locally Sustained Drug Release Enabled Synergistic Combination Therapy for Glioblastoma." Nanomaterials 13, no. 3 (January 19, 2023): 414. http://dx.doi.org/10.3390/nano13030414.

Повний текст джерела
Анотація:
This study aims to propose a new treatment model for glioblastoma (GBM). The combination of chemotherapy, molecular targeted therapy and radiotherapy has been achieved in a highly simultaneous manner through the application of a safe, non-toxic, locally sustained drug-releasing composite Nanofiber mesh (NFM). The NFM consisted of biodegradable poly(ε-caprolactone) with temozolomide (TMZ) and 17-allylamino-17-demethoxygeldanamycin (17AAG), which was used in radiation treatment. TMZ and 17AAG combination showed a synergistic cytotoxicity effect in the T98G cell model. TMZ and 17AAG induced a radiation-sensitization effect, respectively. The NFM containing 17AAG or TMZ, known as 17AAG-NFM and TMZ-NFM, enabled cumulative drug release of 34.1% and 39.7% within 35 days. Moreover, 17AAG+TMZ-NFM containing both drugs revealed a synergistic effect in relation to the NFM of a single agent. When combined with radiation, 17AAG+TMZ-NFM induced in an extremely powerful cytotoxic effect. These results confirmed the application of NFM can simultaneously allow multiple treatments to T98G cells. Each modality achieved a significant synergistic effect with the other, leading to a cascading amplification of the therapeutic effect. Due to the superior advantage of sustained drug release over a long period of time, NFM has the promise of clinically addressing the challenge of high recurrence of GBM post-operatively.
Стилі APA, Harvard, Vancouver, ISO та ін.
21

Węgierek-Ciuk, Aneta, Anna Lankoff, Halina Lisowska, Piotr Kędzierawski, Pamela Akuwudike, Lovisa Lundholm, and Andrzej Wojcik. "Cisplatin Reduces the Frequencies of Radiotherapy-Induced Micronuclei in Peripheral Blood Lymphocytes of Patients with Gynaecological Cancer: Possible Implications for the Risk of Second Malignant Neoplasms." Cells 10, no. 10 (October 9, 2021): 2709. http://dx.doi.org/10.3390/cells10102709.

Повний текст джерела
Анотація:
Gynaecologic cancers are common among women and treatment includes surgery, radiotherapy or chemotherapy, where the last two methods induce DNA damage in non-targeted cells like peripheral blood lymphocytes (PBL). Damaged normal cells can transform leading to second malignant neoplasms (SMN) but the level of risk and impact of risk modifiers is not well defined. We investigated how radiotherapy alone or in combination with chemotherapy induce DNA damage in PBL of cervix and endometrial cancer patients during therapy. Blood samples were collected from nine endometrial cancer patients (treatment with radiotherapy + chemotherapy—RC) and nine cervical cancer patients (treatment with radiotherapy alone—R) before radiotherapy, 3 weeks after onset of radiotherapy and at the end of radiotherapy. Half of each blood sample was irradiated ex vivo with 2 Gy of gamma radiation in order to check how therapy influenced the sensitivity of PBL to radiation. Analysed endpoints were micronucleus (MN) frequencies, apoptosis frequencies and cell proliferation index. The results were characterised by strong individual variation, especially the MN frequencies and proliferation index. On average, despite higher total dose and larger fields, therapy alone induced the same level of MN in PBL of RC patients as compared to R. This result was accompanied by a higher level of apoptosis and stronger inhibition of cell proliferation in RC patients. The ex vivo dose induced fewer MN, more apoptosis and more strongly inhibited proliferation of PBL of RC as compared to R patients. These results are interpreted as evidence for a sensitizing effect of chemotherapy on radiation cytotoxicity. The possible implications for the risk of second malignant neoplasms are discussed.
Стилі APA, Harvard, Vancouver, ISO та ін.
22

Kawano, Yuko, Daniel K. Byun, Benjamin J. Frisch, Hiroki Kawano, Mark W. LaMere, Carl J. Johnston, Jaqueline P. Williams, and Laura M. Calvi. "Local Irradiation Induces Systemic Inflammatory Response and Alteration of the Hematopoietic Stem Cell Niche." Blood 134, Supplement_1 (November 13, 2019): 1213. http://dx.doi.org/10.1182/blood-2019-131809.

