Open Access

Intraoperative radiotherapy in breast cancer: Alterations to the tumor microenvironment and subsequent biological outcomes (Review)

  • Authors:
    • Yang Yang
    • Xiaochen Hou
    • Shujia Kong
    • Zhuocen Zha
    • Mingqing Huang
    • Chenxi Li
    • Na Li
    • Fei Ge
    • Wenlin Chen
  • View Affiliations

  • Published online on: October 25, 2023     https://doi.org/10.3892/mmr.2023.13118
  • Article Number: 231
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Intraoperative radiotherapy (IORT) is a precise, single high‑dose irradiation directly targeting the tumor bed during surgery. In comparison with traditional external beam RT, it minimizes damage to other normal tissues, ensures an adequate dose to the tumor bed and results in improved cosmetic outcomes and quality of life. Furthermore, IORT offers a shorter treatment duration, lower economic costs and therapeutic efficacy comparable with traditional RT. However, its relatively higher local recurrence rate limits its further clinical applications. Identifying effective radiosensitizing drugs and rational RT protocols will improve its advantages. Furthermore, IORT may not only damage DNA to directly kill breast tumor cells but also alter the tumor microenvironment (TME) to exert a sustained antitumor effect. Specific doses of IORT may exert anti‑angiogenic effects, and consequently antitumor effects, by impacting post‑radiation peripheral blood levels of vascular endothelial growth factor and delta‑like 4. IORT may also modify the postoperative wound fluid composition to continuously inhibit tumor growth, e.g. by reducing components such as microRNA (miR)‑21, miR‑221, miR‑115, oncostatin M, TNF‑β, IL‑6 and IL‑8, and by elevating levels of components such as miR‑223, to inhibit the ability of postoperative wound fluid to induce proliferation, invasion and migration of residual cancer cells. IORT can also modify cancer cell glucose metabolism to inhibit the proliferation of residual tumor cells. In addition, IORT can induce a bystander effect, eliminating the postoperative wound fluid‑induced epithelial‑mesenchymal transition and tumor stem cell phenotype. Insights gained at the molecular level may provide new directions for identifying novel therapeutic targets and approaches. A more comprehensive understanding of the effects of IORT on the breast cancer (BC) TME may further its clinical application. Hence, the present article reviews the primary effects of IORT on BC and its impact on the TME, aiming to offer fresh research perspectives for relevant professionals.

Introduction

Breast cancer (BC) currently ranks as the most common malignancy among women worldwide, posing a significant threat to their health, and in 2020, it accounted for 11.7% of all new cancer cases (1). Previous research has reported that BC progression is determined by tumor cells and influenced by the tumor microenvironment (TME) (2). Furthermore, the TME significantly influences the formation of the metabolic and chemical environment, consequently impacting tumor development and progression (3). The complex and multi-level interactions between BC cells and the stromal cells and immune cells in the TME, through direct contact or secretion of factors, can regulate tumor behavior, promoting metastasis and drug resistance (4). For instance, neutrophils have been shown to promote cancer cell EMT and drug resistance, thereby encouraging metastasis (5).

Breast-conserving surgery (BCS) is a prevalent method utilized for the treatment of early-stage BC (6). In 2018, over 266,000 women were diagnosed with early-stage breast cancer in the US, ~60% of whom chose BCS (7). However, surgery can trigger an inflammatory response, modifying the local microenvironment and increasing the invasiveness of residual cancer cells post-surgery (8). In addition, invasive surgical procedures and the related stress responses may facilitate tumor metastasis through the stimulation of angiogenic factor release and concurrent suppression of cellular immunity (9). Thus, the combination of surgery with chemotherapy, hormone therapy and radiotherapy (RT) has become a common treatment approach for BC (7). In 2018, 63% of stage I or II BC patients underwent BCS and radiation therapy (RT) (7).

Currently, BCS combined with postoperative external beam RT (EBRT) is the standard treatment modality and is typically administered using conventional fractionation, with a total dose of 45–50 Gy. Most patients require a tumor bed boost of 10–16 Gy, with the total treatment duration spanning 3–7 weeks (10). However, in comparison with traditional whole-breast irradiation, intraoperative RT (IORT) involves the delivery of high-dose internal brachytherapy radiation therapy to the postoperative region, including the tumor bed and any remaining lesions (11). Compared to EBRT, IORT only requires a few minutes to deliver the necessary radiation dose during surgery, with lower radioactive side effects, reducing the risks of complications such as infection, breast fibrosis, and dermatitis (12). Additionally, IORT can save about $15,000 in total costs compared to EBRT, alleviating patients' psychological stress and improving their psychological and quality of life (12,13). Therefore, IORT is considered to be an effective method for treating BC in clinical practice (12).

A study has reported that IORT not only eliminates neoplastic cells using radiation but also remodels the TME (14). This remodeling occurs via changes in the constituents and biological roles of the exudate at the surgical location, hindering the proliferation and invasion of tumor cells, thereby diminishing local recurrence (14). The efficacy of IORT was reported to be significantly lower in patients who received IORT after a delayed secondary incision than in those who received IORT during their primary surgery (12). This may be due to factors such as the postoperative shift of the tumor bed and the already established TME, highlighting the result that IORT can influence the reshaping process of the postoperative TME (12). Therefore, gaining a deeper and more comprehensive understanding of the impact of IORT on the TME, and using this knowledge as a foundation to seek more personalized treatment plans or radiosensitizing drugs, may be key to further leveraging the advantages of IORT. The present review aims to summarize a range of biological effects and alterations within the TME induced by IORT for BC. These aspects include direct IORT effects, bystander effects, impacts on tumor angiogenesis, miRNA expression and associated cytokines. The potential directions for future research concerning IORT were also proposed, with the goal of integrating these findings into relevant clinical investigations for more precise personalized treatments. The main content of this article is shown in Fig. 1.

