ecancermedicalscience

Review

Anaesthesia and cancer: can anaesthetic drugs modify gene expression?

Aida Raigon Ponferrada1,2, Jose Luis Guerrero Orriach1,2,3,4, Alfredo Malo Manso2, Enrique Sepúlveda Haro2, Salvador Romero Molina2, Ana Fontaneda Heredia2, Manolo Baena Lopez2 and Jose Cruz Mañas2

1Institute of Biomedical Research in Malaga [IBIMA], Malaga 29010, Spain

2Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga 29010, Spain

3Department of Pharmacology and Pediatrics, School of Medicine, University of Malaga, Malaga 29010, Spain

4Member of COST Action 15204

5Department of Urology. Virgen de la Victoria University Hospital, Malaga 29010, Spain

6Department of Anaesthesiology, Complejo Hospitalario de Jaen, 29010, Spain

7Department of Oncology, Virgen de la Victoria University Hospital, Malaga 29010, Spain


Abstract

Cancer remains a primary cause of morbidity and mortality worldwide, and its incidence continues to increase. The most common cause of death in cancer patients is tumour recurrence. Surgery is the gold standard in the treatment of most tumours. However, cancer surgery can lead to the release of tumour cells into the systemic circulation. Surgical stress and several perioperative factors have been suggested to boost tumour growth, thereby increasing the risk of metastatic recurrence.

Preclinical and clinical studies suggest that anaesthetics and adjuvants administered during the perioperative period may impact cancer recurrence and survival. This document summarises the current evidence regarding the effects of anaesthetic drugs and analgesic techniques on the immune system, systemic inflammatory response and tumour cells, as well as their impact on cancer recurrence.

Keywords: anaesthesia, analgesia, cancer recurrence, metastases, anaesthetics, regional anaesthesia, opioid, NSAIDs

Correspondence to: Jose Luis Guerrero Orriach and Aida Raigon Ponferrada
Email: guerreroorriach@gmail.com and aidaraigonp@gmail.com

Published: 27/07/2020
Received: 14/01/2020

Publication costs for this article were supported by ecancer (UK Charity number 1176307).

Copyright: © the authors; licensee ecancermedicalscience. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.


Introduction

Cancer is the second cause of mortality in developed countries, with most deaths caused by metastasis [1]. Surgery is one of the factors involved in metastatic spread since it may facilitate the release of cancer cells into the bloodstream during tumour manipulation. Another factor related to cancer spread is the systemic inflammatory response associated with perioperative surgical stress. Consequently, surgery implies two factors related to metastatic progression, i.e., the presence of cancer cells with metastatic potential and an appropriate microenvironment for their growth. Among that can affect metastatic progression of tumour cells is the anaesthetic technique [2].

The anaesthetic technique can influence patients’ neuroendocrine and immune responses during surgery. Surgical stress can suppress the antitumour immune response and stimulate the hypothalamic-pituitary-adrenal (HPA) axis, which, along with the activation of the sympathetic nervous system (SNS), also regulates the immune response. The activation of the HPA and the SNS leads to the suppression of cell immunity (CI) as well as the release of catecholamines and prostaglandin E2 [3]. In turn, these molecules increase immunosuppressant cytokines such as interleukin (IL): IL-4 and IL-10, TGF-β (transforming growth factor-beta) and vascular endothelial growth factor (VEGF); as well as pro-inflammatory cytokines, such as IL-6 and IL-10, which promote tumour angiogenesis and facilitate the development of metastases [4].

Anaesthetic agents vary in their ability to induce immunomodulation and boosting of tumour growth factors. Pre- and postoperative opioids are able to inhibit the humoral immune response and can have pro-angiogenic effects, which promote tumour cell growth [5, 6]. On the other hand, regional anaesthesia preserves CI and reduces the surgery-induced neuroendocrine response by debilitating afferent activations mediated by the HPA axis and SNS neural responses. Regional anaesthetic techniques have been associated with lower rates of cancer recurrence [7, 8].

Despite the fact that some studies have shown significant benefit in terms of cancer recurrence a recent metaanalysis shows that RA has no overall survival, recurrence- free survival or biochemical recurrence-free survival benefit [9].

In this review, we aim to provide more information regarding the role of surgery and anaesthesia in different aspects of tumour recurrence.


Surgical stress

Surgery remains the primary treatment for most cancer patients. However, the surgical stress leads to immune suppression, allows tumour cell adhesion and increases the release of metalloproteases (MMP) and VEGF. These factors increase the mobility and invasiveness of cancer cells and neovascularisation, and promote cancer progression and metastasis formation.

The first line of defence against the development of primary tumours and metastases are natural killer (NK) cells [10]. The changes in the activity of NK cells depend on both the intensity of the surgery and the magnitude of the stress response, which increases the release of catecholamines and prostaglandins by activating the HPA axis and SNS [11]. From a clinical perspective, surgery leads to decreased circulation of NK and T cells via the induction of apoptosis. Surgical stress increases the amount of Th2 lymphocytes and decreases the amount of Th1 lymphocytes, thereby reducing the Th1/Th2 ratio, which eventually leads to CI suppression [12]. Moreover, the levels of immunomodulatory cytokines such as IL-2, IL-12 and interferon-γ (IFN-γ) decrease, whereas anti-inflammatory cytokines such as IL-10 increase [13].


Oxidative stress, inflammation and molecular regulators

Oxidative stress and inflammatory response are among the most important factors that influence the development, growth and metastatic spread of malignant tumours. Oxidative stress can be induced in tumour cells by overproduction of reactive oxygen species (ROS) due to downregulation of NADPH oxidase [14]. It can also be induced by overexpression of thymidine phosphorylase, as observed in most carcinomas [15].

In general, tumours quickly outgrow their blood supply, leading to glucose deprivation and hypoxia. The lack of glucose induces oxidative stress and depletes intracellular pyruvate in cancer cells, thus preventing the breakdown of endogenous oxygen radicals [16]. Oxygen radicals damage DNA, causing filament tears, guanine and thymine alterations and exchanges of sister chromatids. The genetic instability secondary to oxidative stress increases the malignant potential of tumours. Furthermore, oxidative stress can activate several transcription factors, including NF-κB, AP-1, p53 and hypoxia-inducible factor-1 α (HIF-1) [17].

In tumour cells, the expression of HIF promotes cell proliferation and induces the secretion of angiogenic factors, including VEGF and angiopoietin-2. Therefore, hypoxia is strongly associated with tumour progression and metastatic spread [18]. A comprehensive review by Tavare et al [19] described the direct effects of anaesthetics on HIF-1, which is upregulated by inhalational anaesthetics, and inhibited by propofol. Oxidative stress and inflammation are interrelated. Oxidative stress activates inflammatory pathways that transform normal cells into tumour cells, increasing their chances of survival, proliferation, chemo- and radio-resistance, invasiveness and angiogenesis, as compared to stem cells [20].