Повний текст джерела
Анотація:
Radiotherapy is used in the treatment of ~50% of tumors. We and others have reported long-term suppression of hematopoietic stem and progenitor cells (HSPCs) in the setting of total body irradiation; however, it has been shown that even relatively small irradiation volumes can result in systemic adverse events, such as myeloablation and secondary malignancies. The mechanisms underlying these effects are unclear. We hypothesize that localized radiation may activate a systemic inflammatory response that can acutely alter HSPCs and bone marrow microenvironment (BMME) components, including marrow stromal cells (MSCs), thereby contributing to late effects. We therefore established a murine model of targeted irradiation (TR) using a small animal radiation research platform (SARRP). Methods: We administered local irradiation to a single tibia of 6-8 week old C57BL/6 male mice using a single dose of 15 Gy. Subsequently, we analyzed peripheral blood, BM, BM extracellular fluid (BMEF), collagenase-1 digested bone associated cells of both the irradiated (TR) and non-irradiated, contralateral (CONT) tibiae at 2, 6, 48 hours, 1 and 3 weeks post-TR, performing phenotypic (flow cytometry) and cytokine analyses. For all studies, n = 10-13 mice/time point. Results: In the TR tibia at 2 hours, although total cell numbers were unchanged, there was a significant upregulation of inflammatory cytokines (interleukin 1β (IL1b), IL18), chemokines (CXCL2, CXCL10, CCL2, CCL3) and macrophage colony stimulating factor (M-CSF). Of note, most of these changes normalized by 48 hours (M-CSF at 1 week). Changes in mediator expression were followed, at 6 hours post-TR, by significant increases in macrophage (macs) numbers, including CD206 phagocytic macs, neutrophils (PMNs) and cytotoxic lymphocytes, including CD8+ cells expressing CXCR3+, the receptor for CXCL9 and CXCL10. Interestingly, similar to the TR tibia, CXCL2 expression was also increased significantly in the CONT at 2 hours, followed (6 hours) by significant increases in macs and CD8+ cells, suggesting a systemic or abscopal effect. With respect to the effects of radiation on HSPCs, by 6 hours, most of the stem and progenitor cell (HSPC) populations in the TR marrow were significantly decreased; the decrease in long-term-HSCs was delayed until 48 hours post-TR. All populations remained severely depleted until 3 weeks post RT, demonstrating a rapid and sustained effect of TR on all HSPCs within the irradiation volume. In comparison, in the CONT tibia at 6 hours, CD41+ HSCs were expanded; this is consistent with previous demonstrations that CD41+ LT-HSCs expand with inflammatory signals and suggests that TR-induced signals induced a systemic impact on the non-irradiated HSPCs. By 1 week post-radiation, short term-HSCs were significantly decreased in the CONT marrow, likely due to mobilization since CFU-Cs were correspondingly significantly increased in the circulation. Finally, MSCs, previously shown to support HSCs, were found to be significantly increased in the TR tibia starting at 6 hours and peaking at 48 hours post-radiation. Surprisingly, MSCs were also expanded in the CONT marrow at 48 hours; this expansion was likely associated with the increased CXCL12 levels seen in both TR and CONT marrow, although the CXCL12 levels were higher in the irradiated tibia. Taken together, these changes indicate TR-induced global disruption of the HSC niche. Furthermore, in addition to the transient effects of localized irradiation, we observed a second wave of inflammatory signals, including a significant increase in CCL3, at 1 week post-TR and increased IL1b in the CONT marrow at 3 weeks, changes that may have contributed to the sustained loss of HSPC populations. Conclusions: We present the effects of local irradiation on global hematopoiesis, showing that, in addition to the anticipated acute local changes in the irradiated bone marrow, TR-induced persistent and, more importantly, systemic inflammation. We believe that using this murine model will allow us to dissect the contribution of direct (local) and indirect (systemic) responses to radiation on treatment effects, such as marrow failure and secondary malignancies. Disclosures No relevant conflicts of interest to declare.
Стилі APA, Harvard, Vancouver, ISO та ін.
23

Khaw, Melissa, Zachary Davis, Nicholas Zorko, Greg Berk, Gavin Choy, Margaret MacMillan, Martin Felices, and Jeffrey S. Miller. "Abstract 3435: GTB-5550 (cam16-IL15-camB7H3) trispecific killer engager (TriKE®) drives natural killer cell activation and antibody dependent cellular cytotoxicity against head and neck squamous cell carcinomas." Cancer Research 82, no. 12_Supplement (June 15, 2022): 3435. http://dx.doi.org/10.1158/1538-7445.am2022-3435.

Повний текст джерела
Анотація:
Abstract Worldwide, Head and Neck Squamous Cell Carcinomas (HNSCC) account for about 900,000 cases and 400,000 deaths. In some settings, like Fanconi anemia (FA), patients receive curative treatments (allogeneic stem cell transplantation), only to develop HNSCC in early adulthood at a high rate of incidence. Current treatment strategies for non-FA HNSCC patients include surgery, chemotherapy and radiotherapy. However, these are not viable treatment options for FA HNSCC patients due to their low tolerance for the high toxicity levels of chemotherapy and radiation. Therefore, there is a critical need for novel and targeted therapeutic interventions for the treatment of FA HNSCC patients. B7H3, a checkpoint member of the B7 and CD28 families, is overexpressed on several solid tumors but is absent or not expressed on healthy tissues. It is a promising target for immunotherapy, and recent basket trials, particularly in the prostate cancer, have demonstrated strong clinical signals. Here we developed and tested the ability of GTB-5550, a tri-specific killer engager (TriKE) that includes a B7H3 targeting component, to direct NK cell killing to B7H3-expressing Head and Neck cancer targets. This TriKE molecule includes an NK cell engaging domain containing a humanized camelid nanobody against CD16, a camelid nanobody against B7H3 and a wild type IL-15 sequence between the two engagers. We assessed B7H3 expression by flow cytometry of wild-type HNSCC cells and a paired version with a CRISPER KO of the FANCA gene and determined that the KO had no effect on B7H3 expression. Thus, GTB-5550 activity against HNSCC should be present on both normal HNSCC and FA-HNSCC settings. NK cell responses against HNSCC lines in the presence of GTB-5550 were assessed through either flow cytometry based functional assays, to evaluate NK cell degranulation and cytokine secretion, or IncuCyte imaging assays, to directly assess target killing. NK cell degranulation and IFN-gamma production of GTB-3550-treated samples were higher compared to that of control samples treated with B7H3 single domain or IL-15 alone. GTB-5550 also induced more HNSCC target cell killing by NK cells compared to treatment with the B7H3 single domain or IL-15 alone irrespective of the FANCA gene. Ongoing experiments will evaluate functionality of GTB-5550 on FA patient samples as well as in spheroid assays. Taken together, this data shows that a GTB-5550 is able to drive NK cell activity against B7H3-expressing HNSCC cells, which presents potential for a B7H3-targeted TriKE to be used to be implemented clinically to treat HNSCC or FA-HNSCC patients. Citation Format: Melissa Khaw, Zachary Davis, Nicholas Zorko, Greg Berk, Gavin Choy, Margaret MacMillan, Martin Felices, Jeffrey S. Miller. GTB-5550 (cam16-IL15-camB7H3) trispecific killer engager (TriKE®) drives natural killer cell activation and antibody dependent cellular cytotoxicity against head and neck squamous cell carcinomas [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 3435.
Стилі APA, Harvard, Vancouver, ISO та ін.
24

