Next Article in Journal
Radiotherapeutic Strategies to Overcome Resistance of Breast Cancer Brain Metastases by Considering Immunogenic Aspects of Cancer Stem Cells
Previous Article in Journal
CD3+ and CD8+ T-Cell-Based Immune Cell Score and PD-(L)1 Expression in Pulmonary Metastases of Microsatellite Stable Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Does the Choice of Anaesthesia Affect Cancer? A Molecular Crosstalk between Theory and Practice

by
Wiebrecht Debel
1,
Ali Ramadhan
2,
Caroline Vanpeteghem
1,† and
Ramses G. Forsyth
2,3,*,†
1
Department of Anesthesiology, University Hospital Ghent, 9000 Ghent, Belgium
2
Department of Pathological Anatomy, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium
3
Laboratorium for Experimental Pathology (EXPA), Vrije Universiteit Brussel, 1090 Brussels, Belgium
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(1), 209; https://doi.org/10.3390/cancers15010209
Submission received: 3 September 2022 / Revised: 23 December 2022 / Accepted: 28 December 2022 / Published: 29 December 2022

Abstract

:

Simple Summary

In recent years, there has been an increasing scientific interest in the interaction between anaesthesia and cancer development. Retrospective studies show that the choice of anaesthetics perioperatively may influence cancer outcome and cancer recurrence; however, these studies show contradictory results. Reviewing the recent and relevant literature for the biological effects of anaesthetics on cancer cells in comparison to the clinical effects, it was found that sevoflurane, propofol, opioids and lidocaine are likely to display direct biological effects on cancer cells. However, significant effects are only found in studies with exposure to high concentrations of anaesthetics for longer than practical durations, therefore incomparable to their clinical use.

Abstract

In recent years, there has been an increasing scientific interest in the interaction between anaesthesia and cancer development. Retrospective studies show that the choice of anaesthetics may influence cancer outcome and cancer recurrence; however, these studies show contradictory results. Recently, some large randomized clinical trials have been completed, yet they show no significant effect of anaesthetics on cancer outcomes. In this scoping review, we compiled a body of in vivo and in vitro studies with the goal of evaluating the biological effects of anaesthetics on cancer cells in comparison to clinical effects as described in recent studies. It was found that sevoflurane, propofol, opioids and lidocaine are likely to display direct biological effects on cancer cells; however, significant effects are only found in studies with exposure to high concentrations of anaesthetics and/or during longer exposure times. When compared to clinical data, these differences in exposure and dose–effect relation, as well as tissue selectivity, population selection and unclear anaesthetic dosing protocols might explain the lack of outcome.

1. Introduction

A precise understanding of cancer remains one of the most challenging puzzles of the 21st century. Although surgery for the removal of (large) primary tumours has been regarded as one of the cornerstones in cancer therapy, the exact clinical influence of anaesthetics on cancer biology remains largely unknown. More specifically, interest in this particular subject was already raised two decades ago, and a definitive answer about its exact role in cancer has not been given yet. Several retrospective studies posed a relation between the type of anaesthetics used and the disease-free survival or cancer recurrence. In this point of view, many study reports are available in the literature, but their results are indefinite and oftentimes contradictory. For example, in vitro studies and animal experiments show some benefits for certain types of anaesthetics but solid significant results in clinical trials have yet to be supplied [1]. There are several smaller clinical trials exploring the perioperative effects of anaesthetics on cancer outcomes but these are mostly pilot studies consisting of small sample sizes. Moreover, there are major differences between these study protocols. Interestingly, three recent randomised clinical trials compared different anaesthetic protocols in which certain types of anaesthetics (neuraxial versus opioids, sevoflurane versus propofol) were studied. Nevertheless, these studies still keep lacking significant results [2,3,4]. In this perspective, the methodological aspects of all of these study designs may be possible culprits for the lack of significance. Otherwise unknown confounders might be present, as is the case when few comparable clinical trials are run in the same study field. On the other hand, it is biologically well known that the development of cancer is a multistep process. In this multistep of cancer development certain abilities must be acquired. Moreover, some of these must be kept acquired in order to survive and giving rise to metastasatic disease. Historically, six hallmarks have been described, namely resisting cell death, sustaining proliferative signalling, evading growth suppressors, activating invasion and metastasis, enabling replicative immortality, and inducing angiogenesis. More recently, other hallmarks of cancer have been added as playing a vital role in tumour progression: avoiding immune destruction, tumour-promoting inflammation, genome instability and mutation, and deregulating cellular energetics. Overlooking the recent literature, it is believed that not all hallmarks are affected by anaesthetics. There still is some evidence that certain anaesthetics could influence certain hallmarks. Next to this, it is known that immune responses are regulated by the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS). Therefore, it is of no surprise that activation of these systems induced by surgery or anaesthetics may facilitate tumour activation or distant metastatic disease as such. As for direct effects of anaesthesia, a multitude of mechanisms have been described in the literature. To our knowledge, the mapping of effects of anaesthetics on cancer development is still very limited, and oftentimes molecular pathways are incomplete. The resulting effects can be studied more easily, and the causality can be proven by in vitro knockout models of specific receptors and signalling molecules. The major aim of this review is to get a better sight on whether the currently used anaesthetics have a true direct biological and above all a significant clinical effect, or not, on the outcome during cancer surgery. Furthermore, we aim to discern confounders and methodological errors that help explain the lack of significant results. Therefore, we have opted to use the hallmarks of cancer to facilitate interpretation of the described pathways and mechanisms, and also for categorization of the resulting effects of anaesthesia on cancer cells [5].

2. Materials and Methods

Only publications describing the direct biological effects of anaesthetics on a cellular level, including receptors, pathways and possible mechanisms of action, were included. In addition to this, publications reporting the pathophysiological mechanisms affiliated with tumour cell growth together with a description of the causal effect of the anaesthetics were included. As the first eligible criteria, it was defined that these studies must be clinical trials or observational studies, that the in vivo or in vitro studies must be executed on human cell lines (including the use of xenografts), and that these publications describe the cellular changes during and/or after exposure to anaesthetics. Along with this, the anaesthetics discussed must be relevant and applied in current daily practise. Moreover, inclusion was limited to publication dates not before the year 2000 and to those written in the English language. In the data processed, the focus was foremost on the described biological effect of the anaesthetic agent on the type of cancer cell, including mechanism of action and the signalling pathways if sufficiently investigated. Furthermore, a subanalysis of dosage and exposure of anaesthetics was performed in comparison to the typical clinical use. Data were classified according to the type of anaesthetic agent, the type of tumour and the pathophysiological pathway of cancer initiation and metastasis. The huge amount of publications available in the literature concerning this subject made traditional relevant searches through bibliographic databases very complicated. From this point of view, relevant publications were selected based on title, abstract and full article.
These lists were filtered for double references. One reviewer was used for selection and data extraction. No tools for risk of bias were used. For supplemental data specifically on the cellular effects of propofol, sevoflurane, ketamine, lidocaine, midazolam and dexmedetomidine the bibliographical database PubMed.ncbi was used with the following search strategy: (((**[MeSH Major Topic]) AND (cancer[MeSH Major Topic])) AND (in vitro)) OR (((**[MeSH Major Topic]) AND (cancer[MeSH Major Topic])) AND (in vivo)) **: Independent search queries were made for propofol, sevoflurane, desflurane, ketamine, lidocaine, midazolam and dexmedetomidine.

3. Results

In short, anaesthesia and its direct effects on cancer are subdivided according to the type of anaesthetics (see Table 1).

3.1. The Direct Effects of Midazolam, Dexmedetomidine and Ketamine Are Insufficiently Proven

The results of the database search concerning the direct effects of midazolam, ketamine and dexmedetomidine produced the list of studies available in Table 1.
Concerning midazolam, only three studies describing its direct effects. It was stated that midazolam showed a proapoptotic effect and halts cell cycle progression. Therefore, midazolam reduces tumour growth [6,7,8]. Of interest in this perspective is that Wang et al. [6] (see Table 1) described that lower doses might not have the same effects, and since the doses used far exceed clinical use, these results cannot be extrapolated to human research. Further limitations of these studies are prolonged exposure to the drug compared to clinical use and the use of different cancer tissues, making a comparison between these studies extremely difficult.
The three studies describing the effects of dexmedetomidine show similar concerning issues. Dexmedetomidine showed an effect on apoptosis, although depending on which study was selected a pro- or anti-apoptotic effect was found. This effect is mediated through alpha 2-adrenoreceptors [6,9]. Additionally, a pro-oncogenic effect was described by phosphorylation of the ERK pathway, which activates invasion and metastasis [10]. In line with midazolam, the dosage used in these particular studies is supraphysiological and the exposure time is much longer than typically is the case in an operative setting (48 h). Thus again this is limiting extrapolation to human research. Next to this, other effects are described such as a decrease in the overall survival, and in vivo in mice, and a dose-related worsening in outcome [12,13].
Only one study could be found that describes the direct effects of ketamine. It demonstrated a decreased apoptosis, thus promoting tumour growth. Although in vivo studies have confirmed these effects, both in vivo and in vitro very high doses of ketamine were administered [11,14]. Only one retrospective study of breast cancer describes the possible effects in a true clinical setting and this study showed no significant difference in outcome or recurrence [15].
Taking together, anaesthetics such as midazolam, dexmedetomidine and ketamine are insufficiently investigated to proclaim any possible effect on use during anaesthesia for cancer surgery.

3.2. The Direct Effects of Volatile Anaesthetics Are Dependent on the Type of Anaesthetic and the Biological Type of Tissue

Table 2 shows the collected data on volatile anaesthetics such as isoflurane, desflurane and sevoflurane. Five publications describing the direct effects of isoflurane on cancer cells were found. Generally, it is agreed that HIF-1A is involved which modulates the expression of VEGF-A, and angiopoietin-1, thus increasing tumour angiogenesis, glycolysis and cellproliferation [16,17,18]. In contrast, no direct outcome in prostate tissue could be found [19]. Next to this, a decrease in apoptosis is described in colon tumour cells, conceivably due to the effect on caveolins [20]. These differences in direct effects may be explained by the selection of different tissues used for research as exposure and concentrations were largely similar. Very little information is available on the use of isoflurane and outcome in the tru clinical setting.
Concerning desflurane only two studies were included, showing a prometastatic effect and an effect on cell cycle progression. There is also a dose dependent effect on apoptosis. More specifically, low doses induced a proapoptotic effect, where high doses in contrast demonstrated an antiapoptotic effect. Therefore, more studies are required to determine more strongly the direct effects of desflurane on cancer cells [18,21].
The effects of sevoflurane are more extensively studied resulting in an inclusion of 18 studies describing the direct effects on cancer cells. These results show varying, sometimes very contradictory results. As mentioned earlier, this is possibly due to differences in tissue selection. In colorectal tissue, sevoflurane has an antiproliferative effect and inhibits activation and metastasis through various mechanisms [22,23,24,25]. In lung tissue, a small reduction in viability is found. In gastric cells, sevoflurane weakens proliferative and migratory abilities through yet unknown mechanisms [26,27]. However, pro-oncogenic traits were found in renal cells, cervix cells, and head and neck squamous cell carcinomas treated with sevoflurane [26,28,29]. In breast cancer tissue, a small increase in cancer cells is described. This is likely caused by a change in intracellular calcium homeostasis [30,31]. In studies investigating the influence of sevoflurane on Smad3 signalling, which regulates cell proliferation, differentiation and cell death, proliferation is increased in non-small cell lung carcinomas (NSCLC) whereas in contrast it is decreased in renal cell carcinoma, therefore, producing contrary effects. Again, this further strongly supports the hypothesis that the effect of anaesthetics is fundamentally different, especially depending on the exact type of tissue.
However, even within the same series of tissue samples differences in effects were noted. Additionally, cumulative dosing over time plays an important role as well [31]. For example, in ovarian tissue exposed to sevoflurane an enhanced metastatic potential through CXCR2 is described. Next to this, also inhibition of proliferation and migration through the P38/MAPK pathway was demonstrated [18,32]. In brain tissue cell migration and invasion were repressed in two studies, and invasion potential was increased in one [33,34,35]. This could clearly be explained by differences in study protocols and duration of exposure times. More specifically, retrospective studies were not able to show any clinical significance in lung tissue (NSCLC) and mixed samples [36,37,38]. In breast tissue, however, retrospective studies showed contradictory results in comparison with propofol with either worse outcomes for sevoflurane [39,40] or no significant difference at all [41]. One large retrospective study by Enlund et al. showed no significant difference between propofol and sevoflurane after correction for cofounders [36]. Again, very little is disclosed about anaesthetic dosing in these retrospective studies, and study protocols are very heterogeneous. Moreover, there are no randomised clinical trials (RCT) available that prove the superiority or inferiority of sevoflurane in the daily clinical setting.
Table 2. Studies describing the direct effects of isoflurane, desflurane and sevoflurane on cancer cells, the associated mechanisms of action, and their respective pathways 1.
Table 2. Studies describing the direct effects of isoflurane, desflurane and sevoflurane on cancer cells, the associated mechanisms of action, and their respective pathways 1.
Isoflurane
StudyType of
Cancer
Effect on CancerMechanism of ActionPathway Described
Huang H. et al. [19]ProstateNo direct outcomeIncreased HIF-1A expression (angiogenesis, glycolysis, proliferation)HIF-1A
Benzonana L.L. et al. [16]Kidney
  • Increased proliferation
  • Cytoskeletal rearrangement
  • Migration of cells
Increased HIF-1a and HIF- 2a expressionPI3K/Akt/mTOR pathway
Luo X. et al. [17]OvaryUp-regulation of markers associated with the cell cycle, proliferation, and angiogenesisIncreased VEGF, angiopoietin-1 and MMPs expressionThe IGF1/HIF signalling pathway
Iwasaki M. et al. [18]OvaryEnhanced metastatic potentialSignificant increase in mRNA for CXCR2, VEGF-A, MMP11 and TGF-βCXCR2 plays crucial roll in the pathway, knockdown mitigates anaesthetics effect
Kawarguchi Y. et al. [20]Colon
  • Resistance against apoptosis via a Caveolin dependent mechanism
  • No effect from isoflurane alone
Resistance against TNF-related apoptosis-inducing ligand (TRAIL)-induced apoptosis via Cav-1–dependent mechanismsPossible mechanism: Caveolins are changed in configuration due to effect on lipid membrane of volatiles
Desflurane
StudyType of
Cancer
Effect on CancerMechanism of ActionPathway Described
Iwasaki M. et al. [18]OvaryEnhanced metastatic potentialSignificant increase in mRNA for CXCR2, VEGF-A, MMP11 and TGF-β (change TME)CXCR2 plays a crucial role in the pathway, and knockdown mitigates anaesthetics
Bundscherer A.C. et al. [21]Colon
  • Affection of cell cycle regulation
  • Affection of apoptosis after 6 h exposure
Non describedNon described
Sevoflurane
StudyType of
Cancer
Effect on CancerMechanism of ActionPathway Described
Ecimovic P. et al. [30]BreastSmall increased proliferation and migrationNon describedNon described
Deng, X. et al. [31]BreastSevoflurane, but not propofol, at clinically relevant concentrations and durations:
  • increased survival of breast cancer cells in vitro
  • no effect on cell proliferation, migration or TRPV1 expression.
These findings suggest that changes in intracellular Ca2+ homeostasis play an important role in the general anesthetic-mediated enhancement of breast cancer cell survivalThe TRPV1 channel is a potential site of action of sevoflurane in altering intracellular Ca2+ levels
Iwasaki M. et al. [18]OvaryEnhanced metastatic potential.Significant increase in mRNA for CXCR2, VEGF-A, MMP11 and TGF-βCXCR2 plays a crucial role in the pathway, and knockdown mitigates anaesthetics
Kang K. et al. [32]OvaryInhibition of cell proliferation, migration and invasion, and induced apoptosis of the OC cell linePCNA, Twist, MMP-2 and MMP9 mRNA expressions were significantly decreased while caspase-3 expression was markedly increased in sevoflurane groups compared to that in the control groupDramatical decrease of p-p38/p38 and p-JNK/JNK expressions in OC cells of sevoflurane groups compared to that of the control group, important in p38 MAPK Signaling Pathway
Ciechanowicz S. et al. [26]Lung (NSCLC)
  • Reduced cell viability
  • No effect on metastatic potential
Unchanged levels of TGF-b1, possible homeostatic regulation/sensitizationUpregulation of Smad3 signalling
Ciechanowicz S. et al. [26]Renal cell carcinoma
  • Increases cell viability
  • Promotes metastatic potential
TGF-b1 plays a role in cytoprotection, proliferation and migrationTGF-b and OPN upregulation. Reduced nuclear Smad3
Ferrell J.K. et al. [28]Head and neck SCCIncrease in the expression of pro-oncogenic protein markersExact mechanism unclearStatistically significant increases in the expression of cytoplasmic HIF-2a and nuclear p-p38 MAPK
Yang X. et al. [22]Colon
  • Inhibition of proliferation and invasion of colon cancer cells
  • Promotion of apoptosis in vitro.
  • SW480 cell xenograft tumour experiments: sevoflurane inhibits the tumorigenic ability of cancer cells in vivo
The circ-HMGCS1/miR-34a-5p/SGPP1
axis may play a role in cell viability and apoptosis
circ-HMGCS1 suppression by sevoflurane treatment in a dose-dependent manner
He J. et al. [23]Colon
  • Hindered cell viability and invasion
  • Facilitated cell apoptosis in colon cancer by regulating the circ-HMGCS1/miR-34a-5p/SGPP1 axis
The exosome-transmitted circ-HMGCS1/miR-34a-5p/SGPP1
axis might play a role in cell viability and apoptosis
Circ-HMGCS1 suppression by sevoflurane treatment in a dose-dependent manner
Sun S.Q. et al. [24]ColonInhibition of migration and invasionSignificant decrease in PCNA, Twist, MMP-2 and MMP9 mRNA expressions while caspase-3 expression was markedly increased in sevoflurane groups compared to that in the control groupDramatical decreases of p-p38/p38 and p-JNK/JNK expressions in OC cells of sevoflurane groups compared to that of the control group, important in p38 MAPK Signaling Pathway
Fan L. et al. [25]Colon
  • No significant effect on proliferation
  • Inhibition of migration and invasion
Reduced MMP-9, which plays a role in EMTThrough regulating (inactivating) ERK pathway via regulating miR-203 and Robo1
Bundscherer A.C. et al. [21]Colon
  • Affected cell cycle regulation
  • Increased apoptosis in low and high doses
Non describedNon described
Chen H. et al. [27]GastricWeakening proliferative and migratory abilitiesExact mechanism unclearUpregulation of miR-34a/TGIF2 axis
Zhang W. et al. [29]CervixEnhanced proliferation, migration, and invasion of immortalized cervical cancer cellsIncreased histone deacetylase 6 expression, which leads to decreased acetylation of alpha-tubulinY-phosphatidylinositide 3-kinase/AKT- and ERK1/2-signaling pathway activation
Zhao H. et al. [33]BrainRepressed cell migration and invasionUpregulation of miR-34a-5p, which inhibits MMP-2 thus reducing metastasisNon described
Xu W. et al. [34]Brain (glioma)
  • Suppresion of viability, colony formation, cell cycle, migration and invasion
  • Promotion of apoptosis of glioma cells in vitro, and impeded tumour growth in vivo
  • Downregulation of Circ_0012129 and TGIF2
  • Upregulation miR-761
  • Via regulating the circ_0012129/miR-761/TGIF2 axis.
  • MiR-761 possibly functions through deactivating FGFR1/PI3K/AKT pathway2
Lai R.C. et al. [35]Brain (glioblastoma)
  • Exposure to 1–4% sevoflurane did not change the cell proliferation
  • Concentration-dependent increasement of invasion of human glioblastoma U251 cells
Increased activity of calpains, a group of cysteine proteinases, and CD44 proteinCD 44 regulates intracellular signalling, unsure which pathway is involved
1 The correlated hallmark of cancer is given for reference on the role and importance in cancer development. Sorted by type of tissue.