Impact of IORT on angiogenesis

Angiogenesis is a vital process for cancer cell proliferation and dissemination. Certain concentrations of angiogenic factors within tumor tissue have been associated with tumor aggressiveness and patient prognosis (15). Vascular endothelial growth factor (VEGF) is a stimulator of vascular endothelial cell proliferation and angiogenesis, and delta-like 4 (DLL4) serves as a key angiogenesis inhibitor involved in the regulation of vascular maturation and tumor angiogenesis (16). IORT can impact angiogenesis; however, the effect of radiation on angiogenesis is complex, as it can have both anti-angiogenic and pro-angiogenic effects (1720). A study by Nafissi et al (17) assessed the impact of IORT on angiogenesis and reported that patients with BC who underwent BCS and received 21 Gy of intraoperative electron RT (IOERT) experienced a significant decrease in the blood levels of DLL4 and an significant increase in VEGF levels compared with pre-surgery levels. These findings suggest that IOERT may lead to an increase in the formation of new blood vessels following BCS. However, certain studies have reported contradictory results. For instance, Belletti et al (18) reported a decrease in the levels of angiopoietin and VEGF receptor-3 in the wound fluid (WF) of patients following targeted intraoperative RT (TARGIT) treatment, indicating a potential decrease in neovascularization. Kulcenty et al (21) conducted a study on patients who underwent IORT and reported a significant reduction in the expression of the protein IL-8, which promotes the formation of endothelial cells (20), in the postoperative RT-WF of these patients. This finding suggests that IORT exhibits anti-angiogenic effects. In a mouse model assessing BC recurrence, a single high dose of radiation (20 Gy) was reported to decrease the local vascular density within the breast compared to normal breast tissue (22). Furthermore, higher doses of radiation within the range of 2–15 Gy have been reported to exhibit anti-angiogenic effects (23). However, tumors may also respond to radiation by protecting their blood vessels from radiation-induced damage, leading to a paradoxical pro-angiogenic effect (24).

The aforementioned differing results reported may be a consequence of variations in the RT apparatus and the dosages of RT. Thus, the relationship between IORT and angiogenesis in cancer treatment remains to be fully elucidated. However, the research indicates that IORT can potentially induce an anti-angiogenic effect, which may lead to a decrease in proliferation and metastasis of residual cancer cells. These findings provide new directions for future research on IORT treatment for BC, such as the determination of the impact of different radiation doses on treatment efficacy. Currently, in clinical practice, the mainstream radiation standards are based on TARGIT-A (20 Gy low-energy X-ray) and intraoperative irradiation for early BC (ELIOT; 21 Gy electron beam) (12,25). There are no consensus or guidelines for adjusting the dose according to the specific circumstances of the patient (26). In clinical settings, the intricate relationship between IORT and angiogenesis should be considered when deciding whether to use anti-angiogenic drugs postoperatively, as it is crucial to clarify the potential synergistic or attenuating effects it may have with anti-angiogenic medications.

Effect of IORT on micro (mi)RNA expression

miRNAs are a class of non-coding, single-stranded RNA molecules. Certain miRNAs have been identified as oncogenes or proto-oncogenes, making them important therapeutic cancer targets (27). Research has reported that ionizing radiation may lead to an increase or decrease in the miRNA expression profiles in certain cells such as human lung cancer cell lines, and pre-clinical models like mouse models, therefore making miRNA potential therapeutic targets or biomarkers for the radiation response of cancer (28,29).

In a study by Zaleska et al (30), postoperative WF obtained from BCS alone was compared to the RT-WF from patients who received IORT, and a significant downregulation in the expression of miR-21, miR-221 and miR-155 in BC cells treated with RT-WF was reported. Another study indicated that miR-223 was involved in mediating inflammatory responses and also functioned as an oncogene in tumor cells (31). In addition, Fabris et al (32) reported that 41 miRNAs exhibited differential expression in peritumor tissues between patients with BC who only underwent surgery and those who received IORT. Furthermore, the study reported that miR-223 targeted EGF, and overexpression of miR-223 inhibited residual tumor cell proliferation by reducing post-surgical EGF levels and subsequently suppressing the EGFR signal transduction activation, as demonstrated in Fig. 2. Overexpression of miR-21, an miRNA commonly associated with inflammation, has been reported to promote BC cell proliferation and metastasis in vivo (33). Furthermore, low levels of miR-21 are associated with a more favorable prognosis in patients who are HER-2 positive (34). In addition to miR-21, overexpression of miR-221 and miR-155 has also been associated with tumor growth, invasion and metastasis (3537).

In a previous review on miRNA and radiation (28), it was reported that miR-21 expression was upregulated in BC, esophageal cancer and lung cancer tissue samples after RT. While changes in the expression of miR-221 reported were inconsistent among studies, it tended to decrease according to most studies. Qu et al (38) reported that after undergoing surgery, chemotherapy and RT, the plasma levels of miR-155 significantly decreased in patients with BC compared to before treatment. However, the specific effects of RT could not be determined. Based on these data, it may be inferred that IORT serves a pivotal role in regulating miRNA to exert its antitumor effects. In particular, miR-223 is a noteworthy miRNA. It has been reported to serve a key role in BC proliferation, drug resistance and metastasis (38). Therefore, miR-223 is a potential target in BC therapy. However, current research on the relationship between miR-223 and radiation is limited. In summary, the study by Zaleska et al (30) offers a new clinical therapeutic perspective, suggesting that IORT may have synergistic or antagonistic effects when combined with certain miRNA-targeted treatments. Furthermore, the combined use of IORT with drugs targeting miR-223 may enhance the therapeutic effect of IORT, thereby advancing its application.

Epithelial-mesenchymal transition (EMT) of BC cells using IORT elimination of WF

EMT is a biological process where epithelial or endothelial cells transform into mesenchymal cells. During this transition, cells lose their polarity and exhibit downregulation of epithelial markers, including cadherin (CDH)1, epithelial cell adhesion molecule and keratin, whilst upregulating mesenchymal markers such as CDH2, snail family transcriptional repressor 1 (SNAI1) and vimentin (VIM). EMT confers stem cell-like characteristics to mesenchymal cells, enhances tumor cell aggressiveness and promotes tumor dissemination and metastasis (39). Furthermore, EMT is closely associated with the aggressive phenotype of cancer stem cells (CSCs). Kulcenty et al (40) reported that treatment of BC cell lines with WF resulted in an upregulation of mesenchymal markers (CDH2, SNAI1 and VIM) and downregulation of the epithelial marker CDH1 in vitro. Hence, it can be inferred that WF may have the potential to induce EMT in BC cells. However, in the IORT-treated group, both the BC cells and the induced WF group demonstrated higher levels of epithelial markers and lower levels of mesenchymal markers compared to the RT-WF group. These results indicate that IORT attenuated the EMT process induced by postoperative WF in BC cells. In addition, the study reported that radiation-induced bystander effects counteracted the stimulatory influence of WF on the CSC phenotype and EMT in BC cells.

Impact of IORT on adipose stromal cells

Adipocytes are an essential component of the TME both before and after BCS (41). A study reported that adipocytes can interact with tumor cells and contribute to the development, progression and metastasis of BC (42). The mesenchymal stem cells (MSCs) derived from adipose tissue in the breast are referred to as breast adipose stromal cells (bASC) (43). bASCs have the ability to differentiate into cancer-associated fibroblasts and actively participate in the regulation of the TME for tumor cells (44).