Yahyapour, R., A. Salajegheh, A. Safari, P. Amini, A. Rezaeyan, A. Amraee, and M. Najafi. "Radiation-induced Non-targeted Effect and Carcinogenesis; Implications in Clinical Radiotherapy." Journal of Biomedical Physics and Engineering 8, no. 4 Dec (September 5, 2018). http://dx.doi.org/10.31661/jbpe.v0i0.713.

Повний текст джерела
Анотація:
Bystander or non-targeted effect is known to be an interesting phenomenon in radiobiology. The genetic consequences of bystander effect on non-irradiated cells have shown that this phenomenon can be considered as one of the most important factors involved in secondary cancer after exposure to ionizing radiation. Every year, millions of people around the world undergo radiotherapy in order to cure different types of cancers. The most crucial aim of radiotherapy is to improve treatment efficiency by reducing early and late effects of exposure to clinical doses of radiation. Secondary cancer induction resulted from exposure to high doses of radiation during treatment can reduce the effectiveness of this modality for cancer treatment. The perception of carcinogenesis risk of bystander effects and factors involved in this phenomenon might help reduce secondary cancer incidence years after radiotherapy. Different modalities such as radiation LET, dose and dose rate, fractionation, types of tissue, gender of patients, etc. may be involved in carcinogenesis risk of bystander effects. Therefore, selecting an appropriate treatment modality may improve cost-effectiveness of radiation therapy as well as the quality of life in survived patients. In this review, we first focus on the carcinogenesis evidence of non-targeted effects in radiotherapy and then review physical and biological factors that may influence the risk of secondary cancer induced by this phenomenon.
Стилі APA, Harvard, Vancouver, ISO та ін.
25

Yahyapour, R., A. Salajegheh, A. Safari, P. Amini, A. Rezaeyan, A. Amraee, and M. Najafi. "Radiation-induced Non-targeted Effect and Carcinogenesis; Implications in Clinical Radiotherapy." Journal of Biomedical Physics and Engineering 8, no. 4Dec (September 5, 2018). http://dx.doi.org/10.31661/jbpe.v8i4dec.713.

Повний текст джерела
Анотація:
Bystander or non-targeted effect is known to be an interesting phenomenon in radiobiology. The genetic consequences of bystander effect on non-irradiated cells have shown that this phenomenon can be considered as one of the most important factors involved in secondary cancer after exposure to ionizing radiation. Every year, millions of people around the world undergo radiotherapy in order to cure different types of cancers. The most crucial aim of radiotherapy is to improve treatment efficiency by reducing early and late effects of exposure to clinical doses of radiation. Secondary cancer induction resulted from exposure to high doses of radiation during treatment can reduce the effectiveness of this modality for cancer treatment. The perception of carcinogenesis risk of bystander effects and factors involved in this phenomenon might help reduce secondary cancer incidence years after radiotherapy. Different modalities such as radiation LET, dose and dose rate, fractionation, types of tissue, gender of patients, etc. may be involved in carcinogenesis risk of bystander effects. Therefore, selecting an appropriate treatment modality may improve cost-effectiveness of radiation therapy as well as the quality of life in survived patients. In this review, we first focus on the carcinogenesis evidence of non-targeted effects in radiotherapy and then review physical and biological factors that may influence the risk of secondary cancer induced by this phenomenon.
Стилі APA, Harvard, Vancouver, ISO та ін.
26

Lobachevsky, Pavel, Helen B. Forrester, Alesia Ivashkevich, Joel Mason, Andrew W. Stevenson, Chris J. Hall, Carl N. Sprung, Valentin G. Djonov, and Olga A. Martin. "Synchrotron X-Ray Radiation-Induced Bystander Effect: An Impact of the Scattered Radiation, Distance From the Irradiated Site and p53 Cell Status." Frontiers in Oncology 11 (May 21, 2021). http://dx.doi.org/10.3389/fonc.2021.685598.