Effects of anaesthetic agents on immune function and tumour development

Halogenated anaesthetics

Several studies showed that halogenated anaesthetics inhibit the activity of the immune system. Their impact on the activity of NK and T cells is time- and dose-dependent [21]. Volatile anaesthetics also inhibit various lymphocyte functions, including proliferation and cytokine production [22].

Sevoflurane has been demonstrated to increase the levels of pro-tumourigenic cytokines and MMP [23]. Isoflurane reduces the activity of NK cells, induces T cell and B cell apoptosis and reduces the Th1 /Th2 ratio [24].

Unlike total intravenous anaesthesia (TIVA), exposure to sevoflurane increases the levels of proteins such as cytoplasmic HIF-2α and nuclear p38, which are both associated with a worse prognosis in cancer patients [25]. Halothane reduces the activity of NK cells and increases HIF-1α expression [26]. Conversely, propofol prevents the activation of HIF-1α induced by isoflurane, which is related to a partial reduction of the malignant behaviour of cancer cells [27]. Isoflurane is associated with higher levels of HIF-1α and increased proliferation and migration of cancer cells. Sevoflurane induces T cell apoptosis and increases the rate of expression of HIF-1α [28].

Isoflurane increases the malignant potential of cancer cells through the upregulation of insulin-like growth factor (IGF)-1 and its receptor (IGF-1R), as well as VEGF, angiopoietin-1, MMP-2 and MMP-9. Moreover, isoflurane exposure leads to resistance to apoptosis through a caveoline-1-dependent process [23, 29].

Nitrous oxide (N2O) disrupts the synthesis of DNA, purines and thymidylate, which can induce oncogenesis [30]. As demonstrated in an in vivo model, N2O suppresses chemotaxis, which is potentially the strongest stimulator for the development of liver and lung metastases after surgery [31].

Intravenous anaesthetics: propofol, ketamine and thiopental

Intravenous hypnotics have multiple effects on the immune system. Unlike propofol, ketamine and thiopental suppress the activity of NK cells [32]. Ketamine induces apoptosis in lymphocytes via a mitochondrial pathway and inhibits the functional maturation of dendritic cells, while thiopental protects T cells from apoptosis via the induction of thermic shock proteins [33].

On the one hand, ketamine reduces the synthesis of pro-inflammatory cytokines, such as IL-6, and tumour necrosis factor α (TNF-α). On the other hand, thiopental inhibits the function of neutrophils and suppresses the activation of nuclear factor kappa B (NF-κB). This factor is associated with inhibition of the activity of the NF-κB reporter gene, which leads to activation of T cells, secretion of IL-2, IL-6 and IL-8, as well as overexpression of IFN-γ [34].

Apparently, propofol displays a different profile since its protective effects are exerted by other mechanisms, including anti-inflammatory effects, COX-2 inhibition and PGE-2 reduction, increased cytotoxic T lymphocyte activity, and decreased pro-inflammatory cytokines [35]. Propofol does not affect the Th1/Th2 ratio [36], and is weakly bound to β-adrenoreceptor, producing a β-blocking effect that improves anti-tumour immunity and preserves the function of NK cells [37]. Patients receiving perioperative β-blockers have a lower recurrence of metastases after surgery [38].

Propofol conjugates (propofol-docosahexanoate and propofol-eicosapentanoate) have been shown to inhibit cell adhesion and migration, and to induce apoptosis in cancer cells [39]. Propofol reduces cytokine concentrations (IL-1, TNF-α and IL-6), and stimulates the secretion of nitrous oxide in neutrophils [40]. Propofol concentrations of 1–5 mg/mL were found to decrease the invasiveness of cancer cells. Moreover, continuous infusion of propofol can inhibit the development of lung metastases. Wigmore et al [41] retrospectively compared the long-term survival of patients under general anaesthesia using halogenates versus TIVA in cancer surgery. They concluded that the modality of anaesthesia was significantly associated with patient survival, which was higher in the TIVA group. Working under this assumption, Enlund et al [42] observed that 1-year survival was almost 10% higher in cancer surgeries where propofol was used as an anaesthetic.

In contrast with these findings, a recent retrospective study in lung cancer patients found no outcome improvements when comparing TIVA versus inhalation anaesthesia [43].

Opioids

Opioid analgesics can affect tumour development via the modulation of cell proliferation and apoptosis. Morphine suppresses the activity of NK cells and the differentiation of T cells, promotes lymphocyte apoptosis and decreases toll-like receptor 4 (TLR4), expressed in macrophage membranes [44]. Likewise, fentanyl and sufentanil decrease the activity of NK cells but increase the number of regulatory T cells. Morphine stimulates the proliferation and angiogenesis of endothelial cells by activating mitogen-activated protein kinase/extracellular signal-regulated kinase phosphorylation via Gi/Go-coupled G protein receptors and nitric oxide in these microvascular endothelial cells [45, 46]. The promotion of tumour growth is mediated by AKT; extracellular signalling is mediated by ERK (extracellular signal-regulated kinase); the promoting effects of cell apoptosis are mediated by the inhibition of NF-κB, increase in Fas expression, p53 stabilisation, p38 activation and c-jun-N kinase (JNK) [47]. These effects include apoptosis inhibition via AKT activation, and promotion of cell cycle progression via the increase of cyclin D1 [48].

Studies suggest that sufentanil also inhibits leukocyte migration [49], alfentanil decreases the activity of NK cells and remifentanil was reported to suppress the activity of NK cells and lymphocytic proliferation in an in vitro model [50]. In addition, fentanyl showed anti-tumour effects in colorectal cancer cells in vitro. Its use is associated with reduced tumour cell migration and invasion via the inhibition of downregulation of E-26 transformation-specific sequence-1 into activated serine/threonine-kinase protein kinase B-raf (BRAF) – lncRNA [51].

Opioid-induced cell proliferation is likely to be concentration- and time-dependent. When low concentrations or a single dose of opioids are used, tumour growth is stimulated. Conversely, high concentrations or chronic opioid exposure leads to tumour growth inhibition [52]. In addition, morphine proved to inhibit the expression and secretion of MMP-2 and MMP-9 in breast cancer cells in a time- and dose-dependent manner.

Another study showed that fentanyl inhibits tumour growth and cell invasion in colorectal cancer due to downregulation of miR-182 and MMP-9 expression by β-catenin. A recent study demonstrated that sufentanil does not affect the rate of apoptosis or the cell cycle distribution in colon and pancreatic cancer cells in vitro when clinical concentrations were used. MMP activity cannot be reverted by naloxone, which indicates that the inhibition of MMP secretion by morphine is not mediated by opioid receptors [53].