In a study by Uhlig et al (45), bASCs exposed to IORT demonstrated a senescent-like morphology, indicating a loss of their ability to proliferate when cultured in vitro. This suggests that IORT may have eliminated the capacity of bASCs to adhere and proliferate, demonstrating the radiosensitivity of bASCs. These findings indicate that IORT may influence the active components of stem cells in the tumor bed, thereby reducing tumor recurrence. A further study by the same group reported that IORT-stimulated mammary bASCs demonstrated significantly reduced proliferation, migration and wound-healing compared with that of the group treated with WF alone (46). In addition, the stimulated MSCs demonstrated significantly lower levels of secretory ‘regulated on activation, normal T cell expressed and secreted’ (RANTES), growth-regulated oncogene α (GROα) and VEGF, suggesting a potential mechanism by which IORT influenced the TME. GROα, one of the secretory factors assessed, has been reported to increase the aggressiveness of triple-negative BC when overexpressed, whilst knockdown of GROα attenuated these effects (47). RANTES, also known as chemokine ligand (CCL)5, serves a significant role in the interaction between MSCs and the tumor stroma, thereby facilitating the metastasis of BC cells (41). The mechanism by which IORT affects MSC function may be associated with the bystander effect it produces.

IORT alters WF composition, expression of associated factors and inflammation

Several studies have demonstrated that surgical removal of the primary tumor from the breast induced a wound-healing response and inflammatory process (8,9). This, in turn, altered the local TME and stimulated the proliferation of remaining cancer cells, promoting tumor recurrence and metastasis. Conversely, IORT mitigated these effects.

IORT reduces the stimulatory effect of WF on tumor cell proliferation

Agresti et al (8) reported that the composition of WF varied based on the pathological type of tumors. The study analyzed 34 cytokines, growth factors and chemokines in the WF of 27 patients undergoing BCS. The increased expression of macrophage inflammatory protein (MIP)-1α, MIP-1β, interferon (IFN)γ-induced protein 10 (IP-10), IL-6, granulocyte colony-stimulating factor (G-CSF), monocyte chemoattractant protein-1/monocyte chemotactic and activating factor and bone bridge proteins were reported in more aggressive tumors, specifically those with HER-2 overexpression. In addition, patients who underwent mastectomy had significantly higher levels of IL-1 receptor antagonist, IL-1β, IFN-γ, IL-6, G-CSF, bone bridging protein, IP-10 and MIP-1β in their WF compared with that of those who underwent partial mastectomy. These factors may promote cancer development and progression, and a study by Belletti et al (18) on the effects of postoperative WF on BC cells reported that the fluid promoted the proliferation, movement and invasiveness of the cells. However, when the patients underwent IORT, the fluid collected from their wounds did not demonstrate these effects. It was reported that the WF obtained from patients treated with IORT had diminished levels of numerous proteins linked to tumor development and movement, such as hepatocyte growth factor, leptin and RANTES. This reduction would have resulted in impaired activation of the signaling pathways leading to STAT3 and p70S6 kinases, ultimately inhibiting tumor growth and metastasis. These results demonstrate that IORT may modulate the abundance of growth factors and cytokines within WF, resulting in antitumor effects. Therefore, IORT may be recommended for highly invasive subtypes of BC.

IORT induces tumor cell apoptosis and inhibits their expansion and spread

Tumor necrosis factor (TNF) is a small protein molecule mainly produced by activated macrophages, NK cells, and T lymphocytes (48). This can lead to natural killer cell dysfunction in BC, resulting in the failure of immunotherapy. Death receptor 5 (DR5; also known as TRAIL receptor 2) is a receptor in TNF that induces cancer cell death via exogenous pathway activation by cystatin proteases (49). Kulcenty et al (19) reported that DR5 protein expression was significantly induced in BC cells treated with RT-WF compared to BC cells treated with WF. By contrast, a previous study demonstrated notably diminished levels of IL-6 in the WF of patients following IORT (18), and this is closely associated with tumor stem cell proliferation and drug resistance (50). In addition, IL-8 is an inflammatory chemokine, associated with the EMT and CSC phenotypes of BC cells (51,52). A notable decrease was observed in RT-WF in the protein expression of both IL-6 and IL-8 after treatment (19,21). This reduction in IL-6 expression led to the inhibition of STAT 3 activity, thereby affecting tumor growth, and it also resulted in decreased EMT and CSC phenotypes (21).

Oncostatin M (OSM) is a key factor in the reprogramming of the TME in BC, promoting tumor progression (53). A study reported that OSM can loosen cell-cell and cell-matrix junctions in BC cells, ultimately leading to increased aggressiveness of tumor cells (54), as demonstrated in Fig. 3. IL-1β exhibits a context-dependent function in cancer progression. A study reported that it promotes the development of bone metastases (55), whilst another has reported that it may hinder the colonization of tumor cells that induce metastasis (56). Therefore, the precise role of IL-1β may depend on its specific context. Wuhrer et al (46) reported a significant decrease in OSM levels and a significant increase in leptin and IL-1β levels in BC cells that underwent RT-WF, compared to BC cells treated with WF. The differences in leptin were hypothesized to be due to the adipose tissue being exposed to radiation (18). In addition, it was reported that RT-WF not only reduced the proliferation of MSCs, but also attenuated wound healing.

The aforementioned findings suggest that IORT has the potential to induce apoptosis in tumor cells and prevent their proliferation and metastasis by influencing the composition of WF constituents, modulating the expression of cytokines and affecting signaling pathways. However, the possibility that these effects may coincide with a prolonged wound recovery process following the surgical procedure should be considered and the specific mechanisms underlying this remain to be elucidated through more in-depth research. Furthermore, more research should be conducted on the relationship between OSM and RT, as the OSM/OSM receptor signaling pathway has been reported to be a critical avenue for remodeling the TME of BC and therefore, inhibition of this pathway may offer a new strategy for treating BC (53). The suppressive effect of IORT on OSM could perhaps be an effective measure to realize this therapeutic strategy.

IORT reduces the incidence of radiation inflammation

In a study with a 4-year follow-up period, a single application of IORT did not have a significant impact on the leukocyte count in peripheral blood samples in comparison with conventional external irradiation (57). Meng et al (58) reported that a single high-dose irradiation was more effective in treating cancer compared with multiple irradiations. It was elucidated that a single high-dose radiation treatment diminished the activation cycle of the autotaxin-lysophosphatidic acid axis. Conversely, multiple irradiations facilitated inflammatory responses, which may have protected cancer cells from cell death induced by radiation. In an in vivo mouse model study, Krall et al (59) reported that the systemic inflammatory response initiated after BCS could augment tumor cell proliferation and this process could potentially lead to recurrent metastasis. However, a reduction in tumor resurgence and relapse among patients with BC who received anti-inflammatory treatment during the perioperative period was also reported.