Повний текст джерела
Анотація:
Synchrotron radiation, especially microbeam radiotherapy (MRT), has a great potential to improve cancer radiotherapy, but non-targeted effects of synchrotron radiation have not yet been sufficiently explored. We have previously demonstrated that scattered synchrotron radiation induces measurable γ-H2AX foci, a biomarker of DNA double-strand breaks, at biologically relevant distances from the irradiated field that could contribute to the apparent accumulation of bystander DNA damage detected in cells and tissues outside of the irradiated area. Here, we quantified an impact of scattered radiation to DNA damage response in “naïve” cells sharing the medium with the cells that were exposed to synchrotron radiation. To understand the effect of genetic alterations in naïve cells, we utilised p53-null and p53-wild-type human colon cancer cells HCT116. The cells were grown in two-well chamber slides, with only one of nine zones (of equal area) of one well irradiated with broad beam or MRT. γ-H2AX foci per cell values induced by scattered radiation in selected zones of the unirradiated well were compared to the commensurate values from selected zones in the irradiated well, with matching distances from the irradiated zone. Scattered radiation highly impacted the DNA damage response in both wells and a pronounced distance-independent bystander DNA damage was generated by broad-beam irradiations, while MRT-generated bystander response was negligible. For p53-null cells, a trend for a reduced response to scattered irradiation was observed, but not to bystander signalling. These results will be taken into account for the assessment of genotoxic effects in surrounding non-targeted tissues in preclinical experiments designed to optimise conditions for clinical MRT and for cancer treatment in patients.
Стилі APA, Harvard, Vancouver, ISO та ін.
27

Parisi, Silvana, Ilenia Napoli, Sara Lillo, Alberto Cacciola, Gianluca Ferini, Giuseppe Iatì, Antonio Pontoriero, Consuelo Tamburella, Valerio Davì, and Stefano Pergolizzi. "Spine eburnation in a metastatic lung cancer patient treated with immunotherapy and radiotherapy. The first case report of bystander effect on bone." Journal of Oncology Pharmacy Practice, July 7, 2021, 107815522110273. http://dx.doi.org/10.1177/10781552211027348.

Повний текст джерела
Анотація:
Introduction Metastatic non-small cell lung cancer (NSCLC) is nowadays treated with a multimodal therapeutic approach including immunotherapy, targeted therapy and radiotherapy. Radiation therapy, in addition to immune checkpoint inhibitors, gives rise to a particular radiobiological effect known as “bystander effect” consisting of the radiation-induced damage in nearby unirradiated cells. Case report We report a case of a 79-year-old female patient with stage IV NSCLC treated with concomitant immuno-radiotherapy who showed a bystander effect on bone. Management and outcome: Primary tumour biopsy revealed an adenocarcinoma with a PDL1 expression >50%, while staging exams showed a right pulmonary lesion with a partial involvement of the contiguous rib and a single brain metastasis. The patient refused chemotherapy, so that Pembrolizumab 2 mg/Kg was administered every 3 weeks. After two administrations, the single brain metastasis was treated using stereotactic radiosurgery while the site of primitive lung cancer received an 8 Gy-single fraction 3 D-conformal radiotherapy. Three months after irradiation a chest CT showed a radiological remission of about 10% of the GTV and a partial eburnation of the vertebra located nearby the target volume. The CT images of a PET/CT at six months showed a complete vertebral eburnation. At the last follow-up, the patient was free of disease (brain MRI, spinal MRI and PET/CT). Discussion The present case alerts for unusual side effects provoked by bystander phenomenon in patients treated with a combination of immunotherapy and irradiation. Immune activation exacerbates the bystander effect causing normal tissues toxicities beyond what immunotherapies are causing by themselves.
Стилі APA, Harvard, Vancouver, ISO та ін.
28

Zhang, Jianghong, Yuhong Zhang, Fang Mo, Gaurang Patel, Karl Butterworth, Chunlin Shao та Kevin M. Prise. "The Roles of HIF-1α in Radiosensitivity and Radiation-Induced Bystander Effects Under Hypoxia". Frontiers in Cell and Developmental Biology 9 (25 березня 2021). http://dx.doi.org/10.3389/fcell.2021.637454.

