Next Article in Journal
Oral Papillomatosis: Its Relation with Human Papilloma Virus Infection and Local Immunity—An Update
Next Article in Special Issue
General Anesthetics in Cancer Surgery: Can Anesthesiologists Help the Patient with More than a Safe Sleep
Previous Article in Journal
Does Multisystem Inflammatory Syndrome Only Mimic Acute Appendicitis in Children or Can It Coexist: When Should We Suspect MIS-C?
Previous Article in Special Issue
Impact of Local Anesthetics on Cancer Behavior and Outcome during the Perioperative Period: A Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Anesthetics and Long Term Cancer Outcomes: May Epigenetics Be the Key for Pancreatic Cancer?

1
ESAIC Mentorship Program, BE-1000 Brussels, Belgium
2
The European Platform for Research Outcomes after PerIoperative Interventions in Surgery for Cancer Research Group (Euro-Periscope): The Onco-Anaesthesiology Research Group (RG), BE-1000 Brussels, Belgium
3
Department of Pharmacy, G. d’Annunzio University of Chieti-Pescara, 66100 Chieti, Italy
4
Epidemiology Group, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
5
Department of Anaesthesia, National Health Service (NHS) Grampian, Aberdeen AB25 2ZD, UK
*
Author to whom correspondence should be addressed.
Medicina 2022, 58(8), 1102; https://doi.org/10.3390/medicina58081102
Submission received: 30 May 2022 / Revised: 3 August 2022 / Accepted: 6 August 2022 / Published: 14 August 2022
(This article belongs to the Special Issue Impact of Anesthetics on Cancer Behavior and Outcome)

Abstract

:
Knowledge shows a divergence of results between preclinical and clinical studies regarding anesthesia and postoperative progression of cancer. While laboratory and animal data from then 2000s onwards raised much enthusiasm in this field of research leading to several clinical investigations worldwide, data from randomized trials seem to have killed off hope for many scientists. However several aspects of the actual knowledge should be reevaluated and there is space for new strategies of investigation. In this paper, we perform a critical review of actual knowledge and propose new research strategies with a special focus on anesthetic management and repurposed anesthetic adjuvants for pancreatic cancer.

1. Introduction

Despite considerable efforts, pancreatic cancer (PC) is one of the deadliest cancers and its incidence and mortality is increasing. Exocrine pancreatic ductal adenocarcinomas (PDAC) represent 90–95% of PCs, whereas the remaining 5% arise from the endocrine pancreas. The 5-year survival rate for all stages is 7–11% and only 42% in people with local disease (13%). Surgery remains the best treatment option but resectable cancers represent less than 25% of all cases and only multidisciplinary approaches have shown to be able to improve cure. The development of new drugs is a long, expensive and very challenging process. For these reasons, there is a renewed interest in research towards perioperative and repurposed interventions that may improve mortality after PC. The main strengths of repurposed drugs is the availability of a well-documented clinical use and pharmacological and safety data, the possibility to start from phase II clinical trials to assess their efficacy for the new indication, and the reduced cost [1].
Starting from late the 1970s, experimental and clinical data have suggested that standard anesthetic techniques for cancer surgery, general or regional anesthesia, act differently in cancer biology and may affect its progression-related outcomes [2,3].
During the perioperative period, anesthetics can act either on cancer cells or through modifications in host response. Modifications in host response occur both systemically and locally on the surgical site [2,3]. Regional anesthesia prevents the stress and immune response that follows surgery whereas general anesthesia does not, thus favoring tumor growth in basic research studies. Alterations in cell immunity have been observed up to a week after surgery and are believed to promote local residual micro-metastases as well as a favorable local microenvironment and systemic situation for the invasion of circulating tumor cells [4,5].
However, the situation is extremely complex. This is evident in true life, where basic research findings led to the conduction of several clinical trials worldwide investigating the repurposed action of anesthetics among other drugs [1]. These trials have been mostly negative and this finding is confirmed by meta-analyses [6,7,8,9], but again, to date, there is no clear response as to whether it is the extent of surgery rather than anesthesia management that most influences cancer outcomes. Furthermore, there is some concern, since some negative trial results can be due to high surgical curability that may mask the effects of anesthesia. Factors such as tumor characteristics, extent of surgical damage and the related stress response and surgical curability contribute to determine the long term desired clinical results and should be better taken into account in the design of clinical trials.
Several aspects of the puzzle have not yet been adequately explored and further and new research strategies are needed to explore the basic mechanisms by which anesthesia acts on cancer. The question that remains is which biological mechanism and anesthetic management, lasting a few hours or days, could influence the natural history of cancer changing outcomes for months or years after surgery? In this review, the general aspects of anesthetic role in epigenetics are described with a special focus on pancreatic cancer.

2. Epigenetics

The biological response of cells under the influence of physiological and external factors is driven by epigenetic modulation of gene and protein expression. To make an analogy, genes are the constitution and epigenetic changes are all genetic mechanisms other than DNA sequence changes. Epigenetics are laws, regulatory acts and amendments, or the heritable cellular system that interprets the genome [10,11,12,13,14].
In normal cells, the transcriptional status of most genes, the coding part of the genome, is epigenetically fixed through the binding of DNA with histones. However, other genes reside in a balanced state sensitive to exogenous signals and capable of rapid modulation of DNA transcription to mRNA. These events lead to alterations in gene expression, a consequent differential production of messenger RNAs (mRNAs) and a subsequent modified expression of proteins that are the final biologic effectors. Several short- or long-time-operating factors including external chemical or pharmacological stimuli can alter this balance [15,16]. One of the most significant examples of the effect of epigenetics is synaptic plasticity.
Anesthetics are chemical factors with the potential to induce epigenetic effects. With regard to the nervous system, for example, this is supported by the fact that anesthetics are neurotoxins sharing many molecular mechanisms of action with alcohol and cocaine [17,18]. The effect on epigenetics occurs even after relatively short exposures, but is more intense after prolonged or repeated exposures [19]. Most importantly, the neurocognitive effects associated with epigenetic changes after anesthetic exposure may persist in neonates or be present for weeks to years in the elderly [18]. Moreover, anesthetic exposure may affect not only the exposed subjects, but also future generations [18].
However epigenetics is a relatively new branch of genetics which only began to be investigated 50 years ago. Research on epigenetics and anesthesia is even younger and our understanding of the biological effects of anesthetics and the underlying molecular mechanisms is uncomplete [18].

2.1. Epigenetics and Anesthetics

Research on the epigenetic effects of anesthetics has been mainly focused on the neurodevelopmental brain alterations and ischemia reperfusion injury effects. The first microarray investigation on inhalational anesthetics showed that expression of 1.5% of 10,000 genes in various organs was altered [20]. Sevoflurane has the ability to change the expression of the circadian genes and drug metabolizing enzymes [21,22]. Propofol and sevoflurane show a different protein expression change activity in rat brains [23]. The effects are more pronounced at young ages, when the central nervous system and other tissues are highly susceptible to what is called epigenetic reprogramming [24,25,26,27]. Anesthetics are also capable of inducing prolonged and intergenerational epigenetic effects. One week after bariatric surgery under general anesthesia, DNA methylation of 1509 genes in male spermatozoa remains altered and 1004 of those genes remained altered after 1 year [28]. Reduced DNA methylation in the 5-upstream promoter region of rat mothers exposed to 6 h sevoflurane and upregulation of Arc and JunB mRNA expression, two genes regulating synaptic plasticity and neuronal development, are trans-generationally expressed in offspring male born [29].

2.2. Post-Translational Chromatin Regulation

Post-translational epigenetic modifications on histones and chromatin are regulated by more than 700 enzymes categorized as writers/erasers, readers, movers shapers and insulators. Lysine-rich N-terminal histone “tails” undergo several processes such as acetylation and methylation, ubiquitination, phosphorylation, and sumoylation [30].
Histone acetylation by acetyltransferases allows chromatin liberation and gene transcription, while histone deacetylation by deacetylases results in a stronger histone bond with DNA and inhibition of gene transcription [31]. Repeated sevoflurane exposure of neonatal rats leads to an increased hippocampal deacetylase activity and reduced histone acetylation, with developmental effects that are reversed by histone deacetylase inhibitors [32].
Methylation by DNA methyltransferases (DNMTs) can both induce and repress gene transcription, depending on the chromatin residue modified. Sevoflurane neurotoxicity on rat stem cells is exerted through a concentration-dependent DNA methylation [17]. There is also an increasingly growing evidence of the epigenetic effects of anesthetics on the developing human brain that include both methylation and acetylation [33].
DNA methylation is one of the mechanisms affected by stress and involved in postoperative hyperalgesia [34]. DNA methylation and DNMT expression in skin after incision is changed and controls nociceptive sensitization. DNMT inhibition attenuates incision-induced nociceptive hypersensitivity via up-regulation of Oprm1 gene expression while treatment with naloxone exacerbates incision-induced mechanical hypersensitivity [34].
DNA methylation in blood mononuclear cells after general anesthesia for major breast surgery is globally reduced by 26% while DNMT mRNAs expression is reduced by 65 to 71% [35]. Opiates are thought to increase methylation whereas lidocaine show controversial effects [36,37,38]. These findings support the evidence that anesthesia/surgery may alter the epigenetic status of host’s tissues after surgery.
Aberrant DNA methylation leads to onco-suppressor gene silencing or oncogene activation and has been linked to oncogenesis in a number of tumor types. Cancers present hypermethylation of CpG islands (DNA regions with a high density of cytosine–guanine dinucleotides) in or near promoter regions, whereas gene bodies are hypomethylated.

