Anesth Pain Med Search

CLOSE


Anesth Pain Med > Volume 19(2); 2024 > Article
Ahn: Anesthesia and cancer recurrence: a narrative review

Abstract

Cancer is a leading cause of death worldwide. With the increasingly aging population, the number of emerging cancer cases is expected to increase markedly in the foreseeable future. Surgical resection with adjuvant therapy is the best available option for the potential cure of many solid tumors; thus, approximately 80% of patients with cancer undergo at least one surgical procedure during their disease. Agents used in general anesthesia can modulate cytokine release, transcription factors, and/or oncogenes. This can affect host immunity and the capability of cancer cells to survive and migrate, not only during surgery but for up to several weeks after surgery. However, it remains unknown whether exposure to anesthetic agents affects cancer recurrence or metastasis. This review explores the current literature to explain whether and how the choice of anesthetic and perioperative medication affect cancer surgery outcomes.

INTRODUCTION

Today, the biology and treatment of cancer are better understood than ever before, but cancer remains the leading cause of death, with its number continuously increasing as the population ages [1-3].
Surgical resection with adjuvant therapy is the best available option for the potential cure of many solid tumors; thus, approximately 80% of patients with cancer undergo at least one surgical procedure during their disease [4].
However, even with the best technique, incomplete resection margins or iatrogenic “seeding” of tumor cells with surgical manipulation can lead to a fraction of cancer cells remaining in the body. Some patients already have micrometastases at the time of surgery [5-10]. Whether the remaining cancer cells lead to recurrence and distant metastases depends on the balance between the tumor’s capability to seed, proliferate, and promote angiogenesis and the anti-metastatic immunity of the host [11-13].
To date, numerous in vitro, in vivo and retrospective studies have shown that anesthetics, opioids, and other perioperative medications influence cancer cell activity and survival, either directly and indirectly, through altering the neuroendocrine stress response to surgery, cancer cell signaling, and the host immunity [2,7,14-54].
This review reports the latest data regarding the influence of anesthetic agents on cancer recurrence and metastasis. To date, there are no official recommendations for the best anesthetic for patients with cancer. However, some anesthetic agents may increase this risk, whereas others may reduce it.
To explore how anesthesia can affect cancer progression, I will briefly present tumor biology, potential targets for cancer control, and the effects of various commonly used anesthetics on various cancers.

BASIC TUMOR BIOLOGY

Recurrence and metastatic transition occur via three basic mechanisms [55,56]. Local recurrence occurs when surviving cancer cells proliferate at the resection site through the activation of proinflammatory cytokines, pro-oncogenes, and angiogenic factors. Second, the cancer cells were seeded during surgical manipulation. Third, cancer cells transform and gain the capability to migrate to distant sites via vascular or lymphatic spread through the activation and mutation of oncogenes.
Multiple perioperative factors create a protumor environment. The physiological stress response to surgery and tissue damage causes immunosuppression through the release of various hormonal mediators such as catecholamines, prostaglandins, and growth factors [57]. The release of cortisol, catecholamines, and cytokines (such as interleukin-6 [IL-6] and prostaglandins such as prostaglandin E2 [PGE2]) suppresses the function of immune cells, including natural killer (NK) cells, which are the primary defense against cancer, and CD8+ T cells. In addition, pro-oncogenic cell lines, regulatory T cells, and type 2 helper T cells are activated and proliferated [14,58]. Prostaglandins and catecholamines can activate β2-adrenergic [59] and cyclooxygenase-2 receptors [60] that may help metastasis.
Tissue hypoxia at the site of injury upregulates hypoxia inducible factor 1α (HIF-1α), which promotes cell proliferation, angiogenesis, and metastasis [61]. HIF-1α confers a survival advantage to tumor cells [18,62-64] and stimulates tumor growth by triggering vascular endothelial growth factor (VEGF) [65], which accelerates angiogenesis [66] and can promote remodeling of lymphatics for metastasis [67]. This may cause local tumors to metastasize, which is referred to as epithelial-to-mesenchymal transition [68]. This proinflammatory and immunosuppressive phenomenon usually persists for up to 1 week after surgery [69]. However, its duration may extend beyond 1 week depending on the surgical stress [39,57].
Therefore, it is logical to believe that anesthesiologists use the sympatholytic, anti-inflammatory, and immunomodulatory effects of various anesthetics to inhibit cancer progression and improve patient outcomes. Ideally, anesthetics should 1) attenuate inflammation, 2) promote NK and CD8+ cell activity, and 3) attenuate the transcription factors and oncogenes that promote cancer cell survival and metastasis.

VOLATILE VS. PROPOFOL-BASED TOTAL INTRAVENOUS ANESTHESIA (TIVA)