Повний текст джерела
Анотація:
Radiation-induced bystander effects (RIBE) may have potential implications for radiotherapy, yet the radiobiological impact and underlying mechanisms in hypoxic tumor cells remain to be determined. Using two human tumor cell lines, hepatoma HepG2 cells and glioblastoma T98G cells, the present study found that under both normoxic and hypoxic conditions, increased micronucleus formation and decreased cell survival were observed in non-irradiated bystander cells which had been co-cultured with X-irradiated cells or treated with conditioned-medium harvested from X-irradiated cells. Although the radiosensitivity of hypoxic tumor cells was lower than that of aerobic cells, the yield of micronucleus induced in bystander cells under hypoxia was similar to that measured under normoxia indicating that RIBE is a more significant factor in overall radiation damage of hypoxic cells. When hypoxic cells were treated with dimethyl sulfoxide (DMSO), a scavenger of reactive oxygen species (ROS), or aminoguanidine (AG), an inhibitor of nitric oxide synthase (NOS), before and during irradiation, the bystander response was partly diminished. Furthermore, when only hypoxic bystander cells were pretreated with siRNA hypoxia-inducible factor-1α (HIF-1α), RIBE were decreased slightly but if irradiated cells were treated with siRNA HIF-1α, hypoxic RIBE decreased significantly. In addition, the expression of HIF-1α could be increased in association with other downstream effector molecules such as glucose transporter 1 (GLUT-1), vascular endothelial growth factor (VEGF), and carbonic anhydrase (CA9) in irradiated hypoxic cells. However, the expression of HIF-1α expression in bystander cells was decreased by a conditioned medium from isogenic irradiated cells. The current results showed that under hypoxic conditions, irradiated HepG2 and T98G cells showed reduced radiosensitivity by increasing the expression of HIF-1α and induced a syngeneic bystander effect by decreasing the expression of HIF-1α and regulating its downstream target genes in both the irradiated or bystander cells.
Стилі APA, Harvard, Vancouver, ISO та ін.
29

Feller, G., R. A. G. Khammissa, M. S. Nemutandani, and L. Feller. "Biological consequences of cancer radiotherapy in the context of oral squamous cell carcinoma." Head & Face Medicine 17, no. 1 (August 26, 2021). http://dx.doi.org/10.1186/s13005-021-00286-y.

Повний текст джерела
Анотація:
AbstractApproximately 50% of subjects with cancer have been treated with ionizing radiation (IR) either as a curative, adjuvant, neoadjuvant or as a palliative agent, at some point during the clinical course of their disease. IR kills cancer cells directly by injuring their DNA, and indirectly by inducing immunogenic cell killing mediated by cytotoxic T cells; but it can also induce harmful biological responses to non-irradiated neighbouring cells (bystander effect) and to more distant cells (abscopal effect) outside the primary tumour field of irradiation.Although IR can upregulate anti-tumour immune reactions, it can also promote an immunosuppressive tumour microenvironment. Consequently, radiotherapy by itself is seldom sufficient to generate an effective long lasting immune response that is capable to control growth of metastasis, recurrence of primary tumours and development of second primary cancers. Therefore, combining radiotherapy with the use of immunoadjuvants such as immune checkpoint inhibitors, can potentiate IR-mediated anti-tumour immune reactions, bringing about a synergic immunogenic cell killing effect.The purpose of this narrative review is to discuss some aspects of IR-induced biological responses, including factors that contributes to tumour radiosensitivity/radioresistance, immunogenic cell killing, and the abscopal effect.
Стилі APA, Harvard, Vancouver, ISO та ін.
30

Han, Xueqiong, Yixuan Chen, Nan Zhang, Chengyu Huang, Guangyao He, Ting Li, Mengxin Wei, Qiong Song, Shaowen Mo, and Yufeng Lv. "Single-cell mechanistic studies of radiation-mediated bystander effects." Frontiers in Immunology 13 (October 25, 2022). http://dx.doi.org/10.3389/fimmu.2022.849341.

Повний текст джерела
Анотація:
Ionizing radiation (IR) has been widely used in the diagnosis and treatment of clinical diseases, with radiation therapy (RT) being particularly rapid, but it can induce “bystander effects” that lead to biological responses in non-target cells after their neighboring cells have been irradiated. To help clarify how radiotherapy induces these effects, To help clarify how radiotherapy induces these effects, we analyzed single-cell RNA sequencing data from irradiated intestinal tissues on day 1 (T1 state), day 3 (T3 state), day 7 (T7 state), and day 14 (T14 state) after irradiation, as well as from healthy intestinal tissues (T0 state), to reveal the cellular level, molecular level, and involvement of different time irradiated mouse intestinal tissues in biological signaling pathways. In addition, changes in immune cell subpopulations and myeloid cell subpopulations after different radiation times were further explored, and gene regulatory networks (GRNs) of these cell subpopulations were constructed. Cellular communication between radiation-specific immune cells was explored by cell-to-cell communication events. The results suggest that radiotherapy trigger changes in immune cell subsets, which then reprogram the immune ecosystem and mediate systemic bystander effects. These radiation-specific immune cells participate in a wide range of cell-to-cell communication events. In particular, radiation-specific CD8+T cells appear to be at the core of communication and appear to persist in the body after recovery from radiotherapy, with enrichment analysis showing that radiation-specific CD8+ T cells are associated with ferroptosis. Thus, radiation-specific CD8+ T cells may be involved in cellular ferroptosis-mediated adverse effects caused by RT.
Стилі APA, Harvard, Vancouver, ISO та ін.
31

Nasir, Muhamad Hanis, and Fuaada Mohd Siam. "SIMULATION AND SENSITIVITY ANALYSIS ON THE PARAMETER OF NON-TARGETED IRRADIATION EFFECTS MODEL." Jurnal Teknologi 81, no. 1 (November 4, 2018). http://dx.doi.org/10.11113/jt.v81.12448.