2.3. RNA

Only 1.5% of mammalian DNA encodes for approximately 20,000 genes which will be translated into proteins. Protein synthesis is driven by three main types of RNA: messenger RNA (mRNA), transfer RNA (tRNA) that accounts for the majority of RNA molecules, and ribosomal RNA (rRNA). mRNA is transcribed directly from DNA and contains the “message” that is translated by tRNAs into proteins. rRNA accounts for 90% of total RNA mass and forms ribosomes, which are the protein synthesizers [39]. The rest of the human genome, more than 90%, represents non-coding RNAs (ncRNA). The total number of RNA molecules is estimated at 107 per cell [40].
Non-coding RNAs are classified into long [>200 nucleotides] and short ncRNAs (<200 nucleotides) (lncRNA and sncRNA respectively). A series of biological functions such as regulation of the expression of coding genes, cancero-genesis and regulation of biological processes have been described for many ncRNAs, supporting the hypothesis that they are functional, although for most of them a biological role has not yet been discovered [41,42].
Micro RNAs (miRNA) are non-coding, endogenous and highly conserved sncRNAs across species. They act via post-transcriptional degradation or translational repression by binding to 3′ untranslated regions (UTR) of target mRNAs. Each molecule of mRNA can be regulated by more the one miRNA, and a single miRNA may influence the expression of a wide range of mRNAs [43]. Some miRNAs are expressed ubiquitously, whereas others are tissue-specific and/or stage-specific. miRNAs regulate the activity of 30–50% of protein-coding genes and modulate the expression of 10–30% of human genome [44,45]. RNAs also exist extracellularly in the circulation in exosomes [45].
Anesthetics affect miRNA in various cells across the body. In mice, sevoflurane and propofol affect expression of 46 of 177 miRNAs in liver, 20 miRNAs in lung, and 14 miRNAs in brain, with a specific pattern of expression at each anesthetic exposure [46,47,48]. Sevoflurane inhibits the NF-κB pathway through miRNA-9-5p expression and protects the liver from ischemia-reperfusion injury [49]. Sevoflurane also induces miRNA-155 downregulation that reduces systemic inflammation in acute lung injury models [50]. Propofol attenuates the neuroinflammation induced by lipopolysaccharide through miRNA-155 suppression [51].
Given these demonstrated effects on epigenetics, is it biologically plausible that anesthetics affect cancer’s natural history [18]? It is well known that epigenetic modulation in normal tissues may contrast or favor tumor progression, and anesthesia has the ability to change the epigenetics of surgical tissues [35,36,37,38,52]. Can anesthesia also directly alter cancer’s epigenetics? The question as to which mechanism can drive the anesthetic-induced alterations of tumor biology and host systems during a limited period of time such as the perioperative period, to the point that tumor behavior and its future clinical history change permanently, may have a response in epigenetics. However, to date there is no clear response a to whether it is the extent of surgery or stress response rather than anesthesia management that influences most cancer outcomes.

3. Anesthesia and Cancer Epigenetics

Epigenetics has a crucial role in cancero-genesis, which is a complex biological event. An altered gene and protein expression profile may affect cancer cells and the host response, inducing more or less favourable conditions for tumor progression. Cancer growth itself is driven by a “Darwinian” evolutionary process that starts in normal tissues where advantageous genetic and epigenetic events occur in a series of stages that promote clonal expansions of new tissue. Epigenetic changes can be subject to the same selection forces as genetic events [53]. This “new” tissue progressively misses the features of the original tissue. Every aspect of tumor biology including cell growth and differentiation, cell cycle control, cancer stem cell formation, DNA repair, angiogenesis, migration, and evasion of host immune surveillance is affected by epigenetic alterations [54,55].
Only few sporadic studies have investigated the role of anesthesia on cancer cell epigenetics and much is yet to be discovered (Figure 1). Differential gene expression was shown after ex-vivo exposure of brain cancer cell line SH-SY5Y and of breast cancer cell line MCF-7 to enflurane, isoflurane, desflurane, halothane, sevoflurane, and nitrous oxide [56].
Sevoflurane suppresses viability, invasion, migration, and apoptosis of colorectal cancer cells in a dose-dependent fashion by regulating the circ-HMGCS1/miRNA-34a-5p/SGPP1 axis, via inactivation of the Ras/Raf/MEK/ERK signaling, via regulation of ERK/MMP-9 pathway by up-regulating miRNA-203 and by regulating miRNA-34a/ADAM10 axis [57,58,59,60]. A study also showed a differential and specific impact on circulating exo-somial miRNAs during colon cancer surgery resection [61].
Other anesthetics such as the non-steroidal anti-inflammatory drugs, propofol and ketamine, may also modulate epigenetics. Celecoxib inhibits osteosarcoma cell proliferation, migration, and invasion via miRNA-34a [62]. miRNA-126-5p, −320a and -146a-5p regulate the sensitivity to celecoxib [63].
R-Ketorolac inhibits the activity of Rac1 and Cdc42, which are GTP-ases involved in cell growth and cell cycle regulation, in ovarian cancer patients. Both are believed to be therapeutic targets for ovarian cancer as regulators of migration, adhesion and invasion [64,65]. Studying epigenetics after ketorolac use may help in understanding the mechanism underlying this effect.
Lidocaine at clinical concentrations (1 mM) induced DNA demethylation for 120 h on BT-20 and MCF-7 breast cancer cells in vitro [66]. Lidocaine also reduces the proliferation of pancreatic cancer PaTu8988t cells after 48 h in vitro [67].
Ketamine, a N-methyl-Daspartate [NMDA] receptor antagonist used as a racemic derivative of two enantiomers s[+]ketamine and r[−]ketamine, can also modulate epigenetics. Exposure for 48 h with ketamine and s-ketamine of PaTu8988t pancreatic carcinoma cells significantly inhibited proliferation and expression of nuclear NFATc2 [67]. PaTu8988t and Panc-1 cells express the NMDA type R2a receptor. Ketamine and s-ketamine at 1000 μM concentration for 48 h significantly inhibited the proliferation of pancreatic cancer cells. S-ketamine reduced apoptosis after 3 h in PaTu8988t cells and in PANC-1 cells after 24 h incubation with ketamine [a] (65 ± 17%) and s-ketamine [b] (68 ± 24%). Necrosis increased after 16 h with ketamine and after 6–24 h after s-ketamine [68]. However, clinical doses of ketamine (2 mg/kg iv) produce an average plasma concentration of 41 μg/kg/min, which corresponds to a plasma concentration of 9.3 μM [68]. Ketamine has been recently found to suppress the viability of liver cancer cells and induce ferroptosis through the lncPVT1/miRNA-214-3p/GPX4 pathway [69]. In ovarian cancer, ketamine modulates the P300-mediated H3K27 acetylation activation in the promoter of lncRNA PVT1 that binds histone methyltransferase enhancer of zeste homolog 2 [EZH2], and regulates the expression of target genes, including p57, consequently regulating ovarian cancer cell growth, cell cycle control, apoptosis and colony forming [70].
Propofol (2,6-diisopropylphenol) is an extensively-used sedative anesthetic at 4-20 mcg/mL brain concentrations. Propofol regulates both miRNAs and lncRNAs, and modulates the signaling pathways of important oncogenes/onco-suppressors that are potential therapeutic targets including hypoxia-inducible factor-1α (HIF-1α), mitogen-activated protein kinase (MAPK), nuclear factor-kappaB (NF-κB), and nuclear factor E2-related factor-2 (Nrf2) [71,72].
Propofol upregulates miRNA-34a expression and induces miRNA-34a-dependent apoptosis in SH-SY5Y neuroblastoma cells in vitro [73,74]. At a concentration of 20 μg/mL for 72 h, propofol significantly reduces cell viability and apoptosis in human pancreatic PANC-1 cells increasing the expression of pro-apoptotic caspase-3 and Bax and down-regulating the expression of anti-apoptotic Bcl-2 gene. Apoptosis is induced by miRNA-3-4a-dependent upregulation of LOC285194 [75]. Additionally, propofol inhibits cell growth and metastasis by enhancing miR-328 expression in pancreatic cancer [76]. Propofol also induces a miRNA-34a-dependent E-cadherin upregulation in the PANC-1 cells, and reduces motility of cells is wound healing assays [75].
In human pancreatic Miapaca-2 and Panc-1 cells in vitro and murine pancreatic cancer cell (Panc02) in vivo, propofol shows a concentration-dependent (5, 25, 50, 100 μM) inhibition of cell migration, expression of VEGF and HIF-1α, phosphorylation of (ERK), AKT, (CaMK II), and Ca2+ concentration [77]. Propofol downregulates VEGF and suppresses migration of pancreatic cancer cells by inhibiting the NMDA receptor [77].
Propofol at concentrations of 1 to 10 μg/mL for 48 and 72 h also suppresses the proliferation and invasion of PANC-1 cells by 2.46- and 3.95-fold by upregulating miRNA-133a expression [78,79]. Apoptosis also increases after 1–10 microg/mL propofol exposure in a dose and time-dependent manner and through PUMA pathway [78,79].
Propofol inhibits the growth, invasion and migration of PANC-1 cells in a dose and time-dependent manner via the miRNA-21/Slug pathway and through E-cadherin upregulation [79]. Both PUMA and E-cadherin are upregulated by propofol via miRNA-21 inactivation and subsequent Slug inhibition [79].
The activation and expression of ADAM8 in response to hypoxia in PC is inhibited, thus antagonizing angiogenesis [80]. The downregulation of ADAM8 and upregulation of miRNA-328 mediated by propofol was shown to inhibit pancreatic cancer proliferation and metastasis [81]. Growth of xenograft pancreatic cancer is also inhibited in nude mice models by propofol [80]. Propofol shows a synergistic effect with gemcitabine through downregulation of NF-κB signaling pathway induced by gemcitabine, thereby promoting the chemosensitivity of PC [82].