Direct, bystander and cancer cell metabolism effects of IORT

IORT inhibits the division of cancer cells

A study by Pan et al (60) reported that after in vitro irradiation of BC cells with a single dose of 2/4/6 Gy, the proportion of normal BC cells decreased and the proportion of cells undergoing apoptosis or necrosis increased with increasing doses of irradiation. Furthermore, among the examined cells, there was a rise in the proportion of cells arrested in the G1 phase and a decline in the proportion of cells in the S and G2 phases, indicating an inhibition of the mitotic process. In addition, the total number of newly dead cells gradually increased on days 2 and 3 after RT. Based on the observation of cancer cell survival after four weeks, it was hypothesized that a single dose of brachytherapy may have a long-term inhibitory effect on proliferation and invasion, and a pro-apoptotic effect. Additionally, it was reported that this effect increased with higher doses of brachytherapy.

IORT affects unirradiated cells through bystander effects

In addition to its direct effects, ionizing radiation has the capacity to influence non-irradiated cells situated adjacent to those exposed to radiation. This occurrence is recognized as the radiation-induced bystander effect (RIBE) and is facilitated by intercellular gap junctions, along with the secretion of cytokines and chemokines (61), as illustrated in Fig. 4. Kulcenty et al (40) reported that co-culturing WF-treated BC cells with RT-WF-treated BC cells eliminated the original EMT-inducing effect. Moreover, in vitro scratch assays demonstrated a reduction in the migration of BC cells treated with WF after undergoing RT-WF.

During RT, bystander cells that are not directly exposed to radiation can display effects akin to those of directly irradiated cells, including heightened genomic damage, modified apoptosis rates, increased mutation frequency, DNA damage, diminished cloning efficiency and oncogenic transformation (61). However, there is a distinct difference between the radiation-induced bystander effect and the direct effect of radiation. Al-Abedi et al (62) reported that radiation-generated RIBE enhanced the EMT phenotype of bystander cells and increased the invasiveness of bystander MCF-7 cells. By contrast, Feghhi et al (63) reported that using culture media from electron beam-irradiated cells reduced the survival rate of non-irradiated MCF-7 cells but promoted their migration. It has been hypothesized that, to produce this bystander effect, the radiation dose needs to reach a certain threshold, resulting in an ‘all-or-nothing’ state (61).

The radiation-induced rescue effect (RIRE) is a phenomenon bearing a strong association with RIBE. The occurrence of RIRE permits cells exposed to radiation to derive advantages from signals emitted by nearby, non-irradiated cells. Chen et al (64) reported an improved survival rate for radiation-exposed fibroblasts when grown alongside non-irradiated cells in a shared environment. Similarly, when radiation-exposed HeLa cells were cultivated together with non-irradiated fibroblasts, a reduction in the formation of micronuclei within the HeLa cells was reported. This indicated that bystander cells may rescue irradiated tumor cells through the RIRE, as illustrated in Fig. 4. Consequently, this effect could potentially contribute to the recurrence of residual tumor cells.

In addition, when exposed to radiation stress conditions such as RT, cells can display autophagy-inducing behavior. The induction of autophagy may result in tumor cells transitioning into a reversible dormant condition, allowing them to survive instead of undergoing apoptosis (65,66). However, this behavior may result in later tumor recurrence (66). A study reported that markers of autophagy are notably increased in bystander hepatocellular carcinoma cells exposed to 3 Gy of γ-rays (67). This demonstrates that bystander hepatocellular carcinoma cells can produce an autophagic response.

The mechanism of the RIBE produced during RT is complex. The aforementioned studies demonstrated that diverse cells could generate different biological effects when exposed to varying doses of radiation. However, despite the advantages of IORT, the specific RIBE produced has remained to be fully determined (14,40). Therefore, further research is required to provide an improved understanding of the clinical implications associated with IORT.

Effect of IORT on DNA damage and glucose metabolism in cancer cells

Ionizing radiation primarily damages DNA in cells. Studies conducted by Piotrowski et al (14) and Kulcenty et al (68) reported that both RT with and without RIBE stimulation in cancer cells induced DNA double-strand breaks and heightened the expression of genes responsible for DNA damage repair [such as the genes ERCC excision repair 2, TFIIH core complex helicase subunit (ERCC2), ERCC8 and RAD51 recombinase]. Upon exposure to RT-WF and WF + RIBE stimuli, two DNA repair mechanisms were activated, nucleotide excision repair and homologous recombination.

Moreover, it was reported that BC cells exposed to RT-WF and WF + RIBE treatment exhibited increased oxidative phosphorylation levels in comparison with those treated solely with WF. It has been reported that cancer cells may be more adapted to aerobic glycolysis as their primary means of glucose metabolism instead of oxidative phosphorylation and this phenomenon is known as the Warburg effect (69). During this metabolic process in cancer cells, glucose molecules are converted into pyruvate via glycolysis and then reduced to lactate with the help of lactate dehydrogenase, ultimately leading to a decrease in the pH of the TME (69), as illustrated in Fig. 5. This reduction in pH can enhance the metastatic capacity of cancer cells (70,71). Kulcenty et al (68) hypothesized that it is therefore possible that IORT facilitates a metabolic shift in BC cells from glycolysis to the oxidative phosphorylation pathway, due to direct and bystander effects. The metabolic shift may contribute to a decrease in glucose consumption and lactate secretion in BC cells, which consequently modifies both the cellular metabolism and the pH of the TME. These findings suggest that the alteration of metabolism in cancer cells is another role served by IORT.

Discussion

Recent research from the TARGIT-A and ELIOT trials have reported that the effectiveness of IORT for patients with BC is not inferior to traditional EBRT (12,25). The data from the TARGIT-A trial demonstrated that over an average follow-up period of 8.6 years, the overall mortality rate in the IORT group was significantly lower than that in the EBRT group (3.9 vs. 5.3%; hazard ratio, 0.76), with no significant differences in BC mortality rate and distant metastasis (12). The local recurrence rate in the IORT group was markedly higher than that in the EBRT group (3.3 vs. 1.3%; hazard ratio, 2.55), but this was within an acceptable range. In the TARGIT-A trial, the non-inferiority margin for the difference in local recurrence rates between the two groups was set at 2.5%. In the ELIOT trial (25), after a median follow-up of 12.4 years, the ipsilateral breast tumor recurrence rate was significantly higher in patients receiving single-dose IORET compared to those receiving whole-breast external radiotherapy (12.6 vs. 2.4% at 15 years; hazard ratio, 4.62). However, there was no significant difference in overall survival between the two groups (83.4 vs. 82.4% at 15 years).