Повний текст джерела
Анотація:
Real-life situations showed damage effects on non-targeted cells located in the vicinity of an irradiation region, due to danger signal molecules released by the targeted cells. This effect is widely known as radiation-induced bystander effects (RIBE). The purpose of this paper is to model the interaction of non-targeted cells towards bystander factors released by the irradiated cells by using a system of structured ordinary differential equations. The mathematical model and its simulations are presented in this paper. In the model, the cells are grouped based on the number of double-strand breaks (DSBs) and mis-repair DSBs because the DSBs are formed in non-targeted cells. After performing the model's simulations, the analysis continued with sensitivity analysis. Sensitivity analysis will determine which parameter in the model is the most sensitive to the survival fraction of non-targeted cells. The proposed mathematical model can explain the survival fraction of non-targeted cells affected by the bystander factors.
Стилі APA, Harvard, Vancouver, ISO та ін.
32

Jabbari, Nasrollah, Muhammad Nawaz, and Jafar Rezaie. "Bystander effects of ionizing radiation: conditioned media from X-ray irradiated MCF-7 cells increases the angiogenic ability of endothelial cells." Cell Communication and Signaling 17, no. 1 (December 2019). http://dx.doi.org/10.1186/s12964-019-0474-8.

Повний текст джерела
Анотація:
Abstract Background Non-targeting effects of radiotherapy have become as clinical concern due to secondary tumorigenesis in the patients receiving radiotherapy. Radiotherapy also affects non-tumoral cells present in the tumor microenvironment and surrounding tissues. As such, the irradiated cells are thought to communicate the signals that promote secondary tumorigenesis by affecting the function and fate of non-irradiated cells in the vicinity including endothelial cells. This may include up-regulation of genes in irradiated cells, secretion of paracrine factors and induction of gene expression in surrounding non-irradiated cells, which favor cell survival and secondary tumorigenesis. In the current study, we aimed to investigate whether the conditioned media from X-ray irradiated MCF-7 cells contribute to induction of gene expression in human umbilical vein endothelial cells (HUVECs) in vitro and modulate their angiogenic capability and migration. Methods Following the co-culturing of X-ray irradiated MCF-7 media with HUVECs, the migration and wound healing rate of HUVECs was monitored using Transwell plate and scratch wound healing assay, respectively. The levels of angiogenic protein i.e. vascular endothelial growth factor (VEGF-A) in the conditioned media of MCF-7 cells was measured using ELISA. Additionally, we quantified mRNA levels of VEGFR-2, HSP-70, Ang-2, and Ang-1 genes in HUVECs by real time-PCR. Tubulogenesis capacity of endothelial cells was measured by growth factor reduced Matrigel matrix, whereas expression of CD34 (a marker of angiogenic tip cells) was detected by flow cytometry. Results Data showed that VEGF-A protein content of conditioned media of irradiated MCF-7 cells was increased (P < 0.05) with increase in dose. Data showed that irradiated conditioned media from MCF-7 cells, when incubated with HUVECs, significantly enhanced the cell migration and wound healing rate of HUVECs in a dose-dependent manner (P < 0.05). The mRNA levels of VEGFR-2, HSP-70, Ang-2, and Ang-1 were dose-dependently enhanced in HUVECs incubated with irradiated conditioned media (P < 0.05). Importantly, HUVECs treated with irradiated conditioned media showed a marked increase in the tube formation capability as well as in expression of CD34 marker (P < 0.05). Conclusions Our findings indicate that conditioned media from irradiated MCF-7 cells induce angiogenic responses in endothelial cells in vitro, which could be due to transfer of overexpressed VEGF-A and possibly other factors secreted from irradiated MCF-7 cells to endothelial cells, and induction of intrinsic genes (VEGFR-2, HSP-70, Ang-2, and Ang-1) in endothelial cells. Graphical abstract
Стилі APA, Harvard, Vancouver, ISO та ін.
33

Khozooei, Shayan, Konstanze Lettau, Francesca Barletta, Tina Jost, Simone Rebholz, Soundaram Veerappan, Mirita Franz-Wachtel, et al. "Fisetin induces DNA double-strand break and interferes with the repair of radiation-induced damage to radiosensitize triple negative breast cancer cells." Journal of Experimental & Clinical Cancer Research 41, no. 1 (August 22, 2022). http://dx.doi.org/10.1186/s13046-022-02442-x.