Numerous laboratory studies showed that volatile anesthetics may enhance metastasis by directly promoting cancer cells and inhibiting immune cells [34,70-72], and that propofol has antitumor effects and prevents perioperative immune suppression through its anti-inflammatory and antioxidant actions [73]; in vitro and in vivo studies have shown that volatile anesthetics directly impair macrophages, dendritic cells, neutrophils, T-cells, and NK cells. Volatile anesthetics also upregulate HIF-1α, VEGF, insulin-like growth factor 1, and phosphoinositide 3-kinase- Akt pathway (PI3K-Akt pathway) and have anti-apoptotic effects. All of these factors can accelerate the proliferation and metastasis of minimally invasive cancer cells [33,37,61,69,70,74]. Exposure of breast, ovarian, and kidney cancer cells to volatile anesthetics increases IL-1/6/8 and tumor necrosis factor (TNF), suppresses NK and T-cells, and increases angiogenic and migration factors [3,16,75-77]. A 2016 systematic review of multiple in vivo studies found that volatile anesthetics are associated with an increased incidence of metastasis [78]. In contrast, propofol shows antitumor effects by directly regulating key ribonucleic acid pathways and signaling in laboratory studies [79]. Propofol inhibits the proliferation, invasion, and migration of gastric cancer cell lines [80]. In non-small cell lung cancer (NSCLC), propofol reduced cancer cell migration and invasion by inhibiting HIF-1α [81]. Propofol reduces the expression of neuroepithelial cell transforming gene 1, which increases the migration of breast cancer cell lines [72] and accelerates cell apoptosis through the miR-24/p27 pathway [82]. Propofol decreases the expression of the sex-determining region Y-box (SOX4) gene [83,84], which is associated with poor prognosis in endometrial and esophageal squamous cell carcinoma cells [85].
In animal models (breast cancer), propofol did not increase metastasis or inhibit NK cell activity, whereas halothane did [31]. In another animal model (breast cancer), propofol reduced lung metastasis compared with sevoflurane [86]. Excised breast cancer specimens from patients who received propofol with a paravertebral block showed increased NK cell infiltration into breast cancer tissue compared to patients who received volatile anesthetics [71]. A small study of head and neck cancer resection found that expression of HIF-1α was increased in patients who received volatile anesthetics [87]. Serum from patients who received the regional combined propofol technique for colon cancer resection inhibited the proliferation and invasion of cultured colon cancer cells and showed a higher rate of cell apoptosis than that from patients who received volatile/opioid technique [88]. In another study of breast cancer surgery, application of serum from females who received propofol with paravertebral block preserved NK cell activity and increased cancer cell apoptosis compared to that from females who received sevoflurane/opioid anesthesia [34,35]. In a meta-analysis, in vitro and translational data showed that volatile anesthetics are potent immunosuppressive and tumorigenic agents that promote metastasis, whereas propofol is anti-inflammatory and anti-tumorigenic and prevents metastasis [89].
An important question is whether the known effects of volatile anesthetics have been clinically demonstrated. Most retrospective clinical studies comparing propofol with volatile anesthetics have reported conflicting results. Some studies have shown a beneficial effect of TIVA compared to volatile anesthetics, while others have shown no difference [40-42,45,47,49,90-93].
Wigmore et al. [41] conducted a retrospective propensity score-matched cohort analysis (n = 7,030) on patients who underwent various types of cancer surgery. Overall survival was improved in patients administered propofol rather than volatile anesthesia (15.6% vs. 22.8% 5-year mortality after surgery; hazard ratio [HR], 0.68; 95% CI, 0.60 to 0.78; P < 0.001). These findings are in line with those of other retrospective studies that have shown improved overall survival with propofol-based TIVA in esophageal (n = 922) [44], gastric (n = 2,856) [43], and colon (n = 1,363) [94] cancer surgery. A meta-analysis based primarily on retrospective single-institutional series repeatedly found that propofol-based TIVA was associated with increased disease-free and overall survival in many solid tumors [89]; these associations were stronger when surgery was longer, surgical trauma was more severe, and malignancy was more aggressive [89]. A 2019 meta-analysis from five retrospective studies and one small randomized controlled trial (n > 7,800, breast, esophageal, or NSCLC surgery) reported that the use of propofol-based TIVA was associated with improved recurrence-free survival compared to volatile anesthesia (pooled HR, 0.78; 95% CI, 0.65-0.94) [95]. Enlund et al. [93] identified all patients (n = 6,305) who received anesthesia for breast cancer surgery in the Swedish Breast Cancer Quality Register between 2006 and 2012. In the final model, the 5-year survival rates in the propofol and sevoflurane groups were 91.0% and 81.8%, respectively (P = 0.126). However, the results ranged from ‘non-significant’ to a ‘proposed’ and even ‘determined" difference in survival, with propofol having up to 9.2% higher 5 year survival rate than sevoflurane (HR, 1.46; 95% CI, 1.10-1.95) depending on the statistical adjustment method used [93]. A retrospective Danish database analysis (n > 8,600, propensity-score matched) showed a slight increase in cancer recurrence with volatile anesthetic use compared with propofol-based TIVA in patients undergoing colorectal cancer surgery (HR, 1.12; 95% CI, 1.02-1.13) but no difference in overall survival [46].
However, there are also different results. In a retrospective surgery, Yoo et al. [45] reported no benefit in cancer recurrence and overall survival with propofol-based TIVA compared with volatile anesthesia in breast cancer surgery (n = 5,331). These results are in line with those of other retrospective studies that reported no difference in overall survival for breast (n = 2,645 [96] and n = 1,217 [90]), colorectal (n = 1,297) [90], lung (n = 943) [91], and oral cancer surgery (n = 604) [97]. This may be supported by some preclinical and clinical studies suggesting that volatile anesthetics have no effect on cancer cells or rather have an inhibitory effect. Sevoflurane arrested the cell cycle in the G1 phase and inhibited the proliferation of breast cancer cells [98]. Volatile anesthetics increase the chemosensitivity to cisplatin (a chemotherapeutic agent) [77] and suppress the growth and migration [99] of NSCLC cells. During breast cancer resection, the release of VEGF-C did not differ between propofol-based TIVA and sevoflurane anesthesia [100]. A randomized trial of breast cancer (n = 210) found similar numbers of circulating tumor cells in the blood of patients after anesthesia with sevoflurane and propofol [51]. Among patients undergoing breast cancer surgery, the expression of regulatory T-cell enzymes that promote cancer recurrence did not differ between the patients who received volatile anesthesia and those who received propofol-based TIVA [101]. In a randomized trial of colorectal cancer (n = 153), sevoflurane and propofol anesthesia resulted in similar fractions of circulating NK, helper T, and cytotoxic T cells after surgery [102]. In a meta-analysis of clinical studies, inflammatory biomarkers (IL6, IL10, TNF-α, and C-reactive protein [CRP]) did not differ between propofol-based TIVA and sevoflurane anesthesia groups [103].
Large randomized clinical trials are needed because of the heterogeneity associated with surgical scope, cancer type, and patient characteristics, as well as other limitations associated with the retrospective nature of most previous studies. Only one large randomized trial has been published, the Cancer and Anesthesia Study (CAN NCT01975064), which evaluated the survival of patients with breast cancer after exposure to general anesthesia (n =1 ,670) [52]. Overall survival did not differ between the propofol-based TIVA and sevoflurane anesthesia groups. Five year survival was 773/841 (91.9% [95% CI, 90.1-93.8]) in the propofol-based TIVA group and 764/829 (92.2% [90.3-94.0]) in the sevoflurane group, (HR, 1.03; 95% CI, 0.73-1.44, P = 0.875). Thus, propofol-based TIVA did not increase overall survival compared to sevoflurane anesthesia. Another large multicenter trial by Sessler et al. [53] compared regional (paravertebral block/propofol sedation) and general (sevoflurane) anesthesia in patients with breast cancer (n = 2,132). Regional anesthesia with propofol sedation did not reduce breast cancer recurrence at a median follow-up of 3 years when compared to general anesthesia with sevoflurane [HR, 0.97; 95% CI, 0·74-1·28; P = 0.84]. However, the study design was primarily a comparison between regional and general anesthesia and allowed for supplemental low-dose sevoflurane in the case of insufficient analgesia or sedation in the regional anesthesia group, potentially obscuring the differences between the two groups.
The two previous randomized trials had event rates (death or recurrence) of only 10%, given the relatively large expected differences in outcomes related to the anesthetic regimens (a 5% difference in overall survival [52] and a 30% reduction in cancer recurrence [53] in 1,670 and 2,132 patients, respectively); the statistical power may have been inadequate in these two studies. Additionally, the results of randomized controlled trials conducted on breast cancer may not be related to other cancer types. The potential of anesthetic agents to modify tumor biology, including local recurrence and metastasis, may vary substantially among cancer types [54]. Breast cancer resection procedures tend to be brief and have a commensurately low exposure to anesthetic drugs. Furthermore, the superficial location of these tumors may facilitate convenient surgical management with a low risk of cancer cell dissemination. Therefore, additional clinical trials are needed to evaluate the potential benefits of propofol-based TIVA in patients undergoing major surgery.
Currently, a multicenter study is in progress comparing propofol-based TIVA with volatile anesthetics for various major cancer surgeries (lobectomy or pneumonectomy, esophagectomy, radical cystectomy, pancreatectomy, partial hepatectomy, hyperthermic intraperitoneal chemotherapy, gastrectomy, cholecystectomy, or bile duct resection; n = 1,804), with all-cause mortality as the primary outcome (NCT03034096). The aforementioned CAN trial includes another arm of patients undergoing primary resection of colorectal cancer and is currently in progress [104]. Another upcoming Volatile Anesthesia and Perioperative Outcomes Related to Cancer trial (VAPOR-C trial) is a large randomized trial that began in 2021. This trial is scheduled to be completed by 2025 for patients with lung or colorectal adenocarcinomas. This trial had a 2×2 factorial design comparing propofol and sevoflurane anesthesia with or without intravenous lidocaine (NCT04316013) [105].

OPIOIDS

Laboratory studies have identified three mechanisms through which opioids affect metastasis and tumor growth.
Preclinical studies have shown that opioids have immunosuppressive properties, including impairment of neutrophil chemotaxis and reduction in NK cell cytotoxicity [106,107]. Opioids inhibit lymphocytes and mononuclear phagocytes via opioid or non-opioid receptors, such as toll-like receptor 4 [108]. In rectal cancer resection, the administration of morphine or oxycodone reduced the number of NK cells and T lymphocytes 12 h after injection [109]. Patients with breast cancer who received sevoflurane and opioids had reduced NK cell cytotoxicity [34] and a higher neutrophil-lymphocyte ratio, which is related to an increased risk of recurrence and poor outcome [110], compared to patients who received propofol and regional anesthesia. In pathological samples of resected breast cancer tissue, patients who received propofol/paravertebral anesthesia showed more infiltration of NK cells into their cancer tissue than patients who received general anesthesia with opioid analgesics [71]. However, these clinical studies used combined anesthetic techniques (such as sevoflurane versus opioids versus regional analgesia), making it impossible to determine the effects of opioids alone.
Second, opioids may directly affect cancer cells by activating various transcription factors [111] including VEGF receptors [100,112,113]. However, the most peculiar feature is the overexpression of the mu-opioid receptor (MOR) [108,114,115] on the surface of certain cancer cell lines that are activated by opioids [116-118]. In lung cancer, MOR expression is doubled in patients with metastatic disease compared to that in patients without metastases in tissue biopsy specimens [117]. In prostate cancer, MOR overexpression is associated with decreased overall and progression-free survival [119]. In breast cancer, MOR gene polymorphisms reduced cancer-related mortality over a 10-year follow-up [120]. Consistent with this finding, the MOR antagonist methylnaltrexone has shown beneficial effects in stopping cancer progression; A post hoc analysis of two randomized clinical trials showed that methylnaltrexone, administered for opioid-induced constipation, was associated with improved overall survival in patients with advanced cancer [121]. When NSCLC cell lines were treated with methylnaltrexone, cancer invasion was inhibited [122]. Patients administered mu-opioid receptor antagonists such as naloxone showed improved survival in colorectal and breast cancers [111,113,123].
Third, opioids can interact with inflammatory cytokines (IL1, IL4, IL6, and TNF), which also control gene expression in the MOR [17,124-126].
However, other studies reported contradictory results. Morphine showed antitumor and antimetastatic effects in a mouse model of breast cancer [127-129]. Methadone also showed antitumor effects in glioblastoma [130] and leukemia models [131]. One study showed that morphine suppressed cancer cell proliferation by binding to the opioid growth factor receptor (OGFR) on the surface of lung cancer cell lines [132].
Clinical studies have yielded contradictory results. A retrospective study of about 2800 patients undergoing surgery for renal cell carcinoma found that higher opioid doses during surgery were associated with decreased recurrence-free (HR, 1.06 per 10 MME; 95% CI, 1.03-1.09) and overall survival (HR, 1.05 per 10 MME; 95% CI, 1.02-1.09) [133]. Another retrospective study involving around 900 patients with NSCLC found that higher fentanyl doses during surgery were associated with decreased overall survival in patients with early stage cancer. This result was not applicable for more advanced cancer [134]. However, another study on NSCLC (n = 1,000) found that increased intraoperative opioid use was not associated with increased recurrence or death [135]. Similarly, in patients undergoing colorectal cancer resection (n = 1,680), the amount of intraoperative fentanyl was not associated with recurrence or overall survival [136]. For patients with triple-negative breast cancer (n = 1,100) [137], higher intraoperative opioid doses were associated with improved recurrence free survival (HR, 0.93; 95% CI, 0.88-0.99; per 10 morphine milligram equivalent (MME) increase). A systematic review published in 2018 could not draw conclusions on the relationship between perioperative opioid use and cancer recurrence in colorectal cancer because of inconsistent results and low-quality data [138].
There is no clinical evidence indicating that opioid-sparing techniques affect cancer outcomes. A follow-up analysis of data from a large randomized trial (n = 1,700+) of major abdominal or thoracic surgery found similar overall and recurrence-free survival rates at the 5-year follow-up between patients who received epidural/general anesthesia and those who received only general anesthesia with postoperative opioid analgesia [139]. In a randomized trial (n = 400, video-assisted thoracoscopic surgery for lung cancer), patients who received general anesthesia with postoperative opioid analgesia had similar recurrence-free and overall survival rates to those who received general/epidural anesthesia with postoperative epidural analgesia [140]. Sessler et al. [53] compared regional (paravertebral block with propofol sedation) and general (sevoflurane) anesthesia in patients with breast cancer in a larger multicenter trial (n = 2,132). Regional anesthesia with propofol sedation which used lower opioids, did not reduce breast cancer recurrence when compared with general anesthesia with sevoflurane/opioid at a median follow-up of 3 years [HR, 0.97; 95% CI, 0.74-1·28; P = 0.84]. These study designs make it difficult to isolate the pure effect of opioids on cancer cells owing to the use of combined anesthetic techniques; however, the reduction in opioids did not improve cancer outcomes.
Several retrospective database studies have suggested potentially different effects of opioids on different cancer subtypes [137,141]. Additionally, different opioids may have different effects on the immune system. For example, morphine and fentanyl suppress NK cell activity and lymphocyte proliferation, whereas oxycodone causes minimal immunosuppression [142]. A recent review highlighted the dose-dependent and short-lived nature of the immunosuppressive effects of opioids [143]. Therefore, currently, there is no basis for changing perioperative opioid prescriptions because of concerns about cancer progression.