3.3. The Direct Effects of Propofol Show a Multitude of Cellular Changes Dependent on Tissue Type

This study has yielded 41 articles describing the direct results of propofol on cancer cells. When comparing the direct effects per type of tissue, the most frequent described effects of propofol are anti-oncogenic and anti-metastatic (see Table 3). Exceptions are a study on prostate cancer, where no effect was found, and studies on breast cancer showing an increase in proliferation and migration of cancer cells [19,42]. The mechanisms linked to these effects varied greatly, again mostly and especially depending on the type of tissue used. The most important effects are inhibition of matrix metalloproteinases (MMPs) causing inhibition of invasion, EMT, and metastasis [43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61] and increased apoptosis [42,55,62,63,64,65,66,67,68,69,70,71]. Some studies reported an effect on the cell cycle [68,69,72] or even deregulation of cellular genetics [73].
Overall, it is clear that propofol directly affects cancer cells through a multitude of translational changes and modulation of cellular pathways. However, there are a considerable amount of important limits to these studies. First, most studies describe long periods of exposure to propofol (more than 24 h) before any significant outcomes can be found. Furthermore, the study protocols implemented exposure to a relatively high concentration of propofol. Some significant effects started clinically at a relevant dose up to 5 µg/mL [48,49,64,66,68,73,74,75,76], but most significant effects started at 10 µg/mL or even higher. In shorter periods of exposure or at lower concentrations, these studies show no significant effect at all. This is of particular importance to the reference of the clinical use of propofol, in which typically shorter periods of exposure are custom during cancer surgery. Although exposure periods of 24 h may be clinically applied, concentrations of 5 µg/mL are rarely used during sedation in the intensive care unit. Currently, there are no RCTs available that could show anti-oncogenic effects of propofol with differences in the outcome on overall survival or disease-free survival in the clinical setting. Next to this, there is a multitude of retrospective studies with varying outcomes. Most describe any significant difference in outcome for propofol versus other anaesthetics in a collection of tissue samples [37,38,41,77,78,79]. Others described superior effects of propofol versus volatile anaesthetics in the breast [39,40] hepatocellular [80] or mixed [41,81] tissue samples. As was mentioned earlier, a large difference in anaesthetics and study protocols is clearly noted in comparison of these studies as well as the presence of multiple confounders and biases that inherently trouble retrospective research seriously. More specifically, the concentration of anaesthetic used, combinations of possible influential drugs used perioperatively and the time of exposure may be important factors. Very little about this is documented in these retrospective studies.
Table 3. Studies describing the direct effects of propofol on cancer cells, the associated mechanisms of action and their respective pathways 1.
Table 3. Studies describing the direct effects of propofol on cancer cells, the associated mechanisms of action and their respective pathways 1.
Propofol
StudyType of CancerEffect on CancerMechanism of ActionPathway Described
Huang H. et al. [19]ProstateNo direct effectStimulation of HIF-1a PathwayHIF-1A
Yang C. et al. [69]Gastric
  • Significantly inhibited cell proliferation, invasion and migration
  • Enhanced apoptosis
Upregulation of expression of inhibitor of growth 3 (ING3)Non described
Peng Z. et al. [51]GastricSignificant elevation of miR-451 expression levels, inhibition of cell proliferation and promotion of apoptosisOverexpression of miR-451 inhibited MMP-2 protein expressionStimulated expression of miR-451, exact mechanism unclear
Liu F. et al. [53]GastricPropofol treatment reduced the invaded and migrated SGC-7901 and NCI-N87 cell numbers
  • After treatment, reduction of the Snail and vimentin levels while the E-cadherin level increased in SGC-7901 cells
  • Similar trends in NCI-N87 cells. This could inhibit EMT
Elevated expression of miR-195-5p, pathway unclear
Yang N. et al. [43]Lung (NSCLC)Decreased tumour aggressiveness
  • Downregulation of HIF1A
  • Suppression of upregulation of EMT-TFs
Propofol suppressed the LPS-induced transcriptional activity of HIF-1α
Wu KC. et al. [47]Lung (NSCLC)Significant inhibition of migration and invasion of human lung cancer A549 cells (inhibited MMP-2 activity)Suppressed MMP-2 and -9 expression and invasion of A549 cellsThe downstream regulation of MAPK (p38 and JNK) pathway
Yang N. et al. [64]Lung (NSCLC)Inhibited cell viability and induced cell apoptosis by upregulating miR-486 expressionSignificant increase in protein levels of FOXO1, FOXO3, Bim, pro-caspase-3 and activated caspase-3, initiating growth halt and apoptosisPossible relation between miRNA-486 upregulation and FOXO pathway
Zheng X. et al. [66]Lung (NSCLC)
  • Inhibition of A549 cell growth
  • Acceleration apoptosis in vitro and in vivo.
  • Suppression NSCLC tumour cell growth
Downstream regulation of apoptotic pathways, exact mechanism unclearmiR-21/PTEN/AKT pathway
Zhao H. et al. [73]Lung (NSCLC)Inhibition of cell proliferation, migration, Invasion and aerobic glycolysisAll three glycolysis enzymes, HK2, PKM2, and LDHA and a Glucose transporter GLUT1 decreased by propofol treatment, suppressing aerobic glycolysiscircTADA2A/miR-455-3p/FOXM1 axis
Cui W.Y. et al. [55]Lung (NSCLC)Inhibition of proliferation and induction of apoptosis in H460 cells both in vivo and in vitroPropofol causes ER stress, leading to increased apoptosis, inflammation and reduced cell growthPossible effect on JNK signalling pathway in H460 cells
Sun H. et al. [68]Lung (NSCLC)
  • Inhibition of lung cancer A549 cell viability, proliferation, migration
  • Promotion of A549 cell apoptosis
  • Downregulation of pro-proliferative protein, Cyclin D1, cell migration- and invasion-related proteins, MMP-9, Vimentin, as well as anti-apoptotic protein Bcl-2
  • Upregulation of anti-pro liferative proteins, p53 and p16, as well as pro-apoptotic proteins, Bax, cleaved-Caspase-3, and cleaved-Caspase-9
Suppressed Wnt/β-catenin and mTOR signalling pathways by down-regulating miR-372
Gao J. et al. [72]Lung (NSCLC)
  • Suppression of cell proliferation, invasion and glycolysis
  • Expedited apoptosis cells
FOXM1 (transcription factor belonging to the Forkhead box family) essential for cell cycle progressioncirc-ERBB2/miR-7-5p/FOXM1 axis
Ecimovic P. et al. [61]Breast
  • No effect on proliferation
  • Reduced migration and reduced invasion of MCF7 but not of MDA-MB-231 cells.
The Neuroepithelial Cell Transforming Gene 1 (NET1) gene is associated with promoting migration in adenocarcinoma in vitro.Propofol reduced expression of NET1
Yu B. et al. [62]BreastSignificantly induces apoptosis.Downregulation of miR-24, upregulation of p27 expression and cleaved caspase-3 expressionInactivation of miR-24/p27 signal pathway
Meng C. et al. [42]Breast
  • Increasement of proliferation of human breast cancer MDA-MB-231 cells
  • Inducion of cell migration
  • Antioxidative effect of propofol inhibited the expression of p53 decreasing cell apoptosis
  • Attenuation of apoptosis by Nrf2 (an important redox-sensitive transcription factor for inducing antioxidant defense system)
  • Activation mitochondrial apoptosis pathway and Nrf2 pathway
  • Possible role of Nrf2 on migration.
Garib V. et al. [74]BreastActivation of GABA-A receptor correlated with an increased migration of MDA-MB-468 breast carcinoma cellsMediated by calcium influx and reorganization of the actin cytoskeletonGABA-A receptor activation results in activation of voltage-gated L-type calcium channels
Li Q. et al. [49]BreastInhibition of the invasion and migration of breast cancer cellsDown-regulation of MMP-2 and MMP-9Reduction phosphorylation of IKKb (Ser180), which is an important upstream kinase for IkB degradation and subsequent NF-kB activation
Liu Y.P. et al. [59]BreastReduced invasion and migration ability of breast cancer cellsInhibition of circNOLC1 by repressing STAT3 in a feedback mechanismTargeting miR-365a-3p/STAT3 axis.
Huang X. et al. [48]Ovary
  • Inhibition of cell growth and invasion
  • Induced cell apoptosis in a dose dependent manner
Role of miR-9 is not well understood, inhibition of MMP-9Upregulation of miR-9 expression and inhibition of NF-kB activation
Su Z. et al. [70]OvaryInhibition of proliferation and induction of apoptosisExact mechanism unclearIncreased expression of miR-let-7i, no pathway described
Lu H. et al. [71]Ovary
  • Suppression of proliferation, cell cycle, migration and invasion
  • Induction of apoptosis of ovarian cancer cells
Up-regulating miR-145 via down-regulating circVPS13CInhibited the activation of MEK/ERK signalling
Liu Z. et al. [63]Pancreas
  • Inhibition tumour growth and invasion
  • Induction apoptosis in a dose- and time-dependent manner in PANC-1 cells.
Increase in PUMA induces apoptosis and increase in E-cadherin prevents epithelial to mesenchymal transitionInhibition miR-21 levels and decrease in Slug expression, resulting in an increase in Slug-dependent PUMA and E-cadherin expression in PANC-1 cells
Wang Z.T. et al. [82]Pancreas
  • Significant inhibition of Panc-1 cell proliferation and invasion
  • Promotion of apoptosis
The antitumor effect on pancreatic cancer cells may be partly due to the upregulation of miR-133aNo pathway described
Wang H. et al. [67]PancreasInduction of apoptosis and inhibited cell migration PANC-1 cells in vitro
  • Upregulation miR-34a expression, which in turn upregulates LOC285194 expression, resulting in PANC-1 cell apoptosis and growth inhibition
  • Upregulation miR-34a expression, which in turn upregulates E-cadherin expression, resulting in cell migration inhibition
No clear pathway described
Chen X. et al. [76]PancreasInhibition of VEGF expression, cell migration and tumour growthInhibition of NMDA receptor, attenuated intracellular Ca2+ concentration, thus suppressing VEGF expression. Relation to tumour suppression unclearVia inhibiting CaMK II activity, attenuated AKT, ERK phosphorylation and HIF-1α expression
Miao Y. et al. [50]ColonInhibition of cancer cell invasionThrough activation of GABA A receptors. MMP’s are crucial proteinases for invasion and metastasisInhibition of MAPK pathway. Especially deactivation of ERK1/2 suppressed MMP production
Zhang Y.F. et al. [54]Colon
  • Inhibition of cell invasion
  • Promotion of cell apoptosis
HOTAIR regulates E-cadherin, MMP-9 and vimentin expressions. STAT 3 can also regulate HOTAIRRegulating STAT3/HOTAIR by activating WIF-1 and suppressing Wnt pathway
Takabuchi S. et al. [83]HepatocellularSuppression of HIF-1a protein expression was significant at 20%and 5% O2 but not at 1% O2Suppression of the translation of HIF-1amRNA into proteinPossible role of MAPK pathway
Zhang J. et al. [65]Hepatocellular
  • Induction of apoptosis of HCC cells
  • Modulation of miR-199a contributing to the antitumor action of propofol
Induction of apoptosis and activation of caspase-8 and caspase-9 in a dose-dependent mannerStimulation of miR-199a expression in HepG2 cells
Song F. et al. [75]Hepatocellular
  • Significant suppression of cell proliferation and metastasis
  • Significant promotional effect on cell apoptosis after propofol treatment in vitro in HCC
Downregulation of HOXA11-AS and upregulation of miR-4458 in HCC. This may serve a tumour suppressive effectExact mechanism unclear
Gong T. et al. [56]Hepatocellular
  • Suppression of proliferation, invasion and migration of HCC in vitro.
  • Significant decrease in tumour volumes, growth rates and the liver orthotopic xenograft tumour in vivo
Reversal of EMT transitionUpregulated expression levels of the candidate tumour suppressor miR-219-5p. miR-219-5p inhibits HCC cell progression by targeting glypican-3 and subsequently results in the inhibition of Wnt/β-catenin signalling
Zhang J. et al. [60]HepatocellularInhibition of the invasiveness of HepG2 cells
  • Significant decrease in expression of MMPs
  • Significant inhibition of the activity of MMP-9 in HepG2 cells but no effect on MMP-2 expression
Exact mechanism unclear, possible role of miR-199a
Du Q. et al. [44]EndometriumPropofol inhibits proliferation, migration, invasion and promotes apoptosis.Downregulation of SOX4 gene expression.Propofol inhibited Sox4 expression via inactivation of Wnt/b-catenin signal pathway
Zhang D. et al. [80]Cervix
  • Inhibition cervical cancer cell growth
  • Induction of apoptosis
Decreased HOTAIR expression of cervical cancer cells in a dose-dependent manner. HOTAIR is an lncRNA which is noticeably increased in multiple carcinomas.HOTAIR activates mTOR/p70S6K pathway leading to cell growth
Ye Z. et al. [46]Osteosarcoma
  • Significant inhibition of cell proliferation and invasion
  • Promotion of apoptosis
Decreased protein expression of matrix metalloproteinase 13 (MMP-13)Elevated expression of miR-143, which decreases expression of MMP-13
Xu J. et al. [52]GliomaEffective suppression of proliferation and invasion, and induction of apoptosis of glioma cellsIncrease in caspase 3, reduction in MMPIncreased miR-218 expression, no clear pathway described
Zhang L. et al. [84]GliomaRepression of cell growth and metastasis in glioma cells in vitro and in vivoExact mechanism unclearMediated by the circNCAPG/miR-200a-3p/RAB5A axis
Zhou C. et al. [45]EsophagusInhibition of migration and invasion
  • Decreased expression of MMP-2 and MMP-9
  • Increased expression of TIMP-1
Decreased expression SOX4. No other pathways described
Xu Y.B. et al. [58]EsophagusSignificant promotion of cell apoptosis and inhibition of proliferation, invasion and angiogenesis in a dose and time-dependent mannerDown regulation gene expression and protein production of VEGF and MMP-9ERK/VEGF and ERK/MMP-9 signalling pathways
Du Y. et al. [57]Bladder
  • Significant suppression of proliferation, migration and invasion of BC cells in vitro
  • Suppressed of tumour xenograft growth
  • TOP2A expression promotes tumour growth, metastasis and chemotherapeutic drug resistance by regulating DNA topological states
  • Propofol suppressed the EMT of BC cells
Induced miR-145-5p expression in a time-dependent manner. Topoisomerase II α (TOP2A) was a direct target of miR-145-5p
1 The correlated hallmark of cancer is given for reference on the role and importance in cancer development and sorted by type of tissue.

