Next Article in Journal
Cytokine Release Syndrome Associated with T-Cell-Based Therapies for Hematological Malignancies: Pathophysiology, Clinical Presentation, and Treatment
Next Article in Special Issue
Cytostatic Effect of a Novel Mitochondria-Targeted Pyrroline Nitroxide in Human Breast Cancer Lines
Previous Article in Journal
Magnetic Particle Imaging: Current and Future Applications, Magnetic Nanoparticle Synthesis Methods and Safety Measures
Previous Article in Special Issue
Optical Redox Imaging of Treatment Responses to Nampt Inhibition and Combination Therapy in Triple-Negative Breast Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Breast Cancer and Anaesthesia: Genetic Influence

1
Institute of Biomedical Research in Malaga (IBIMA), 29010 Malaga, Spain
2
Department of Anaesthesiology, Virgen de la Victoria University Hospital, 29010 Malaga, Spain
3
Department of Pharmacology and Pediatrics, School of Medicine, University of Malaga, 29010 Malaga, Spain
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(14), 7653; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147653
Submission received: 28 May 2021 / Revised: 5 July 2021 / Accepted: 6 July 2021 / Published: 17 July 2021
(This article belongs to the Special Issue New Drugs for Breast Cancer Treatment)

Abstract

:
Breast cancer is the leading cause of mortality in women. It is a heterogeneous disease with a high degree of inter-subject variability even in patients with the same type of tumor, with individualized medicine having acquired significant relevance in this field. The clinical and morphological heterogeneity of the different types of breast tumors has led to a diversity of staging and classification systems. Thus, these tumors show wide variability in genetic expression and prognostic biomarkers. Surgical treatment is essential in the management of these patients. However, the perioperative period has been found to significantly influence survival and cancer recurrence. There is growing interest in the pro-tumoral effect of different anaesthetic and analgesic agents used intraoperatively and their relationship with metastatic progression. There is cumulative evidence of the influence of anaesthetic techniques on the physiopathological mechanisms of survival and growth of the residual neoplastic cells released during surgery. Prospective randomized clinical trials are needed to obtain quality evidence on the relationship between cancer and anaesthesia. This document summarizes the evidence currently available about the effects of the anaesthetic agents and techniques used in primary cancer surgery and long-term oncologic outcomes, and the biomolecular mechanisms involved in their interaction.

1. Introduction

Breast cancer is the leading cause of mortality in women [1]. Even when the tumor is completely resected, tumor recurrence occurs in up to one third of patients, with metastatic disease being the direct cause of death [2]. Surgery may generate systemic inflammatory response syndrome, which causes oxidative stress, and in turn impairs the anti-tumor immunologic response [3]. Surgical stress activates a neuroendocrine response in the hypothalamic–pituitary–adrenal axis (HPA axis) and sympathetic nervous system (SNS), which results in the suppression of cell-mediated immunity (CMI); this suppression is induced by the release of neuroendocrine mediators such as catecholamines, cortisol, and cytokines [4].
These mediators, including vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMPs) and interleukin (IL) 6 and 8, are endogenous regulators that promote tumor growth and angiogenesis, thereby favoring metastasis.
Recent studies reveal that the type of anaesthesia administered during cancer surgery may influence the course of the disease [5]. However, there is limited evidence available that supports the modification of standard anaesthetic approaches.
Further studies are necessary to identify the mechanisms by which each agent interacts with tumor cells. The aim of this article is to provide a review of the current knowledge on how anaesthetic drugs and techniques affect breast cancer recurrence.

2. Anaesthetics and Cancer Relapse

2.1. Hypnotics

Propofol

Propofol may have beneficial effects on survival in cancer patients, including breast cancer patients. This agent inhibits tumor cell migration and proliferation, promotes tumor apoptosis and has anti-inflammatory activity [5,6,7]. This agent acts on the immune system at the level of natural killer lymphocytes (NK), which belong to innate immunity. Cho et al. [8] conducted a prospective study comparing a group of patients who received propofol-ketorolac vs. sevoflurane-fentanyl. The authors observed a reduction in NK activity in the sevoflurane group, whereas this activity was increased in the propofol group. Although evidence consistently shows that propofol has tumor-killing activity, two recent studies associate it with pro-tumor activity in breast and bladder cancer, mediated by the activation of the Nrf2 pathway and the reduction in p53 levels [9,10].
Propofol favors tumor cell apoptosis by affecting matrix metalloproteinase (MMPs) expression, which play a crucial role in extracellular protein degradation and epithelial-mesenchymal transition (EMT), activate vascular endothelial growth factor (VEGF) [11], and inhibit intrinsic apoptosis pathways [12]. Two pathways have been identified to be involved in the inhibition of the synthesis of these proteins: the MAP-kinase pathway (ERK1/2, JNK y p38) in colon cancer [13], and NF-κB in breast cancer [14].
As for the ability to induce tumor apoptosis, several pathways have been investigated, such as the inhibition of anti-apoptosis mechanisms, including Bcl-2, Sox4, Akt/mTOR and Wnt/β-catenin, and the increase in pathways involving tumor suppressor genes such as the Bax, ING3, Fox01 and caspase pathways [15,16,17,18,19,20]. In addition, Wang et al. [21] demonstrated that propofol induces the intrinsic apoptotic signaling pathway by the release of reactive oxygen species (ORS).
Propofol may inhibit surgery-induced systemic inflammatory response syndrome throughout decreasing cell concentrations of hypoxia-inducible factor 1 (HIF1A), which is elevated in the tumor’s hypoxic micro-environment and promotes cell migration and invasion [22]. Ecimovic et al. [23] demonstrated in vitro the role of NET-1 (neuroepithelial cell transforming gene-1) overexpression, a gene that is associated with tumor dissemination and which decreases with propofol exposure [24].

2.2. Halogenated

It has been suggested that inhalation agents, for the most part, have a tumorigenic effect since they both inhibit tumor cell apoptosis and stimulate tumor cell proliferation and migration. More specifically, in advanced breast cancer, elevated caveolin-1 concentrations have been associated with lower survival. This protein has been linked to higher resistance to apoptosis, migration and elevated invasivity in breast cancer cells. Ecimovic et al. [25] analyzed, in vitro, the ability of sevoflurane to stimulate tumor cell proliferation, migration and invasion in patients with positive (ER+) and negative (ER-) estrogen receptor breast cancer (with the latter not having invasive capacity).
However, Kawaraguchi et al. [26] documented that isoflurane confers a protective effect on tumor cells in colon cancer against TNF-mediated apoptosis (TRAIL or TNF-related apoptosis-inducing ligand) by interacting with caveolin-1.
Sevoflurane reduces NK activity, thereby reducing immunosurveillance and favoring progression of micrometastases. In contrast, a range of studies have been conducted to compare immunosuppression induced by halogenated anaesthetics vs. general intravenous anaesthesia, with inconsistent results [27,28,29,30,31,32]. Enlund et al. [27] found no significant differences in 1-year and 5-year survival in a sample of 1837 breast cancer patients. Kim et al. [29] compared propofol with a variety of halogenated agents (sevoflurane, desflurane, isoflurane and enflurane), without significant differences. Recent retrospective studies [30,31,32] provide cumulative evidence of the absence of significant differences between intravenous and inhalation anaesthetics in terms of recurrence or survival.

3. Analgesics

3.1. Opioids

Opioids have an immunosuppressive effect that influences cellular and humoral immunity, as they reduce NK lymphocyte activity and proliferation, citokine production, phagocytic activity, and antibody release [33]. The type and degree of immunosuppression depends on the type, dose and time of exposure to the opioid. All synthetic opioids reduce NK activity [34,35].
Morphine inhibits T and NK lymphocyte activity, promotes lymphocyte apoptosis, reduces toll-like 4 factor in macrophages [36] and has angiogenic activity [37]. In addition, the tumorigenic activity of morphine is mediated by two independent mechanisms, namely: by direct stimulation of mu receptors in tumor cells, the overexpression of which has been associated with poor prognosis, and indirectly by promoting neo-angiogenesis through metabolic signaling pathways similar to those used by VEGF factor [38,39].
In breast tumor cells, fentanyl exhibits an antitumoral effect by reducing levels of proteins involved in cell apoptosis and differentiation mechanisms (Bax, Bcl2, Oct4, Sox2, and Nanog) [40]. Although tramadol is a µ-receptor agonist, its analgesic effect is prevailingly mediated by the inhibition of noradrenaline and serotonin reuptake. Sacerdote et al. [41] assessed the relationship between tramadol and immune response in patients with uterine carcinoma. The authors found that tramadol not only inhibits but also stimulates NK lymphocyte activity. Thereupon, Xia et al. [42] demonstrated, in vitro, in breast tumor cells that tramadol reduces tumor cell proliferation, migration and invasion by up to 28 days through the inhibition of the α2-adrenergic receptor. In a retrospective study, Kim et al. [43] observed lower rates of mortality and tumor recurrence in the group of breast cancer patients treated surgically who received tramadol. This effect is conferred by the inhibition of tumor cell proliferation, induction of apoptosis, and action on serotonergic receptors and transient receptor potential channel V1 or TRPV1.

3.2. Regional Anaesthesia and Local Anaesthetics

There is a variety of locoregional anaesthesia techniques in breast cancer surgery that have good analgesic outcomes. Paravertebral block (PVB) is the most widely used technique, although it is associated with a higher risk of severe complications. New techniques have been developed, with pectoral block type II having shown good effectiveness, and having been employed in a similar context as PVB [44]. Several studies have been published comparing general anaesthesia and combined anaesthesia: five retrospective studies, two prospective studies and a systematic review. These studies provide evidence of the beneficial effects of not using opioids and/or local anaesthetics per se [45,46,47,48,49,50,51,52].
Exadaktylos et al. [45] published the first retrospective study assessing the outcomes of 129 patients with breast cancer treated surgically, of whom 50 received combined anaesthesia (PVB+Propofol) and 79 balanced general anaesthesia. The rate of recurrence was lower in the group that received combined anaesthesia. In contrast, a recent systematic review conducted by Pérez-González et al. [52] did not show statistically significant differences between combined and general anaesthesia.
Local anaesthetics block afferent and efferent nerve response and effectively suppress sympathic stimulation through the inhibition of hypothalamic–pituitary–adrenal (HPA) stimulation induced by surgical stress, thereby reducing HPA activity [52].
Lidocaine has been proven to exert beneficial effects in vivo and in vitro, and it is associated with a reduction in tumor cell proliferation, migration and invasion in breast, liver and lung cancer [53,54,55]. Lidocaine inhibits the proto-oncogen that releases Src, an intracellular non- tyrosine kinase protein that is involved in cell proliferation and migration processes through ICAM-1 phosphorylation, which enables neutrophils to cross the endothelium and increase immune response [55]. It has been reported to have effects on other signaling pathways such as TRPV-6 inhibition [56] or DNA demethylation in breast cancer cells [57]. Chang et al. [58] demonstrated in vitro that both lidocaine and bupivacaine induce breast cancer cell apopotosis through the activation of caspases 7, 8 and 9. In the same line, D’Agositino et al. showed that lidocaine inhibits cytoskeletal modification in breast cancer cells [59].
Evidence has been provided that lidocaine infiltration in the peritumoral region inhibits tumor growth by binding EGFR [60]. As for the immune system, lidocaine, at clinically relevant concentrations, stimulates the cytotoxic effect of NK lymphocytes [61]. A prospective, randomized trial conducted by Galoș et al. revealed that lidocaine reduced neutrophil extracellular traps, a phenomenon that has been associated with tumor recurrence [62].

3.3. NSAIDs

The enzyme cyclooxygenase (COX-2) causes an increase in prostaglandins, which are involved in immune system control and angiogenesis. Ketorolac is the most extensively studied NSAID in relation to cancer. It is a COX-1 and COX-2 inhibitor that is commonly used in the perioperative period. Evidence from retrospective studies demonstrates that perioperative administration of ketorolac reduces breast cancer recurrence by diminishing the production of prostaglandins and VEGF. Forget et al. attempted to replicate these results in patients with breast cancer at high risk of recurrence (triple negative, neutrophil/lymphocyte ratio ≥ 4) [63] in a prospective study [64] of 203 patients, without differences having been found between treatment groups.