NITROUS OXIDE

There are insufficient data for nitrous oxide. In a secondary analysis of a trial on colorectal cancer (n = 400), there was no difference in cancer recurrence or death between patients who received nitrous oxide or nitrogen during isoflurane/remifentanil anesthesia and those who did not when followed for 4-8 years after surgery [144].

OTHER INTRAVENOUS AGENTS

Both ketamine and thiopental inhibited NK cell activity in animal models [31,145,146]. In an animal model, ketamine induced tumor metastasis more efficiently than other intravenous agents. Thiopental treatment also increased metastasis, whereas propofol treatment prevented metastasis [31]. Ketamine upregulates the levels of anti-apoptotic proteins in human breast cancer cell lines, which helps the cells invade and proliferate [147].

α2 ADRENOCEPTOR AGONISTS

Clonidine and dexmedetomidine are potent α2-adrenoceptor agonists with analgesic, opioid-sparing, sedative, and anxiolytic effects. Therefore, they are widely used in general anesthesia and intensive care units. Clonidine, an older α2-adrenoceptor agonist, is used as a part of multimodal analgesia in oral form. Compared with clonidine, dexmedetomidine, a more selective α2 adrenoceptor agonist, is administered intravenously and is much more effective with fewer side effects. As a sympatholytic agent, α2-adrenoceptor agonist is theoretically an attractive option for general anesthesia in patients with cancer owing to its analgesic effects and reduction of catecholamines. In addition, some studies have found that dexmedetomidine is anti-inflammatory and reduces serum TNF-α, IL-6, PI3K, and AKT [148-150]. However, dexmedetomidine reduces both pro- and anti-inflammatory cytokines, but a greater reduction in anti-inflammatory cytokines (IL6/IL10 and IL8/IL1) by dexmedetomidine leads to a proinflammatory state [151].
Laboratory studies in murine models (lung, colon, and breast cancer) have shown a dose-dependent increase in tumor cell retention and metastasis following stimulation with α2-adrenergic receptors [152]. Numerous in vivo and in vitro studies have shown that dexmedetomidine increases the risk of recurrence. Dexmedetomidine increased cancer cell survival through activation of HIF-1α, upregulating the expression of survivin, metalloproteinases, and signal transducer and activator of transcription 3 (STAT3). These processes are related to cell migration and metastatic transition [15,18-21,62-64,76,149,153,154]. In addition, dexmedetomidine induces the proliferation of myeloid-derived suppressor cells, increases VEGF production, and promotes tumor metastasis [18].
A retrospective single-center study (n = 250) found that dexmedetomidine was associated with a decrease in overall survival, but not in recurrence-free survival, in NSCLC [64]. Another retrospective study of nearly 650 patients (breast or lung cancer) found that patients who received low-dose clonidine during surgery had similar recurrence-free and overall survival rates to those who did not [155].
There is no high-quality evidence supporting the use of α2-adrenoceptor agonists in patients with cancer. Prospective randomized controlled trials are needed to determine whether the effects observed in in vitro and in vivo studies can be observed in patients. However, given the potential harm caused by dexmedetomidine, avoiding its use when safe alternatives are available may be prudent. Several randomized studies are being conducted, one of which is expected to be completed by 2024 for breast cancer surgery (NCT03109990) [156].

LOCAL ANESTHETICS

Local anesthetics are used for both systemic intravenous infusions and neuraxial and peripheral nerve blocks. Lidocaine is a short acting sodium channel blocker that acts to decrease nerve conduction [125,157-159]. Continuous lidocaine infusion is usually administered during anesthesia as a component of multimodal analgesia and as part of the enhanced recovery after surgery (ERAS) protocol to aid in the recovery of bowel movements and decrease opioid consumption.
In in vitro studies, systemic lidocaine has been shown to protect against cancer recurrence. Lidocaine has anti-inflammatory and antitumor effects through multiple pathways [160-164]. In addition to directly affecting cancer cells through the blockade of voltage-gated sodium channels in tumor cells, lidocaine reduced IL1, IL8, and TNF-α [165,166]. Lidocaine reduced cancer cell viability and migration in laboratory studies and improved the survival of 4T1 syngeneic breast cancer mice [167]. In that study, intravenous lidocaine reduced the number of pulmonary metastases when used in combination with volatile anesthesia, thus reducing the pro-cancer effects of volatile anesthesia in a mouse model [168]. In another mouse study, the addition of lidocaine to the chemotherapy drug cisplatin reduced the number of lung metastases compared with the control or cisplatin alone [86].
There are numerous suggested pathways responsible for the antitumor effects of lidocaine [86,169-174]. However, clinical data are limited. In a retrospective study of approximately 2,000 patients (pancreatic cancer), intravenous lidocaine infusion was associated with a modest increase in 1- and 3-year overall survival (68% vs. 63% for 1-year overall survival and 34% vs. 27% for 3-year overall survival), with no difference in disease-free survival [172]. Another retrospective study on radical cystectomy for bladder cancer (n = 144) found that intraoperative lidocaine administration was associated with a reduction in overall mortality (adjusted HR, 0.36; 95% CI, 0.12-0.83) and cancer recurrence (30% vs. 47%) within 2 years compared to patients who did not receive lidocaine [175].
No studies have shown that lidocaine infusions have anticancer effects [159,160,171,176]. The upcoming VAPOR-C trial will investigate the effectiveness of lidocaine (lung and colorectal adenocarcinoma) in a 2×2 factorial design comparing propofol and sevoflurane general anesthesia with or without intravenous (IV) lidocaine (NCT04316013) [105].
To avoid overdose toxicity, IV lidocaine infusions have dose-limitation [159,160]. Most clinical studies have used a bolus of 1.5 mg/kg IV at induction, followed by 2 mg/kg/h IV infusion [172], or a bolus of 1 mg/kg IV, followed by 1.5 mg/kg/h IV infusion [175].

GLUCOCORTICOIDS

Glucocorticoids are widely used as adjuvant therapy in patients with metastatic cancer to prevent chemotherapy-induced nausea and vomiting and to reduce pain. Dexamethasone is commonly administered during anesthesia to prevent postoperative nausea and vomiting and for analgesia.
Glucocorticoids have anti-inflammatory effects that can reduce cancer recurrence; however, they also have immunomodulatory effects that can impair cancer cell destruction.
The existing evidence is inconsistent. In preclinical studies, dexamethasone was associated with increased proliferation of some solid cancer cell lines but not others [177].
A retrospective study of patients who underwent cytoreductive surgeries for ovarian [178] and endometrial cancer [179] showed that cancer recurrence did not differ between patients who received dexamethasone (4-10 mg IV) during surgery and those who did not [178,179]. In another retrospective study of NSCLC, intraoperative administration of dexamethasone was associated with prolonged postoperative survival [180]. In contrast, a retrospective study of nearly 500 patients with rectal cancer found that the administration of dexamethasone during surgery was associated with reduced disease-free survival [181].
Therefore, there is insufficient evidence to suggest that practices should be changed based on concerns regarding cancer recurrence.