3.4. The Direct Effects of µ-Receptor Opioids on Cancer Cells Remain Uncertain and Are Difficult to Explore

Sixteen articles reporting the direct effects of opioids on cancer cells could be selected by this study (see Table 4). Most frequently described are the effects on breast, lung or colon tissue. However, these results are often contradictory, and multiple mechanisms varying from apoptosis, EMT and increased proliferation or activation of microenvironment and inflammation are found as direct effects. For example, some authors claimed no effect of morphine in breast tissue [85,86,87], where others described possible anti-oncogenic effects [88,89,90,91,92]. In lung tissue, the same conflicting results between oncogenic [93,94] and anti-oncogenic [90,95] effects were found. Little outcome was found in colon tissue inclusive one article describing an anti-invasive effect [89,96,97]. Large variations in outcome were noticeable for these in vitro trials. The trials describing apoptosis and necrosis did use high supraphysiological doses of morphine, possibly partially explaining the difference in outcome [90,97]. In addition, there are large differences when comparing cell lines of similar tissue. Some authors used commercially available cell lines, while others used samples of cells in their own microenvironment. Even within the same cell line set-up conflicting evidence was found [93,95].
There are no RCTs demonstrating an effect of µ-receptor opioids on overall survival or disease-free outcome. Some RCTs and experimental studies described an immunomodulatory effect of opioids and a possible decrease in natural killer (NK) Cell function and neutrophils [4,98,99,100]. However, a weakness in these studies is the lack of knowledge of influence of opioids and immune function together with the surgery related stress response. As both affect immune function, and stress levels are hard to objectify, this may be an extra challenge in interpreting these results as reliable.
Retrospective studies described a correlation between the amount of µ-opioid receptors (MOR) and clinical outcomes [101,102,103]. However, in the case of NSCLC, retrospective studies could not show significant differences in the risk of cancer recurrence. Only a small decrease in overall survival for stage I NSCLC patients could be noticed [104,105,106]. Moreover, no impact could be demonstrated on the overall survival and disease-free survival in colorectal cancer [105]. An important possible cofounder in these cases is the chronic use of opioids during cancer treatment and the impact on the outcome.
Generally, it is largely uncertain if opioids directly affect cancer cells in vivo. Stronger paracrine and endocrine effects play a vital role in this type of research with opioids. Factors such as the effect of the tumour microenvironment, inflammation and stress response make it extremely difficult to interpret study results adequately and reliable.
Table 4. Studies describing the direct effects of MOR opioids on cancer cells, the associated mechanisms of action and their respective pathways 1.
Table 4. Studies describing the direct effects of MOR opioids on cancer cells, the associated mechanisms of action and their respective pathways 1.
µ-Opioid Receptor Agonists
StudyType of CancerEffect on CancerMechanism of ActionPathway Described
Nguyen J. et al. [85]Breast
  • No influence on initiation
  • Increased progression
  • Decreased survival
µ-opioid receptors on large tumours, possibly stimulated by VEGF and cytokinesStimulation of MAST cells in tumours, release of substance P, increased pain and inflammation
Doornebal C. et al. [86]BreastAnalgesic doses of morphine do not affect mammary tumour growthDifference in effect is explained by heterogeneity of primary tumour compared to selected cell lines chosen for metastatic potentialNon described
Afsharimani B. et al. [88]BreastReduced expression of matrix-degrading enzymes in cocultures with macrophages or endothelial cellsReduced the level of MMP-9 and increased its endogenous inhibitor, TIMP-1Modulation of paracrine communication between cancer cells and non-malignant cells in the tumour microenvironment
Gupta K. et al. [91]Breast
  • Statistically significant increase in tumour volume and vascularization
  • Significantly increased migration in vitro
  • Increased vascularisation similar to VEGF
  • Increased growth through inhibition of apoptosis
Stimulation of the MAPK/ERK signalling pathway and activation of the cell survival signal Akt and increasing cell cycle protein cyclin D1
Tegeder I. et al. [89]BreastMorphine significantly reduced the growth of MCF-7 and MDA-MB231 tumoursInhibition of cell cycle progression in low dose, activation of apoptosis in high dosesp53 activation and up-regulation of p53-dependent genes (including CD95/Fas)
Ecimovic P. et al. [92]BreastIncrease in both expression of NET1 and cell migration but not when NET1 was silencedThe NET1 gene has a key role in organization of the actin cytoskeleton and thus in the ability of cancer cells to migrate and invadeMechanism unclear
Gach K. et al. [87]BreastIncrease in secretion of urokinase plasminogen activator, no results on migrationOpioid agonists greatly increase the secretion of uPA from MCF-7 human breast cancer cells, as well asup-regulate the expression of uPA and uPAR genesThrough MOR increases the expression of uPA and uPAR
Hatsukari I. et al. [90]lung (NSCLC) and breastA clinical concentration of morphine induced apoptosis and necrosis in human tumour cell linesThrough activation of opioid receptors, no clear arguments for different mechanisms (control group with naloxone)Non described
Mathew B. et al. [93]Lung (NSCLC)
  • Increased MOR expression in NSCLC
  • Morphine increases cell growth
  • Methylnaltrexone decreases cell growth and invasion
Knockout of MOR receptor reduces cell growth and metastasisDirect inhibition of MOR and activation of tyrosine phosphatase activity
Koodie L. et al. [95]Lung/ Ovaries
  • Inhibition of migration of tumour infiltrating leukocytes
  • Decrease in angiogenesis
Altering cell adhesion molecule expression on both the leukocyte and endothelial cells. Impairs mobilization of endothelial progenitors and neutrophils, thus decreasing inflammation and angiogenesisThe mechanism is unclear Possible mechanism: decreases the tight junction protein zonula occludens protein 1 expression.
Lennon F.E. et al. [94]lung (NSCLC)The data suggest a possible direct effect of MOR on opioid and growth factor-signalling and consequent proliferation, migration and epithelial mesenchymal transition (EMT) during lung cancer progression. DAMGO, morphine and fentanyl were used as MOR agonistsMOR regulates opioid and growth factor-induced EGF receptor signalling through Grb2-associated-binding protein 1 (Gab-1)Activation of Src, Gab-1, PI3K, Akt and STAT3
Tegeder I. et al. [89]ColonNo significant effect on HT-29 tumour growthNon describedLess expression of P53, therefore less effect
Nylund. et al. [96]ColonMorphine largely fails to affect the proliferation of the HT-29 cell line, but causes a markedly increased secretion of uPa. No results on migrationuPa plays an important role in activating invasion and metastasisThrough MOR
Harimaya Y. et al. [97]Colon
  • Significant reduction of the number of tumour colonies and of the weight of the tumour-containing lung
  • Inhibition of adhesion and migration of cells to the extracellular matrix, without affecting the cell proliferation in vitro
Suppression of tumour cell adhesion, invasion and migration, partly through opioid receptors, partly through reducing enzymatic degradation of the ECMInhibition of the production of MMP-2and MMP-9 in tumour cells, no clear pathway was described
Friesen C. et al. [103]Leukemia (ALL)Methadone induces apoptosis, increases doxorubicin triggered cell death, reduces tumour growth in vivo
  • Opioid-receptor activation induces the downregulation of cAMP, resulting in apoptosis in cells with high levels of expression (not in moderate levels)
  • Increases concentration of doxorubicin in cells
Downregulation of the antiapoptotic proteins XIAP and Bcl-xL and cAMP. cAMP activates
PI3K-dependent Akt
Friesen C. et al. [107]GlioblastomaActivation of opioid receptors sensitizes glioblastoma cells for therapyOpioid receptor signalling pathway is involved in apoptosis induction by chemotherapy.Opioid receptor stimulation activates inhibitory Gi-proteins, which, in turn, block adenylyl cyclase activity, reducing cAMP. Downregulation of Bcl-x and XIAP
1 The correlated hallmark of cancer is given for reference on the role and importance in cancer development. Sorted by type of tissue.