4. Anesthetic Management

It remains uninvestigated how epigenetics changes after anesthesia and whether it may lead to an influence on cancer biology during the years after surgery, modifying its natural history and determining relevant clinical effects. To date, most available clinical studies are retrospective, small randomized trials or heterogeneous meta-analyses with negative or inconclusive results [6,7,8,9,83,84,85,86].
The major available trials have evaluated regional and general anesthesia showing that locoregional or epidural anesthesia do not affect cancer progression-related outcomes in breast (2132 patients), lung (400 patients) and cancer thoraco-abdominal surgery (1802 patients) [6,7,8]. These trials seem to definitively suggest that regional anesthesia-analgesia does not reduce recurrence after potentially curative cancer surgery [87,88].
However these trials were not constructed to detect differences between cancer subtypes or cancer grade. In one trial, only respectively did 13.5% and 12.5% of patients in the general anesthesia and epidural groups have advanced cancers [T3-4], whereas the other trial included multiple cancer types (gastrointestinal, hepatobiliary-pancreatic, lung-esophageal-thymic and genito-urinary and other cancers) [7,8].
The largest randomized controlled trial (The breast cancer recurrence trial, BCR), compared paravertebral to general anesthesia for breast cancer surgery. It included 2132 patients and showed no difference in cancer recurrence and survival between anesthetic techniques [6]. Contrarily to the study hypothesis based on residual disease, this trial included only low stage disease which has two implications. Firstly, surgery is highly curative at five years (>90%) in low grade disease as shown by the Mindact trial [89]. Secondly, surgery is these cases is less invasive leading to a lower stress response, lower pain and use of opioid, and reduced recurrence risk [90,91,92]. This is consistent with the finding that there is no metastatic progression after resection of primary breast cancer in murine orthotopic models if surgery is not associated with a large resection (laparotomy) [92]. In the human setting, patients having more invasive breast surgery (modified radical mastectomy), showed a significantly improved recurrence-free survival with propofol–total intravenous anesthesia compared with volatile-based anesthesia (HR, 0.55; 95% CI, 0.31 to 0.97; p = 0.037) [93].
60% of the participants of the BCR trial were enrolled from one hospital in China, which may have influenced the final results. Moreover, the study did not take into account surgical variability, was not able to detect patients with higher risk of recurrence, included the use of opiates for both groups, assessed only progesterone receptor status, and did not have an adequate follow-up time to assess recurrence in slow progression cancer phenotypes.
Based on the available preclinical and clinical data, amide local anesthetics and propofol are a plausible repurposing strategy frequently used on intravenous infusion for analgesia during cancer surgery. They are also a fundamental part in the setting of the opioid-free anesthesia strategy, which may have an important role in cancer outcomes [94]. Local anesthetics can modulate autonomic receptors, have a cytotoxic activity, and have an anti-inflammatory effect and anti-proliferative effects on mesenchymal stem cells [38,66]. However, whether the findings of preclinical research can have a clinical value depends on plasmatic concentration reachable after administration of anesthetics at human dose range. For example, the antitumor growth effect of ropivacaine and bupivacaine, two local anesthetics, occur at high concentrations and they do not affect apoptosis rate of pancreatic cancer cells at clinical concentrations in vitro [95]. To date only a retrospective study has reported that intraoperative intravenous lidocaine afforded a longer overall survival (HR = 0.616; 95% CI, 0.290–0.783; p = 0.013) in patients undergoing pancreatic cancer resection [96].
In the meantime, several randomized trials are being conducted on different cancer types (Table 1). The question is not simple, due also to the multifactorial and very complex situation including surgical, anesthetic, disease-specific and patients-linked factors. In this regard, the study of the influence of anesthesia on pancreatic cancer is at a very early phase of study and is based on retrospective studies.
Propofol afforded a better survival pattern than desflurane in pancreatic cancer surgery in a retrospective cohort study [97]. In this study, the overall mortality rate was 60.0% with propofol and 82.0% with desflurane (p = 0.006). Cancer-specific mortality rate was 57.0% with propofol and 78.0% with desflurane (p = 0.014) (HR 0.63) (95% CI, 0.42–0.95; p = 0.028). Recurrence was 43.0% with propofol and 66.0% with desflurane (p = 0.010) (HR 0.53 (95% CI, 0.34–0.84; p = 0.007) with no difference in postoperative metastases. Propensity score-matched HR for mortality and recurrence were respectively 0.63 (95% CI, 0.40–0.97; p = 0.037) and 0.55 (95% CI, 0.34–0.90; p = 0.028). Survival was 40.0% with propofol but 18% with desflurane (crude hazard ratio (HR 0.63 (95% confidence interval (CI), 0.42–0.93; p = 0.021) and HR, 0.53; 95% CI, 0.32–0.86; p = 0.010 in the multivariable analysis after adjustment). All cause survival was better with propofol than with desflurane (HR 0.63 (95% CI, 0.42–0.93; p = 0.021), propensity score matched HR 0.65 (95% CI, 0.42–0.99; p = 0.047). Conversely, desflurane anesthesia increased all-cause mortality, cancer-specific mortality and cancer progression [97].
Perioperative epidural anesthesia and analgesia and regional anesthesia have been evaluated for cancer survival after pancreatic surgery [98]. Epidural and regional anesthesia block nociception and reduce adrenergic activity, thus favoring the parasympathetic system [1]. However to date results of clinical trials are negative. In a retrospective study on 366 patients, epidural-TIVA (propofol-lidocaine-dexmedetomidine-ketamine) showed no differences in recurrence, metastasis, mortality, platelet-lymphocyte ratios (PLR) and neutrophil-lymphocyte ratios (NLR), compared with epidural-volatile-opioid anesthesia [midazolam-propofol-fentanyl-desflurane-hydromorphone] in pancreatic cancer surgery [99]. Recently, a heterogeneous meta-analysis including three RCT revealed no difference between regional and general anesthesia for late stage abdominal cancers [100].
Perineural invasion is a common feature of PC and is driven by the adrenergic system through neurotrophins. The sympathetic system acts though beta-adrenergic receptors (β-ARs) to favor PC activity. Conversely, beta-blocking agents such as propranolol inhibit PC progression [1]. To date three trials are evaluating the role of propranolol as an adjuvant to other anticancer treatments for PC (Table 1).
Finally, on the basis of retrospective studies, dexamethasone associated to peridural anesthesia may be associated with a reduced mortality risk and an improved recurrence-free after pancreatic cancer surgery [97,98].

5. Conclusions

Based on preclinical observations, it is to be hoped that the epigenetic effects of anesthetics on the biological mechanisms that determine the natural history of cancers in the perioperative period will be better investigated. Previous preclinical studies show that anesthetics induce epigenetic changes that can last for months and can be intergenerational [18]. This is an important observation, since such an epigenetic effect may be possible in residual cancer disease after surgery. Moreover, anesthesia alters host response and epigenetics of surgical tissues [35,36,37,38]. Most studies have investigated RNAs and there are only few studies investigating the other epigenetic mechanisms. The ability of anesthetics to affect epigenetics in germinal cells can be also a plausible sign that immature cancer cells may be susceptible to anesthetics.
Most preclinical or clinical studies still address single gene products and results are difficult to interpret in a setting where thousands of other genes interact with each other, along with other perioperative factors. One of the most investigated anesthetics is propofol and its role in the modulation of miRNAs. There is thus a need for studies investigating the whole genome. Microarrays, new generation sequencing (NGS) and RNAseq techniques can capture all the RNA-related information in a single test. Their integration into clinical trials may uncover unanticipated findings whose clinical impact is, at present, impossible to quantify. Several ‘proof-of-principle’ works show that gene expression profiling can predict important clinical outcomes and therefore have the potential to evolve into true diagnostic tests or drive clinical decision and implement personalized clinical care [89,101,102,103]. To date, there are no available studies of genome-wide analysis of the expression changes in tumors and surgical tissues induced by different anesthesia techniques.
Secondly, there is a need for robust prospective clinical studies, exploring anesthetics (propofol, lidocaine, nonsteroidal anti-inflammatory drugs, ketamine and propranolol), according to standardized outcomes, stratified in order to detect differences in the type and stage of cancer, and integrating genetic characterization of cancers [89,104,105]. The Standardizing Endpoints for Perioperative Medicine Group used a Delphi process to recommend, as appropriate endpoints for prospective trials of anesthetic technique on cancer outcomes, the cancer health related quality of life, days alive and out of hospital at 90 days, time to cancer progression, disease-free survival, cancer-specific survival, and overall survival (and 5-yr overall survival) [105]. The hypothesis that anesthetics may play a role in the recurrence of neuroendocrine pancreatic cancers and after endoscopic ultrasound-guided tumor ablation for pancreatic cancer should also be investigated [106,107].
Genetic profiling is a fundamental tool in order to take the disease into account in terms of clinical and genetic variability [89]. There is thus a need for randomized trials comparing different anesthetic techniques that integrate genetic profiling and genome-wide analysis of the gene expression changes [103,108]. A prospective trial design for genomic studies minimizes experimental bias and ensures that all samples are handled and processed in a standardized fashion [101,109]. To date only one study protocol (NCT03779685: GENe EXpression After Regional or General ANesthesia in Patients Undergoing Breast Cancer Surgery (GENEXAN)) has been built to incorporate gene expression analysis into a randomized trial comparing regional to general anesthesia for breast cancer surgery.
Rather than a single disease, cancer is an extremely variable disease from a genetic point of view. The higher the variability of the disease or population, the more the risk of losing effects on subpopulations in large trials who do not show variability. The MINDACT trial, for example, stratified breast cancer patients according to clinical and genetic risk instead of clinical risk alone, finding that patients with a high clinical risk but with a low genomic risk, randomized to chemotherapy or not, showed only a 2.6 percentage points benefit on the treatment group at 8 years follow-up, signifying that nearly 46% of high clinical risk patients can be treated with endocrine therapy alone [89]. A crucial aspect of generalizing clinical trials is that, in complex situations (cancer’s multiple phenotype, a long disease history, lack of a receptorial mechanism), we cannot expect a linear relationship between the action (anesthesia) and the outcome.
Another problem that may cover the effect of anesthesia and reduce the enthusiasm for further investigation is surgical curability, which is high today, especially in low grade cancers [3]. On the other hand the main cause of postoperative cancer death is metastasis, which occurs in one third of operated patients [110,111,112]. Only a small number of patients with pancreatic cancer will be operable and, among these, 7% will survive at 5 years, indicating a high recurrence rate and a lack of effective treatments [113]. It is well known that epigenetic mechanisms drive cancero-genesis in pancreatic cancer [114]. For these reasons, studies on epigenetics and pancreatic cancer with different anesthetics can be a promising field of research.