NONSTEROIDAL ANTI-INFLAMMATORY DRUGS

Several retrospective studies have shown that intraoperative administration of non-steroidal anti-inflammatory drugs (NSAIDs) reduces cancer recurrence and improves survival. A single-center retrospective study (n = 720 patients with breast cancer) found that intraoperative administration of ketorolac or diclofenac was associated with prolonged disease-free and overall survival [182]. Another retrospective study (n = 327 patients with breast cancer) found that patients who received ketorolac during surgery had a lower rate of cancer recurrence than in those who received sufentanil, ketamine, or clonidine [183]. Yeh et al. [184] conducted a nationwide cohort study (n = 15,574 patients with hepatocellular carcinoma) that revealed an association between perioperative administration of NSAIDs and a reduced risk of cancer recurrence (HR, 0.81; 95% CI, 0.73-0.90; P < 0.001).
In a small prospective study (n = 38 patients with breast cancer), patients randomly received propranolol and etodolac or placebo for 5 days before and after surgery [185]. Those receiving propranolol and etodolac showed reduced levels of cancer recurrence markers [185].
Regarding anticancer mechanisms, NSAIDs may play a significant role by inhibiting a pro-inflammatory tumor microenvironment, downregulating VEGF, and reducing regulatory T cell infiltration. This leads to a decrease in angiogenesis and lymphangiogenesis, which are critical processes for tumor growth and spread. Additionally, NSAIDs have been shown to reduce NK cell suppression and metastasis in mouse models, further hindering cancer progression. Together, these mechanisms underscore the potential of NSAIDs in preventing cancer recurrence by targeting various aspects of tumor biology and immune response [39].

CONCLUSION

In this review, I briefly discussed cancer cell biology and the interaction between residual tumor cells, growth and migration factors, and the host immune system, in relation to the effects of commonly used anesthetics and adjuvants. The currently available data are insufficient to form a definitive recommendation for the choice of anesthetics despite the numerous studies that have been published.
The apparent protumor or antitumor effect shown in in vitro or in vivo studies was not clear in clinical settings. Therefore, this process appears to be much more complex than we initially believed, probably owing to the heterogeneous biology of different malignancies and surgeries, as well as patient populations.
Several multicenter, randomized controlled clinical trials are currently underway to shed light on this topic. Given the current state of evidence, the clinical impact of anesthetics and adjuvants on cancer recurrence and metastasis should be investigated by conducting high-quality randomized studies.

Notes

FUNDING

None.

CONFLICTS OF INTEREST

Hyun Joo Ahn has been the associate editor of the Anesthesia and Pain Medicine since 2015. However, She was not involved in the peer reviewer selection, evaluation, or decision process of this article. No other potential conflicts of interest relevant to this article were reported.

DATA AVAILABILITY STATEMENT

Data sharing is not applicable to this article, as no datasets were used or analyzed for this work.