3.5. Lidocaine Affects Cancer Cells Directly through Various Mechanisms

Twenty-one articles were included demonstrating the direct effects of lidocaine (see Table 5). Of particular interest is that lidocaine showed the highest concentrations perioperatively among all modern local anaesthetics, thus most likely to produce a significant observable clinical effect. Other local anaesthetics are mainly used perioperatively as part of the locoregional anaesthesia procedure, thus limiting the concentration. Like propofol, various mechanisms and pathways are described as direct effects of anaesthesia. Interestingly, all these described effects are anti-oncogenic, making lidocaine a prime candidate for further investigation. The most recurring effects were the evasion of growth suppressors [108,109,110,111], increased activation of apoptosis mechanisms [112,113,114,115,116], and decreased invasion, EMT and/or metastasis [117,118,119,120,121].
Unfortunately, all these in vitro studies used very high concentrations of local anaesthetic drugs. If extrapolated to the daily clinical use, the concentrations used are approximate to that applied to local anaesthesia with infiltration approximate to the target tissue, whereas the clinically used concentrations for locoregional or intravenous anaesthesia are considerably lower. Chamaraux-Tran. et al. (see Table 5) described a difference in sensitivity for local anaesthetics in different cell lines. Additionally, differences were noted concerning toxic effects in higher concentrations, which were frequently applied in other studies [122]. The studies that had used lower concentrations closer to clinically relevant intravenous doses (lower or equal to 10 µM) reported no significant effects at all [109,111,114,115,118,119,121,123,124,125,126,127]. At low concentrations, long exposure times were absolutely needed before significant results could be found. Yang. et al. (see Table 5) found significant results at 10 µM concentration, but only starting after 24 h of exposure time, and also depending on the type of cell line used [110]. Lirk. et al. (see Table 5) described a significant effect starting at least 24 h of incubation time but only for specific breast cell lines. This fits again with the hypothesis of different levels of sensitivity per cell line [123]. A few studies, however, found significant effects at lower concentrations in line with the exposure time comparable to routine clinical use [116,124]. Interestingly, in three of these low concentration studies, breast tissue was involved.
No RCTs have currently proven the superiority of lidocaine over other anaesthetics or analgesics. Zhang. et al. published a large sample retrospective study that claims a potential anti-oncogenic effect of lidocaine in pancreatic cancer with increased overall survival [128]. In vivo studies with mice, xenograft models showed a possible diminishing effect on metastasis as well [129,130].
From this point of view, it is plausible that lidocaine displays several anti-oncogenic effects by direct interaction with cancer cells. However, once again, concentrations used in these studies strongly deviates from daily clinical application.
Table 5. Studies describing the direct effects of Lidocaine on cancer cells, the associated mechanisms of action, and their respective pathways 1.
Table 5. Studies describing the direct effects of Lidocaine on cancer cells, the associated mechanisms of action, and their respective pathways 1.
Lidocaine/Local Anaesthetics (-Ester and -Amide)
StudyType of CancerEffect on CancerMechanism of ActionPathway Described
Piegeler T. et al. [117]Lung
(NSCLC)
Both ropivacaine and lidocaine blocked tumour cell invasion and MMP-9 secretionAttenuation of Src-dependent inflammatory signalling eventsSrc-dependent activation of Akt and focal adhesion kinase (FAK) and phosphorylation of caveolin-1 (Cav-1) by Src, resulting in reduced MMP-9 synthesis
Piegeler T. et al. [118]Lung (NSCLC)This study indicates that amide-, but not ester-linked local anaesthetics may inhibit migration of tumour cellsIndependent mechanism of voltage gated Sodium channel inhibitionThe inhibition of Tumour Necrosis Factor-α-induced Src-activation and Intercellular Adhesion Molecule-1 (ICAM1) phosphorylation
Wang H.W. et al. [108]Lung (NSCLC)
  • Lidocaine and ropivacaine inhibited cell growth and arrested cell cycle at G0/G1 phase
  • Lidocaine and ropivacaine treatment induced apoptosis
  • Downregulation of cyclin D1 expression, inhibiting cell growth
  • Local anaesthetics could activate the mitochondrial apoptotic pathway.
The phosphorylation of ERK1/2 and JNK increased, suggesting that ERK1/2, JNK, and p38 MAPK may have different effects on apoptosis
Sun H. et al. [121]Lung
(NSCLC)
Inhibition of lung cancer cell growth and metastasis possibly through regulating mitochondria-dependent and EMT-related signallingUpregulation of miR-539, which blocked EGFR signalling by directly binding with EGFRPI3K/AKT signalling
Chamaraux-Tran T.N. et al. [122]Breast
  • Reduction in tumour viability, cell growth and migration in vitro
  • Reduction in cell growth and increased survival in vivo
Non describedNon described
Lirk P. et al. [123]BreastDemethylation of DNA of breast cancer cell lines in vitro (in clinical relevant concentrations)
  • No effect on three known tumour suppressor genes (RASSF1A, MYOD1 and GSTP1)
  • Demethylating effects are dependent on the type of cancer cell
Methylation of DNA changes epigenetic expression which affects expression
Li R. et al. [124]BreastSignificant cytotoxic effect in high concentrations(1 mM), none in physiological concentrations (10 µM). arrest of MDA-MB-231 cells in the S phase for both concentrations. Most significant effect was found in the levobupivacaine group.Non describedNon described
D’Agostino G. et al. [120]BreastInhibition of CXCL12-induced in vitro migration of MDA-MB-231 cells
  • Lidocaine, in clinical concentrations, inhibits CXCL12-induced CXCR4 signalling, which impairs the essential cascade of cytoskeleton remodelling, leading to a reduced migration of breast cancer cells
  • Lidocaine treatment promotes upregulation of CD44 expression (a transmembrane glycoprotein important for cancer interaction with hyaluronic acid), an essential component of the extracellular matrix
Exact mechanism unclear
Jiang Y. et al. [119]Breast, prostate and ovarium
  • Lidocaine was able to Significant decrease in cell viability of MDA-MB-231, PC-3 and ES-2 cells in a concentration-dependent manner
  • Inhibition of the migration and invasion of cancer cells at concentrations that are much lower than clinical concentrations
The influx of calcium plays a role in invasion and migrationNo described pathway for cytotoxicity, could occur partly as a result of the downregulation of TRPV6 expression, with reduced rate of calcium influx
Xuan W. et al. [112]Ovarium and prostateIncreased apoptosis in both cell lines after 24 h of exposure, both through intrinsic and extrinsic pathways in ovarian cells, but only intrinsic in prostate cells.
  • Caspase 8 and 9 inhibition generated partial cell death reversal in SKOV-3, whilst only caspase 9 was effective in PC-3
  • Bupivacaine increased the phosphorylation of GSK-3βTyr216 in SKOV-3 but without measurable effect in PC3
  • Caspase 8 and 9 inhibition generated partial cell death reversal in SKOV-3, whilst only caspase 9 was effective in PC-3
  • Bupivacaine increased the phosphorylation of GSK-3βTyr216 in SKOV-3 but without measurable effect in PC3
Chang YC. et al. [113]ThyroidLidocaine and bupivacaine induced apoptosis, and necrosis in high concentrations in thyroid cancer cells
  • Possible direct effect of local anaesthetics on mitochondria
  • Increased pro-apoptotic Bax expression and downregulated Bcl-2 expression, facilitating release of cytochrome C
MAPK phosphorylation influences caspase 3 release
Xing W. et al. [114]Hepato-cellular
  • Suppression of tumour growth and induction of apoptosis in human HepG2 cells in vitro
  • In vivo, lidocaine not only suppressed hepatocellular carcinoma development but also sensitized hepatocellular carcinoma to cisplatin
  • Increased ratio of Bax/Bcl-2
  • Increased caspase 3 activation
  • Activation of apoptosis
Phosphorylation of ERK1/2 and P38 through the MAPK pathway
Le Gac G. et al. [125]Hepato-cellularLocal anaesthetics decreased viability and proliferation of HuH7 cells and HepaRG progenitor cells
  • Ropivacaine stops the G2 phase of the cell cycle in HCC cells by decreasing key cell cycle regulators
  • Increased apoptosis marked by increased caspase 3
  • Lidocaine increased mRNA levels of APC and of DKK1, which both act as antagonists of the Wnt/β-catenin pathway
  • Ropivacaine decreased the mRNA level of cyclin A2, cyclin B1, cyclin B2, and cyclin-dependent kinase 1, and the expression of the nuclear marker of cell proliferation MKI67
Zhao L. et al. [126]Hepato-cellular
  • Repression of hepatocellular carcinoma cell proliferation, migration, and invasion
  • Promotion of apoptosis
UnclearVia regulating circ_ITCH/miR-421/CPEB3 axis.
Liu H. et al. [127]Hepato-cellularDecrease in HepG2 cell viability and colony formation in a dose-dependent mannerUnclearCPEB3 as a critical mediator of lidocaine-induced repression of HepG2 cell proliferation
Bundscherer AC. et al. [109]Colon
  • Induction of cell-cycle arrest in both colon carcinoma cell lines in vitro, but no effect on apoptosis
  • Small increase in proliferation between 10–100 µM lidocaine in SW480 cells
Cell cycle arrest, exact mechanism unclearUnclear
Yang W. et al. [110]GastricLidocaine and ropivacaine inhibited the proliferation of AGS and HGC-27 cells within 72 h. Especially lidocaine at doses of 10 μM or above (which is safe as the blood level for clinical use)Significant reduction of expression of p-ERK1/2 in AGS and HGC-27 cellsMAPK pathway
Ye L. et al. [115]Gastric
  • Significant suppression of proliferation, migration and invasion
  • Induction of apoptosis in a dose-dependent manner in human gastric cancer cells
  • Simultaneous p-p38 increasement, while the level of p38 was not affected
  • Increased Bax level and decreased Bcl-2 level in a dose-dependent manner
MAPK pathway
Sakaguchi M. et al. [131]Tongue
  • In a clinical concentration of lidocaine (400 μM): suppressed proliferation and without cytotoxicity
  • In a larger concentration of lidocaine (4000 μM): cytotoxicity with an antiproliferative effect
Inhibition of EGF-stimulated tyrosine kinase activity of EGFRDirect inhibition tyrosine kinase activity
Chen J. et al. [111]Melanoma
  • Inhibition of A375 melanoma cell proliferation in a dose-and time-dependent manner
  • Colony formation in a dose-dependent inhibition
Arrest of cell-cycle progression in the G1 phase and inhibited Ki-67 expression in a dose-dependent mannerInhibition of ERK signalling pathway
Bezu L. et al. [116]Colon, breast, cervix, osteosarcoma, fibrosarcomaLidocaine and other anaesthetics induced signs of cancer cell stress including inhibition of oxidative phosphorylation, and induction of autophagy as well as endoplasmic reticulum (ER) stressInduction of ER stress, resulting in eIF2α phosphorylation, causing activation of autophagyEIF2AK3/PERK-dependent eIF2α phosphorylation leading to ATF4 translation, IRE1-mediated XBP1 activation, as well as activation of the latent transcription factor ATF6
1 The correlated hallmark of cancer is given for reference on the role and importance in cancer development. Sorted by type of tissue.

4. Discussion

In the multistep development of human cancers, certain abilities must be acquired along the way in order to fully differentiate as a malignant cancer cell. Next to this, some of these abilities must be kept acquired in order to be able to further survive and metastasize. These abilities are at present times best described by the model known as the hallmarks of cancer, specifying the particular importance of the different mechanisms discerning malignant cells from healthy cells. Historically, six hallmarks have been described, namely resisting cell death, sustaining proliferative signalling, evading growth suppressors, activating invasion and metastasis, enabling replicative immortality, and inducing angiogenesis. More recently, other hallmarks of cancer have been suggested as playing a vital role in tumour progression. Hallmarks such as avoiding immune destruction, tumour-promoting inflammation, genome instability and mutation, and deregulating cellular energetics. Where the abilities to sustain proliferative signalling and evade growth suppressors lie at the start of the development of a malignant cell, by dividing without limitation, other hallmarks such as genome instability and mutation, resisting cell death, and enabling replicative immortality help it by further differentiating from regular cells and mutating more favourable genes. The other hallmarks help to further amplify growth signalling, regulate the delivery of nutrients and oxygen, and create a favourable microenvironment for the tumour to thrive in. This includes increasing vascular growth, recruiting stromal cells for further positive signalling, and manipulating the immune response to gain a more favourable outcome for the cancer cells. Finally, the ability to activate invasion and metastasis helps the spread to further satellites and increases the mortality of cancer itself [5]. In this point of view, cancer cells are present in a complex tissue microenvironment whereby there is an interaction of surrounding different types of cells including noncancerous cells, cells of the immune system, the extracellular matrix, chemokines, cytokines, and other factors [132]. The microenvironment provides positive growth signalling but manages also energy delivery through vascular growth and immune modulation. Interactions between circulating tumour cells and the microenvironmental components of circulation determine survival and the ability of these cells to eventually extravasate in distant sites [133,134,135]. A final step in differentiating between primary tumour and metastasis is the induction of epithelial to mesenchymal transition (EMT), where the cell loses its polarity and cell–cell adhesions through multiple biological transformations and changes into a mesenchymal cell type, facilitating migration and invasion, ultimately resulting in increased metastatic potential [136]. Spontaneous epithelial-mesenchymal transition EMT in primary tumour cells shifts between different intermediate stages with different characteristics [137]. It has been reported that the EMT programme is a gamut of transitional steps between the epithelial and mesenchymal phenotypes. In fact, studies suggest that the nature of the primary cancer cell determines the different metastatic properties with respect to growth and response to therapy [138,139].
Generally, not all hallmarks seem to be affected by anaesthetics, but there is evidence that some anaesthetics are able to influence certain hallmarks. As it is known that immune responses are regulated by the hypothalamic-pituitary-adrenal (HPA) axis and the sympathetic nervous system (SNS), it is no surprise that activation of these systems induced by surgery or anaesthetics may facilitate tumour activation or distant metastasis through several tumour-derived soluble factors suppressing the HPA axis, activating SNS cellular immune responses (CMI) and releasing catecholamines and prostaglandin E2 (PGE2) [140]. Subsequently, these factors will increase immunosuppressive cytokines, and soluble factors (for example interleukin-4 [IL-4], IL-10, transforming growth factor Beta [TGF-β], vascular endothelial growth factor VEGF]), and pro-inflammatory cytokines (e.g., IL-6 and IL-8), and ends up promoting angiogenesis and metastasis [141,142,143,144].