Furthermore, overexpression of μ-opioid receptor (MOR) promotes tumour growth and metastasis in several cancer cell types [54]. The activation of AKT and mTOR is associated with cell proliferation and migration which, in turn, are related to MOR overexpression [55].

Treatment with methyl-naltrexone (MNTX), an opioid antagonist, inhibits tumour cell invasion and implantation, while continuous infusion of MNTX decreases primary tumour growth and development of lung metastasis. Clinically, MNTX has been found to be associated with higher overall survival rates in patients with advanced cancer, which supports the hypothesis that MOR is involved in tumour progression [56]. Treatment with morphine, both prior to and after surgery, significantly reduced stress-induced corticosteroids in rats [57]. This finding suggests that the preoperative administration of morphine may play a key role in protection against surgery-induced metastasis [58].

NSAIDs and COX-2 inhibitors

The induction of COX-2, which is frequently observed in cancer, has a role in immune resistance. COX-2 inhibitors increase the cytotoxicity of NK cells [59]. Moreover, when combined with β-adrenergic antagonists, they proved to decrease metastases in animal models [18]. A selective COX-2 inhibitor can suppress the release of prostaglandin E2 (PGE2) and promote immune responses against cancer cells [60].

The use of non-steroid anti-inflammatory drugs (NSAIDs) in preoperative medication increases cell immunity in cancer tissues [61]. PGE2 is a tumour-derived angiogenic factor independent of VEGF. PGE2 synthesis is controlled by COX-2 expression, and COX-2 inhibition blocks VEGF, leading to angiogenesis inhibition, tumour growth and metastasis formation [62].

Recently, study indicated that morphine enhanced TNBC metastasis and angiogenesis while ketorolac suppressed this effect. Mechanistically, this may be related to the enhancement of TSP-1 synthesis after ketorolac administration which further de-activated PI3K/AKT/c-Myc pathway [63].

However, in a recent clinical trial reported that a single administration of 30 mg of ketorolac tromethamine before surgery does not increase disease-free survival in high-risk breast cancer patients. Overall survival difference between ketorolac tromethamine group and placebo group was not statistically significant [64]

Local anaesthetics

Local anaesthetics block voltage-dependent sodium channels and may inhibit tumour growth. Lidocaine, ropivacaine and bupivacaine inhibit cell proliferation and differentiation, are cytotoxic for mesenchymal stem cells in vitro, and play a key role in tumour growth and metastasis development in cancer cells [65].

Locally-administered lidocaine inhibits epidermal growth factor receptor, which is a target molecule of many cancer drugs. A study that assessed the direct effect of local anaesthetics showed that lidocaine and bupivacaine induce apoptosis in cancer cells both in vivo and in vitro, suggesting potential benefits for cancer surgery [66]. Lidocaine and tetracaine, which inhibit motor proteins of kinesin, reduce the formation and activity of tubulin microtentacles. Therefore, these drugs may have a new ability to decrease metastatic spread in cancer [67]. In addition, lidocaine produces DNA demethylation in breast cancer cells in vitro [68]. Moreover, lidocaine, ropivacaine and bupivacaine reduce cell proliferation at concentrations of 100 μM, leading to cell cycle delay or arrest at G0/S-1 phases [69].

A recent study demonstrated that topic lidocaine increases the activity of NK cells against cancer in vitro by releasing lytic granules [70]. Furthermore, results from basic science studies reveal a promising role of local anaesthetics regarding the reduction of tumour recurrence [71].


Anaesthetics and genetic implications

The possibility that many of the beneficial or harmful effects related to the drugs used in the anaesthetic procedure, have a genetic and molecular basis is increasingly accepted [72]. The oncological disease could be mediated by specific genes or molecular pathways.

The relationship between genes and protein coding is widely known (DNA -> mRNA -> protein). Variations in the signalling pathways and the cells genetic material made by cancer is one of the objectives of study and development of current treatment.

Nevertheless, less than 2% of the mammalian genome encodes proteins, which means that >90% represents non-coding RNA (ncRNA) [73]. ncRNAs are involved in the control of development, differentiation, metabolism, cell growth and tumour progression [74]. In general, ncRNAs are classified into two groups based on their length: small ncRNAs (sncRNAs) and long ncRNAs (lncRNAs). sncRNAs include microRNA (miRNA), transfer RNA (tRNA) and some ribosomal RNA transcripts. Interplay patterns between lncRNAs and miRNAs appear to be crucial events in cancer progression.

Emerging data support the involvement of lncRNA in tumour-stroma communication, a potentially important landmark in cancer progression. Recently, Sang et al [75] demonstrated that lncRNA participates in the activation of calcium-dependent kinase (CamK-A), which is highly activated in several cancers and involved in remodelling the tumour microenvironment through the activation of calcium (Ca2 ), thereby promoting macrophage recruitment, angiogenesis and cancer progression.

Different miRNAs have been identified in biological fluids, such as urine and blood [76], serving as potential biomarkers in the diagnosis and prognosis of cancer. More recently, lncRNAs have been highlighted as potential biomarkers and cancer targets in precision medicine due to their specific expression patterns in tumour cells [77]. The role of LncRNAs in cancer disease has been known as different studies have shown its role as a diagnostic and prognostic element related to resistance to several drugs, through the modulation in the expression of drug transporters, and have been identified in signalling pathways that contribute to oncogenic survival, cell cycle, and apoptosis. In this line, lncRNA-specific therapeutic approaches target lncRNA-mediated functions and pathways through gene silencing and structure disruption mechanisms[78].

The genetic modifications in LncRNAs, trying to modulate a group of genes and acting on their functions, are the basis of future therapeutic objectives that could be reached.

In the regulation of gene expression by circular RNAs (circRNAs) through disease-related miRNAs, a complex network of miRNA-circRNA-ncRNA interacting at a protein level is formed, affecting a wide range of human diseases, particularly cancer. Likewise, circRNAs also have oncogenic and proto-oncogenic roles. Fusion circRNAs (f-circRNAs) result from cancer-associated genomic translocations and have tumour-promoting properties, including an increase in cell viability, a higher resistance to cancer therapy, and in vivo cell transformation into leukaemia cells [79]. circRNA expression has been demonstrated in different types of human cells [80], playing important physiological roles by regulating cell proliferation and haematopoiesis [81]. In addition, circRNAs have been implicated in the mediation of immune responses. For instance, circRasGEF1B upregulates the expression of TLR4/LPS-induced ICAM-1 mature mRNA stability, thus controlling the innate immune responses. Moreover, cellular mechanism allows to recognise foreign circRNAs through intron identity, which enables the identification of self and foreign circRNAs within the cytoplasm of the host [82].