REFERENCES

1. Luo J, Shi Y, Wang X, Zhang R, Chen S, Yu W, et al. A 20-year research trend analysis of the influence of anesthesia on tumor prognosis using bibliometric methods. Front Oncol 2021; 11: 683232.
crossref pmid pmc
2. Sekandarzad MW, Van Zundert AAJ, Lirk PB, Doornebal CW, Hollmann MW. Perioperative anesthesia care and tumor progression. Anesth Analg 2017; 124: 1697-708.
crossref pmid
3. Forget P, Aguirre JA, Bencic I, Borgeat A, Cama A, Condron C, et al. How anesthetic, analgesic and other non-surgical techniques during cancer surgery might affect postoperative oncologic outcomes: A summary of current state of evidence. Cancers (Basel) 2019; 11: 592.
crossref pmid pmc
4. Montejano J, Jevtovic-Todorovic V. Anesthesia and cancer, friend or foe? A narrative review. Front Oncol 2021; 11: 803266.
crossref pmid pmc
5. Vona G, Sabile A, Louha M, Sitruk V, Romana S, Schütze K, et al. Isolation by size of epithelial tumor cells: a new method for the immunomorphological and molecular characterization of circulating tumor cells. Am J Pathol 2000; 156: 57-63.
pmid pmc
6. 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 pdf
7. 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 pdf
8. 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
9. Foss OP, Brennhovd IO, Messelt OT, Efskind J, Liverud K. Invasion of tumor cells into the bloodstream caused by palpation or biopsy of the tumor. Surgery 1966; 59: 691-5.
pmid
10. Denis MG, Lipart C, Leborgne J, LeHur PA, Galmiche JP, Denis M, et al. Detection of disseminated tumor cells in peripheral blood of colorectal cancer patients. Int J Cancer 1997; 74: 540-4.
crossref pmid
11. L Holmgren L, O'Reilly MS, Folkman J. Dormancy of micrometastases: balanced proliferation and apoptosis in the presence of angiogenesis suppression. Nat Med 1995; 1: 149-53.
crossref pmid pdf
12. Shakhar G, Ben-Eliyahu S. Potential prophylactic measures against postoperative immunosuppression: could they reduce recurrence rates in oncological patients? Ann Surg Oncol 2003; 10: 972-92.
crossref pmid pdf
13. Smyth MJ, Godfrey DI, Trapani JA. A fresh look at tumor immunosurveillance and immunotherapy. Nat Immunol 2001; 2: 293-9.
crossref pmid pdf
14. Coussens LM, Werb Z. Inflammation and cancer. Nature 2002; 420: 860-7.
crossref pmid pmc pdf
15. Chen P, Luo X, Dai G, Jiang Y, Luo Y, Peng S. Dexmedetomidine promotes the progression of hepatocellular carcinoma through hepatic stellate cell activation. Exp Mol Med 2020; 52: 106274.
crossref pmid pmc pdf
16. Deng F, Ouyang M, Wang X, Yao X, Chen Y, Tao T, et al. Differential role of intravenous anesthetics in colorectal cancer progression: implications for clinical application. Oncotarget 2016; 7: 77087-95.
crossref pmid pmc
17. Nguyen J, Luk K, Vang D, Soto W, Vincent L, Robiner S, et al. Morphine stimulates cancer progression and mast cell activation and impairs survival in transgenic mice with breast cancer. Br J Anaesth 2014; 113 Suppl 1(Suppl 1): i4-13.
crossref pmid
18. Su X, Fan Y, Yang L, Huang J, Qiao F, Fang Y, et al. Dexmedetomidine expands monocytic myeloid-derived suppressor cells and promotes tumour metastasis after lung cancer surgery. J Transl Med 2018; 16: 347.
crossref pmid pmc pdf
19. Szpunar MJ, Burke KA, Dawes RP, Brown EB, Madden KS. The antidepressant desipramine and alpha2-adrenergic receptor activation promote breast tumor progression in association with altered collagen structure. Cancer Prev Res (Phila) 2013; 6: 1262-72.
pmid
20. 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: 4655-64.
crossref pmid pmc
21. 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.
crossref pmid pmc
22. Kim R. Anesthetic technique and cancer recurrence in oncologic surgery: unraveling the puzzle. Cancer Metastasis Rev 2017; 36: 159-77.
crossref pmid pdf
23. Wall T, Sherwin A, Ma D, Buggy DJ. Influence of perioperative anaesthetic and analgesic interventions on oncological outcomes: a narrative review. Br J Anaesth 2019; 123: 135-50.
crossref pmid pmc
24. Bar-Yosef S, Melamed R, Page GG, Shakhar G, Shakhar K, Ben-Eliyahu S. Attenuation of the tumor-promoting effect of surgery by spinal blockade in rats. Anesthesiology 2001; 94: 1066-73.
crossref pmid pdf
25. Sacerdote P, Bianchi M, Gaspani L, Manfredi B, Maucione A, Terno G, et al. The effects of tramadol and morphine on immune responses and pain after surgery in cancer patients. Anesth Analg 2000; 90: 1411-4.
crossref pmid
26. Yeager MP, Colacchio TA, Yu CT, Hildebrandt L, Howell AL, Weiss J, et al. Morphine inhibits spontaneous and cytokine-enhanced natural killer cell cytotoxicity in volunteers. Anesthesiology 1995; 83: 500-8.
crossref pmid pdf
27. Schlagenhauff B, Ellwanger U, Breuninger H, Stroebel W, Rassner G, Garbe C. Prognostic impact of the type of anaesthesia used during the excision of primary cutaneous melanoma. Melanoma Res 2000; 10: 165-9.
crossref pmid
28. Brand JM, Kirchner H, Poppe C, Schmucker P. The effects of general anesthesia on human peripheral immune cell distribution and cytokine production. Clin Immunol Immunopathol 1997; 83: 190-4.
crossref pmid
29. Markovic SN, Knight PR, Murasko DM. Inhibition of interferon stimulation of natural killer cell activity in mice anesthetized with halothane or isoflurane. Anesthesiology 1993; 78: 700-6.
crossref pmid pdf
30. Shapiro J, Jersky J, Katzav S, Feldman M, Segal S. Anesthetic drugs accelerate the progression of postoperative metastases of mouse tumors. J Clin Invest 1981; 68: 678-85.
crossref pmid pmc
31. 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
32. Looney M, Doran P, Buggy DJ. Effect of anesthetic technique on serum vascular endothelial growth factor C and transforming growth factor beta in women undergoing anesthesia and surgery for breast cancer. Anesthesiology 2010; 113: 1118-25.
pmid
33. Tavare AN, Perry NJS, Benzonana LL, Takata M, Ma D. Cancer recurrence after surgery: direct and indirect effects of anesthetic agents. Int J Cancer 2012; 130: 1237-50.
crossref pmid
34. Buckley A, McQuaid S, Johnson P, Buggy DJ. 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: i56-62.
crossref pmid
35. Jaura AI, Flood G, Gallagher HC, Buggy DJ. Differential effects of serum from patients administered distinct anaesthetic techniques on apoptosis in breast cancer cells in vitro: a pilot study. Br J Anaesth 2014; 113 Suppl 1: i63-7.
crossref pmid
36. Cho JS, Lee MH, Kim SI, Park S, Park HS, Oh E. The effects of perioperative anesthesia and analgesia on immune function in patients undergoing breast cancer resection: A prospective randomized study. Int J Med Sci 2017; 14: 970-6.
crossref pmid pmc
37. Iwasaki M, Zhao H, Jaffer T, Unwith S, Benzonana L, Lian Q. Volatile anaesthetics enhance the metastasis related cellular signalling including CXCR2 of ovarian cancer cells. Oncotarget 2016; 7: 26042-56.
crossref pmid pmc
38. Markovic-Bozic J, Karpe B, Potocnik I, Jerin A, Vranic A, Novak-Jankovic V. Effect of propofol and sevoflurane on the inflammatory response of patients undergoing craniotomy. BMC Anesthesiol 2016; 16: 18.
crossref pmid pmc
39. 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 pdf
40. Lai HC, Lee MS, Lin C, Lin KT, Huang YH, Wong CS. 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: 151-60.
crossref pmid pmc
41. 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.
pmid
42. 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
43. 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 pdf
44. 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 pdf
45. 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.
pmid
46. Hasselager RP, Hallas J, Gögenur I. Inhalation or total intravenous anaesthesia and recurrence after colorectal cancer surgery: a propensity score matched Danish registry-based study. Br J Anaesth 2021; 126: 921-30.
crossref pmid
47. Enlund M, Berglund A, Enlund A, Bergkvist L. Volatile versus propofol general anesthesia and long-term survival after breast cancer surgery: A national registry retrospective cohort study. Anesthesiology 2022; 137: 315-26.
crossref pmid pdf
48. Yoon S, Jung SY, Kim MS, Yoon D, Cho Y, Jeon Y. Impact of propofol-based total intravenous anesthesia versus inhalation anesthesia on long-term survival after cancer surgery in a nationwide cohort. Ann Surg 2023; 278: 1024-31.
crossref pmid
49. Makito K, Matsui H, Fushimi K, Yasunaga H. Volatile versus total intravenous anesthesia for cancer prognosis in patients having digestive cancer surgery. Anesthesiology 2020; 133: 764-73.
crossref pmid pdf
50. Chang CY, Wu MY, Chien YJ, Su IM, Wang SC, Kao MC. Anesthesia and long-term oncological outcomes: A systematic review and meta-analysis. Anesth Analg 2021; 132: 623-34.
crossref pmid
51. Hovaguimian F, Braun J, Schläpfer M, Puhan MA, Beck-Schimmer B. Anesthesia and circulating tumor cells: Reply. Anesthesiology 2021; 134: 507-8.
crossref pmid pdf
52. Enlund M, Berglund A, Enlund A, Lundberg J, Wärnberg F, Wang DX, et al. Impact of general anaesthesia on breast cancer survival: a 5-year follow up of a pragmatic, randomised, controlled trial, the CAN-study, comparing propofol and sevoflurane. EClinicalMedicine 2023; 60: 102037.