4. Dexmedetomidine

Dexmedetomidine is a selective α2 agonist with sympatholytic and anti-inflammatory activity that reduces IL-6, IL-8 and TNF-α concentrations and increases anti-inflammatory cytokine IL-10 levels [65].
Despite its anti-inflammatory effect, a pro-tumoral activity is attributed to dexmedetomidine. Lavon et al. [66] demonstrated in animal models that it promotes metastasis in breast, lung and colon cancer. This effect is credited to the transient immunosuppression induced by dexmedetomidine, added to the effects of surgical stress and changes in vascular patency.
In the same line, Xia et al. [67] investigated the effect of dexmedetomidine in breast cancer cells, in vitro and in vivo, in mice and concluded that dexmedetomidine promotes tumor cell proliferation, migration and invasion through the inhibition of the α2/ERK adrenergic receptor pathway [42].
Cata et al. [68] performed a retrospective study involving 1404 patients with non-small lung cancer (NSCLC) treated surgically to investigate a potential relationship between tumor recurrence and the use of dexmedetomidine. This relationship was not confirmed. Indeed, the results showed a significant relationship between dexmedetomidine and lower survival.
Beta-blockers and lipid lowering drugs are two of the main groups of drugs among patients undergoing a surgical procedure.

5. Beta-Blockers

Beta-adrenergic receptors are found both in tumor cells and the immune system [69], and seem to play a key role in carcinogenesis [70]. Beta-blockers have been proven to be involved in angiogenesis and cellular neoproliferation [71]. Exposure to beta-agonists inhibits lymphocyte NK activity [72,73] and induces an increase in T-regulator lymphocytes [74], leading to immunosuppression. Kang et al. documented that adrenergic stimulation activated the MAP-kinase cascade and, more specifically, the DUSP1 cascade, which causes resistance to chemotherapy and apoptosis [75]. Recently, Zhou et al. [76] observed that propranolol prevented T-regulator lymphocyte elevation. Another potential cellular signaling pathway is adrenergic activation of PI3K/AKT and HIF-1 α, which is also inhibited by propranolol [77].
Contradictory results were obtained in five retrospective [78,79,80,81,82] and two cohort studies [83,84] assessing recurrence in breast cancer patients after surgery due to the lack of a standard treatment administration protocol [85,86,87,88].

6. Lipid Lowering Drugs

The increased prevalence of cardiovascular disease in the recent years has resulted in an increase in the use of lipid lowering drugs, with statins being the most common pharmaceutical group. As a component of the cellular membrane, cholesterol plays an essential role in cellular division; therefore, a reduction in extracellular cholesterol should cause an inhibition of tumor cell proliferation. Cholesterol metabolites such as 27-hydroxycholesterol and 25-hydroxycholesterol may stimulate estrogen receptors (ERs) [89,90]. Alikhani et al. [91] reported an increase in breast tumor growth mediated by the PI3K/AKT pathway in hyperlipidemic mice.
Cholesterol favors a pro-inflammatory environment by the activation of macrophage toll-like receptors [92] and the inhibition of CCR7 expression in dendritic cells, which explains their antigenic effects [93]. On the other hand, cholesterol modulates lymphocyte T activity through the liver X receptor (LXR) [94].
As for the use of statins, inconsistent results were obtained in six retrospective [80,95,96,97,98,99] and five prospective studies [85,100,101,102,103] (four supporting its use and the remaining seven having not provided clinically relevant results). In contrast, the three meta-analyses retrieved [103,104,105,106] provide consistent evidence that statins reduce breast cancer recurrence. However, these studies were conducted using non-standard methods, and prospective randomized studies are needed.

7. Biomarkers, Anaesthetic Technique and Cancer

In previous paragraphs, we have described different routes and mechanisms by which drugs administered in the perioperative period affect breast cancer recurrence. In the next section, we will discuss the current knowledge of how these drugs and techniques affect the most important biomarkers, as well as their clinical relevance.
Early detection, disease monitoring during treatment and subsequent follow-up to detect recurrences are still critical for improving the outcomes, morbidity and mortality of cancer patients. This is hampered by the absence of obvious symptoms and insufficiently sensitive biomarkers. Biopsy and imaging examination applications are limited; in addition, traditional tumor diagnostic markers exhibit low sensitivity. Therefore, there is an exponential development of evidence for the detection and validation of novel, more sensitive, and easy-to-detect biomarkers which can be used in the diagnosis and prognosis of cancers.
A multiplicity of anaesthetic techniques are used in clinical practice in patients undergoing cancer surgery. The pharmacokinetics and pharmacodynamics of common anaesthetic agents are thoroughly understood and their use is determined based on the characteristics of each patient. On the other hand, the retrospective analysis of the results in oncological surgery showed that the use of total intravenous anaesthesia (TIVA) was associated with improved recurrence-free survival and overall survival [107].
Thanks to the growing evidence, special attention has been given to the molecules having remarkable ability to target oncogenic protein network, restore drug sensitivity and induce apoptosis in cancer cells. The mechanisms through which general anaesthetics modulated wide-ranging deregulated cell signaling pathways involved in angiogenesis, metastasis, drug resistance, immune-related responses, cytokine secretion, proliferation, invasion and prognosis remained unclear.
The beneficial effects and clinical implications of the different pharmacological groups in cardiovascular surgery are widely known. These include the direct organ-protective effects of sevoflurane mediated by the modulation of metabolic cascades [108,109,110]. In neuroscience, the analysis of the mechanisms of effects of isoflurane on the brain for short periods showed that differentially expressed genes, such as homeobox containing 1, APC, MAPK1 and miRNA-17-5p, were involved [111].
Propofol is one of the most studied drugs in onco-anaesthesia, and there is evidence of its ability to modulate molecular targets such as the Wnt/β-catenin, JAK/STAT and mTOR-driven pathways [112]. Targeted therapies based on molecular markers have potential for the diagnosis and prognosis of a diversity of diseases, including cancer. The development of drugs that act on these molecular pathways show promising results in breast cancer [113].
The identification of biomarkers helps to better understand the influence of the anaesthetic technique on tumor progression and select the optimal anaesthetic plan for each cancer patient. For example, exposure to halogenated agents increased Lipocalin 2 expression over a prolonged period of time [114], and this over-expression also promotes cell migration and invasion through activating ERK signaling to increase SLUG [115], whereas knockdown suppresses the growth and invasion of cancer cells [116]. On the contrary, propofol inhibits growth and invasion through the regulation of the Slug signaling pathway [117].
Lipocalin 2 (LCN2), also known as neutrophil gelatinase associated lipocalin (NGAL), regulates angiogenesis [118,119] and has been reported to be elevated in several types of cancer, including breast cancer. The evidence obtained in vitro and in vivo demonstrates that the tumorigenic and metastatic potential of Lcn2 is induced by TEM promotion, cell migration, invasion, VEGF production, and angiogenesis. The mechanisms that mediate the metastatic and oncogenic potential of Lcn2 include the PI3K/Akt/NF-κB and HIF-1a/ERK signaling pathways, and the protective formation of the MMP-9/Lcn2 complex [120,121].
When its activity is knocked down, angiogenesis decreases through the inhibition of the MMP-9/Lcn complex, thereby reducing MMP-9 activity, cell migration and invasion capacity [118,119,121,122,123,124,125]. In addition, ICAM-targeted Lcn2 sRNA (small interfering RNA) significantly reduced the angiogenic activity and migration of triple-negative breast tumor cells [126,127]. Leng et al. reported the use of anti-Lcn2I policlonal antibodies and they achieved the inhibition of 4T1 cellular metastases [128].
A further alternative molecule that has been associated with the interaction between anaesthetics and cancer is the endothelial adhesion molecule. The main function of endothelial adhesion molecules is mediated by immune response. A range of studies demonstrate that intercellular adhesion molecule-1 (ICAM-1) triggers the activation of multiple cellular signaling pathways that promote tumor cell proliferation, migration and resistance to apoptosis, as well as the development of drug resistance induced by cellular adhesion molecules [129,130]. Where inflammation occurs, the activated leukocytes adhere to the endothelium through ICAM-1, and cell transmigration follows through vessel walls. A similar mechanism mediates extravasation of circulating tumor cells during metastasis, where endothelial ICAM-1 might play a crucial role [129].
ICAM-1 is expressed in several types of tumors and, through the mediation of its main ligand, LFA-1, it may facilitate immunosurveillance [131,132]. Several studies associate ICAM-1 expression with a more aggressive tumor phenotype and higher metastatic potential [133,134]. The capacity of infiltration of breast cancer cells is related to ICAM-1 expression [130]. Serum levels are elevated in breast cancer patients [135] and correlate with advanced-stage or recurrent disease [136,137] and metastasis. Schröder et al. documented the association between ICAM-1 mRNA in breast cancer, high levels of urokine plasminogen (uPA) and uPA-1 inhibitor (PAI 1), and Ki67 and VEGF mRNA overexpression [134].
Expression of ICAM-1 is upregulated by tumor necrosis factor-alfa (TNF-α); meanwhile, local anaesthetics may have an anti-inflammatory effect, since they effectively block the inflammatory signaling of TNFα. The resulting decrease in Akt, endothelial nitric oxide synthase, and Src phosphorylation reduced neutrophil adhesion and endothelial hyperpermeability. Lidocaine and ropivacaine also impair cancer cell migration by inhibiting Src activation that is induced by TNF-α and the phosphorylation of ICAM-1 [138,139].
The roles of the immune system in cancer, from tumor initiation to metastatic progression, remain unclear. Neutrophil extracellular traps (NETosis) form part of the immune response to cancer antigens. NETosis originates from neutrophil degranulation, which results in the release of neutrophil contents into the bloodstream, where they can be detected [140,141]. NETosis processes have been described in the tumor microenvironment [142,143].
Tumor cells can interact direct or indirectly with neutrophils to induce NET production. Intra-and inter-cellular molecular communications generate micro-environments that act as a premetastatic niche to recruit primary tumor cells and form multiple metastases [144,145]. Park et al., described the relationship between metastatic cancer and NET formation through the use of anti-G-CSF blocking antibodies, which reduce the ability of 4T1 cells to induce NETosis [146].
The close cooperation between tumor cells, neutrophils and NETosis in the tumor microenvironment demonstrates the role of NETosis in cancer progression and metastasis [143,147,148]. The myeloperoxidase enzyme (MPO) and citrullinated histone-3 are specific proteins released during NETosis and can be easily measured in serum [143]. The release of chromatin by tumor cells and neutrophils influences tumor progression and thrombosis associated with cancer [149].
This phenomenon is associated with poor prognosis in esophageal cancer and a higher risk for tumor infiltration in patients with higher NETosis activity [150]. The presence of circulating NETs has been described, in patients with advanced esophageal, gastric and lung cancer, at higher levels than healthy controls. The authors further demonstrated, in murine models, that NETs can regulate disease progression, since blocking their formation reduced the occurrence of metastases [151].
Firstly, the relationship between NETs and anaesthetics emerges in patients with septic shock. Propofol shows that clinically relevant concentrations inhibit ROS-dependent NET production by human neutrophils [152]. Chen et al., compare the effects of four intravenous anaesthetics. Through this study, they define a new anti-inflammatory effect of intravenous anaesthetics. Propofol appears to decrease NET formation by activated neutrophils through the inhibition of hypochlorus acid production and MAPK/ERK [153]. Local anaesthetics in clinically used doses, decreased, in a dose-dependent manner, the time to onset of maximal ROS production and half-maximal NETosis [154].
Two randomized clinical trials have been conducted to assess the influence of different anaesthetic techniques in NETosis. Aghamelu et al. found no significant differences between the halogenated agents and propofol in patients with breast cancer treated surgically [155] In addition, Galos et al. incorporated the differential use of intravenous lidocaine in their treatment protocol and observed significant differences in NETosis markers between groups [62].