Повний текст джерела
Анотація:
Abstract Background Triple-negative breast cancer (TNBC) is associated with aggressiveness and a poor prognosis. Besides surgery, radiotherapy serves as the major treatment modality for TNBC. However, response to radiotherapy is limited in many patients, most likely because of DNA damage response (DDR) signaling mediated radioresistance. Y-box binding protein-1 (YB-1) is a multifunctional protein that regulates the cancer hallmarks among them resisting to radiotherapy-induced cell death. Fisetin, is a plant flavonol of the flavonoid family of plant polyphenols that has anticancer properties, partially through inhibition of p90 ribosomal S6 kinase (RSK)-mediated YB-1 phosphorylation. The combination of fisetin with radiotherapy has not yet been investigated. Methods Activation status of the RSK signaling pathway in total cell lysate and in the subcellular fractions was analyzed by Western blotting. Standard clonogenic assay was applied to test post-irradiation cell survival. γH2AX foci assay and 3 color fluorescence in situ hybridization analyses were performed to study frequency of double-strand breaks (DSB) and chromosomal aberrations, respectively. The underlying repair pathways targeted by fisetin were studied in cells expressing genomically integrated reporter constructs for the DSB repair pathways via quantifying the expression of green fluorescence protein by flow cytometry. Flow cytometric quantification of sub-G1 cells and the protein expression of LC3-II were employed to measure apoptosis and autophagy, respectively. Kinase array and phosphoproteomics were performed to study the effect of fisetin on DDR response signaling. Results We showed that the effect of fisetin on YB-1 phosphorylation in TNBC cells is comparable to the effect of the RSK pharmacological inhibitors. Similar to ionizing radiation (IR), fisetin induces DSB. Additionally, fisetin impairs repair of IR-induced DSB through suppressing the classical non-homologous end-joining and homologous recombination repair pathways, leading to chromosomal aberration as tested by metaphase analysis. Effect of fisetin on DSB repair was partially dependent on YB-1 expression. Phosphoproteomic analysis revealed that fisetin inhibits DDR signaling, which leads to radiosensitization in TNBC cells, as shown in combination with single dose or fractionated doses irradiation. Conclusion Fisetin acts as a DSB-inducing agent and simultaneously inhibits repair of IR-induced DSB. Thus, fisetin may serve as an effective therapeutic strategy to improve TNBC radiotherapy outcome.
Стилі APA, Harvard, Vancouver, ISO та ін.
34

Tubin, Slavisa, Mohammad K. Khan, Gerardo Salerno, Waleed F. Mourad, Weisi Yan, and Branislav Jeremic. "Mono-institutional phase 2 study of innovative Stereotactic Body RadioTherapy targeting PArtial Tumor HYpoxic (SBRT-PATHY) clonogenic cells in unresectable bulky non-small cell lung cancer: profound non-targeted effects by sparing peri-tumoral immune microenvironment." Radiation Oncology 14, no. 1 (November 26, 2019). http://dx.doi.org/10.1186/s13014-019-1410-1.

Повний текст джерела
Анотація:
Abstract Background Radiotherapy-induced lymphopenia may be limiting the success of therapy and could also negatively affect the ability of immune system in mediating the bystander (BE) and abscopal effects (AE). A novel SBRT-based PArtial Tumor irradiation of HYpoxic clonogenic cells (SBRT-PATHY) for induction of the tumoricidal BE and AE by sparing the peritumoral immune microenvironment and regional circulating lymphocytes has been developed to enhance the radiotherapy therapeutic ratio of advanced lung cancer. The aim of this retrospective review of prospectively collected mono-institutional phase 2 study was to compare the outcomes between unconventional SBRT-PATHY and standard of care in unresectable stage IIIB/IV bulky NSCLC. Materials and methods Sixty patients considered inoperable or unsuitable for radical radio-chemotherapy were enrolled and treated using the following 3 regimens: SBRT-PATHY (group I, n = 20 patients), recommended standard of care chemotherapy (group II, n = 20 patients), and institutional conventional palliative radiotherapy (group III, n = 20 patients). Results Median follow-up was 13 months. The 1-year overall survival was 75, 60, and 20% in groups 1, 2 and 3, respectively (p = 0.099). The 1-year cancer specific survival was 90, 60, and 20% in groups 1, 2, and 3, respectively (p = 0.049). Bulky tumor control rate was 95% for SBRT-PATHY compared with 20% in the other two groups. BE and AE were seen by SBRT-PATHY in 95 and 45% of patients, respectively. Multi-variate analysis for cancer specific survival was significant for treatment effect with SBRT-PATHY (p < 0.001) independent of age, sex, performance status, histology, stage, treated bulky site and tumor diameter. SBRT-PATHY resulted in lower toxicity (p = 0.026), and improved symptom control (p = 0.018) when compared to other two treatment options. Conclusion SBRT-PATHY improved treatment outcomes in unresectable NSCLC and should be investigated in larger trials. Present study has been retrospectively registered on 8th of August 2019 by the ethic committee for Austrian region „Kärnten “in Klagenfurt (AUT), under study number A 31/19.
Стилі APA, Harvard, Vancouver, ISO та ін.
35

Ku, Anthony, Valerie J. Facca, Zhongli Cai, and Raymond M. Reilly. "Auger electrons for cancer therapy – a review." EJNMMI Radiopharmacy and Chemistry 4, no. 1 (October 11, 2019). http://dx.doi.org/10.1186/s41181-019-0075-2.

