Korean J Anesthesiol Search

CLOSE


Korean J Anesthesiol > Volume 74(3); 2021 > Article
Alam, Rampes, Patel, Hana, and Ma: Anesthetics or anesthetic techniques and cancer surgical outcomes: a possible link

Abstract

As of 2018 cancer is responsible for almost 9.6 million deaths annually and, with an aging population, the incidence of cancer is expected to continue to rise. Surgery is an important treatment modality for patients with solid organ cancers. It has been postulated that, due to potentially overlapping processes underlying the development of malignancy and the therapeutic pathways of various anesthetic agents, the choice of anesthetic type and method of administration may affect post-operative outcomes in patients with cancer. This is a literature review of the most recent evidence extracted from various databases including PubMed, EMBASE, and the Cochrane, as well as journals and book reference lists. The review highlights the pathophysiological processes underpinning cancer development and the molecular actions of anesthetic agents, pre-clinical and retrospective studies investigating cancer and anesthetics, as well as ongoing clinical trials. Overall, there are conflicting results regarding the impact of regional vs. general anesthesia on cancer recurrence, whilst the majority of data suggest a benefit of the use of intravenous propofol over inhalational volatile anesthetics. The biological changes associated with the surgical inflammatory response offer a unique opportunity to intervene to counteract any potentially cancer-promoting effects.

General introduction: anesthesia and cancer outcomes

Cancer was responsible for 9.6 million deaths worldwide in 2018 and is the second leading cause of mortality globally [1]. In the context of aging populations, the incidence of cancer is expected to continue to rise from 14 million in 2012 to an estimated 24 million in 2035 [2]. Surgery is a central treatment modality for patients with solid organ cancers; it is estimated that over 80% of cancer patients will undergo a surgical procedure as part of their treatment [3]. Recurrence of cancer after surgery is affected by numerous factors including primary cancer organ type, Tumor Node Metastasis staging, and surgical technique.
Surgery results in a complex inflammatory response involving both the innate and adaptive immune system and is thought to result in a period of postoperative immunosuppression that predisposes to infection [4]. The inflammatory response has wide ranging systemic effects from impacting postoperative recovery to sleep quality [4,5]. Perioperative interventions such as anesthesia and anesthetic techniques have been hypothesized to play a role in modifying the surgical inflammatory response and thus cancer recurrence. The role of anesthesia on cancer recurrence has been the subject of increasing attention over the past decade and has resulted in many retrospective studies and pre-clinical research into the area. An increasing body of pre-clinical laboratory data suggests that general anesthetic agents have the ability to influence key hallmarks of cancer involved in tumorigenesis and metastasis [6]. There are two key classes of general anesthetic agents used in clinical practice: intravenous propofol and inhalational volatile anesthetic agents such as sevoflurane. Inhalation anesthetics have been shown to enhance proliferation, migration, invasion, and angiogenesis across a range of cancer cell types, whereas propofol has been shown to antagonize these same pathways [6]. Several retrospective studies have demonstrated an association between inhalational anesthesia and reduced recurrence-free survival in cancer patients undergoing elective surgery compared to survival in cancer patients who receive propofol-based anesthesia [7,8]. However, there are smaller retrospective studies that show no association, highlighting the need for future prospective and randomized controlled trials.
In this literature review we present the most recent evidence extracted from various databases including PubMed, EMBASE, and the Cochrane, as well as journals and book reference lists.

Cancer development

Cancer results from the proliferation of a clonal population of cells—a multistage process termed carcinogenesis. A single cell undergoes a mutation in critical genes responsible for the control of cell division, cell death, and the maintenance of genetic integrity thereby rendering the cell susceptible to the acquisition of further mutations [9]. The tumor cell becomes refractory to regulatory biochemical cell signaling pathways, which results in the progressive loss of differentiation and in turn, uncontrolled cellular proliferation ensues [9]. The expanding pre-neoplastic cell clone outgrows the capacity of the host vasculature and subsequent tumor progression is dependent on angiogenesis for the supply of growth factors and oxygen [10]. Pro-angiogenic factors including vascular endothelial growth factor (VEGF), platelet-derived endothelial growth factor (PDGF), and fibroblast growth factor are released from the tumor, establishing a new capillary network that promotes tumor growth, local invasion, and metastasis [11].

Metastasis and tumor progression

The pathogenesis of cancer metastases is complex, of which a series of tumor-host interactions are outlined by the metastatic cascade. Tumor cells lose their cell polarity and cell-cell adhesion properties, allowing for a subclone of cells with metastatic potential to invade the surrounding stroma and dissociate from the primary tumor mass (invasion) [12]. The detached cells, known as circulating tumor cells (CTCs), enter the systemic circulation via the established blood vessel network formed through angiogenesis and the lymphatic system (intravasation). During this process, most CTCs are rapidly destroyed by the immune system as a result of host immunosurveillance carried out by NK cells and CD8+ T cells, with only 0.1% of cells viable after 24 h [13]. The surviving tumor cells arrest in the capillary beds of a distant organ and adhere to capillary endothelial cells and thus penetrate the endothelium and basement membrane (extravasation). Consequently, proliferation within the secondary organ parenchyma achieves metastasis and the formation of a malignant tumor.
It is becoming increasingly recognized that various anesthetic agents used in the perioperative setting for primary cancer surgery have a role in cancer recurrence through postoperative metastasis [14]. In this narrative review, we aim to present the current state of evidence linking anesthetic techniques and cancer surgical outcomes.

Molecular actions of anesthetics and cancer

Inhalational anesthetics

Volatile anesthesia

Rapidly acting volatile anesthetic agents, such as sevoflurane and isoflurane, are commonly used for the maintenance of general anesthesia. It is well-established that these agents have pro-inflammatory and immune modulatory effects, and therefore, may have deleterious effects in cancer recurrence, although the exact molecular mechanisms are incompletely understood [6,15,16].
In particular, volatile anesthetics have been shown to suppress NK cell cytotoxicity and induce T-lymphocyte apoptosis, of which both cells have a vital role in immune surveillance and achieving anti-metastatic immunity after cancer surgery [17,18]. Thus, volatile agents may promote immunosuppression and the metastatic spread of residual cancer cells postoperatively.
Moreover, volatile anesthetics have a protective role against ischemia-reperfusion injury in various organs and tissues [19]. These cytoprotective features, however, are associated with the upregulation of hypoxia-inducible factor 1-alpha (HIF-1α) in tumor cells, causing increased transcription of genes encoding VEGF and PDGF [20] and thereby facilitate tumor angiogenesis, residual cell survival, and tumor cell migration.

Nitrous oxide

Nitrous oxide is an anesthetic gas used for the maintenance of anesthesia often in combination with more potent general anesthetic techniques for surgical anesthesia. This inhaled agent has been related to a number of immunosuppressive effects, primarily through impaired neutrophil chemotaxis and suppressed NK cell and macrophage function [21,22]. This is mediated by its interaction with vitamin B12, causing selective inactivation of methionine synthase, which is critical for DNA, purine, and thymidylate synthesis [23]. Consequently, there is impaired synthesis of hematopoietic cells involved in tumor surveillance.

Intravenous anesthetics

Propofol

Propofol is the most extensively used intravenous anesthetic agent for induction and maintenance of general anesthesia. It has been demonstrated to possess a range of antitumor properties and may seem to have protective effects against cancer cell dissemination and development of metastasis. This is achieved directly by regulating key cell signaling pathways implicated in tumorigenesis, such as the MAPK and NF-κB pathways [24], as well as regulating expression of miRNA and HIF-1α [24,25]. Indirectly, propofol has been shown to minimize perioperative immunosuppression by preserving NK cell and cytotoxic T cell function [26].