crossref pmid pmc
53. 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.
pmid
54. Tsuchiya Y, Sawada S, Yoshioka I, Ohashi Y, Matsuo M, Harimaya Y, et al. Increased surgical stress promotes tumor metastasis. Surgery 2003; 133: 547-55.
crossref pmid
55. Ganesh K, Massague J. Targeting metastatic cancer. Nat Med 2021; 27: 34-44.
crossref pmid pmc pdf
56. Suhail Y, Cain MP, Vanaja K, Kurywchak PA, Levchenko A, Kalluri R, et al. Systems biology of cancer metastasis. Cell Syst 2019; 9: 109-27.
crossref pmid pmc
57. Alazawi W, Pirmadjid N, Lahiri R, Bhattacharya S. Inflammatory and immune responses to surgery and their clinical impact. Ann Surg 2016; 264: 73-80.
crossref pmid
58. Angka L, Khan ST, Kilgour MK, Xu R, Kennedy MA, Auer RC. Dysfunctional natural killer cells in the aftermath of cancer surgery. Int J Mol Sci 2017; 18: 1787.
crossref pmid pmc
59. Perez-Sayans M, Pérez-Sayáns M, Somoza-Martín JM, Barros-Angueira F, Diz PG, Rey JMG, García-García A. Beta-adrenergic receptors in cancer: therapeutic implications. Oncol Res 2010; 19: 45-54.
crossref pmid
60. Wu WKK, Sung JJY, Lee CW, Yu J, Cho CH. Cyclooxygenase-2 in tumorigenesis of gastrointestinal cancers: an update on the molecular mechanisms. Cancer Lett 2010; 295: 7-16.
crossref pmid
61. Huang H, Benzonana LL, Zhao H, Watts HR, Perry NJS, Bevan C, et al. Prostate cancer cell malignancy via modulation of HIF-1alpha pathway with isoflurane and propofol alone and in combination. Br J Cancer 2014; 111: 1338-49.
crossref pmid pmc pdf
62. Zhu L, Zhang Y, Zhang Z, Ding X, Gong C, Qian Y. Activation of PI3K/Akt/HIF-1alpha signaling is involved in lung protection of dexmedetomidine in patients undergoing video-assisted thoracoscopic surgery: A pilot study. Drug Des Devel Ther 2020; 14: 5155-66.
pmid pmc
63. Chen HY, Li GH, Tan GC, Liang H, Lai XH, Huang Q, et al. Dexmedetomidine enhances hypoxia-induced cancer cell progression. Exp Ther Med 2019; 18: 4820-8.
crossref pmid pmc
64. Cata JP, Singh V, Lee BM, Villarreal J, Mehran JR, Yu J, et al. Intraoperative use of dexmedetomidine is associated with decreased overall survival after lung cancer surgery. J Anaesthesiol Clin Pharmacol 2017; 33: 317-23.
crossref pmid pmc
65. Darby IA, Hewitson TD. Hypoxia in tissue repair and fibrosis. Cell Tissue Res 2016; 365: 553-62.
crossref pmid pdf
66. Schito L, Semenza GL. Hypoxia-inducible factors: master regulators of cancer progression. Trends Cancer 2016; 2: 758-70.
crossref pmid
67. Karnezis T, Shayan R, Caesar C, Roufail S, Harris NC, Ardipradja K, et al. VEGF-D promotes tumor metastasis by regulating prostaglandins produced by the collecting lymphatic endothelium. Cancer Cell 2012; 21: 181-95.
crossref pmid
68. Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell 2008; 14: 818-29.
crossref pmid
69. Stollings LM, Jia LJ, Tang P, Dou H, Lu B, Xu Y. Immune modulation by volatile anesthetics. Anesthesiology 2016; 125: 399-411.
crossref pmid pmc pdf
70. Benzonana LL, Perry NJS, 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 pdf
71. Desmond F, McCormack J, Mulligan N, Stokes M, Buggy DJ. Effect of anaesthetic technique on immune cell infiltration in breast cancer: a follow-up pilot analysis of a prospective, randomised, investigator-masked study. Anticancer Res 2015; 35: 1311-9.
pmid
72. Ecimovic P, Blaithnaid McHugh B, Murray D, Doran P, Buggy DJ. Effects of sevoflurane on breast cancer cell function in vitro. Anticancer Res 2013; 33: 4255-60.
pmid
73. Inada T, Kubo K, Shingu K. Promotion of interferon-gamma production by natural killer cells via suppression of murine peritoneal macrophage prostaglandin E2 production using intravenous anesthetic propofol. Int Immunopharmacol 2010; 10: 1200-8.
crossref pmid
74. Luo X, Zhao H, Hennah L, Ning J, Liu J, Tu H, et al. Impact of isoflurane on malignant capability of ovarian cancer in vitro. Br J Anaesth 2015; 114: 831-9.
crossref pmid
75. Deng X, Vipani M, Liang G, Gouda D, Wang B, Wei H. Sevoflurane modulates breast cancer cell survival via modulation of intracellular calcium homeostasis. BMC Anesthesiol 2020; 20: 253.
crossref pmid pmc pdf
76. Liu Y, Sun J, Wu T, Lu X, Du Y, Duan H, et al. Effects of serum from breast cancer surgery patients receiving perioperative dexmedetomidine on breast cancer cell malignancy: A prospective randomized controlled trial. Cancer Med 2019; 8: 7603-12.
crossref pmid pmc pdf
77. Ciechanowicz S, Zhao H, Chen Q, Cui J, Mi E, Mi E, et al. Differential effects of sevoflurane on the metastatic potential and chemosensitivity of non-small-cell lung adenocarcinoma and renal cell carcinoma in vitro. Br J Anaesth 2018; 120: 368-75.
crossref pmid
78. Hooijmans CR, Geessink FJ, Ritskes-Hoitinga M, Scheffer GJ. A systematic review of the modifying effect of anaesthetic drugs on metastasis in animal models for cancer. PLoS One 2016; 11: e0156152.
crossref pmid pmc
79. 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
80. Yang C, Gao J, Yan N, Wu B, Ren Y, Li H, et al. Propofol inhibits the growth and survival of gastric cancer cells in vitro through the upregulation of ING3. Oncol Rep 2017; 37: 587-93.
crossref pmid
81. Yang N, Liang Y, Yang P, Ji F. Propofol suppresses LPS-induced nuclear accumulation of HIF-1α and tumor aggressiveness in non-small cell lung cancer. Oncol Rep 2017; 37: 2611-9.
crossref pmid pmc
82. Yu B, Gao W, Zhou H, Miao X, Chang Y, Wang L, et al. Propofol induces apoptosis of breast cancer cells by downregulation of miR-24 signal pathway. Cancer Biomark 2018; 21: 513-9.
crossref pmid
83. Du Q, Liu J, Zhang X, Zhang X, Zhu H, Wei M, et al. Propofol inhibits proliferation, migration, and invasion but promotes apoptosis by regulation of Sox4 in endometrial cancer cells. Braz J Med Biol Res 2018; 51: e6803.
crossref pmid pmc
84. Zhou CL, Li JJ, Ji P. Propofol suppresses esophageal squamous cell carcinoma cell migration and invasion by down-regulation of sex-determining region Y-box 4 (SOX4). Med Sci Monit 2017; 23: 419-27.
crossref pmid pmc
85. Chen J, Ju HL, Yuan XY, Wang TJ, Lai BQ. SOX4 is a potential prognostic factor in human cancers: a systematic review and meta-analysis. Clin Transl Oncol 2016; 18: 65-72.
crossref pmid pdf
86. Freeman J, Crowley PD, Foley AG, Gallagher HC, Iwasaki M, Ma D, et al. Effect of perioperative lidocaine, propofol and steroids on pulmonary metastasis in a murine model of breast cancer surgery. Cancers (Basel) 2019; 11: 613.
crossref pmid pmc
87. Ferrell JK, Cattano D, Brown RE, Patel CB, Karni RJ. The effects of anesthesia on the morphoproteomic expression of head and neck squamous cell carcinoma: a pilot study. Transl Res 2015; 166: 674-82.
crossref pmid
88. Xu YJ, Li SY, Cheng Q, Chen WK, Wang SL, Ren Y, et al. Effects of anaesthesia on proliferation, invasion and apoptosis of LoVo colon cancer cells in vitro. Anaesthesia 2016; 71: 147-54.
crossref pmid
89. Selby LV, Fernandez-Bustamante A, Ejaz A, Gleisner A, Pawlik TM, Douin DJ. Association between anesthesia delivered during tumor resection and cancer survival: a systematic review of a mixed picture with constant themes. J Gastrointest Surg 2021; 25: 2129-41.
crossref pmid pdf
90. Enlund M, Berglund A, Andreasson K, Cicek C, Enlund A, Bergkvist J. 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
91. Oh TK, Kim K, Jheon S, Lee J, Do SH, Hwang JW. 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
92. Oh TK, Kim HH, Jeon YT. Retrospective analysis of 1-year mortality after gastric cancer surgery: Total intravenous anesthesia versus volatile anesthesia. Acta Anaesthesiol Scand 2019; 63: 1169-77.
crossref pmid pdf
93. Enlund M, Berglund A, Ahlstrand R, Walldén J, Lundberg J, Wärnberg F, et al. Survival after primary breast cancer surgery following propofol or sevoflurane general anesthesia-A retrospective, multicenter, database analysis of 6305 Swedish patients. Acta Anaesthesiol Scand 2020; 64: 1048-54.
crossref pmid pdf
94. 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 pdf
95. Yap A, Lopez-Olivo MA, Dubowitz J, Hiller J, Riedel B; Global Onco-Anesthesia Research Collaboration Group. Anesthetic technique and cancer outcomes: a meta-analysis of total intravenous versus volatile anesthesia. Can J Anaesth 2019; 66: 546-61.
pmid
96. 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
97. Miao L, Lv X, Huang C, Li P, Sun Y, Jiang H. Long-term oncological outcomes after oral cancer surgery using propofol-based total intravenous anesthesia versus sevoflurane-based inhalation anesthesia: A retrospective cohort study. PLoS One 2022; 17: e0268473.
crossref pmid pmc
98. Liu J, Yang L, Guo X, Jin G, Wang Q, Lv D, et al. Sevoflurane suppresses proliferation by upregulating microRNA-203 in breast cancer cells. Mol Med Rep 2018; 18: 455-60.
crossref pmid
99. Wang L, Wang T, Gu JQ, Su HB. Volatile anesthetic sevoflurane suppresses lung cancer cells and miRNA interference in lung cancer cells. Onco Targets Ther 2018; 11: 5689-93.
crossref pmid pmc pdf
100. Yan T, Zhang GH, Wang BN, Sun L, Zheng H. Effects of propofol/remifentanil-based total intravenous anesthesia versus sevoflurane-based inhalational anesthesia on the release of VEGF-C and TGF-beta and prognosis after breast cancer surgery: a prospective, randomized and controlled study. BMC Anesthesiol 2018; 18: 131.
pmid pmc
101. Oh CS, Lee J, Yoon TG, Seo EH, Park HJ, Piao L, et al. Effect of equipotent doses of propofol versus sevoflurane anesthesia on regulatory T cells after breast cancer surgery. Anesthesiology 2018; 129: 921-31.
crossref pmid pdf
102. Oh CS, Park HJ, Piao L, Sohn KM, Koh SE, Hwang DY, et al. Expression profiles of immune cells after propofol or sevoflurane anesthesia for colorectal cancer surgery: A prospective double-blind randomized trial. Anesthesiology 2022; 136: 448-58.
crossref pmid pdf
103. O'Bryan LJ, Atkins KJ, Lipszyc A, Scott DA, Silbert BS, Evered LA. Inflammatory biomarker levels after propofol or sevoflurane anesthesia: A meta-analysis. Anesth Analg 2022; 134: 69-81.
crossref pmid
104. Enlund M, Enlund A, Berglund A, Bergkvist L. Rationale and design of the CAN study: an RCT of survival after propofol- or sevoflurane-based anesthesia for cancer surgery. Curr Pharm Des 2019; 25: 3028-33.
crossref pmid
105. Dubowitz JA, Cata JP, De Silva AP, Braat S, Shan D, Yee K, et al. Volatile anaesthesia and peri-operative outcomes related to cancer: a feasibility and pilot study for a large randomised control trial. Anaesthesia 2021; 76: 1198-206.
crossref pmid pdf
106. Beilin B, Martin FC, Shavit Y, Gale RP, Liebeskind JC. Suppression of natural killer cell activity by high-dose narcotic anesthesia in rats. Brain Behav Immun 1989; 3: 129-37.
crossref pmid
107. Shavit Y, Terman GW, Lewis JW, Zane CJ, Gale RP, Liebeskind JC. Effects of footshock stress and morphine on natural killer lymphocytes in rats: studies of tolerance and cross-tolerance. Brain Res 1986; 372: 382-5.
crossref pmid
108. Boland JW, Pockley AG. Influence of opioids on immune function in patients with cancer pain: from bench to bedside. Br J Pharmacol 2018; 175: 2726-36.
crossref pmid pmc pdf
109. Cui JH, Jiang WW, Liao YJ, Wang QH, Min Xu, Li Y. Effects of oxycodone on immune function in patients undergoing radical resection of rectal cancer under general anesthesia. Medicine (Baltimore) 2017; 96: e7519.
crossref pmid pmc
110. Ni Eochagain A, Burns D, Riedel B, Sessler DI, Buggy DJ. The effect of anaesthetic technique during primary breast cancer surgery on neutrophil-lymphocyte ratio, platelet-lymphocyte ratio and return to intended oncological therapy. Anaesthesia 2018; 73: 603-11.
crossref pmid pdf
111. Wu Q, Chen X, Wang J, Sun P, Weng M, Chen W, et al. Nalmefene attenuates malignant potential in colorectal cancer cell via inhibition of opioid receptor. Acta Biochim Biophys Sin (Shanghai) 2018; 50: 156-63.
crossref pmid pdf
112. Sen Y, Xiyang H, Yu H. Effect of thoracic paraspinal block-propofol intravenous general anesthesia on VEGF and TGF-beta in patients receiving radical resection of lung cancer. Medicine (Baltimore) 2019; 98: e18088.
pmid pmc
113. 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.
crossref pmid
114. Saurer TB, Ijames SG, Carrigan KA, Lysle DT. Neuroimmune mechanisms of opioid-mediated conditioned immunomodulation. Brain Behav Immun 2008; 22: 89-97.
crossref pmid pmc
115. Kraus J. Regulation of mu-opioid receptors by cytokines. Front Biosci (Schol Ed) 2009; 1: 164-70.
crossref pmid
116. Wigmore T, Farquhar-Smith P. Opioids and cancer: friend or foe? Curr Opin Support Palliat Care 2016; 10: 109-18.
pmid
117. Singleton PA, Mirzapoiazova T, Hasina R, Salgia R, Moss J. Increased mu-opioid receptor expression in metastatic lung cancer. Br J Anaesth 2014; 113 Suppl 1(Suppl 1): i103-8.
pmid
118. Zhang H, Sun M, Zhou D, Gorur A, Sun Z, Zeng W, et al. Increased mu-opioid receptor expression is associated with reduced disease-free and overall survival in laryngeal squamous cell carcinoma. Br J Anaesth 2020; 125: 722-9.
crossref pmid
119. 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 pmc
120. Bortsov AV, Millikan RC, Belfer I, Boortz-Marx RL, Arora H, McLean SA. μ-Opioid receptor gene A118G polymorphism predicts survival in patients with breast cancer. Anesthesiology 2012; 116: 896-902.
crossref pmid pmc pdf
121. 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
122. Mathew B, Lennon FE, Siegler J, Mirzapoiazova T, Mambetsariev N, Sammani S, et al. The novel role of the mu opioid receptor in lung cancer progression: a laboratory investigation. Anesth Analg 2011; 112: 558-67.
pmid
123. Bimonte S, Barbieri A, Cascella M, Rea D, Palma G, Vecchio VD, et al. The effects of naloxone on human breast cancer progression: in vitro and in vivo studies on MDA.MB231 cells. Onco Targets Ther 2018; 11: 185-91.
crossref pmid pmc pdf
124. Connolly C, Buggy DJ. Opioids and tumour metastasis: does the choice of the anesthetic-analgesic technique influence outcome after cancer surgery? Curr Opin Anaesthesiol 2016; 29: 468-74.
pmid
125. Bugada D, Lorini LF, Lavand'homme P. Opioid free anesthesia: evidence for short and long-term outcome. Minerva Anestesiol 2021; 87: 230-7.
crossref pmid
126. Chen J, Luo F, Lei M, Chen Z. A study on cellular immune function of patients treated with radical resection of pulmonary carcinoma with two different methods of anesthesia and analgesia. J BUON 2017; 22: 1416-21.
pmid
127. 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
128. Doornebal CW, Vrijland K, Hau CS, Coffelt SB, Ciampricotti M, Jonkers J, et al. Morphine does not facilitate breast cancer progression in two preclinical mouse models for human invasive lobular and HER2+ breast cancer. Pain 2015; 156: 1424-32.
crossref pmid
129. Afsharimani B, Baran J, Watanabe S, Lindner D, Cabot PJ, Parat MO. Morphine and breast tumor metastasis: the role of matrix-degrading enzymes. Clin Exp Metastasis 2014; 31: 149-58.
crossref pmid pdf
130. Friesen C, Hormann I, Roscher M, Fichtner I, Alt A, Hilger R, et al. Opioid receptor activation triggering downregulation of cAMP improves effectiveness of anti-cancer drugs in treatment of glioblastoma. Cell Cycle 2014; 13: 1560-70.
crossref pmid pmc
131. Friesen C, Roscher M, Hormann I, Fichtner I, Alt A, Hilger RA, et al. Cell death sensitization of leukemia cells by opioid receptor activation. Oncotarget 2013; 4: 677-90.
crossref pmid pmc
132. Kim JY, Ahn HJ, Jin Kim K, Kim J, Lee SH, Chae HB. 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: 1429-36.
pmid
133. Silagy AW, Hannum ML, Mano R, Attalla K, Scarpa JR, DiNatale RG, et al. Impact of intraoperative opioid and adjunct analgesic use on renal cell carcinoma recurrence: role for onco-anaesthesia. Br J Anaesth 2020; 125: e402-4.
crossref pmid pmc
134. Cata JP, Keerty V, Keerty D, Feng L, Norman PH, Gottumukkala V, et al. A retrospective analysis of the effect of intraoperative opioid dose on cancer recurrence after non-small cell lung cancer resection. Cancer Med 2014; 3: 900-8.
crossref pmid pmc pdf
135. Oh TK, Jeon JH, Lee JM, Kim MS, Kim JH, Cho H, et al. Investigation of opioid use and long-term oncologic outcomes for non-small cell lung cancer patients treated with surgery. PLoS One 2017; 12: e0181672.
crossref pmid pmc
136. 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 pdf
137. Montagna G, Gupta HV, Hannum M, Tan KS, Lee J, Scarpa JR, et al. Intraoperative opioids are associated with improved recurrence-free survival in triple-negative breast cancer. Br J Anaesth 2021; 126: 367-76.
crossref pmid pmc
138. Diaz-Cambronero O, Mazzinari G, Cata JP. Perioperative opioids and colorectal cancer recurrence: a systematic review of the literature. Pain Manag 2018; 8: 353-61.
crossref pmid
139. Du YT, Li YW, Zhao BJ, Guo XY, Feng Y, Zuo MZ, et al. Long-term survival after combined epidural-general anesthesia or general anesthesia alone: follow-up of a randomized trial. Anesthesiology 2021; 135: 233-45.
crossref pmid pdf
140. Xu ZZ, Li HJ, Li MH, Huang SM, Li X, Liu QH, et al. Epidural anesthesia-analgesia and recurrence-free survival after lung cancer surgery: a randomized trial. Anesthesiology 2021; 135: 419-32.
crossref pmid pdf
141. Yuval JB, Lee J, Wu F, Thompson HM, Verheij FS, Gupta HV, et al. Intraoperative opioids are associated with decreased recurrence rates in colon adenocarcinoma: a retrospective observational cohort study. Br J Anaesth 2022; 129: 172-81.
crossref pmid pmc
142. Franchi S, Moschetti G, Amodeo G, Sacerdote P. Do all opioid drugs share the same immunomodulatory properties? A review from animal and human studies. Front Immunol 2019; 10: 2914.
crossref pmid pmc
143. Cata JP, Sessler DI. Lost in translation: Failure of preclinical studies to accurately predict the effect of regional analgesia on cancer recurrence. Anesthesiology 2024; 140: 361-74.
crossref pmid pdf
144. Fleischmann E, Marschalek C, Schlemitz K, Dalton JE, Gruenberger T, Herbst F, et al. Nitrous oxide may not increase the risk of cancer recurrence after colorectal surgery: a follow-up of a randomized controlled trial. BMC Anesthesiol 2009; 9: 1.
crossref pmid pmc pdf
145. Forget P, Collet V, Lavand'homme P, Kock MD. 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
146. Nishina K, Akamatsu H, Mikawa K, Shiga M, Maekawa N, Obara H, et al. The inhibitory effects of thiopental, midazolam, and ketamine on human neutrophil functions. Anesth Analg 1998; 86: 159-65.
crossref pmid
147. 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
148. Zhang J, Liu G, Zhang F, Fang H, Zhang D, Liu S, et al. Analysis of postoperative cognitive dysfunction and influencing factors of dexmedetomidine anesthesia in elderly patients with colorectal cancer. Oncol Lett 2019; 18: 3058-64.
crossref pmid pmc
149. Liu M, Yi Y, Zhao M. Effect of dexmedetomidine anesthesia on perioperative levels of TNF-alpha and IL-6 in patients with ovarian cancer. Oncol Lett 2019; 17: 5517-22.
pmid pmc
150. Huyan T, Hu X, Peng H, Zhu Z, Li Q, Zhang W. Perioperative dexmedetomidine reduces delirium in elderly patients after lung cancer surgery. Psychiatr Danub 2019; 31: 95-101.
crossref pmid
151. Kim JA, Ahn HJ, Yang M, Lee SH, Jeong H, Seong BG. Intraoperative use of dexmedetomidine for the prevention of emergence agitation and postoperative delirium in thoracic surgery: a randomized-controlled trial. Can J Anaesth 2019; 66: 371-9.
crossref pmid pdf
152. Lavon H, Matzner P, Benbenishty A, Sorski L, Rossene E, Haldar R, et al. Dexmedetomidine promotes metastasis in rodent models of breast, lung, and colon cancers. Br J Anaesth 2018; 120: 188-96.
crossref pmid pmc
153. Yi XL, Wang JT, Chu CQ, Li YX, Yin JH, Liu SL. Cardiocerebral protective effects of dexmedetomidine as anesthetic in colorectal cancer surgery. Eur Rev Med Pharmacol Sci 2018; 22: 3570-6.
pmid
154. Xia M, Ji NN, Duan ML, Tong JH, Xu JG, Zhang YM, et al. Dexmedetomidine regulate the malignancy of breast cancer cells by activating alpha2-adrenoceptor/ERK signaling pathway. Eur Rev Med Pharmacol Sci 2016; 20: 3500-6.
pmid
155. Forget P, Berlière M, Poncelet A, Kock MD. Effect of clonidine on oncological outcomes after breast and lung cancer surgery. Br J Anaesth 2018; 121: 103-4.
crossref pmid
156. Wall T, Sherwin A, Ma D, Buggy DJ. Influence of perioperative anaesthetic and analgesic interventions on oncological outcomes: a narrative review. Br J Anaesth 2019; 123: 135-50.
crossref pmid pmc
157. Weibel S, Jelting Y, Pace NL, Helf A, Eberhart LH, Hahnenkamp K, et al. Continuous intravenous perioperative lidocaine infusion for postoperative pain and recovery in adults. Cochrane Database Syst Rev 2018; 6: CD009642.
crossref pmid
158. Masic D, Liang E, Long C, Sterk EJ, Barbas B, Rech MA. Intravenous lidocaine for acute pain: a systematic review. Pharmacotherapy 2018; 38: 1250-9.
crossref pmid pdf
159. Khan JS, Hodgson N, Choi S, Reid S, Paul JE, Hong NJL, et al. Perioperative pregabalin and intraoperative lidocaine infusion to reduce persistent neuropathic pain after breast cancer surgery: A multicenter, factorial, randomized, controlled pilot trial. J Pain 2019; 20: 980-93.
crossref pmid
160. Lee JT, Sanderson CR, Xuan W, Agar M. Lidocaine for cancer pain in adults: a systematic review and meta-analysis. J Palliat Med 2019; 22: 326-34.
crossref pmid
161. Hermanns H, Hollmann MW, Stevens MF, Lirk P, Brandenburger T, Piegeler T, et al. Molecular mechanisms of action of systemic lidocaine in acute and chronic pain: a narrative review. Br J Anaesth 2019; 123: 335-49.
crossref pmid
162. Galoș EV, Tat TF, Popa R, Efrimescu CI, Finnerty D, Buggy DJ, et al. Neutrophil extracellular trapping and angiogenesis biomarkers after intravenous or inhalation anaesthesia with or without intravenous lidocaine for breast cancer surgery: a prospective, randomised trial. Br J Anaesth 2020; 125: 712-21.
crossref pmid
163. Van Haren F, Van den Heuvel S, Radema S, Van Erp N, Van den Bersselaar L, Vissers K, et al. Intravenous lidocaine affects oxaliplatin pharmacokinetics in simultaneous infusion. J Oncol Pharm Pract 2020; 26: 1850-6.
crossref pmid pdf
164. Chamaraux-Tran TN, Piegeler T. The amide local anesthetic lidocaine in cancer surgery-potential antimetastatic effects and preservation of immune cell function? A narrative review. Front Med (Lausanne) 2017; 4: 235.
crossref pmid pmc
165. Soto G, Gonzalez MN, Calero F. Intravenous lidocaine infusion. Rev Esp Anestesiol Reanim (Engl Ed) 2018; 65: 269-74.
crossref pmid
166. Piegeler T, Schläpfer M, Dull RO, Schwartz DE, Borgeat A, Minshall RD, et al. Clinically relevant concentrations of lidocaine and ropivacaine inhibit TNFalpha-induced invasion of lung adenocarcinoma cells in vitro by blocking the activation of Akt and focal adhesion kinase. Br J Anaesth 2015; 115: 784-91.
pmid pmc
167. Chamaraux-Tran TN, Mathelin C, Aprahamian M, Joshi GP, Tomasetto C, Diemunsch P, et al. Antitumor effects of lidocaine on human breast cancer cells: an in vitro and in vivo experimental trial. Anticancer Res 2018; 38: 95-105.
pmid
168. 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
169. Wall TP, Crowley PD, Buggy DJ. The effect of lidocaine and bosutinib on 4T1 murine breast cancer cell behaviour in vitro. Anticancer Res 2021; 41: 2835-40.
crossref pmid
170. Wall TP, Crowley PD, Sherwin A, Foley AG, Buggy DJ. Effects of lidocaine and Src inhibition on metastasis in a murine model of breast cancer surgery. Cancers (Basel) 2019; 11: 1414.
crossref pmid pmc
171. Chong PH, Yeo ZZ. Parenteral lidocaine for complex cancer pain in the home or inpatient hospice setting: a review and synthesis of the evidence. J Palliat Med 2021; 24: 1154-60.
crossref pmid pmc
172. Zhang H, Yang L, Zhu X, Zhu M, Sun Z, Cata JP, et al. Association between intraoperative intravenous lidocaine infusion and survival in patients undergoing pancreatectomy for pancreatic cancer: a retrospective study. Br J Anaesth 2020; 125: 141-8.
crossref pmid
173. Ji W, Zhang X, Sun G, Wang X, Liu J, Bian J, et al. Effect of perioperative intravenous lidocaine on postoperative outcomes in patients undergoing resection of colorectal cancer: a protocol for systematic review and meta-analysis. BMJ Open 2021; 11: e048803.
crossref pmid pmc
174. Hou YH, Shi WC, Cai S, Liu H, Zheng Z, Qi FW, et al. Effect of intravenous lidocaine on serum interleukin-17 after video-assisted thoracic surgery for non-small-cell lung cancer: a randomized, double-blind, placebo-controlled trial. Drug Des Devel Ther 2021; 15: 3379-90.
crossref pmid pmc pdf
175. Cazenave L, Faucher M, Tourret M, Marques M, Tezier M, Mokart D. Intravenous lidocaine and cancer outcomes after radical cystectomy. Eur J Anaesthesiol 2022; 39: 396-9.
crossref pmid
176. Wall TP, Buggy DJ. Perioperative intravenous lidocaine and metastatic cancer recurrence - a narrative review. Front Oncol 2021; 11: 688896.
crossref pmid pmc
177. Gündisch s, Boeckeler E, Behrends U, Amtmann E, Ehrhardt H, Jeremias I. Glucocorticoids augment survival and proliferation of tumor cells. Anticancer Res 2012; 32: 4251-61.
pmid
178. De Oliveira GS Jr, McCarthy R, Turan A, Schink JC, Fitzgerald PC, Sessler DI. Is dexamethasone associated with recurrence of ovarian cancer? Anticancer Res 2014; 118: 1213-8.
crossref
179. Merk BA, Havrilesky LJ, Ehrisman JA, Broadwater G, Habib AS. Impact of postoperative nausea and vomiting prophylaxis with dexamethasone on the risk of recurrence of endometrial cancer. Curr Med Res Opin 2016; 32: 453-8.
crossref pmid
180. Huang WW, Zhu WZ, Mu DL, Ji XQ, Nie XL, Li XY, et al. Perioperative management may improve long-term survival in patients after lung cancer surgery: a retrospective cohort study. Anesth Analg 2018; 126: 1666-74.
crossref pmid pmc
181. Yu HC, Luo YX, Peng H, Kang L, Huang MJ, Wang JP. Avoiding perioperative dexamethasone may improve the outcome of patients with rectal cancer. Eur J Surg Oncol 2015; 41: 667-73.
crossref pmid
182. Forget P, Bentin C, Machiels JP, Berliere M, Coulie PG, Kock MD. Intraoperative use of ketorolac or diclofenac is associated with improved disease-free survival and overall survival in conservative breast cancer surgery. Br J Anaesth 2014; 113 Suppl 1: i82-7.
crossref pmid
183. Forget P, Vandenhende J, Berliere M, Machiels JP, Nussbaum B, Legrand C, et al. Do intraoperative analgesics influence breast cancer recurrence after mastectomy? A retrospective analysis. Anesth Analg 2010; 110: 1630-5.
crossref pmid
184. Yeh CC, Lin JT, Jeng LB, Ho HJ, Yang HR, Wu MS, et al. Nonsteroidal anti-inflammatory drugs are associated with reduced risk of early hepatocellular carcinoma recurrence after curative liver resection: a nationwide cohort study. Ann Surg 2015; 261: 521-6.
pmid
185. Shaashua L, Shabat-Simon M, Haldar R, Matzner P, Zmora O, Shabtai M, et al. Perioperative COX-2 and β-adrenergic blockade improves metastatic biomarkers in breast cancer patients in a phase-II randomized trial. Clin Cancer Res 2017; 23: 4651-61.
crossref pmid pmc pdf


ABOUT
ARTICLE & TOPICS
Article category

Browse all articles >

Topics

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: apm@anesthesia.or.kr                

Copyright © 2024 by Korean Society of Anesthesiologists.

Developed in M2PI

Close layer
prev next