It is known, and widely accepted that a number of factors that occur in the perioperative period have a significant and direct impact on the status of cancer cells on the one hand and the body’s cellular immunity on the other hand. This concept enhances the possibility of cancer spreading or reactivating cancer cells from a state of inactivity and dormancy, and turning them into functional and active cancer cells. Since the perioperative changes to immune function, inflammation, stress response and cancer activity are complex and multifactorial, there has been significant difficulty in describing causality or associative connections between anaesthesia and cancer, especially since microenvironmental regulation and interactions between host reactions and cancer cells are difficult mechanisms to recreate in a research environment. A limitation of this study is that only the direct cellular effects of anaesthetics on tumour mechanisms and development were evaluated through extensive research of literature. Recent clinical trials have tried to study the multifactorial indirect effects of anaesthesia during surgery. Comparing perioperative epidural analgesia with opioid analgesia in thoracic and abdominal surgery showed no difference in overall survival and disease-free survival either [2,3]. The anaesthetic protocols of the latter study were matched for the hypothesis that the effect of opioids might be immunomodulatory and that locoregional anaesthesia could possibly inhibit stress response in major surgery. However, both studies allowed for opioids to be used in epidural groups, and showed similar opioid consumption. When the proposed mechanisms of those studies were compared to literature search findings of this review, several conclusions can be made. Since this study was limited to the direct effects of anaesthetics, we cannot exclude a possible effect of the shift in immune function or changes in stress response. Stress response refers to the situation in which a complex series of physiological events occurs following an injury or trauma. It is, therefore, best defined as the natural response to physiological stress that occurs as a result of surgery and associated perioperative events. Beside the activation of the neuroendocrine system, there will be some inflammatory changes and activation of the hypothalamic-pituitary-adrenal axis. The complex interplay of these events, in turn, leads to the formation of a constellation of immune, hemodynamic and metabolic changes. Surgical stress is induced by the release of norepinephrine and epinephrine that may interact with β1 and 2 receptors expressed by cancer cells increasing their invasive and proliferative capacity [141,145,146]. Although it has been believed that the stress response plays a beneficial role in recovery and survival after injury, several publications have appeared in recent years documenting that with regard to cancer recurrence and metastases after surgery, the effect of the stress response may not be beneficial. Certain elements of the stress response are thought to boost cancer growth [1]. In addition, the inflammatory component of the stress response includes the production and release of cytokines, prostaglandins and cyclooxygenase. The chronic release of such mediators are thought to play a role in cancer development through inhibition of apoptosis, promotion of angiogenesis and immunosuppression [147]. It has also been suggested that acute release of such mediators during the perioperative period may promote cancer growth [148]. Finally, pain activates the hypothalamic-pituitary-adrenal (HPA) axis, which has been implicated in immunosuppression, reduction of natural killer cell activity and enhancement of tumour cell activity in animals [149,150].
Forget. et al. showed that surgical trauma in rats works as a very powerful metastatic stimulus [14]. Since greater surgical stress is associated with more pain and worse surgical outcome, it is very difficult to objectively measure its role on cancer in human subjects. To our knowledge, there are no human trials on the causality of stress response and cancer outcome. Surrogate markers can be used such as postoperative pain, changes in vital signs attributable to sympathetic stimuli or endocrine measurements. In animal studies, there is evidence to back up this theory [13]. For pain and postoperative morphine consumption, there were no significant differences in the comparison of epidural and opioid analgesia, and no data on stress hormones, thus making it difficult to interpret the effect of the stress response on outcome. The lack of arguments for changed stress responses in all three RCTs might explain the lack of significant results [2,3,4]. Opioids may display an immunomodulatory effect, as mentioned earlier. They possibly decrease NK cell function and modulate the function and differentiation of T cells. It is well known that NK cells play a major role in the first immune defence against malignancy and that factors such as neutrophil-to-lymphocyte ratio (NLR) have a negative prognostic value in cancer outcomes. In this point of view, morphine suppresses neutrophil functions such as phagocytosis, respiratory burst, and complement receptor expression by stimulating NO release via μ3 receptors [151]. T lymphocyte functions and B lymphocyte functions are also suppressed by morphine in vivo. The mitogenic response of B-lymphocytes plasma cells [152] is suppressed by morphine administration in vivo. Moreover, T-lymphocyte proliferation is decreased by both acute and chronic morphine administration [153]. In contrast to the inhibitory effects induced by morphine on immune cells, synthetic opioids such as fentanyl and remifentanil have no effect in attenuating immune cell responses because of a reduced interaction of synthetic opioids with specific opioid receptors. Fentanyl, remifentanil, and alfentanil do not impair functions of neutrophils such as respiratory burst [154] and phagocytosis [155] Sevoflurane may also affect NK cell function and has an immunomodulatory effect [14,98,156,157,158,159]. However, the effect of sevoflurane on immune function is unsure, as some studies cannot find any difference in immune function [41,160]. Furthermore, local anaesthetics in high concentration have recently been proven to trigger T lymphocyte-dependent tumour growth reduction through ER stress induction and eliciting other immunostimulatory stress signals including the release of ATP and HMGB1 from cancer cells [116]. These finding, together with the abovementioned possible changes in stress response further complicate translation of in vitro studies to the in vivo setting, and interpretation of aforementioned RCT’s.
To the best of our knowledge, no clinical study has shown the direct effect of immunomodulation by opioids or sevoflurane on cancer outcomes in a surgical setting. Xu. et al. have shown that serum treated with opioids and sevoflurane has a detrimental effect on cancer development compared to serum treated with propofol and locoregional anaesthesia, but only in vitro [100]. In summary, it is unsure what exactly the importance of immunomodulation is in the perioperative setting on cancer outcomes taking the direct effects of anaesthetics into account. However, since previously mentioned RCTs have not produced significant results, it is possible that the effect of this immunomodulation is nearly as important as previously thought, especially compared to the larger effect of the surgical stress response.
When considering the direct effects of sevoflurane, it can be stated that several discrepancies exist throughout the body of evidence concerning the effect of sevoflurane on cancer cells. It is possible that sevoflurane increases angiogenesis through mechanisms such as HIF1a; however, this is uncertain. Differences between tissue selection, time of exposure or concentrations used may explain these discrepancies. The concentration of sevoflurane turns out to be especially important since Deng et al. described a duality in action depending on the concentration of sevoflurane, where higher doses were more cytotoxic [31]. Since most recent clinical studies leave sevoflurane dosage to physician’s discretion, it might be a possible cofounder skewing results.
When overviewing the results of the performed literature search, there are several arguments to suspect possible effects on cancer cells by the anaesthetics researched. Very few of these mechanisms are evaluated in current RCTs. Additionally, there is retrospective evidence that the use of some anaesthetics would be beneficial in cancer surgery. Therefore, it is very surprising that no benefits have been found in recent randomised clinical trials. A number of explanations can be formulated concerning these results. The first is the absence of effect on cancer cells in study designs with low concentrations of anaesthetics and short periods of exposure. We observed that most of the anaesthetic study drugs were used in vitro at supraphysiological concentrations with long exposure times. Although this makes sense for in vitro studies to increase the chance of significant outcomes, it makes translation to in vivo research very challenging. Most studies report lack of significant outcome when doses are lowered or exposure time is shortened such as observed with midazolam, dexmedetomidine, lidocaine and propofol [6,10,48,49,64,66,68,73,74,75,76,109,111,114,115,118,119,121,123,124,125,126,127]. There are studies that prove possible effects in physiological doses and exposure time for lidocaine, but to a lesser extent [120,129]. The effects of opioids in anaesthesia for cancer surgery are highly debatable, since direct effects in vitro are contradictory, and the chronic use of opioids is frequent in cancer patients. It may be possible that the immunomodulatory effects play a role in cancer outcome, but this is not clearly proven in clinical studies so far. Sevoflurane is inconclusive as well due to the duality of its effect and lack of transparency on dosing protocols in clinical studies. Thus, a possible scenario could be that when these anaesthetics are used in clinically relevant concentrations and duration, there is an absence of a true direct effect on cancer cells. The second explanation for these conflicting results found in literature is the complex interaction between tumour and microenvironment and the biochemical changes of EMT. These are complex physiological interactions that are very hard to recreate in vitro, thus making research more complicated and difficult. Additionally, it may have a huge effect on the behaviour of cancer exposed to anaesthetics. This microenvironment is easily perturbed by any tissue trauma, and surgical intervention aimed at eliminating the disease may unintentionally create conditions that not only promote survival, but also the propagation, multiplication and spread of residual cancer cells. Such surgical-induced physiological changes are many and may include among which inflammation, tissue hypoxia, angiogenesis, surgical stress response, and immunosuppression [161]. All opioids, volatile and local anaesthetics are able to influence, directly or indirectly, the rate of proliferation, epithelial-mesenchymal transition, and the invasiveness properties of cancer cells as well as various elements of the tumour microenvironment. Nevertheless, none of these drugs has proven to have a direct causal relationship between their use in the perioperative period and a diminution of cancer recurrence rates or an increase in cancer-specific survival. As the last argument, stronger and more strict study protocols are needed to find consistent significant results.
In this point of view, a number of limitations can frequently be found in study designs and in recommendations that define how to solve these problems for future study design. The first limitation is matching for population and tumour confounders. It is important to note that these do not address the influence of the histopathological parameters such as tumour histology, size, margins, and the number of positive nodes as key factors that determines the risk of cancer recurrence. Another major drawback is that factors such as patient’s age, ethnicity, and gender, the patient’s general health status, and factors affecting the quality and efficiency of the surgical procedure such as the experience of the surgeons as well as patient pre, peri, and postoperative care have been scarcely investigated from the theoretical point of view. Apart from this is that the efficiency of metastatic detection techniques has been improved in recent years through the developing of novel methods for early detection, monitoring, and surveillance. Nonetheless, molecular-genetic imaging approaches allowing the visualisation and quantification of biochemical processes at the cellular and molecular level are needed to improve the tools and methodologies for monitoring circulating tumour cells. Additionally, when achieved, this might significantly enhance the power and accuracy of this approach for monitoring patient cancers noninvasively [162,163].
The second limitation is the transparency of drug dosing. In recent trials, there is transparency on the drugs used in the anaesthetic protocol, as well as documentation about perioperative consumption of possible relevant drugs. However, the dosing of applied drugs is left to the discretion of the treating anaesthesiologist. Since we now have various arguments on why the exact dose is also important. Since sevoflurane might have a different effect in low or high concentrations, knowing the dosage used perioperatively might help differentiate the effect on the outcome. Anaesthesia is rarely executed with a single anaesthetic, and study interventions might change dosing regiments of concomitant anaesthetics during surgery, further complicating interpretation of results. Additionally, dosing of analgesics compared to hemodynamic monitoring or nociceptive monitoring might give an idea of the relative surgical stress levels of the patient during surgery. As this may play an important role in cancer outcomes, collecting data on these surrogates might be interesting. Morphine consumption is frequently described as a measure for postoperative pain, but collected data perioperatively are generally lacking.
The third limitation is optimizing the study design for primary tumour or metastasis. A distinction must be made between targeting a primary tumour and targeting microsatellites or metastasis. As mentioned above, the microenvironment of the tumour is a complex synergistic interaction between many different cells. It is well known that concentrations and exposure are important factors in determining the effects of anaesthesia on cancer cells. Keeping the pharmacokinetics of anaesthetic drugs in mind, it may be very unlikely that a significant concentration of any kind is reached at the location of the primary tumour and its microenvironment. In vitro, it is obvious to reach the desired concentration of the drug at the target site, thus producing significant results. In the case of in vivo studies, this is more complicated and therefore troubled. Most studies that show anti-oncogenic effects on the primary tumour in vivo are animal studies dealing with very high nonclinical doses with a long exposure period [6,8,9,32,59,66,72,85,91,114,164]. Studies that apply clinically more comparable doses described more often an effect on metastasis and invasion than on reduction of the primary tumour [7,22,53,88,89,117,165]. It is possible that the in vivo concentration is higher intravenously or in highly vascularized areas. Additionally, circulating tumour satellites that are exposed to these concentrations are more strongly affected. In the clinical setting, the concentrations of anaesthetics are highest intravenously, depending on the diffusion coefficient of the drug used. This difference is mediated as diffusion to other tissues reaches equilibrium, but in practice, intravenous concentrations are higher. To conclude, it may be of major interest to harvest significant results if the focus is on the EMT and microsatellites leading to invasion and metastasis, since the concentrations of anaesthetics are highest in blood circulation and not at the site of the primary tumour.
The fourth limitation is the true importance of cancer tissue selection for research. In this literature research, several types of tissue were found that display different sensitivity to different anaesthetic drugs. For sevoflurane and lidocaine, different thresholds of a significant effect were found depending on the type of cell line and type of tissue [29,30,35,109,115,119,122]. This may play an important role in case selection in clinical studies. By limiting studies to only one type of tumour, and by correcting the tumour for known differences in differentiation or receptor expression, significant differences in outcome may be found when comparing anaesthetics.