The application of IORT has yet to be standardized internationally, primarily due to insufficient large-scale clinical trials and foundational research on this technique. IORT is an RT technique that has become more commonly used over the last 30 years and has filled certain gaps between conventional surgical treatments and traditional EBRT (72). For instance, in the traditional surgical + EBRT treatment, patients are required to endure long periods of psychological distress and complications, whereas implementing IORT is able to markedly shorten patients' treatment cycles, enhance their quality of life and reduce economic losses (73).

However, for optimal therapeutic and economic benefits, whilst ensuring patient quality of life and mental health, integrating IORT with EBRT may be a novel paradigm worth exploring. The ongoing TARGIT-B trial is a large-scale clinical trial evaluating the combination of IORT with post-operative tumor bed boost (74). However, there is still insufficient research supporting the wider clinical application of IORT and therefore, understanding the potential mechanisms of IORT is pivotal for the advancement of this technology.

Furthermore, the present review demonstrated variations in the composition of RT-WF across different studies, possibly due to differences in IORT equipment or radiation doses. Belletti et al (18) reported a notable decrease in hepatocyte growth factor (HGF) and MIP-1α levels and an increase in IL-13 concentration in RT-WF, whereas Kulcenty et al (21) reported a significant increase in HGF and MIP-1α levels and a decrease in the IL-13 concentration in RT-WF, as illustrated in Fig. 6. Thus, it is hypothesized that these changes in the TME may explain the differences in the results of the TARGIT-A and ELIOT trials. Furthermore, WF had significantly higher concentrations of HGF and MIP-1α, and lower levels of IL-13 in both studies. Kulcenty et al (21) additionally noted a marked disparity in the levels of HGF in the RT-WF between patients with luminal A and B BC. The concentrations of small molecules, including IL-9, platelet-derived growth factor-BB, RANTES, TNF-β, CCL2 and CCL7, were reported to differ between the WFs of the two groups. This suggested that there may be differences in how different types of BC respond to IORT.

The aforementioned studies illustrate the intricate biological effects that result from direct irradiation of the tumor bed and surrounding tissues through IORT during surgery, which may be the primary mechanism underlying its antitumor effects. However, the entire biological foundation underlying this phenomenon has remained to be fully elucidated, necessitating additional studies. Detailed molecular-level information obtained from these studies would form a foundation for further investigations into tumor recurrence and metastasis, and for the identification of new therapeutic targets and treatment modalities. Through comprehending the interplay between BC cells and their TME following IORT, novel approaches to BC therapy may be identified.

Lastly, it is noteworthy to mention some limitations in this review. Firstly, due to time constraints, we may not have incorporated the most recent research findings. Secondly, the heterogeneous quality of reviews might lead to a lack of depth or comprehensiveness in the interpretation of certain literature. Additionally, given the constraints in length and focus, this review may not have encompassed all pertinent literature, thereby possibly omitting some crucial information or research.

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant nos. 82060543 and 82060538), the Yunnan Provincial Science and Technology Plan Kunming Medical Joint Special Project (grant no. 202001AY070001 079) and the Middle-aged and Young Academic and Technical Leader Reserve Talent Program of Yunnan Province Science and Technology Department (grant no. 202205AC160008).

Availability of data and materials

Not applicable.

Authors' contributions

YY, XH, SK, ZZ, MH, CL and NL searched the literature. YY, XH and SK wrote the manuscript. ZZ, MH, CL and NL critically revised the manuscript. FG and WC conceived the idea for the review and provided the final approval. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Guo S and Deng CX: Effect of stromal cells in tumor microenvironment on metastasis initiation. Int J Biol Sci. 14:2083–2093. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Simmons A, Burrage PM, Nicolau DV Jr, Lakhani SR and Burrage K: Environmental factors in breast cancer invasion: A mathematical modelling review. Pathology. 49:172–180. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Terceiro LEL, Edechi CA, Ikeogu NM, Nickel BE, Hombach-Klonisch S, Sharif T, Leygue E and Myal Y: The breast tumor microenvironment: A key player in metastatic spread. Cancers (Basel). 13:47982021. View Article : Google Scholar : PubMed/NCBI

5 

Güç E and Pollard JW: Redefining macrophage and neutrophil biology in the metastatic cascade. Immunity. 54:885–902. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Si J, Guo R, Lu X, Han C, Xue L, Xing D and Chen C: Decision aids on breast conserving surgery for early stage breast cancer patients: A systematic review. BMC Med Inform Decis Mak. 20:2752020. View Article : Google Scholar : PubMed/NCBI

7 

Giaquinto AN, Sung H, Miller KD, Kramer JL, Newman LA, Minihan A, Jemal A and Siegel RL: Breast cancer statistics, 2022. CA Cancer J Clin. 72:524–541. 2022. View Article : Google Scholar : PubMed/NCBI

8 

Agresti R, Triulzi T, Sasso M, Ghirelli C, Aiello P, Rybinska I, Campiglio M, Sfondrini L, Tagliabue E and Bianchi F: Wound healing fluid reflects the inflammatory nature and aggressiveness of breast tumors. Cells. 8:1812019. View Article : Google Scholar : PubMed/NCBI

9 

Kim R: Effects of surgery and anesthetic choice on immunosuppression and cancer recurrence. J Transl Med. 16:82018. View Article : Google Scholar : PubMed/NCBI

10 

Feng K, Meng X, Liu J, Xing Z, Zhang M and Wang X, Feng Q and Wang X: Update on intraoperative radiotherapy for early-stage breast cancer. Am J Cancer Res. 10:2032–2042. 2020.PubMed/NCBI

11 

Stoll A, van Oepen A and Friebe M: Intraoperative delivery of cell-killing boost radiation-a review of current and future methods. Minim Invasive Ther Allied Technol. 25:176–187. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Vaidya JS, Bulsara M, Baum M, Wenz F, Massarut S, Pigorsch S, Alvarado M, Douek M, Saunders C, Flyger HL, et al: Long term survival and local control outcomes from single dose targeted intraoperative radiotherapy during lumpectomy (TARGIT-IORT) for early breast cancer: TARGIT-A randomised clinical trial. BMJ. 370:m28362020. View Article : Google Scholar : PubMed/NCBI