Ketamine and thiopental

Ketamine and thiopental are alternative suitable intravenous anesthetic agents indicated in emergency medicine and for patients with high intracranial pressure respectively. Both agents have exhibited immunomodulatory effects by suppressing NK cell activity and increasing tumor cell viability [27]. In particular, ketamine can upregulate anti-apoptotic proteins such as Bcl-2 enabling tumor cell proliferation and promotes production of pro-inflammatory cytokines such as IL-6 and TNFα [28,29].

Local anesthesia

Local anesthetics cause reversible, local inhibition of nociception, providing targeted anesthesia and analgesia. Local anesthetics have been shown to exert anti-tumor growth activity [30], although exact mechanisms are widely conflicted in studies. Possible mechanisms include their well-established inhibitory actions on voltage-gated sodium channels, which are expressed by cancer cells and correlate with tumor growth and metastatic formation [31]. Other evidence suggests these agents have protective effects on cell-mediated immunity and administration of lidocaine in particular can directly inhibit the epidermal growth factor receptor (EGFR) involved in cellular proliferation [30]. Intravenous infusions of lidocaine are a popular component of multimodal analgesia, particularly for major surgery, and therefore, are a feasible adjunct.

Sedatives

Common benzodiazepines, consisting of midazolam, lorazepam, and diazepam, are primarily used for preoperative sedation. The effect of benzodiazepines on tumor recurrence is disputed between studies. Early studies show that these sedatives, especially midazolam, have negative immunomodulatory effects and potentiate tumor occurrence [32]. Other studies report there is no association [33].

Opioids

Opioid analgesics are widely used in the perioperative period to supplement general anesthetic agents during induction and maintenance of anesthesia. Evidence from experimental studies investigating the role of opioids in tumor growth and metastasis is conflicting. Multiple animal studies have found that some opioids promote immunosuppression and in turn, tumor recurrence post-operatively, with the effects on immune function varying between the different types of opioids. In particular, morphine has largely been shown to suppress NK cell cytotoxicity and T-cell proliferation [34,35]; however, a small number of studies contradict these findings and instead propose the antitumor effects of morphine [36,37]. Likewise, fentanyl has been shown to inhibit NK cells and promote apoptosis of lymphocytes and macrophages in various laboratory studies [38,39]. Yet, a recent retrospective cohort study of 1,679 patients with stage I-III colorectal cancer showed no association between fentanyl and oncological outcomes/prognosis [40]. Alternatively, tramadol has been shown to have immune stimulatory properties by enhancing NK cell cytotoxicity [41].
There is also evidence that mu-opioid receptors (MORs) are overexpressed in certain cancers. Consequently, opioid binding at the MOR directly promotes cancer cell growth via growth-factor induced receptor signaling and potentiation of angiogenesis [42]. A study of lung samples from 34 patients with lung cancer demonstrated there was a two-fold increase in MOR expression in patients with metastatic lung disease [43]. Clinical studies further support the role of MOR in cancer progression. In a retrospective study of 113 prostate cancer patients, overexpression of MOR was associated with reduced overall survival and progression-free survival, especially prominent in those with metastatic disease [44]. In keeping with these results, two randomized controlled trials have shown treatment with Methylnaltrexone (a MOR antagonist) is associated with increased overall survival in end-stage cancer patients [45].
Overall, the role of opioids in facilitating tumor recurrence and metastasis are variable and conflicting with opioid type, dosage, and administration also influencing outcomes. Greater quality clinical evidence in the form of prospective randomized controlled trials is needed.

Pre-clinical in vitro and in vivo studies of cancer and anesthetics

The effects of anesthesia on various cancers have been extensively studied in vitro, although in vivo studies are limited in comparison. The molecular actions of anesthetic agents and lignocaine are summarized in Table 1. Understanding the underlying mechanism of anesthetics and their potential effects on cell cycle arrest and apoptosis will give us more insight into the clinical implications.

Sevoflurane

Literature studying the effects of volatile anesthetics on cancer proliferation, migration, apoptosis, and tumor aggression remains inconsistent. Yang et al. [46] incubated SW480 colon cells with different concentrations of sevoflurane (1.7%, 3.4% and 5.1%) for 6 h, and the results have shown sevoflurane’s capacity to induce apoptosis and inhibit the proliferation and invasion of colon cancer cells by inactivating the Ras/Raf/MEK/ERK signaling pathway. However, research by Bundscherer et al. [47] had revealed sevoflurane’s and desflurane’s limited effect on SW480 colon cancer cells, albeit at lower concentrations of drug during incubation (1% or 2.5% sevoflurane).
Sevoflurane was also found to inhibit the viability of SKOV3 and OVCAR3 cells in a dose-dependent manner, by reducing the migration and invasion ability of these cells. In addition, MMP-9 and stanniocalcin 1 (STC1) were also downregulated. These factors in combination have alluded to sevoflurane’s involvement in inhibiting the progression of ovarian cancer (concentrations of sevoflurane ranging from 0.5% to 10%, depending on cell type) [48]. The effects of sevoflurane were corroborated in a study by Kang and Wang [49], which had also shown an inhibition of ovarian cancer proliferation (sevoflurane low concentration (1.7%), medium concentration (3.4%) and high concentration (5.1%) groups); however, this was through the repression of the phosphorylation of JNK and p38 MAPK signaling pathways. In contrast, a study that utilized higher concentrations of volatile anesthetics (sevoflurane 3.6%, isoflurane 2%, and desflurane 10.3%), but with a shorter incubation period, had revealed a significant increase in VEGF-A, MMP-11, CXCR2, and TGF-β genes, which collectively may enhance ovarian cancer proliferation [50]. Cervical cancer Caski and HeLa lines were incubated with sevoflurane (1-3%) for 2–4 h, which resulted in the proliferation, migration, and invasion of immortalized cervical cancer cells by increasing histone deacetylase 6 expression via the ERK1/2 and phosphatidylinositide 3-kinase/AKT signaling pathways [51,52].
Data from Chen et al. [53] has shown sevoflurane’s inhibition of osteosarcoma cell invasion and proliferation through regulating miR-203/WNT2B/Wnt/β-catenin axis; cells were exposed to 0%, 1%, 2%, 5% and 10% sevoflurane. Further evidence of sevoflurane’s involvement (0%, 2%, 4% or 8% sevoflurane) in the inhibition of Wnt/β-catenin is noted in the inhibition of leukemia cell proliferation [54], and involvement in cognitive dysfunction (3.6% sevoflurane) [55]. Furthermore, sevoflurane (0.2 mM) has been reported to promote lung metastases through the overexpression of IL-6 in pre-metastatic lung during the perioperative phase [56]. Results from Hurmath et al. [57] have highlighted that the inhibitory role of sevoflurane (2.5%), and different concentrations of thiopental, in glioma cells is dependent on regulating MMP-2 migration and activity. Another volatile anesthetic, isoflurane, has been shown to be involved in the inhibition of hepatic carcinoma aggression, as achieved through the regulation of NF-κB and PI3K/AKT signaling pathways [58], in addition to having detrimental effects in glioblastoma by promoting tumor and migration capacities (1.2% isoflurane) [59].