Author Contributions

Conceptualization, Z.M. and P.F.; methodology, Z.M.; writing—original draft preparation, Z.M.; writing—review and editing, A.C. and P.F.; visualization, Z.M.; supervision, P.F. and A.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We wish to thank Fabio Verginelli for sharing his expertise.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. De Lellis, L.; Veschi, S.; Tinari, N.; Mokini, Z.; Carradori, S.; Brocco, D.; Florio, R.; Grassadonia, A.; Cama, A. Drug Repurposing, an Attractive Strategy in Pancreatic Cancer Treatment: Preclinical and Clinical Updates. Cancers 2021, 13, 3946. [Google Scholar] [CrossRef] [PubMed]
  2. Hiller, J.G.; Perry, N.J.; Poulogiannis, G.; Riedel, B.; Sloan, E.K. Perioperative events influence cancer recurrence risk after surgery. Nat. Rev. Clin. Oncol. 2018, 15, 205–218. [Google Scholar] [CrossRef] [PubMed]
  3. Sessler, I.D.; Riedel, B. Anesthesia and Cancer Recurrence: Context for Divergent Study Outcomes. Anesthesiology 2019, 130, 3–5. [Google Scholar] [CrossRef] [PubMed]
  4. Lin, D.; Shen, L.; Luo, M.; Zhang, K.; Li, J.; Yang, Q.; Zhu, F.; Zhou, D.; Zheng, S.; Chen, Y.; et al. Circulating tumor cells: Biology and clinical significance. Signal Transduct. Target. Ther. 2021, 6, 404. [Google Scholar] [CrossRef] [PubMed]
  5. Forget, P.; Aguirre, J.A.; Bencic, I.; Borgeat, A.; Cama, A.; Condron, C.; Eintrei, C.; Eroles, P.; Gupta, A.; Hales, T.G.; 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 2019, 11, 592. [Google Scholar] [CrossRef]
  6. Sessler, I.D.; Pei, L.; Huang, Y.; Fleischmann, E.; Marhofer, P.; Kurz, A.; Mayers, D.B.; Meyer-Treschan, A.T.; Grady, M.; Tan, E.Y.; et al. Recurrence of breast cancer after regional or general anaesthesia: A randomised controlled trial. Lancet 2019, 394, 1807–1815. [Google Scholar] [CrossRef]
  7. Du, Y.-T.; Li, Y.-W.; Zhao, B.-J.; Guo, X.-Y.; Feng, Y.; Zuo, M.-Z.; Fu, C.; Zhou, W.-J.; Li, H.-J.; Liu, Y.-F.; et al. Long-term Survival after Combined Epidural–General Anesthesia or General Anesthesia alone: Follow-up of a Randomized Trial. Anesthesiology 2021, 135, 233–245. [Google Scholar] [CrossRef]
  8. Xu, Z.-Z.; Li, H.-J.; Li, M.-H.; Huang, S.-M.; Li, X.; Liu, Q.-H.; Li, J.; Wang, D.-X.; Sessler, D.I. Epidural Anesthesia–Analgesia and Recurrence-free Survival after Lung Cancer Surgery: A Randomized Trial. Anesthesiology 2021, 135, 419–432. [Google Scholar] [CrossRef]
  9. Lilley, R.; Chan, E.; Ng, N.; Orr, A.; Szostok, M.; Yeh, G.T.T.; Tulloch, R.; Ramsay, G.; Mokini, Z.; Forget, P. Recurrence Kinetics after Laparoscopic versus Open Surgery in Colon Cancer. A Meta-Analysis. J. Clin. Med. 2021, 10, 4163. [Google Scholar] [CrossRef]
  10. Jenuwein, T.; Allis, C.D. Translating the Histone Code. Science 2001, 293, 1074–1080. [Google Scholar] [CrossRef]
  11. Berger, S.L. The complex language of chromatin regulation during transcription. Nature 2007, 447, 407–412. [Google Scholar] [CrossRef] [PubMed]
  12. Kouzarides, T. Chromatin modifications and their function. Cell 2007, 128, 693–705. [Google Scholar] [CrossRef] [PubMed]
  13. Jaenisch, R.; Bird, A. Epigenetic regulation of gene expression: How the genome integrates intrinsic and environmental signals. Nat. Genet. 2003, 33, 245–254. [Google Scholar] [CrossRef] [PubMed]
  14. Ozanne, E.S.; Constancia, M. Mechanisms of Disease: The developmental origins of disease and the role of the epigenotype. Nat. Clin. Pract. Endocrinol. Metab. 2007, 3, 539–546. [Google Scholar] [CrossRef] [PubMed]
  15. Kangaspeska, S.; Stride, B.; Métivier, R.; Polycarpou-Schwarz, M.; Ibberson, D.; Carmouche, R.P.; Benes, V.; Gannon, F.; Reid, G. Transient cyclical methylation of promoter DNA. Nature 2008, 452, 112–115. [Google Scholar] [CrossRef]
  16. Métivier, R.; Gallais, R.; Tiffoche, C.; Le Péron, C.; Jurkowska, R.Z.; Carmouche, R.P.; Ibberson, D.; Barath, P.; Demay, F.; Reid, G.; et al. Cyclical DNA methylation of a transcriptionally active promoter. Nature 2008, 452, 45–50. [Google Scholar] [CrossRef]
  17. Wang, K.; Tian, Y.; Zhang, Y.; Li, X.; Wei, X.; Hu, H.; Xu, S. Toxicity mechanism of sevoflurane in neural stem cells of rats through DNA methylation. Exp. Ther. Med. 2019, 18, 237–241. [Google Scholar] [CrossRef]
  18. Martynyuk, E.A.; Ju, L.-S.; Morey, E.T.; Zhang, J.-Q. Neuroendocrine, epigenetic, and intergenerational effects of general anesthetics. World J. Psychiatry 2020, 10, 81–94. [Google Scholar] [CrossRef]
  19. Lin, E.P.; Lee, J.-R.; Lee, C.S.; Deng, M.; Loepke, A.W. Do anesthetics harm the developing human brain? An integrative analysis of animal and human studies. Neurotoxicology Teratol. 2017, 60, 117–128. [Google Scholar] [CrossRef]
  20. Sakamoto, A.; Imai, J.-I.; Nishikawa, A.; Honma, R.; Ito, E.; Yanagisawa, Y.; Kawamura, M.; Ogawa, R.; Watanabe, S. Influence of inhalation anesthesia assessed by comprehensive gene expression profiling. Gene 2005, 356, 39–48. [Google Scholar] [CrossRef]
  21. Kobayashi, K.; Takemori, K.; Sakamoto, A. Circadian gene expression is suppressed during sevoflurane anesthesia and the suppression persists after awakening. Brain Res. 2007, 1185, 1–7. [Google Scholar] [CrossRef] [PubMed]
  22. Nakazato, K.; Yoshida, Y.; Takemori, K.; Kobayashi, K.; Sakamoto, A. Expressions of genes encoding drug-metabolizing enzymes are altered after sevoflurane, isoflurane, propofol or dexmedetomidine anesthesia. Biomed. Res. 2009, 30, 17–24. [Google Scholar] [CrossRef] [PubMed]
  23. Tsuboko, Y.; Sakamoto, A. Propofol anaesthesia alters the cerebral proteome differently from sevoflurane anaesthesia. Biomed. Res. 2011, 32, 55–65. [Google Scholar] [CrossRef]
  24. Heindel, J.J.; Balbus, J.; Birnbaum, L.; Brune-Drisse, M.N.; Grandjean, P.; Gray, K.; Landrigan, P.J.; Sly, P.D.; Suk, W.A.; Cory Slechta, D.; et al. Developmental Origins of Health and Disease: Integrating Environmental Influences. Endocrinology 2015, 156, 3416–3421. [Google Scholar] [CrossRef] [PubMed]
  25. Barker, D.J.P. The origins of the developmental origins theory. J. Intern. Med. 2007, 261, 412–417. [Google Scholar] [CrossRef] [PubMed]
  26. Gluckman, P.D.; Hanson, A.M.; Mitchell, M.D. Developmental origins of health and disease: Reducing the burden of chronic disease in the next generation. Genome Med. 2010, 2, 14. [Google Scholar] [CrossRef] [PubMed]
  27. Baird, J.; Jacob, C.; Barker, M.; Fall, C.H.D.; Hanson, M.; Harvey, N.C.; Inskip, H.M.; Kumaran, K.; Cooper, C. Developmental Origins of Health and Disease: A Lifecourse Approach to the Prevention of Non-Communicable Diseases. Healthcare 2017, 5, 14. [Google Scholar] [CrossRef]
  28. Donkin, I.; Versteyhe, S.; Ingerslev, L.R.; Qian, K.; Mechta, M.; Nordkap, L.; Mortensen, B.; Appel, E.V.R.; Jørgensen, N.; Kristiansen, V.B.; et al. Obesity and Bariatric Surgery Drive Epigenetic Variation of Spermatozoa in Humans. Cell Metab. 2016, 23, 369–378. [Google Scholar] [CrossRef]
  29. Chastain-Potts, S.E.; Tesic, V.; Tat, Q.L.; Cabrera, O.H.; Quillinan, N.; Jevtovic-Todorovic, V. Sevoflurane Exposure Results in Sex-Specific Transgenerational Upregulation of Target IEGs in the Subiculum. Mol. Neurobiol. 2020, 57, 11–22. [Google Scholar] [CrossRef]
  30. Bates, S.E. Epigenetic Therapies for Cancer. New Engl. J. Med. 2020, 383, 650–663. [Google Scholar] [CrossRef]
  31. Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef] [PubMed]
  32. Jia, M.; Liu, W.-X.; Yang, J.-J.; Xu, N.; Xie, Z.-M.; Ju, L.-S.; Ji, M.-H.; Martynyuk, A.E.; Yang, J.-J. Role of histone acetylation in long-term neurobehavioral effects of neonatal Exposure to sevoflurane in rats. Neurobiol. Dis. 2016, 91, 209–220. [Google Scholar] [CrossRef] [PubMed]
  33. Cabrera, O.H.; Useinovic, N.; Jevtovic-Todorovic, V. Neonatal anesthesia and dysregulation of the epigenome. Biol. Reprod. 2021, 105, 720–734. [Google Scholar] [CrossRef]
  34. Sun, Y.; Sahbaie, P.; Liang, D.; Li, W.; Shi, X.; Kingery, P.; Clark, J.D. DNA Methylation Modulates Nociceptive Sensitization after Incision. PLoS ONE 2015, 10, e0142046. [Google Scholar] [CrossRef] [PubMed]
  35. Caputi, F.F.; Carboni, L.; Rullo, L.; Alessandrini, I.; Balzani, E.; Melotti, R.M.; Romualdi, P.; Candeletti, S.; Fanelli, A. An Exploratory Pilot Study of Changes in Global DNA Methylation in Patients Undergoing Major Breast Surgery under Opioid-Based General Anesthesia. Front. Pharmacol. 2021, 12, 733577. [Google Scholar] [CrossRef]
  36. Doehring, A.; Oertel, B.G.; Sittl, R.; Lötsch, J. Chronic opioid use is associated with increased DNA methylation correlating with increased clinical pain. Pain 2013, 154, 15–23. [Google Scholar] [CrossRef]
  37. Fragou, D.; Zanos, P.; Kouidou, S.; Njau, S.; Kitchen, I.; Bailey, A.; Kovatsi, L. Effect of chronic heroin and cocaine administration on global DNA methylation in brain and liver. Toxicol. Lett. 2013, 218, 260–265. [Google Scholar] [CrossRef]
  38. Lirk, P.; Fiegl, H.; Weber, N.C.; Hollmann, M.W. Epigenetics in the perioperative period. Br. J. Pharmacol. 2015, 172, 2748–2755. [Google Scholar] [CrossRef]
  39. Liu, Y.; Ding, M.; Gao, Q.; He, A.; Liu, Y.; Mei, H. Current Advances on the Important Roles of Enhancer RNAs in Gene Regulation and Cancer. BioMed Res. Int. 2018, 2018, 2405351. [Google Scholar] [CrossRef]
  40. Palazzo, A.F.; Lee, E.S. Non-coding RNA: What is functional and what is junk? Front. Genet. 2015, 6, 2. [Google Scholar] [CrossRef]
  41. Amin, N.; McGrath, A.; Chen, Y.-P.P. Author Correction: Evaluation of deep learning in non-coding RNA classification. Nat. Mach. Intell. 2020, 2, 236. [Google Scholar] [CrossRef]
  42. Panni, S.; Lovering, R.C.; Porras, P.; Orchard, S. Non-coding RNA regulatory networks. Biochim. Biophys. Acta. Gene Regul. Mech. 2020, 1863, 194417. [Google Scholar] [CrossRef] [PubMed]