7.1. miRNAs and Drugs Used during Anaesthetic Procedures

Even though enzymes play a key role in describing the effects of anaesthetic drugs and techniques on breast cancer recurrence, the codifying process may also have major importance in this issue. Non-coding RNAs, particularly MicroRNAs, are playing a growing role in the field of oncology and anaesthesia.
There is growing interest in the identification of biomarkers for assessing individual risk of cancer progression and response to treatment. MicroRNAs (miRNAs) are endogenous non-coding RNA sequences that regulate gene expression and suppress or activate multiple genes at pre- and post-transcriptional levels [156,157]. In recent years, evidence of the relevant role of miRNA in the molecular mechanisms of carcinogenesis in healthy patients has accumulated [158,159,160,161]. Although these changes are detectable in tumor tissue, levels of deregulated miRNAs can also be determined in blood [162]. Likewise, there is wide inter-subject variability in deregulated miRNA levels in healthy subjects and in cancer patients treated surgically [163].
Current evidence suggests that the beneficial or deleterious effects of anaesthetics during cancer surgery are mediated by genetic and molecular mechanisms [164]. Multiple studies have been published on the interaction of propofol with the genetic mechanisms that mediate miRNA suppression/overexpression in different types of tumors: propofol increases miRNA let7 expression and apoptosis in ovarian cancer cells [165]; it increases miRNA 218 and miRNA 451 expression, thereby resulting in the reduction in MMP-2 expression and cellular proliferation [166]. Finally, propofol reduces the invasive capacity of liver cell carcinoma through miRNA-199a and its action on MMP-9 [167].
Sun et al. concluded that propofol reduces miRNA-374a expression and modulates the forkhead box O1 pathway response to reduce proliferation and resistance to cisplatin in ovarian cancer cells [168].
Propofol has demonstrated, in vitro and in vivo, its ability to facilitate sensitivization to trastuzumab-resistant tumor cells through epigenetic changes mediated by the miRNA-149-5p/IL-6 axis [169]. Propofol induces apoptosis in breast cancer cell lines through the inactivation of the miRNA-24/p27 signaling pathway [170]. Propofol has also been associated with one of the major types of onco-miRNA-2; it is overexpressed in early-stage pancreatic cancer. The action of propofol on miRNA-21 involves the suppression of Slugh expression, thereby increasing the proapoptotic effect of the p53 gene and inducing e-cadherin overexpression [171].
Buschmann et al. recently published the first prospective study comparing the serum of patients with colon cancer—who were treated surgically and exposed to propofol or sevoflurane—and miRNA expression patterns. Significant differences were observed between groups, with propofol showing an inhibitory effect on signaling pathways involved in carcinogenesis [172]. Deng et al. exposed breast cancer cell lines to sevofluorane vs. propofol, and observed an increase in tumor survival in the propofol group, mediated by the modulation of intracellular calcium homeostasis [173]. In contrast, Liu et al. observed that sevoflurane suppressed breast cancer cell proliferation through an increase in miRNA-203 overexpression [174]. Its potential anti-tumor activity is demonstrated by miRNA-155 suppression in papillary thyroid carcinoma [175].
Interactions have been observed in other drugs commonly used in clinical practice. Li et al. documented that ropivacaine inhibits cellular viability and induces apoptosis, whereas concomitant use of sufentanil mitigates its action by means of miRNA-182-5p/BCL10/CYCS regulation [176]. Qu et al. suggest that the beneficial effects of lidocaine on colorectal cancer could be mediated by the upregulation of miRNA-520 a-3p and targeting EGFR [177].
Sequencing of the genome of macrophages treated with morphine shows a differential expression pattern of 26 miRNAs. Of these, miRNA-15b-5p and miRNA-181b-5p are involved in inflammatory and infectious processes, thereby promoting the suppression of immune functions [178]. A diversity of studies reveals the upregulation of 9 miRNAs and downregulation of 17 miRNAs. More specifically, the miRNAs of the Let-7 family have been found to be associated with the MOR pathway [179,180,181]. Low doses of morphine promote non-differentiated cell proliferation, thus reducing miRNA-133b expression. In contrast, at high doses, they have the opposite effect and reduce miRNA-133b and miRNA-128 levels; this effect cannot be countered by naloxone, which suggests an independent mediation by MOR [182]. Exposure to morphine upregulates miRNA-543 expression and contributes to tumor progression through MARCKs inhibition [183].

7.1.1. MiRNAs and Breast Cancer

Different studies document the association between breast cancer and a variety of miRNA expression patterns, which enables the analysis, classification and characterization of different types of tumors [184,185,186]. Mar-Aguilar et al. reported the potential usefulness of three miARNs in breast cancer screening [187]. The anti-tumor effect of propofol and pro-tumor effect of sevoflurane have been demonstrated in in vitro studies. These effects are mediated by the modulation of miRNA- 24/p27 and miRNA-203 expression, respectively [170,174].

7.1.2. MiRNA-21

There is consistent evidence supporting the oncogenic role of miR-21, which is involved in the inhibition of tumor cell apoptosis and contributes to tumor growth. MiR-21 is significantly overexpressed in a wide variety of solid tumors, including breast cancer. MiR-21 overexpression has been associated with tumor progression and poor prognosis [188]. It is involved in the inhibition of tropomyosin alpha-1, the TP53 and TAp63 pathways (regulating LKB1 transcription) [189], programmed cell death 4 (related to apoptosis) [190], and phosphatase tensin homolog (PTEN) [191,192,193,194].
In vitro studies reveal that miR-21 is overexpressed in tissue and cells of triple-negative breast cancer models. This confirms its relationship with MDA-MB-468 cell proliferation and invasion, and the regulation of PTEN expression [195]. MiR-21 knockdown reduced proliferation, viability and invasivity, demonstrating that PTEN is a target of this mRNA [196]. Its overexpression is also linked to poor survival due to the development of resistance to trastuzumab [197,198,199].

7.1.3. MicroRNA-202

MiR-202 has been recently associated with multiple types of cancer, including breast cancer. It belongs to the Let-7 miRNA family, and its downregulation has been associated with poor differentiation and higher tumor aggressiveness [200].
The let-7 family regulates estrogen receptor alpha in breast cancer growth [201] and is involved in the self-renovation and tumorigenicity of breast cancer cells [202]. Joosse et al. detected a relationship between miR-202 levels and prognosis, which suggests a potential role in tumor staging [203].

7.1.4. MicroRNA-155

MiRNA-155 is involved in immune function through cancer development and the production of cytokines. It is overexpressed in different types of cancer, including breast, liver, uterine and lung cancer [204,205,206]. It favors breast cancer progression by acting on TP53 [207,208] and accelerating metastasis through tumor protein p53-inducible nuclear protein 1 (TP53INP1) [209].