Повний текст джерела
Анотація:
Abstract Background Auger electrons (AEs) are very low energy electrons that are emitted by radionuclides that decay by electron capture (e.g. 111In, 67Ga, 99mTc, 195mPt, 125I and 123I). This energy is deposited over nanometre-micrometre distances, resulting in high linear energy transfer (LET) that is potent for causing lethal damage in cancer cells. Thus, AE-emitting radiotherapeutic agents have great potential for treatment of cancer. In this review, we describe the radiobiological properties of AEs, their radiation dosimetry, radiolabelling methods, and preclinical and clinical studies that have been performed to investigate AEs for cancer treatment. Results AEs are most lethal to cancer cells when emitted near the cell nucleus and especially when incorporated into DNA (e.g. 125I-IUdR). AEs cause DNA damage both directly and indirectly via water radiolysis. AEs can also kill targeted cancer cells by damaging the cell membrane, and kill non-targeted cells through a cross-dose or bystander effect. The radiation dosimetry of AEs considers both organ doses and cellular doses. The Medical Internal Radiation Dose (MIRD) schema may be applied. Radiolabelling methods for complexing AE-emitters to biomolecules (antibodies and peptides) and nanoparticles include radioiodination (125I and 123I) or radiometal chelation (111In, 67Ga, 99mTc). Cancer cells exposed in vitro to AE-emitting radiotherapeutic agents exhibit decreased clonogenic survival correlated at least in part with unrepaired DNA double-strand breaks (DSBs) detected by immunofluorescence for γH2AX, and chromosomal aberrations. Preclinical studies of AE-emitting radiotherapeutic agents have shown strong tumour growth inhibition in vivo in tumour xenograft mouse models. Minimal normal tissue toxicity was found due to the restricted toxicity of AEs mostly on tumour cells targeted by the radiotherapeutic agents. Clinical studies of AEs for cancer treatment have been limited but some encouraging results were obtained in early studies using 111In-DTPA-octreotide and 125I-IUdR, in which tumour remissions were achieved in several patients at administered amounts that caused low normal tissue toxicity, as well as promising improvements in the survival of glioblastoma patients with 125I-mAb 425, with minimal normal tissue toxicity. Conclusions Proof-of-principle for AE radiotherapy of cancer has been shown preclinically, and clinically in a limited number of studies. The recent introduction of many biologically-targeted therapies for cancer creates new opportunities to design novel AE-emitting agents for cancer treatment. Pierre Auger did not conceive of the application of AEs for targeted cancer treatment, but this is a tremendously exciting future that we and many other scientists in this field envision.
Стилі APA, Harvard, Vancouver, ISO та ін.
36

Sasi, Sharath, Daniel Park, Maria A. Zuriaga, Kenneth Walsh, Xinhua Yan, and David A. Goukassian. "Abstract 134: Low Dose Particle Radiation Affects Long-Term Survival of Bone Marrow Progenitor Cell Populations." Circulation Research 115, suppl_1 (July 18, 2014). http://dx.doi.org/10.1161/res.115.suppl_1.134.

Повний текст джерела
Анотація:
Radiation-induced decreases in the number of bone marrow (BM)-derived endothelial progenitor cell (BM-EPCs) and their lineage precursors which include Early- and Late-Multi-Potent Progenitor cells (E-MPP and L-MPP) could contribute to the pathogenesis of ischemic and vascular diseases. We examined the effect of full-body single dose of proton (1H) at 0.5 Gy, 1 GeV and 0.15 Gy, 1 GeV/nucleon of iron (56Fe) - ionizing radiation (IR) on survival and proliferation of BM-EPCs. The survival of E-MPPs and L-MPPs in the BM after particle IR in C57BL/6 mice were determined at 1, 2, 4, 8, 12, 28 and 40 weeks post-IR. BM-derived mononuclear cells were triple-stained with RAM34 (CD34, c-kit, and Sca1), AC133, and hematopoietic lineage negative cocktail, then sorted by FASC for E- and L-MPP. BM EPCs ex-vivo - There was a transient 2.5-3.5-fold increase in BM-EPC apoptosis, with 3.5-fold increases for 56Fe and 1H at 5hrs and 24hrs, respectively that was no longer detected by day 7. Subsequently, there was a 3-fold increase in BM-EPC apoptosis on day 28 for both ion-IR mice. Compared to 24 hrs, there was a ~20% (1H) and ~45% (56Fe) increase in the rate of EPC proliferation on day 14 that returned to control levels on day 28. BM E-MPP and L-MPP in vivo - Compared to control mice, 1H-IR increased the number of both E-MPPs (665%) and L-MPPs (203%), whereas 56Fe-IR decreased E-MPP (74%) and L-MPPs (65%) at 1 week post-IR, suggesting stimulation by 1H but overt damage by 56Fe in the BM milieu. In 56Fe-IR mice, E-MPPs recovered between 4 and 12 weeks, followed by declines at later time points. In 1H-IR mice, E-MPPs were near control levels up to 4 weeks, but declined at later time points. The long-lasting and cyclical effects of IR on the BM E- and L-MPPs after a single 1H or 56Fe IR dose suggests the presence of prolonged and non-targeted effects in BM milieu, that occur in cells that were not traversed by IR, rather induced by signals from IR cells. Our studies showed that, both 1H- and 56Fe-IR has profound and long-lasting (28-40 months) negative effects on the number of E- and L-MPPs. Future longitudinal studies are necessary to determine whether BM progenitor cells may be affected after terrestrial IR exposure, such as cancer radiotherapy, CT and PET scans, and in astronauts after exploration-type space missions.
Стилі APA, Harvard, Vancouver, ISO та ін.
Ми пропонуємо знижки на всі преміум-плани для авторів, чиї праці увійшли до тематичних добірок літератури. Зв'яжіться з нами, щоб отримати унікальний промокод!

До бібліографії