Retrospective analyses suggest that an anaesthetic-analgesic technique during cancer surgery may affect recurrence/metastasis. This could involve direct effects on cancer cells. An interesting research objective is the modification of gene expression as a target of anaesthetic-analgesic drugs.

On the other hand, propofol has a potential antitumour effect, mainly due to the regulation of miRNA expression and transference. Accordingly, Wang et al [83] found that propofol suppresses cell proliferation and invasion in pancreatic cancer cells through the upregulation of miRNA-133a expression.

Furthermore, Xu et al [84] found that propofol induces the upregulation of miRNA let-7i expression and cell apoptosis in epithelial ovarian cancer cells in vitro.

Propofol increases miRNA-218 and miRNA-451 expression, whereas it reduces MMP-2 protein expression and cancer cell proliferation in vitro [85]. Likewise, Zhang et al [86] found that propofol reduces the invasiveness of hepatocellular carcinoma cells, partly due to the downregulation of MMP-9 by miRNA-199a.

In addition, the low expression of miRNA contributes to the antitumour effect of propofol. Accordingly, miRNA-21 is overexpressed in the early stages of pancreatic cancer [87]. Propofol inhibits miRNA-21 and suppresses the invasion of pancreatic cancer cells. In fact, it is thought that propofol inhibits the expression of miRNA-21 and reduces the expression of Slug, resulting in an increase in Slug-dependent PUMA (p53 proapoptotic target gene) and in the expression of E-cadherin [88]. The activation of PUMA and E-cadherin is involved in the inhibition of cell apoptosis. Therefore, propofol induces apoptosis and inhibits the invasion of pancreatic cancer cells through miR-21/Slug/E-cadherin and miR-21/Slug/PUMA signalling pathways [87].

Anaesthetic drugs may affect oncogene overexpression in certain types of cancer. Propofol can inhibits the expression of the androgen receptor in vitro, thus indicating a potential positive effect, as androgenic stimulation is involved in prostate cancer progression [16]. Furthermore, propofol has been shown to reduce of HIF-1a levels in cancer cells in vitro, with potential inhibitory effects on angiogenesis and, therefore, on tumour growth [89]. Volatile anaesthetics are known to be protective against ischaemia-reperfusion injury in a variety of clinical contexts and organ systems. This protection is associated with an induced expression of the angiogenesis-regulating factor HIF-1a, a mechanism that may be protective in the setting of reperfusion injury, but which promotes malignant recurrence in cancer surgery. Isoflurane has been found to increase the expression of HIF in prostate and renal cell carcinoma cells in different studies, and this has been associated with increased cancer cell migration and proliferation.

Opioids are related to modulation in cancer progression. Morphine, even when it was given in a low dose, can change the expression of gene groups, and induce metastasis (in vitro) [90]. The NET1 gene, belongs to this genetic group, overexpressed in breast and gastric adenocarcinoma cells through the Serial Analysis of Gene Expression database. The NET1 gene has a key role in the organisation of the actin cytoskeleton and, thus, in the ability of cancer cells to migrate and invade tissues, and even more, can help identify breast cancer patients at high risk of metastasis [91].

There is evidence supporting that a single dose of morphine can alter the expression of two major gene groups: regulators of proteins involved in mitochondrial pathways, and cytoskeleton-related proteins [92,93]. However, it has not been determined whether it modifies any cell function.

TNF-α increases the expression of intracellular adhesion molecule-1 (ICAM-1), a receptor required for leukocyte adhesion and tumour invasion. TNF-α also activates Src protein tyrosine kinase, which is a regulator of endothelial permeability. Src is involved in extravasation of cancerous cells, which is necessary for solid tumour metastasis [94]. Src is also involved with regulation of the cytoskeletal changes required for cell migration by phosphorylation of the proteins involved in focal adhesions and actin binding [95]. Lidocaine and ropivacaine have been reported to decrease cancer cell migration by inhibiting Src activation induced by tumour necrosis factor-α and phosphorylation of intercellular adhesion molecule-1 [96]. A later study showed that these local anaesthetics may have an anti-inflammatory effect, since in endothelial cells they effectively block inflammatory signaling of TNFα by attenuating the recruitment of p85 in the TNF-1 receptor. The resulting decrease in Akt, endothelial nitric oxide synthase, and Src phosphorylation reduced neutrophil adhesion and endothelial hyperpermeability [97].


Conclusion

There is growing evidence that the anaesthetic technique and anaesthetics may play a relevant role in tumour dissemination and relapse in the long term. The perioperative use of anaesthetic/analgesic techniques with protective effects on antimetastatic immune response may reduce tumour progression. To understand the mechanisms is important in order to study the genetic implications regarding anaesthetics drugs. New studies are necessary, the effects of anaesthetic drugs on the genetics of cancer need to be defined. The results of these studies may provide an answer if the data in animal models and in vitro studies can be applied in clinical practice and these studies may provide a new therapeutic target in cancer cells.


Sources of funding

This paper has been supported by the Anaesthesia Department and the Institution for Clinical Science. No external funding has been received.


Competing interests

The authors declare that they have no competing interests.


References

1. Pilleron S, Sarfati D, and Janssen-Heijnen M, et al (2019) Global cancer incidence in older adults, 2012 and 2035: a population-based study Int J Cancer 144 49–58 https://doi.org/10.1002/ijc.31664

2. Schreiber RD, Old LJ, and Smyth MJ (2011) Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion Science 331 1565–1570 https://doi.org/10.1126/science.1203486 PMID: 21436444

3. Reijmena E, Vannucci L, and De Couckc M, et al (2018) Therapeutic potential of the vagus nerve in cancer Immunol Lett 202 38–43 https://doi.org/10.1016/j.imlet.2018.07.006

4. Baum M, Demicheli R, and Hrushesky W, et al (2005) Does surgery unfavourably perturb the “natural history” of early breast cancer by accelerating the appearance of distant metastases? Eur J Cancer 41(4) 508–515 https://doi.org/10.1016/j.ejca.2004.09.031 PMID: 15737554

5. Gupta K, Kshirsagar S, and Chang L, et al (2002) Morphine stimulates angiogenesis by activating proangiogenic and survival-promoting signaling and promotes breast tumor growth Cancer Res 62 4491–4498 PMID: 12154060

6. Lennon FE, Moss J, and Singleton PA (2012) The mu-opioid receptor in cancer progression: is there a direct effect? Anesthesiology 116 940–945 https://doi.org/10.1097/ALN.0b013e31824b9512 PMID: 22357347