8. Conclusions

The perioperative use of anaesthetic/analgesic techniques with protective effects on anti- metastatic immune response may reduce tumor progression. A limitation of most studies published to date is that they were performed in vitro, where conditions are not the same as in vivo. The use of biomarkers could be useful to identify scenarios in which there is a high risk of cancer progression. Randomized clinical trials are required to investigate whether anesthetic/analgesic techniques and anaesthetic agents are effective in reducing cancer relapse and increasing survival in oncological patients.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest, financial or otherwise.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Kim, R. Effects of surgery and anesthetic choice on immunosuppression and cancer recurrence. J. Transl. Med. 2018, 16, 1–13. [Google Scholar] [CrossRef]
  3. Horowitz, M.; Neeman, E.; Sharon, E.; Ben-Eliyahu, S. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat. Rev. Clin. Oncol. 2015, 12, 213–226. [Google Scholar] [CrossRef] [Green Version]
  4. Hiller, J.G.; Perry, N.J.; Poulogiannis, G.; Riedel, B.; Sloan, E.K. Perioperative events influence cancer recurrence risk after surgery. Nat. Rev. Clin. Oncol. 2018, 15, 205–218. [Google Scholar] [CrossRef]
  5. Li, R.; Liu, H.; Dilger, J.; Lin, J. Effect of Propofol on breast Cancer cell, the immune system, and patient outcome. BMC Anesthesiol. 2018, 18, 1–8. [Google Scholar] [CrossRef]
  6. Jiang, S.; Liu, Y.; Huang, L.; Zhang, F.; Kang, R. Effects of propofol on cancer development and chemotherapy: Potential mechanisms. Eur. J. Pharmacol. 2018, 831, 46–51. [Google Scholar] [CrossRef] [PubMed]
  7. Xu, Y.; Pan, S.; Jiang, W.; Xue, F.; Zhu, X. Effects of propofol on the development of cancer in humans. Cell Prolif. 2020, 53, e12867. [Google Scholar] [CrossRef] [PubMed]
  8. Cho, J.S.; Lee, M.H.; Kim, S.I.; Park, S.; Park, H.S.; Oh, E.; Lee, J.H.; Koo, B.-N. The effects of perioperative anesthesia and analgesia on immune function in patients undergoing breast cancer resection: A prospective randomized study. Int. J. Med. Sci. 2017, 14, 970–976. [Google Scholar] [CrossRef] [Green Version]
  9. Zhang, L.; Wang, N.; Zhou, S.; Ye, W.; Jing, G.; Zhang, M. Propofol induces proliferation and invasion of gallbladder cancer cells through activation of Nrf2. J. Exp. Clin. Cancer Res. 2012, 31, 66. [Google Scholar] [CrossRef] [Green Version]
  10. Chao, M.; Linlin, S.; Juan, W.; Li, D.; Liu, Y.; Cui, X. Propofol induces proliferation partially via downregulation of p53 protein and promotes migration via activation of the Nrf2 pathway in human breast cancer cell line MDA-MB-231. Oncol. Rep. 2017, 37, 841–848. [Google Scholar] [CrossRef]
  11. Gialeli, C.; Theocharis, A.D.; Karamanos, N.K. Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J. 2010, 278, 16–27. [Google Scholar] [CrossRef]
  12. Mitsiades, N.; Yu, W.-H.; Poulaki, V.; Tsokos, M.; Stamenkovic, I. Matrix metalloproteinase-7-mediated cleavage of Fas ligand protects tumor cells from chemotherapeutic drug cytotoxicity. Cancer Res. 2001, 61, 577–581. [Google Scholar]
  13. Miao, Y.; Zhang, Y.; Wan, H.; Chen, L.; Wang, F. GABA-receptor agonist, propofol inhibits invasion of colon carcinoma cells. Biomed. Pharmacother. 2010, 64, 583–588. [Google Scholar] [CrossRef]
  14. Li, Q.; Zhang, L.; Han, Y.; Jiang, Z.; Wang, Q. Propofol reduces MMPs expression by inhibiting NF-κB activity in human MDA-MB-231 cells. Biomed. Pharmacother. 2012, 66, 52–56. [Google Scholar] [CrossRef]
  15. Zhang, Z.; Zang, M.; Wang, S.; Wang, C. Effects of propofol on human cholangiocarcinoma and the associated mechanisms. Exp. Ther. Med. 2018, 17, 472–478. [Google Scholar] [CrossRef] [Green Version]
  16. Kang, F.; Wang, S.; So, E.C.; Chang, M.; Wong, K.; Cheng, K.S.; Chen, Y.; Huang, B. Propofol may increase caspase and MAPK pathways, and suppress the Akt pathway to induce apoptosis in MA-10 mouse Leydig tumor cells. Oncol. Rep. 2019, 41, 3565–3574. [Google Scholar] [CrossRef] [PubMed]
  17. Chen, L.; Wan, Y.; Liu, Y.; Li, T. Propofol inhibits biological functions of leukaemia stem and differentiated cells through suppressing Wnt/β-catenin and Akt/ mTOR. Clin. Exp. Pharmacol. Physiol. 2020, 47, 127–134. [Google Scholar] [CrossRef]
  18. Yang, C.; Gao, J.; Yan, N.; Wu, B.; Ren, Y.; Li, H.; Liang, J. Propofol inhibits the growth and survival of gastric cancer cells in vitro through the upregulation of ING3. Oncol. Rep. 2017, 37, 587–593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Gao, C.; Ren, C.; Liu, Z.; Zhang, L.; Tang, R.; Li, X. GAS5, a FoxO1-actived long noncoding RNA, promotes propofol-induced oral squamous cell carcinoma apoptosis by regulating the miR-1297-GSK3β axis. Artif. Cells Nanomed. Biotechnol. 2019, 47, 3985–3993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Du, Q.; Liu, J.; Zhang, X.; Zhang, X.; Zhu, H.; Wei, M.; Wang, S. Propofol inhibits proliferation, migration, and invasion but promotes apoptosis by regulation of Sox4 in endometrial cancer cells. Braz. J. Med Biol. Res. 2018, 51. [Google Scholar] [CrossRef] [Green Version]
  21. Wang, H.; Zhao, L.; Wu, J.; Hong, J.; Wang, S. Propofol induces ROS-mediated intrinsic apoptosis and migration in triple-negative breast cancer cells. Oncol. Lett. 2020, 20, 810–816. [Google Scholar] [CrossRef]
  22. Tanaka, T.; Takabuchi, S.; Nishi, K.; Oda, S.; Wakamatsu, T.; Daijo, H.; Fukuda, K.; Hirota, K. The intravenous anesthetic propofol inhibits lipopolysaccharide-induced hypoxia-inducible factor 1 activation and suppresses the glucose metabolism in macrophages. J. Anesth. 2009, 24, 54–60. [Google Scholar] [CrossRef]
  23. Ecimovic, P.; Murray, D.; Doran, P.; McDonald, J.; Lambert, D.; Buggy, D.J. Direct effect of morphine on breast cancer cell function in vitro: Role of the NET1 gene. Br. J. Anaesth. 2011, 107, 916–923. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ecimovic, P.; Murray, D.; Doran, P.; Buggy, D.J. Propofol and bupivacaine in breast cancer cell function in vitro—Role of the NET1 gene. Anticancer. Res. 2014, 34, 1321–1331. [Google Scholar] [PubMed]
  25. Ecimovic, P.; McHugh, B.; Murray, D.; Doran, P.; Buggy, D. Direct effect of sevoflurane on breast cancer cell function in vitro: BAPCPC1–1. Eur. J. Anaesthesiol. 2010, 27, 1. [Google Scholar] [CrossRef]
  26. Kawaraguchi, Y.; Horikawa, Y.T.; Murphy, A.N.; Murray, F.; Miyanohara, A.; Ali, S.S.; Head, B.P.; Patel, P.M.; Roth, D.M. Patel, H.H. Volatile anesthetics protect cancer cells against tumor necrosis factor-related apoptosis- inducing ligand-induced apoptosis via caveolins. Anesthesiology 2011, 115, 499–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Enlund, M.; Berglund, A.; Andreasson, K.; Cicek, C.; Enlund, A.; Bergkvist, L. The choice of anaesthetic—Sevoflurane or propofol—and outcome from cancer surgery: A retrospective analysis. Upsala J. Med Sci. 2014, 119, 251–261. [Google Scholar] [CrossRef] [Green Version]
  28. Lee, J.H.; Kang, S.H.; Kim, Y.; Kim, H.-A.; Kim, B.S. Effects of propofol-based total intravenous anesthesia on recurrence and overall survival in patients after modified radical mastectomy: A retrospective study. Korean J. Anesthesiol. 2016, 69, 126–132. [Google Scholar] [CrossRef] [PubMed]
  29. Kim, M.; Kim, D.W.; Kim, J.H.; Lee, K.-Y.; Park, S.; Yoo, Y.C. Does the type of anesthesia really affect the recurrence-free survival after breast cancer surgery? Oncotarget 2017, 8, 90477–90487. [Google Scholar] [CrossRef]
  30. Yoo, S.; Lee, H.-B.; Han, W.; Noh, D.-Y.; Park, S.-K.; Kim, W.H.; Kim, J.-T. Total intravenous anesthesia versus inhalation anesthesia for breast cancer surgery: A retrospective cohort study. Anesthesiology 2019, 130, 31–40. [Google Scholar] [CrossRef]
  31. Huang, Y.-H.; Lee, M.-S.; Lou, Y.-S.; Lai, H.-C.; Yu, J.-C.; Lu, C.-H.; Wong, C.-S.; Wu, Z.-F. Propofol-based total intravenous anesthesia did not improve survival compared to desflurane anesthesia in breast cancer surgery. PLoS ONE 2019, 14, e0224728. [Google Scholar] [CrossRef] [Green Version]
  32. Shiono, S.; Shibata, S.C.; Kabata, D.; Shintani, A.; Ikeda, T.; Fujino, Y. Comparison of 1-year recurrence-free survival between sevoflurane and propofol use for general anesthesia management in primary breast cancer surgery. J. Anesth. 2020, 34, 694–701. [Google Scholar] [CrossRef] [PubMed]
  33. Snyder, G.L.; Greenberg, S. Effect of anaesthetic technique and other perioperative factors on cancer recurrence. Br. J. Anaesth. 2010, 105, 106–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Gong, L.; Qin, Q.; Zhou, L.; Ouyang, W.; Li, Y.; Wu, Y.; Li, Y. Effects of fentanyl anesthesia and sufentanil anesthesia on regulatory T cells frequencies. Int. J. Clin. Exp. Pathol. 2014, 7, 7708–7716. [Google Scholar] [PubMed]
  35. Sacerdote, P.; Gaspani, L.; Rossoni, G.; Panerai, A.; Bianchi, M. Effect of the opioid remifentanil on cellular immune response in the rat. Int. Immunopharmacol. 2001, 1, 713–719. [Google Scholar] [CrossRef]
  36. Franchi, S.; Moretti, S.; Castelli, M.; Lattuada, D.; Scavullo, C.; Panerai, A.E.; Sacerdote, P. Mu opioid receptor activation modulates Toll like receptor 4 in murine macrophages. Brain Behav. Immun. 2012, 26, 480–488. [Google Scholar] [CrossRef] [PubMed]
  37. Cheng, S.; Guo, M.; Liu, Z.; Fu, Y.; Wu, H.; Wang, C.; Cao, M. Morphine Promotes the Angiogenesis of Postoperative Recurrent Tumors and Metastasis of Dormant Breast Cancer Cells. Pharmacol. 2019, 104, 276–286. [Google Scholar] [CrossRef] [PubMed]
  38. Heaney, Á.; Buggy, D.J. Can anaesthetic and analgesic techniques affect cancer recurrence or metastasis? Br. J. Anaesth. 2012, 109, i17–i28. [Google Scholar] [CrossRef] [Green Version]
  39. Gupta, K.; Kshirsagar, S.; Chang, L.; Schwartz, R.; Law, P.-Y.; Yee, D.; Hebbel, R.P. Morphine stimulates angiogenesis by activating proangiogenic and survival-promoting signaling and promotes breast tumor growth. Cancer Res. 2002, 62, 4491–4498. [Google Scholar] [PubMed]
  40. Kocak, N.; Ozen, F.; Yildirim, I.H.; Duran, Y. Fentanyl Inhibits Tumorigenesis from Human Breast Stem Cells by Inducing Apoptosis. Asian Pac. J. Cancer Prev. 2017, 18, 735–739. [Google Scholar]
  41. Sacerdote, P.; Bianchi, M.; Gaspani, L.; Manfredi, B.; Maucione, A.; Terno, G.; Ammatuna, M.; Panerai, A.E. The Effects of Tramadol and Morphine on Immune Responses and Pain After Surgery in Cancer Patients. Anesth. Analg. 2000, 90, 1411–1414. [Google Scholar] [CrossRef] [Green Version]
  42. Xia, M.; Tong, J.H.; Zhou, Z.Q.; Duan, M.L.; Xu, J.G.; Zeng, H.J.; Wang, S.H. Tramadol inhibits proliferation, migration and invasion via α2-adrenoceptor signaling in breast cancer cells. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 157–165. [Google Scholar]
  43. Kim, M.H.; Oh, J.E.; Park, S.; Kim, J.H.; Lee, K.-Y.; Bai, S.J.; Song, H.; Hwang, H.J.; Kim, D.W.; Yoo, Y.C. Tramadol use is associated with enhanced postoperative outcomes in breast cancer patients: A retrospective clinical study with in vitro confirmation. Br. J. Anaesth. 2019, 123, 865–876. [Google Scholar] [CrossRef] [PubMed]
  44. Versyck, B.; van Geffen, G.-J.; Chin, K.-J. Analgesic efficacy of the Pecs II block: A systematic review and meta-analysis. Anaesthesia 2019, 74, 663–673. [Google Scholar] [CrossRef] [PubMed]