13 

Eisavi M, Rezapour A, Alipour V, Mirzaei HR and Arabloo J: Cost-effectiveness analysis of intraoperative radiation therapy versus external beam radiation therapy for the adjuvant treatment of early breast cancer: A systematic review. Med J Islam Repub Iran. 34:1672020.PubMed/NCBI

14 

Piotrowski I, Kulcenty K, Murawa D and Suchorska W: Surgical wound fluids from patients treated with intraoperative radiotherapy induce radiobiological response in breast cancer cells. Med Oncol. 36:142018. View Article : Google Scholar : PubMed/NCBI

15 

Lee E, Lee EA, Kong E, Chon H, Llaiqui-Condori M, Park CH, Park BY, Kang NR, Yoo JS, Lee HS, et al: An agonistic anti-Tie2 antibody suppresses the normal-to-tumor vascular transition in the glioblastoma invasion zone. Exp Mol Med. 55:470–484. 2023. View Article : Google Scholar : PubMed/NCBI

16 

Baharlou R, Tajik N, Habibi-Anbouhi M, Shokrgozar MA, Zarnani AH, Shahhosseini F and Behdani M: Generation and characterization of an anti-delta like ligand-4 nanobody to induce non-productive angiogenesis. Anal Biochem. 544:34–41. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Nafissi N, Mohammadlou M, Akbari ME, Mahdavi SR, Sheikh M, Borji M, Babaee E and Baharlou R: The impact of intraoperative radiotherapy on breast cancer: Focus on the levels of angiogenic factors. World J Surg Oncol. 20:1912022. View Article : Google Scholar : PubMed/NCBI

18 

Belletti B, Vaidya JS, D'Andrea S, Entschladen F, Roncadin M, Lovat F, Berton S, Perin T, Candiani E, Reccanello S, et al: Targeted intraoperative radiotherapy impairs the stimulation of breast cancer cell proliferation and invasion caused by surgical wounding. Clin Cancer Res. 14:1325–1332. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Kulcenty KI, Piotrowski I, Zaleska K, Murawa D and Suchorska WM: Wound fluids collected from patients after IORT treatment activates extrinsic apoptotic pathway in MCF7 breast cancer cell line. Ginekol Pol. 89:175–182. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Végran F, Boidot R, Michiels C, Sonveaux P and Feron O: Lactate influx through the endothelial cell monocarboxylate transporter MCT1 supports an NF-κB/IL-8 pathway that drives tumor angiogenesis. Cancer Res. 71:2550–2560. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Kulcenty K, Piotrowski I, Wróblewska JP, Wasiewicz J and Suchorska AWM: The composition of surgical wound fluids from breast cancer patients is affected by intraoperative radiotherapy treatment and depends on the molecular subtype of breast cancer. Cancers (Basel). 12:112019. View Article : Google Scholar : PubMed/NCBI

22 

Kuonen F, Laurent J, Secondini C, Lorusso G, Stehle JC, Rausch T, Faes-Van't Hull E, Bieler G, Alghisi GC, Schwendener R, et al: Inhibition of the Kit ligand/c-Kit axis attenuates metastasis in a mouse model mimicking local breast cancer relapse after radiotherapy. Clin Cancer Res. 18:4365–4374. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Abdollahi A, Griggs DW, Zieher H, Roth A, Lipson KE, Saffrich R, Gröne HJ, Hallahan DE, Reisfeld RA, Debus J, et al: Inhibition of alpha(v)beta3 integrin survival signaling enhances antiangiogenic and antitumor effects of radiotherapy. Clin Cancer Res. 11:6270–6279. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Goedegebuure RSA, de Klerk LK, Bass AJ, Derks S and Thijssen VLJL: Combining radiotherapy with anti-angiogenic therapy and immunotherapy; a therapeutic triad for cancer? Front Immunol. 9:31072019. View Article : Google Scholar : PubMed/NCBI

25 

Orecchia R, Veronesi U, Maisonneuve P, Galimberti VE, Lazzari R, Veronesi P, Jereczek-Fossa BA, Cattani F, Sangalli C, Luini A, et al: Intraoperative irradiation for early breast cancer (ELIOT): Long-term recurrence and survival outcomes from a single-centre, randomised, phase 3 equivalence trial. Lancet Oncol. 22:597–608. 2021. View Article : Google Scholar : PubMed/NCBI

26 

Harris EER and Small W Jr: Intraoperative radiotherapy for breast cancer. Front Oncol. 7:3172017. View Article : Google Scholar : PubMed/NCBI

27 

Dhawan A, Scott JG, Harris AL and Buffa FM: Pan-cancer characterisation of microRNA across cancer hallmarks reveals microRNA-mediated downregulation of tumour suppressors. Nat Commun. 9:52282018. View Article : Google Scholar : PubMed/NCBI

28 

Mueller AK, Lindner K, Hummel R, Haier J, Watson DI and Hussey DJ: MicroRNAs and their impact on radiotherapy for cancer. Radiat Res. 185:668–677. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Metheetrairut C and Slack FJ: MicroRNAs in the ionizing radiation response and in radiotherapy. Curr Opin Genet Dev. 23:12–19. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Zaleska K, Przybyla A, Kulcenty K, Wichtowski M, Mackiewicz A, Suchorska W and Murawa D: Wound fluids affect miR-21, miR-155 and miR-221 expression in breast cancer cell lines, and this effect is partially abrogated by intraoperative radiation therapy treatment. Oncol Lett. 14:4029–4036. 2017. View Article : Google Scholar : PubMed/NCBI

31 

Jeffries J, Zhou W, Hsu AY and Deng Q: miRNA-223 at the crossroads of inflammation and cancer. Cancer Lett. 451:136–141. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Fabris L, Berton S, Citron F, D'Andrea S, Segatto I, Nicoloso MS, Massarut S, Armenia J, Zafarana G, Rossi S, et al: Radiotherapy-induced miR-223 prevents relapse of breast cancer by targeting the EGF pathway. Oncogene. 35:4914–4926. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Wang H, Tan Z, Hu H, Liu H, Wu T, Zheng C, Wang X, Luo Z, Wang J, Liu S, et al: microRNA-21 promotes breast cancer proliferation and metastasis by targeting LZTFL1. BMC Cancer. 19:7382019. View Article : Google Scholar : PubMed/NCBI