7. Jaura AI, Flood G, and Gallagher HC, et al (2014) Differential effects of serum from patients administered distinct anaesthetic techniques on apoptosis in breast cancer cells in vitro: a pilot study Br J Anaesth 113(1) 9 https://doi.org/10.1093/bja/aet581

8. Conrick-Martin I, Kell MR, and Buggy DJ (2012) Meta-analysis of the effect of central neuraxial regional anesthesia compared with general anesthesia on postoperative natural killer T lymphocyte function J Clin Anesth 24 3–7 https://doi.org/10.1016/j.jclinane.2011.09.001 PMID: 22284311

9. Grandhi RK, Lee S, and Abd-Elsayed A (2017) The relationship between regional Anesthesia and cancer: a metaanalysis Ochsner J 17 345–361 PMID: 29230120 PMCID: 5718448

10. Schreiber RD, Old LJ, and Smyth MJ (2011) Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion Science 331 1565–1570 https://doi.org/10.1126/science.1203486 PMID: 21436444

11. Buckley A, McQuaid S, and Johnson P, et al (2014) 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 113 (S1) i56–i62 https://doi.org/10.1093/bja/aeu200 PMID: 25009196

12. Wang YC, He F, and Feng F, et al (2010) Notch signaling determines the M1 versus M2 polarization of macrophages in antitumor immune responses Cancer Res 70 4840–4849 https://doi.org/10.1158/0008-5472.CAN-10-0269 PMID: 20501839

13. Deegan CA, Murray D, and Doran P, et al (2010) Anesthetic technique and the cytokine and matrix metalloproteinase response to primary breast cancer surgery Reg Anesth Pain Med 35 490–495 https://doi.org/10.1097/AAP.0b013e3181ef4d05 PMID: 20975461

14. Heaney A and Buggy DJ (2012) Can anaesthetic and analgesic techniques affect cancer recurrence or metastasis? Br J Anaesth 109(Suppl 1) i17–i28 https://doi.org/10.1093/bja/aes421

15. Ecimovic P, Mchugh B, and Murray D, et al (2013) Effects of sevoflurane on breast cancer cell function in vitro Anticancer Res 33 4255–4260 PMID: 24122989

16. Vogelaar FJ, Lips DJ, and Frank RC, et al (2016) Impact of anaesthetic technique on survival in colon cancer: a review of the literature Gastroenterol Rep 4(1) 30–34

17. Wall T, Sherwin A, and Buggy DJ (2019) Influence of perioperative anaesthetic and analgesic interventions on oncological outcomes: a narrative review Br J Anaesth 123(2) 135–150 https://doi.org/10.1016/j.bja.2019.04.062 PMID: 31255291 PMCID: 6676329

18. Brand L, Munding J, and Pox CP, et al (2013) ss-catenin, Cox-2 and p53 immunostaining in colorectal adenomas to predict recurrence after endoscopic polypectomy Int J Colorectal Dis 28(8) 1091–1098 https://doi.org/10.1007/s00384-013-1667-z PMID: 23516071

19. Tavare AN, Perry NJ, and Benzonana LL, et al (2012) Cancer recurrence after surgery: direct and indirect effects of anesthetic agents Int J Cancer 130(6) 1237–1250 https://doi.org/10.1002/ijc.26448

20. Retsky M, Rogers R, and Demicheli R, et al (2012) NSAID analgesic ketorolac used perioperatively may suppress early breast cancer relapse: particular relevance to triple negative subgroup Breast Cancer Res Treat 134 881–888 https://doi.org/10.1007/s10549-012-2094-5 PMID: 22622810

21. Kvolik S, Dobrosevic B, and Marczi S, et al (2009) Different apoptosis ratios and gene expresions in two human cell lines after sevofluorane anaesthesia Acta Anaesthesiol Scand 53 1192–1199 https://doi.org/10.1111/j.1399-6576.2009.02036.x PMID: 19572936

22. Huitink JM, Heimerikxs M, and Nieuwland M, et al (2010) Volatile anesthetics modulate gene expression in breast and brain tumor cells Anesth Analg 111(6) 1411–1415 https://doi.org/10.1213/ANE.0b013e3181fa3533 PMID: 20889943

23. Wigmore TJ, Mohammed K, and Jhanji S (2016) Long-term survival for patients undergoing volatile versus IV anesthesia for cancer surgery: a retrospective analysis Anesthesiology 124(1) 69–79 https://doi.org/10.1097/ALN.0000000000000936

24. Benzonana LL, Perry NJS, and Watts HR, et al (2013) Isoflurane, a commonly used volatile anesthetic, enhances renal cancer growth and malignant potential via the hypoxia inducible factor cellular signalling pathway in vitro Anesthesiology 119 593–605 https://doi.org/10.1097/ALN.0b013e31829e47fd PMID: 23774231

25. Huang H, Benzonana LL, and Zhao H, et al (2014) Prostate cancer cell malignancy via modulation of HIF-1α pathway with isoflurane and propofol alone and in combination Br J Cancer 111 1338–1349 https://doi.org/10.1038/bjc.2014.426 PMID: 25072260 PMCID: 4183852

26. Melamed R, Bar-Yosef S, and Shakhar G, et al (2003) 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 Anesth Analg 97 1331–1339 https://doi.org/10.1213/01.ANE.0000082995.44040.07 PMID: 14570648

27. Lee JH, Kang SH, and Kim Y, et al (2016) Effects of propofol-based total intravenous anesthesia on recurrence and overall survival in patients after modified radical mastectomy: a retrospective study Korean J Anesthesiol 69(2) 126–132 https://doi.org/10.4097/kjae.2016.69.2.126 PMID: 27066202 PMCID: 4823406

28. Jun IJ, Jo JY, and Kim JI, et al (2017) Impact of anesthetic agents on overall and recurrence-free survival in patients undergoing esophageal cancer surgery: a retrospective observational study Sci Rep 7(1) 14020 https://doi.org/10.1038/s41598-017-14147-9 PMID: 29070852 PMCID: 5656640

29. Soltanizadeh S, Degett TH, and Gögenur I (2017) Outcomes of cancer surgery after inhalational and intravenous anesthesia: a systematic review J Clin Anesth 42 19–25 https://doi.org/10.1016/j.jclinane.2017.08.001 PMID: 28797751

30. Byrne K, Levins KJ, and Buggy DJ (2016) Can anesthetic-analgesic technique during primary cancer surgery affect recurrence or metastasis? Can J Anesth 63(2) 184–192 https://doi.org/10.1007/s12630-015-0523-8