Propofol

A variety of mechanisms have been proposed to explain the role of propofol in cancer cells. Liang and Dong [60] incubated human colon cancer line SW480 with propofol (2, 4, and 8 μg/ml) and propofol with colivelin, which resulted in the inhibition of JAK2/STAT3 signaling pathway and the proliferation, migration, and invasion of human colon cancer cells. Similarly, Zhang et al. [61] incubated LOVO and SW480 cells with propofol (8 μg/ml), exerting an inhibition of cell invasion and induction of apoptosis through STAT3/HOTAIR by activation of WIF-1 and the suppression of the Wnt pathway.
The A549 cancer line, which are adenocarcinoma alveolar basal epithelial cells, was incubated with propofol (0, 2, 5, and 10 μg/ml). Propofol demonstrated inhibition of A549 cell growth in a concentrated and time-dependent manner, by accelerating apoptosis via the miR-21/PTEN/AKT pathway [62]. Wang et al. [63] exposed pancreatic cancer PANC-1 cells to a relatively higher concentration of propofol (20 μg/ml). Consequently, propofol was seen to inhibit the migration and apoptosis induction of PANC-1 cells via miR-34a-mediated E-cadherin and LOC285194 signals. In another study, PANC-1 cells were treated with 5 or 10 μg/ml of propofol, resulting in a reduced expression of ADAM8 and inhibition of cell proliferation and migration of PANC-1 via downregulation of β1, ERK1/2, MMP2, and MMP9 [64]. Human gastric cells, SGC-7901 and NCI-N87, were exposed to different concentrations of propofol (5, 10, 20 μM), in which inhibition of epithelial to mesenchymal transition, migration, and invasion of gastric cells were noted in a dose-dependent manner [65]. The inhibitory effects of propofol (5, 10, 20 mg/ml) on papillary thyroid cancer cells were reported, in which an upregulation of miR-320a and downregulation of ANRIL and inactivation of Wnt/β-catenin and NF-κB pathways all played a role [66].
U251 and A172 glioma cell lines were incubated with different concentrations of propofol (5, 10 μg/ml) for 24 h. This consequently resulted in the inhibition of cell proliferation, invasion, and migration through the mir-410-3p/transforming growth factor-β receptor type 2 axis [67]. Finally, Su et al. [68] utilized higher concentrations of propofol (12.5, 25, and 50 μg/ml) in their incubation of cardia cancer cells and had reported an inhibition of proliferation of cancer cell growth and induction of apoptosis via inhibition of the MAPK/ERK signaling pathway. Our group has shown that propofol (4 μg/ml) reduced cell viability and inhibited proliferation migration and invasion of lung cancer cells, but not in neuroglioma cells. In lung cancer cells, propofol downregulated glucose transporter 1, mitochondrial pyruvate carrier 1, p-Akt, p-Erk1/2, and HIF-1α, and upregulated pigment epithelium derived factor expression [69]. The reason for the disparity in behavior of lung cancer cells and neuroglioma cells from our experiments is uncertain and warrants further study.

Lidocaine

Lidocaine, a local anesthetic, has also been investigated for its role in cancer involvement. For instance, lidocaine (50, 100, 500 or 1000 μM) was shown to inhibit cervical cancer growth through the modulation of the IncRNA-MEG3/miR-421/BTG1 pathway [70]. The large-cell cancer line, 95D, was exposed to different concentrations of lidocaine (2, 5, and 10 μg/ml). In a dose-dependent manner , lidocaine demonstrated anti-tumor activity by inhibiting the PI3K/AKT/mTOR signaling pathway [71]. Lidocaine (50, 100, 500 or 1000 μM) has been seen to also inhibit the growth of retinoblastomas by modulation of the miR-520a-3p/EGFR axis [72] and human gastric cancers by alteration of the MAPK pathway [73].
Overall, current literature does indicate a possible association between anesthetics and anti-tumor properties [74]. Although this may provide us with potential clinical implications, we must be cautious in any interpretation as there are considerable discrepancies in the methodologies between studies. This can be ultimately reduced to different concentrations of anesthetic drugs and varying length of incubation time.

Retrospective studies

Numerous retrospective clinical studies have investigated the potential relationship between anesthetic technique and the outcomes of patients following oncological surgery. As surgical stress is thought to produce a pro-inflammatory response that favors tumor growth and metastasis, optimizing perioperative interventions, including anesthesia, may confer an improvement in long-term cancer outcomes. Furthermore, surgical resection in patients with solid tumors can lead to tumor cell release into the circulation [75,76].
There is a significant lack of prospective evidence regarding the putative relationship between anesthetic technique and post-operative outcomes in oncological surgery. The only such randomized controlled trial studied the efficacy of regional paravertebral anesthesia in combination with propofol-based total intravenous anesthesia (TIVA) vs. sevoflurane inhalational anesthesia plus opioid analgesia [77]. The study was conducted in thirteen countries and recruited 2,100 women due for primary breast cancer surgery. The authors found that propofol anesthesia with paravertebral block had no impact breast cancer recurrence compared with inhalational anesthesia and opioids (HR: 0.97, 95% CI: 0.74, 1.3; P = 0.84) [77]. As the first and potentially largest RCT of its kind, these findings are pivotal, particularly in the context of the significant amount of retrospective evidence purporting a relationship between the use of TIVA and an improvement in post-operative survival and disease recurrence compared to inhalational anesthesia.
Wigmore et al. [7] conducted the largest retrospective series of 7,030 patients over a 3-year period from one cancer center, with around half of the patients receiving TIVA with propofol and the remainder receiving volatile inhalational anesthesia. The hazard ratio (HR) for death within the inhalational cohort compared to the TIVA cohort was 1.46 (95% CI: 1.29, 1.66; P < 0.001), after multivariable analysis of known confounders and a median follow-up of 2.6 years. Furthermore, within the inhalational cohort, 87.9% of patients survived at one year, compared to 94.1% in the TIVA cohort. The authors found that the decreased survival within the inhalational group was present regardless of American Society of Anesthesiologists grade, surgical severity, or the presence of metastases at the point of operation.
These findings are supported by similar studies. Using data from a Swedish database, a retrospective study of 2,838 patients who received surgery for colon, rectal, or breast cancer found that the survival rate for patients in the propofol group was 4.7% higher at one year and 5.6% higher at five years compared to patients receiving volatile inhalational anesthesia [78]. It is important to note that the differences in this study were not significant after adjustment for confounders. An additional retrospective observational study of 922 patients who underwent esophagectomy found the inhalational anesthesia cohort had reduced overall survival (HR: 1.58, 95% CI: 1.24, 2.01; P < 0.001) and recurrence-free survival (HR: 1.42, 95% CI: 1.12, 1.79; P = 0.003) after multivariate adjustment. Similar favorable long-term outcomes with propofol–TIVA have also been found in patients undergoing gastrectomy [79] and colectomy [80].
Overall, it seems unclear whether tumor type plays a critical role in this apparent correlation; evidence suggests that the degree of surgical stress is an important determining factor. This theory seems to be supported by a retrospective analysis of 383 patients receiving modified radical mastectomy, rather than commoner and less invasive breast-conserving procedures, which found a statistically significant decrease in cancer recurrence in the group that received propofol-based TIVA (HR: 0.550, 95% CI: 0.311, 0.973; P = 0.037) [81]. However, there was no difference in overall survival between the propofol-based TIVA group and the sevoflurane group, and the study did not directly compare the outcomes of patients receiving mastectomy compared to those having breast-conserving surgery [81].
Despite the various studies that seem to suggest improved outcomes in patients receiving TIVA, it is important to note that there is a limited amount of prospective evidence, whilst the only RCT conducted suggests no benefit in post-operative outcomes with TIVA [77]. Furthermore, other retrospective studies have also reported no benefit in overall survival in patients receiving intravenous anesthesia for breast [78,82,83], lung [84], and colorectal surgery [78].
With regards to regional anesthesia, early, predominantly retrospective studies suggest that the use of regional anesthesia is associated with an improvement in overall and disease-free survival for colorectal, prostate, breast, ovarian, and head and neck malignancies [8588]. Furthermore, a randomized trial of 177 patients with colorectal cancer demonstrated a benefit associated with epidural analgesia, but this was limited to 1–5 years post-operatively [89]. A randomized study of 132 patients with cancer of abdominal organs treated with abdominal surgery receiving epidural analgesia showed a non-statistically significant improvement in recurrence-free survival, although the study was clinically underpowered [90]. Although the precise reasons for this benefit remain to be elucidated, it has been postulated it may be due to the avoidance of opioids, which have previously been shown to potentiate tumor cell survival and angiogenesis [7,91].
Despite this, post-hoc analyses of previous clinical studies, as well as randomized trials, suggest that there is limited benefit associated with regional anesthesia in the context of oncological surgery. Reanalysis of the MASTER trial is the first and largest post-hoc analysis of nearly 500 patients who had abdominal malignancy who were randomized to general anesthesia or epidural anesthesia. The study demonstrated no significant impact of epidural anesthesia on the recurrence of cancer [92]. Additionally a recent large multi-country randomized controlled trial investigating the impact of regional anesthesia-analgesia (paravertebral blocks and propofol) or general anesthesia (sevoflurane) and opioid analgesia on local or metastatic breast cancer recurrence in 2,132 women found no difference between the two groups (HR for regional anesthesia: 0.97, 95% CI: 0.74, 1.28; P = 0.84) [77].
In conclusion, studies investigating the relationship between regional anesthesia, cancer recurrence, and overall survival have yielded mixed results, with many studies suggesting no benefit [85,86]. The results of clinical studies investigating propofol-based TIVA versus inhalational anesthesia are summarized in Table 2. However, the heterogeneous, non-randomized, retrospective natures of the majority of these studies are key limiting factors.

