Next Article in Journal
The NOTCH-RIPK4-IRF6-ELOVL4 Axis Suppresses Squamous Cell Carcinoma
Next Article in Special Issue
Immune Phenotypic Characterization of a TRAIL-Knockout Mouse
Previous Article in Journal
An Update of G-Protein-Coupled Receptor Signaling and Its Deregulation in Gastric Carcinogenesis
Previous Article in Special Issue
Tumor Necrosis Factor Receptor 2 (TNFR2): An Emerging Target in Cancer Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumor Necrosis Factor Alpha: Implications of Anesthesia on Cancers

1
Department of Anesthesiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
2
Department of Anesthesiology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
3
Department of Anesthesiology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
4
Center for Regional Anesthesia and Pain Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(3), 739; https://doi.org/10.3390/cancers15030739
Submission received: 26 December 2022 / Revised: 20 January 2023 / Accepted: 23 January 2023 / Published: 25 January 2023
(This article belongs to the Special Issue Tumor Necrosis Factor: Molecular Insights and Clinical Implications)

Abstract

:

Simple Summary

Cancer ranks as a leading cause of death worldwide, which is often the result of recurrence and metastasis. Surgical resection is the mainstay of cancer treatment for potentially removable solid tumors. However, surgery-induced stress responses may lead to immunosuppression and subsequent cancer regrowth and spread. Evidence shows that the tumor microenvironment plays an essential role in disease progression through mechanisms such as inflammation promotion. Tumor necrosis factor alpha (TNF-α) is one of the pro-inflammatory cytokines found in cancer patients and is reported to be involved in the immune system as well as in the surveillance of tumor growth. To date, several studies have demonstrated that various anesthetic agents or techniques and perioperative management have varying effects on innate and cellular immunity, the enhancement of adrenergic-inflammatory responses, and the activation of cancer-promoting cellular signaling pathways, which may be associated with long-term cancer outcomes. This review outlines the current knowledge of anesthesia implications on TNF-α release and provides potential anesthetic strategies for improving patient survival.

Abstract

Cancer remains a major public health issue and a leading cause of death worldwide. Despite advancements in chemotherapy, radiation therapy, and immunotherapy, surgery is the mainstay of cancer treatment for solid tumors. However, tumor cells are known to disseminate into the vascular and lymphatic systems during surgical manipulation. Additionally, surgery-induced stress responses can produce an immunosuppressive environment that is favorable for cancer relapse. Up to 90% of cancer-related deaths are the result of metastatic disease after surgical resection. Emerging evidence shows that the interactions between tumor cells and the tumor microenvironment (TME) not only play decisive roles in tumor initiation, progression, and metastasis but also have profound effects on therapeutic efficacy. Tumor necrosis factor alpha (TNF-α), a pleiotropic cytokine contributing to both physiological and pathological processes, is one of the main mediators of inflammation-associated carcinogenesis in the TME. Because TNF-α signaling may modulate the course of cancer, it can be therapeutically targeted to ameliorate clinical outcomes. As the incidence of cancer continues to grow, approximately 80% of cancer patients require anesthesia during cancer care for diagnostic, therapeutic, or palliative procedures, and over 60% of cancer patients receive anesthesia for primary surgical resection. Numerous studies have demonstrated that perioperative management, including surgical manipulation, anesthetics/analgesics, and other supportive care, may alter the TME and cancer progression by affecting inflammatory or immune responses during cancer surgery, but the literature about the impact of anesthesia on the TNF-α production and cancer progression is limited. Therefore, this review summarizes the current knowledge of the implications of anesthesia on cancers from the insights of TNF-α release and provides future anesthetic strategies for improving oncological survival.

1. Introduction

Cancer is a major public health issue and a leading cause of death worldwide, accounting for 19.3 million new cases and almost 10.0 million deaths in 2020 [1]. Despite advancements in chemotherapy, radiation therapy, and immunotherapy, surgery is the mainstay of cancer treatment for solid tumors [2,3]. However, up to 90% of cancer-related deaths are the result of metastatic disease after surgical resection [4]. Tumor cells are known to disseminate into the vascular and lymphatic systems during surgical manipulation and subsequently migrate to distant organs and initiate tumor regrowth [5,6]. In addition, surgery-induced stress responses can lead to the immunosuppression and upregulation of adhesion molecules through mechanisms involving inflammation, ischemia–reperfusion injury, sympathetic nervous system activation, and increased cytokine release, significantly increasing the risk of metastatic recurrence [5]. Ultimately, the combination of potential tumor cell dissemination and an immunosuppressive state may result in the growth of residual cancer and metastatic seeding. Thus, modulating the cellular and molecular profiles that contribute to tumor recurrence and metastasis during surgery is critical to improve the outcomes for cancer patients (Figure 1).
The tumor microenvironment (TME) is the cellular environment in which the tumor exists and is composed of various cell types (fibroblasts, endothelial cells, immune cells, etc.) and extracellular components (cytokines, growth factors, extracellular matrix, etc.) [7]. The interactions between tumor cells and the TME not only play decisive roles in tumor initiation, progression, and metastasis but also have profound effects on therapeutic efficacy [8]. Evidence shows that the TME can harbor tumor cells to invade healthy tissues and spread to other body parts through the lymphatic or circulatory system; however, non-malignant cells in the TME perform a pro-tumorigenic function in all stages of carcinogenesis by stimulating and facilitating uncontrolled cell proliferation [9]. Notably, surgery itself and the resultant stress responses have direct impacts on tumor cells and can form a supportive TME through platelet activation, neutrophil extracellular traps, and immunosuppression, promoting occult tumor growth and the metastatic process [10,11].
Tumor necrosis factor alpha (TNF-α) is a pleiotropic cytokine contributing to both physiological and pathological processes. Its superfamily members include TNF beta (TNF-β), CD40 ligand (CD40L), Fas ligand (FasL), TNF-related apoptosis-inducing ligand (TRAIL), and TNF superfamily member 14 (LIGHT) [12]. As a pro-inflammatory cytokine, TNF-α is one of the main mediators of inflammation-associated carcinogenesis in the TME. It exerts diverse effects through two receptors: TNF receptor type 1 (TNFR1) and TNF receptor type 2 (TNFR2). The major role of TNFR1 is the initiation of cell apoptosis, but the activation of TNFR1 can also induce cell survival mechanisms [13,14]. The determination of the final activity of TNFR1 is based on the concentration of TNF-α in the TME, as well as the effects of other involved cytokines [13]. As for TNFR2, its activation assists in regulating the immune and inflammatory responses, which can promote tumor growth and progression [13,14]. TNF-α is reported to have the potential to act as a diagnostic biomarker in oncological diseases [15,16,17,18,19]. In addition, previous studies have demonstrated that the increased expression of TNF-α is associated with poor survival in endometrial, colorectal, and hepatocellular cancers [17,19,20]. These findings indicate that TNF-α signaling may modulate the course of cancer, which could be therapeutically targeted to ameliorate clinical outcomes.
Because of the growing incidence of cancer, approximately 80% of cancer patients require anesthesia during cancer care for diagnostic, therapeutic, or palliative procedures, and over 60% of cancer patients receive anesthesia for primary surgical resection [4]. Clearly, anesthesia has become a requisite component of surgery and even cancer treatment. Growing evidence has shown that various anesthetic agents or techniques and perioperative management have distinct effects on innate and cellular immunity, the enhancement of adrenergic-inflammatory responses, and the activation of cancer-promoting cellular signaling pathways [2,4,5,6]. Moreover, there are retrospective studies reporting significant differences in survival among clinical anesthetic settings in different types of cancer surgeries [21,22,23,24,25,26,27]. A meta-analysis also concludes that the choice of anesthesia may be associated with long-term cancer outcomes [28]. Therefore, the following review aims to summarize an up-to-date overview of the current experimental and clinical evidence of the anesthesia implications on cancers based on the insights of TNF-α release. It also aims to provide protective anesthesia strategies for improving patient survival in the future.

2. General Anesthesia

2.1. Volatile Anesthetics

Volatile anesthetics (VAs) are used extensively in surgical oncology. Numerous in vitro studies have investigated the effects of VAs on human cancer cell lines and demonstrated that VAs seem to enhance the malignant potential of tumor cells and accelerate cancer progression [29,30,31,32,33,34]. Similar results are also observed in animal models [29,30]. However, some studies report inconsistent findings due to varied experimental protocols, environments, and subject species [35,36,37,38,39]. In addition, VAs have been reported to modulate the innate as well as the adaptive immune responses [40,41]. As a result, the effects of VAs on tumorigenesis may depend on the balance between the cellular signaling pathways and the immunosuppressive microenvironment formation.

2.1.1. Laboratory Research

Research on the TNF-α release and its impact on cancer cell biology with the exposure to VAs is lacking (Table 1). Using human colon cancer cell lines, the exposure of isoflurane was reported to enhance resistance against TRAIL (a member of the TNF-α superfamily)-induced apoptosis through mechanisms related to caveolin-1, which suggested that the exposure of isoflurane may inadvertently decrease sensitivity to postoperative chemotherapeutic agents and even aggravate oncological outcomes [42].

2.1.2. Clinical Studies

During cancer surgery, the effect of VAs on the TNF-α release is still uncertain. Chen et al. conducted a clinical study, which demonstrated that sevoflurane anesthesia significantly improved intraoperative hemodynamics and reduced the serum level of TNF-α in elderly patients undergoing lobectomy for lung cancer, but the incidence of postoperative complications simultaneously increased compared to propofol-based total intravenous anesthesia (TIVA) [43]. Moreover, Mahmoud et al. found that isoflurane anesthesia decreased the alveolar and plasma concentrations of TNF-α associated with one-lung ventilation (OLV) during open thoracic cancer surgery and had better postoperative outcomes compared with propofol anesthesia [44]. However, Jin et al. reported that, compared with propofol-based TIVA, sevoflurane anesthesia in patients undergoing lung cancer surgery under OLV exacerbated the injury to pulmonary function during the perioperative period via the increased release of serum TNF-α, aggravated lung edema, and inhibited hypoxic pulmonary vasoconstriction [45]. Qiao et al. also demonstrated that elderly patients receiving sevoflurane anesthesia during esophageal cancer surgery had a higher incidence of postoperative cognitive dysfunction (POCD) and an elevated plasma concentration of TNF-α than those maintained on propofol anesthesia [46]. Additionally, sevoflurane anesthesia and propofol-based TIVA did not make a difference in serum TNF-α production in patients undergoing major colorectal surgery for cancer disease or inflammatory bowel disease [47]. Similarly, an in vitro analysis study revealed no obvious impacts on TNF-α release during different types of anesthesia for breast cancer surgery [48]. Due to conflicting results, further large-scale and high-quality randomized control trials are required to determine the significance of these observations and the impact on prognosis after cancer surgery (Table 2; Figure 2).

2.2. Propofol

Propofol, a commonly used intravenous anesthetic, is widely administered in clinical anesthetic practice for cancer surgery. It has been reported to exert anti-tumor effects through various mechanisms in different cancer types [49]. Previous in vitro studies have shown that propofol can not only suppress the aggressiveness of human cancer cells [50,51,52,53,54,55,56,57], but can also alter the resistance against chemotherapeutic agents [51,58,59,60,61]. Animal studies provide similar information on the effects of propofol on tumor growth [50,58,62,63,64]. Furthermore, propofol can preserve the integrity of host immune function and mitigate surgery-induced immunosuppression [6,49]. Although there are few experimental investigations showing discrepancies [65,66], propofol is generally regarded to beneficially affect cancer development.

2.2.1. Laboratory Research

In mouse neuroblastoma cell lines, propofol pretreatment ameliorated oxygen-glucose deprivation/reperfusion-induced inflammation and inhibited TNF-α production by modulating the nuclear transcription factor κB (NF-κB) pathway, indicating that propofol may be considered as a potential therapeutic approach to reduce inflammation in neuroblastoma [67]. However, Yan et al. revealed that treatment with propofol during tumor resection in murine models of breast cancer exhibited similar effects on reducing T lymphocyte cell counts, upregulating serum TNF-α production, and promoting distant metastasis compared with sevoflurane, which implied that the two common anesthetics modulated cancer progression through their immunosuppressive activity [68]. Current laboratory data about the effect of propofol on the release of TNF-α in cancer cell biology are limited, and further investigations are required (Table 1).

2.2.2. Clinical Studies

During cancer surgery, propofol anesthesia had an inhibitory effect on the release of TNF-α in esophageal cancer patients [46] and contradictory results in lung cancer patients compared to inhalation anesthesia [43,44,45]. Moreover, the effect of propofol anesthesia on the plasma concentration of TNF-α did not differ from that of sevoflurane anesthesia during colorectal and breast cancer surgery [47,48]. Thus, although propofol anesthesia is recommended as a better choice for cancer surgery, prospective randomized control trials are needed to clarify its influence on the TNF-α release and cancer outcomes in the future (Table 2; Figure 2).

2.3. Ketamine

Ketamine is an N-methyl-D-aspartate receptor antagonist clinically used for anesthetic and analgesic purposes, which may provide anti-inflammatory and immunomodulatory effects in cancer surgery. Based on previous in vitro research, it has been determined that ketamine possesses the capacity to attenuate the malignant potential of various cancer cells [69,70,71,72,73,74]. However, there are inconsistent findings surrounding the relationship between ketamine administration and tumor metastasis from in vivo studies [64,75,76]. Additionally, ketamine seems to negatively influence cell-mediated immunity, especially nature killer (NK) cell activity, in animal models [64,76] but does not exhibit direct immunomodulation in clinical investigations [77,78]. Accordingly, there is a lack of evidence to advocate for the perioperative use of ketamine to improve outcomes in cancer patients.

2.3.1. Laboratory Research

Basic research on the impact of ketamine administration on TNF-α production in cancer cell biology is limited (Table 1). Using mouse neuroblastoma cell lines, the administration of ketamine provided an anti-inflammatory effect on the lipopolysaccharide (LPS)-induced production of TNF-α through the inhibition of NF-κB and inducible nitric oxide synthase pathways [79].

2.3.2. Clinical Studies

Ali et al. showed that ketamine administration could significantly suppress serum TNF-α production, especially when given in repeated doses, in patients undergoing radical prostatectomy [80]. During the laparoscopic radical resection of colorectal cancer, the addition of a single subanesthetic dose of ketamine was also reported to decrease the plasma level of TNF-α, prevent brain injury, and improve postoperative fatigue syndrome [81]. Similarly, Ren et al. showed that ketamine exerted an inhibitory effect on the release of serum TNF-α and improved the postoperative anxiety and depression of colorectal cancer patients [82]. In contrast, Cho et al. reported that subanesthetic doses of ketamine as an adjunct to desflurane anesthesia during colorectal cancer surgery did not convey any favorable impacts on the postoperative plasma level of TNF-α, NK cell activity, or long-term prognosis [77]. With ketamine as an anesthetic adjuvant, TIVA with propofol and remifentanil also failed to provide a direct immunomodulatory effect on the release of serum TNF-α during minimally invasive robotic radical prostatectomies [78]. Although the anti-inflammatory effect of ketamine on the TNF-α release is predominated, the impact of ketamine administration in cancer surgery is still debated (Table 2; Figure 2).

2.4. Dexmedetomidine

Dexmedetomidine (DEX), a highly selective α-2 adrenergic receptor agonist, exerts sedative, hypnotic, anxiolytic, sympatholytic, and analgesic properties. Due to its opioid-sparing and hemodynamic stabilizing effects, DEX has become an essential component of multimodal analgesia in major oncological surgeries. DEX is reported to restrain biological behaviors in human cancer cell lines [83,84,85,86] and suppress tumor progression in animal models through various pathways [83,86]. Nevertheless, there are contradictory results observed [87,88,89,90]. Moreover, the immunomodulatory effects of DEX from α-2 adrenergic receptors on various immune cells have been increasingly recognized [91]. Taken together, the impacts of DEX administration on cancer biology and immunity remain inconclusive, and further laboratory and clinical studies are warranted in the future.

2.4.1. Laboratory Research

In rat models of ovarian cancer, DEX treatment could effectively decrease the plasma concentration of TNF-α and enhance the immunity through various signaling pathways, which eventually inhibited tumor growth, invasion, and migration [92,93]. Shin et al. also showed that DEX infusion during ovarian cancer surgery suppressed postoperative levels of serum TNF-α and cortisol, sped up the recovery of NK cell activity, and lowered the tumor burden after surgery in mouse models of ovarian cancer xenograft [94]. In addition, using rat pheochromocytoma cell lines, DEX significantly decreased oxygen-glucose deprivation/reperfusion-induced inflammation and apoptosis and reduced the release of TNF-α [95]. Based on the available data, DEX seems to have beneficial effects on the production of TNF-α and tumor progression (Table 1).

2.4.2. Clinical Studies

In a meta-analysis, Wang et al. reported that DEX administration attenuated perioperative inflammation with a decreased TNF-α production and maintained the integrity of the immune function, which may contribute to diminish postoperative complications and improve clinical outcomes in surgical patients [96]. Pretreatment with DEX before the induction of general anesthesia (GA) was reported to protect lung cancer patients against lung injury during OLV by upregulating the expression of heme oxygenase-1 and reducing the serum production of TNF-α and reactive oxygen species [97]. Compared with GA plus epidural anesthesia (EA), adjunctive DEX in GA provided better hemodynamic control, dismissed inflammatory response with a reduction in serum TNF-α, protected intestinal function, and shortened the length of hospital stay in elderly patients with a gastrointestinal tumor after laparoscopic radical gastrectomy [98]. Dong et al. demonstrated that DEX could effectively reduce the release of plasma TNF-α by downregulating the expression of NF-κB and ameliorate the impaired immune function in patients receiving radical resections of gastric cancer [99]. Furthermore, DEX infusion decreased the plasma TNF-α concentration and improved the occurrence of POCD in patients undergoing surgical resections of colon, gynecological, gastric, and esophageal cancer [100,101,102,103]. Patients administered with DEX in colorectal cancer surgery also had better cardiocerebral protection with a reduced expression level of serum TNF-α [104]. Similarly, DEX could significantly stabilize hemodynamics, reduce inflammation via the downregulation of serum TNF-α, and inhibit free radical generation, playing a vital role in brain protection in patients undergoing craniotomy resections of glioma [105]. Liu et al. conducted a retrospective analysis and showed that DEX anesthesia could effectively stabilize hemodynamics and reduce the release of serum TNF-α compared with midazolam anesthesia for patients undergoing radical resections of ovarian cancer [106].
In addition to administration during cancer surgery, DEX could be an adjuvant for postoperative analgesia in cancer patients. During the postoperative period of patients undergoing a combined thoracoscopic–laparoscopic esophagectomy, the addition of DEX to sufentanil in intravenous patient-controlled analgesia (IVPCA) exhibited better postoperative analgesia, fewer inflammatory responses with a reduced plasma TNF-α level, and lower postoperative delirium categories and better health statuses [107]. Likewise, a combination of the intraoperative usage of DEX and postoperative IVPCA of DEX and ketorolac could provide adequate postoperative analgesia, reduce opioid consumption and relative complications, and alleviate immunosuppression and inflammation with a decrease in the serum TNF-α level in patients with lung cancer undergoing thoracoscopic surgery [108]. Song et al. also reported that DEX combined with oxycodone in IVPCA had better sedative and analgesic effects with fewer adverse reactions and reduced the release of serum TNF-α compared with oxycodone alone for patients with laparoscopic radical resections of rectal cancer [109]. In summation, perioperative DEX treatment seemed to have a beneficial effect on anti-inflammation by reducing the expression of TNF-α and improved postoperative outcomes in patients receiving cancer surgery (Table 2; Figure 2).

2.5. Systemic Lidocaine

Lidocaine is an amide local anesthetic and possesses an analgesic effect when used intravenously in perioperative settings. The administration of intravenous lidocaine during anesthesia has been associated with lowering the use of opioids, reducing the incidence of postoperative nausea and vomiting, and enhancing patients’ postoperative recovery [110]. Like other adjunctive agents, lidocaine has an inhibitory effect on carcinogenesis in experimental observations [111,112,113,114,115]. Apart from affecting cancer cell behavior, lidocaine can exhibit beneficial effects on components of inflammatory and immune responses [116]. Although results from in vitro and in vivo studies indicate that lidocaine may prevent against tumor progression, whether lidocaine can improve outcomes of surgical cancer patients needs to be investigated in prospective trials.

2.5.1. Laboratory Research

Laboratory research on the impact of lidocaine treatment on the release of TNF-α in cancer cell biology is limited (Table 1). Ferreira et al. demonstrated that lidocaine in vitro and in vivo provided an anti-proliferative effect on tumor cells without affecting the release of TNF-α [117].

2.5.2. Clinical Studies

During supratentorial tumor resections, systemic lidocaine administration improved early recovery quality and had neuroprotective effects, which may be attributed to its analgesic and inflammation-alleviating properties with a reduction in serum TNF-α [118]. Systemic lidocaine was also reported to improve postoperative recovery, alleviate inflammation and immunosuppression with a decrease in plasma TNF-α, and accelerate the return of bowel function in patients with gastric cancer undergoing laparoscopic radical gastrectomies [119]. Nevertheless, Herroeder et al. suggested that systemic lidocaine could significantly accelerate the return of bowel function and shorten the length of hospital stay after colorectal surgery by modulating pro-inflammatory cytokines other than TNF-α [120]. Accordingly, further investigations about the effects of systemic lidocaine on inflammatory responses and outcomes in patients receiving cancer surgery are needed (Table 2; Figure 2).

2.6. Midazolam

Midazolam is a common benzodiazepine medication, which can be used intravenously as an anesthetic for conscious sedation or as an adjunct for general and regional anesthesia. It has a rapid onset and short duration of action, and it causes relatively mild cardiovascular and respiratory effects. Previous in vitro and in vivo studies have shown an anti-tumorigenic property of midazolam [90,121,122,123,124]. However, midazolam is reported to provide distinct effects on various types of innate immune cells [63,124,125]. Hence, further findings from prospective studies are necessary before valid conclusions about the effects of perioperative midazolam use on the prognosis of cancer patients can be made.

2.6.1. Laboratory Research

Basic research on the impact of midazolam on the release of TNF-α in cancer cell biology is limited (Table 1). Kang et al. conducted an in vitro and in vivo study and reported that midazolam could reduce the progression of hepatocellular carcinoma by inhibiting the NF-κB pathway and influence the immune microenvironment with a decrease in the secretion of TNF-α [124]. However, midazolam inhibited the growth of murine myeloid leukemia cell lines with an upregulated expression of TNF-α in a dose-dependent manner [126].

2.6.2. Clinical Studies

Anesthetic induction with a higher dosage of midazolam in patients undergoing thoracoscopic resections of lung cancer was reported to provide better inhibition of the production of serum TNF-α and remarkably decrease the requirement for anesthetics, which may stabilize the hemodynamics of patients in the perioperative period and mitigate the severity of postoperative pain [127]. However, a retrospective analysis revealed that midazolam anesthesia was associated with less stable hemodynamics and an increased level of serum TNF-α than DEX anesthesia in patients undergoing radical resections of ovarian cancer [106]. Therefore, the role of midazolam in the TNF-α release and cancer progression is currently questionable and further investigations should be conducted (Table 2; Figure 2).

2.7. Thiopental

Thiopental is a rapid-onset and short-acting barbiturate agent, which is widely used in the treatment of seizure disorders and the induction of GA. Experimental studies have demonstrated that thiopental can enhance the aggressiveness of cancer cells [64,75]. Notably, thiopental affects immune cells to different extents. Thiopental is reported to suppress NK cell activity and impair neutrophil function [64,125] while protecting T lymphocytes from apoptosis [128]. Overall, we lack the evidence required to make recommendations about the perioperative use of thiopental in improving the prognosis of surgical cancer patients.

2.7.1. Laboratory Research

Laboratory research on the impact of thiopental administration on TNF-α production in cancer cell biology is limited (Table 1). Using LPS-treated human glioma cell lines and murine brain inflammation models, thiopental was reported to exert anti-inflammatory effects with the suppression of TNF-α production by inhibiting NF-κB pathway activation [129].

2.7.2. Clinical Studies

To the best of our knowledge, there is no clinical study on the relationship among thiopental, TNF-α, and cancer outcomes, which may contribute to the limited use of thiopental for anesthetic induction (Table 2).

2.8. Opioids

Opioids are often used in cancer patients for acute perioperative and chronic pain management. It is believed that opioids exert their influence on tumor growth and progression, but the role of opioids in cancer dissemination is conflicting [130,131,132,133,134,135,136]. Moreover, there is evidence that not all opioids have the same effect on immunity [137]. Previous prospective trials have failed to detect any correlation between opioid prescription and cancer progression [138,139]. However, results from retrospective studies exhibit a negative association between opioid exposure and survival [140,141,142]. The current state of knowledge suggests that pain is related to poor outcomes in cancer patients, but opioids should be judiciously prescribed due to their potential impact on cancer development [143]. Further prospective studies are needed to clarify possible effects of perioperative opioid administration on cancer prognosis.

2.8.1. Laboratory Research

Using human cancer cell lines, morphine could dose-dependently suppress tumor cell growth via the inhibition of TNF-α release [144], which may be associated with reduced NF-κB pathway activation [145]. Fentanyl administration in human astrocytoma cell lines was also shown to inhibit TNF-α-induced chemokine expression, which may thereby be protective in response to inflammation-mediated neuropathogenesis [146]. However, Bastami et al. conducted an in vitro study and reported that various opioids had diverse influences on the release of TNF-α in LPS-treated human histiocytic lymphoma cell lines, in which tramadol and morphine had an inhibitory effect on TNF-α release and fentanyl had no effect [147]. In mouse models of breast cancer, tramadol attenuated tumor growth and hormone receptor expression, reduced the serum level of TNF-α, and preserved the NK cell activity compared with morphine [148]. Interestingly, Ma et al. declared that, when using mouse models of colorectal cancer, low-dose naltrexone (an opioid antagonist) decreased proliferation and promoted the apoptosis of cancer cells with increased TNF-α expression [149]. Results from laboratory research about the effects of opioids on the TNF-α release and cancer cell biology are inconclusive, and further investigations are required (Table 1).

2.8.2. Clinical Studies

Compared with fentanyl, remifentanil administration during laparoscopic surgery for colon cancer was reported to significantly alleviate inflammatory responses with reduced secretion of serum TNF-α, improve oxidative stress indices, and decrease the frequency of adverse reactions [150]. In addition, oxycodone combined with flurbiprofen axetil applied for IVPCA could effectively reduce pain intensity, reverse immunosuppression, inhibit the release of serum TNF-α, and enhance postoperative intestinal recovery compared with sufentanil combined with flurbiprofen axetil in patients undergoing radical resections of colorectal cancer [151]. During thoracoscopic lobectomies for lung cancer, nalbuphine treatment prior to the induction of anesthesia also provided a significant analgesic effect, reduced the incidence of adverse reactions, and alleviated postoperative inflammatory responses with a reduced serum level of TNF-α [152]. However, Titon et al. demonstrated that opioid-free anesthesia may influence acute inflammation through a reduction of plasma interleukin (IL)-12 rather than TNF-α and other pro-inflammatory cytokines in the perioperative period of patients undergoing oncological surgery [153]. In summary, whether opioids possess an immunomodulatory capability and whether different types of opioids exert distinct effects on the release of TNF-α and subsequent cancer outcomes have been inconclusive thus far (Table 2; Figure 2).

2.9. Nonsteroidal Anti-Inflammatory Drugs

Nonsteroidal anti-inflammatory drugs (NSAIDs) play an essential role in multimodal analgesia during the perioperative period, and their main mechanism of action is the inhibition of cyclooxygenase. NSAIDs provide inhibitory effects on the development, growth, and invasion of human cancer cells [154,155,156,157]. The influence of NSAID treatment on inflammatory TME and cancer immunity is also becoming clearer [6,158]. However, retrospective data have shown an uncertain relationship between the perioperative use of NSAIDs and oncological outcomes in patients undergoing different types of cancer surgery [159,160,161]. Therefore, the current evidence is insufficient to encourage the routine use of NSAIDs in surgical cancer patients, and further prospective studies should be conducted.

2.9.1. Laboratory Research

NSAID administration in vitro was reported to enhance TNF-α-mediated and TRAIL-induced cell death in different cancer cells through various mechanisms [162,163,164,165,166,167], of which aspirin and ibuprofen may be the least potent, while celecoxib and tamoxifen may be the most potent anti-inflammatory and anti-proliferative agents [168]. In addition, NSAIDs effectively inhibited the TNF-α-induced upregulation of cancer cell migration [166,169]. Setia et al. revealed that celecoxib administration prevented colitis-mediated colon carcinogenesis through the downregulation of inflammation with a decreased release of TNF-α and inactivation of the NF-κB pathway in mouse models [170]. NSAIDs in vivo could also reduce tumor growth via TNF-α-mediated and TRAIL-induced apoptosis [162,165,167,171]. In contrast, Vaish et al. reported that NSAIDs exerted anti-inflammatory and anti-neoplastic actions to reverse inflammation and carcinogenesis by upregulating the expression of TNF-α in rat models of colorectal cancer [172]. Moreover, Axiak-Bechtel et al. conducted a dog model of osteosarcoma and declared that NSAID use did not affect the stimulated production of TNF-α [173]. Based on the published data, further investigations are required to determine the effects of NSAIDs on the TNF-α release and cancer cell biology (Table 1).

2.9.2. Clinical Studies

In clinical cancer surgical settings, the effect of perioperative NSAID prescription for pain management on the release of TNF-α is not fully discussed. Wen et al. demonstrated that postoperative analgesia with flurbiprofen axetil combined with fentanyl provided similar analgesic effects compared with fentanyl alone but significantly decreased the serum concentration of TNF-α in female patients undergoing breast cancer surgery [174]. Notably, preincisional parecoxib administration, compared with postincisional administration, was reported to reduce postoperative morphine consumption and attenuate serum IL-6 production rather than TNF-α after surgery but did not affect morphine-related adverse effects for patients undergoing colorectal cancer surgery [175]. Overall, NSAIDs seem to have beneficial effects on cancer progression through their anti-inflammatory actions, but further research discussing the relationship among NSAIDs, TNF-α, and cancer prognosis should be executed (Table 2; Figure 2).

3. Regional Anesthesia/Analgesia

3.1. Regional Anesthesia/Analgesia

Regional anesthesia/analgesia (RA), used either alone or in combination with GA, can reduce perioperative pain as well as opioid requirements and attenuate surgery-induced stress responses [4]. The regional techniques include the neuraxial block (EA or spinal anesthesia [SA]) and peripheral nerve block. Previous in vitro studies have shown a beneficial effect of RA in inhibiting the malignant potential of tumor cells [176,177]. Furthermore, the anti-inflammatory and analgesic effects of RA suggest a theoretical framework in which the perioperative immunity is optimally preserved [137]. There are observational studies showing better outcomes in patients receiving RA during cancer surgery [178,179], but prospective studies and meta-analyses reveal no significant benefit of RA in oncological outcomes [138,180,181,182,183,184]. Thus, RA may be reasonable for optimizing cancer patients’ comfort, but it does not seem to improve their outcomes based on controversial evidence.

3.1.1. Laboratory Research

Research on the effect of RA on the release of TNF-α and cancer outcomes in experimental animal models is limited (Table 1). Inoue et al. recently conducted an in vivo study, which reported that GA combined with SA may decrease the total number of circulating tumor cells and reduce the stress response to surgery with a decreased level of serum TNF-α compared with GA alone in mouse models of prostate cancer [185].

3.1.2. Clinical Studies

The thoracic paravertebral block was associated with lower incidence of postoperative delirium in elderly lung cancer patients undergoing thoracoscopic lobectomies compared with IVPCA, which may result from its opioid-sparing and anti-neuroinflammatory effects with a decreased surgery-induced production of TNF-α and neurofilament light [186]. Furthermore, the continuous wound infusion of local anesthetics significantly alleviated systemic inflammation with a reduced level of plasma TNF-α, decreased pain scores and opioid intake, and accelerated the recovery of respiratory function in patients of lung cancer resections [187]. During gastric cancer surgery, EA combined with GA could also preserve innate tumor immunity and mitigate stress responses with a reduced plasma concentration of TNF-α compared with GA alone [188,189]. In elderly patients undergoing liver cancer surgery, the addition of EA in GA similarly improved postoperative recovery and cognitive function and relieved inflammatory responses with a lower plasma level of TNF-α [190]. Recently, Geng et al. reported that a combination of a pectoral nerve block and stellate ganglion block effectively blunted perioperative inflammatory responses with a decreased level of serum TNF-α, alleviated acute postoperative pain, stabilized perioperative hemodynamics, and provided better postoperative sleep quality than a pectoral nerve block alone in breast cancer patients undergoing modified radical mastectomy [191]. Nevertheless, Okuda et al. demonstrated that thoracic EA during lung cancer surgery with OLV could attenuate local inflammation by decreasing the production of IL-6 rather than TNF-α [192]. A combination of thoracic paravertebral block or EA and GA was also reported to decrease pain scores, reduce the use of opioids and vasoactive agents, and improve perioperative immune function and long-term outcomes in patients undergoing esophageal cancer surgery, but it did not affect the release of serum TNF-α [193]. Karadeniz et al. similarly reported that GA combined with EA plus patient-controlled epidural analgesia provided lower intraoperative opioid consumption and shorter hospital stay in comparison with GA plus IVPCA in bladder cancer patients undergoing radical cystectomy, but it had no effect on the serum level of TNF-α [194]. Additionally, during radical prostatectomies, thoracic EA could significantly blunt the early neuroendocrine response to surgery by preventing an increase in plasma cortisol and glucose levels and reducing postoperative pain without affecting the secretion of plasma TNF-α [195]. Notably, Siekmann et al. reported that surgical and analgesic techniques had no impact on the postoperative release of plasma TNF-α following colorectal cancer surgery [196]. In summary, RA seems to provide beneficial effects on surgical cancer patients, but further prospective trials are needed to investigate the impacts of RA on cancer progression by modulating perioperative TNF-α release (Table 2; Figure 2).

4. Perioperative Care

4.1. Body Temperature

Body temperature control is crucial for optimal metabolism and physiological processes, especially during surgery. Perioperative hypothermia may lead to detrimental complications and contribute to perioperative morbidity and mortality [197]. In addition, perioperative hypothermia is reported to exacerbate surgery-induced immunosuppression, which may worsen prognoses in cancer patients [198,199,200]. However, an in vivo study has demonstrated that hypothermia appears to have no significant effects on NK activity and resultant tumor metastasis [64]. Although there is no sufficient evidence elucidating the influence of hypothermia on cancer progression, maintaining normothermia during cancer surgery is still strongly recommended.

4.1.1. Laboratory Research

The effect of body temperature on the TNF-α release in cancer cell biology has been the subject of debate (Table 1). Du et al. reported that hypothermia in vitro and in vivo activated adipocytes to stimulate lung cancer progression by increasing the release of TNF-α [201]. However, mild hyperthermia significantly enhanced the efficacy of immunotherapy and reduced the risk of cancer metastasis in mouse models of pancreatic cancer, which may contribute to increasing drug accumulation and improving the anti-tumor immune activity with increased secretion of TNF-α [202].

4.1.2. Clinical Studies

Preoperative hyperthermia could improve the immune responses to surgical stress by decreasing the plasma concentration of TNF-α without enhancing the quality of recovery after colorectal cancer surgery [203]. In contrast, Atanackovic et al. found that performing hyperthermia showed a redistribution of innate immune cells with an increase in serum TNF-α in patients with solid cancers [204]. During cytoreductive surgery and hyperthermic intraperitoneal chemotherapy, Coccolini et al. reported that serum and peritoneal concentrations of TNF-α did not change significantly throughout the whole procedure [205]. Furthermore, warmed and humidified carbon dioxide (CO2) insufflation was more effective than standard CO2 insufflation in maintaining patients’ heat homeostasis, but it showed no benefit in terms of pain scores and plasma TNF-α and other cytokine levels during robotic radical prostatectomies [206]. As for hypothermia, Hansen et al. did not detect a significant difference in the serum level of TNF-α in patients with perioperative moderate hypothermia scheduled for the resection of malignant melanoma [207]. Consequently, although maintaining normothermia during cancer surgery is recommended, the effect of body temperature control on the release of TNF-α and cancer outcomes warrants further investigations (Table 2; Figure 2).

4.2. Hyperglycemia

Hyperglycemia is associated with increased postoperative complications in different types of surgery, including oncological resections. Perioperative hyperglycemia can enhance surgery-induced immunosuppression, which may increase patients’ vulnerability to postoperative wound infection and potential tumor dissemination [199,208]. Moreover, the hyperglycemic condition can induce the activation of signaling pathways in cancer progression and confer resistance against chemotherapy [209]. As a result, perioperative hyperglycemia in surgical cancer patients should be avoided, and further studies are warranted to confirm the association between perioperative hyperglycemia and tumor spread or metastasis.

4.2.1. Laboratory Research

Laboratory research on the impact of hyperglycemia on TNF-α production in cancer cell biology is limited (Table 1). Otto et al. conducted an in vitro study and showed that the interaction between hyperglycemia and macrophages could promote the expression of TNF-α, as well as epithelial–mesenchymal transition transcription factors in pancreatic ductal epithelial cells, which may aggravate malignancy-associated alterations [210].
Table 1. Experimental studies on the effects of anesthetics/analgesics and perioperative care on TNF-α release and cancer cell biology.
Table 1. Experimental studies on the effects of anesthetics/analgesics and perioperative care on TNF-α release and cancer cell biology.
Anesthetics/Analgesics and Perioperative ManagementTNF-α ReleaseEffects
VAsNot applicableThere is no relevant experimental study.
PropofolDecreasedPropofol ameliorated oxygen-glucose deprivation/reperfusion-induced inflammation in mouse neuroblastoma cell lines [67].
IncreasedPropofol suppressed the proliferation and killing activity of anti-tumor immune cells during tumor resection in murine models of breast cancer [68].
KetamineDecreasedKetamine attenuated LPS-induced inflammatory responses in mouse neuroblastoma cell lines [79].
DEXDecreasedDEX improved the immune function and decreased the tumor invasion and migration in rat models of ovarian cancer [92]; DEX enhanced the immune function and suppressed the tumor growth in rat models of ovarian cancer [93]; DEX suppressed surgical stress responses, sped up the recovery of NK cell activity, and lowered the tumor burden after surgery in mouse models of ovarian cancer [94]; DEX attenuated oxygen-glucose deprivation/reperfusion-induced inflammation and apoptosis in rat pheochromocytoma cell lines [95].
LidocaineNo differenceLidocaine contributed to tumor reduction but had no influence on tumor-induced inflammatory responses [117].
MidazolamDecreasedMidazolam reduced the progression of hepatocellular carcinoma cell lines and influenced the immune microenvironment in mouse models [124].
IncreasedMidazolam inhibited the growth and induced the differentiation of murine myeloid leukemia cell lines [126].
ThiopentalDecreasedThiopental exerted anti-inflammatory effects on LPS-treated human glioma cell lines and murine brain inflammation models [129].
OpioidsDecreasedMorphine dose-dependently suppressed tumor cell growth in human cancer cell lines [144]; morphine and its derivatives induced the apoptosis of human cancer cell lines [145]; tramadol and morphine inhibited cytokine release in LPS-treated human histiocytic lymphoma cell lines [147].
IncreasedNaltrexone decreased proliferation and promoted apoptosis in mouse models of colorectal cancer [149].
No differenceFentanyl had no effect on the production of cytokines in LPS-treated human histiocytic lymphoma cell lines [147].
NSAIDsDecreasedCelecoxib downregulated inflammation and prevented against colitis-mediated colon carcinogenesis in mouse models [170].
IncreasedSulindac and celecoxib exerted anti-inflammatory and anti-neoplastic actions in rat models of colorectal cancer [172].
No differenceNSAIDs had no influence on cytokine release in dog models of osteosarcoma [173].
RADecreasedThe combination of GA and SA reduced stress responses to surgery and attenuated the suppression of innate tumor immunity compared with GA alone in mouse models of prostate cancer [185].
Body temperatureIncreasedHypothermia stimulated lung cancer boost [201]; mild hyperthermia enhanced the efficacy of immunotherapy and reduced the risk of cancer metastasis in mouse models of pancreatic cancer [202].
HyperglycemiaIncreasedThe interaction between hyperglycemia and macrophages promoted malignancy-associated alterations in pancreatic ductal epithelial cells [210].
Blood transfusionNot applicableThere is no relevant experimental study.
DEX = dexmedetomidine; GA = general anesthesia; LPS = lipopolysaccharide; NK cell = nature killer cell; NSAID = non-steroidal anti-inflammatory drug; SA = spinal anesthesia; TNF-α = tumor necrosis factor alpha; VA = volatile anesthetic; RA = regional anesthesia/analgesia.

4.2.2. Clinical Studies

The degree of perioperative glucose fluctuation was positively associated with the postoperative level of serum TNF-α and poor prognosis in patients with supratentorial neoplasms undergoing intracranial excision [211]. Notably, perioperative hyperglycemia was shown to attenuate postoperative immune activation with a significant suppression of serum TNF-α release in cancer patients of esophageal or pancreatic resections [212]. Collectively, the limited data show that the glucose fluctuation and hyperglycemia may affect the TNF-α expression and subsequent cancer progression, which should be clarified through further investigations (Table 2; Figure 2).

4.3. Blood Transfusion

Blood transfusion is a commonly used adjunctive therapy for surgical diseases. Cancer patients often suffer from anemia prior to operation and require a blood transfusion during the perioperative period. However, blood transfusion can modulate the immune responses and may increase the incidence of cancer recurrence and postoperative infection [198,199]. Available clinical data summarized in meta-analyses show that blood transfusion seems to enhance the risks of postoperative complications, cancer recurrence, and worse patient survival [213,214,215,216]. Taken together, although more aggressive types of cancer may need more requirements for blood transfusion during surgery, using a restrictive threshold of blood transfusion in cancer patients should be mandatory.

4.3.1. Laboratory Research

To the best of our knowledge, there is no experimental study about the relationship among blood transfusion, TNF-α release, and cancer cell biology (Table 1).

4.3.2. Clinical Studies

During colorectal cancer surgery, patients who received an allogenic red blood cell (RBC) transfusion had a significantly increased concentration of plasma TNF-α after surgery compared to those who did not [217]. Surinenaite et al. also showed that the serum concentration of TNF-α was increased in RBC-transfused patients with advanced colorectal cancer after surgical treatment but did not significantly change in those with early colorectal cancer [218]. Compared with homologous RBC transfusion, autologous RBC transfusion was reported to alleviate transfusion-related immunosuppression by increasing the plasma TNF-α level in esophageal and colorectal cancer patients undergoing radical resections [219,220]. In addition, Geng et al. reported that, compared with allogeneic suspended RBC transfusion, allogeneic leukocyte-depleted RBC transfusion could effectively alleviate inflammation with a less decreased level of serum TNF-α, improve coagulation function, reduce stress response, and enhance wound healing without increasing adverse reactions or the postoperative infection rate in patients with recurrences of colon cancer after operation [221]. However, Benson et al. revealed that, although stored leukocyte-depleted or non-leukocyte-depleted packed RBC both contained tumorigenic mediators that may enhance tumor progression, their TNF-α levels did not differ [222]. Beck-Schimmer et al. similarly demonstrated that the retransfusion of irradiated intraoperative cell salvage blood did not upregulate the concentrations of serum TNF-α and other inflammatory mediators in patients undergoing gynecological cancer surgery [223]. Interestingly, a low plasma TNF-α level before surgery was associated with a high perioperative transfusion rate in glioma surgical patients, which may indicate that the preoperative immune response could influence perioperative transfusion requirements [224]. In summation, according to the available data, perioperative RBC transfusion seems to influence the release of TNF-α and even cancer prognosis; however, further prospective trials are needed to build definitive evidence (Table 2; Figure 2).
Table 2. Clinical studies on the effects of anesthetics/analgesics and perioperative care on TNF-α release and clinical outcomes.
Table 2. Clinical studies on the effects of anesthetics/analgesics and perioperative care on TNF-α release and clinical outcomes.
Anesthetics/Analgesics and Perioperative ManagementTNF-α ReleaseEffects
VAsDecreasedSevoflurane improved hemodynamics and inflammatory responses in lung lobectomies but increased the incidence of postoperative complications compared with propofol [43]; isoflurane decreased inflammatory responses associated with OLV during open thoracic cancer surgery and had better postoperative outcomes compared with propofol [44].
IncreasedSevoflurane exacerbated the injury to pulmonary function in lung cancer surgery compared with propofol [45]; sevoflurane increased the incidence of POCD after esophageal cancer surgery compared with propofol [46].
No differenceSevoflurane had no effect on complement activation and cytokine release in major colorectal cancer surgery compared with propofol [47]; sevoflurane had no effect on NK cell and cytotoxic T lymphocyte counts or the apoptosis rate in breast cancer surgery compared with propofol [48].
PropofolDecreasedPropofol attenuated the injury to pulmonary function in lung cancer surgery compared with sevoflurane [45]; propofol decreased the incidence of POCD after esophageal cancer surgery compared with sevoflurane [46].
IncreasedPropofol enhanced unstable hemodynamics and inflammatory responses in lung lobectomies but decreased the incidence of postoperative complications compared with sevoflurane [43]; propofol increased inflammatory responses associated with OLV during open thoracic cancer surgery and had worse postoperative outcomes compared with isoflurane [44].
No differencePropofol had no effect on complement activation and cytokine release in major colorectal surgery compared with sevoflurane [47]; propofol had no effect on NK cell and cytotoxic T lymphocyte counts or the apoptosis rate in breast cancer surgery compared with sevoflurane [48].
KetamineDecreasedKetamine suppressed pro-inflammatory cytokine production in radical prostatectomies [80]; ketamine prevented brain injury and improved postoperative fatigue syndrome after laparoscopic colorectal cancer surgery [81]; ketamine reduced inflammatory responses and improved postoperative anxiety and depression after colorectal cancer surgery [82].
No differenceKetamine in colorectal cancer surgery had no impact on postoperative NK cell activity, inflammatory responses, or long-term prognosis [77]; ketamine had no direct immunomodulation during minimally invasive robotic radical prostatectomies [78].
DEXDecreasedDEX reduced the oxidative stress and inflammation in lung lobectomies with OLV [97]; DEX in GA provided better hemodynamic control, dismissed inflammatory responses, protected intestinal function, and shortened the length of hospital stay after laparoscopic radical gastrectomies compared with GA plus EA [98]; DEX attenuated systemic inflammation and ameliorated the impaired immune function in radical gastric cancer surgery [99]; DEX improved postoperative cognitive function after laparoscopic colon cancer surgery [100]; DEX reduced adverse responses and the occurrence of POCD after laparoscopic total hysterectomies [101]; DEX reduced postoperative inflammation and promoted the recovery of postoperative cognitive function after gastric cancer surgery [102]; DEX, during esophageal cancer surgery, alleviated the incidence of POCD [103]; DEX provided better cardiocerebral protection in colorectal cancer surgery [104]; DEX stabilized hemodynamics, reduced inflammation, and inhibited free radical generation in glioma surgery [105]; DEX stabilized hemodynamics and alleviated stress responses in ovarian cancer surgery compared with midazolam [106].
LidocaineDecreasedLidocaine, during supratentorial tumor resections, improved early recovery quality and had brain-injury alleviation effects [118]; lidocaine improved postoperative recovery, alleviated inflammation and immunosuppression, and accelerated the return of bowel function after laparoscopic radical gastrectomies [119].
No differenceLidocaine accelerated the return of bowel function and shortened the length of hospital stay after colorectal surgery [120].
MidazolamDecreasedMidazolam, during thoracoscopic lung cancer surgery, inhibited inflammatory responses, decreased the requirement of anesthetics, stabilized perioperative hemodynamics, and mitigated postoperative pain [127].
IncreasedMidazolam enhanced unstable hemodynamics and stress responses in ovarian cancer surgery compared with DEX [106].
ThiopentalNot applicableThere is no relevant clinical study.
OpioidsDecreasedNalbuphine enhanced analgesic effects, reduced the incidence of adverse reactions, and alleviated postoperative inflammatory responses in thoracoscopic lung lobectomies [152].
No differenceOpioid-free anesthesia influenced acute inflammation in the perioperative period of oncological surgery [153].
NSAIDsDecreasedFlurbiprofen axetil plus fentanyl provided similar postoperative analgesia in breast cancer surgery compared with fentanyl alone [174].
RADecreasedThoracic paravertebral blocks decreased the incidence of postoperative delirium and enhanced pain control and recovery quality compared with IVPCA in thoracoscopic lung lobectomies [186]; continuous wound analgesia alleviated systemic inflammation, decreased pain scores and opioid intake, and accelerated the recovery of respiratory function in lung cancer surgery [187]; EA, during gastric cancer surgery, mitigated inflammatory responses, preserved innate tumor immunity, and decreased the incidence of postoperative adverse reactions [188]; EA, during gastric cancer surgery, reduced the stress reactions and maintained the integrity of immune function [189]; EA improved postoperative recovery and cognitive function and relieved inflammatory responses in liver cancer surgery [190].
No differenceThoracic EA attenuated local inflammation but did not affect systemic inflammation during lung cancer surgery [192]; thoracic paravertebral blocks or EA decreased pain scores, reduced the use of opioids and vasoactive agents, and improved perioperative immune function and postoperative survival after esophageal cancer surgery [193]; GA plus EA and patient-controlled epidural analgesia provided lower opioid consumption and shorter hospital stay in radical cystectomies compared with GA plus IVPCA [194]; thoracic EA blunted early postoperative stress responses and reduced postoperative pain after radical prostatectomies [195]; surgical rather than analgesic techniques had a great impact on postoperative inflammation in colorectal cancer surgery [196].
Body temperatureDecreasedPreoperative hyperthermia improved immune responses to surgical stress but did not enhance the quality of recovery after colorectal cancer surgery [203].
IncreasedCancer patients treated with hyperthermia had a redistribution of innate immune cells [204].
No differenceThe cytokine levels paralleled hemodynamic and metabolic derangements in the hyperthermic phase of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy [205]; warmed and humidified CO2 insufflation had no benefit in terms of pain scores and cytokine levels during robotic radical prostatectomies [206]; moderate hypothermia during malignant melanoma resection affected the release of circulating cytokines and adhesion molecules [207].
HyperglycemiaDecreasedPerioperative hyperglycemia attenuated postoperative immune activation during esophageal or pancreatic cancer resections [212].
IncreasedThe degree of perioperative glucose fluctuation was positively related to postoperative cytokine levels and poor short-term prognosis after supratentorial tumor resection [211].
Blood transfusionIncreasedAllogenic RBC transfusion increased the serum levels of cytokines in colorectal cancer surgery [217]; RBC transfusion in advanced colorectal cancer surgery exacerbated the antioxidative and immune systems and increased serum cytokine levels [218].
CO2 = carbon dioxide; DEX = dexmedetomidine; EA = epidural anesthesia; GA = general anesthesia; IVPCA = intravenous patient-controlled analgesia; NK cell = nature killer cell; NSAID = non-steroidal anti-inflammatory drug; OLV = one-lung ventilation; POCD = postoperative cognitive dysfunction; TNF-α = tumor necrosis factor alpha; VA = volatile anesthetic; RA = regional anesthesia/analgesia; RBC = red blood cell.

5. TNF-α in Cancer

TNF-α is reported to be associated with inflammation-associated carcinogenesis. It is mainly secreted by immune cells such as monocytes, macrophages, NK cells, T lymphocytes, mast cells, and neutrophils, but can also be produced by non-immune cells such as endothelial cells, adipocytes, neurons, fibroblasts, and smooth muscle [14]. Additionally, TNF-α not only induces its own secretion, but also stimulates the production of other inflammatory cytokines and chemokines [12]. TNF-α and its receptors exert biological functions in cancer cells by activating distinct signaling pathways, including NF-κB and c-Jun N-terminal kinase (JNK), in which NF-κB is an anti-apoptotic signal while sustained JNK activation contributes to cell death [225]. The crosstalk between the NF-κB and JNK signaling is involved in determining cellular outcomes in response to TNF-α, which depends on cytokine concentrations. In high concentrations, TNF-α provides an anti-tumor action by mediating cellular apoptosis, directing tumor-associated macrophages to anti-tumoral phenotypes, guiding neutrophils and monocytes to tumor sites, activating macrophages and inhibiting monocyte differentiation to immunosuppressive phenotypes, and inducing the disruption of tumor vasculature; in contrast, TNF-α expression at low levels can be pro-tumorigenic [14].
The TME is a complex biological ecosystem of solid tumors. As mentioned above, anesthetics/analgesic techniques and perioperative management may affect the release of TNF-α, but there are inconsistent results. The potential causes include the following: first, TNF-α has been described as having dual effects on almost every type of cancer [14]; second, more detailed and complicated mechanisms about the effects of TNF-α on cancer cells beyond the NF-κB and JNK signaling pathways are increasingly elucidated [13,14]; third, perioperative anesthetic strategies may have impacts to different extents on various immune and non-immune cells that can produce TNF-α; and last, differences in clinical surgical settings, including cancer types, patient conditions, and surgical procedures, probably confound the study consequences. Due to these reasons, the effects of anesthesia on TNF-α release and resultant cancer outcomes eventually become minimal or none.

6. Conclusions

Cancer remains the leading cause of death worldwide, and surgical resection is currently the mainstay treatment for solid tumors. However, tumor recurrence and/or metastasis after surgical resection can lead to morbidities and mortalities in most cancer patients. During surgery, perioperative anesthetic strategies may alter the TME and even affect cancer outcomes. Herein, we endeavor to summarize the role of anesthesia in cancers from the insights of TNF-α release and subsequent cancer progression. The scarce evidence from the available data suggests that patients maintained on VA-based anesthesia or propofol-based TIVA during cancer surgery have no clear significance in the TNF-α release and patient outcomes, but adjunctive agents including ketamine, DEX, systemic lidocaine, and opioids appear to have a beneficial effect on the release of TNF-α and may have a tendency towards the suppression of carcinogenesis. Additionally, the addition of RA and the reduction of blood transfusion provide immunomodulatory effects in cancer patients undergoing surgical resections. Currently, it remains challenging to recommend specific anesthetics/analgesic techniques or perioperative management during cancer surgery for optimal risk reduction with regard to tumor recurrence or metastasis. Therefore, further in-depth experimental research and high-quality clinical trials are needed to explore the detailed molecular mechanisms involved in the relationship between anesthesia, TNF-α release, and cancer progression.

Author Contributions

Conceptualization, W.-C.T. and Z.-F.W.; methodology, H.-C.L.; software, H.-C.L.; validation, Y.-H.H. and S.-M.C.; formal analysis, Y.-H.H. and S.-M.C.; investigation, W.-C.T.; resources, Z.-F.W.; data curation, Y.-H.H. and S.-M.C.; writing—original draft preparation, W.-C.T.; writing—review and editing, Z.-F.W.; visualization, H.-C.L.; supervision, Z.-F.W.; project administration, W.-C.T.; funding acquisition, none. 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. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
  2. Yuki, K. The Role of General Anesthetic Drug Selection in Cancer Outcome. Biomed Res. Int. 2021, 2021, 2563093. [Google Scholar] [CrossRef] [PubMed]
  3. Tang, F.; Tie, Y.; Tu, C.; Wei, X. Surgical trauma-induced immunosuppression in cancer: Recent advances and the potential therapies. Clin. Transl. Med. 2020, 10, 199–223. [Google Scholar] [CrossRef]
  4. Dubowitz, J.A.; Sloan, E.K.; Riedel, B.J. Implicating anaesthesia and the perioperative period in cancer recurrence and metastasis. Clin. Exp. Metastasis 2018, 35, 347–358. [Google Scholar] [CrossRef] [PubMed]
  5. Chen, Z.; Zhang, P.; Xu, Y.; Yan, J.; Liu, Z.; Lau, W.B.; Lau, B.; Li, Y.; Zhao, X.; Wei, Y.; et al. Surgical stress and cancer progression: The twisted tango. Mol. Cancer 2019, 18, 132. [Google Scholar] [CrossRef] [Green Version]
  6. Kim, R. Effects of surgery and anesthetic choice on immunosuppression and cancer recurrence. J. Transl. Med. 2018, 16, 8. [Google Scholar] [CrossRef] [Green Version]
  7. Anderson, N.M.; Simon, M.C. The tumor microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef] [PubMed]
  8. Xiao, Y.; Yu, D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol. Ther. 2021, 221, 107753. [Google Scholar] [CrossRef]
  9. Arneth, B. Tumor Microenvironment. Medicina 2019, 56, 15. [Google Scholar] [CrossRef] [Green Version]
  10. Cheng, X.; Zhang, H.; Hamad, A.; Huang, H.; Tsung, A. Surgery-mediated tumor-promoting effects on the immune microenvironment. Semin. Cancer Biol. 2022, 86, 408–419. [Google Scholar] [CrossRef]
  11. Onuma, A.E.; Zhang, H.; Gil, L.; Huang, H.; Tsung, A. Surgical Stress Promotes Tumor Progression: A Focus on the Impact of the Immune Response. J. Clin. Med. 2020, 9, 4096. [Google Scholar] [CrossRef]
  12. Chu, W.M. Tumor necrosis factor. Cancer Lett. 2013, 328, 222–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Laha, D.; Grant, R.; Mishra, P.; Nilubol, N. The Role of Tumor Necrosis Factor in Manipulating the Immunological Response of Tumor Microenvironment. Front. Immunol. 2021, 12, 656908. [Google Scholar] [CrossRef] [PubMed]
  14. Mercogliano, M.F.; Bruni, S.; Mauro, F.; Elizalde, P.V.; Schillaci, R. Harnessing Tumor Necrosis Factor Alpha to Achieve Effective Cancer Immunotherapy. Cancers 2021, 13, 564. [Google Scholar] [CrossRef] [PubMed]
  15. Sha, J.; Du, J.; Yang, J.; Hu, X.; Li, L. Changes of serum levels of tumor necrosis factor (TNF-α) and soluble interleukin-2 receptor (SIL 2R) in patients with cervical cancer and their clinical significance. Am. J. Transl. Res. 2021, 13, 6599–6604. [Google Scholar] [PubMed]
  16. Pawlik, W.; Pawlik, J.; Kozłowski, M.; Łuczkowska, K.; Kwiatkowski, S.; Kwiatkowska, E.; Machaliński, B.; Cymbaluk-Płoska, A. The Clinical Importance of IL-6, IL-8, and TNF-α in Patients with Ovarian Carcinoma and Benign Cystic Lesions. Diagnostics 2021, 11, 1625. [Google Scholar] [CrossRef] [PubMed]
  17. Wieser, V.; Abdel Azim, S.; Sprung, S.; Knoll, K.; Kögl, J.; Hackl, H.; Marth, C.; Zeimet, A.G.; Fiegl, H. TNFα signalling predicts poor prognosis of patients with endometrial cancer. Carcinogenesis 2020, 41, 1065–1073. [Google Scholar] [CrossRef]
  18. Erturk, K.; Tastekin, D.; Serilmez, M.; Bilgin, E.; Bozbey, H.U.; Vatansever, S. Clinical significance of serum interleukin-29, interleukin-32, and tumor necrosis factor alpha levels in patients with gastric cancer. Tumor Biol. 2016, 37, 405–412. [Google Scholar] [CrossRef]
  19. Stanilov, N.; Miteva, L.; Dobreva, Z.; Stanilova, S. Colorectal cancer severity and survival in correlation with tumour necrosis factor-alpha. Biotechnol. Biotechnol. Equip. 2014, 28, 911–917. [Google Scholar] [CrossRef]
  20. Wang, H.; Liu, J.; Hu, X.; Liu, S.; He, B. Prognostic and Therapeutic Values of Tumor Necrosis Factor-Alpha in Hepatocellular Carcinoma. Med. Sci. Monit. 2016, 22, 3694–3704. [Google Scholar] [CrossRef] [Green Version]
  21. Sun, T.Y.; Hsu, C.L.; Lee, M.S.; Yeh, T.T.; Lai, H.C.; Wu, K.L.; Wu, Z.F.; Tseng, W.C. Propofol-based total intravenous anesthesia is associated with better survival than desflurane anesthesia in limb-salvage surgery for osteosarcoma: A retrospective analysis. Medicine 2022, 101, e30840. [Google Scholar] [CrossRef] [PubMed]
  22. Tseng, W.C.; Lee, M.S.; Lin, Y.C.; Lai, H.C.; Yu, M.H.; Wu, K.L.; Wu, Z.F. Propofol-Based Total Intravenous Anesthesia is Associated with Better Survival than Desflurane Anesthesia in Epithelial Ovarian Cancer Surgery: A Retrospective Cohort Study. Front. Pharmacol. 2021, 12, 685265. [Google Scholar] [CrossRef] [PubMed]
  23. Huang, Y.H.; Wu, Z.F.; Lee, M.S.; Lou, Y.S.; Wu, K.L.; Cheng, K.I.; Lai, H.C. Propofol-based total intravenous anesthesia is associated with better survival than desflurane anesthesia in glioblastoma surgery. PLoS ONE 2021, 16, e0255627. [Google Scholar] [CrossRef] [PubMed]
  24. Lai, H.C.; Lee, M.S.; Lin, K.T.; Huang, Y.H.; Chen, J.Y.; Lin, Y.T.; Hung, K.C.; Wu, Z.F. Propofol-based total intravenous anesthesia is associated with better survival than desflurane anesthesia in robot-assisted radical prostatectomy. PLoS ONE 2020, 15, e0230290. [Google Scholar] [CrossRef]
  25. Lai, H.C.; Lee, M.S.; Liu, Y.T.; Lin, K.T.; Hung, K.C.; Chen, J.Y.; Wu, Z.F. Propofol-based intravenous anesthesia is associated with better survival than desflurane anesthesia in pancreatic cancer surgery. PLoS ONE 2020, 15, e0233598. [Google Scholar] [CrossRef] [PubMed]
  26. 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] [PubMed]
  27. Wu, Z.F.; Lee, M.S.; Wong, C.S.; Lu, C.H.; Huang, Y.S.; Lin, K.T.; Lou, Y.S.; Lin, C.; Chang, Y.C.; Lai, H.C. Propofol-based Total Intravenous Anesthesia Is Associated with Better Survival Than Desflurane Anesthesia in Colon Cancer Surgery. Anesthesiology 2018, 129, 932–941. [Google Scholar] [CrossRef]
  28. Chang, C.Y.; Wu, M.Y.; Chien, Y.J.; Su, I.M.; Wang, S.C.; Kao, M.C. Anesthesia and Long-term Oncological Outcomes: A Systematic Review and Meta-analysis. Anesth. Analg. 2021, 132, 623–634. [Google Scholar] [CrossRef]
  29. Lu, N.; Piao, M.H.; Feng, C.S.; Yuan, Y. Isoflurane promotes epithelial-to-mesenchymal transition and metastasis of bladder cancer cells through HIF-1α-β-catenin/Notch1 pathways. Life Sci. 2020, 258, 118154. [Google Scholar] [CrossRef]
  30. 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]
  31. Zhang, W.; Shao, X. Isoflurane Promotes Non-Small Cell Lung Cancer Malignancy by Activating the Akt-Mammalian Target of Rapamycin (mTOR) Signaling Pathway. Med. Sci. Monit. 2016, 22, 4644–4650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. 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]
  33. 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, 4255–4260. [Google Scholar]
  34. Benzonana, L.L.; Perry, N.J.; 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]
  35. 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] [PubMed]
  36. Zhang, C.; Wang, B.; Wang, X.; Sheng, X.; Cui, Y. Sevoflurane inhibits the progression of ovarian cancer through down-regulating stanniocalcin 1 (STC1). Cancer Cell Int. 2019, 19, 339. [Google Scholar] [CrossRef] [Green Version]
  37. 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]
  38. Liang, H.; Yang, C.X.; Zhang, B.; Wang, H.B.; Liu, H.Z.; Lai, X.H.; Liao, M.J.; Zhang, T. Sevoflurane suppresses hypoxia-induced growth and metastasis of lung cancer cells via inhibiting hypoxia-inducible factor-1α. J. Anesth. 2015, 29, 821–830. [Google Scholar] [CrossRef]
  39. Müller-Edenborn, B.; Roth-Zʼgraggen, B.; Bartnicka, K.; Borgeat, A.; Hoos, A.; Borsig, L.; Beck-Schimmer, B. Volatile anesthetics reduce invasion of colorectal cancer cells through down-regulation of matrix metalloproteinase-9. Anesthesiology 2012, 117, 293–301. [Google Scholar] [CrossRef] [Green Version]
  40. Stollings, L.M.; Jia, L.J.; Tang, P.; Dou, H.; Lu, B.; Xu, Y. Immune Modulation by Volatile Anesthetics. Anesthesiology 2016, 125, 399–411. [Google Scholar] [CrossRef] [Green Version]
  41. Yuki, K.; Eckenhoff, R.G. Mechanisms of the Immunological Effects of Volatile Anesthetics: A Review. Anesth. Analg. 2016, 123, 326–335. [Google Scholar] [CrossRef] [Green Version]
  42. Kawaraguchi, Y.; Horikawa, Y.T.; Murphy, A.N.; Murray, F.; Miyanohara, A.; Ali, S.S.; Head, B.P.; Patel, P.M.; Roth, D.M.; Patel, H.H. Volatile anesthetics protect cancer cells against tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis via caveolins. Anesthesiology 2011, 115, 499–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Chen, X.; Li, M.; Zheng, R.; Huang, Q.; Li, Y.; Zhu, Y.; Chen, Z.; Lin, J. Effects of sevoflurane inhalation anesthesia on IL-6, TNF-α and MMP-9 expression and hemodynamics in elderly patients undergoing lobectomy for lung cancer. Cell. Mol. Biol. 2020, 66, 49–53. [Google Scholar] [CrossRef] [PubMed]
  44. Mahmoud, K.; Ammar, A. Immunomodulatory Effects of Anesthetics during Thoracic Surgery. Anesthesiol. Res. Pract. 2011, 2011, 317410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Jin, Y.; Zhao, X.; Li, H.; Wang, Z.; Wang, D. Effects of sevoflurane and propofol on the inflammatory response and pulmonary function of perioperative patients with one-lung ventilation. Exp. Ther. Med. 2013, 6, 781–785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Qiao, Y.; Feng, H.; Zhao, T.; Yan, H.; Zhang, H.; Zhao, X. Postoperative cognitive dysfunction after inhalational anesthesia in elderly patients undergoing major surgery: The influence of anesthetic technique, cerebral injury and systemic inflammation. BMC Anesthesiol. 2015, 15, 154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Kvarnström, A.L.; Sarbinowski, R.T.; Bengtson, J.P.; Jacobsson, L.M.; Bengtsson, A.L. Complement activation and interleukin response in major abdominal surgery. Scand. J. Immunol. 2012, 75, 510–516. [Google Scholar] [CrossRef]
  48. Lim, J.A.; Oh, C.S.; Yoon, T.G.; Lee, J.Y.; Lee, S.H.; Yoo, Y.B.; Yang, J.H.; Kim, S.H. The effect of propofol and sevoflurane on cancer cell, natural killer cell, and cytotoxic T lymphocyte function in patients undergoing breast cancer surgery: An in vitro analysis. BMC Cancer 2018, 18, 159. [Google Scholar] [CrossRef]
  49. Jiang, S.; Liu, Y.; Huang, L.; Zhang, F.; Kang, R. Effects of propofol on cancer development and chemotherapy: Potential mechanisms. Eur. J. Pharmacol. 2018, 831, 46–51. [Google Scholar] [CrossRef]
  50. Wu, Z.; Wang, H.; Shi, Z.A.; Li, Y. Propofol Prevents the Growth, Migration, Invasion, and Glycolysis of Colorectal Cancer Cells by Downregulating Lactate Dehydrogenase Both In Vitro and In Vivo. J. Oncol. 2022, 2022, 8317466. [Google Scholar] [CrossRef]
  51. Sun, Y.; Peng, Y.B.; Ye, L.L.; Ma, L.X.; Zou, M.Y.; Cheng, Z.G. Propofol inhibits proliferation and cisplatin resistance in ovarian cancer cells through regulating the microRNA-374a/forkhead box O1 signaling axis. Mol. Med. Rep. 2020, 21, 1471–1480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Liu, W.Z.; Liu, N. Propofol Inhibits Lung Cancer A549 Cell Growth and Epithelial-Mesenchymal Transition Process by Upregulation of MicroRNA-1284. Oncol. Res. 2018, 27, 1–8. [Google Scholar] [CrossRef] [PubMed]
  53. 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, 10.4238. [Google Scholar] [CrossRef] [PubMed]
  54. Xu, Y.B.; Jiang, W.; Zhao, F.R.; Li, G.; Du, Q.H.; Zhang, M.Y.; Guo, X.G. Propofol suppresses invasion and induces apoptosis of osteosarcoma cell in vitro via downregulation of TGF-β1 expression. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 1430–1435. [Google Scholar]
  55. Wang, Z.T.; Gong, H.Y.; Zheng, F.; Liu, D.J.; Dong, T.L. Propofol suppresses proliferation and invasion of pancreatic cancer cells by upregulating microRNA-133a expression. Genet. Mol. Res. 2015, 14, 7529–7537. [Google Scholar] [CrossRef]
  56. 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]
  57. 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]
  58. Ling, Q.; Wu, S.; Liao, X.; Liu, C.; Chen, Y. Anesthetic propofol enhances cisplatin-sensitivity of non-small cell lung cancer cells through N6-methyladenosine-dependently regulating the miR-486-5p/RAP1-NF-κB axis. BMC Cancer 2022, 22, 765. [Google Scholar] [CrossRef]
  59. Qian, J.; Shen, S.; Chen, W.; Chen, N. Propofol Reversed Hypoxia-Induced Docetaxel Resistance in Prostate Cancer Cells by Preventing Epithelial-Mesenchymal Transition by Inhibiting Hypoxia-Inducible Factor 1α. Biomed Res. Int. 2018, 2018, 4174232. [Google Scholar] [CrossRef] [Green Version]
  60. Li, H.; Lu, Y.; Pang, Y.; Li, M.; Cheng, X.; Chen, J. Propofol enhances the cisplatin-induced apoptosis on cervical cancer cells via EGFR/JAK2/STAT3 pathway. Biomed. Pharmacother. 2017, 86, 324–333. [Google Scholar] [CrossRef]
  61. Du, Q.H.; Xu, Y.B.; Zhang, M.Y.; Yun, P.; He, C.Y. Propofol induces apoptosis and increases gemcitabine sensitivity in pancreatic cancer cells in vitro by inhibition of nuclear factor-κB activity. World J. Gastroenterol. 2013, 19, 5485–5492. [Google Scholar] [CrossRef]
  62. 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]
  63. Kushida, A.; Inada, T.; Shingu, K. Enhancement of antitumor immunity after propofol treatment in mice. Immunopharmacol. Immunotoxicol. 2007, 29, 477–486. [Google Scholar] [CrossRef]
  64. 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. Anesth. Analg. 2003, 97, 1331–1339. [Google Scholar] [CrossRef] [PubMed]
  65. 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. 2017, 37, 841–848. [Google Scholar] [CrossRef] [Green Version]
  66. Zhang, L.; Wang, N.; Zhou, S.; Ye, W.; Jing, G.; Zhang, M. Propofol induces proliferation and invasion of gallbladder cancer cells through activation of Nrf2. J. Exp. Clin. Cancer Res. 2012, 31, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Hu, F.; Jiang, J.; Yu, G.; Zang, H.; Sun, H. Propofol Pretreatment Prevents Oxygen-Glucose Deprivation/Reoxygenation (OGD/R)-induced Inflammation Through Nuclear Transcription Factor κB (NF-κB) Pathway in Neuroblastoma Cells. Curr. Neurovasc. Res. 2020, 17, 27–34. [Google Scholar] [CrossRef] [PubMed]
  68. Yan, R.; Song, T.; Wang, W.; Tian, J.; Ma, X. Immunomodulatory roles of propofol and sevoflurane in murine models of breast cancer. Immunopharmacol. Immunotoxicol. 2022, 1–7. [Google Scholar] [CrossRef]
  69. Li, H.; Liu, W.; Zhang, X.; Wu, F.; Sun, D.; Wang, Z. Ketamine suppresses proliferation and induces ferroptosis and apoptosis of breast cancer cells by targeting KAT5/GPX4 axis. Biochem. Biophys. Res. Commun. 2021, 585, 111–116. [Google Scholar] [CrossRef]
  70. He, G.N.; Bao, N.R.; Wang, S.; Xi, M.; Zhang, T.H.; Chen, F.S. Ketamine Induces Ferroptosis of Liver Cancer Cells by Targeting lncRNA PVT1/miR-214-3p/GPX4. Drug Des. Dev. Ther. 2021, 15, 3965–3978. [Google Scholar] [CrossRef]
  71. Li, T.; Yang, J.; Yang, B.; Zhao, G.; Lin, H.; Liu, Q.; Wang, L.; Wan, Y.; Jiang, H. Ketamine Inhibits Ovarian Cancer Cell Growth by Regulating the lncRNA-PVT1/EZH2/p57 Axis. Front. Genet. 2020, 11, 597467. [Google Scholar] [CrossRef]
  72. Zhao, S.; Shao, L.; Wang, Y.; Meng, Q.; Yu, J. Ketamine exhibits anti-gastric cancer activity via induction of apoptosis and attenuation of PI3K/Akt/mTOR. Arch. Med. Sci. 2020, 16, 1140–1149. [Google Scholar] [CrossRef]
  73. Duan, W.; Hu, J.; Liu, Y. Ketamine inhibits colorectal cancer cells malignant potential via blockage of NMDA receptor. Exp. Mol. Pathol. 2019, 107, 171–178. [Google Scholar] [CrossRef] [PubMed]
  74. Zhou, X.; Zhang, P.; Luo, W.; Zhang, L.; Hu, R.; Sun, Y.; Jiang, H. Ketamine induces apoptosis in lung adenocarcinoma cells by regulating the expression of CD69. Cancer Med. 2018, 7, 788–795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Shapiro, J.; Jersky, J.; Katzav, S.; Feldman, M.; Segal, S. Anesthetic drugs accelerate the progression of postoperative metastases of mouse tumors. J. Clin. Investig. 1981, 68, 678–685. [Google Scholar] [CrossRef] [PubMed]
  76. 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]
  77. Cho, J.S.; Kim, N.Y.; Shim, J.K.; Jun, J.H.; Lee, S.; Kwak, Y.L. The immunomodulatory effect of ketamine in colorectal cancer surgery: A randomized-controlled trial. Can. J. Anaesth. 2021, 68, 683–692. [Google Scholar] [CrossRef]
  78. Kawaguchi, J.; Ota, D.; Niwa, H.; Sugo, Y.; Kushikata, T.; Hirota, K. Immunomodulation by ketamine as an adjunct to total intravenous anesthesia in patients undergoing minimally invasive radical prostatectomy: A randomized pilot trial. Mol. Clin. Oncol. 2020, 13, 203–208. [Google Scholar] [CrossRef]
  79. Yang, C.; Jiang, R.Y.; Shen, J.; Hong, T.; Liu, N.; Ding, L.C.; Wang, D.M.; Chen, L.J.; Xu, B.; Zhu, B. Ketamine attenuates the lipopolysaccharide-induced inflammatory response in cultured N2a cells. Mol. Med. Rep. 2013, 8, 217–220. [Google Scholar] [CrossRef] [Green Version]
  80. Ali, H.M.; Mokhtar, A.M. Effect of Single Compared to Repeated Doses of Intravenous S(+) Ketamine on the Release of Pro-inflammatory Cytokines in Patients Undergoing Radical Prostatectomy. Anesth. Essays Res. 2017, 11, 282–286. [Google Scholar] [CrossRef] [Green Version]
  81. Zhao, L.; Zhang, H.; Cheng, H. Effect of a single sub-dose of ketamine on postoperative fatigue syndrome in colorectal cancer patients undergoing radical laparoscopic surgery: A double-blind, pilot study. J. Affect. Disord. 2022, 312, 146–151. [Google Scholar] [CrossRef]
  82. Ren, Q.; Hua, L.; Zhou, X.; Cheng, Y.; Lu, M.; Zhang, C.; Guo, J.; Xu, H. Effects of a Single Sub-Anesthetic Dose of Ketamine on Postoperative Emotional Responses and Inflammatory Factors in Colorectal Cancer Patients. Front. Pharmacol. 2022, 13, 818822. [Google Scholar] [CrossRef]
  83. Tian, H.; Hou, L.; Xiong, Y.; Cheng, Q. Dexmedetomidine upregulates microRNA-185 to suppress ovarian cancer growth via inhibiting the SOX9/Wnt/β-catenin signaling pathway. Cell Cycle 2021, 20, 765–780. [Google Scholar] [CrossRef]
  84. Xu, B.; Qian, Y.; Hu, C.; Wang, Y.; Gao, H.; Yang, J. Dexmedetomidine upregulates the expression of miR-493-5p, inhibiting growth and inducing the apoptosis of lung adenocarcinoma cells by targeting RASL11B. Biochem. Cell Biol. 2021, 99, 457–464. [Google Scholar] [CrossRef] [PubMed]
  85. Yan, R.; Jin, S.; Liu, H.; Le, C.; Gao, J.; Cheng, J.; Chen, L.; Li, N. Dexmedetomidine inhibits cell malignancy in osteosarcoma cells via miR-520a-3p-YOD1 interactome. Biochem. Biophys. Res. Commun. 2021, 543, 56–64. [Google Scholar] [CrossRef]
  86. 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. Anticancer Drugs 2020, 31, 693–701. [Google Scholar] [CrossRef] [PubMed]
  87. Chi, M.; Shi, X.; Huo, X.; Wu, X.; Zhang, P.; Wang, G. Dexmedetomidine promotes breast cancer cell migration through Rab11-mediated secretion of exosomal TMPRSS2. Ann. Transl. Med. 2020, 8, 531. [Google Scholar] [CrossRef] [PubMed]
  88. Chen, H.Y.; Li, G.H.; Tan, G.C.; Liang, H.; Lai, X.H.; Huang, Q.; Zhong, J.Y. Dexmedetomidine enhances hypoxia-induced cancer cell progression. Exp. Ther. Med. 2019, 18, 4820–4828. [Google Scholar] [CrossRef] [Green Version]
  89. Lavon, H.; Matzner, P.; Benbenishty, A.; Sorski, L.; Rossene, E.; Haldar, R.; Elbaz, E.; Cata, J.P.; 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]
  90. Wang, C.; Datoo, T.; Zhao, H.; Wu, L.; Date, A.; Jiang, C.; Sanders, R.D.; Wang, G.; Bevan, C.; Ma, D. Midazolam and Dexmedetomidine Affect Neuroglioma and Lung Carcinoma Cell Biology In Vitro and In Vivo. Anesthesiology 2018, 129, 1000–1014. [Google Scholar] [CrossRef]
  91. Yuki, K. The immunomodulatory mechanism of dexmedetomidine. Int. Immunopharmacol. 2021, 97, 107709. [Google Scholar] [CrossRef]
  92. Tian, H.; Hou, L.; Xiong, Y.; Cheng, Q.; Huang, J. Effect of Dexmedetomidine-Mediated Insulin-Like Growth Factor 2 (IGF2) Signal Pathway on Immune Function and Invasion and Migration of Cancer Cells in Rats with Ovarian Cancer. Med. Sci. Monit. 2019, 25, 4655–4664. [Google Scholar] [CrossRef]
  93. Cai, Q.H.; Tang, Y.; Fan, S.H.; Zhang, Z.F.; Li, H.; Huang, S.Q.; Wu, D.M.; Lu, J.; Zheng, Y.L. In vivo effects of dexmedetomidine on immune function and tumor growth in rats with ovarian cancer through inhibiting the p38MAPK/NF-κB signaling pathway. Biomed. Pharmacother. 2017, 95, 1830–1837. [Google Scholar] [CrossRef]
  94. Shin, S.; Kim, K.J.; Hwang, H.J.; Noh, S.; Oh, J.E.; Yoo, Y.C. Immunomodulatory Effects of Perioperative Dexmedetomidine in Ovarian Cancer: An In Vitro and Xenograft Mouse Model Study. Front. Oncol. 2021, 11, 722743. [Google Scholar] [CrossRef] [PubMed]
  95. Suo, L.; Wang, M. Dexmedetomidine attenuates oxygen-glucose deprivation/ reperfusion-induced inflammation through the miR-17-5p/ TLR4/ NF-κB axis. BMC Anesthesiol. 2022, 22, 126. [Google Scholar] [CrossRef]
  96. Wang, K.; Wu, M.; Xu, J.; Wu, C.; Zhang, B.; Wang, G.; Ma, D. Effects of dexmedetomidine on perioperative stress, inflammation, and immune function: Systematic review and meta-analysis. Br. J. Anaesth. 2019, 123, 777–794. [Google Scholar] [CrossRef] [PubMed]
  97. Gao, S.; Wang, Y.; Zhao, J.; Su, A. Effects of dexmedetomidine pretreatment on heme oxygenase-1 expression and oxidative stress during one-lung ventilation. Int. J. Clin. Exp. Pathol. 2015, 8, 3144–3149. [Google Scholar] [PubMed]
  98. Guo, L.; Liu, Y.; Wang, M. Effect of Perioperative Dexmedetomidine Anesthesia on Prognosis of Elderly Patients with Gastrointestinal Tumor Surgery. Comput. Math. Methods Med. 2022, 2022, 7889372. [Google Scholar] [CrossRef]
  99. Dong, W.; Chen, M.H.; Yang, Y.H.; Zhang, X.; Huang, M.J.; Yang, X.J.; Wang, H.Z. The effect of dexmedetomidine on expressions of inflammatory factors in patients with radical resection of gastric cancer. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 3510–3515. [Google Scholar] [PubMed]
  100. Ding, M.; Xu, X.; Xia, L.; Cao, Y. Magnetic Resonance Imaging Characteristic Evaluation of Dexmedetomidine on Neurocognitive Dysfunction in Elderly Patients with Colorectal Tumors after Laparoscopic Operation. Comput. Math. Methods Med. 2022, 2022, 1345695. [Google Scholar] [CrossRef]
  101. Huang, H.; Xu, X.; Xiao, Y.; Jia, J. The Influence of Different Dexmedetomidine Doses on Cognitive Function at Early Period of Patients Undergoing Laparoscopic Extensive Total Hysterectomy. J. Healthc. Eng. 2021, 2021, 3531199. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, Z.; Shen, Z.; Wang, H.; Zhang, L.; Dong, R. Effect of dexmedetomidine on the cognitive function of patients undergoing gastric cancer surgery by regulating the PI3K/AKT signaling pathway. Oncol. Lett. 2020, 19, 1151–1156. [Google Scholar] [CrossRef] [Green Version]
  103. Zhang, H.; Wu, Z.; Zhao, X.; Qiao, Y. Role of dexmedetomidine in reducing the incidence of postoperative cognitive dysfunction caused by sevoflurane inhalation anesthesia in elderly patients with esophageal carcinoma. J. Cancer Res. Ther. 2018, 14, 1497–1502. [Google Scholar] [CrossRef] [PubMed]
  104. Yi, X.L.; Wang, J.T.; Chu, C.Q.; Li, Y.X.; Yin, J.H.; Liu, S.L. Cardiocerebral protective effects of dexmedetomidine as anesthetic in colorectal cancer surgery. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 3570–3576. [Google Scholar] [CrossRef]
  105. Luo, X.; Zheng, X.; Huang, H. Protective effects of dexmedetomidine on brain function of glioma patients undergoing craniotomy resection and its underlying mechanism. Clin. Neurol. Neurosurg. 2016, 146, 105–108. [Google Scholar] [CrossRef]
  106. Liu, M.; Yi, Y.; Zhao, M. Effect of dexmedetomidine anesthesia on perioperative levels of TNF-α and IL-6 in patients with ovarian cancer. Oncol. Lett. 2019, 17, 5517–5522. [Google Scholar] [CrossRef] [Green Version]
  107. Tang, C.; Hu, Y.; Zhang, Z.; Wei, Z.; Wang, H.; Geng, Q.; Shi, S.; Wang, S.; Wang, J.; Chai, X. Dexmedetomidine with sufentanil in intravenous patient-controlled analgesia for relief from postoperative pain, inflammation and delirium after esophageal cancer surgery. Biosci. Rep. 2020, 40, BSR20193410. [Google Scholar] [CrossRef]
  108. Miao, Z.; Wu, P.; Wang, J.; Zhou, F.C.; Lin, Y.; Lu, X.Y.; Lv, R.; Hou, Q.H.; Wen, Q.P. Whole-Course Application of Dexmedetomidine Combined with Ketorolac in Nonnarcotic Postoperative Analgesia for Patients with Lung Cancer Undergoing Thoracoscopic Surgery: A Randomized Control Trial. Pain Physician 2020, 23, E185–E193. [Google Scholar] [PubMed]
  109. Song, J.; Wei, N.; Zhang, J.; Wang, G. Effect of dexmedetomidine combined with oxycodone patient-controlled intravenous analgesia on the levels of inflammatory cytokine in patients with rectal cancer. Pak. J. Pharm. Sci. 2019, 32, 1381–1385. [Google Scholar]
  110. Beaussier, M.; Delbos, A.; Maurice-Szamburski, A.; Ecoffey, C.; Mercadal, L. Perioperative Use of Intravenous Lidocaine. Drugs 2018, 78, 1229–1246. [Google Scholar] [CrossRef] [PubMed]
  111. Wang, H.; Zhang, X.; Li, Y.; Li, Y.; Pang, T. Lidocaine hampers colorectal cancer process via circITFG2/miR-1204/SOCS2 axis. Anticancer Drugs 2022, 33, 235–244. [Google Scholar] [CrossRef]
  112. Guan, E.; Liu, H.; Xu, N. Lidocaine Suppresses Gastric Cancer Development Through Circ_ANO5/miR-21-5p/LIFR Axis. Dig. Dis. Sci. 2022, 67, 2244–2256. [Google Scholar] [CrossRef]
  113. Li, B.; Xu, H.; He, C.; Zou, W.; Tu, Y. Lidocaine prevents breast cancer growth by targeting neuronatin to inhibit nerve fibers formation. J. Toxicol. Sci. 2021, 46, 329–339. [Google Scholar] [CrossRef] [PubMed]
  114. 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] [PubMed]
  115. 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]
  116. Wall, T.P.; Buggy, D.J. Perioperative Intravenous Lidocaine and Metastatic Cancer Recurrence—A Narrative Review. Front. Oncol. 2021, 11, 688896. [Google Scholar] [CrossRef]
  117. Ferreira, L.E.N.; Abdalla, H.B.; da Costa, J.P.; de Freitas Domingues, J.S.; Burga-Sánchez, J.; Groppo, F.C.; Volpato, M.C. Effects of 2-Hydroxypropil-Β-Cyclodextrin-Lidocaine on Tumor Growth and Inflammatory Response. Curr. Drug Deliv. 2020, 17, 588–598. [Google Scholar] [CrossRef]
  118. Zhao, K.; Dong, Y.; Su, G.; Wang, Y.; Ji, T.; Wu, N.; Cui, X.; Li, W.; Yang, Y.; Chen, X. Effect of Systemic Lidocaine on Postoperative Early Recovery Quality in Patients Undergoing Supratentorial Tumor Resection. Drug Des. Dev. Ther. 2022, 16, 1171–1181. [Google Scholar] [CrossRef]
  119. Lv, X.; Li, X.; Guo, K.; Li, T.; Yang, Y.; Lu, W.; Wang, S.; Liu, S. Effects of Systemic Lidocaine on Postoperative Recovery Quality and Immune Function in Patients Undergoing Laparoscopic Radical Gastrectomy. Drug Des. Dev. Ther. 2021, 15, 1861–1872. [Google Scholar] [CrossRef]
  120. Herroeder, S.; Pecher, S.; Schönherr, M.E.; Kaulitz, G.; Hahnenkamp, K.; Friess, H.; Böttiger, B.W.; Bauer, H.; Dijkgraaf, M.G.; Durieux, M.E.; et al. Systemic lidocaine shortens length of hospital stay after colorectal surgery: A double-blinded, randomized, placebo-controlled trial. Ann. Surg. 2007, 246, 192–200. [Google Scholar] [CrossRef]
  121. 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] [PubMed] [Green Version]
  122. 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 2020, 72, 452–464. [Google Scholar] [CrossRef] [PubMed]
  123. Oshima, Y.; Sano, M.; Kajiwara, I.; Ichimaru, Y.; Itaya, T.; Kuramochi, T.; Hayashi, E.; Kim, J.; Kitajima, O.; Masugi, Y.; et al. Midazolam exhibits antitumour and anti-inflammatory effects in a mouse model of pancreatic ductal adenocarcinoma. Br. J. Anaesth. 2022, 128, 679–690. [Google Scholar] [CrossRef] [PubMed]
  124. Kang, J.; Zheng, Z.; Li, X.; Huang, T.; Rong, D.; Liu, X.; Qin, M.; Wang, Y.; Kong, X.; Song, J.; et al. Midazolam exhibits antitumour and enhances the efficiency of Anti-PD-1 immunotherapy in hepatocellular carcinoma. Cancer Cell Int. 2022, 22, 312. [Google Scholar] [CrossRef]
  125. Nishina, K.; Akamatsu, H.; Mikawa, K.; Shiga, M.; Maekawa, N.; Obara, H.; Niwa, Y. The inhibitory effects of thiopental, midazolam, and ketamine on human neutrophil functions. Anesth. Analg. 1998, 86, 159–165. [Google Scholar] [CrossRef] [Green Version]
  126. Mak, N.K.; Szeto, Y.Y.; Fung, M.C.; Leung, K.N.; Kwan, S.K. Effects of midazolam on the differentiation of murine myeloid leukemia cells. Chemotherapy 1997, 43, 272–281. [Google Scholar] [CrossRef]
  127. Zhang, L.; Wang, G.; Gan, J.; Dou, Z.; Bai, L. Analgesic effect of the midazolam-induced anesthesia in different doses on the patients after the thoracoscopic resection of lung cancer. Saudi J. Biol. Sci. 2019, 26, 2064–2067. [Google Scholar] [CrossRef]
  128. Roesslein, M.; Schibilsky, D.; Muller, L.; Goebel, U.; Schwer, C.; Humar, M.; Schmidt, R.; Geiger, K.K.; Pahl, H.L.; Pannen, B.H.; et al. Thiopental protects human T lymphocytes from apoptosis in vitro via the expression of heat shock protein 70. J. Pharmacol. Exp. Ther. 2008, 325, 217–225. [Google Scholar] [CrossRef]
  129. Ichiyama, T.; Nishikawa, M.; Lipton, J.M.; Matsubara, T.; Takashi, H.; Furukawa, S. Thiopental inhibits NF-kappaB activation in human glioma cells and experimental brain inflammation. Brain Res. 2001, 911, 56–61. [Google Scholar] [CrossRef]
  130. Huang, Y.H.; Sue, S.H.; Wu, Z.S.; Huang, S.M.; Lee, S.Y.; Wu, Z.F. Antitumorigenic Effect of Tramadol and Synergistic Effect with Doxorubicin in Human Breast Cancer Cells. Front. Oncol. 2022, 12, 811716. [Google Scholar] [CrossRef]
  131. Liu, X.; Yang, J.; Yang, C.; Huang, X.; Han, M.; Kang, F.; Li, J. Morphine promotes the malignant biological behavior of non-small cell lung cancer cells through the MOR/Src/mTOR pathway. Cancer Cell Int. 2021, 21, 622. [Google Scholar] [CrossRef]
  132. Li, C.; Qin, Y.; Zhong, Y.; Qin, Y.; Wei, Y.; Li, L.; Xie, Y. Fentanyl inhibits the progression of gastric cancer through the suppression of MMP-9 via the PI3K/Akt signaling pathway. Ann. Transl. Med. 2020, 8, 118. [Google Scholar] [CrossRef] [PubMed]
  133. Gong, S.; Ying, L.; Fan, Y.; Sun, Z. Fentanyl Inhibits Lung Cancer Viability and Invasion via Upregulation of miR-331-3p and Repression of HDAC5. OncoTargets Ther. 2020, 13, 13131–13141. [Google Scholar] [CrossRef] [PubMed]
  134. Chen, Y.; Qin, Y.; Li, L.; Chen, J.; Zhang, X.; Xie, Y. Morphine Can Inhibit the Growth of Breast Cancer MCF-7 Cells by Arresting the Cell Cycle and Inducing Apoptosis. Biol. Pharm. Bull. 2017, 40, 1686–1692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Kim, J.Y.; Ahn, H.J.; Kim, J.K.; Kim, J.; Lee, S.H.; Chae, H.B. Morphine Suppresses Lung Cancer Cell Proliferation Through the Interaction with Opioid Growth Factor Receptor: An In Vitro and Human Lung Tissue Study. Anesth. Analg. 2016, 123, 1429–1436. [Google Scholar] [CrossRef] [PubMed]
  136. Bimonte, S.; Barbieri, A.; Rea, D.; Palma, G.; Luciano, A.; Cuomo, A.; Arra, C.; Izzo, F. Morphine Promotes Tumor Angiogenesis and Increases Breast Cancer Progression. Biomed Res. Int. 2015, 2015, 161508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Sekandarzad, M.W.; van Zundert, A.A.J.; Lirk, P.B.; Doornebal, C.W.; Hollmann, M.W. Perioperative Anesthesia Care and Tumor Progression. Anesth. Analg. 2017, 124, 1697–1708. [Google Scholar] [CrossRef]
  138. Falk, W.; Magnuson, A.; Eintrei, C.; Henningsson, R.; Myrelid, P.; Matthiessen, P.; Gupta, A. Comparison between epidural and intravenous analgesia effects on disease-free survival after colorectal cancer surgery: A randomised multicentre controlled trial. Br. J. Anaesth. 2021, 127, 65–74. [Google Scholar] [CrossRef]
  139. 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]
  140. Connolly, J.G.; Tan, K.S.; Mastrogiacomo, B.; Dycoco, J.; Caso, R.; Jones, G.D.; McCormick, P.J.; Sanchez-Vega, F.; Irie, T.; Scarpa, J.R.; et al. Intraoperative opioid exposure, tumour genomic alterations, and survival differences in people with lung adenocarcinoma. Br. J. Anaesth. 2021, 127, 75–84. [Google Scholar] [CrossRef] [PubMed]
  141. 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]
  142. Zylla, D.; Gourley, B.L.; Vang, D.; Jackson, S.; Boatman, S.; Lindgren, B.; 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]
  143. Ramirez, M.F.; Gorur, A.; Cata, J.P. Opioids and cancer prognosis: A summary of the clinical evidence. Neurosci. Lett. 2021, 746, 135661. [Google Scholar] [CrossRef]
  144. Sueoka, N.; Sueoka, E.; Okabe, S.; Fujiki, H. Anti-cancer effects of morphine through inhibition of tumour necrosis factor-alpha release and mRNA expression. Carcinogenesis 1996, 17, 2337–2341. [Google Scholar] [CrossRef] [PubMed]
  145. Sueoka, E.; Sueoka, N.; Kai, Y.; Okabe, S.; Suganuma, M.; Kanematsu, K.; Yamamoto, T.; Fujiki, H. Anticancer activity of morphine and its synthetic derivative, KT-90, mediated through apoptosis and inhibition of NF-kappaB activation. Biochem. Biophys. Res. Commun. 1998, 252, 566–570. [Google Scholar] [CrossRef] [PubMed]
  146. Davis, R.L.; Buck, D.J.; Saffarian, N.; Stevens, C.W. The opioid antagonist, beta-funaltrexamine, inhibits chemokine expression in human astroglial cells. J. Neuroimmunol. 2007, 186, 141–149. [Google Scholar] [CrossRef] [Green Version]
  147. Bastami, S.; Norling, C.; Trinks, C.; Holmlund, B.; Walz, T.M.; Ahlner, J.; Uppugunduri, S. Inhibitory effect of opiates on LPS mediated release of TNF and IL-8. Acta Oncol. 2013, 52, 1022–1033. [Google Scholar] [CrossRef]
  148. Kim, M.H.; Lee, J.R.; Kim, K.J.; Jun, J.H.; Hwang, H.J.; Lee, W.; Nam, S.H.; Oh, J.E.; Yoo, Y.C. Identification for antitumor effects of tramadol in a xenograft mouse model using orthotopic breast cancer cells. Sci. Rep. 2021, 11, 22113. [Google Scholar] [CrossRef]
  149. Ma, M.; Wang, X.; Liu, N.; Shan, F.; Feng, Y. Low-dose naltrexone inhibits colorectal cancer progression and promotes apoptosis by increasing M1-type macrophages and activating the Bax/Bcl-2/caspase-3/PARP pathway. Int. Immunopharmacol. 2020, 83, 106388. [Google Scholar] [CrossRef]
  150. Ding, S.; Ma, H.; Wang, G.; Yu, Z.; Li, K.; Huang, A. Effect of Remifentanil Combined Anesthesia on Cytokines and Oxidative Stress in Patients undergoing Laparoscopic Surgery for Colon Cancer. J. Coll. Physicians Surg. Pak. 2019, 29, 8–11. [Google Scholar] [CrossRef]
  151. Wan, Z.; Chu, C.; Zhou, R.; Que, B. Effects of Oxycodone Combined with Flurbiprofen Axetil on Postoperative Analgesia and Immune Function in Patients Undergoing Radical Resection of Colorectal Cancer. Clin. Pharmacol. Drug Dev. 2021, 10, 251–259. [Google Scholar] [CrossRef] [PubMed]
  152. Zhang, Y.; Jiang, Q.; Li, T. Nalbuphine analgesic and anti-inflammatory effects on patients undergoing thoracoscopic lobectomy during the perioperative period. Exp. Ther. Med. 2017, 14, 3117–3121. [Google Scholar] [CrossRef] [Green Version]
  153. Titon, O.J.; Titon, J.P.; Silva, J.C.D.; Ferreira, M.O.; Garbim, M.R.; Rech, D.; Souza, J.A.; Panis, C. Influence of exogenous opioids on the acute inflammatory response in the perioperative period of oncological surgery: A clinical study. Braz. J. Anesthesiol. 2021; online ahead of print. [Google Scholar] [CrossRef] [PubMed]
  154. Ninomiya, I.; Nagai, N.; Oyama, K.; Hayashi, H.; Tajima, H.; Kitagawa, H.; Fushida, S.; Fujimura, T.; Ohta, T. Antitumor and anti-metastatic effects of cyclooxygenase-2 inhibition by celecoxib on human colorectal carcinoma xenografts in nude mouse rectum. Oncol. Rep. 2012, 28, 777–784. [Google Scholar] [CrossRef] [Green Version]
  155. Valle, B.L.; D′Souza, T.; Becker, K.G.; Wood, W.H., 3rd; Zhang, Y.; Wersto, R.P.; Morin, P.J. Non-steroidal anti-inflammatory drugs decrease E2F1 expression and inhibit cell growth in ovarian cancer cells. PLoS ONE 2013, 8, e61836. [Google Scholar] [CrossRef] [Green Version]
  156. Garcia, M.; Velez, R.; Romagosa, C.; Majem, B.; Pedrola, N.; Olivan, M.; Rigau, M.; Guiu, M.; Gomis, R.R.; Morote, J.; et al. Cyclooxygenase-2 inhibitor suppresses tumour progression of prostate cancer bone metastases in nude mice. BJU Int. 2014, 113, E164–E177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Henry, W.S.; Laszewski, T.; Tsang, T.; Beca, F.; Beck, A.H.; McAllister, S.S.; Toker, A. Aspirin Suppresses Growth in PI3K-Mutant Breast Cancer by Activating AMPK and Inhibiting mTORC1 Signaling. Cancer Res. 2017, 77, 790–801. [Google Scholar] [CrossRef] [Green Version]
  158. Zappavigna, S.; Cossu, A.M.; Grimaldi, A.; Bocchetti, M.; Ferraro, G.A.; Nicoletti, G.F.; Filosa, R.; Caraglia, M. Anti-Inflammatory Drugs as Anticancer Agents. Int. J. Mol. Sci. 2020, 21, 2605. [Google Scholar] [CrossRef] [Green Version]
  159. 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]
  160. Forget, P.; Tombal, B.; Scholtès, J.L.; Nzimbala, J.; Meulders, C.; Legrand, C.; Van Cangh, P.; Cosyns, J.P.; De Kock, M. Do intraoperative analgesics influence oncological outcomes after radical prostatectomy for prostate cancer? Eur. J. Anaesthesiol. 2011, 28, 830–835. [Google Scholar] [CrossRef]
  161. Choi, J.E.; Villarreal, J.; Lasala, J.; Gottumukkala, V.; Mehran, R.J.; Rice, D.; Yu, J.; Feng, L.; Cata, J.P. Perioperative neutrophil:lymphocyte ratio and postoperative NSAID use as predictors of survival after lung cancer surgery: A retrospective study. Cancer Med. 2015, 4, 825–833. [Google Scholar] [CrossRef] [PubMed]
  162. Liu, G.; Yu, M.Y.; Huang, X.; Zhu, D.; Cheng, S.; Ma, R.; Gu, G. Synergistic effect of celecoxib in tumor necrosis factor-related apoptosis-inducing ligand treatment in osteosarcoma cells. Mol. Med. Rep. 2014, 10, 2198–2202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Todo, M.; Horinaka, M.; Tomosugi, M.; Tanaka, R.; Ikawa, H.; Sowa, Y.; Ishikawa, H.; Fujiwara, H.; Otsuji, E.; Sakai, T. Ibuprofen enhances TRAIL-induced apoptosis through DR5 upregulation. Oncol. Rep. 2013, 30, 2379–2384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Sareddy, G.R.; Geeviman, K.; Ramulu, C.; Babu, P.P. The nonsteroidal anti-inflammatory drug celecoxib suppresses the growth and induces apoptosis of human glioblastoma cells via the NF-κB pathway. J. Neurooncol. 2012, 106, 99–109. [Google Scholar] [CrossRef] [PubMed]
  165. Lu, M.; Strohecker, A.; Chen, F.; Kwan, T.; Bosman, J.; Jordan, V.C.; Cryns, V.L. Aspirin sensitizes cancer cells to TRAIL-induced apoptosis by reducing survivin levels. Clin. Cancer Res. 2008, 14, 3168–3176. [Google Scholar] [CrossRef] [Green Version]
  166. Katerinaki, E.; Haycock, J.W.; Lalla, R.; Carlson, K.E.; Yang, Y.; Hill, R.P.; Lorigan, P.C.; MacNeil, S. Sodium salicylate inhibits TNF-alpha-induced NF-kappaB activation, cell migration, invasion and ICAM-1 expression in human melanoma cells. Melanoma Res. 2006, 16, 11–22. [Google Scholar] [CrossRef]
  167. Yasui, H.; Adachi, M.; Imai, K. Combination of tumor necrosis factor-alpha with sulindac augments its apoptotic potential and suppresses tumor growth of human carcinoma cells in nude mice. Cancer 2003, 97, 1412–1420. [Google Scholar] [CrossRef]
  168. Takada, Y.; Bhardwaj, A.; Potdar, P.; Aggarwal, B.B. Nonsteroidal anti-inflammatory agents differ in their ability to suppress NF-kappaB activation, inhibition of expression of cyclooxygenase-2 and cyclin D1, and abrogation of tumor cell proliferation. Oncogene 2004, 23, 9247–9258. [Google Scholar] [CrossRef] [Green Version]
  169. Redpath, M.; Marques, C.M.; Dibden, C.; Waddon, A.; Lalla, R.; Macneil, S. Ibuprofen and hydrogel-released ibuprofen in the reduction of inflammation-induced migration in melanoma cells. Br. J. Dermatol. 2009, 161, 25–33. [Google Scholar] [CrossRef]
  170. Setia, S.; Nehru, B.; Sanyal, S.N. Activation of NF-κB: Bridging the gap between inflammation and cancer in colitis-mediated colon carcinogenesis. Biomed. Pharmacother. 2014, 68, 119–128. [Google Scholar] [CrossRef]
  171. Aboelella, N.S.; Brandle, C.; Okoko, O.; Gazi, M.Y.; Ding, Z.C.; Xu, H.; Gorman, G.; Bollag, R.; Davila, M.L.; Bryan, L.J.; et al. Indomethacin-induced oxidative stress enhances death receptor 5 signaling and sensitizes tumor cells to adoptive T-cell therapy. J. Immunother. Cancer 2022, 10, e004938. [Google Scholar] [CrossRef] [PubMed]
  172. Vaish, V.; Sanyal, S.N. Chemopreventive effects of NSAIDs on cytokines and transcription factors during the early stages of colorectal cancer. Pharmacol. Rep. 2011, 63, 1210–1221. [Google Scholar] [CrossRef] [PubMed]
  173. Axiak-Bechtel, S.M.; Mathew, L.M.; Amorim, J.R.; DeClue, A.E. Dogs with osteosarcoma have altered pro- and anti-inflammatory cytokine profiles. Vet. Med. Sci. 2019, 5, 485–493. [Google Scholar] [CrossRef] [Green Version]
  174. Wen, Y.; Wang, M.; Yang, J.; Wang, Y.; Sun, H.; Zhao, J.; Liu, W.; Zhou, Z.; Deng, H.; Castillo-Pedraza, C.; et al. A Comparison of Fentanyl and Flurbiprofen Axetil on Serum VEGF-C, TNF-α, and IL-1ß Concentrations in Women Undergoing Surgery for Breast Cancer. Pain Pract. 2015, 15, 530–537. [Google Scholar] [CrossRef] [PubMed]
  175. Pandazi, A.; Kapota, E.; Matsota, P.; Paraskevopoulou, P.; Dervenis, C.; Kostopanagiotou, G. Preincisional versus postincisional administration of parecoxib in colorectal surgery: Effect on postoperative pain control and cytokine response. A randomized clinical trial. World J. Surg. 2010, 34, 2463–2469. [Google Scholar] [CrossRef] [PubMed]
  176. Xu, Y.J.; Li, S.Y.; Cheng, Q.; Chen, W.K.; Wang, S.L.; Ren, Y.; Miao, C.H. Effects of anaesthesia on proliferation, invasion and apoptosis of LoVo colon cancer cells in vitro. Anaesthesia 2016, 71, 147–154. [Google Scholar] [CrossRef]
  177. 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 (Suppl. 1), i56–i62. [Google Scholar] [CrossRef] [Green Version]
  178. Hirano, Y.; Kaneko, H.; Konishi, T.; Itoh, H.; Matsuda, S.; Kawakubo, H.; Uda, K.; Matsui, H.; Fushimi, K.; Daiko, H.; et al. Short-Term Outcomes of Epidural Analgesia in Minimally Invasive Esophagectomy for Esophageal Cancer: Nationwide Inpatient Data Study in Japan. Ann. Surg. Oncol. 2022, 29, 8225–8234. [Google Scholar] [CrossRef]
  179. Yen, F.Y.; Chang, W.K.; Lin, S.P.; Lin, T.P.; Chang, K.Y. Association Between Epidural Analgesia and Cancer Recurrence or Survival After Surgery for Renal Cell Carcinoma: A Propensity Weighted Analysis. Front. Med. 2021, 8, 782336. [Google Scholar] [CrossRef]
  180. Xu, Z.Z.; Li, H.J.; Li, M.H.; Huang, S.M.; Li, X.; Liu, Q.H.; Li, J.; Li, X.Y.; 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]
  181. Karmakar, M.K.; Samy, W.; Lee, A.; Li, J.W.; Chan, W.C.; Chen, P.P.; Tsui, B.C.H. Survival Analysis of Patients with Breast Cancer Undergoing a Modified Radical Mastectomy with or Without a Thoracic Paravertebral Block: A 5-Year Follow-up of a Randomized Controlled Trial. Anticancer Res. 2017, 37, 5813–5820. [Google Scholar] [CrossRef] [PubMed]
  182. Myles, P.S.; Peyton, P.; Silbert, B.; Hunt, J.; Rigg, J.R.; Sessler, D.I. Perioperative epidural analgesia for major abdominal surgery for cancer and recurrence-free survival: Randomised trial. Bmj 2011, 342, d1491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Grandhi, R.K.; Lee, S.; Abd-Elsayed, A. The Relationship Between Regional Anesthesia and Cancer: A Metaanalysis. Ochsner J. 2017, 17, 345–361. [Google Scholar]
  184. Cakmakkaya, O.S.; Kolodzie, K.; Apfel, C.C.; Pace, N.L. Anaesthetic techniques for risk of malignant tumour recurrence. Cochrane Database Syst Rev. 2014, 11, Cd008877. [Google Scholar] [CrossRef] [PubMed]
  185. Inoue, G.N.C.; Pimenta, R.; Camargo, J.A.; Viana, N.I.; Guimarães, V.R.; Srougi, M.; Nahas, W.C.; Leite, K.R.M.; Reis, S.T. Combined spinal and general anesthesia attenuate tumor promoting effects of surgery. An experimental animal study. Ann. Med. Surg. 2022, 75, 103398. [Google Scholar] [CrossRef] [PubMed]
  186. Wei, W.; Zheng, X.; Gu, Y.; Fu, W.; Tang, C.; Yao, Y. Effect of general anesthesia with thoracic paravertebral block on postoperative delirium in elderly patients undergoing thoracoscopic lobectomy: A randomized-controlled trial. BMC Anesthesiol. 2022, 22, 1. [Google Scholar] [CrossRef] [PubMed]
  187. Fiorelli, A.; Izzo, A.C.; Frongillo, E.M.; Del Prete, A.; Liguori, G.; Di Costanzo, E.; Vicidomini, G.; Santini, M. Efficacy of wound analgesia for controlling post-thoracotomy pain: A randomized double-blind study. Eur. J. Cardiothorac. Surg. 2016, 49, 339–347. [Google Scholar] [CrossRef] [Green Version]
  188. Zhou, M.; Liu, W.; Peng, J.; Wang, Y. Impact of propofol epidural anesthesia on immune function and inflammatory factors in patients undergoing gastric cancer surgery. Am. J. Transl. Res. 2021, 13, 3064–3073. [Google Scholar] [PubMed]
  189. Zhao, J.; Mo, H. The Impact of Different Anesthesia Methods on Stress Reaction and Immune Function of the Patients with Gastric Cancer during Peri-Operative Period. J. Med. Assoc. Thail. 2015, 98, 568–573. [Google Scholar]
  190. Su, Y.; Pu, Y.; Zhao, Z.; Yang, X. Influence of combined epidural anesthesia on cognitive function, inflammation and stress response in elderly liver cancer patients undergoing surgery. Oncol. Lett. 2020, 19, 2733–2738. [Google Scholar] [CrossRef] [Green Version]
  191. Geng, J.; Wang, J.; Zhang, Y.; Song, W.; Zhu, J.; Chen, J.; Wu, Z. The Effect of a Combined Modified Pectoral and Stellate Ganglion Block on Stress and Inflammatory Response in Patients Undergoing Modified Radical Mastectomy. Int. J. Breast Cancer 2022, 2022, 3359130. [Google Scholar] [CrossRef] [PubMed]
  192. Okuda, J.; Suzuki, T.; Wakaizumi, K.; Kato, J.; Yamada, T.; Morisaki, H. Effects of Thoracic Epidural Anesthesia on Systemic and Local Inflammatory Responses in Patients Undergoing Lung Cancer Surgery: A Randomized Controlled Trial. J. Cardiothorac. Vasc. Anesth. 2022, 36, 1380–1386. [Google Scholar] [CrossRef] [PubMed]
  193. Cong, X.; Huang, Z.; Zhang, L.; Sun, M.; Chang, E.; Zhang, W.; Zhang, J. Effect of different anaesthesia methods on perioperative cellular immune function and long-term outcome in patients undergoing radical resection of esophageal cancer: A prospective cohort study. Am. J. Transl. Res. 2021, 13, 11427–11438. [Google Scholar] [PubMed]
  194. Karadeniz, M.S.; Mammadov, O.; Çiftci, H.; Usta, S.A.; Pembeci, K. Comparing the Effects of Combined General/Epidural Anaesthesia and General Anaesthesia on Serum Cytokine Levels in Radical Cystectomy. Turk. J. Anaesthesiol. Reanim. 2017, 45, 203–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Fant, F.; Tina, E.; Sandblom, D.; Andersson, S.O.; Magnuson, A.; Hultgren-Hörnkvist, E.; Axelsson, K.; Gupta, A. Thoracic epidural analgesia inhibits the neuro-hormonal but not the acute inflammatory stress response after radical retropubic prostatectomy. Br. J. Anaesth. 2013, 110, 747–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Siekmann, W.; Eintrei, C.; Magnuson, A.; Sjölander, A.; Matthiessen, P.; Myrelid, P.; Gupta, A. Surgical and not analgesic technique affects postoperative inflammation following colorectal cancer surgery: A prospective, randomized study. Color. Dis. 2017, 19, O186–O195. [Google Scholar] [CrossRef] [PubMed]
  197. Rauch, S.; Miller, C.; Bräuer, A.; Wallner, B.; Bock, M.; Paal, P. Perioperative Hypothermia—A Narrative Review. Int. J. Environ. Res. Public Health 2021, 18, 8749. [Google Scholar] [CrossRef] [PubMed]
  198. Snyder, G.L.; Greenberg, S. Effect of anaesthetic technique and other perioperative factors on cancer recurrence. Br. J. Anaesth. 2010, 105, 106–115. [Google Scholar] [CrossRef] [Green Version]
  199. Kurosawa, S. Anesthesia in patients with cancer disorders. Curr. Opin. Anaesthesiol. 2012, 25, 376–384. [Google Scholar] [CrossRef]
  200. Horowitz, M.; Neeman, E.; Sharon, E.; Ben-Eliyahu, S. Exploiting the critical perioperative period to improve long-term cancer outcomes. Nat. Rev. Clin. Oncol. 2015, 12, 213–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Du, G.; Zhao, B.; Zhang, Y.; Sun, T.; Liu, W.; Li, J.; Liu, Y.; Wang, Y.; Li, H.; Hou, X. Hypothermia activates adipose tissue to promote malignant lung cancer progression. PLoS ONE 2013, 8, e72044. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Yu, Q.; Tang, X.; Zhao, W.; Qiu, Y.; He, J.; Wan, D.; Li, J.; Wang, X.; He, X.; Liu, Y.; et al. Mild hyperthermia promotes immune checkpoint blockade-based immunotherapy against metastatic pancreatic cancer using size-adjustable nanoparticles. Acta Biomater. 2021, 133, 244–256. [Google Scholar] [CrossRef] [PubMed]
  203. Sulyok, I.; Fleischmann, E.; Stift, A.; Roth, G.; Lebherz-Eichinger, D.; Kasper, D.; Spittler, A.; Kimberger, O. Effect of preoperative fever-range whole-body hyperthermia on immunological markers in patients undergoing colorectal cancer surgery. Br. J. Anaesth. 2012, 109, 754–761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Atanackovic, D.; Pollok, K.; Faltz, C.; Boeters, I.; Jung, R.; Nierhaus, A.; Braumann, K.M.; Hossfeld, D.K.; Hegewisch-Becker, S. Patients with solid tumors treated with high-temperature whole body hyperthermia show a redistribution of naive/memory T-cell subtypes. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2006, 290, R585–R594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Coccolini, F.; Corbella, D.; Finazzi, P.; Brambillasca, P.; Benigni, A.; Prussiani, V.; Ceresoli, M.; Manfredi, R.; Poiasina, E.; Bertoli, P.; et al. Time course of cytokines, hemodynamic and metabolic parameters during hyperthermic intraperitoneal chemotherapy. Minerva Anestesiol. 2016, 82, 310–319. [Google Scholar]
  206. Oderda, M.; Cerutti, E.; Gontero, P.; Manetta, T.; Mengozzi, G.; Meyer, N.; Munegato, S.; Noll, E.; Rampa, P.; Piéchaud, T.; et al. The impact of warmed and humidified CO(2) insufflation during robotic radical prostatectomy: Results of a randomized controlled trial. Urologia 2019, 86, 130–140. [Google Scholar] [CrossRef] [PubMed]
  207. Hansen, D.; Bogatzki, S.; Syben, R.; Bechrakis, N.E.; Dopjans, D.; Spies, C.; Welte, M.; Schaffartzik, W. Effect of profound normovolemic hypotension and moderate hypothermia on circulating cytokines and adhesion molecules. Shock 1999, 12, 335–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Chang, S.C.; Yang, W.V. Hyperglycemia, tumorigenesis, and chronic inflammation. Crit. Rev. Oncol. Hematol. 2016, 108, 146–153. [Google Scholar] [CrossRef]
  209. Duan, W.; Shen, X.; Lei, J.; Xu, Q.; Yu, Y.; Li, R.; Wu, E.; Ma, Q. Hyperglycemia, a neglected factor during cancer progression. Biomed Res. Int. 2014, 2014, 461917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Otto, L.; Rahn, S.; Daunke, T.; Walter, F.; Winter, E.; Möller, J.L.; Rose-John, S.; Wesch, D.; Schäfer, H.; Sebens, S. Initiation of Pancreatic Cancer: The Interplay of Hyperglycemia and Macrophages Promotes the Acquisition of Malignancy-Associated Properties in Pancreatic Ductal Epithelial Cells. Int. J. Mol. Sci. 2021, 22, 5086. [Google Scholar] [CrossRef] [PubMed]
  211. Liu, T.C.; Liu, Q.R.; Huang, Y. Effect of intraoperative glucose fluctuation and postoperative IL-6, TNF-α, CRP levels on the short-term prognosis of patients with intracranial supratentorial neoplasms. Asian Pac. J. Cancer Prev. 2014, 15, 10879–10882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Lachmann, G.; von Haefen, C.; Wollersheim, T.; Spies, C. Severe perioperative hyperglycemia attenuates postoperative monocytic function, basophil count and T cell activation. Minerva Anestesiol. 2017, 83, 921–929. [Google Scholar] [CrossRef] [PubMed]
  213. Amato, A.; Pescatori, M. Perioperative blood transfusions for the recurrence of colorectal cancer. Cochrane Database Syst Rev. 2006, 2006, Cd005033. [Google Scholar] [CrossRef] [PubMed]
  214. Cata, J.P.; Lasala, J.; Pratt, G.; Feng, L.; Shah, J.B. Association between Perioperative Blood Transfusions and Clinical Outcomes in Patients Undergoing Bladder Cancer Surgery: A Systematic Review and Meta-Analysis Study. J. Blood Transfus. 2016, 2016, 9876394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Li, S.L.; Ye, Y.; Yuan, X.H. Association between Allogeneic or Autologous Blood Transfusion and Survival in Patients after Radical Prostatectomy: A Systematic Review and Meta-Analysis. PLoS ONE 2017, 12, e0171081. [Google Scholar] [CrossRef]
  216. Agnes, A.; Lirosi, M.C.; Panunzi, S.; Santocchi, P.; Persiani, R.; D′Ugo, D. The prognostic role of perioperative allogeneic blood transfusions in gastric cancer patients undergoing curative resection: A systematic review and meta-analysis of non-randomized, adjusted studies. Eur. J. Surg. Oncol. 2018, 44, 404–419. [Google Scholar] [CrossRef] [PubMed]
  217. Milasiene, V.; Stratilatovas, E.; Characiejus, D.; Kazbariene, B.; Norkiene, V. TGF-beta1 and TNF-alpha after red blood cell transfusion in colorectal cancer patients. Exp. Oncol. 2007, 29, 67–70. [Google Scholar]
  218. Surinenaite, B.; Prasmickiene, G.; Milasiene, V.; Stratilatovas, E.; Didziapetriene, J. The influence of surgical treatment and red blood cell transfusion on changes in antioxidative and immune system parameters in colorectal cancer patients. Medicina 2009, 45, 785–791. [Google Scholar] [CrossRef] [Green Version]
  219. Xing, Y.L.; Wang, Y.C. Influence of autologous and homologous blood transfusion on interleukins and tumor necrosis factor-α in peri-operative patients with esophageal cancer. Asian Pac. J. Cancer Prev. 2014, 15, 7831–7834. [Google Scholar] [CrossRef] [Green Version]
  220. Heiss, M.M.; Fraunberger, P.; Delanoff, C.; Stets, R.; Allgayer, H.; Ströhlein, M.A.; Tarabichi, A.; Faist, E.; Jauch, K.W.; Schildberg, F.W. Modulation of immune response by blood transfusion: Evidence for a differential effect of allogeneic and autologous blood in colorectal cancer surgery. Shock 1997, 8, 402–408. [Google Scholar] [CrossRef] [Green Version]
  221. Geng, Y.; Liu, L. Impact of Allogeneic Leukocyte-Depleted Red Blood Cell Transfusion on Inflammatory Response and Blood Coagulation in Patients with Recurrence of Colon Cancer after Operation. Evid. Based Complement. Alternat. Med. 2021, 2021, 6957569. [Google Scholar] [CrossRef] [PubMed]
  222. Benson, D.D.; Beck, A.W.; Burdine, M.S.; Brekken, R.; Silliman, C.C.; Barnett, C.C., Jr. Accumulation of pro-cancer cytokines in the plasma fraction of stored packed red cells. J. Gastrointest. Surg. 2012, 16, 460–468. [Google Scholar] [CrossRef] [Green Version]
  223. Beck-Schimmer, B.; Romero, B.; Booy, C.; Joch, H.; Hallers, U.; Pasch, T.; Spahn, D.R. Release of inflammatory mediators in irradiated cell salvage blood and their biological consequences in human beings following transfusion. Eur. J. Anaesthesiol. 2004, 21, 46–52. [Google Scholar] [CrossRef] [PubMed]
  224. Zhang, Q.; Wu, H.; Zhang, J.; Qi, Q.; Zhang, W.; Xia, R. Preoperative Immune Response is Associated with Perioperative Transfusion Requirements in Glioma Surgery. J. Cancer 2019, 10, 3526–3532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Wang, X.; Lin, Y. Tumor necrosis factor and cancer, buddies or foes? Acta Pharm. Sin. 2008, 29, 1275–1288. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Overview of surgery and surgical stress response, which may jointly contribute to cancer recurrence and metastasis. IL = interleukin; MMP = matrix metalloproteinase; NK cell = nature killer cell; TNF-α = tumor necrosis factor alpha.
Figure 1. Overview of surgery and surgical stress response, which may jointly contribute to cancer recurrence and metastasis. IL = interleukin; MMP = matrix metalloproteinase; NK cell = nature killer cell; TNF-α = tumor necrosis factor alpha.
Cancers 15 00739 g001
Figure 2. A theory of balance between TNF-α-promoting and -inhibiting factors contributing to cancer recurrence/metastasis. (A) Surgical stress and blood transfusion may promote cancer recurrence/metastasis by increasing the TNF-α level; (B) ketamine, DEX, lidocaine, opioids, and RA may reduce cancer recurrence/metastasis by decreasing the TNF-α release. DEX = dexmedetomidine; NSAID = non-steroidal anti-inflammatory drug; TNF-α = tumor necrosis factor alpha; VA = volatile anesthetic; RA = regional anesthesia/analgesia.
Figure 2. A theory of balance between TNF-α-promoting and -inhibiting factors contributing to cancer recurrence/metastasis. (A) Surgical stress and blood transfusion may promote cancer recurrence/metastasis by increasing the TNF-α level; (B) ketamine, DEX, lidocaine, opioids, and RA may reduce cancer recurrence/metastasis by decreasing the TNF-α release. DEX = dexmedetomidine; NSAID = non-steroidal anti-inflammatory drug; TNF-α = tumor necrosis factor alpha; VA = volatile anesthetic; RA = regional anesthesia/analgesia.
Cancers 15 00739 g002
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

Tseng, W.-C.; Lai, H.-C.; Huang, Y.-H.; Chan, S.-M.; Wu, Z.-F. Tumor Necrosis Factor Alpha: Implications of Anesthesia on Cancers. Cancers 2023, 15, 739. https://doi.org/10.3390/cancers15030739

AMA Style

Tseng W-C, Lai H-C, Huang Y-H, Chan S-M, Wu Z-F. Tumor Necrosis Factor Alpha: Implications of Anesthesia on Cancers. Cancers. 2023; 15(3):739. https://doi.org/10.3390/cancers15030739

Chicago/Turabian Style

Tseng, Wei-Cheng, Hou-Chuan Lai, Yi-Hsuan Huang, Shun-Ming Chan, and Zhi-Fu Wu. 2023. "Tumor Necrosis Factor Alpha: Implications of Anesthesia on Cancers" Cancers 15, no. 3: 739. https://doi.org/10.3390/cancers15030739

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