  43. Catalanotto, C.; Cogoni, C.; Zardo, G. MicroRNA in Control of Gene Expression: An Overview of Nuclear Functions. Int. J. Mol. Sci. 2016, 17, 1712. [Google Scholar] [CrossRef] [PubMed]
  44. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef] [PubMed]
  45. Shu, Z.; Tan, J.; Miao, Y.; Zhang, Q. The role of microvesicles containing microRNAs in vascular endothelial dysfunction. J. Cell. Mol. Med. 2019, 23, 7933–7945. [Google Scholar] [CrossRef] [PubMed]
  46. Ishikawa, M.; Tanaka, S.; Arai, M.; Genda, Y.; Sakamoto, A. Differences in microRNA Changes of Healthy Rat Liver between Sevoflurane and Propofol Anesthesia. Anesthesiology 2012, 117, 1245–1252. [Google Scholar] [CrossRef] [PubMed]
  47. Tanaka, S.; Ishikawa, M.; Arai, M.; Genda, Y.; Sakamoto, A. Changes in microRNA expression in rat lungs caused by sevoflurane anesthesia: A TaqMan® low-density array study. Biomed. Res. 2012, 33, 255–263. [Google Scholar] [CrossRef]
  48. Goto, G.; Hori, Y.; Ishikawa, M.; Tanaka, S.; Sakamoto, A. Changes in the gene expression levels of microRNAs in the rat hippocampus by sevoflurane and propofol anesthesia. Mol. Med. Rep. 2014, 9, 1715–1722. [Google Scholar] [CrossRef]
  49. Liao, X.; Zhou, S.; Zong, J.; Wang, Z. Sevoflurane exerts protective effects on liver ischemia/reperfusion injury by regulating NFKB3 expression via miR-9-5p. Exp. Ther. Med. 2019, 17, 2632–2640. [Google Scholar] [CrossRef]
  50. Otsuki, T.; Ishikawa, M.; Hori, Y.; Goto, G.; Sakamoto, A. Volatile anesthetic sevoflurane ameliorates endotoxin-induced acute lung injury via microRNA modulation in rats. Biomed. Rep. 2015, 3, 408–412. [Google Scholar] [CrossRef]
  51. Lu, Y.; Jian, M.-Y.; Ouyang, Y.-B.; Han, R.-Q. Changes in Rat Brain MicroRNA Expression Profiles Following Sevoflurane and Propofol Anesthesia. Chin. Med J. 2015, 128, 1510–1515. [Google Scholar] [CrossRef] [PubMed]
  52. Li, E. Chromatin modification and epigenetic reprogramming in mammalian development. Nat. Rev. Genet. 2002, 3, 662–673. [Google Scholar] [CrossRef] [PubMed]
  53. Nowell, P.C. The clonal evolution of tumor cell populations. Science 1976, 194, 23–28. [Google Scholar] [CrossRef] [PubMed]
  54. Lund, A.H.; van Lohuizen, M. Epigenetics and cancer. Genes Dev. 2004, 18, 2315–2335. [Google Scholar] [CrossRef] [PubMed]
  55. Wainwright, E.N.; Scaffidi, P. Epigenetics and Cancer Stem Cells: Unleashing, Hijacking, and Restricting Cellular Plasticity. Trends Cancer 2017, 3, 372–386. [Google Scholar] [CrossRef] [PubMed]
  56. Huitink, J.M.; Heimerikxs, M.; Nieuwland, M.; Loer, S.A.; Brugman, W.; Velds, A.; Sie, D.; Kerkhoven, R.M. Volatile Anesthetics Modulate Gene Expression in Breast and Brain Tumor Cells. Anesth. Analg. 2010, 111, 1411–1415. [Google Scholar] [CrossRef] [PubMed]
  57. He, J.; Zhao, H.; Liu, X.; Wang, D.; Wang, Y.; Ai, Y.; Yang, J. Sevoflurane suppresses cell viability and invasion and promotes cell apoptosis in colon cancer by modulating exosome-mediated circ-HMGCS1 via the miR-34a-5p/SGPP1 axis. Oncol. Rep. 2020, 44, 2429–2442. [Google Scholar] [CrossRef]
  58. Yang, X.; Zheng, Y.-T.; Rong, W. Sevoflurane induces apoptosis and inhibits the growth and motility of colon cancer in vitro and in vivo via inactivating Ras/Raf/MEK/ERK signaling. Life Sci. 2019, 239, 116916. [Google Scholar] [CrossRef]
  59. Fan, L.; Wu, Y.; Wang, J.; He, J.; Han, X. Sevoflurane inhibits the migration and invasion of colorectal cancer cells through regulating ERK/MMP-9 pathway by up-regulating miR-203. Eur. J. Pharmacol. 2019, 850, 43–52. [Google Scholar] [CrossRef]
  60. Sun, S.Q.; Ren, L.J.; Liu, J.; Wang, P.; Shan, S.M. Sevoflurane inhibits migration and invasion of colorectal cancer cells by regulating microRNA-34a/ADAM10 axis. Neoplasma 2019, 66, 887–895. [Google Scholar] [CrossRef]
  61. Buschmann, D.; Brandes, F.; Lindemann, A.; Maerte, M.; Ganschow, P.; Chouker, A.; Schelling, G.; Pfaffl, M.W.; Reithmair, M. Propofol and Sevoflurane Differentially Impact MicroRNAs in Circulating Extracellular Vesicles during Colorectal Cancer Resection: A Pilot Study. Anesthesiology 2020, 132, 107–120. [Google Scholar] [CrossRef] [PubMed]
  62. Chen, X.; Peng, D.; Shen, Y.; Liu, B.; Zhou, H.; Tao, H.; Huang, J. The potential combinational effect of miR-34a with celecoxib in osteosarcoma. Anti-Cancer Drugs 2017, 28, 888–897. [Google Scholar] [CrossRef] [PubMed]
  63. Dong, Z.; Jiang, H.; Jian, X.; Zhang, W. Change of miRNA expression profiles in patients with knee osteoarthritis before and after celecoxib treatment. J. Clin. Lab. Anal. 2019, 33, e22648. [Google Scholar] [CrossRef]
  64. Guo, Y.; Kenney, S.R.; Cook, L.; Adams, S.F.; Rutledge, T.; Romero, E.; Oprea, T.I.; Sklar, L.A.; Bedrick, E.; Wiggins, C.L.; et al. A Novel Pharmacologic Activity of Ketorolac for Therapeutic Benefit in Ovarian Cancer Patients. Clin. Cancer Res. 2015, 21, 5064–5072. [Google Scholar] [CrossRef] [PubMed]
  65. Guo, Y.; Kenney, S.R.; Muller, C.Y.; Adams, S.; Rutledge, T.; Romero, E.; Murray-Krezan, C.; Prekeris, R.; Sklar, L.A.; Hudson, L.G.; et al. R-Ketorolac Targets Cdc42 and Rac1 and Alters Ovarian Cancer Cell Behaviors Critical for Invasion and Metastasis. Mol. Cancer Ther. 2015, 14, 2215–2227. [Google Scholar] [CrossRef]
  66. Lirk, P.; Berger, R.; Hollmann, M.W.; Fiegl, H. Lidocaine time- and dose-dependently demethylates deoxyribonucleic acid in breast cancer cell lines in vitro. Br. J. Anaesth. 2012, 109, 200–207. [Google Scholar] [CrossRef]
  67. Malsy, M.; Graf, B.; Bundscherer, A. The Effects of Analgesics and Local Anesthetics on Gene Transcription Mediated by NFATc2 and Sp1 in Pancreatic Carcinoma. Anticancer Res. 2019, 39, 4721–4728. [Google Scholar] [CrossRef]
  68. Malsy, M.; Gebhardt, K.; Gruber, M.; Wiese, C.; Graf, B.; Bundscherer, A. Effects of ketamine, s-ketamine, and MK 801 on proliferation, apoptosis, and necrosis in pancreatic cancer cells. BMC Anesthesiol. 2015, 15, 111. [Google Scholar] [CrossRef]
  69. He, G.-N.; Bao, N.-R.; Wang, S.; Xi, M.; Zhang, T.-H.; Chen, F.-S. Ketamine Induces Ferroptosis of Liver Cancer Cells by Targeting lncRNA PVT1/miR-214-3p/GPX4. Drug Des. Dev. Ther. 2021, 15, 3965–3978. [Google Scholar] [CrossRef]
  70. Li, T.; Yang, J.; Yang, B.; Zhao, G.; Lin, H.; Liu, Q.; Wang, L.; Wan, Y.; Jiang, H. Ketamine Inhibits Ovarian Cancer Cell Growth by Regulating the lncRNA-PVT1/EZH2/p57 Axis. Front. Genet. 2021, 11, 597467. [Google Scholar] [CrossRef]
  71. Jiang, S.; Liu, Y.; Huang, L.; Zhang, F.; Kang, R. Effects of propofol on cancer development and chemotherapy: Potential mechanisms. Eur. J. Pharmacol. 2018, 831, 46–51. [Google Scholar] [CrossRef] [PubMed]
  72. Mirzaei, S.; Zarrabi, A.; Hashemi, F.; Zabolian, A.; Saleki, H.; Ranjbar, A.; Saleh, S.H.; Bagherian, M.; Sharifzadeh, S.O.; Hushmandi, K.; et al. Regulation of Nuclear Factor-KappaB (NF-κB) signaling pathway by non-coding RNAs in cancer: Inhibiting or promoting carcinogenesis? Cancer Lett. 2021, 509, 63–80. [Google Scholar] [CrossRef]
  73. Xing, N.; Xing, F.; Li, Y.; Li, P.; Zhang, J.; Wang, D.; Zhang, W.; Yang, J. Dexmedetomidine improves propofol-induced neuronal injury in rat hippocampus with the involvement of miR-34a and the PI3K/Akt signaling pathway. Life Sci. 2020, 247, 117359. [Google Scholar] [CrossRef] [PubMed]
  74. Li, G.F.; Li, Z.B.; Zhuang, S.J. Inhibition of microRNA-34a protects against propofol anesthesia-induced neurotoxicity and cognitive dysfunction via the MAPK/ERK signaling pathway. Neurosci. Lett. 2018, 675, 152–159. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, H.; Jiao, H.; Jiang, Z.; Chen, R. Propofol inhibits migration and induces apoptosis of pancreatic cancer PANC-1 cells through miR-34a-mediated E-cadherin and LOC285194 signals. Bioengineered 2020, 11, 510–521. [Google Scholar] [CrossRef] [PubMed]
  76. Yu, X.; Gao, Y.; Zhang, F. Propofol inhibits pancreatic cancer proliferation and metastasis by up-regulating miR-328 and down-regulating ADAM8. Basic Clin. Pharmacol. Toxicol. 2019, 125, 271–278. [Google Scholar] [CrossRef]
  77. Chen, X.; Wu, Q.; You, L.; Chen, S.; Zhu, M.; Miao, C. Propofol attenuates pancreatic cancer malignant potential via inhibition of NMDA receptor. Eur. J. Pharmacol. 2017, 795, 150–159. [Google Scholar] [CrossRef]
  78. Wang, Z.; Gong, H.; Zheng, F.; Liu, D.; Dong, T. Propofol suppresses proliferation and invasion of pancreatic cancer cells by upregulating microRNA-133a expression. Genet. Mol. Res. 2015, 14, 7529–7537. [Google Scholar] [CrossRef]
  79. Liu, Z.; Zhang, J.; Hong, G.; Quan, J.; Zhang, L.; Yu, M. Propofol Inhibits Growth and Invasion of Pancreatic Cancer Cells through Regulation of the MiR-21/Slug Signaling Pathway. Am. J. Transl. Res. 2016, 8, 4120–4133. [Google Scholar]
  80. Gao, Y.; Yu, X.; Zhang, F.; Dai, J. Propofol inhibits pancreatic cancer progress under hypoxia via ADAM 8. J. Hepato-Biliary-Pancreatic Sci. 2019, 26, 219–226. [Google Scholar] [CrossRef]
  81. Yu, X.; Shi, J.; Wang, X.; Zhang, F. Propofol affects the growth and metastasis of pancreatic cancer via ADAM8. Pharmacol. Rep. 2019, 72, 418–426. [Google Scholar] [CrossRef] [PubMed]
  82. Du, Q.-H. Propofol induces apoptosis and increases gemcitabine sensitivity in pancreatic cancer cells in vitro by inhibition of nuclear factor-κB activity. World J. Gastroenterol. 2013, 19, 5485–5492. [Google Scholar] [CrossRef] [PubMed]
  83. Grandhi, R.K.; Lee, S.; Abd-Elsayed, A. The Relationship between Regional Anesthesia and Cancer: A Metaanalysis. Ochsner J. 2017, 17, 345–361. [Google Scholar] [PubMed]
  84. Cakmakkaya, O.S.; Kolodzie, K.; Apfel, C.C.; Pace, N.L. Anaesthetic techniques for risk of malignant tumour recurrence. Cochrane Database Syst. Rev. 2014, 11, CD008877. [Google Scholar] [CrossRef] [PubMed]