  45. Exadaktylos, A.K.; Buggy, D.J.; Moriarty, D.C.; Mascha, E.; Sessler, D.I. Can anesthetic technique for primary breast cancer surgery affect recurrence or metastasis? Anesthesiology 2006, 105, 660–664. [Google Scholar] [CrossRef] [Green Version]
  46. Starnes-Ott, K.; Goravanchi, F.; Meininger, J.C. Anesthetic Choices and Breast Cancer Recurrence: A Retrospective Pilot Study of Patient, Disease, and Treatment Factors. Crit. Care Nurs. Q. 2015, 38, 200–210. [Google Scholar] [CrossRef] [PubMed]
  47. Kairaluoma, P.; Mattson, J.; Heikkilä, P.; Pere, P.; Leidenius, M. Perioperative Paravertebral Regional Anaesthesia and Breast Cancer Recurrence. Anticancer Res. 2016, 36, 415–418. [Google Scholar]
  48. Tsigonis, A.M.; Al-Hamadani, M.; Linebarger, J.H.; Vang, C.A.; Krause, F.J.; Johnson, J.M.; Marchese, E.; Marcou, K.A.; Hudak, J.M.; Landercasper, J. Are cure rates for breast cancer improved by local and regional anesthesia? Reg. Anesth. Pain Med. 2016, 41, 339–347. [Google Scholar] [CrossRef] [PubMed]
  49. Cata, J.P.; Mac Gregor, M.C.; Valero, V.; Black, W.; Black, D.M.; Goravanchi, F.; Ifeanyi, I.C.; Hernandez, M.; Rodriguez-Restrepo, A.; Gottumukkala, V. The Impact of Paravertebral Block Analgesia on Breast Cancer Survival After Surgery. Reg. Anesth. Pain Med. 2016, 41, 696–703. [Google Scholar] [CrossRef] [PubMed]
  50. Finn, D.M.; Ilfeld, B.M.; Unkart, J.; Madison, S.J.; Suresh, P.J.; Sandhu, N.P.S.; Kormylo, N.J.; Malhotra, N.; Loland, V.J.; Wallace, M.S.; et al. Post-mastectomy cancer recurrence with and without a continuous paravertebral block in the immediate postoperative period: A prospective multi-year follow-up pilot study of a randomized, triple-masked, placebo-controlled investigation. J. Anesth. 2017, 31, 374–379. [Google Scholar] [CrossRef] [PubMed]
  51. I Sessler, D.; Pei, L.; Huang, Y.; Fleischmann, E.; Marhofer, P.; Kurz, A.; Mayers, D.B.; A Meyer-Treschan, T.; Grady, M.; Tan, E.Y.; et al. Recurrence of breast cancer after regional or general anaesthesia: A randomised controlled trial. Lancet 2019, 394, 1807–1815. [Google Scholar] [CrossRef]
  52. Pérez-González, O.; Cuéllar-Guzmán, L.F.; Soliz, J.; Cata, J.P. Impact of Regional Anesthesia on Recurrence, Metastasis, and Immune Response in Breast Cancer Surgery: A Systematic Review of the Literature. Reg. Anesth. Pain Med. 2017, 42, 751–756. [Google Scholar] [CrossRef] [PubMed]
  53. Kim, R. Anesthetic technique and cancer recurrence in oncologic surgery: Unraveling the puzzle. Cancer Metastasis Rev. 2017, 36, 159–177. [Google Scholar] [CrossRef] [PubMed]
  54. Freeman, J.; Crowley, P.D.; Foley, A.G.; Gallagher, H.C.; Iwasaki, M.; Ma, D.; Buggy, D.J. Effect of Perioperative Lidocaine, Propofol and Steroids on Pulmonary Metastasis in a Murine Model of Breast Cancer Surgery. Cancers 2019, 11, 613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Wall, T.P.; Crowley, P.D.; Sherwin, A.; Foley, A.G.; Buggy, D.J. Effects of Lidocaine and Src Inhibition on Metastasis in a Murine Model of Breast Cancer Surgery. Cancers 2019, 11, 1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Jiang, Y.; Gou, H.; Zhu, J.; Tian, S.; Yu, L. Lidocaine inhibits the invasion and migration of TRPV6-expressing cancer cells by TRPV6 downregulation. Oncol. Lett. 2016, 12, 1164–1170. [Google Scholar] [CrossRef] [Green Version]
  57. Li, K.; Yang, J.; Han, X. Lidocaine Sensitizes the Cytotoxicity of Cisplatin in Breast Cancer Cells via Up-Regulation of RARβ2 and RASSF1A Demethylation. Int. J. Mol. Sci. 2014, 15, 3519. [Google Scholar] [CrossRef]
  58. Chang, Y.-C.; Liu, C.-L.; Chen, M.-J.; Hsu, Y.-W.; Chen, S.-N.; Lin, C.-H.; Chen, C.-M.; Yang, F.-M.; Hu, M.-C. Local Anesthetics Induce Apoptosis in Human Breast Tumor Cells. Anesth. Analg. 2014, 118, 116–124. [Google Scholar] [CrossRef] [Green Version]
  59. D’Agostino, G.; Saporito, A.; Cecchinato, V.; Silvestri, Y.; Borgeat, A.; Anselmi, L.; Uguccioni, M. Lidocaine inhibits cytoskeletal remodelling and human breast cancer cell migration. Br. J. Anaesth. 2018, 121, 962–968. [Google Scholar] [CrossRef] [Green Version]
  60. Mammoto, T.; Higashiyama, S.; Mukai, M.; Mammoto, A.; Ayaki, M.; Mashimo, T.; Hayashi, Y.; Kishi, Y.; Nakamura, H.; Akedo, H. Infiltration anesthetic lidocaine inhibits cancer cell invasion by modulating ectodomain shedding of heparin-binding epidermal growth factor-like growth factor (HB-EGF). J. Cell. Physiol. 2002, 192, 351–358. [Google Scholar] [CrossRef]
  61. Ramirez, M.F.; Tran, P.; Cata, J.P. The Effect of Clinically Therapeutic Plasma Concentrations of Lidocaine on Natural Killer Cell Cytotoxicity. Reg. Anesth. Pain Med. 2015, 40, 43–48. [Google Scholar] [CrossRef] [PubMed]
  62. Galoș, E.V.; Tat, T.-F.F.; Popa, R.; Efrimescu, C.-I.I.; Finnerty, D.; Buggy, D.J.; Ionescu, D.C.; Mihu, C.M. Neutrophil extracellular trapping and angiogenesis biomarkers after intravenous or inhalation anaesthesia with or without intravenous lidocaine for breast cancer surgery: A prospective, randomised trial. Br. J. Anaesth. 2020, 125, 712–721. [Google Scholar] [CrossRef]
  63. Forget, P.; Machiels, J.-P.; Coulie, P.G.; Berlière, M.; Poncelet, A.J.; Tombal, B.; Stainier, A.; Legrand, C.; Canon, J.-L.; Kremer, Y.; et al. Neutrophil:Lymphocyte Ratio and Intraoperative Use of Ketorolac or Diclofenac are Prognostic Factors in Different Cohorts of Patients Undergoing Breast, Lung, and Kidney Cancer Surgery. Ann. Surg. Oncol. 2013, 20, 650–660. [Google Scholar] [CrossRef] [PubMed]
  64. Forget, P.; Bouche, G.; Duhoux, F.P.; Coulie, P.G.; Decloedt, J.; Dekleermaker, A.; Guillaume, J.-E.; Ledent, M.; Machiels, J.-P.; Mustin, V.; et al. Intraoperative ketorolac in high-risk breast cancer patients. A prospective, randomized, placebo-controlled clinical trial. PLoS ONE 2019, 14, e0225748. [Google Scholar] [CrossRef] [PubMed]
  65. Li, B.; Li, Y.; Tian, S.; Wang, H.; Wu, H.; Zhang, A.; Gao, C. Anti-inflammatory Effects of Perioperative Dexmedetomidine Administered as an Adjunct to General Anesthesia: A Meta-analysis. Sci. Rep. 2015, 5, 12342. [Google Scholar] [CrossRef] [Green Version]
  66. Lavon, H.; Matzner, P.; Benbenishty, A.; Sorski, L.; Rossene, E.; Haldar, R.; Elbaz, E.; Cata, J.; Gottumukkala, V.; Ben-Eliyahu, S. Dexmedetomidine promotes metastasis in rodent models of breast, lung, and colon cancers. Br. J. Anaesth. 2018, 120, 188–196. [Google Scholar] [CrossRef] [Green Version]
  67. Xia, M.; Ji, N.-N.; Duan, M.-L.; Tong, J.-H.; Xu, J.-G.; Zhang, Y.-M.; Wang, S.-H. Dexmedetomidine regulate the malignancy of breast cancer cells by activating α2-adrenoceptor/ERK signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 3500–3506. [Google Scholar]
  68. Cata, J.P.; Singh, V.; Lee, B.M.; Villarreal, J.; Mehran, J.R.; Yu, J.; Gottumukkala, V.; Lavon, H.; Ben-Eliyahu, S. Intraoperative use of dexmedetomidine is associated with decreased overall survival after lung cancer surgery. J. Anaesthesiol. Clin. Pharmacol. 2017, 33, 317–323. [Google Scholar] [CrossRef]
  69. Cole, S.W.; Sood, A.K. Molecular Pathways: Beta-Adrenergic Signaling in Cancer. Clin. Cancer Res. 2012, 18, 1201–1206. [Google Scholar] [CrossRef] [Green Version]
  70. Phadke, S.; Clamon, G. Beta blockade as adjunctive breast cancer therapy: A review. Crit. Rev. Oncol. 2019, 138, 173–177. [Google Scholar] [CrossRef]
  71. Coelho, M.; Soares-Silva, C.; Brandão, D.; Marino, F.; Cosentino, M.; Ribeiro, L. β-Adrenergic modulation of cancer cell proliferation: Available evidence and clinical perspectives. J. Cancer Res. Clin. Oncol. 2017, 143, 275–291. [Google Scholar] [CrossRef]
  72. Shakhar, G.; Ben-Eliyahu, S. In vivo beta-adrenergic stimulation suppresses natural killer activity and compromises resistance to tumor metastasis in rats. J. Immunol. 1998, 160, 3251–3258. [Google Scholar]
  73. Chung, J.F.; Lee, S.J.; Sood, A.K. Immunological and pleiotropic effects of individual β- blockers and their relevance in cancer therapies. Expert Opin. Investig. Drugs 2016, 25, 501–505. [Google Scholar] [CrossRef] [Green Version]
  74. Kohm, A.P.; Sanders, V.M. Norepinephrine and β2-adrenergic receptor stimulation regulate CD4+ T and B lymphocyte function in vitro and in vivo. Pharmacol. Rev. 2001, 53, 487–525. [Google Scholar]
  75. Kang, Y.; Nagaraja, A.; Armaiz-Pena, G.N.; Dorniak, P.L.; Hu, W.; Rupaimoole, R.; Liu, T.; Gharpure, K.; Previs, R.A.; Hansen, J.M.; et al. Adrenergic Stimulation of DUSP1 Impairs Chemotherapy Response in Ovarian Cancer. Clin. Cancer Res. 2016, 22, 1713–1724. [Google Scholar] [CrossRef] [Green Version]
  76. Zhou, L.; Li, Y.; Li, X.; Chen, G.; Liang, H.; Wu, Y.; Tong, J.; Ouyang, W. Propranolol attenuates surgical stress–induced elevation of the regulatory T cell response in patients undergoing radical mastectomy. J. Immunol. 2016, 196, 3460–3469. [Google Scholar] [CrossRef] [Green Version]
  77. Lin, Z.; Wang, L.; Huang, G.; Wang, W.; Lin, H. Propranolol inhibits the activity of PI3K, AKT, and HIF-1α in infantile hemangiomas. Pediatr. Surg. Int. 2018, 34, 1233–1238. [Google Scholar] [CrossRef] [PubMed]
  78. Ganz, P.A.; Habel, L.A.; Weltzien, E.K.; Caan, B.; Cole, S.W. Examining the influence of beta blockers and ACE inhibitors on the risk for breast cancer recurrence: Results from the LACE cohort. Breast Cancer Res. Treat. 2011, 129, 549–556. [Google Scholar] [CrossRef] [Green Version]
  79. Melhem-Bertrandt, A.; Chavez-MacGregor, M.; Lei, X.; Brown, E.N.; Lee, R.T.; Meric-Bernstam, F.; Sood, A.K.; Conzen, S.D.; Hortobagyi, G.N.; Gonzalez-Angulo, A.-M. Beta-blocker use is associated with improved relapse-free survival in patients with triple-negative breast cancer. J. Clin. Oncol. 2011, 29, 2645–2652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Botteri, E.; Munzone, E.; Rotmensz, N.; Cipolla, C.; De Giorgi, V.; Santillo, B.; Zanelotti, A.; Adamoli, L.; Colleoni, M.; Viale, G.; et al. Therapeutic effect of β-blockers in triple-negative breast cancer postmenopausal women. Breast Cancer Res. Treat. 2013, 140, 567–575. [Google Scholar] [CrossRef] [PubMed]
  81. Sakellakis, M.; Kostaki, A.; Starakis, I.; Koutras, A. β-Blocker Use and Risk of Recurrence in Patients with Early Breast Cancer. Chemotherapy 2015, 60, 288–289. [Google Scholar] [CrossRef] [PubMed]
  82. Chen, L.; Chubak, J.; Boudreau, D.M.; Barlow, W.E.; Weiss, N.S.; Li, C.I. Use of Antihypertensive Medications and Risk of Adverse Breast Cancer Outcomes in a SEER–Medicare Population. Cancer Epidemiol. Biomark. Prev. 2017, 26, 1603–1610. [Google Scholar] [CrossRef] [Green Version]
  83. Powe, D.G.; Voss, M.J.; Zänker, K.S.; Habashy, H.O.; Green, A.R.; Ellis, I.; Entschladen, F. Beta-Blocker Drug Therapy Reduces Secondary Cancer Formation in Breast Cancer and Improves Cancer Specific Survival. Oncotarget 2010, 1, 628–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Sørensen, G.V.; Ganz, P.A.; Cole, S.W.; Pedersen, L.A.; Sørensen, H.T.; Cronin-Fenton, D.P.; Garne, J.P.; Christiansen, P.M.; Lash, T.L.; Ahern, T. Use of β-Blockers, Angiotensin-Converting Enzyme Inhibitors, Angiotensin II Receptor Blockers, and Risk of Breast Cancer Recurrence: A Danish Nationwide Prospective Cohort Study. J. Clin. Oncol. 2013, 31, 2265–2272. [Google Scholar] [CrossRef] [PubMed]