5. Conclusions

There is evidence that anaesthesia can influence the biology of cancer through different hallmark mechanisms, although this is mostly described in vitro with the usage of very high concentrations that do not match daily clinical use. Most significant effects are only found in long durations of exposure, much longer than in most clinical anaesthetic procedures. For most anaesthetics, the effects are clearly correlated to drug concentration and time of exposure. These effects are likely dependent on various confounders, such as type of tumour, applied concentration, population, stress response and time of exposure. Yet, none of these have demonstrated a direct correlative causality between their use in the perioperative period and a reduction in cancer recurrence or an increase in cancer-specific survival. Moreover, a potential individual drug effect still remains complex to clarify in a clinical situation where different anaesthetics and drugs are similarly administered. In this review, no strict evidence of a significant clinical effect on cancer outcomes during surgery could be found. As a consequence, more transparent study protocols are absolutely necessary together with the exact description of drug dose and duration used during surgery for a better evaluation of the effect on cancer outcome, since these effects are strongly dose- and duration-dependent.

Author Contributions

Conceptualization, W.D., A.R., C.V. and R.G.F.; methodology, W.D., C.V. and R.G.F.; resources, W.D., A.R. and R.G.F.; writing—original draft preparation, W.D. and A.R.; writing—review and editing, C.V. and R.G.F.; supervision, C.V. and R.G.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Heaney, A.; Buggy, D.J. Can anaesthetic and analgesic techniques affect cancer recurrence or metastasis? Br. J. Anaesth. 2012, 109, i17–i28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Du, Y.-T.; Li, Y.-W.; Zhao, B.-J.; Guo, X.-Y.; Feng, Y.; Zuo, M.-Z.; Fu, C.; Zhou, W.-J.; Li, H.-J.; Liu, Y.-F.; et al. Long-term Survival after Combined Epidural–General Anesthesia or General Anesthesia Alone: Follow-up of a Randomized Trial. Anesthesiology 2021, 135, 233–245. [Google Scholar] [CrossRef] [PubMed]
  3. Xu, Z.-Z.; Li, H.-J.; Li, M.-H.; Huang, S.-M.; Li, X.; Liu, Q.-H.; Li, J.; Wang, D.-X.; Sessler, D.I. Epidural Anesthesia–Analgesia and Recurrence-free Survival after Lung Cancer Surgery: A Randomized Trial. Anesthesiology 2021, 135, 419–432. [Google Scholar] [CrossRef] [PubMed]
  4. Sessler, D.I.; Pei, L.; Huang, Y.; Fleischmann, E.; Marhofer, P.; Kurz, A.; Mayers, D.B.; Meyer-Treschan, T.A.; 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] [PubMed]
  5. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  6. Wang, C.; Datoo, M.T.; Zhao, H.; Wu, M.L.; Date, M.A.; Jiang, M.C.; Sanders, M.R.D.; Wang, G.; Bevan, C.; Ma, M.D. Midazolam and Dexmedetomidine Affect Neuroglioma and Lung Carcinoma Cell Biology In Vitro and In Vivo. Anesthesiology 2018, 129, 1000–1014. [Google Scholar] [CrossRef]
  7. Qi, Y.; Yao, X.; Du, X. Midazolam inhibits proliferation and accelerates apoptosis of hepatocellular carcinoma cells by elevating microRNA-124-3p and suppressing PIM-1. IUBMB Life 2019, 72, 452–464. [Google Scholar] [CrossRef]
  8. Mishra, S.K.; Kang, J.-H.; Lee, C.W.; Oh, S.H.; Ryu, J.S.; Bae, Y.S.; Kim, H.M. Midazolam induces cellular apoptosis in human cancer cells and inhibits tumor growth in xenograft mice. Mol. Cells 2013, 36, 219–226. [Google Scholar] [CrossRef] [Green Version]
  9. Zhang, P.; He, H.; Bai, Y.; Liu, W.; Huang, L. Dexmedetomidine suppresses the progression of esophageal cancer via miR-143-3p/epidermal growth factor receptor pathway substrate 8 axis. Anti-Cancer Drugs 2020, 31, 693–701. [Google Scholar] [CrossRef]
  10. 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]
  11. He, H.; Chen, J.; Xie, W.; Cao, S.; Hu, H.; Yang, L.; Gong, B. Ketamine used as an acesodyne in human breast cancer therapy causes an undesirable side effect, upregulating anti-apoptosis protein Bcl-2 expression. Genet. Mol. Res. 2013, 12, 1907–1915. [Google Scholar] [CrossRef]
  12. 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]
  13. Glasner, A.; Avraham, R.; Rosenne, E.; Benish, M.; Zmora, O.; Shemer, S.; Meiboom, H.; Ben-Eliyahu, S. Improving Survival Rates in Two Models of Spontaneous Postoperative Metastasis in Mice by Combined Administration of a β-Adrenergic Antagonist and a Cyclooxygenase-2 Inhibitor. J. Immunol. 2010, 184, 2449–2457. [Google Scholar] [CrossRef] [Green Version]
  14. Forget, P.; Collet, V.; Lavand’homme, P.; De Kock, M. Does analgesia and condition influence immunity after surgery? Effects of fentanyl, ketamine and clonidine on natural killer activity at different ages. Eur. J. Anaesthesiol. 2010, 27, 233–240. [Google Scholar] [CrossRef]
  15. Forget, P.; Vandenhende, J.; Berliere, M.; Machiels, J.-P.; Nussbaum, B.; Legrand, C.; De Kock, M. Do intraoperative analgesics influence breast cancer recurrence after mastectomy? A retrospective analysis. Anesth. Analg. 2010, 110, 1630–1635. [Google Scholar] [CrossRef] [Green Version]
  16. Benzonana, L.L.; Perry, N.J.S.; Watts, H.R.; Yang, B.; Perry, I.A.; Coombes, C.; Takata, M.; Ma, D. Isoflurane, a Commonly Used Volatile Anesthetic, Enhances Renal Cancer Growth and Malignant Potential via the Hypoxia-inducible Factor Cellular Signaling Pathway In Vitro. Anesthesiology 2013, 119, 593–605. [Google Scholar] [CrossRef]
  17. Luo, X.; Zhao, H.; Hennah, L.; Ning, J.; Liu, J.; Tu, H.; Ma, D. Impact of isoflurane on malignant capability of ovarian cancer in vitro. Br. J. Anaesth. 2015, 114, 831–839. [Google Scholar] [CrossRef] [Green Version]
  18. Iwasaki, M.; Zhao, H.; Jaffer, T.; Unwith, S.; Benzonana, L.; Lian, Q.; Sakamoto, A.; Ma, D. Volatile anaesthetics enhance the metastasis related cellular signalling including CXCR2 of ovarian cancer cells. Oncotarget 2016, 7, 26042–26056. [Google Scholar] [CrossRef] [Green Version]
  19. Huang, H.; Benzonana, L.L.; Zhao, H.; Watts, H.R.; Perry, N.J.S.; Bevan, C.L.; Brown, R.; Ma, D. Prostate cancer cell malignancy via modulation of HIF-1α pathway with isoflurane and propofol alone and in combination. Br. J. Cancer 2014, 111, 1338–1349. [Google Scholar] [CrossRef] [Green Version]
  20. Kawaraguchi, M.Y.; Horikawa, Y.T.; Murphy, A.N.; Murray, F.; Miyanohara, A.; Ali, S.S.; Head, B.P.; Patel, M.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] [Green Version]
  21. Bundscherer, A.C.; Ullrich, V.; Malsy, M.; Gruber, M.A.; Graf, B.M.; Brockhoff, G.; Sinner, B. Effects of Volatile Anesthetics on Proliferation and Viability of SW480 Colon Cancer Cells In Vitro. Anticancer Res. 2019, 39, 6049–6055. [Google Scholar] [CrossRef] [PubMed]
  22. Yang, X.; Zheng, Y.-T.; Rong, W. Sevoflurane induces apoptosis and inhibits the growth and motility of colon cancer in vitro and in vivo via inactivating Ras/Raf/MEK/ERK signaling. Life Sci. 2019, 239, 116916. [Google Scholar] [CrossRef] [PubMed]
  23. He, J.; Zhao, H.; Liu, X.; Wang, D.; Wang, Y.; Ai, Y.; Yang, J. Sevoflurane suppresses cell viability and invasion and promotes cell apoptosis in colon cancer by modulating exosome-mediated circ-HMGCS1 via the miR-34a-5p/SGPP1 axis. Oncol. Rep. 2020, 44, 2429–2442. [Google Scholar] [CrossRef] [PubMed]
  24. Sun, S.Q.; Ren, L.J.; Liu, J.; Wang, P.; Shan, S.M. Sevoflurane inhibits migration and invasion of colorectal cancer cells by regulating microRNA-34a/ADAM10 axis. Neoplasma 2019, 66, 887–895. [Google Scholar] [CrossRef] [PubMed]
  25. Fan, L.; Wu, Y.; Wang, J.; He, J.; Han, X. Sevoflurane inhibits the migration and invasion of colorectal cancer cells through regulating ERK/MMP-9 pathway by up-regulating miR-203. Eur. J. Pharmacol. 2019, 850, 43–52. [Google Scholar] [CrossRef]
  26. Ciechanowicz, S.; Zhao, H.; Chen, Q.; Cui, J.; Mi, E.; Lian, Q.; Ma, D. Differential effects of sevoflurane on the metastatic potential and chemosensitivity of non-small-cell lung adenocarcinoma and renal cell carcinoma in vitro. Br. J. Anaesth. 2017, 120, 368–375. [Google Scholar] [CrossRef] [Green Version]
  27. Chen, H.; Zhu, X.M.; Luo, Z.L.; Hu, Y.J.; Cai, X.C.; Gu, Q.H. Sevoflurane induction alleviates the progression of gastric cancer by upregulating the miR-34a/TGIF2 axis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 11883–11890. [Google Scholar] [CrossRef]
  28. Ferrell, J.K.; Cattano, D.; Brown, R.E.; Patel, C.B.; Karni, R.J. The effects of anesthesia on the morphoproteomic expression of head and neck squamous cell carcinoma: A pilot study. Transl. Res. 2015, 166, 674–682. [Google Scholar] [CrossRef]
  29. Zhang, M.W.; Sheng, M.B.; Chen, M.S.; Zhao, B.H.; Wu, B.L.; Sun, M.Y.; Cui, B.J.; Zhu, M.X.; Ma, M.D. Sevoflurane Enhances Proliferation, Metastatic Potential of Cervical Cancer Cells via the Histone Deacetylase 6 Modulation In Vitro. Anesthesiology 2020, 132, 1469–1481. [Google Scholar] [CrossRef]
  30. Ecimovic, P.; McHugh, B.; Murray, D.; Doran, P.; Buggy, D.J. Effects of sevoflurane on breast cancer cell function in vitro. Anticancer Res. 2013, 33. [Google Scholar]
  31. 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, 253. [Google Scholar] [CrossRef]
  32. Kang, K.; Wang, Y. Sevoflurane Inhibits Proliferation and Invasion of Human Ovarian Cancer Cells by Regulating JNK and p38 MAPK Signaling Pathway. Drug Des. Dev. Ther. 2019, 13, 4451–4460. [Google Scholar] [CrossRef]
  33. Zhao, H.; Xing, F.; Yuan, J.; Li, Z.; Zhang, W. Sevoflurane inhibits migration and invasion of glioma cells via regulating miR-34a-5p/MMP-2 axis. Life Sci. 2020, 256, 117897. [Google Scholar] [CrossRef]
  34. Xu, W.; Xue, R.; Xia, R.; Liu, W.-W.; Zheng, J.-W.; Tang, L.; Kang, L.-Y.; Wang, W.; Wei, W.-T. Sevoflurane impedes the progression of glioma through modulating the circular RNA has_circ_0012129/miR-761/TGIF2 axis. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 5534–5548. [Google Scholar] [CrossRef]
  35. Lai, R.-C.; Shan, W.-R.; Zhou, D.; Zeng, X.-Q.; Zuo, K.; Pan, D.-F.; Zeng, W.-A.; Zuo, Z.-Y. Sevoflurane promotes migration, invasion, and colony-forming ability of human glioblastoma cells possibly via increasing the expression of cell surface protein 44. Acta Pharmacol. Sin. 2019, 40, 1424–1435. [Google Scholar] [CrossRef]
  36. 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]
  37. Oh, T.K.; Kim, K.; Jheon, S.; Lee, J.; Do, S.-H.; Hwang, J.-W.; Song, I.-A. Long-Term Oncologic Outcomes for Patients Undergoing Volatile Versus Intravenous Anesthesia for Non-Small Cell Lung Cancer Surgery. Cancer Control 2018, 25. [Google Scholar] [CrossRef] [Green Version]
  38. Watson, S.d.L.M.; Puxty, K.; Moran, D.; Morrison, D.S.; Sloan, B.; Buggy, D.; Shelley, B. Association between Anesthetic Dose and Technique and Oncologic Outcomes after Surgical Resection of Non-Small Cell Lung Cancer. J. Cardiothorac. Vasc. Anesth. 2021, 35, 3265–3274. [Google Scholar] [CrossRef]
  39. 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]
  40. Enlund, M.; Berglund, A.; Ahlstrand, R.; Walldén, J.; Lundberg, J.; Wärnberg, F.; Ekman, A.; Widfeldt, N.S.; Enlund, A.; Bergkvist, L. Survival after primary breast cancer surgery following propofol or sevoflurane general anesthesia—A retrospective, multicenter, database analysis of 6305 Swedish patients. Acta Anaesthesiol. Scand. 2020, 64, 1048–1054. [Google Scholar] [CrossRef]
  41. Hovaguimian, F.; Braun, J.; Z’Graggen, B.R.; Schläpfer, M.; Dumrese, C.; Ewald, C.; Dedes, K.J.; Fink, D.; Rölli, U.; Seeberger, M.; et al. Anesthesia and Circulating Tumor Cells in Primary Breast Cancer Patients. Anesthesiology 2020, 133, 548–558. [Google Scholar] [CrossRef] [PubMed]
  42. Meng, C.; Song, L.; Wang, J.; 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. 2016, 37, 841–848. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, N.; Liang, Y.; Yang, P.; Ji, F. Propofol suppresses LPS-induced nuclear accumulation of HIF-1α and tumor aggressiveness in non-small cell lung cancer. Oncol. Rep. 2017, 37, 2611–2619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. 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, e6803. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Zhou, C.-L.; Li, J.-J.; Ji, P. Propofol Suppresses Esophageal Squamous Cell Carcinoma Cell Migration and Invasion by Down-Regulation of Sex-Determining Region Y-box 4 (SOX4). J. Pharmacol. Exp. Ther. 2017, 23, 419–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ye, Z.; Jingzhong, L.; Yangbo, L.; Lei, C.; Jiandong, Y. Propofol Inhibits Proliferation and Invasion of Osteosarcoma Cells by Regulation of MicroRNA-143 Expression. Oncol. Res. Featur. Preclin. Clin. Cancer Ther. 2014, 21, 201–207. [Google Scholar] [CrossRef]
  47. Wu, K.-C.; Yang, S.-T.; Hsia, T.-C.; Yang, J.-S.; Chiou, S.-M.; Lu, C.-C.; Wu, R.S.-C.; Chung, J.-G. Suppression of cell invasion and migration by propofol are involved in down-regulating matrix metalloproteinase-2 and p38 MAPK signaling in A549 human lung adenocarcinoma epithelial cells. Anticancer Res. 2012, 32, 4833–4842. [Google Scholar]
  48. Huang, X.; Teng, Y.; Yang, H.; Ma, J. Propofol inhibits invasion and growth of ovarian cancer cells via regulating miR-9/NF-κB signal. Braz. J. Med. Biol. Res. 2016, 49, e5717. [Google Scholar] [CrossRef] [Green Version]
  49. 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. 2011, 66, 52–56. [Google Scholar] [CrossRef]
  50. 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]
  51. Peng, Z.; Zhang, Y. Propofol inhibits proliferation and accelerates apoptosis of human gastric cancer cells by regulation of microRNA-451 and MMP-2 expression. Genet. Mol. Res. 2016, 15, gmr.15027078. [Google Scholar] [CrossRef]
  52. Xu, J.; Xu, W.; Zhu, J. Propofol suppresses proliferation and invasion of glioma cells by upregulating microRNA-218 expression. Mol. Med. Rep. 2015, 12, 4815–4820. [Google Scholar] [CrossRef] [Green Version]
  53. Liu, F.; Qiu, F.; Fu, M.; Chen, H.; Wang, H. Propofol Reduces Epithelial to Mesenchymal Transition, Invasion and Migration of Gastric Cancer Cells through the MicroRNA-195-5p/Snail Axis. J. Pharmacol. Exp. Ther. 2020, 26, e920981. [Google Scholar] [CrossRef]
  54. Zhang, Y.; Li, C.; Zhou, Y.; Lu, X. Effects of propofol on colon cancer metastasis through STAT3/HOTAIR axis by activating WIF-1 and suppressing Wnt pathway. Cancer Med. 2020, 9, 1842–1854. [Google Scholar] [CrossRef] [Green Version]
  55. Cui, W.-Y.; Liu, Y.; Zhu, Y.-Q.; Song, T.; Wang, Q.-S. Propofol induces endoplasmic reticulum (ER) stress and apoptosis in lung cancer cell H460. Tumor Biol. 2014, 35, 5213–5217. [Google Scholar] [CrossRef]
  56. Gong, T.; Ning, X.; Deng, Z.; Liu, M.; Zhou, B.; Chen, X.; Huang, S.; Xu, Y.; Chen, Z.; Luo, R. Propofol-induced miR-219-5p inhibits growth and invasion of hepatocellular carcinoma through suppression of GPC3-mediated Wnt/β-catenin signalling activation. J. Cell. Biochem. 2019, 120, 16934–16945. [Google Scholar] [CrossRef]
  57. Du, Y.; Zhang, X.; Zhang, H.; Chen, Y.; Zhu, S.; Shu, J.; Pan, H. Propofol modulates the proliferation, invasion and migration of bladder cancer cells through the miR-145-5p/TOP2A axis. Mol. Med. Rep. 2021, 23, 1–11. [Google Scholar] [CrossRef]