Ongoing clinical trials

As discussed previously, evidence regarding the effects of various anesthetic techniques on surgical outcomes in patients with cancer is almost exclusively from observational, retrospective studies. The single RCT that has been conducted suggests regional anesthesia is unlikely to impact recurrence after breast cancer surgery; other tumor types may show a difference based on anesthetic technique [77]. Furthermore, an interesting new pre-clinical development suggests that peri-operative systemically administered lidocaine decreases pulmonary metastases when combined with inhalational anesthesia, thus potentially heralding a new avenue for clinical trial development [93].
There are a several large, randomized controlled trials investigating the effect of inhalational anesthetic agents vs. propofol on cancer recurrence following surgery. Results from these trials are eagerly awaited and will be highly informative in providing high quality evidence to answer and provide greater certainty as to the impact of anesthetic choice on cancer recurrence (NCT01975064 [94], NCT02660411 [95], NCT03034096 [96], ACTRN12617001065381 [97], NCT02660411 [98]).

Implications and conclusions

There is an increasing body of evidence investigating the impact of anesthesia and anesthetic techniques on cancer recurrence and survival in cancer patients. The impact of regional anesthesia vs. general anesthesia on cancer recurrence is also uncertain, with conflicting results from retrospective studies and small clinical trials. A recent large multi-country randomized controlled trial failed to show a benefit of regional anesthesia on either local or metastatic recurrence of breast cancer following surgery [77]. Further studies are required across a greater range of cancer types and more diverse patient populations to definitively prove any benefit of regional over general anesthesia on postoperative cancer recurrence.
The majority of evidence thus far suggests a benefit of the use of intravenous propofol over inhalational volatile anesthetics such as sevoflurane. This evidence is mainly pre-clinical and retrospective in nature. A recent meta-analysis that examined the effect of propofol vs. volatile anesthesia on cancer recurrence and survival found the use of propofol-based TIVA was associated with improved recurrence-free survival in all cancer types (pooled HR: 0.78, 95% CI: 0.65, 0.94; P < 0.01) and improved overall survival (pooled HR: 0.76, 95% CI: 0.63, 0.92; P < 0.01)[99]. Although this provides support that propofol is superior to volatile anesthesia in reducing cancer recurrence, the meta-analysis has several limitations. Notably nine of the ten studies included were observational studies, and heterogeneity in studies included in terms of study population, the stages of cancer and differences in use of regional anesthesia. Therefore, the results of four large randomized controlled trials investigating this question will be eagerly anticipated and will provide more definitive results as to whether propofol is superior to volatile anesthesia .
The perioperative period is characterized by physiological changes induced by surgery and perioperative interventions. These biological changes associated with the surgical inflammatory response, and the pharmacological actions of anesthetic drugs, may promote the recurrence of cancer in postoperative cancer patients. This highlights an opportunity to intervene to counteract any potentially cancer-promoting effects. Anesthesia, anesthetic technique, and other strategies such as the use of anti-adrenergic, anti-inflammatory, and anti-thrombotic therapies (which haven’t been discussed in this review) offer the potential to promote recurrence-free survival of postoperative cancer patients [6,100].

NOTES

Conflicts of Interest

No potential conflict of interest relevant to this article was reported.

Author Contributions

Azeem Alam (Data curation; Formal analysis; Investigation; Methodology; Project administration; Resources; Supervision; Writing – original draft; Writing – review & editing)

Sanketh Rampes (Methodology; Writing – original draft; Writing – review & editing)

Sonam Patel (Writing – original draft; Writing – review & editing)

Zac Hana (Writing – original draft; Writing – review & editing)

Daqing Ma (Conceptualization; Project administration; Supervision)