31. Oh TK, Kim K, and Jheon S, et al (2018) Long-term oncologic outcomes for patients undergoing volatile versus intravenous anesthesia for non-small cell lung cancer surgery: a retrospective propensity matching analysis Cancer Control 25(1) 1073274818775360 https://doi.org/10.1177/1073274818775360 PMID: 29739245 PMCID: 6028176

32. Zheng X, Wang Y, and Dong L, et al (2018) Effects of propofol-based total intravenous anesthesia on gastric cancer: a retrospective study Onco Targets Ther 11 1141–1148 https://doi.org/10.2147/OTT.S156792 PMID: 29535538 PMCID: 5840299

33. Lee JH, Kang SH, and Kim Y, et al (2016) Effects of propofol-based total intravenous anesthesia on recurrence and overall survival in patients after modified radical mastectomy: a retrospective study Korean J Anesthesiol 69(2) 126–132 https://doi.org/10.4097/kjae.2016.69.2.126 PMID: 27066202 PMCID: 4823406

34. Huang H, Benzonana LL, and Zhao H, et al (2014) Prostate cancer cell malignancy via modulation of HIF-1α pathway with isoflurane and propofol alone and in combination Br J Cancer 111 1338–1349 https://doi.org/10.1038/bjc.2014.426 PMID: 25072260 PMCID: 4183852

35. Hooijmansa CR, Geessinkb FJ, and Ritskes-Hoitingaa M, et al (2015) A systematic reviewand meta-analysis of the ability of analgesic drugs to reduce metastasis in experimental cancer models Pain 156 1835–1844 https://doi.org/10.1097/j.pain.0000000000000296

36. Wada H, Seki S, and Takahashi T, et al (2007) Combined spinal and general anesthesia attenuates liver metastasis by preserving TH1/TH2 cytokine balance Anesthesiology 106 499–506 https://doi.org/10.1097/00000542-200703000-00014 PMID: 17325508

37. Sakamoto A, Yagi K, and Okamura T, et al (2019) Perioperative administration of an intravenous beta-blocker landiolol hydrochloride in patients with lung cancer: a japanese retrospective exploratory clinical study Sci Rep 9 5217 https://doi.org/10.1038/s41598-019-41520-7 PMID: 30914694 PMCID: 6435747

38. Raytis JL and Lew MW (2013) Surgical stress response and cancer metastasis: the potential benefit of perioperative beta blockade Madame Curie Bioscience Database [Internet]

39. Jaura AI, Flood G, and Gallagher HC, et al (2014) Differential effects of serum from patients administered distinct anaesthetic techniques on apoptosis in breast cancer cells in vitro: a pilot study Br J Anaesth 113(S1) i63–i67 https://doi.org/10.1093/bja/aet581 PMID: 25009197

40. Zhao H, Zhang X, and Zheng Y, et al (2018) Propofol protects rat cardiomyocytes from anthracycline-induced apoptosis by regulating MicroRNA-181a in vitro and in vivo Oxid Med Cell Long Article ID 2109216 11 https://doi.org/10.1155/2018/2109216

41. Wigmore TJ, Mohammed K, and Jhanji S (2016) Long-term survival for patients undergoing volatile versus IV anesthesia for cancer surgery: a retrospective analysis Anesthesiology 124(1) 69–79 https://doi.org/10.1097/ALN.0000000000000936

42. Enlund M, Berglund A, and Andreasson K, et al (2014) The choice of anaesthetic--sevoflurane or propofol--and outcome from cancer surgery: a retrospective analysis Ups J Med Sci 119(3) 251–261 https://doi.org/10.3109/03009734.2014.922649 PMID: 24857018 PMCID: 4116765

43. Soltanizadeh S, Degett TH, and Gögenur I (2017) Outcomes of cancer surgery after inhalational and intravenous anesthesia: a systematic review J Clin Anesth 42 19–25 https://doi.org/10.1016/j.jclinane.2017.08.001 PMID: 28797751

44. Lennon FE, Mirzapoiazova T, and Mambetsariev B, et al (2012) Overexpression of the mu-opioid receptor in human non-small cell lung cancer promotes Akt and mTOR activation, tumor growth, and metastasis Anesthesiology 116(4) 857–867 https://doi.org/10.1097/ALN.0b013e31824babe2 PMID: 22343475

45. Singleton PA, Mirzapoiazova T, and Hasina R, et al (2014) Increased μ-opioid receptor expression in metastatic lung cancer Br J Anaesth 113(Suppl 1) i103–i108 https://doi.org/10.1093/bja/aeu165 PMCID: 4111280

46. Cieślińska A, Sienkiewicz-Szøapka E, and Kostyra E, et al (2015) M-Opioid receptor gene (OPRM1) polymorphism in patients with breast cancer Tumour Biol 36 4655–4660 https://doi.org/10.1007/s13277-015-3113-z

47. Nguyen J, Luk K, and Vang D, et al (2014) Morphine stimulates cancer progression and mast cell activation and impairs survival in transgenic mice with breast cancer Br J Anaesth 113(Suppl 1) i4–i13 https://doi.org/10.1093/bja/aeu090 PMID: 24861561 PMCID: 4111281

48. Heaney A and Buggy DJ (2012) Can anaesthetic and analgesic techniques affect cancer recurrence or metastasis? Br J Anaesth 109(Suppl 1) i17–i28 https://doi.org/10.1093/bja/aes421

49. Jaura AI, Flood G, and Gallagher HC, et al (2014) Differential effects of serum from patients administered distinct anaesthetic techniques on apoptosis in breast cancer cells in vitro: a pilot study Br J Anaesth 113(1) 9 https://doi.org/10.1093/bja/aet581

50. Cosinella F, Prieto I, and del Olmo M, et al (2015) Cancer surgery: how may anaesthesia influence outcome? J Clin Anaesth 27 262–272 https://doi.org/10.1016/j.jclinane.2015.02.007

51. Zhu Y, Zhang X, and Liu Y, et al (2012) Antitumor effect of the mTOR inhibitor everolimus in combination with trastuzumab on human breast cancer stem cells in vitro and in vivo Tumour Biol 33(5) 1349–1362 https://doi.org/10.1007/s13277-012-0383-6 PMID: 22492237

52. Kumar K, Kirksey MA, and Silvia D, et al (2017) A review of opioid-sparing modalities in perioperative pain management: methods to decrease opioid use postoperatively Anesth Analg 125(5) 1749–1760 https://doi.org/10.1213/ANE.0000000000002497 PMID: 29049119

53. Deegan CA, Murray D, and Doran P, et al (2010) Anesthetic technique and the cytokine and matrix metalloproteinase response to primary breast cancer surgery Reg Anesth Pain Med 35 490–495 https://doi.org/10.1097/AAP.0b013e3181ef4d05 PMID: 20975461