  58. Xu, Y.-B.; Du, Q.-H.; Zhang, M.-Y.; Yun, P.; He, C.-Y. Propofol suppresses proliferation, invasion and angiogenesis by down-regulating ERK-VEGF/MMP-9 signaling in Eca-109 esophageal squamous cell carcinoma cells. Eur. Rev. Med. Pharmacol. Sci. 2013, 17, 2486–2494. [Google Scholar]
  59. Liu, Y.-P.; Heng, J.-Y.; Zhao, X.-Y.; Li, E.-Y. The inhibition of circular RNA circNOLC1 by propofol/STAT3 attenuates breast cancer stem cells function via miR-365a-3p/STAT3 signaling. J. Transl. Med. 2021, 19, 1–16. [Google Scholar] [CrossRef]
  60. Zhang, J.; Zhang, D.; Wu, G.-Q.; Feng, Z.-Y.; Zhu, S.-M. Propofol inhibits the adhesion of hepatocellular carcinoma cells by upregulating microRNA-199a and downregulating MMP-9 expression. Hepatobiliary Pancreat. Dis. Int. 2013, 12, 305–309. [Google Scholar] [CrossRef]
  61. 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. [Google Scholar]
  62. Yu, B.; 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] [PubMed]
  63. 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]
  64. Yang, N.; Liang, Y.; Yang, P.; Yang, T.; Jiang, L. Propofol inhibits lung cancer cell viability and induces cell apoptosis by upregulating microRNA-486 expression. Braz. J. Med. Biol. Res. 2017, 50, e5794. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, J.; Wu, G.-Q.; Zhang, Y.; Feng, Z.-Y.; Zhu, S.-M. Propofol induces apoptosis of hepatocellular carcinoma cells by upregulation of microRNA-199a expression. Cell Biol. Int. 2012, 37, 227–232. [Google Scholar] [CrossRef]
  66. Zheng, X.; Dong, L.; Zhao, S.; Li, Q.; Liu, D.; Zhu, X.; Ge, X.; Li, R.; Wang, G. Propofol Affects Non–Small-Cell Lung Cancer Cell Biology by Regulating the miR-21/PTEN/AKT Pathway In Vitro and In Vivo. Obstet. Anesth. Dig. 2020, 131, 1270–1280. [Google Scholar] [CrossRef]
  67. Wang, H.; Jiao, H.; Jiang, Z.; Chen, R. Propofol inhibits migration and induces apoptosis of pancreatic cancer PANC-1 cells through miR-34a-mediated E-cadherin and LOC285194 signals. Bioengineered 2020, 11, 510–521. [Google Scholar] [CrossRef] [Green Version]
  68. Sun, H.; Gao, D. Propofol suppresses growth, migration and invasion of A549 cells by down-regulation of miR-372. BMC Cancer 2018, 18, 1–11. [Google Scholar] [CrossRef] [Green Version]
  69. 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. 2016, 37, 587–593. [Google Scholar] [CrossRef] [Green Version]
  70. Su, Z.; Hou, X.K.; Wen, Q.P. Propofol induces apoptosis of epithelial ovarian cancer cells by upregulation of microRNA let-7i expression. Eur. J. Gynaecol. Oncol. 2014, 35, 688–691. [Google Scholar]
  71. Lu, H.; Zheng, G.; Gao, X.; Chen, C.; Zhou, M.; Zhang, L. Propofol suppresses cell viability, cell cycle progression and motility and induces cell apoptosis of ovarian cancer cells through suppressing MEK/ERK signaling via targeting circVPS13C/miR-145 axis. J. Ovarian Res. 2021, 14, 1–11. [Google Scholar] [CrossRef]
  72. Gao, J.; Ding, C.; Zhou, J.; Wu, G.; Han, Z.; Li, J.; Hei, F. Propofol suppresses lung cancer tumorigenesis by modulating thecirc-ERBB2/miR-7-5p/FOXM1axis. Thorac. Cancer 2021, 12, 824–834. [Google Scholar] [CrossRef]
  73. Zhao, H.; Wei, H.; He, J.; Wang, D.; Li, W.; Wang, Y.; Ai, Y.; Yang, J. Propofol disrupts cell carcinogenesis and aerobic glycolysis by regulating circTADA2A/miR-455-3p/FOXM1 axis in lung cancer. Cell Cycle 2020, 19, 2538–2552. [Google Scholar] [CrossRef]
  74. Garib, V.; Lang, K.; Niggemann, B.; Zänker, K.S.; Brandt, L.; Dittmar, T. Propofol-induced calcium signalling and actin reorganization within breast carcinoma cells. Eur. J. Anaesthesiol. 2005, 22, 609–615. [Google Scholar] [CrossRef]
  75. Song, F.; Liu, J.; Feng, Y.; Jin, Y. Propofol-induced HOXA11-AS promotes proliferation, migration and invasion, but inhibits apoptosis in hepatocellular carcinoma cells by targeting miR-4458. Int. J. Mol. Med. 2020, 46, 1135–1145. [Google Scholar] [CrossRef]
  76. Chen, X.; Wu, Q.; You, L.; Chen, S.; Zhu, M.; Miao, C. Propofol attenuates pancreatic cancer malignant potential via inhibition of NMDA receptor. Eur. J. Pharmacol. 2017, 795, 150–159. [Google Scholar] [CrossRef]
  77. Oh, T.K.; Kim, H.; Jeon, Y. Retrospective analysis of 1-year mortality after gastric cancer surgery: Total intravenous anesthesia versus volatile anesthesia. Acta Anaesthesiol. Scand. 2019, 63, 1169–1177. [Google Scholar] [CrossRef]
  78. 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. Anesthesiology 2019, 130, 31–40. [Google Scholar] [CrossRef]
  79. Makito, K.; Matsui, H.; Fushimi, K.; Yasunaga, H. Volatile versus Total Intravenous Anesthesia for Cancer Prognosis in Patients Having Digestive Cancer Surgery. Anesthesiology 2020, 133, 764–773. [Google Scholar] [CrossRef]
  80. Lai, H.-C.; Lee, M.-S.; Lin, C.; Lin, K.-T.; Huang, Y.-H.; Wong, C.-S.; Chan, S.-M.; Wu, Z.-F. Propofol-based total intravenous anaesthesia is associated with better survival than desflurane anaesthesia in hepatectomy for hepatocellular carcinoma: A retrospective cohort study. Br. J. Anaesth. 2019, 123, 151–160. [Google Scholar] [CrossRef]
  81. Wigmore, T.J.; Mohammed, K.; Jhanji, S. Long-term Survival for Patients Undergoing Volatile versus IV Anesthesia for Cancer Surgery. Anesthesiology 2016, 124, 69–79. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, Z.; Gong, H.; Zheng, F.; Liu, D.; Dong, T. Propofol suppresses proliferation and invasion of pancreatic cancer cells by upregulating microRNA-133a expression. Genet. Mol. Res. 2015, 14, 7529–7537. [Google Scholar] [CrossRef] [PubMed]
  83. Takabuchi, S.; Hirota, K.; Nishi, K.; Oda, S.; Oda, T.; Shingu, K.; Takabayashi, A.; Adachi, T.; Semenza, G.L.; Fukuda, K. The intravenous anesthetic propofol inhibits hypoxia-inducible factor 1 activity in an oxygen tension-dependent manner. FEBS Lett. 2004, 577, 434–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Zhang, L.; Chen, H.; Tian, C.; Zheng, D. Propofol Represses Cell Growth and Metastasis by Modulating the Circular RNA Non-SMC Condensin I Complex Subunit G/MicroRNA-200a-3p/RAB5A Axis in Glioma. World Neurosurg. 2021, 153, e46–e58. [Google Scholar] [CrossRef] [PubMed]
  85. Nguyen, J.; Luk, K.; Vang, D.; Soto, W.; Vincent, L.; Robiner, S.; Saavedra, R.; Li, Y.; Gupta, P.; Gupta, K. Morphine stimulates cancer progression and mast cell activation and impairs survival in transgenic mice with breast cancer. Br. J. Anaesth. 2014, 113, i4–i13. [Google Scholar] [CrossRef] [Green Version]
  86. Doornebal, C.W.; Vrijland, K.; Hau, C.-S.; Coffelt, S.B.; Ciampricotti, M.; Jonkers, J.; de Visser, K.E.; Hollmann, M.W. Morphine does not facilitate breast cancer progression in two preclinical mouse models for human invasive lobular and HER2+ breast cancer. Pain 2015, 156, 1424–1432. [Google Scholar] [CrossRef]
  87. Gach, K.; Szemraj, J.; Fichna, J.; Piestrzeniewicz, M.; Delbro, D.S.; Janecka, A. The Influence of Opioids on Urokinase Plasminogen Activator on Protein and mRNA Level in MCF-7 Breast Cancer Cell Line. Chem. Biol. Drug Des. 2009, 74, 390–396. [Google Scholar] [CrossRef]
  88. Afsharimani, B.; Baran, J.; Watanabe, S.; Lindner, D.; Cabot, P.J.; Parat, M.-O. Morphine and breast tumor metastasis: The role of matrix-degrading enzymes. Clin. Exp. Metastasis 2013, 31, 149–158. [Google Scholar] [CrossRef]
  89. Tegeder, I.; Grösch, S.; Schmidtko, A.; Häussler, A.; Schmidt, H.; Niederberger, E.; Scholich, K.; Geisslinger, G. G protein-independent G1 cell cycle block and apoptosis with morphine in ade-nocarcinoma cells: Involvement of p53 phosphorylation. Cancer Res. 2003, 63, 1846–1852. [Google Scholar]
  90. Hatsukari, I.; Hitosugi, N.; Ohno, R.; Hashimoto, K.; Nakamura, S.; Satoh, K.; Nagasaka, H.; Matsumoto, I.; Sakagami, H. Induction of apoptosis by morphine in human tumor cell lines in vitro. Anticancer Res. 2007, 27, 857–864. [Google Scholar]
  91. Gupta, K.; Kshirsagar, S.; Chang, L.; Schwartz, R.; Law, P.-Y.; Yee, D.; Hebbel, R.P. Morphine stimulates angiogenesis by activating proangiogenic and surviv-al-promoting signaling and promotes breast tumor growth. Cancer Res. 2002, 62, 4491–4498. [Google Scholar]
  92. 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] [Green Version]
  93. Mathew, B.; Lennon, F.E.; Siegler, J.; Mirzapoiazova, T.; Mambetsariev, N.; Sammani, S.; Gerhold, L.M.; LaRiviere, P.J.; Chen, C.-T.; Garcia, J.G.N.; et al. The Novel Role of the Mu Opioid Receptor in Lung Cancer Progression. Obstet. Anesth. Dig. 2011, 112, 558–567. [Google Scholar] [CrossRef] [Green Version]
  94. Lennon, F.E.; Mirzapoiazova, T.; Mambetsariev, B.; Poroyko, V.A.; Salgia, R.; Moss, J.; Singleton, P.A. The Mu Opioid Receptor Promotes Opioid and Growth Factor-Induced Proliferation, Migration and Epithelial Mesenchymal Transition (EMT) in Human Lung Cancer. PLoS ONE 2014, 9, e91577. [Google Scholar] [CrossRef]
  95. Koodie, L.; Yuan, H.; Pumper, J.A.; Yu, H.; Charboneau, R.; Ramkrishnan, S.; Roy, S. Morphine Inhibits Migration of Tumor-Infiltrating Leukocytes and Suppresses Angiogenesis Associated with Tumor Growth in Mice. Am. J. Pathol. 2014, 184, 1073–1084. [Google Scholar] [CrossRef] [Green Version]
  96. Nylund, G.; Pettersson, A.; Bengtsson, C.; Khorram-Manesh, A.; Nordgren, S.; Delbro, D.S. Functional Expression of μ-Opioid Receptors in the Human Colon Cancer Cell Line, HT-29, and their Localization in Human Colon. Dig. Dis. Sci. 2007, 53, 461–466. [Google Scholar] [CrossRef]
  97. Harimaya, Y.; Koizumi, K.; Andoh, T.; Nojima, H.; Kuraishi, Y.; Saiki, I. Potential ability of morphine to inhibit the adhesion, invasion and metastasis of metastatic colon 26-L5 carcinoma cells. Cancer Lett. 2002, 187, 121–127. [Google Scholar] [CrossRef]
  98. Buckley, A.; McQuaid, S.; Johnson, P.; Buggy, D.J. Effect of anaesthetic technique on the natural killer cell anti-tumour activity of serum from women undergoing breast cancer surgery: A pilot study. Br. J. Anaesth. 2014, 113, i56–i62. [Google Scholar] [CrossRef] [Green Version]
  99. Desmond, F.; McCormack, J.; Mulligan, N.; Stokes, M.; Buggy, D.J. Effect of anaesthetic technique on immune cell infiltration in breast cancer: A follow-up pilot analysis of a prospective, randomised, investigator-masked study. Anticancer. Res. 2015, 35, 1311–1319. [Google Scholar]
  100. Xu, Y.J.; Li, S.Y.; Cheng, Q.; Chen, W.K.; Wang, S.L.; Ren, Y.; Miao, C. Effects of anaesthesia on proliferation, invasion and apoptosis of LoVo colon cancer cells in vitro. Anaesthesia 2015, 71, 147–154. [Google Scholar] [CrossRef]
  101. Zhang, H.; Sun, M.; Zhou, D.; Gorur, A.; Sun, Z.; Zeng, W.; Cata, J.P.; Chen, W.; Miao, C. Increased mu-opioid receptor expression is associated with reduced disease-free and overall survival in laryngeal squamous cell carcinoma. Br. J. Anaesth. 2020, 125, 722–729. [Google Scholar] [CrossRef] [PubMed]
  102. Zylla, D.; Gourley, B.L.; Vang, D.; Jackson, S.; Boatman, S.; Ms, B.L.; Kuskowski, M.A.; Le, C.; Gupta, K.; Gupta, P. Opioid requirement, opioid receptor expression, and clinical outcomes in patients with advanced prostate cancer. Cancer 2013, 119, 4103–4110. [Google Scholar] [CrossRef] [PubMed]
  103. Friesen, C.; Roscher, M.; Hormann, I.; Fichtner, I.; Alt, A.; Hilger, R.A.; Debatin, K.-M.; Miltner, E. Cell death sensitization of leukemia cells by opioid receptor activation. Oncotarget 2013, 4, 677–690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Cata, J.P.; Keerty, V.; Keerty, D.; Feng, L.; Norman, P.H.; Gottumukkala, V.; Mehran, J.R.; Engle, M. A retrospective analysis of the effect of intraoperative opioid dose on cancer recurrence after non-small cell lung cancer resection. Cancer Med. 2014, 3, 900–908. [Google Scholar] [CrossRef] [PubMed]
  105. Tai, Y.-H.; Wu, H.-L.; Chang, W.-K.; Tsou, M.-Y.; Chen, H.-H.; Chang, K.-Y. Intraoperative Fentanyl Consumption Does Not Impact Cancer Recurrence or Overall Survival after Curative Colorectal Cancer Resection. Sci. Rep. 2017, 7, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Oh, T.K.; Jeon, J.H.; Lee, J.M.; Kim, M.S.; Kim, J.H.; Cho, H.; Kim, S.-E.; Eom, W. Investigation of opioid use and long-term oncologic outcomes for non-small cell lung cancer patients treated with surgery. PLoS ONE 2017, 12, e0181672. [Google Scholar] [CrossRef] [Green Version]
  107. Friesen, C.; Hormann, I.; Roscher, M.; Fichtner, I.; Alt, A.; Hilger, R.; Debatin, K.-M.; Miltner, E. Opioid receptor activation triggering downregulation of cAMP improves effectiveness of anti-cancer drugs in treatment of glioblastoma. Cell Cycle 2014, 13, 1560–1570. [Google Scholar] [CrossRef] [Green Version]
  108. Wang, H.-W.; Wang, L.-Y.; Jiang, L.; Tian, S.-M.; Zhong, T.-D.; Fang, X.-M. Amide-linked local anesthetics induce apoptosis in human non-small cell lung cancer. J. Thorac. Dis. 2016, 8, 2748–2757. [Google Scholar] [CrossRef] [Green Version]
  109. Bundscherer, A.C.; Malsy, M.; Bitzinger, D.I.; Wiese, C.H.; Gruber, M.A.; Graf, B.M. Effects of Lidocaine on HT-29 and SW480 Colon Cancer Cells In Vitro. Anticancer Res. 2017, 37, 1941–1945. [Google Scholar] [CrossRef]
  110. Yang, W.; Cai, J.; Zhang, H.; Wang, G.; Jiang, W. Effects of Lidocaine and Ropivacaine on Gastric Cancer Cells through Down-regulation of ERK1/2 Phosphorylation In Vitro. Anticancer Res. 2018, 38, 6729–6735. [Google Scholar] [CrossRef]
  111. Chen, J.; Jiao, Z.; Wang, A.; Zhong, W. Lidocaine inhibits melanoma cell proliferation by regulating ERK phosphorylation. J. Cell. Biochem. 2018, 120, 6402–6408. [Google Scholar] [CrossRef]
  112. Xuan, W.; Zhao, H.; Hankin, J.; Chen, L.; Yao, S.; Ma, D. Local anesthetic bupivacaine induced ovarian and prostate cancer apoptotic cell death and underlying mechanisms in vitro. Sci. Rep. 2016, 6, 26277. [Google Scholar] [CrossRef] [Green Version]
  113. Chang, Y.-C.; Hsu, Y.-C.; Liu, C.-L.; Huang, S.-Y.; Hu, M.-C.; Cheng, S.-P. Local Anesthetics Induce Apoptosis in Human Thyroid Cancer Cells through the Mitogen-Activated Protein Kinase Pathway. PLoS ONE 2014, 9, e89563. [Google Scholar] [CrossRef] [Green Version]
  114. Xing, W.; Chen, D.-T.; Pan, J.-H.; Chen, Y.-H.; Yan, Y.; Li, Q.; Xue, R.-F.; Yuan, Y.-F.; Zeng, W.-A. Lidocaine Induces Apoptosis and Suppresses Tumor Growth in Human Hepatocellular Carcinoma Cells In Vitro and in a Xenograft Model In Vivo. Anesthesiology 2017, 126, 868–881. [Google Scholar] [CrossRef]
  115. Ye, L.; Zhang, Y.; Chen, Y.J.; Liu, Q. Anti-tumor effects of lidocaine on human gastric cancer cells in vitro. Bratisl. Med. J. 2019, 120, 212–217. [Google Scholar] [CrossRef]
  116. Bezu, L.; Chuang, A.W.; Sauvat, A.; Humeau, J.; Xie, W.; Cerrato, G.; Liu, P.; Zhao, L.; Zhang, S.; Le Naour, J.; et al. Local anesthetics elicit immune-dependent anticancer effects. J. Immunother. Cancer 2022, 10, e004151. [Google Scholar] [CrossRef]
  117. Piegeler, T.; Schläpfer, M.; Dull, R.O.; Schwartz, D.E.; Borgeat, A.; Minshall, R.D.; Beck-Schimmer, B. Clinically relevant concentrations of lidocaine and ropivacaine inhibit TNFα-induced invasion of lung adenocarcinoma cells in vitro by blocking the activation of Akt and focal adhesion kinase. Br. J. Anaesth. 2015, 115, 784–791. [Google Scholar] [CrossRef] [Green Version]