Table 1.
Summary of the Molecular Actions of Anesthetics Found in in vitro and in vivo Studies
Anesthetics Oncological effects
Sevoflurane Colon cancer cells:
Induces apoptosis
Inhibits proliferation and invasion as it inhibits Ras/Raf/MEK/ERK signaling pathway [46]
Ovarian cancer cells:
Inhibits migration and invasion
↓ MMP-9 and STC1 [48]
Inhibits proliferation via ↓ phosphorylation of JNK and p38 MAPK signaling pathways [49]
Potential enhanced cancer proliferation via ↑ VEGF-A, MMP-11, CXCR2, and TGF-β genes [50]
Cervical cancer cells:
Enhanced proliferation, migration, and invasion of cells via ↑ histone deacetylase 6 expression via the ERK1/2 and phosphatidylinositide 3-kinase/AKT signaling pathways [52]
Osteosarcoma cells:
Inhibits invasion and proliferation via ↓ miR-203/WNT2B/Wnt/β-catenin axis [53]
Leukemia cells:
Inhibits proliferation via ↓ Wnt/β-catenin [54]
Induces cognitive dysfunction via Wnt/β-catenin-Annexin A1 pathway [55]
Lung cancer cells:
Promotes metastases via ↑ IL-6 [56]
Glioma cells:
Inhibits growth via ↓ MMP-2 migration and activity [57]
Isoflurane Hepatic carcinoma cells:
Inhibits growth via NF-κB and PI3K/Akt signaling pathways [58]
Glioblastoma cells:
Promotes tumor and migration [59]
Propofol Human colon cancer cells:
Inhibits JAK2/STAT3 pathway
Inhibits proliferation, migration, and invasion [60]
Induces apoptosis via STAT3/HOTAIR by ↑WIF-1 and ↓Wnt pathway [61]
Adenocarcinoma alveolar basal epithelial cells:
Accelerates apoptosis via miR-21/PTEN/AKT pathway [62]
Pancreatic cancer cells:
Inhibits migration and induces apoptosis via miR-34a-mediated E-cadherin and LOC285194 signals [63]
↓ expression of ADAM8
Inhibits cell proliferation and migration via ↓ β1, ERK1/2, MMP2, and MMP9 [64]
Human gastric cells:
Inhibition of EMT, migration, and invasion [65]
Papillary thyroid cancer cells:
Inhibits proliferation and migration
↑ miR-320a and ↓ ANRIL
↓ Wnt/β-catenin and NF-κB [66]
Glioma cells:
Inhibits cell proliferation, invasion, and migration via mir-410-3p/TGFBR2 2 axis [67]
Cardia cancer cells:
Inhibits proliferation of cell growth
Induces apoptosis via inhibition of the MAPK/ERK signaling pathway [68]
Lidocaine Cervical cancer cells:
Inhibits growth via modulation of IncRNA-MEG3/miR-421/BTG1 pathway [70]
Lung cancer cells:
Inhibits proliferation, migration, and invasion via ↓ TNFα, MMP-9 secretion, and ↓ GOLPH2 in NSCLC A549 cells [74]
Retinoblastoma cells:
Inhibits tumor growth via modulation of miR-520a-3p/EGFR axis [72]
Human gastric cancer cells:
Inhibits growth via altering MAPK pathway [73]

Ras/Raf/MEK/ERK: Ras/Raf/Mitogen-activated protein kinase/ERK kinase (MEK)/extracellular-signal-regulated kinase (ERK), MMP: Matrix metalloproteinase, STC1: stanniocalcin 1, JNK: c-Jun N-terminal kinase, p38 MAPK: p38 mitogen-activated protein kinase, VEGF-A: vascular endothelial growth factor-A, CXCR2: CXC chemokine receptor 2, TGF-β: Transforming growth factor beta, miR-203: microRNA-203, WNT: wingless-type MMTV integration site, IL: interleukin, NF-κB: Nuclear factor kappa B, PI3K: phosphatidylinositol 3-kinase, Akt: protein kinase B, JAK2: Janus kinase 2, STAT3: signal transducer and activator of transcription 3, HOTAIR: HOX transcript antisense RNA, WIF1: WNT Inhibitory Factor 1, PTEN: phosphatase and tensin homolog deleted on chromosome 10, ADAM8: A Disintegrin and metalloproteinase domain-containing protein 8, ERK: extracellular signal-regulated kinase, EMT: Epithelial-mesenchymal transition, ANRIL: antisense non-coding RNA in the INK4 locus, IncRNA: long non-coding RNA, BTG1: B-cell translocation gene 1, GOLPH2: Golgi phosphoprotein 2, NSCLC: non-small cell lung cancer, EGFR: epidermal growth factor.

Table 2.
Summary of the Clinical Studies Evaluating Relative Benefit of Propofol-based TIVA vs. Inhalational Anesthesia on Cancer Recurrence and Overall Survival
Study type Anesthesia Cancer type Results
Randomized controlled trial [77] Inhalational anesthesia plus opioids vs. propofol-based TIVA Breast Propofol-based TIVA had no impact on breast cancer recurrence compared with inhalational anesthesia and opioids: HR 0.97 (95% CI: 0.74, 1.3; P = 0.84)
Retrospective analysis [7] Inhalational anesthesia vs. propofol-based TIVA Solid organ Inhalational anesthesia associated with greater HR of death: HR 1.46 (95% CI 1.29, 1.66; P < 0.001)
Retrospective analysis [78] Inhalational anesthesia vs. propofol-based TIVA Breast, colorectal Differences in overall one- and five-year survival rates for all three sites combined were 4.7% (P = 0.004) and 5.6% (P < 0.001), respectively, in favor of propofol.
Retrospective analysis [79] Inhalational anesthesia vs. propofol-based TIVA Gastric TIVA was associated with a HR of 0.67 (95% CI: 0.58, 0.77) for death in univariate analysis and 0.65 (95% CI: 0.56, 0.75) after a multivariate analysis of known confounders in the matched group.
Retrospective analysis [80] Inhalational anesthesia vs. propofol-based TIVA Colon (HR: 0.22, 95% CI: 0.11, 0.42; P < 0.001) or higher tumor-node-metastasis stage (HR: 0.42, 95% CI: 0.32, 0.55; P < 0.001) and presence of metastases (HR: 0.67, 95% CI: 0.51, 0.86; P = 0.002) or absence of metastases (HR: 0.08, 95% CI: 0.01, 0.62; P = 0.016)
Retrospective analysis [81] Inhalational anesthesia vs. propofol-based TIVA Breast Propofol group showed a lower rate of cancer recurrence (P = 0.037), with an estimated HR of 0.550 (95% CI: 0.311, 0.973).
Retrospective analysis [82] Inhalational anesthesia vs. propofol-based TIVA Breast No association found using Cox regression analyses and propensity matching.
Retrospective analysis [83] Inhalational anesthesia vs. propofol-based TIVA Breast Kaplan-Meier survival curves showed no significant difference in recurrence-free or overall survival between the two groups.
Retrospective analysis [84] Inhalational anesthesia vs. propofol-based TIVA Lung No significant difference in HR for recurrence (P = 0.233) or HR for death (P = 0.551) between the two groups.

TIVA: total intravenous anesthesia, HR: hazard ratio.