54. Liu Y, Zhang X, and Liu J, et al (2014) Everolimus in combination with letrozole inhibit human breast cancer MCF-7/Aro stem cells via PI3K/mTOR pathway: an experimental study Tumour Biol 35(2) 1275–1286 https://doi.org/10.1007/s13277-013-1170-8

55. Lennon FE, Mirzapoiazova T, and Mambetsariev B, et al (2012) Overexpression of the mu-opioid receptor in human non-small cell lung cancer promotes AKT and mTOR activation, tumor growth and metastasis Anaesthesiology 116(4) 857–867 https://doi.org/10.1097/ALN.0b013e31824babe2

56. Janku F, Johnson LK, and Karp DD, et al (2016) Treatment with methylnaltrexone is associated with increased survival in patients with advanced cancer Ann Oncol 27(11) 2032–2038 https://doi.org/10.1093/annonc/mdw317 PMID: 27573565 PMCID: 6267944

57. Connolly C, Madden SF, and Buggy DJ, et al (2017) Expression of anaesthetic and analgesic drug target genes in excised breast tumour tissue: association with clinical disease recurrence or metastasis PLoS One 12(5) e0177105 https://doi.org/10.1371/journal.pone.0177105 PMID: 28558008 PMCID: 5448742

58. Mulier J and De Kock M (2017) Opioid free general anesthesia, a new paradigm? Best Pract Res Clin Anaesthesiol 31(4) 441–443 https://doi.org/10.1016/j.bpa.2017.11.005

59. Farooqui M, Li Y, and Rogers T, et al (2007) COX-2 inhibitor celecoxib prevents chronic morphine-induced promotion of angiogenesis, tumour growth, metastasis and mortality, without compromising analgesia Br J Cancer 97(11) 1523–1531 https://doi.org/10.1038/sj.bjc.6604057 PMID: 17971769 PMCID: 2360252

60. Lee JY, Myung SK, and Song YS (2013) Prognostic role of cyclooxygenase-2 in epithelial ovarian cancer: a meta-analysis of observational studies Gynecol Oncol 129(3) 613–619 https://doi.org/10.1016/j.ygyno.2013.02.011 PMID: 23422504

61. Desmedt C, Demicheli R, and Fornili M, et al (2018) Potential benefit of intra-operative administration of ketorolac on breast cancer recurrence according to the patient’s body mass index J Natl Cancer Inst 110(10) 1115–1122 https://doi.org/10.1093/jnci/djy042 PMID: 29718396

62. Forget P, Berlière M, and Maanen AV, et al (2013) Perioperative ketorolac in high risk breast cancer patients. Rationale, feasibility and methodology of prospective randomized placebo-controlled trial Med Hypotheses 81(4) 707–712 https://doi.org/10.1016/j.mehy.2013.07.033 PMID: 23937996

63. Madan HK, Singh R, and Sodhi GS, et al (2016) Comparsion of intravenous lignocaine, tramadol and keterolac for attenuation of propofol injection pain J Clin Diagn Res 10(7) UC05–UC08 PMID: 27630928 PMCID: 5020294

64. Liu Z, Chengab S, and Fu G, et al (2020) Postoperative administration of ketorolac averts morphine-induced angiogenesis and metastasis in triple-negative breast cancer Life Sci 251 117604 https://doi.org/10.1016/j.lfs.2020.117604 PMID: 32243929

65. D’Agostino G, Saporito A, and Cecchinato V, et al (2018) Lidocaine inhibits cytoskeletal remodelling and human breast cancer cell migration BJA 121(4) 962–968 https://doi.org/10.1016/j.bja.2018.07.015

66. Tiouririne M, et al IV Lidocaine for Patients Undergoing Primary Breast Cancer Surgery: Effects on Postoperative Recovery and Cancer Recurrence ClinicalTrials.gov Identifier: NCT01204242

67. Wall T, Sherwin A, and Ma D, et al (2019) Influence of perioperative anaesthetic and analgesic interventions on oncological outcomes: a narrative review Br J Anaesth 123(2) 135–150 https://doi.org/10.1016/j.bja.2019.04.062 PMID: 31255291 PMCID: 6676329

68. Missair A, Cata JP, and Votta-Velis G, et al (2019) Impact of perioperative pain management on cancer recurrence: an ASRA/ESRA special article Reg Anesth Pain Med 44(1) 13–28 https://doi.org/10.1136/rapm-2018-000001 PMID: 30640648

69. Chamaraux-Tran TN, Mathelin C, and Aprahamian M, et al (2018) Antitumor effects of lidocaine on human breast Cancer cells: an in vitro and in vivo experimental trial Anticancer Res 38(1) 95–105

70. Aird J, Baird AM, and Lim MC, et al (2018) Carcinogenesis in prostate cancer: the role of long noncoding RNAs Non-coding RNA Res 3 29–38 https://doi.org/10.1016/j.ncrna.2018.01.001

71. Bhat SA, Ahmad SM, and Mumtaz PT, et al (2016) Long non-coding RNAs: mechanism of action and functional utility Non-coding RNA Res 1 43–50 https://doi.org/10.1016/j.ncrna.2016.11.002

72. Guo Y, Yang J, and Huang Q, et al (2019) Circular RNAs and their roles in head and neck cancers Mol Cancer 18 44 https://doi.org/10.1186/s12943-019-1003-5 PMID: 30898135 PMCID: 6427840

73. Liu Y, Ding M, and Gao Q, et al (2018) Current advances on the important roles of enhancer rnas in gene regulation and cancer BioMed Res Int Article ID 2405351

74. Azofeifa JG, Allen MA, and Hendrix JR, et al (2018) Enhancer RNA profiling predicts transcription factor activity Genome Res 28(3) 334–344 https://doi.org/10.1101/gr.225755.117 PMCID: 5848612

75. Sang L-J, Ju H-Q, Liu G-P, et al (2018) LncRNA CamK-A regulates Ca2 -signaling-mediated tumor microenvironment remodeling Mol Cell 72(1) 4 https://doi.org/10.1016/j.molcel.2018.10.024

76. Sweeney BP (2003) Do genes influence outcome from anaesthesia? Br J Anaesth 90(6) 725–727 https://doi.org/10.1093/bja/aeg103 PMID: 12765885

77. Lin Y-H, Wu M-H, and Yeh C-T, et al (2018) Long non-coding rnas as mediators of tumor microenvironment and liver cancer cell communication Int J Mol Sci 19 3742 https://doi.org/10.3390/ijms19123742

78. Kolendowski B, Hassan H, and Krstic M, et al (2018) Genome-wide analysis reveals a role for TDG in estrogen receptor-mediated enhancer RNA transcription and 3-dimensional reorganization Epigenet Chromatin 11(1) article 5 https://doi.org/10.1186/s13072-018-0176-2

79. Jiang S, Liu Y, Huang L (2018) Effects of propofol on cancer development and chemotherapy: potential mechanisms Eur J Pharmacol 831 46–51 https://doi.org/10.1016/j.ejphar.2018.04.009 PMID: 29654781

80. Yuan Y, Jiang YC, and Sun CK, et al (2016) Role of the tumor microenvironment in tumor progression and the clinical applications (Review) Oncol Rep 35 2499–2515 https://doi.org/10.3892/or.2016.4660 PMID: 26986034

81. He M, Xu Z, and Ding T, et al (2009) MicroRNA-155 regulates inflammatory cytokine production in tumor-associated macrophages via targeting C/EBPbeta Cell Mol Immunol 6 343–352 https://doi.org/10.1038/cmi.2009.45 PMID: 19887047 PMCID: 4003217

82. Chai ZT, Zhu XD, and Ao JY, et al (2015) microRNA-26a suppresses recruitment of macrophages by down-regulating macrophage colony-stimulating factor expression through the PI3K/Akt pathway in hepatocellular carcinoma J Hematol Oncol 8 56 https://doi.org/10.1186/s13045-015-0150-4 PMID: 26021873 PMCID: 4455972

83. Wang ZT, Gong HY, and Zheng F, et al (2015) Propofol suppresses proliferation and invasion of pancreatic cancer cells by upregulating microRNA-133a expression GMR 14(3) 7529–7537 https://doi.org/10.4238/2015.July.3.28 PMID: 26214431

84. Xu S, Lou F, and Wu Y, et al (2016) Circulating tumor DNA identified by targeted sequencing in advanced-stage non-small cell lung cancer patients Cancer Lett 370(2) 324–331 https://doi.org/10.1016/j.canlet.2015.11.005

85. Peng K, Liu HY, and Wu SR, et al (2016) Does propofol anesthesia lead to less postoperative pain compared with inhalational anesthesia? A systematic review and meta-analysis Anesth Analg 123(4) 846–858 https://doi.org/10.1213/ANE.0000000000001504 PMID: 27636574

86. Zhang Y, Yang L, and Kucherlapati M, et al (2018) A pan-cancer compendium of genes deregulated by somatic genomic rearrangement across more than 1,400 cases Cell Rep 24(2) 515–527 https://doi.org/10.1016/j.celrep.2018.06.025 PMID: 29996110 PMCID: 6092947

87. Li B, Tsao SW, and Li YY, et al. (2009) Id-1 promotes tumorigenicity and metastasis of human esophageal cancer cells through activation of PI3K/AKT signaling pathway Int J Cancer 125 2576–2585 https://doi.org/10.1002/ijc.24675 PMID: 19551863

88. Liu X, Ranganathan R, and Jiang S, et al (2016) A chimeric switch-receptor targeting pd1 augments the efficacy of second-generation car t cells in advanced solid tumors Cancer Res 76(6) 1578–1590 https://doi.org/10.1158/0008-5472.CAN-15-2524 PMID: 26979791 PMCID: 4800826

89. Cai Q, Wang Z, and Wang S, et al (2017) Long non-coding RNA LINC00152 promotes gallbladder cancer metastasis and epithelial–mesenchymal transition by regulating HIF-1a via miR-138 Open Biol 7 160247 https://doi.org/10.1098/rsob.160247

90. Jun JC, Rathore A, and Younas H, et al (2017) Hypoxia-inducible factors and cancer Curr Sleep Med Rep 3(1) 1–10 https://doi.org/10.1007/s40675-017-0062-7 PMID: 28944164 PMCID: 5607450

91. Tao L, Huang G, and Song H, et al (2017) Cancer associated fibroblasts: an essential role in the tumor microenvironment Oncol Lett 14 2611–2620 https://doi.org/10.3892/ol.2017.6497 PMID: 28927027 PMCID: 5588104

92. Izdebska M, Zielińska W, and Grzanka D, et al (2018) The role of actin dynamics and actin-binding proteins expression in epithelial-to-mesenchymal transition and its association with cancer progression and evaluation of possible therapeutic targets Biomed Res Int 2018 4578373 https://doi.org/10.1155/2018/4578373 PMID: 29581975 PMCID: 5822767

93. Wu Y, Ding J, and Sun Q, et al (2018) Long noncoding RNA hypoxia-inducible factor 1 alpha-antisense RNA 1 promotes tumor necrosis factor-alpha-induced apoptosis through caspase 3 in Kupffer cells Medicine 97 e9483 https://doi.org/10.1097/MD.0000000000009483

94. Piegeler T, Gina Votta-Velis E, and Bakhshi FR, et al (2014) Endothelial barrier protection by local anesthetics: ropivacaine and lidocaine block tumor necrosis Factor-α-induced endothelial cell Src activation Anesthesiology 120(6) 1414–1428 https://doi.org/10.1097/ALN.0000000000000174 PMID: 24525631 PMCID: 4284094

95. Wang W, Liu Y, and Liao K (2011) Tyrosine phosphorylation of cortactin by the FAK-Src complex at focal adhesions regulates cell motility BMC Cell Biol 12 49 https://doi.org/10.1186/1471-2121-12-49 PMID: 22078467 PMCID: 3245448

96. Grandhi RK and Perona B (2020) Mechanisms of action by which local anesthetics reduce cancer recurrence: a systematic review Pain Med 21(2) 401–414

97. Piegeler T, Gina Votta-Velis E, and Liu G, et al (2012) Anti-metastatic potential of amide-linked local anesthetics: inhibition of lung adenocarcinoma cell migration and inflammatory Src signaling independent of sodium channel blockade Anesthesiology 117(3) 548–559 https://doi.org/10.1097/ALN.0b013e3182661977 PMID: 22846676 PMCID: 3482823

 

Artículos relacionados

Estefania Moreno-Luna, Patricia Alonso, Javier De Santiago, Ignacio Zapardiel
Carmen Herrero-Vicent, Isidro Machado, Carmen Illueca, Amparo Avaria, Claudia Salazar, Abraham Hernandez, Sergio Sandiego, Javier Lavernia
Ana Vaz da Conceição, Dora Bernardo, Lygia Vieira Lopes, Fernando Miguel, Fernanda Bessa, Fernando Monteiro, Cristina Santos, Blasques Oliveira, Lúcio Lara Santos
Elkin Muñoz, Naira González, Luis Muñoz, Jesús Aguilar, Juan A García Velasco
Table of Contents
Table of Contents