  118. Piegeler, M.T.; Votta-Velis, M.E.G.; Liu, G.; Place, A.T.; Schwartz, M.D.E.; Beck-Schimmer, M.B.; Minshall, R.D.; Borgeat, M.A. Antimetastatic Potential of Amide-linked Local Anesthetics. Anesthesiology 2012, 117, 548–559. [Google Scholar] [CrossRef] [Green Version]
  119. 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]
  120. 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]
  121. Sun, H.; Sun, Y. Lidocaine inhibits proliferation and metastasis of lung cancer cell via regulation of miR-539/EGFR axis. Artif. Cells Nanomed. Biotechnol. 2019, 47, 2866–2874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Chamaraux-Tran, T.-N.; Mathelin, C.; Aprahamian, M.; Joshi, G.P.; Tomasetto, C.; Diemunsch, P.; Akladios, C. Antitumor Effects of Lidocaine on Human Breast Cancer Cells: An In Vitro and In Vivo Experimental Trial. Anticancer Res. 2018, 38, 95–105. [Google Scholar] [CrossRef] [PubMed]
  123. Lirk, P.; Berger, R.; Hollmann, M.; Fiegl, H. Lidocaine time- and dose-dependently demethylates deoxyribonucleic acid in breast cancer cell lines in vitro. Br. J. Anaesth. 2012, 109, 200–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Li, R.; Xiao, C.; Liu, H.; Huang, Y.; Dilger, J.P.; Lin, J. Effects of local anesthetics on breast cancer cell viability and migration. BMC Cancer 2018, 18, 666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Le Gac, G.; Angenard, G.; Clément, B.; Laviolle, B.; Coulouarn, C.; Beloeil, H. Local Anesthetics Inhibit the Growth of Human Hepatocellular Carcinoma Cells. Anesth. Analg. 2017, 125, 1600–1609. [Google Scholar] [CrossRef]
  126. Zhao, L.; Ma, N.; Liu, G.; Mao, N.; Chen, F.; Li, J. Lidocaine Inhibits Hepatocellular Carcinoma Development by Modulating circ_ITCH/miR-421/CPEB3 Axis. Dig. Dis. Sci. 2021, 66, 4384–4397. [Google Scholar] [CrossRef]
  127. Liu, H.; Wang, Y.; Chen, B.; Shen, X.; Li, W. Effects of Lidocaine-Mediated CPEB3 Upregulation in Human Hepatocellular Carcinoma Cell Proliferation In Vitro. BioMed Res. Int. 2018, 2018, 1–7. [Google Scholar] [CrossRef]
  128. Zhang, H.; Yang, L.; Zhu, X.; Zhu, M.; Sun, Z.; Cata, J.P.; Chen, W.; Miao, C. Association between intraoperative intravenous lidocaine infusion and survival in patients undergoing pancreatectomy for pancreatic cancer: A retrospective study. Br. J. Anaesth. 2020, 125, 141–148. [Google Scholar] [CrossRef]
  129. Freeman, J.; Crowley, P.D.; Foley, A.G.; Gallagher, H.C.; Iwasaki, M.; Ma, D.; Buggy, D.J. Effect of Perioperative Lidocaine and Cisplatin on Metastasis in a Murine Model of Breast Cancer Surgery. Anticancer Res. 2018, 38, 5599–5606. [Google Scholar] [CrossRef]
  130. Johnson, M.; Crowley, P.; Foley, A.; Xue, C.; Connolly, C.; Gallagher, H.; Buggy, D. Effect of perioperative lidocaine on metastasis after sevoflurane or ketamine-xylazine anaesthesia for breast tumour resection in a murine model. Br. J. Anaesth. 2018, 121, 76–85. [Google Scholar] [CrossRef]
  131. Sakaguchi, M.; Kuroda, Y.; Hirose, M. The Antiproliferative Effect of Lidocaine on Human Tongue Cancer Cells with Inhibition of the Activity of Epidermal Growth Factor Receptor. Anesth. Analg. 2006, 102, 1103–1107. [Google Scholar] [CrossRef]
  132. Casey, S.C.; Amedei, A.; Aquilano, K.; Azmi, A.S.; Benencia, F.; Bhakta, D.; Bilsland, A.E.; Boosani, C.S.; Chen, S.; Ciriolo, M.R.; et al. Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin. Cancer Biol. 2015, 35, S199–S223. [Google Scholar] [CrossRef]
  133. Pantel, K.; Speicher, M. The biology of circulating tumor cells. Oncogene 2015, 35, 1216–1224. [Google Scholar] [CrossRef]
  134. Gao, H.; Chakraborty, G.; Lee-Lim, A.P.; Mo, Q.; Decker, M.; Vonica, A.; Shen, R.; Brogi, E.; Brivanlou, A.H.; Giancotti, F.G. The BMP Inhibitor Coco Reactivates Breast Cancer Cells at Lung Metastatic Sites. Cell 2012, 150, 764–779. [Google Scholar] [CrossRef] [Green Version]
  135. Lu, X.; Mu, E.; Wei, Y.; Riethdorf, S.; Yang, Q.; Yuan, M.; Yan, J.; Hua, Y.; Tiede, B.J.; Lu, X.; et al. VCAM-1 Promotes Osteolytic Expansion of Indolent Bone Micrometastasis of Breast Cancer by Engaging α4β1-Positive Osteoclast Progenitors. Cancer Cell 2011, 20, 701–714. [Google Scholar] [CrossRef] [Green Version]
  136. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [Green Version]
  137. Pastushenko, I.; Brisebarre, A.; Sifrim, A.; Fioramonti, M.; Revenco, T.; Boumahdi, S.; Van Keymeulen, A.; Brown, D.; Moers, V.; Lemaire, S.; et al. Identification of the tumour transition states occurring during EMT. Nature 2018, 556, 463–468. [Google Scholar] [CrossRef]
  138. Tabassum, D.P.; Polyak, K. Tumorigenesis: It takes a village. Nat. Rev. Cancer 2015, 15, 473–483. [Google Scholar] [CrossRef]
  139. Gundem, G.; Van Loo, P.; Kremeyer, B.; Alexandrov, L.B.; Tubio, J.M.; Papaemmanuil, E.; Brewer, D.S.; Kallio, H.M.; Högnäs, G.; Annala, M. The evolutionary history of lethal metastatic prostate cancer. Nature 2015, 520, 353–357. [Google Scholar] [CrossRef] [Green Version]
  140. Zappalà, G.; McDonald, P.G.; Cole, S.W. Tumor dormancy and the neuroendocrine system: An undisclosed connection? Cancer Metastasis Rev. 2012, 32, 189–200. [Google Scholar] [CrossRef]
  141. Sood, A.K.; Bhatty, R.; Kamat, A.A.; Landen, C.N.; Han, L.; Thaker, P.H.; Li, Y.; Gershenson, D.M.; Lutgendorf, S.; Cole, S.W. Stress Hormone–Mediated Invasion of Ovarian Cancer Cells. Clin. Cancer Res. 2006, 12, 369–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Wong, H.P.; Ho, J.W.; Koo, M.W.; Yu, L.; Wu, W.K.; Lam, E.K.; Tai, E.K.; Ko, J.K.; Shin, V.Y.; Chu, K.M.; et al. Effects of adrenaline in human colon adenocarcinoma HT-29 cells. Life Sci. 2011, 88, 1108–1112. [Google Scholar] [CrossRef] [PubMed]
  143. Bernabé, D.G.; Tamae, A.C.; Biasoli, .R.; Oliveira, S.H. Stress hormones increase cell proliferation and regulates interleukin-6 secretion in human oral squamous cell carcinoma cells. Brain Behav. Immun. 2011, 25, 574–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Yang, E.V.; Kim, S.; Donovan, E.L.; Chen, M.; Gross, A.C.; Webster Marketon, J.I.; Barsky, S.H.; Glaser, R. Norepinephrine upregulates VEGF, IL-8, and IL-6 expression in human melanoma tumor cell lines: Implications for stress-related enhancement of tumor progression. Brain Behav. Immun. 2009, 23, 267–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Gottschalk, A.; Sharma, S.; Ford, J.; Durieux, M.E.; Tiouririne, M. The Role of the Perioperative Period in Recurrence after Cancer Surgery. Obstet. Anesth. Dig. 2010, 110, 1636–1643. [Google Scholar] [CrossRef]
  146. Lutgendorf, S.K.; Cole, S.; Costanzo, E.; Bradley, S.; Coffin, J.; Jabbari, S.; Rainwater, K.; Ritchie, J.M.; Yang, M.; Sood, A.K. Stress-related mediators stimulate vascular endothelial growth factor secretion by two ovarian cancer cell lines. Clin. Cancer Res. 2003, 9, 4514–4521. [Google Scholar]
  147. Aguilar-Cazares, D.; Chavez-Dominguez, R.; Carlos-Reyes, A.; Lopez-Camarillo, C.; de la Cruz, O.N.H.; Lopez-Gonzalez, J.S. Contribution of Angiogenesis to Inflammation and Cancer. Front. Oncol. 2019, 9, 1399. [Google Scholar] [CrossRef] [Green Version]
  148. Goldfarb, Y.; Ben-Eliyahu, S. Surgery as a Risk Factor for Breast Cancer Recurrence and Metastasis: Mediating Mechanisms and Clinical Prophylactic Approaches. Breast Dis. 2007, 26, 99–114. [Google Scholar] [CrossRef]
  149. Sacerdote, P.; Manfredi, B.; Bianchi, M.; Panerai, A. Intermittent but Not Continuous Inescapable Footshock Stress Affects Immune Responses and Immunocyte Beta-Endorphin Concentrations in the Rat. Brain Behav. Immun. 1994, 8, 251–260. [Google Scholar] [CrossRef] [Green Version]
  150. Shavit, Y.; Martin, F.; Yirmiya, R.; Ben-Eliyahu, S.; Terman, G.; Weiner, H.; Gale, R.; Liebeskind, J. Effects of a single administration of morphine or footshock stress on natural killer cell cytotoxicity. Brain Behav. Immun. 1987, 1, 318–328. [Google Scholar] [CrossRef]
  151. Bryant, H.U.; Roudebush, R.E. Suppressive effects of morphine pellet implants on in vivo parameters of immune function. J. Pharmacol. Exp. Ther. 1990, 255. [Google Scholar]
  152. Jaeger, K.; Scheinichen, D.; Heine, J.; André, M.; Bund, M.; Piepenbrock, S.; Leuwer, M. Remifentanil, fentanyl, and alfentanil have no influence on the respiratory burst of human neutrophilsin vitro. Acta Anaesthesiol. Scand. 1998, 42, 1110–1113. [Google Scholar] [CrossRef]
  153. Krumholz, W.; Endrass, J.; Hempelmann, G. Inhibition of phagocytosis and killing of bacteria by anaesthetic agents in vitro. Br. J. Anaesth. 1995, 75, 66–70. [Google Scholar] [CrossRef] [Green Version]
  154. Lysle, D.T.; Coussons, M.E.; Watts, V.J.; Bennett, E.H.; Dykstra, L.A. Morphine-induced alterations of immune status: Dose dependency, compartment specificity and antagonism by naltrexone. J. Pharmacol. Exp. Ther. 1993, 265, 1071–1078. [Google Scholar]
  155. Welters, I.; Menzebach, A.; Goumon, Y.; Langefeld, T.; Teschemacher, H.; Hempelmann, G.; Stefano, G. Morphine suppresses complement receptor expression, phagocytosis, and respiratory burst in neutrophils by a nitric oxide and μ3 opiate receptor-dependent mechanism. J. Neuroimmunol. 2000, 111, 139–145. [Google Scholar] [CrossRef]
  156. Cui, J.-H.; Jiang, W.-W.; Liao, Y.-J.; Wang, Q.-H.; Xu, M.; Li, Y. Effects of oxycodone on immune function in patients undergoing radical resection of rectal cancer under general anesthesia. Medicine 2017, 96, e7519. [Google Scholar] [CrossRef]
  157. Chen, Y.; Liang, M.; Zhu, Y.; Zhou, D. The effect of propofol and sevoflurane on the perioperative immunity in patients un-der laparoscopic radical resection of colorectal cancer. Zhonghua Yi Xue Za Zhi 2015, 95, 3440–3444. [Google Scholar]
  158. Liu, S.; Gu, X.; Zhu, L.; Wu, G.; Zhou, H.; Song, Y.; Wu, C. Effects of propofol and sevoflurane on perioperative immune response in patients undergoing laparoscopic radical hysterectomy for cervical cancer. Medicine 2016, 95, e5479. [Google Scholar] [CrossRef]
  159. Eochagáin, A.N.; Burns, D.; Riedel, B.; Sessler, D.I.; Buggy, D.J. The effect of anaesthetic technique during primary breast cancer surgery on neutrophil-lymphocyte ratio, platelet-lymphocyte ratio and return to intended oncological therapy. Anaesthesia 2018, 73, 603–611. [Google Scholar] [CrossRef] [Green Version]
  160. Oh, C.-S.; Lee, J.; Yoon, T.-G.; Seo, E.-H.; Park, H.-J.; Piao, L.; Lee, S.-H.; Kim, S.-H. Effect of Equipotent Doses of Propofol versus Sevoflurane Anesthesia on Regulatory T Cells after Breast Cancer Surgery. Anesthesiology 2018, 129, 921–931. [Google Scholar] [CrossRef]
  161. 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] [PubMed]
  162. Menezes, M.; Das, S.; Minn, I.; Emdad, L.; Wang, X.-Y.; Sarkar, D.; Pomper, M.; Fisher, P. Detecting Tumor Metastases. Adv. Cancer Res. 2016, 132, 1–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Hong, B.; Zu, Y. Detecting Circulating Tumor Cells: Current Challenges and New Trends. Theranostics 2013, 3, 377–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Zhang, D.; Zhou, X.-H.; Zhang, J.; Zhou, Y.-X.; Ying, J.; Wu, G.-Q.; Qian, J.-H. Propofol promotes cell apoptosis via inhibiting HOTAIR mediated mTOR pathway in cervical cancer. Biochem. Biophys. Res. Commun. 2015, 468, 561–567. [Google Scholar] [CrossRef]
  165. Melamed, R.; Bar-Yosef, S.; Shakhar, G.; Shakhar, K.; Ben-Eliyahu, S. Suppression of Natural Killer Cell Activity and Promotion of Tumor Metastasis by Ketamine, Thiopental, and Halothane, but Not by Propofol: Mediating Mechanisms and Prophylactic Measures. Obstet. Anesth. Dig. 2003, 97, 1331–1339. [Google Scholar] [CrossRef]
Table 1. Studies describing the direct effects of midazolam, dexmedetomidine, and ketamine on cancer cells, the associated mechanisms of action and their respective pathways 1.
Table 1. Studies describing the direct effects of midazolam, dexmedetomidine, and ketamine on cancer cells, the associated mechanisms of action and their respective pathways 1.
Midazolam
StudyType of
Cancer
Effect on CancerMechanism of ActionPathway Described
Wang C. et al. [6]Glioma, Lung
  • Anti-tumourigenic properties in very high doses. No effect in low (physiological) concentrations.
  • Significant reduction in tumour size compared with tumours from naïve animals
Peripheral Benzodiazepine Receptor (PBR) on mitochondria resulting in reduction tumour burden,
Ki67 expression and cyclin D expression
Intrinsic apoptotic pathway (exact mechanism unclear)
Qi Y. et al. [7]Hepato-cellular
  • Inhibition of invasion and migration
  • Repression of tumour growth
Overexpression of miR-124-3p and subsequent inhibition of PIM-1 resulting in cell cycle arrest and increased apoptosismiR-124-3p/PIM- axis
Mishra SK. et al. [8]Colon,
leukemia
  • Growth inhibition of cancer cells
  • Inhibited HT29 tumour growth in xenografts mice
Activation of caspase-9, capspase-3 and PARP indicating induction of the mitochondrial intrinsic pathway of apoptosisInhibition of pERK1/2 signalling leading to inhibition of the anti-apoptotic proteins Bcl-XL and XIAP and phosphorylation activation of the pro-apoptotic protein Bid
Dexmedetomidine
StudyType of
cancer
Effect on CancerMechanism of ActionPathway Described
Wang C. et al. [6]Glioma, Lung
  • Promotes cancer cell survival in vitro
  • No significant effect in vivo
Increased Ki67 and cyclin D expression leading to cell proliferationVia α2–adrenergic signalling and upregulation of antiapoptotic proteins Bcl-2 and Bcl-xL
Zhang P. et al. [9]EsophagusSuppressed tumour growth and metastasisIncreased apoptosis of esophageal cancer cells in vivo and in vitroUpregulation of miR-143-3p and reducing the level of EPS8
Xia M. et al. [10]BreastIncreased proliferation, migration and
invasion ability of MDA-MB-231 cells in a
dose-dependent manner in vitro
  • Up-regulation of the protein level of α2-adrenoceptor
  • Enhancement of phosphorylation of ERK without changing the total level of it
ERK Pathway.
Ketamine
StudyType of
Cancer
Effect on CancerMechanism of ActionPathway Described
He H. et al. [11]BreastDecreased apoptosisUpregulation of Bcl-2 expressionNon described
1 The correlated hallmark of cancer is given for reference on the role and importance in cancer development.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Debel, W.; Ramadhan, A.; Vanpeteghem, C.; Forsyth, R.G. Does the Choice of Anaesthesia Affect Cancer? A Molecular Crosstalk between Theory and Practice. Cancers 2023, 15, 209. https://doi.org/10.3390/cancers15010209

AMA Style

Debel W, Ramadhan A, Vanpeteghem C, Forsyth RG. Does the Choice of Anaesthesia Affect Cancer? A Molecular Crosstalk between Theory and Practice. Cancers. 2023; 15(1):209. https://doi.org/10.3390/cancers15010209

Chicago/Turabian Style

Debel, Wiebrecht, Ali Ramadhan, Caroline Vanpeteghem, and Ramses G. Forsyth. 2023. "Does the Choice of Anaesthesia Affect Cancer? A Molecular Crosstalk between Theory and Practice" Cancers 15, no. 1: 209. https://doi.org/10.3390/cancers15010209

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