References

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018; 68: 394-424.
crossref pmid
2. Pilleron S, Sarfati D, Janssen-Heijnen M, Vignat J, Ferlay J, Bray F, et al. Global cancer incidence in older adults, 2012 and 2035: a population-based study. Int J Cancer 2019; 144: 49-58.
crossref pmid
3. Sullivan R, Alatise OI, Anderson BO, Audisio R, Autier P, Aggarwal A, et al. Global cancer surgery: delivering safe, affordable, and timely cancer surgery. Lancet Oncol 2015; 16: 1193-224.
crossref pmid
4. Dąbrowska AM, Słotwiński R. The immune response to surgery and infection. Cent Eur J Immunol 2014; 39: 532-7.
crossref pmid pmc
5. Rampes S, Ma K, Divecha YA, Alam A, Ma D. Postoperative sleep disorders and their potential impacts on surgical outcomes. J Biomed Res 2019; 34: 271-80.
crossref pmid
6. Perry NJ, Buggy D, Ma D. Can anesthesia influence cancer outcomes after surgery? JAMA Surg 2019; 154: 279-80.
crossref pmid
7. Wigmore TJ, Mohammed K, Jhanji S. Long-term survival for patients undergoing volatile versus IV anesthesia for cancer surgery: a retrospective analysis. Anesthesiology 2016; 124: 69-79.
crossref pmid
8. Jun IJ, Jo JY, Kim JI, Chin JH, Kim WJ, Kim HR, et al. Impact of anesthetic agents on overall and recurrence-free survival in patients undergoing esophageal cancer surgery: a retrospective observational study. Sci Rep 2017; 7: 14020.
crossref pmid pmc
9. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144: 646-74.
crossref pmid
10. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971; 285: 1182-6.
crossref pmid
11. Tonini T, Rossi F, Claudio PP. Molecular basis of angiogenesis and cancer. Oncogene 2003; 22: 6549-56.
crossref pmid
12. Behrens J. The role of cell adhesion molecules in cancer invasion and metastasis. Breast Cancer Res Treat 1993; 24: 175-84.
crossref pmid
13. Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer 2003; 3: 453-8.
crossref pmid
14. 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: 213-26.
crossref pmid pmc
15. Lee YM, Song BC, Yeum KJ. Impact of volatile anesthetics on oxidative stress and inflammation. Biomed Res Int 2015; 2015: 242709.
crossref pmid pmc
16. Kurosawa S, Kato M. Anesthetics, immune cells, and immune responses. J Anesth 2008; 22: 263-77.
crossref pmid
17. Welden B, Gates G, Mallari R, Garrett N. Effects of anesthetics and analgesics on natural killer cell activity. AANA J 2009; 77: 287-92.
pmid
18. Loop T, Dovi-Akue D, Frick M, Roesslein M, Egger L, Humar M, et al. Volatile anesthetics induce caspase-dependent, mitochondria-mediated apoptosis in human T lymphocytes in vitro. Anesthesiology 2005; 102: 1147-57.
crossref pmid
19. Wu L, Zhao H, Wang T, Pac-Soo C, Ma D. Cellular signaling pathways and molecular mechanisms involving inhalational anesthetics-induced organoprotection. J Anesth 2014; 28: 740-58.
crossref pmid
20. Benzonana LL, Perry NJ, Watts HR, Yang B, Perry IA, Coombes C, et al. 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.
crossref pmid
21. Kripke BJ, Kupferman A, Luu KC. Suppression of chemotaxis to corneal inflammation by nitrous oxide. Zhonghua Min Guo Wei Sheng Wu Ji Mian Yi Xue Za Zhi 1987; 20: 302-10.
pmid
22. Schneemilch CE, Hachenberg T, Ansorge S, Ittenson A, Bank U. Effects of different anaesthetic agents on immune cell function in vitro. Eur J Anaesthesiol 2005; 22: 616-23.
crossref pmid
23. Weimann J. Toxicity of nitrous oxide. Best Pract Res Clin Anaesthesiol 2003; 17: 47-61.
crossref pmid
24. 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.
crossref pmid
25. Huang H, Benzonana LL, Zhao H, Watts HR, Perry NJ, Bevan C, et al. Prostate cancer cell malignancy via modulation of HIF-1α pathway with isoflurane and propofol alone and in combination. Br J Cancer 2014; 111: 1338-49.
crossref pmid pmc
26. 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: 1331-9.
crossref pmid
27. 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: 233-40.
crossref pmid
28. He H, Chen J, Xie WP, Cao S, Hu HY, Yang LQ, et al. Ketamine used as an acesodyne in human breast cancer therapy causes an undesirable side effect, upregulating anti-apoptosis protein Bcl-2 expression. Genet Mol Res 2013; 12: 1907-15.
crossref pmid
29. Beilin B, Rusabrov Y, Shapira Y, Roytblat L, Greemberg L, Yardeni IZ, et al. Low-dose ketamine affects immune responses in humans during the early postoperative period. Br J Anaesth 2007; 99: 522-7.
crossref pmid
30. Sakaguchi M, Kuroda Y, Hirose M. The antiproliferative effect of lidocaine on human tongue cancer cells with inhibition of the activity of epidermal growth factor receptor. Anesth Analg 2006; 102: 1103-7.
crossref pmid
31. Brackenbury WJ. Voltage-gated sodium channels and metastatic disease. Channels (Austin) 2012; 6: 352-61.
crossref pmid pmc
32. Galley HF, Dubbels AM, Webster NR. The effect of midazolam and propofol on interleukin-8 from human polymorphonuclear leukocytes. Anesth Analg 1998; 86: 1289-93.
crossref pmid
33. Halapy E, Kreiger N, Cotterchio M, Sloan M. Benzodiazepines and risk for breast cancer. Ann Epidemiol 2006; 16: 632-6.
crossref pmid
34. Franchi S, Panerai AE, Sacerdote P. Buprenorphine ameliorates the effect of surgery on hypothalamus-pituitary-adrenal axis, natural killer cell activity and metastatic colonization in rats in comparison with morphine or fentanyl treatment. Brain Behav Immun 2007; 21: 767-74.
crossref pmid
35. Das J, Kumar S, Khanna S, Mehta Y. Are we causing the recurrence-impact of perioperative period on long-term cancer prognosis: review of current evidence and practice. J Anaesthesiol Clin Pharmacol 2014; 30: 153-9.
crossref pmid pmc
36. Khabbazi S, Nassar ZD, Goumon Y, Parat MO. Morphine decreases the pro-angiogenic interaction between breast cancer cells and macrophages in vitro. Sci Rep 2016; 6: 31572.
crossref pmid pmc
37. Koodie L, Yuan H, Pumper JA, Yu H, Charboneau R, Ramkrishnan S, et al. Morphine inhibits migration of tumor-infiltrating leukocytes and suppresses angiogenesis associated with tumor growth in mice. Am J Pathol 2014; 184: 1073-84.
crossref pmid pmc
38. Beilin B, Shavit Y, Hart J, Mordashov B, Cohn S, Notti I, et al. Effects of anesthesia based on large versus small doses of fentanyl on natural killer cell cytotoxicity in the perioperative period. Anesth Analg 1996; 82: 492-7.
crossref pmid
39. Shavit Y, Ben-Eliyahu S, Zeidel A, Beilin B. Effects of fentanyl on natural killer cell activity and on resistance to tumor metastasis in rats. Dose and timing study. Neuroimmunomodulation 2004; 11: 255-60.
crossref pmid
40. Tai YH, Wu HL, Chang WK, Tsou MY, Chen HH, Chang KY. Intraoperative fentanyl consumption does not impact cancer recurrence or overall survival after curative colorectal cancer resection. Sci Rep 2017; 7: 10816.
crossref pmid pmc
41. Gaspani L, Bianchi M, Limiroli E, Panerai AE, Sacerdote P. The analgesic drug tramadol prevents the effect of surgery on natural killer cell activity and metastatic colonization in rats. J Neuroimmunol 2002; 129: 18-24.
crossref pmid
42. Wigmore T, Farquhar-Smith P. Opioids and cancer: friend or foe? Curr Opin Support Palliat Care 2016; 10: 109-18.
pmid
43. Singleton PA, Mirzapoiazova T, Hasina R, Salgia R, Moss J. Increased μ-opioid receptor expression in metastatic lung cancer. Br J Anaesth 2014; 113 Suppl 1(Suppl 1): i103-8.
crossref pmid
44. Zylla D, Gourley BL, Vang D, Jackson S, Boatman S, Lindgren B, et al. Opioid requirement, opioid receptor expression, and clinical outcomes in patients with advanced prostate cancer. Cancer 2013; 119: 4103-10.
crossref pmid
45. Janku F, Johnson LK, Karp DD, Atkins JT, Singleton PA, Moss J. Treatment with methylnaltrexone is associated with increased survival in patients with advanced cancer. Ann Oncol 2016; 27: 2032-8.
crossref pmid pmc
46. Yang X, Zheng YT, Rong W. Sevoflurane induces apoptosis and inhibits the growth and motility of colon cancer in vitro and in vivo via inactivating Ras/Raf/MEK/ERK signaling. Life Sci 2019; 239: 116916.
crossref pmid
47. Bundscherer AC, Ullrich V, Malsy M, Gruber MA, Graf BM, Brockhoff G, et al. Effects of volatile anesthetics on proliferation and viability of SW480 colon cancer cells in vitro. Anticancer Res 2019; 39: 6049-55.
crossref pmid
48. 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.
crossref pmid pmc
49. Kang K, Wang Y. Sevoflurane inhibits proliferation and invasion of human ovarian cancer cells by regulating JNK and p38 MAPK signaling pathway. Drug Des Devel Ther 2019; 13: 4451-60.
pmid pmc
50. Iwasaki M, Zhao H, Jaffer T, Unwith S, Benzonana L, Lian Q, et al. Volatile anaesthetics enhance the metastasis related cellular signalling including CXCR2 of ovarian cancer cells. Oncotarget 2016; 7: 26042-56.
crossref pmid pmc
51. Xue F, Xu Y, Song Y, Zhang W, Li R, Zhu X. The effects of sevoflurane on the progression and cisplatinum sensitivity of cervical cancer cells. Drug Des Devel Ther 2019; 13: 3919-28.
pmid pmc
52. Zhang W, Sheng B, Chen S, Zhao H, Wu L, Sun Y, et al. Sevoflurane enhances proliferation, metastatic potential of cervical cancer cells via the histone deacetylase 6 modulation in vitro. Anesthesiology 2020; 132: 1469-81.
crossref pmid
53. Chen M, Zhou L, Liao Z, Ye X, Xuan X, Gu B, et al. Sevoflurane inhibited osteosarcoma cell proliferation and invasion via targeting miR-203/WNT2B/Wnt/β-catenin axis. Cancer Manag Res 2019; 11: 9505-15.
pmid pmc
54. Ruan X, Jiang W, Cheng P, Huang L, Li X, He Y, et al. Volatile anesthetics sevoflurane targets leukemia stem/progenitor cells via Wnt/β-catenin inhibition. Biomed Pharmacother 2018; 107: 1294-301.
crossref pmid
55. Hu N, Wang C, Zheng Y, Ao J, Zhang C, Xie K, et al. The role of the Wnt/β-catenin-annexin A1 pathway in the process of sevoflurane-induced cognitive dysfunction. J Neurochem 2016; 137: 240-52.
crossref pmid
56. Li R, Huang Y, Lin J. Distinct effects of general anesthetics on lung metastasis mediated by IL-6/JAK/STAT3 pathway in mouse models. Nat Commun 2020; 11: 642.
crossref pmid pmc
57. Hurmath FK, Mittal M, Ramaswamy P, Umamaheswara Rao GS, Dalavaikodihalli Nanjaiah N. Sevoflurane and thiopental preconditioning attenuates the migration and activity of MMP-2 in U87MG glioma cells. Neurochem Int 2016; 94: 32-8.
crossref pmid
58. Hu J, Hu J, Jiao H, Li Q. Anesthetic effects of isoflurane and the molecular mechanism underlying isoflurane‑inhibited aggressiveness of hepatic carcinoma. Mol Med Rep 2018; 18: 184-92.
crossref pmid pmc
59. Zhu M, Li M, Zhou Y, Dangelmajer S, Kahlert UD, Xie R, et al. Isoflurane enhances the malignant potential of glioblastoma stem cells by promoting their viability, mobility in vitro and migratory capacity in vivo. Br J Anaesth 2016; 116: 870-7.
crossref pmid
60. Liang B, Dong T. Effects of propofol on invasion and migration of colon cancer cells and JAK2/STAT3 signaling pathway. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2020; 45: 290-6.
pmid
61. Zhang YF, Li CS, Zhou Y, Lu XH. Effects of propofol on colon cancer metastasis through STAT3/HOTAIR axis by activating WIF-1 and suppressing Wnt pathway. Cancer Med 2020; 9: 1842-54.
crossref pmid pmc
62. Zheng X, Dong L, Zhao S, Li Q, Liu D, Zhu X, et al. Propofol affects non-small-cell lung cancer cell biology by regulating the miR-21/PTEN/AKT pathway in vitro and in vivo. Anesth Analg 2020; 131: 1270-80.
crossref pmid
63. Wang H, Jiao H, Jiang Z, Chen R. Propofol inhibits migration and induces apoptosis of pancreatic cancer PANC-1 cells through miR-34a-mediated E-cadherin and LOC285194 signals. Bioengineered 2020; 11: 510-21.
crossref pmid pmc
64. Yu X, Shi J, Wang X, Zhang F. Propofol affects the growth and metastasis of pancreatic cancer via ADAM8. Pharmacol Rep 2020; 72: 418-26.
crossref pmid
65. Liu F, Qiu F, Fu M, Chen H, Wang H. Propofol reduces epithelial to mesenchymal transition, invasion and migration of gastric cancer cells through the microRNA-195-5p/Snail axis. Med Sci Monit 2020; 26: e920981.
crossref pmid pmc
66. Li M, Qu L, Chen F, Zhu X. Propofol upregulates miR-320a and reduces HMGB1 by downregulating ANRIL to inhibit PTC cell malignant behaviors. Pathol Res Pract 2020; 216: 152856.
crossref pmid
67. Li F, Li F, Chen W. Propofol inhibits cell proliferation, migration, and invasion via mir-410-3p/transforming growth factor-β receptor type 2 (TGFBR2) axis in glioma. Med Sci Monit 2020; 26: e919523.
crossref pmid pmc
68. Su Z, Liu HL, Qi B, Liu Y. Effects of propofol on proliferation and apoptosis of cardia cancer cells via MAPK/ERK signaling pathway. Eur Rev Med Pharmacol Sci 2020; 24: 428-33.
pmid
69. Hu C, Iwasaki M, Liu Z, Wang B, Li X, Lin H, et al. Lung but not brain cancer cell malignancy inhibited by commonly used anesthetic propofol during surgery: implication of reducing cancer recurrence risk. J Adv Res 2021. Advance Access published on Jan 6, 2021. doi: 10.1016/j.jare.2020.12.007.
crossref
70. Zhu J, Han S. Lidocaine inhibits cervical cancer cell proliferation and induces cell apoptosis by modulating the lncRNA-MEG3/miR-421/BTG1 pathway. Am J Transl Res 2019; 11: 5404-16.
pmid pmc
71. Dong Q, Mao Z. The local anaesthetic lignocaine exhibits potent antilung cancer cell activity by inhibiting the phosphoinositide 3-kinases/mammalian target of rapamycin/mammalian target of rapamycin pathway. Pharmacology 2019; 104: 139-46.
crossref pmid
72. Xia W, Wang L, Yu D, Mu X, Zhou X. Lidocaine inhibits the progression of retinoblastoma in vitro and in vivo by modulating the miR‑520a‑3p/EGFR axis. Mol Med Rep 2019; 20: 1333-42.
crossref pmid pmc
73. Ye L, Zhang Y, Chen YJ, Liu Q. Anti-tumor effects of lidocaine on human gastric cancer cells in vitro. Bratisl Lek Listy 2019; 120: 212-7.
crossref pmid
74. Zhou D, Wang L, Cui Q, Iftikhar R, Xia Y, Xu P. Repositioning lidocaine as an anticancer drug: the role beyond anesthesia. Front Cell Dev Biol 2020; 8: 565.
crossref pmid pmc
75. Eschwège P, Dumas F, Blanchet P, Le Maire V, Benoit G, Jardin A, et al. Haematogenous dissemination of prostatic epithelial cells during radical prostatectomy. Lancet 1995; 346: 1528-30.
crossref pmid
76. Yamaguchi K, Takagi Y, Aoki S, Futamura M, Saji S. Significant detection of circulating cancer cells in the blood by reverse transcriptase-polymerase chain reaction during colorectal cancer resection. Ann Surg 2000; 232: 58-65.
crossref pmid pmc
77. Sessler DI, Pei L, Huang Y, Fleischmann E, Marhofer P, Kurz A, et al. Recurrence of breast cancer after regional or general anaesthesia: a randomised controlled trial. Lancet 2019; 394: 1807-15.
crossref pmid
78. Enlund M, Berglund A, Andreasson K, Cicek C, Enlund A, Bergkvist L. The choice of anaesthetic--sevoflurane or propofol--and outcome from cancer surgery: a retrospective analysis. Ups J Med Sci 2014; 119: 251-61.
crossref pmid pmc
79. Zheng X, Wang Y, Dong L, Zhao S, Wang L, Chen H, et al. Effects of propofol-based total intravenous anesthesia on gastric cancer: a retrospective study. Onco Targets Ther 2018; 11: 1141-8.
crossref pmid pmc
80. Wu ZF, Lee MS, Wong CS, Lu CH, Huang YS, Lin KT, et al. Propofol-based total intravenous anesthesia is associated with better survival than desflurane anesthesia in colon cancer surgery. Anesthesiology 2018; 129: 932-41.
crossref pmid
81. Lee JH, Kang SH, Kim Y, Kim HA, Kim BS. Effects of propofol-based total intravenous anesthesia on recurrence and overall survival in patients after modified radical mastectomy: a retrospective study. Korean J Anesthesiol 2016; 69: 126-32.
crossref pmid pmc
82. Kim MH, Kim DW, Kim JH, Lee KY, Park S, Yoo YC. Does the type of anesthesia really affect the recurrence-free survival after breast cancer surgery? Oncotarget 2017; 8: 90477-87.
crossref pmid pmc
83. Yoo S, Lee HB, Han W, Noh DY, Park SK, Kim WH, et al. Total intravenous anesthesia versus inhalation anesthesia for breast cancer surgery: a retrospective cohort study. Anesthesiology 2019; 130: 31-40.
crossref pmid
84. Oh TK, Kim K, Jheon S, Lee J, Do SH, Hwang JW, et al. Long-term oncologic outcomes for patients undergoing volatile versus intravenous anesthesia for non-small cell lung cancer surgery: a retrospective propensity matching analysis. Cancer Control 2018; 25: 1073274818775360.
pmid pmc
85. Heaney A, Buggy DJ. Can anaesthetic and analgesic techniques affect cancer recurrence or metastasis? Br J Anaesth 2012; 109 Suppl 1: i17-28.
crossref pmid
86. Grandhi RK, Lee S, Abd-Elsayed A. The relationship between regional anesthesia and cancer: a metaanalysis. Ochsner J 2017; 17: 345-61.
pmid pmc
87. Biki B, Mascha E, Moriarty DC, Fitzpatrick JM, Sessler DI, Buggy DJ. Anesthetic technique for radical prostatectomy surgery affects cancer recurrence: a retrospective analysis. Anesthesiology 2008; 109: 180-7.
pmid
88. Chen WK, Miao CH. The effect of anesthetic technique on survival in human cancers: a meta-analysis of retrospective and prospective studies. PLoS One 2013; 8: e56540.
crossref pmid pmc
89. Christopherson R, James KE, Tableman M, Marshall P, Johnson FE. Long-term survival after colon cancer surgery: a variation associated with choice of anesthesia. Anesth Analg 2008; 107: 325-32.
crossref pmid
90. Binczak M, Tournay E, Billard V, Rey A, Jayr C. Major abdominal surgery for cancer: does epidural analgesia have a long-term effect on recurrence-free and overall survival? Ann Fr Anesth Reanim 2013; 32: e81-8.
crossref pmid
91. Juneja R. Opioids and cancer recurrence. Curr Opin Support Palliat Care 2014; 8: 91-101.
crossref pmid
92. Myles PS, Peyton P, Silbert B, Hunt J, Rigg JR, Sessler DI. Perioperative epidural analgesia for major abdominal surgery for cancer and recurrence-free survival: randomised trial. BMJ 2011; 342: d1491.
crossref pmid
93. Johnson MZ, Crowley PD, Foley AG, Xue C, Connolly C, Gallagher HC, et al. Effect of perioperative lidocaine on metastasis after sevoflurane or ketamine-xylazine anaesthesia for breast tumour resection in a murine model. Br J Anaesth 2018; 121: 76-85.
crossref pmid
94. Enlund M. Cancer and anaesthesia: survival after radical surgery—a comparison between propofol or sevoflurane anaesthesia (CAN) [Internet]. Bethesda (MD): clinicaltrials.gov; 2013 Nov 3 [updated 2019 May 6; cited 2020 Dec 26]. Available from https://clinicaltrials.gov/ct2/show/NCT01975064

95. Wang DX. Impact of anaesthesia maintenance methods on long-term survival [Internet]. Bethesda (MD): clinicaltrials.gov; 2016 Jan 21 [updated 2021 Feb 10; cited 2020 Dec 26]. Available from https://clinicaltrials.gov/ct2/show/NCT02660411

96. Bennett-Guerrero E. General anesthetics in CAncer REsection Surgery (GA-CARES) trial: (GA-CARES) [Internet]. Bethesda (MD): clinicaltrials.gov; 2017 Jan 27 [updated 2020 Dec 30; cited 2020 Dec 26]. Available from https://clinicaltrials.gov/ct2/show/NCT03034096

97. Riedel B. Volatile anaesthesia and perioperative outcomes related to cancer (VAPOR-C): a feasibility study [Internet]. Camperdown: Australian New Zealand Clinical Trials Registry 2017 Jul 21 [updated 2021 May 25; cited 2020 Dec 26]. Available from https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=373249&isReview=true

98. Zhang Y, Li HJ, Wang DX, Jia HQ, Sun XD, Pan LH, et al. Impact of inhalational versus intravenous anaesthesia on early delirium and long-term survival in elderly patients after cancer surgery: study protocol of a multicentre, open-label, and randomised controlled trial. BMJ Open 2017; 7: e018607.
crossref pmid pmc
99. Yap A, Lopez-Olivo MA, Dubowitz J, Hiller J, Riedel B. Anesthetic technique and cancer outcomes: a meta-analysis of total intravenous versus volatile anesthesia. Can J Anaesth 2019; 66: 546-61.
crossref pmid
100. Hiller JG, Perry NJ, Poulogiannis G, Riedel B, Sloan EK. Perioperative events influence cancer recurrence risk after surgery. Nat Rev Clin Oncol 2018; 15: 205-18.
crossref pmid


ABOUT
ARTICLE CATEGORY

Browse all articles >

BROWSE ARTICLES
AUTHOR INFORMATION
Editorial Office
101-3503, Lotte Castle President, 109 Mapo-daero, Mapo-gu, Seoul 04146, Korea
Tel: +82-2-792-5128    Fax: +82-2-792-4089    E-mail: journal@anesthesia.or.kr                

Copyright © 2024 by Korean Society of Anesthesiologists.

Developed in M2PI

Close layer
prev next