  85. Boudreau, D.M.; Yu, O.; Chubak, J.; Wirtz, H.S.; Bowles, E.J.A.; Fujii, M.; Buist, D.S.M. Comparative safety of cardiovascular medication use and breast cancer outcomes among women with early stage breast cancer. Breast Cancer Res. Treat. 2014, 144, 405–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Li, C.; Li, T.; Tang, R.; Yuan, S.; Zhang, W. β-Blocker use is not associated with improved clinical outcomes in women with breast cancer: A meta-analysis. Biosci. Rep. 2020, 40. [Google Scholar] [CrossRef]
  87. Childers, W.K.; Hollenbeak, C.S.; Cheriyath, P. β-blockers reduce breast cancer recurrence and breast cancer death: A meta-analysis. Clin. Breast Cancer 2015, 15, 426–431. [Google Scholar] [CrossRef]
  88. Kim, H.Y.; Jung, Y.J.; Lee, S.H.; Jung, H.J. Pak, K. Is Beta-Blocker Use Beneficial in Breast Cancer? A Meta-Analysis. Oncology 2017, 92, 264–268. [Google Scholar] [CrossRef] [PubMed]
  89. Baek, A.E.; Nelson, E.R. The Contribution of Cholesterol and Its Metabolites to the Pathophysiology of Breast Cancer. Horm. Cancer 2016, 7, 219–228. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. DuSell, C.D.; Umetani, M.; Shaul, P.W.; Mangelsdorf, D.J.; McDonnell, D.P. 27-Hydroxycholesterol is an endogenous selective estrogen receptor modulator. Mol. Endocrinol. 2008, 22, 65–77. [Google Scholar] [CrossRef] [Green Version]
  91. Alikhani, N.; Ferguson, R.D.; Novosyadlyy, R.; Gallagher, E.J.; Scheinman, E.J.; Yakar, S.; LeRoith, D. Mammary tumor growth and pulmonary metastasis are enhanced in a hyperlipidemic mouse model. Oncogene 2012, 32, 961–967. [Google Scholar] [CrossRef] [Green Version]
  92. Strachan, D.C.; Ruffell, B.; Oei, Y.; Bissell, M.J.; Coussens, L.M.; Pryer, N.; Daniel, D. CSF1R inhibition delays cervical and mammary tumor growth in murine models by attenuating the turnover of tumor-associated macrophages and enhancing infiltration by CD8+T cells. OncoImmunology 2013, 2, e26968. [Google Scholar] [CrossRef] [Green Version]
  93. Villablanca, E.J.; Raccosta, L.; Zhou, D.; Fontana, R.; Maggioni, D.; Negro, A.; Sanvito, F.; Ponzoni, M.; Valentinis, B.; Bregni, M.; et al. Tumor- mediated liver X receptor-α activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens antitumor responses. Nat. Med. 2010, 16, 98–105. [Google Scholar] [CrossRef] [PubMed]
  94. Bensinger, S.J.; Bradley, M.N.; Joseph, S.B.; Zelcer, N.; Janssen, E.M.; Hausner, M.A.; Shih, R.; Parks, J.S.; Edwards, P.A.; Jamieson, B.D.; et al. LXR Signaling Couples Sterol Metabolism to Proliferation in the Acquired Immune Response. Cell 2008, 134, 97–111. [Google Scholar] [CrossRef] [Green Version]
  95. Brewer, T.M.; Masuda, H.; Liu, D.D.; Shen, Y.; Liu, P.; Iwamoto, T.; Kai, K.; Barnett, C.M.; Woodward, W.A.; Reuben, J.M.; et al. Statin use in primary inflammatory breast cancer: A cohort study. Br. J. Cancer 2013, 109, 318–324. [Google Scholar] [CrossRef] [Green Version]
  96. Chae, Y.K.; Valsecchi, M.E.; Kim, J.; Bianchi, A.L.; Khemasuwan, D.; Desai, A.; Tester, W. Reduced risk of breast cancer recurrence in patients using ACE inhibitors, ARBs, and/or statins. Cancer Invest. 2011, 29, 585–593. [Google Scholar] [CrossRef]
  97. Li, Y.R.; Ro, V.; Steel, L.; Carrigan, E.; Nguyen, J.; Williams, A.; So, A.; Tchou, J. Impact of long-term lipid-lowering therapy on clinical outcomes in breast cancer. Breast Cancer Res. Treat. 2019, 176, 669–677. [Google Scholar] [CrossRef] [PubMed]
  98. Sakellakis, M.; Akinosoglou, K.; Kostaki, A.; Spyropoulou, D.; Koutras, A. Statins and risk of breast cancer recurrence. Breast Cancer Targets Ther. 2016, 8, 199–205. [Google Scholar] [CrossRef] [Green Version]
  99. Shaitelman, S.F.; Stauder, M.C.; Allen, P.; Reddy, S.; Lakoski, S.; Atkinson, B.; Reddy, J.; Amaya, D.; Guerra, W.; Ueno, N.; et al. Impact of statin use on outcomes in triple negative breast cancer. J. Cancer 2017, 8, 2026–2032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Ahern, T.P.; Pedersen, L.; Tarp, M.; Cronin-Fenton, D.P.; Garne, J.P.; Silliman, R.A.; Sørensen, H.T.; Lash, T.L. Statin prescriptions and breast cancer recurrence risk: A Danish nationwide prospective cohort study. JNCI J. Natl. Cancer Inst. 2011, 103, 1461–1468. [Google Scholar] [CrossRef] [Green Version]
  101. Harborg, S.; Heide-Jørgensen, U.; Ahern, T.P.; Ewertz, M.; Cronin-Fenton, D.; Borgquist, S. Statin use and breast cancer recurrence in postmenopausal women treated with adjuvant aromatase inhibitors: A Danish population-based cohort study. Breast Cancer Res. Treat. 2020, 183, 153–160. [Google Scholar] [CrossRef]
  102. Kwan, M.L.; Habel, L.A.; Flick, E.D.; Quesenberry, C.P.; Caan, B. Post-diagnosis statin use and breast cancer recurrence in a prospective cohort study of early stage breast cancer survivors. Breast Cancer Res. Treat. 2007, 109, 573–579. [Google Scholar] [CrossRef] [Green Version]
  103. Tryggvadottir, H.; Huzell, L.; Gustbée, E.; Simonsson, M.; Markkula, A.; Jirström, K.; Rose, C.; Ingvar, C.; Borgquist, S.; Jernström, H. Interactions Between ABCB1 Genotype and Preoperative Statin Use Impact Clinical Outcomes Among Breast Cancer Patients. Front. Oncol. 2018, 8, 428. [Google Scholar] [CrossRef] [Green Version]
  104. Lv, H.; Shi, D.; Fei, M.; Chen, Y.; Xie, F.; Wang, Z.; Wang, Y.; Hu, P. Association Between Statin Use and Prognosis of Breast Cancer: A Meta-Analysis of Cohort Studies. Front. Oncology 2020, 10, 1461–1468. [Google Scholar] [CrossRef] [PubMed]
  105. Manthravadi, S.; Shrestha, A.; Madhusudhana, S. Impact of statin use on cancer recurrence and mortality in breast cancer: A systematic review and meta-analysis [Internet]. Int. J. Cancer 2016, 139, 1281–1288. [Google Scholar] [CrossRef]
  106. Mansourian, M.; Haghjoo-Javanmard, S.; Eshraghi, A.; Vaseghi, G.; Hayatshahi, A.; Thomas, J. Statins use and risk of breast cancer recurrence and death: A systematic review and meta-analysis of observational studies. J. Pharm. Pharm. Sci. 2016, 19, 72–81. [Google Scholar] [CrossRef] [PubMed]
  107. Yap, A.; The Global Onco-Anesthesia Research Collaboration Group; Lopez-Olivo, M.; Dubowitz, J.; Hiller, J.; Riedel, B. Anesthetic technique and cancer outcomes: A meta-analysis of total intravenous versus volatile anesthesia. Can. J. Anesth. /J. Can. D’anesthésie 2019, 66, 546–561. [Google Scholar] [CrossRef] [Green Version]
  108. Inamura, Y.; Miyamae, M.; Sugioka, S.; Kaneda, K.; Okusa, C.; Onishi, A.; Domae, N.; Kotani, J.; Figueredo, V.M. Aprotinin Abolishes Sevoflurane Postconditioning by Inhibiting Nitric Oxide Production and Phosphorylation of Protein Kinase C-δ and Glycogen Synthase Kinase 3β. Anesthesiology 2009, 111, 1036–1043. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Lemoine, S.; Zhu, L.; Buléon, C.; Massetti, M.; Gérard, J.-L.; Galera, P.; Hanouz, J.-L. Mechanisms involved in the desflurane-induced post-conditioning of isolated human right atria from patients with type 2 diabetes. Br. J. Anaesth. 2011, 107, 510–518. [Google Scholar] [CrossRef] [Green Version]
  110. Lemoine, S.; Zhu, L.; Beauchef, G.; Lepage, O.; Babatasi, G.; Ivascau, C.; Massetti, M.; Galera, P.; Gérard, J.-L.; Hanouz, J.-L. Role of 70-kDa Ribosomal Protein S6 Kinase, Nitric Oxide Synthase, Glycogen Synthase Kinase-3β, and Mitochondrial Permeability Transition Pore in Desflurane-induced Postconditioning in Isolated Human Right Atria. Anesthesiol. 2010, 112, 1355–1363. [Google Scholar] [CrossRef] [Green Version]
  111. Chen, Y.; Zhou, Z.-F.; Wang, Y. Prediction and analysis of weighted genes in isoflurane induced general anesthesia based on network analysis. Int. J. Neurosci. 2019, 130, 610–620. [Google Scholar] [CrossRef]
  112. Farooqi, A.A.; Adylova, A.; Sabitaliyevich, U.Y.; Attar, R.; Sohail, M.I.; Yilmaz, S. Recent updates on true potential of an anesthetic agent as a regulator of cell signaling pathways and non-coding RNAs in different cancers: Focusing on the brighter side of propofol. Gene 2020, 737, 144452. [Google Scholar] [CrossRef] [PubMed]
  113. Inoue, J.; Inazawa, J. Cancer-associated miRNAs and their therapeutic potential. J. Hum. Genet. 2021, 1–9. [Google Scholar] [CrossRef]
  114. Gibbs, K.M.; Izer, J.M.; Reeves, W.B.; Wilson, R.P.; Cooper, T.K. Effects of General Anesthesia on 2 Urinary Biomarkers of Kidney Injury—Hepatitis A Virus Cellular Receptor 1 and Lipocalin 2—in Male C57BL/6J Mice. J. Am. Assoc. Lab. Anim. Sci. 2019, 58, 21–29. [Google Scholar] [CrossRef] [PubMed]
  115. Ding, G.; Fang, J.; Tong, S.; Qu, L.; Jiang, H.; Ding, Q.; Liu, J. Over-expression of lipocalin 2 promotes cell migration and invasion through activating ERK signaling to increase SLUG expression in prostate cancer. Prostate 2015, 75, 957–968. [Google Scholar] [CrossRef] [PubMed]
  116. Tung, M.C.; Hsieh, S.C.; Yang, S.F.; Cheng, C.W.; Tsai, R.T.; Wang, S.C.; Hsieh, Y.H. Knockdown of lipocalin-2 suppresses the growth and invasion of prostate cancer cells. Prostate 2013, 73, 1281–1290. [Google Scholar] [CrossRef]
  117. Liu, Z.; Zhang, J.; Hong, G.; Quan, J.; Zhang, L.; Yu, M. Propofol inhibits growth and invasion of pancreatic cancer cells through regulation of the miR-21/Slug signaling pathway. Am. J. Transl. Res. 2016, 8, 4120–4133. [Google Scholar] [PubMed]
  118. Fernández, C.A.; Yan, L.; Louis, G.; Yang, J.; Kutok, J.L.; Moses, M.A. The Matrix Metalloproteinase-9/Neutrophil Gelatinase-Associated Lipocalin Complex Plays a Role in Breast Tumor Growth and Is Present in the Urine of Breast Cancer Patients. Clin. Cancer Res. 2005, 11, 5390–5395. [Google Scholar] [CrossRef] [Green Version]
  119. Yang, J.; McNeish, B.; Butterfield, C.; Moses, M.A. Lipocalin 2 is a novel regulator of angiogenesis in human breast cancer. FASEB J. 2013, 27, 45–50. [Google Scholar] [CrossRef] [Green Version]
  120. Yang, J.; Moses, M.A. Lipocalin 2: A multifaceted modulator of human cancer. Cell Cycle 2009, 8, 2347–2352. [Google Scholar] [CrossRef]
  121. Yang, J.; Bielenberg, D.R.; Rodig, S.J.; Doiron, R.; Clifton, M.C.; Kung, A.L.; Moses, M.A. Lipocalin 2 promotes breast cancer progression. Proc. Natl. Acad. Sci. USA 2009, 106, 3913–3918. [Google Scholar] [CrossRef] [Green Version]
  122. Cheng, G.; Sun, X.; Wang, J.; Xiao, G.; Wang, X.; Fan, X.; Zu, L.; Hao, M.; Qu, Q.; Mao, Y.; et al. HIC1 Silencing in Triple-Negative Breast Cancer Drives Progression through Misregulation of LCN2. Cancer Res. 2014, 74, 862–872. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Leng, X.; Wu, Y.; Arlinghaus, R.B. Relationships of lipocalin 2 with breast tumorigenesis and metastasis. J. Cell. Physiol. 2010, 226, 309–314. [Google Scholar] [CrossRef]
  124. Ören, B.; Urosevic, J.; Mertens, C.; Mora, J.; Guiu, M.; Gomis, R.R.; Weigert, A.; Schmid, T.; Grein, S.; Brüne, B.; et al. Tumour stroma-derived lipocalin-2 promotes breast cancer metastasis. J. Pathol. 2016, 239, 274–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hu, C.; Yang, K.; Li, M.; Huang, W.; Zhang, F.; Wang, H. Lipocalin 2: A potential therapeutic target for breast cancer metastasis. OncoTargets Ther. 2018, 11, 8099–8106. [Google Scholar] [CrossRef] [Green Version]
