scholarly journals EP-1717: Impact of radiation induced cell death kinetics on reoxygenation and tumour response

2016 ◽  
Vol 119 ◽  
pp. S803
Author(s):  
A. Gago-Arias ◽  
I. Espinoza ◽  
B. Sánchez-Nieto ◽  
J. Pardo-Montero
2017 ◽  
Author(s):  
Thomas D. Lewin ◽  
Philip K Maini ◽  
Eduardo G Moros ◽  
Heiko Enderling ◽  
Helen M Byrne

AbstractCurrent protocols for delivering radiotherapy are based primarily on tumour stage and nodal and metastases status, even though it is well known that tumours and their microenvironments are highly heterogeneous. It is well established that the local oxygen tension plays an important role in radiation-induced cell death, with hypoxic tumour regions responding poorly to irradiation. Therefore, to improve radiation response, it is important to understand more fully the spatiotemporal distribution of oxygen within a growing tumour before and during fractionated radiation. To this end, we have extended a spatially-resolved mathematical model of tumour growth first proposed by Greenspan (Stud. Appl. Math., 1972) to investigate the effects of oxygen heterogeneity on radiation-induced cell death. In more detail, cell death due to radiation at each location in the tumour, as determined by the well-known linear-quadratic model, is assumed also to depend on the local oxygen concentration. The oxygen concentration is governed by a reaction-diffusion equation that is coupled to an integro-differential equation that determines the size of the assumed spherically-symmetric tumour. We combine numerical and analytical techniques to investigate radiation response of tumours with different intra-tumoral oxygen distribution profiles. Model simulations reveal a rapid transient increase in hypoxia upon re-growth of the tumour spheroid post-irradiation. We investigate the response to different radiation fractionation schedules and identify a tumour-specific relationship between inter-fraction time and dose per fraction to achieve cure. The rich dynamics exhibited by the model suggest that spatial heterogeneity may be important for predicting tumour response to radiotherapy for clinical applications.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Jiraporn Kantapan ◽  
Siwaphon Paksee ◽  
Aphidet Duangya ◽  
Padchanee Sangthong ◽  
Sittiruk Roytrakul ◽  
...  

Abstract Background Radioresistance can pose a significant obstacle to the effective treatment of breast cancers. Epithelial–mesenchymal transition (EMT) is a critical step in the acquisition of stem cell traits and radioresistance. Here, we investigated whether Maprang seed extract (MPSE), a gallotannin-rich extract of seed from Bouea macrophylla Griffith, could inhibit the radiation-induced EMT process and enhance the radiosensitivity of breast cancer cells. Methods Breast cancer cells were pre-treated with MPSE before irradiation (IR), the radiosensitizing activity of MPSE was assessed using the colony formation assay. Radiation-induced EMT and stemness phenotype were identified using breast cancer stem cells (CSCs) marker (CD24−/low/CD44+) and mammosphere formation assay. Cell motility was determined via the wound healing assay and transwell migration. Radiation-induced cell death was assessed via the apoptosis assay and SA-β-galactosidase staining for cellular senescence. CSCs- and EMT-related genes were confirmed by real-time PCR (qPCR) and Western blotting. Results Pre-treated with MPSE before irradiation could reduce the clonogenic activity and enhance radiosensitivity of breast cancer cell lines with sensitization enhancement ratios (SERs) of 2.33 and 1.35 for MCF7 and MDA-MB231cells, respectively. Pretreatment of breast cancer cells followed by IR resulted in an increased level of DNA damage maker (γ-H2A histone family member) and enhanced radiation-induced cell death. Irradiation induced EMT process, which displayed a significant EMT phenotype with a down-regulated epithelial marker E-cadherin and up-regulated mesenchymal marker vimentin in comparison with untreated breast cancer cells. Notably, we observed that pretreatment with MPSE attenuated the radiation-induced EMT process and decrease some stemness-like properties characterized by mammosphere formation and the CSC marker. Furthermore, pretreatment with MPSE attenuated the radiation-induced activation of the pro-survival pathway by decrease the expression of phosphorylation of ERK and AKT and sensitized breast cancer cells to radiation. Conclusion MPSE enhanced the radiosensitivity of breast cancer cells by enhancing IR-induced DNA damage and cell death, and attenuating the IR-induced EMT process and stemness phenotype via targeting survival pathways PI3K/AKT and MAPK in irradiated breast cancer cells. Our findings describe a novel strategy for increasing the efficacy of radiotherapy for breast cancer patients using a safer and low-cost natural product, MPSE.


2003 ◽  
Vol 1593 (2-3) ◽  
pp. 219-229 ◽  
Author(s):  
Yoon-Jin Lee ◽  
Jae-Won Soh ◽  
Doo-Il Jeoung ◽  
Chul-Koo Cho ◽  
Gil Ja Jhon ◽  
...  

Genetics ◽  
1972 ◽  
Vol 72 (4) ◽  
pp. 777-782
Author(s):  
A V Carrano

ABSTRACT A formula, based on the Poisson distribution of radiation-induced chromosomal deletions, was derived to predict the frequency of transmission of acentric fragments between subsequent mitoses. The frequency of deletions observed in the i  th + 1 division subsequent to fragment distribution at the i  th division anaphase is independent of the cell death resulting from fragment loss. Further, the transmission frequency of chromosome acentric fragments is mathematically equal to the fragment frequency observed in the i  th + 1 generation divided by the mean fragment frequency in the i  th generation. The formula was also extended to chromatid deletions.


2018 ◽  
Vol 447 (1-2) ◽  
pp. 9-19 ◽  
Author(s):  
Poonam Malhotra ◽  
Ashutosh K. Gupta ◽  
Darshana Singh ◽  
Saurabh Mishra ◽  
Shravan K. Singh ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document