34 

Badr M, Said H, Louka ML, Elghazaly HA, Gaballah A and Atef Abd El Mageed M: MicroRNA-21 as a predictor and prognostic factor for trastuzumab therapy in human epidermal growth factor receptor 2-positive metastatic breast cancer. J Cell Biochem. 120:3459–3466. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Di Martino MT, Arbitrio M, Caracciolo D, Cordua A, Cuomo O, Grillone K, Riillo C, Caridà G, Scionti F, Labanca C, et al: miR-221/222 as biomarkers and targets for therapeutic intervention on cancer and other diseases: A systematic review. Mol Ther Nucleic Acids. 27:1191–1224. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Wang J, Wang Q, Guan Y, Sun Y, Wang X, Lively K, Wang Y, Luo M, Kim JA, Murphy EA, et al: Breast cancer cell-derived microRNA-155 suppresses tumor progression via enhancing immune cell recruitment and antitumor function. J Clin Invest. 132:e1572482022. View Article : Google Scholar : PubMed/NCBI

37 

Khalighfard S, Alizadeh AM, Irani S and Omranipour R: Plasma miR-21, miR-155, miR-10b, and Let-7a as the potential biomarkers for the monitoring of breast cancer patients. Sci Rep. 8:179812018. View Article : Google Scholar : PubMed/NCBI

38 

Qu H, Zhu F, Dong H, Hu X and Han M: Corrigendum: Upregulation of CCT-3 induces breast cancer cell proliferation through miR-223 competition and Wnt/b-catenin signaling pathway activation. Front Oncol. 12:9173782022. View Article : Google Scholar : PubMed/NCBI

39 

Park M, Kim D, Ko S, Kim A, Mo K and Yoon H: Breast cancer metastasis: Mechanisms and therapeutic implications. Int J Mol Sci. 23:68062022. View Article : Google Scholar : PubMed/NCBI

40 

Kulcenty K, Piotrowski I, Zaleska K, Wichtowski M, Wróblewska J, Murawa D and Suchorska WM: Wound fluids collected postoperatively from patients with breast cancer induce epithelial to mesenchymal transition but intraoperative radiotherapy impairs this effect by activating the radiation-induced bystander effect. Sci Rep. 9:78912019. View Article : Google Scholar : PubMed/NCBI

41 

Zhao C, Wu M, Zeng N, Xiong M, Hu W, Lv W, Yi Y, Zhang Q and Wu Y: Cancer-associated adipocytes: Emerging supporters in breast cancer. J Exp Clin Cancer Res. 39:1562020. View Article : Google Scholar : PubMed/NCBI

42 

Iwase T, Wang X, Shrimanker TV, Kolonin MG and Ueno NT: Body composition and breast cancer risk and treatment: Mechanisms and impact. Breast Cancer Res Treat. 186:273–283. 2021. View Article : Google Scholar : PubMed/NCBI

43 

Bunnell BA, Martin EC, Matossian MD, Brock CK, Nguyen K, Collins-Burow B and Burow ME: The effect of obesity on adipose-derived stromal cells and adipose tissue and their impact on cancer. Cancer Metastasis Rev. 41:549–573. 2022. View Article : Google Scholar : PubMed/NCBI

44 

Eckel-Mahan K, Ribas Latre A and Kolonin MG: Adipose stromal cell expansion and exhaustion: Mechanisms and consequences. Cells. 9:8632020. View Article : Google Scholar : PubMed/NCBI

45 

Uhlig S, Wuhrer A, Berlit S, Tuschy B, Sutterlin M and Bieback K: Intraoperative radiotherapy for breast cancer treatment efficiently targets the tumor bed preventing breast adipose stromal cell outgrowth. Strahlenther Onkol. 196:398–404. 2020. View Article : Google Scholar : PubMed/NCBI

46 

Wuhrer A, Uhlig S, Tuschy B, Berlit S, Sperk E, Bieback K and Sütterlin M: Wound fluid from breast cancer patients undergoing intraoperative radiotherapy exhibits an altered cytokine profile and impairs mesenchymal stromal cell function. Cancers (Basel). 13:21402021. View Article : Google Scholar : PubMed/NCBI

47 

Bhat K, Sarkissyan M, Wu Y and Vadgama JV: GROα overexpression drives cell migration and invasion in triple negative breast cancer cells. Oncol Rep. 38:21–30. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Slattery K, Woods E, Zaiatz-Bittencourt V, Marks S, Chew S, Conroy M, Goggin C, MacEochagain C, Kennedy J, Lucas S, et al: TGFβ drives NK cell metabolic dysfunction in human metastatic breast cancer. J Immunother Cancer. 9:e0020442021. View Article : Google Scholar : PubMed/NCBI

49 

Pan L, Fu TM, Zhao W, Zhao L, Chen W, Qiu C, Liu W, Liu Z, Piai A, Fu Q, et al: Higher-order clustering of the transmembrane anchor of DR5 drives signaling. Cell. 176:1477–1489.e14. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Wang T, Fahrmann JF, Lee H, Li YJ, Tripathi SC, Yue C, Zhang C, Lifshitz V, Song J, Yuan Y, et al: JAK/STAT3-regulated fatty acid β-oxidation is critical for breast cancer stem cell self-renewal and chemoresistance. Cell Metab. 27:136–150.e5. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Valeta-Magara A, Gadi A, Volta V, Walters B, Arju R, Giashuddin S, Zhong H and Schneider RJ: Inflammatory breast cancer promotes development of M2 tumor-associated macrophages and cancer mesenchymal cells through a complex chemokine network. Cancer Res. 79:3360–3371. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Deng F, Weng Y, Li X, Wang T, Fan M and Shi Q: Overexpression of IL-8 promotes cell migration via PI3K-Akt signaling pathway and EMT in triple-negative breast cancer. Pathol Res Pract. 223:1528242021. View Article : Google Scholar : PubMed/NCBI

53 

Araujo AM, Abaurrea A, Azcoaga P, López-Velazco JI, Manzano S, Rodriguez J, Rezola R, Egia-Mendikute L, Valdés-Mora F, Flores JM, et al: Stromal oncostatin M cytokine promotes breast cancer progression by reprogramming the tumor microenvironment. J Clin Invest. 132:e1486672022. View Article : Google Scholar : PubMed/NCBI

54 

Junk DJ, Bryson BL, Smigiel JM, Parameswaran N, Bartel CA and Jackson MW: Oncostatin M promotes cancer cell plasticity through cooperative STAT3-SMAD3 signaling. Oncogene. 36:4001–4013. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Tulotta C and Ottewell P: The role of IL-1B in breast cancer bone metastasis. Endocr Relat Cancer. 25:R421–R434. 2018. View Article : Google Scholar : PubMed/NCBI