  126. Guo, P.; Yang, J.; Jia, D.; Moses, M.A.; Auguste, D.T. ICAM-1-Targeted, Lcn2 siRNA- Encapsulating Liposomes are Potent Anti-angiogenic Agents for Triple Negative Breast Cancer. Theranostics 2016, 6, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Guo, P.; Yang, J.; Huang, J.; Auguste, D.T.; Moses, M.A. Therapeutic genome editing of triple-negative breast tumors using a noncationic and deformable nanolipogel. Proc. Natl. Acad. Sci. USA 2019, 116, 18295–18303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Leng, X.; Ding, T.; Lin, H.; Wang, Y.; Hu, L.; Hu, J.; Feig, B.; Zhang, W.; Pusztai, L.; Symmans, W.F.; et al. Inhibition of Lipocalin 2 Impairs Breast Tumorigenesis and Metastasis. Cancer Res. 2009, 69, 8579–8584. [Google Scholar] [CrossRef] [Green Version]
  129. Roland, C.L.; Harken, A.H.; Sarr, M.G.; Barnett, C.C., Jr. ICAM-1 expression determines malignant potential of cancer. Surgery 2007, 141, 705–707. [Google Scholar] [CrossRef] [PubMed]
  130. Rosette, C.; Roth, R.B.; Oeth, P.; Braun, A.; Kammerer, S.; Ekblom, J.; Denissenko, M.F. Role of ICAM1 in invasion of human breast cancer cells. Carcinog. 2005, 26, 943–950. [Google Scholar] [CrossRef] [Green Version]
  131. Maeda, K.; Kang, S.-M.; Sawada, T.; Nishiguchi, Y.; Yashiro, M.; Ogawa, Y.; Ohira, M.; Ishikawa, T.; Chung, K.H.-Y. Expression of intercellular adhesion molecule-1 and prognosis in colorectal cancer. Oncol. Rep. 2002, 9, 511–514. [Google Scholar] [CrossRef] [PubMed]
  132. Tachimori, A.; Yamada, N.; Sakate, Y.; Yashiro, M.; Maeda, K.; Ohira, M.; Nishino, H.; Hirakawa, K. Up regulation of ICAM-1 gene expression inhibits tumour growth and liver metastasis in colorectal carcinoma. Eur. J. Cancer 2005, 41, 1802–1810. [Google Scholar] [CrossRef] [PubMed]
  133. Maruo, Y.; Gochi, A.; Kaihara, A.; Shimamura, H.; Yamada, T.; Tanaka, N.; Orita, K. ICAM-1 expression and the soluble ICAM-1 level for evaluating the metastatic potential of gastric cancer. Int. J. Cancer 2002, 100, 486–490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Schröder, C.; Witzel, I.; Müller, V.; Krenkel, S.; Wirtz, R.M.; Jänicke, F.; Schumacher, U.; Milde-Langosch, K. Prognostic value of intercellular adhesion molecule (ICAM)-1 expression in breast cancer. J. Cancer Res. Clin. Oncol. 2011, 137, 1193–1201. [Google Scholar] [CrossRef] [PubMed]
  135. A Regidor, P.; Callies, R.; Regidor, M.; E Schindler, A. Expression of the cell adhesion molecules ICAM-1 and VCAM-1 in the cytosol of breast cancer tissue, benign breast tissue and corresponding sera. Eur. J. Gynaecol. Oncol. 1998, 19, 377–383. [Google Scholar]
  136. Köstler, W.J.; Tomek, S.; Brodowicz, T.; Budinsky, A.C.; Flamm, M.; Hejna, M.; Krainer, M.; Wiltschke, C.; Zielinski, C.C. Soluble ICAM-1 in breast cancer: Clinical significance and biological implications. Cancer Immunol. Immunother. 2001, 50, 483–490. [Google Scholar] [CrossRef]
  137. O’Hanlon, D.M.; Fitzsimons, H.; Lynch, J.; Tormey, S.; Malone, C.; Given, H.F. Soluble adhesion molecules (E-selectin, ICAM-1 and VCAM-1) in breast carcinoma. Eur. J. Cancer 2002, 38, 2252–2257. [Google Scholar] [CrossRef]
  138. Piegeler, T.; Votta-Velis, E.G.; Bakhshi, F.R.; Mao, M.; Carnegie, G.; Bonini, M.G.; E Schwartz, D.; Borgeat, A.; Beck-Schimmer, B.; Minshall, R.D. Endothelial barrier protection by local anesthetics: Ropivacaine and lidocaine block tumor necrosis factor-α-induced endothelial cell Src activation. Anesthesiology 2014, 120, 1414–1428. [Google Scholar] [CrossRef] [Green Version]
  139. Piegeler, T.; Votta-Velis, E.G.; Liu, G.; Place, A.T.; E Schwartz, D.; Beck-Schimmer, B.; Minshall, R.D.; Borgeat, A. Antimetastatic potential of amide-linked local anesthetics: Inhibition of lung adenocarcinoma cell migration and inflammatory Src signaling independent of sodium channel blockade. Anesthesiology 2012, 117, 548–559. [Google Scholar] [CrossRef] [Green Version]
  140. Berger-Achituv, S.; Brinkmann, V.; Abu-Abed, U.; Kühn, L.I.; Ben-Ezra, J.; Elhasid, R.; Zychlinsky, A. A proposed role for neutrophil extracellular traps in cancer immunoediting. Front Immunol. 2013, 4, 48. [Google Scholar] [CrossRef] [Green Version]
  141. Dąbrowska, D.; Jabłońska, E.; Garley, M.; Wrona, W.R.; Iwaniuk, A. New Aspects of the Biology of Neutrophil Extracellular Traps. Scand. J. Immunol. 2016, 84, 317–322. [Google Scholar] [CrossRef]
  142. Gonzalez-Aparicio, M.; Alfaro, C. Influence of Interleukin-8 and Neutrophil Extracellular Trap (NET) Formation in the Tumor Microenvironment: Is There a Pathogenic Role? J. Immunol. Res. 2019, 2019, 1–7. [Google Scholar] [CrossRef] [Green Version]
  143. Snoderly, H.T.; Boone, B.A.; Bennewitz, M.F. Neutrophil extracellular traps in breast cancer and beyond: Current perspectives on NET stimuli, thrombosis and metastasis, and clinical utility for diagnosis and treatment. Breast Cancer Res. 2019, 21, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Wang, W.; Zhang, J.; Zheng, N.; Li, L.; Wang, X.; Zeng, Y. The role of neutrophil extracellular traps in cancer metastasis. Clin. Transl. Med. 2020, 10, 199–223. [Google Scholar] [CrossRef] [PubMed]
  145. Tang, F.; Tie, Y.; Tu, C.; Wei, X. Surgical trauma-induced immunosuppression in cancer: Recent advances and the potential therapies. Clin. Transl. Med. 2020, 10, 199–223. [Google Scholar] [CrossRef] [PubMed]
  146. Park, J.; Wysocki, R.; Amoozgar, Z.; Maiorino, L.; Fein, M.R.; Jorns, J.; Schott, A.F.; Kinugasa-Katayama, Y.; Lee, Y.; Won, N.H.; et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl. Med. 2016, 8, 361ra138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Cedervall, J.; Zhang, Y.; Olsson, A.-K. Tumor-Induced NETosis as a Risk Factor for Metastasis and Organ Failure. Cancer Res. 2016, 76, 4311–4315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Demers, M.; Wong, S.L.; Martinod, K.; Gallant, M.; Cabral, J.E.; Wang, Y.; Wagner, D.D. Priming of neutrophils toward NETosis promotes tumor growth. OncoImmunology 2016, 5, e1134073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Demers, M.; Wagner, D.D. NETosis: A New Factor in Tumor Progression and Cancer-Associated Thrombosis. Semin. Thromb. Hemost. 2014, 40, 277–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Zhang, H.; Lv, H.; Weng, M.; Wang, H.; Cata, J.P.; Chen, W.; Miao, C. Preoperative leukocytosis is associated with increased tumor-infiltrating neutrophil extracellular traps and worse outcomes in esophageal cancer. Ann. Transl. Med. 2020, 8, 441. [Google Scholar] [CrossRef]
  151. Rayes, R.F.; Mouhanna, J.G.; Nicolau, I.; Bourdeau, F.; Giannias, B.; Rousseau, S.; Quail, D.; Walsh, L.; Sangwan, V.; Bertos, N.; et al. Primary tumors induce neutrophil extracellular traps with targetable metastasis-promoting effects. JCI Insight 2019, 4. [Google Scholar] [CrossRef] [Green Version]
  152. Meier, A.; Chien, J.; Hobohm, L.; Patras, K.A.; Nizet, V.; Corriden, R. Inhibition of Human Neutrophil Extracellular Trap (NET) Production by Propofol and Lipid Emulsion. Front. Pharmacol. 2019, 10, 323. [Google Scholar] [CrossRef] [Green Version]
  153. Chen, M.-S.; Lin, W.-C.; Yeh, H.-T.; Hu, C.-L.; Sheu, S.-M. Propofol specifically reduces PMA-induced neutrophil extracellular trap formation through inhibition of p-ERK and HOCl. Life Sci. 2019, 221, 178–186. [Google Scholar] [CrossRef]
  154. Kolle, G.; Metterlein, T.; Gruber, M.; Seyfried, T.; Petermichl, W.; Pfaehler, S.-M.; Bitzinger, D.; Wittmann, S.; Bredthauer, A. Potential Impact of Local Anesthetics Inducing Granulocyte Arrest and Altering Immune Functions on Perioperative Outcome. J. Inflamm. Res. 2021, 14, 1–12. [Google Scholar] [CrossRef]
  155. Aghamelu, O.; Buggy, P.; Smith, G.; Inzitari, R.; Wall, T.; Buggy, D.J. Serum NETosis expression and recurrence risk after regional or volatile anaesthesia during breast cancer surgery: A pilot, prospective, randomised single-blind clinical trial. Acta Anaesthesiol. Scand. 2021, 65, 313–319. [Google Scholar] [CrossRef] [PubMed]
  156. Osada, H.; Takahashi, T. MicroRNAs in biological processes and carcinogenesis. Carcinogen 2007, 28, 2–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Zhang, L.; Huang, J.; Yang, N.; Greshock, J.; Megraw, M.; Giannakakis, A.; Liang, S.; Naylor, T.L.; Barchetti, A.; Ward, M.R.; et al. microRNAs exhibit high frequency genomic alterations in human cancer. Proc. Natl. Acad. Sci. USA 2006, 103, 9136–9141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Hossain, A.; Kuo, M.T.; Saunders, G.F. Mir-17-5p Regulates Breast Cancer Cell Proliferation by Inhibiting Translation of AIB1 mRNA. Mol. Cell. Biol. 2006, 26, 8191–8201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Kong, W.; Yang, H.; He, L.; Zhao, J.-J.; Coppola, D.; Dalton, W.S.; Cheng, J.Q. MicroRNA-155 Is Regulated by the Transforming Growth Factor β/Smad Pathway and Contributes to Epithelial Cell Plasticity by Targeting RhoA. Mol. Cell. Biol. 2008, 28, 6773–6784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Kloosterman, W.P.; Plasterk, R.H. The Diverse Functions of MicroRNAs in Animal Development and Disease. Dev. Cell 2006, 11, 441–450. [Google Scholar] [CrossRef] [Green Version]
  161. Stefani, G.; Slack, F. Small non-coding RNAs in animal development. Nat. Rev. Mol. Cell Biol. 2008, 9, 219–230. [Google Scholar] [CrossRef]
  162. Heneghan, H.; Miller, N.; Lowery, A.; Sweeney, K.J.; Newell, J.; Kerin, M. Circulating microRNAs as Novel Minimally Invasive Biomarkers for Breast Cancer. Ann. Surg. 2010, 251, 499–505. [Google Scholar] [CrossRef] [PubMed]
  163. Filipów, S.; Łaczmański, Ł. Blood Circulating miRNAs as Cancer Biomarkers for Diagnosis and Surgical Treatment Response. Front. Genet. 2019, 10, 705–707. [Google Scholar] [CrossRef] [PubMed]
  164. Guo, Y.; Yang, J.; Huang, Q.; Hsueh, C.; Zheng, J.; Wu, C.; Chen, H.; Zhou, L. Circular RNAs and their roles in head and neck cancers. Mol. Cancer 2019, 18, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Xu, S.; Lou, F.; Wu, Y.; Sun, D.-Q.; Zhang, J.-B.; Chen, W.; Ye, H.; Liu, J.-H.; Wei, S.; Zhao, M.-Y.; et al. Circulating tumor DNA identified by targeted sequencing in advanced-stage non-small cell lung cancer patients. Cancer Lett. 2016, 370, 324–331. [Google Scholar] [CrossRef] [PubMed]
  166. Peng, K.; Liu, H.-Y.; Wu, S.-R.; Liu, H.; Zhang, Z.-C.; Ji, F.-H. Does Propofol Anesthesia Lead to Less Postoperative Pain Compared With Inhalational Anesthesia?: A Systematic Review and Meta-analysis. Anesth. Analg. 2016, 123, 846–858. [Google Scholar] [CrossRef]
  167. Zhang, Y.; Yang, L.; Kucherlapati, M.; Chen, F.; Hadjipanayis, A.; Pantazi, A.; Bristow, C.A.; Lee, E.A.; Mahadeshwar, H.S.; Tang, J.; et al. A Pan-Cancer Compendium of Genes Deregulated by Somatic Genomic Rearrangement across More Than 1400 Cases. Cell Rep. 2018, 24, 515–527. [Google Scholar] [CrossRef] [Green Version]
  168. Sun, Y.; Peng, Y.; Ye, L.; Ma, L.; Zou, M.; Cheng, Z. Propofol inhibits proliferation and cisplatin resistance in ovarian cancer cells through regulating the microRNA-374a/forkhead box O1 signaling axis. Mol. Med. Rep. 2020, 21, 1471–1480. [Google Scholar] [CrossRef] [PubMed]
  169. Tian, D.; Tian, M.; Ma, Z.-M.; Zhang, L.-L.; Cui, Y.-F.; Li, J.-L. Anesthetic propofol epigenetically regulates breast cancer trastuzumab resistance through IL-6/miR-149-5p axis. Sci. Rep. 2020, 10, 8858. [Google Scholar] [CrossRef] [PubMed]