  85. Ishikawa, M.; Iwasaki, M.; Sakamoto, A.; Ma, D. Anesthetics may modulate cancer surgical outcome: A possible role of miRNAs regulation. BMC Anesthesiol. 2021, 21, 71. [Google Scholar] [CrossRef] [PubMed]
  86. Ponferrada, A.R.; Orriach, J.L.G.; Manso, A.M.; Haro, E.S.; Molina, S.R.; Heredia, A.F.; Lopez, M.B.; Mañas, J.C. Anaesthesia and cancer: Can anaesthetic drugs modify gene expression? Ecancermedicalscience 2020, 14, 1080. [Google Scholar] [CrossRef] [PubMed]
  87. Buggy, D.J.; Riedel, B.; Sessler, D.I. Can anaesthetic technique influence cancer outcome? The next step. Br. J. Anaesth. 2021, 127, 5–7. [Google Scholar] [CrossRef]
  88. Enlund, M. Is It Definitely Clear That Long-Term Survival after Breast Cancer Surgery Is Not Affected by Anaesthetics? Cancers 2021, 13, 3390. [Google Scholar] [CrossRef]
  89. Piccart, M.; Veer, L.J.V.; Poncet, C.; Cardozo, J.M.N.L.; Delaloge, S.; Pierga, J.-Y.; Vuylsteke, P.; Brain, E.; Vrijaldenhoven, S.; Neijenhuis, A.P.; et al. 70-gene signature as an aid for treatment decisions in early breast cancer: Updated results of the phase 3 randomised MINDACT trial with an exploratory analysis by age. Lancet Oncol. 2021, 22, 476–488. [Google Scholar] [CrossRef]
  90. Wigmore, T.J.; 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. [Google Scholar] [CrossRef]
  91. Lacy, A.M.; García-Valdecasas, J.C.; Delgado, S.; Castells, A.; Taurá, P.; Piqué, J.M.; Visa, J. Laparoscopy-assisted colectomy versus open colectomy for treatment of non-metastatic colon cancer: A randomised trial. Lancet 2002, 359, 2224–2229. [Google Scholar] [CrossRef]
  92. Martin, M.A.; Meyricke, R.; O’Neill, T.; Roberts, S. Breast-Conserving Surgery Versus Mastectomy for Survival from Breast Cancer: The Western Australian Experience. Ann. Surg. Oncol. 2007, 14, 157–164. [Google Scholar] [CrossRef] [PubMed]
  93. Lee, J.H.; Kang, S.H.; Kim, Y.; Kim, H.-A.; Kim, B.S. Effects of propofol-based total intravenous anesthesia on recurrence and overall survival in patients after modified radical mastectomy: A retrospective study. Korean J. Anesthesiol. 2016, 69, 126–132. [Google Scholar] [CrossRef] [PubMed]
  94. Malo-Manso, A.; Raigon-Ponferrada, A.; Diaz-Crespo, J.; Escalona-Belmonte, J.J.; Cruz-Mañas, J.; Guerrero-Orriach, J.L. Opioid Free Anaesthesia and Cancer. Curr. Pharm. Des. 2019, 25, 3011–3019. [Google Scholar] [CrossRef]
  95. Bundscherer, A.; Malsy, M.; Gebhardt, K.; Metterlein, T.; Plank, C.; Wiese, C.; Gruber, M.; Graf, B. Effects of ropivacaine, bupivacaine and sufentanil in colon and pancreatic cancer cells in vitro. Pharmacol. Res. 2015, 95–96, 126–131. [Google Scholar] [CrossRef]
  96. Zhang, H.; Yang, L.; Zhu, X.; Zhu, M.; Sun, Z.; Cata, J.P.; Chen, W.; Miao, C. Association between intraoperative intravenous lidocaine infusion and survival in patients undergoing pancreatectomy for pancreatic cancer: A retrospective study. Br. J. Anaesth. 2020, 125, 141–148. [Google Scholar] [CrossRef]
  97. Lai, H.-C.; Lee, M.-S.; Liu, Y.-T.; Lin, K.-T.; Hung, K.-C.; Chen, J.-Y.; Wu, Z.-F. Propofol-based intravenous anesthesia is associated with better survival than desflurane anesthesia in pancreatic cancer surgery. PLoS ONE 2020, 15, e0233598. [Google Scholar] [CrossRef]
  98. Call, T.R.; Pace, N.L.; Thorup, D.B.; Maxfield, D.; Chortkoff, B.; Christensen, J.; Mulvihill, S.J. Factors Associated with Improved Survival after Resection of Pancreatic Adenocarcinoma. Anesthesiology 2015, 122, 317–324. [Google Scholar] [CrossRef]
  99. Soliz, J.M.; Ifeanyi, I.C.; Katz, M.H.; Wilks, J.; Cata, J.P.; McHugh, T.; Fleming, J.B.; Feng, L.; Rahlfs, T.; Bruno, M.; et al. Comparing Postoperative Complications and Inflammatory Markers Using Total Intravenous Anesthesia Versus Volatile Gas Anesthesia for Pancreatic Cancer Surgery. Anesthesiol. Pain Med. 2017, 7, e13879. [Google Scholar] [CrossRef]
  100. Zhang, Y.-L.; Pei, L.-J.; Sun, C.; Zhao, M.-Y.; Che, L.; Huang, Y.-G. Regional anesthesia and cancer recurrence in patients with late-stage cancer: A systematic review and meta-analysis. Chin. Med J. 2021, 134, 2403–2411. [Google Scholar] [CrossRef]
  101. Pusztai, L.; Hess, K.R. Clinical trial design for microarray predictive marker discovery and assessment. Ann. Oncol. 2004, 15, 1731–1737. [Google Scholar] [CrossRef] [PubMed]
  102. Cardoso, F.; Piccart-Gebhart, M.; van’t Veer, L.; Rutgers, E. The MINDACT trial: The first prospective clinical validation of a genomic tool. Mol. Oncol. 2007, 1, 246–251. [Google Scholar] [CrossRef] [PubMed]
  103. Vassy, J.L.; Christensen, K.; Schonman, E.F.; Blout, C.L.; Robinson, J.O.; Krier, J.B.; Diamond, P.M.; Lebo, M.; Machini, K.; Azzariti, D.R.; et al. The Impact of Whole-Genome Sequencing on the Primary Care and Outcomes of Healthy Adult Patients: A Pilot Randomized Trial. Ann. Intern. Med. 2017, 167, 159–169. [Google Scholar] [CrossRef]
  104. Dubowitz, J.; Hiller, J.; Riedel, B. Anesthetic technique and cancer surgery outcomes. Curr. Opin. Anaesthesiol. 2021, 34, 317–325. [Google Scholar] [CrossRef]
  105. Buggy, D.; Freeman, J.; Johnson, M.; Leslie, K.; Riedel, B.; Sessler, D.; Kurz, A.; Gottumukkala, V.; Short, T.; Pace, N.; et al. Systematic review and consensus definitions for standardised endpoints in perioperative medicine: Postoperative cancer outcomes. Br. J. Anaesth. 2018, 121, 38–44. [Google Scholar] [CrossRef] [PubMed]
  106. Pérez-Aguado, G.; de la Mata, D.M.-A.; Valenciano, C.M.-L.; Sainz, I.F.-U. Endoscopic ultrasonography-guided celiac plexus neurolysis in patients with unresectable pancreatic cancer: An update. World J. Gastrointest. Endosc. 2021, 13, 460–472. [Google Scholar] [CrossRef] [PubMed]
  107. Citterio, D.; Pusceddu, S.; Facciorusso, A.; Coppa, J.; Milione, M.; Buzzoni, R.; Bongini, M.; Debraud, F.; Mazzaferro, V. Primary tumour resection may improve survival in functional well-differentiated neuroendocrine tumours metastatic to the liver. Eur. J. Surg. Oncol. (EJSO) 2016, 43, 380–387. [Google Scholar] [CrossRef]
  108. Cardoso, T.A.A.M.; Kunst, G.; Neto, C.N.; Júnior, J.D.R.C.; Silva, C.G.S.; Bastos, G.M.; Borges, J.B.; Hirata, M.H. Effect of sevoflurane on the inflammatory response during cardiopulmonary bypass in cardiac surgery: The study protocol for a randomized controlled trial. Trials 2021, 22, 25. [Google Scholar] [CrossRef]
  109. Yang, Z.; Zhang, M.; Wang, G.; Wei, P.; Gao, S. Identification of anesthetic-induced expression changes using DNA microarray. Mol. Med. Rep. 2014, 11, 589–596. [Google Scholar] [CrossRef]
  110. Fokas, E.; Engenhart-Cabillic, R.; Daniilidis, K.; Rose, F.; An, H.-X. Metastasis: The seed and soil theory gains identity. Cancer Metastasis Rev. 2007, 26, 705–715. [Google Scholar] [CrossRef]
  111. Mehlen, P.; Puisieux, A. Metastasis: A question of life or death. Nat. Cancer 2006, 6, 449–458. [Google Scholar] [CrossRef]
  112. Nepogodiev, D.; Martin, J.; Biccard, B.; Makupe, A.; Bhangu, A.; Ademuyiwa, A.; Adisa, A.O.; Aguilera, M.-L.; Chakrabortee, S.; Fitzgerald, J.; et al. Global burden of postoperative death. Lancet 2019, 393, 401. [Google Scholar] [CrossRef]
  113. Lucas, A.L.; Malvezzi, M.; Carioli, G.; Negri, E.; La Vecchia, C.; Boffetta, P.; Bosetti, C. Global Trends in Pancreatic Cancer Mortality From 1980 Through 2013 and Predictions for 2017. Clin. Gastroenterol. Hepatol. 2016, 14, 1452–1462.e4. [Google Scholar] [CrossRef] [PubMed]
  114. Lomberk, G.; Dusetti, N.; Iovanna, J.; Urrutia, R. Emerging epigenomic landscapes of pancreatic cancer in the era of precision medicine. Nat. Commun. 2019, 10, 3875. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the effect of anesthesia on epigenetics and cancer cells.
Figure 1. Schematic representation of the effect of anesthesia on epigenetics and cancer cells.
Medicina 58 01102 g001
Table 1. Ongoing trials on pancreatic cancer and recurrence-related outcomes with repurposed anesthetics.
Table 1. Ongoing trials on pancreatic cancer and recurrence-related outcomes with repurposed anesthetics.
IdentifierStatusInterventionTypeConditionOutcomeTitle
NCT02335151CDesflurane
Propofol
Randomized
Parallel assignment
Double blind
86 patients
Adenocarcinoma
Circulating Tumor Cells
Pancreatic Neoplasms
Surgery
Peak of CTC in the postoperative phase after curative tumor removal
Kinetics of CTC after surgery up to day 7
Month to Tumor recurrence
Number of surviving patients
CTC Pancreatic Adenocarcinoma
NCT04048278RIntravenous (IV) lidocaine
Saline Solution for Injection
Randomized
Parallel Assignment
Double blind
46 patients
Pancreatic Cancer
Surgery
Enzymatic activity of SCR TK on CTC
Cytokine levels
Chemokine levels
Gene expression
CTC number
Lidocaine Infusion in Pancreatic Cancer
NCT04449289NYRIntravenous (IV) lidocaine
Epidural ropivacaine
Randomized
Parallel Assignment
Open label
100 patients
Pancreatic Cancer
Surgery
1- and 3-years recurrence rate after surgery
Lidocaine and ropivacaine concentration
Complication rate after surgery
Influence of Local Anesthetic Administration on the Cancer Recurrence Rate After Pancreatic Oncologic Surgery
NCT03245346RGEA
PCEA
GA
PCIA
Randomized
Parallel Assignment
Open label
540 patients
Cancer of Pancreas
Surgery
Overall survival (OS)
Disease-free survival (DFS)
Postoperative pain score and side effects of patient-controlled
Incidence of delirium
Incidence of persistent post-surgical pain (PPSP) after surgery
Length of stay in hospital after surgery and total costs after surgery
Return of bowel function
Start of enteral tube feeding
Removal of Perianastomotic drains
Removal of Urinary drainage
Removal of nasogastric tube
Removal of enteral feeding tube
Blood level of neuroendocrine, stress and inflammatory response (blood epinephrine, norepinephrine, cortisol, VEGF, interleukin-6 (IL-6), interleukin-8 (IL-8), peripheral blood NLR (neutrophil-lymphocyte ratio))

Serum CA19-9, CA125, CEA, CA72-4, CA242, AFP, CA15-3, CA50 levels
Plasma levels of ropivacaine and sufentanil
Effects of Epidural Anesthesia and Analgesia on the Prognosis in Patients Undergoing Pancreatic Cancer Surgery
NCT04025840REpidural Block Dexamethasone
Randomized
Intervention
Factorial Assignment
Intervention Model
Single blind
260 patients
Pancreatic Cancer
Surgery
2-year overall survival
Postoperative gastrointestinal complications
Overall postoperative complications
Length of stay in hospital after surgery
All-cause 30-day mortality
Quality of life in 1- and 2-year survivors
Hospital readmission within 2 years after surgery
2-year progression-free survival
Subjective sleep quality:
Pain severity
Time to ambulation after surgery
Time to oral intake after surgery
Perioperative Epidural Block and Dexamethasone in Pancreatic Cancer Surgery
NCT03447691RDesflurane
Total Intravenous Anesthesia with Propofol and Remifentanil
Randomized
Parallel Assignment
Triple blind
132 patients
Pancreatic Cancer
Distal CBD Cancer
Surgery
Score of QoR40
Score of QoR40
Comparison Between Volatile Anesthetic-desflurane and Total Intravenous Anesthesia With Propofol and Remifentanil on Early Recovery Quality and Long Term Prognosis of Patients Undergoing Pancreatic Cancer and Common Bile Duct Cancer Surgery
NCT03838029RPropranolol
Etodolac
Other: Placebo
Randomized
Intervention Parallel Assignment
Quadruple blind
210 patients
Pancreatic Neoplasms
Surgery
Rate of cancer recurrence
Biomarkers in extracted tumor tissue samples
Number of patients with treatment related adverse events
Depression, Anxiety, Global distress
Fatigue
Perioperative Intervention to Reduce Metastatic Processes in Pancreatic Cancer Patients Undergoing Curative Surgery (BC-PC)
NCT03034096RPropofol
Volatile Agent (sevoflurane, isoflurane, or desflurane)
Randomized
Parallel assignment
Double blind
2000 patients
Patients with known or suspected cancer and scheduled to undergo any of the following oncologic surgical procedures:

Lobectomy or pneumonectomy
Esophagectomy
Radical (total) cystectomy
Pancreatectomy
Partial hepatectomy
Hyperthermic intraperitoneal chemotherapy (HIPEC)
Gastrectomy (subtotal or total)
Cholecystectomy or bile duct resection
All-cause mortality (Time Frame: 2 year minimum)
Time to event
Secondary Outcome Measures:
Recurrence free survival (Time Frame: Minimum 2 years)
Time to event
All-cause mortality as a binary outcome (Time Frame: 2 years)
General Anesthetics in CAncer REsection Surgery (GA-CARES) Trial (GA-CARES)
NCT02660411CSevoflurane
Propofol
Randomized
Parallel Assignment
Triple blind
1228 patients
Age ≥ 65 years and < 90 years;
Primary malignant tumor;
Do not receive radiation therapy or chemotherapy before surgery;
Scheduled to undergo surgery for the treatment of tumors, with an expected duration of 2 h or more, under general anesthesia;
Over survival after surgery. (Time Frame: Up to 5 years after surgery.)
Time from surgery to the date of all-cause death.
Secondary Outcome Measures:
Recurrence-free survival after surgery (Time Frame: Up to 5 years after surgery)
Time from surgery to the date of cancer recurrence/metastasis or all-cause death, whichever occurs first.
Event-free survival after surgery (Time Frame: Up to 5 years after surgery)
Time from surgery to the date of cancer recurrence/metastasis, new cancer, new serious non-cancer disease, or all-cause death, whichever occurs first.
Quality of life in 3-year survivors after surgery. (Time Frame: Assessed at the end of the 3rd year after surgery.)
Quality of life is assessed with the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-C30).
Cognitive function in 3-year survivors after surgery. (Time Frame: Assessed at the end of the 3rd year after surgery.)
Cognitive function is assessed with the Telephone Interview for Cognitive Status-Modified (TICS-m).
Impact of Inhalational Versus Intravenous Anesthesia Maintenance Methods on Long-term Survival in Elderly Patients After Cancer Surgery: a Randomized Controlled Trial
NCT03838029RPropranolol
Etodolac
Placebo
Randomized
Parallel Assignment
Quadruple blind
210 patients
Pancreatic Neoplasms
Primary:
-Rate of cancer recurrence (Time Frame: From the date of surgery until malignant disease is identified, assessed up to 60 months post-surgery)
-Data regarding post-surgical recurrence will be recorded at 1, 3, 6, 12, 18, 24, 36, 48, and 60 following surgery
-Biomarkers in extracted tumor tissue samples (Time Frame: An average of one year following surgery)
Epithelial-to-mesenchymal-transition (EMT) status and natural-killer cell, macrophage, T-cell, and B-cell infiltration levels into tumor tissue (as assessed by messenger RNA profiling of tissue samples.
-Biomarkers in blood samples (Time Frame: An average of one year following surgery) Cytokine levels in blood samples (interleukin-6, interleukin-10, C-reactive protein, interferon-gamma, and vascular endothelial growth factor and additional exploratory analysis of other cytokines)
Perioperative Intervention to Reduce Metastatic Processes in Pancreatic Cancer Patients Undergoing Curative Surgery
NCT05451043NYRBiological: Durvalumab
Drug: Gemcitabine
Drug: Nab paclitaxel
Biological: Tremelimumab
Drug: Propranolol
Drug: Cisplatin
Single group
Single Group Assignment
Open label
62 patients
Pancreatic Cancer
Hepatocellular Cancer
Biliary Tract Cancer
Cholangiocarcinoma
Primary:
-Investigating and establishing the efficacy of propranolol in boosting the effects of immunotherapy in pancreatic adenocarcinoma (Time Frame: Assessed one year after enrollment of last participant) combination of gemcitabine+nab-paclitaxel+propranolol+durvalumab+tremelimumab’s objective response rate is greater than or equal to 50%
-Investigating and establishing the efficacy of propranolol in boosting the effects of immunotherapy in hepatocellular carcinoma (Time Frame: Assessed one year after enrollment of last participant) propranolol + durvalumab + tremelimumab objective response rate is greater than 45%
Investigating and establishing the efficacy of propranolol in boosting the effects of immunotherapy in biliary tract tumors (Time Frame: Assessed one year after enrollment of last participant)
-To demonstrate in unresectable BTC (gallbladder, cholangiocarcinoma of the biliary tracts including ampullary carcinomas) that the combination of gemcitabine + cisplatin + propranolol + durvalumab + tremelimumab’s objective response rate is greater than 50%.
Durvalumab and Tremelimumab in Combination With Propranolol and Chemotherapy for Treatment of Advanced Hepato-pancreabiliary Tumors (BLOCKED)
EudraCT number: 2018-000415-25 Propranolol
Etodolac
Placebo
Randomized
Parallel Assignment
Single blind
100 patients
Non-metastatic head PC of the head undergoing elective pancreatoduodenectomy.
Primary:
serious adverse events and reactions within 3 months.
Secondary: utility of the two drugs in
improving survival.
Pancreatic resection with perioperative drug repurposing of propranolol and etodolac: trial protocol of the phase-II randomized placebo controlled PROSPER trial.
R: recruiting; NYR: non yet recruiting; C: completed; CTC: Circulating Tumor Cells.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mokini, Z.; Cama, A.; Forget, P. Anesthetics and Long Term Cancer Outcomes: May Epigenetics Be the Key for Pancreatic Cancer? Medicina 2022, 58, 1102. https://doi.org/10.3390/medicina58081102

AMA Style

Mokini Z, Cama A, Forget P. Anesthetics and Long Term Cancer Outcomes: May Epigenetics Be the Key for Pancreatic Cancer? Medicina. 2022; 58(8):1102. https://doi.org/10.3390/medicina58081102

Chicago/Turabian Style

Mokini, Zhirajr, Alessandro Cama, and Patrice Forget. 2022. "Anesthetics and Long Term Cancer Outcomes: May Epigenetics Be the Key for Pancreatic Cancer?" Medicina 58, no. 8: 1102. https://doi.org/10.3390/medicina58081102

Article Metrics

Back to TopTop