56 

Castaño Z, San Juan BP, Spiegel A, Pant A, DeCristo MJ, Laszewski T, Ubellacker JM, Janssen SR, Dongre A, Reinhardt F, et al: IL-1β inflammatory response driven by primary breast cancer prevents metastasis-initiating cell colonization. Nat Cell Biol. 20:1084–1097. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Wersal C, Keller A, Weiss C, Giordano FA, Abo-Madyan Y, Tuschy B, Sütterlin M, Wenz F and Sperk E: Long-term changes in blood counts after intraoperative radiotherapy for breast cancer-single center experience and review of the literature. Transl Cancer Res. 8:1882–1903. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Meng G, Wuest M, Tang X, Dufour J, Zhao Y, Curtis JM, McMullen TPW, Murray D, Wuest F and Brindley DN: Repeated fractions of X-radiation to the breast fat pads of mice augment activation of the autotaxin-lysophosphatidate-inflammatory cycle. Cancers (Basel). 11:18162019. View Article : Google Scholar : PubMed/NCBI

59 

Krall JA, Reinhardt F, Mercury OA, Pattabiraman DR, Brooks MW, Dougan M, Lambert AW, Bierie B, Ploegh HL, Dougan SK and Weinberg RA: The systemic response to surgery triggers the outgrowth of distant immune-controlled tumors in mouse models of dormancy. Sci Transl Med. 10:eaan34642018. View Article : Google Scholar : PubMed/NCBI

60 

Pan L, Wan M, Zheng W, Wu R, Tang W, Zhang X, Yang T and Ye C: Intrabeam radiation inhibits proliferation, migration, and invasiveness and promotes apoptosis of MCF-7 breast cancer cells. Technol Cancer Res Treat. 18:15330338198407062019. View Article : Google Scholar : PubMed/NCBI

61 

Tang H, Cai L, He X, Niu Z and Huang H, Hu W, Bian H and Huang H: Radiation-induced bystander effect and its clinical implications. Front Oncol. 13:11244122023. View Article : Google Scholar : PubMed/NCBI

62 

Al-Abedi R, Tuncay Cagatay S, Mayah A, Brooks SA and Kadhim M: Ionising radiation promotes invasive potential of breast cancer cells: The role of exosomes in the process. Int J Mol Sci. 22:115702021. View Article : Google Scholar : PubMed/NCBI

63 

Feghhi M, Rezaie J, Mostafanezhad K and Jabbari N: Bystander effects induced by electron beam-irradiated MCF-7 cells: A potential mechanism of therapy resistance. Breast Cancer Res Treat. 187:657–671. 2021. View Article : Google Scholar : PubMed/NCBI

64 

Chen S, Zhao Y, Han W, Chiu SK, Zhu L, Wu L and Yu KN: Rescue effects in radiobiology: Unirradiated bystander cells assist irradiated cells through intercellular signal feedback. Mutat Res. 706:59–64. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Amaravadi RK and Thompson CB: The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res. 13:7271–7279. 2007. View Article : Google Scholar : PubMed/NCBI

66 

Lu Z, Luo RZ, Lu Y, Zhang X, Yu Q, Khare S, Kondo S, Kondo Y, Yu Y, Mills GB, et al: The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells. J Clin Invest. 118:3917–3929. 2008.PubMed/NCBI

67 

Wang X, Zhang J, Fu J, Wang J, Ye S, Liu W and Shao C: Role of ROS-mediated autophagy in radiation-induced bystander effect of hepatoma cells. Int J Radiat Biol. 91:452–458. 2015. View Article : Google Scholar : PubMed/NCBI

68 

Kulcenty K, Piotrowski I, Rucinski M, Wroblewska JP, Jopek K, Murawa D and Suchorska WM: Surgical wound fluids from patients with breast cancer reveal similarities in the biological response induced by intraoperative radiation therapy and the radiation-induced bystander effect-transcriptomic approach. Int J Mol Sci. 21:11592020. View Article : Google Scholar : PubMed/NCBI

69 

Vaupel P and Multhoff G: Revisiting the Warburg effect: Historical dogma versus current understanding. J Physiol. 599:1745–1757. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Vaupel P, Schmidberger H and Mayer A: The Warburg effect: Essential part of metabolic reprogramming and central contributor to cancer progression. Int J Radiat Biol. 95:912–919. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Estrella V, Chen T, Lloyd M, Wojtkowiak J, Cornnell HH, Ibrahim-Hashim A, Bailey K, Balagurunathan Y, Rothberg JM, Sloane BF, et al: Acidity generated by the tumor microenvironment drives local invasion. Cancer Res. 73:1524–1535. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Wenz F: Keynote address at the american society of breast surgeons 18th annual meeting: Current and future application of intraoperative radiotherapy (IORT) in the curative and palliative treatment of breast cancer. Ann Surg Oncol. 24:2811–2817. 2017. View Article : Google Scholar : PubMed/NCBI

73 

Omosule M, De Silva-Minor S and Coombs N: Case report: Intraoperative radiotherapy as the new standard of care for breast cancer patients with disabling health conditions or impairments. Front Oncol. 13:11566192023. View Article : Google Scholar : PubMed/NCBI

74 

Hochhertz F, Hass P, Röllich B, Ochel HJ and Gawish A: A single-institution retrospective analysis of intraoperative radiation boost during breast-conservation treatment for breast cancer. J Cancer Res Clin Oncol. 149:5743–5749. 2023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2023
Volume 28 Issue 6

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang Y, Hou X, Kong S, Zha Z, Huang M, Li C, Li N, Ge F and Chen W: Intraoperative radiotherapy in breast cancer: Alterations to the tumor microenvironment and subsequent biological outcomes (Review). Mol Med Rep 28: 231, 2023
APA
Yang, Y., Hou, X., Kong, S., Zha, Z., Huang, M., Li, C. ... Chen, W. (2023). Intraoperative radiotherapy in breast cancer: Alterations to the tumor microenvironment and subsequent biological outcomes (Review). Molecular Medicine Reports, 28, 231. https://doi.org/10.3892/mmr.2023.13118
MLA
Yang, Y., Hou, X., Kong, S., Zha, Z., Huang, M., Li, C., Li, N., Ge, F., Chen, W."Intraoperative radiotherapy in breast cancer: Alterations to the tumor microenvironment and subsequent biological outcomes (Review)". Molecular Medicine Reports 28.6 (2023): 231.
Chicago
Yang, Y., Hou, X., Kong, S., Zha, Z., Huang, M., Li, C., Li, N., Ge, F., Chen, W."Intraoperative radiotherapy in breast cancer: Alterations to the tumor microenvironment and subsequent biological outcomes (Review)". Molecular Medicine Reports 28, no. 6 (2023): 231. https://doi.org/10.3892/mmr.2023.13118