  170. Yuan, C.; Gao, W.; Zhou, H.; Miao, X.; Chang, Y.; Wang, L.; Xu, M.; Ni, G. Propofol induces apoptosis of breast cancer cells by downregulation of miR-24 signal pathway. Cancer Biomark. 2018, 21, 513–519. [Google Scholar] [CrossRef]
  171. Liu, X.; Ranganathan, R.; Jiang, S.; Fang, C.; Sun, J.; Kim, S.; Newick, K.; Lo, A.; June, C.H.; Zhao, Y.; et al. A Chimeric Switch-Receptor Targeting PD1 Augments the Efficacy of Second-Generation CAR T Cells in Advanced Solid Tumors. Cancer Res. 2016, 76, 1578–1590. [Google Scholar] [CrossRef] [Green Version]
  172. Buschmann, D.; Brandes, F.; Lindemann, A.; Maerte, M.; Ganschow, P.; Chouker, A.; Schelling, G.; Pfaffl, M.W.; Reithmair, M. Propofol and Sevoflurane Differentially Impact MicroRNAs in Circulating Extracellular Vesicles during Colorectal Cancer Resection: A Pilot Study. Anesthesiology 2020, 132, 107–120. [Google Scholar] [CrossRef] [PubMed]
  173. Deng, X.; Vipani, M.; Liang, G.; Gouda, D.; Wang, B.; Wei, H. Sevoflurane modulates breast cancer cell survival via modulation of intracellular calcium homeostasis. BMC Anesthesiol. 2020, 20, 1–9. [Google Scholar] [CrossRef]
  174. Liu, J.; Yang, L.; Guo, X.; Jin, G.; Wang, Q.; Lv, D.; Liu, J.; Chen, Q.; Song, Q.; Li, B. Sevoflurane suppresses proliferation by upregulating microRNA-203 in breast cancer cells. Mol. Med. Rep. 2018, 18, 455–460. [Google Scholar] [CrossRef]
  175. Li, Y.; Zeng, Q.-G.; Qiu, J.-L.; Pang, T.; Wang, H.; Zhang, X.-X. Sevoflurane inhibits the progression of PTC by downregulating miR-155. Eur. Rev. Med. Pharm. Sci 2019, 23, 6579–6587. [Google Scholar]
  176. Li, L.; Sun, Y.; Zhang, N.; Qiu, X.; Wang, L.; Luo, Q. By regulating miR-182-5p/BCL10/CYCS, sufentanil reduces the apoptosis of umbilical cord mesenchymal stem cells caused by ropivacaine. Biosci. Trends 2019, 13, 49–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Qu, X.; Yang, L.; Shi, Q.; Wang, X.; Wang, D.; Wu, G. Lidocaine inhibits proliferation and induces apoptosis in colorectal cancer cells by upregulating mir-520a-3p and targeting EGFR. Pathol. Res. Pract. 2018, 214, 1974–1979. [Google Scholar] [CrossRef] [PubMed]
  178. Dave, R.S.; Khalili, K. Morphine treatment of human monocyte-derived macrophages induces differential miRNA and protein expression: Impact on inflammation and oxidative stress in the central nervous system. J. Cell. Biochem. 2010, 110, 834–845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Wu, Q.; Zhang, L.; Law, P.-Y.; Wei, L.-N.; Loh, H.H. Long-Term Morphine Treatment Decreases the Association of μ-Opioid Receptor (MOR1) mRNA with Polysomes through miRNA23b. Mol. Pharmacol. 2009, 75, 744–750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Melo, Z.; Ishida, C.; Goldaraz MD la, P.; Rojo, R.; Echavarria, R. Novel roles of non-coding RNAs in opioid signaling and cardioprotection. Non-coding RNA. 2018, 4, 22. [Google Scholar] [CrossRef] [Green Version]
  181. He, Y.; Yang, C.; Kirkmire, C.M.; Wang, Z.J. Regulation of opioid tolerance by let-7 family microRNA targeting the μ opioid receptor. J. Neurosci. 2010, 30, 10251–10258. [Google Scholar] [CrossRef] [Green Version]
  182. Gonzalez-Nunez, V.; Prieto, J.A.N.; Rodríguez, R.E. Morphine modulates cell proliferation through mir133b & mir128 in the neuroblastoma SH-SY5Y cell line. Biochim. Biophys. Acta Mol. Basis Dis. 2014, 1842, 566–572. [Google Scholar] [CrossRef] [Green Version]
  183. Luo, J.; Chen, Y.; Xu, Y.; Tang, M.; Zhang, X. Morphine contributed to the deterioration of cancer via miR-543/MARCKS/FcγR-mediated phagocytosis pathway. J. Pharm. Pharmacol. 2019, 71, 1584–1598. [Google Scholar] [CrossRef]
  184. Chen, X.; Wang, L.; Qu, J.; Guan, N.; Li, J.-Q. Predicting miRNA–disease association based on inductive matrix completion. Bioinform. 2018, 34, 4256–4265. [Google Scholar] [CrossRef]
  185. Kim, S.; Dougherty, E.R.; Shmulevich, I.; Hess, K.R.; Hamilton, S.R.; Trent, J.M.; Fuller, G.N.; Zhang, W. Identification of Combination Gene Sets for Glioma Classification 1 Supported in part by the Tobacco Settlement Funds as appropriated by the Texas State Legislature, by a generous donation from the Michael and Betty Kadoorie Foundation, and by a grant fro. Mol Cancer Ther. 2002, 1, 1229–1236. [Google Scholar]
  186. Lu, J.; Getz, G.; Miska, E.A.; Alvarez-Saavedra, E.; Lamb, J.; Peck, D.; Sweet-Cordero, A.; Ebert, B.L.; Mak, R.H.; Ferrando, A.A.; et al. MicroRNA expression profiles classify human cancers. Nature 2005, 435, 834–838. [Google Scholar] [CrossRef]
  187. Mar-Aguilar, F.; Mendoza-Ramírez, J.A.; Malagón-Santiago, I.; Espino-Silva, P.K.; Santuario-Facio, S.K.; Ruiz-Flores, P.; Baldassarre, G.; Croce, C.M.; Vecchione, A. Serum circulating microRNA profiling for identification of potential breast cancer biomarkers. Dis. Markers 2013, 34, 163–169. [Google Scholar] [CrossRef] [PubMed]
  188. Bautista-Sánchez, D.; Arriaga-Canon, C.; Pedroza-Torres, A.; De La Rosa-Velázquez, I.A.; González-Barrios, R.; Contreras-Espinosa, L.; Montiel-Manríquez, R.; Castro-Hernández, C.; Fragoso-Ontiveros, V.; Álvarez-Gómez, R.M.; et al. The Promising Role of miR-21 as a Cancer Biomarker and Its Importance in RNA-Based Therapeutics. Mol. Ther. Nucleic Acids 2020, 20, 409–420. [Google Scholar] [CrossRef] [PubMed]
  189. Papagiannakopoulos, T.; Shapiro, A.; Kosik, K.S. MicroRNA-21 Targets a Network of Key Tumor-Suppressive Pathways in Glioblastoma Cells. Cancer Res. 2008, 68, 8164–8172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Asangani, I.; Rasheed, S.A.K.; A Nikolova, D.; Leupold, J.H.; Colburn, N.H.; Post, S.; Allgayer, H. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 2007, 27, 2128–2136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Frankel, L.B.; Christoffersen, N.R.; Jacobsen, A.; Lindow, M.; Krogh, A.; Lund, A.H. Programmed cell death 4 (PDCD4) is an important functional target of the microRNA miR-21 in breast cancer cells. J. Biol. Chem. 2008, 283, 1026–1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Meng, F.; Henson, R.; Wehbe–Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 Regulates Expression of the PTEN Tumor Suppressor Gene in Human Hepatocellular Cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Petrocca, F.; Visone, R.; Onelli, M.R.; Shah, M.H.; Nicoloso, M.S.; de Martino, I.; Iliopoulos, D.; Pilozzi, E.; Liu, C.-G.; Negrini, M.; et al. E2F1-regulated microRNAs impair TGFbeta-dependent cell-cycle arrest and apoptosis in gastric cancer. Cancer Cell 2008, 13, 272–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Zhu, S.; Si, M.-L.; Wu, H.; Mo, Y.-Y. MicroRNA-21 Targets the Tumor Suppressor Gene Tropomyosin 1 (TPM1). J. Biol. Chem. 2007, 282, 14328–14336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Fang, H.; Xie, J.; Zhang, M.; Zhao, Z.; Wan, Y.; Yao, Y. miRNA-21 promotes proliferation and invasion of triple-negative breast cancer cells through targeting PTEN. Am. J. Transl. Res. 2017, 9, 953–961. [Google Scholar]
  196. Si, H.; Sun, X.; Chen, Y.; Cao, Y.; Chen, S.; Wang, H.; Hu, C. Circulating microRNA-92a and microRNA-21 as novel minimally invasive biomarkers for primary breast cancer. J. Cancer Res. Clin. Oncol. 2012, 139, 223–229. [Google Scholar] [CrossRef] [Green Version]
  197. Asaga, S.; Kuo, C.; Nguyen, T.; Terpenning, M.; Giuliano, A.E.; Hoon, D.S. Direct Serum Assay for MicroRNA-21 Concentrations in Early and Advanced Breast Cancer. Clin. Chem. 2011, 57, 84–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Gong, C.; Yao, Y.; Wang, Y.; Liu, B.; Wu, W.; Chen, J.; Su, F.; Yao, H.; Song, E. Up-regulation of miR-21 Mediates Resistance to Trastuzumab Therapy for Breast Cancer. J. Biol. Chem. 2011, 286, 19127–19137. [Google Scholar] [CrossRef] [Green Version]
  199. Yadav, P.; Mirza, M.; Nandi, K.; Jain, S.K.; Kaza, R.C.M.; Khurana, N.; Ray, P.C.; Saxena, A. Serum microRNA-21 expression as a prognostic and therapeutic biomarker for breast cancer patients. Tumor Biol. 2016, 37, 15275–15282. [Google Scholar] [CrossRef]
  200. Su, J.-L.; Chen, P.-S.; Johansson, G.; Kuo, M.-L. Function and Regulation of Let-7 Family microRNAs. MicroRNA 2012, 1, 34–39. [Google Scholar] [CrossRef]
  201. Zhao, Y.; Deng, C.; Wang, J.; Xiao, J.; Gatalica, Z.; Recker, R.R.; Xiao, G.G. Let-7 family miRNAs regulate estrogen receptor alpha signaling in estrogen receptor positive breast cancer. Breast Cancer Res. Treat. 2011, 127, 69–80. [Google Scholar] [CrossRef]
  202. Yu, F.; Yao, H.; Zhu, P.; Zhang, X.; Pan, Q.; Gong, C.; Huang, Y.; Hu, X.; Su, F.; Lieberman, J.; et al. let-7 Regulates Self Renewal and Tumorigenicity of Breast Cancer Cells. Cell 2007, 131, 1109–1123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. A Joosse, S.; Müller, V.; Steinbach, B.; Pantel, K.; Schwarzenbach, H. Circulating cell-free cancer-testis MAGE-A RNA, BORIS RNA, let-7b and miR-202 in the blood of patients with breast cancer and benign breast diseases. Br. J. Cancer 2014, 111, 909–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Liu, F.; Kong, X.; Lv, L.; Gao, J. MiR-155 targets TP53INP1 to regulate liver cancer stem cell acquisition and self-renewal. FEBS Lett. 2015, 589, 500–506. [Google Scholar] [CrossRef] [Green Version]
  205. Mattiske, S.; Suetani, R.J.; Neilsen, P.; Callen, D.F. The Oncogenic Role of miR-155 in Breast Cancer. Cancer Epidemiol. Biomark. Prev. 2012, 21, 1236–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Thai, T.-H.; Calado, D.; Casola, S.; Ansel, K.M.; Xiao, C.; Xue, Y.; Murphy, A.; Frendewey, D.; Valenzuela, D.; Kutok, J.L.; et al. Regulation of the Germinal Center Response by MicroRNA-155. Science 2007, 316, 604–608. [Google Scholar] [CrossRef]
  207. Hemmatzadeh, M.; Mohammadi, H.; Jadidi-Niaragh, F.; Asghari, F.; Yousefi, M. The role of oncomirs in the pathogenesis and treatment of breast cancer. Biomed. Pharmacother. 2016, 78, 129–139. [Google Scholar] [CrossRef]
  208. Kim, S.; Song, J.H.; Kim, S.; Qu, P.; Martin, B.K.; Sehareen, W.S.; Haines, D.C.; Lin, P.C.; Sharan, S.K.; Chang, S. Loss of oncogenic miR-155 in tumor cells promotes tumor growth by enhancing C/EBP-β-mediated MDSC infiltration. Oncotarget 2016, 7, 11094–11112. [Google Scholar] [CrossRef] [Green Version]
  209. Li, N.; Cui, T.; Guo, W.; Wang, D.; Mao, L. MiR-155-5p accelerates the metastasis of cervical cancer cell via targeting TP53INP1. OncoTargets Ther. 2019, 12, 3181–3196. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Raigon Ponferrada, A.; Guerrero Orriach, J.L.; Molina Ruiz, J.C.; Romero Molina, S.; Gómez Luque, A.; Cruz Mañas, J. Breast Cancer and Anaesthesia: Genetic Influence. Int. J. Mol. Sci. 2021, 22, 7653. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147653

AMA Style

Raigon Ponferrada A, Guerrero Orriach JL, Molina Ruiz JC, Romero Molina S, Gómez Luque A, Cruz Mañas J. Breast Cancer and Anaesthesia: Genetic Influence. International Journal of Molecular Sciences. 2021; 22(14):7653. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147653

Chicago/Turabian Style

Raigon Ponferrada, Aida, Jose Luis Guerrero Orriach, Juan Carlos Molina Ruiz, Salvador Romero Molina, Aurelio Gómez Luque, and Jose Cruz Mañas. 2021. "Breast Cancer and Anaesthesia: Genetic Influence" International Journal of Molecular Sciences 22, no. 14: 7653. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22147653

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop