Acessibilidade / Reportar erro

Lidocaine in oncological surgery: the role of blocking in voltage-gated sodium channels. A narrative review

Abstract

Background:

The current evidence suggests that oncological surgery, which is a therapy used in the treatment of solid tumors, increases the risk of metastasis. In this regard, a wide range of tumor cells express Voltage-Gated Sodium Channels (VGSC), whose biological roles are not related to the generation of action potentials. In epithelial tumor cells, VGSC are part of cellular structures named invadopodia, involved in cell proliferation, migration, and metastasis. Recent studies showed that lidocaine could decrease cancer recurrence through its direct effects on tumor cells and immunomodulatory properties on the stress response.

Objective:

The aim of this narrative review is to highlight the role of VGSC in tumor cells, and to describe the potential antiproliferative effect of lidocaine during the pathogenesis of metastasis.

Contents:

A critical review of literature from April 2017 to April 2019 was performed. Articles found on PubMed (2000–2019) were considered. A free text and MeSH-lidocaine; voltage-gated sodium channels; tumor cells; invadopodia; surgical stress; cell proliferation; metastasis; cancer recurrence – for articles in English, Spanish and Portuguese language – was used. A total of 62 were selected.

Conclusion:

In animal studies, lidocaine acts by blocking VGSC and other receptors, decreasing migration, invasion, and metastasis. These studies need to be replicated in humans in the context of oncological surgery.

KEYWORDS
Lidocaine; Voltage-gated sodium channels; Tumor cells; Invadopodia; Surgical stress; Cell proliferation; Metastasis; Cancer recurrence

Resumo

Justificativa:

As evidências atuais sugerem que a cirurgia oncológica, usada no tratamento de tumores sólidos, aumenta o risco de metástase. Nesse sentido, uma ampla gama de células tumorais expressa Canais de Sódio Dependentes de Voltagem (CSDV), cujos papéis biológicos não estão relacionados à produção de potencial de ação. Nas células epiteliais tumorais, o CSDV é parte integrante de estruturas celulares denominadas invadópodes, que participam da proliferação, migração e metástase celular. Estudos recentes mostraram que a lidocaína pode diminuir a recorrência do câncer através de efeitos diretos nas células tumorais e de propriedades imunomoduladoras na resposta ao estresse.

Objetivo:

O objetivo desta revisão narrativa é analisar o papel do CSDV nas células tumorais e descrever o possível efeito antiproliferativo da lidocaína na patogênese das metástases.

Conteúdo:

Foi realizada uma revisão crítica da literatura de Abril de 2017 a Abril de 2019. Os artigos encontrados no PubMed (2000 − 2019) foram analisados. Pesquisamos textos de linguagem livre e descritores MeSH-lidocaína; canais de sódio dependentes de voltagem; células tumorais; invadópodes; estresse cirúrgico; proliferação celular; metástase; recorrência do câncer − em artigos publicados em inglês, espanhol e português. Foram selecionadas 62 publicações.

Conclusão:

Em estudos empregando animais, a lidocaína atua bloqueando o CSDV e outros receptores, diminuindo a migração, invasão e metástase. Esses estudos precisam ser replicados em humanos submetidos a cirurgia oncológica.

PALAVRAS-CHAVE
Lidocaína; Canais de sódio dependentes de voltagem; Células tumorais; Invadópodes; Estresse cirúrgico; Proliferação celular; Metástase; Recorrência do câncer

Introduction

Despite the advances in cancer therapies, cancer remains to be a major cause of morbidity and mortality globally.11 Jemal A, Bray F, Center MM, et al. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90. The current evidence suggests that oncological surgery, which is a therapy used in the treatment of solid tumors, increases the risk of metastasis.22 Exadaktylos AK, Buggy DJ, Moriarty DC, et al. Can anesthetic technique for primary breast cancer surgery affect recurrence or metastasis?. Anesthesiology. 2006;105:660-4.44 Heaney A, Buggy DJ. Can anaesthetic and analgesic techniquesaffect cancer recurrence or metastasis? Br J Anaesth. 2012;109.S1:i17-i28. The surgical stress causes perioperative immunosuppression and a release of angiogenic factors which enables the spread of Circulating Tumor Cells (CTC) and the subsequent distance reimplantation.55 Kim J. Effects of surgery and anesthetic choice on immunosuppression and cancer recurrence. J Transl Med. 2018;16:8. The tumor cell escape, in combination with the impairment immunity in perioperative time, increases the susceptibility to recurrences, which is considered the “perfect storm” for cancer progression.66 Lee BM, Cata JP. Impact of anesthesia on cancer recurrence. Rev Esp Anestesiol Reanim. 2015;62:570-5.88 McCausland K, Martin N, Missair A. Anaesthetic technique and cancer recurrence: current understanding. OA Anaesth. 2014;18:1-7. In this regard, a wide range of tumor cells express Voltage-Gated Sodium Channels (VGSC), whose biological roles are not related to the generation of action potentials.99 Besson P, Driffort V, Bon E, et al. How do voltage. gated sodium channels enhance migration and invasiveness in cancer cells?. Biochim Biophys. 2015;1848:2493-501.,1010 Brackenbury WJ. Voltage-gated sodium channels and metastatic disease. Channels (Austin). 2012;6:352-61. In epithelial tumor cells, VGSC are part of cellular structures named invadopodia, involved in cell proliferation, migration, and metastasis.1010 Brackenbury WJ. Voltage-gated sodium channels and metastatic disease. Channels (Austin). 2012;6:352-61.,1111 Fife M, McCarroll JA, Kavallaris M. Movers and shakers: cell cytoskeleton in cancer metastasis. Br J Pharm. 2014;171:5507-23. Recent studies showed that lidocaine could decrease cancer recurrence through its direct effects on tumor cells and immunomodulatory properties on stress response.1212 Sekandarzad MW, Van Zundert AAJ, Lirk PB, et al. Perioperative anesthesia care and tumor progression. Anesth Analg. 2017;124:1697-708.,1313 Weinberg L, Peake B, Tan C, et al. Pharmacokinetics and pharmacodynamics of lignocaine: a review. World J Anesthesiol. 2015;4:17-29. Lidocaine acts by blocking VGSC and other receptors, and it is used in different scenarios in anesthetic practice, such as local infiltration, regional blocks (epidural, spinal, peripheral nerves), and intravenous infusion in the context of multimodal analgesia.1414 Soto G, Naranjo Gonzalez M, Calero F. Intravenous lidocaine infusion. Rev Esp Anestesiol Reanim. 2018;65:269-74. The aim of this narrative review is to highlight the role of VGSC in tumor cells and to describe the potential antiproliferative effect of lidocaine during the pathogenesis of metastasis.

Methodology

A critical review of the literature from April 2017 to April 2019 was performed. Articles found on PubMed (2000–2019) were considered. A free text and MeSH-lidocaine; voltage-gated sodium channels; tumor cells; invadopodia; surgical stress; cell proliferation; metastasis; cancer recurrence – for articles in English, Spanish and Portuguese language – was used. Additional studies from bibliographies of retrieved trials and previous reviews were recruited. Abstracts, case reports, and letters were excluded. Of the 156 articles screened, 96 were excluded. A total of 62 were selected, and these were reviewed with the propose to describe the role of VGSC in tumor cells and the different effects of lidocaine in cancer cells. In addition, we describe the clinical application of intravenous lidocaine.

Contents

Mechanisms of tumoral progression and metastasis

Both surgery and anesthesia stimulate the hypothalamic-pituitary axis and sympathetic nervous system, whose activation suppresses Cell-Mediated Immunity (CMI), releases catecholamines and prostaglandins, inducing perioperative immunosuppression.55 Kim J. Effects of surgery and anesthetic choice on immunosuppression and cancer recurrence. J Transl Med. 2018;16:8. Some of the mediators released during the inflammatory process included Interleukin 6 (IL-6), Interleukin-1β (IL-1β), Tumor Necrosis Factor-α (TNF-α), angiogenic factors such as Vascular Endothelial Growth Factor (VEGF), reactive oxygen species, hypoxia-inducible factor-α and β (HIF-1α and HIF-2α), and factor NF-kB, which are directly or indirectly involved in the survival of tumor cells. The immune system plays a pivotal role in clearing new forming malignant cells and it does so by favoring anti-tumor mechanisms such as the production of cytokines. Th1 cells release IFN-γ, IL-2, and TNF-α; in contrast, Th2 cells secrete IL-4, IL-5, IL-10, and IL-13 cytokines. A shift towards Th1 polarization is the expected response against cancer. In contrast, surgical stress, volatile anesthetics, opioids, and blood transfusions are known to favor a Th2 response that manifests as immune suppression.1515 Ramírez MF, Huitink JM, Cata JP. Perioperative clinical interventions that modify the immune response in cancer patients. Op J Anesth. 2013;3:133-9. In this way, oncological surgery increases the risk of tumor dissemination, a complex process that involves the detachment of cancer cells from the primary tumor, local invasion, and their subsequent seeding in distant organs.1616 Missair A, Cata JP, Votta-Velis G, et al. Impact of perioperative pain management on cancer recurrence: an ASRA/ESRA special article. Reg Anesth Pain Med. 2019;44:13-28.,1717 Cassinello F, Prieto I, del Olmo M, et al. Cancer surgery: how may anesthesia influence outcome?. J Clin Anesth. 2015;27:262-72. CTC carried in blood or lymph can reach distant tissues.1818 Gupta GP, Massague J. Cancer metastasis: building a framework. Cell. 2006;127:679-95. Cancer progression is linked to a subset of tumor cells known as “cancer stem cells” residing in a specific environment within the tumor microenvironment called the cancer stem cell niche. The niche plays a major role in maintaining cancer stem cell activity, protects the cell from the host defense mechanisms and facilitates metastasis.1919 Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147:992-1009. The prevalence of escape mechanisms is greater in metastatic niche than in the primary tumor, indicating a higher selection pressure during the metastatic process. Taken together, the risk factors described above, which are all common in oncological surgery, occur simultaneously during the perioperative period.

Voltage-Gated Sodium Channels (VGSC)

VGSCs are typical of excitable cells, which are compound of one α subunit (Nav1.1-Nav1.9) and one or more β subunits (β1–β4).2020 Catterall WA. From ionic currents to molecular mechanisms: the structure and function of voltage-gated sodium channels. Neuron. 2000;26:13-25. The α subunit is an integral heteromultimeric protein complex consisting of four homologous domains (D1–D4), each containing six α-helical transmembrane segments (S1–S6).2121 Catterall WA. Voltage-gated sodium channel: structure, function, and pathophysiology. J Physiol. 2012;590:2577-89. Both terminus, C- and N-terminal, and ligand binding domains are intra cytoplasmic. S5 and S6 segments and a P-loop in each domain form the channel pore, penetrating the interior of the membrane. Seven of the voltage-gated sodium channels (Nav1.1–Nav1.3 and Nav1.6–Nav1.9) play major roles in electrogenesis in neurons, whereas Nav1.4 is the muscle sodium channel and Nav1.5 is the predominant cardiac myocyte channel. The canonical role of sodium channels in impulse electrogenesis and conduction in these excitable cells has been well established and is relatively well understood.2222 Black JA, Waxman SG. Non-canonical roles of voltage-gated sodium channels. Neuron. 2013;80:280-91. Lidocaine penetrates the neuronal membrane becoming in its not ionized form by the pH effect, joining the S6 portion of domain 4 α-subunit within the sodium channel. The analgesic and antihyperalgesic properties of lidocaine have been proven in vivo and in vitro studies.1212 Sekandarzad MW, Van Zundert AAJ, Lirk PB, et al. Perioperative anesthesia care and tumor progression. Anesth Analg. 2017;124:1697-708. The current evidence shows that VGSC are expressed in a wide range of non-excitable cells, such as astrocytes, oligodendrocytes, immune cells, dendritic cells, endothelial cells, macrophages, osteoblasts, odontoblasts, keratinocytes, and tumor cells.2222 Black JA, Waxman SG. Non-canonical roles of voltage-gated sodium channels. Neuron. 2013;80:280-91. In non-excitable cells, the VGSC function is different from the generation of action potential since they participate in cell survival, differentiation, proliferation, migration, endosome acidification, and phagocytosis. All these functions are known as non-canonical functions of VGSC.2222 Black JA, Waxman SG. Non-canonical roles of voltage-gated sodium channels. Neuron. 2013;80:280-91.,2323 Roger S, Gillet L, LeGuennec JY, et al. Voltage-gated sodium channels and cancer: is excitability their primary role?. Front Pharmacol. 2015;6:152.

VGSC in tumor cells

Tumor cells use VGSC for migrating, invading and metastasizing.2424 Lambert AW, Pattabiraman DR, Weinberg RA. Emerging biological principles of metastasis. Cell. 2017;168:670-91. The Epithelial-Mesenchymal Transition (EMT) program is a developmental cell-biological program in which tumor cell lose its epithelial morphology and turns into mesenchymal, acquiring invasiveness.2525 Jobe N, Rösel D, Tolde O, et al. Complex 3D models to study drug targeting of invadopodias. Clin Cancer Drugs. 2014;1:85-9.2727 Eckert MA, Yang J. Targeting invadopodia to block breast cancer metastasis. Oncotarget. 2011;2:562-8. In the course of this process, tumor cells degrade the extracellular matrix (ECM) through invasive protrusions referred to as invadopodia2525 Jobe N, Rösel D, Tolde O, et al. Complex 3D models to study drug targeting of invadopodias. Clin Cancer Drugs. 2014;1:85-9. (Fig. 1). The role of invadopodia in the ECM degradation is to facilitate the invasion of adjacent tissues.2626 Linder S. The matrix corroded: podosomes and invadopodias in extracellular matrix degradation. Trends Cell Biol. 2007;17:107-17. VGSCs are precisely located in invadopodia along with the Na+/H+ type 1 exchanger pump (NHE1) and the Na+/Ca++ exchanger pump (NXC).2828 Brisson L, Driffort V, Benoist L, et al. Nav1.5 Na+ channels allosterically regulate the NHE-1 exchanger and promote the activity of breast cancer cell invadopodia. J Cell Sci. 2013;126:4835-42. In a meticulously orchestrated process, the NHE1 releases H+ by acidifying the medium while the NXC increases the intracellular Ca++ concentration. This leads to the acidification of the peri-invadopodia, cathepsins secreted by tumor cells, and the consistent degradation of ECM. Furthermore, VGSC action sustains enzyme Src kinase activity, promoting the polymerization of actin filaments. These results suggest that VGSC activity in cancer cells enhances both the formation and ECM degradative activity of invadopodia. One of the most VGSC studied in breast cancer in its neonatal splice variant is the VGSC 1.5.2929 Brackenbury WJ, Chioni AM, Diss JK, et al. The neonatal splice variant of Nav1.5 potentiates in vitro invasive behavior of MDA-MB-231 human breast cancer cells. Breast Cancer Res Treat. 2007;101:149-60. In vitro studies have shown that VGSC 1.5 block with shRNA reduces cell invasion. On the contrary, VGSC activation with veratridine increases the ECM invasion, showing the importance of these channels during the invasion process. In vitro, VGSC activation has shown to increase the metastatic power due to greater motility and invasion. Therefore, tumor cells behave as “electrically excitable cells”, becoming hyperactive and gaining aggressiveness, an event that is called CELEX, which represents a hypothesis of metastatic tumor progression.3030 Djamgoz MBA. Biophysics of cancer: cellular excitability (“CELEX”) hypothesis of metastasis. J Clin Exp Oncol. 2014.S1:005. Several drugs that block the VGSC are being investigated due to their antiproliferative actions on tumor cells, including phenytoin, carbamazepine, divalproex sodium, lamotrigine, gabapentin, and lidocaine.3131 Martin F, Ufodiama C, Watt I, et al. Therapeutic value of voltage-gated sodium channel inhibitors in breast, colorectal, and prostate cancer: a systematic review. Front Pharmacol. 2015;6:273.,3232 Koltai T. Voltage-gated sodium channel as a target for metastatic risk reduction with re-purposed drugs. F1000Res. 2015;4:297. Taken together, these results suggest that VGSCs and Src kinase have a critical role, both in invadopodia formation and in proteolytic activity, promoting the mesenchymal invasion. Both migration and invasion occur during the early stage of metastasis.

Figure 1
Voltage Gate-Sodium Channels (VGSC) in invadopodia of cancer cell. Blocking by lidocaine. The Epithelial-Mesenchymal Transition (EMT) program is a developmental cell-biological program in which tumor cell lose its epithelial morphology and turns into mesenchymal, acquiring invasiveness. VGSCs are precisely located in invadopodia. VGSC action sustains enzyme Src kinase activity, promoting the polymerization of actin filaments. VGSC activity enhances both the formation and ECM degradative activity of invadopodia. Adapted from Roger et al. Front Pharm, 2015.

Direct effects of lidocaine on tumor cells

Lidocaine and other local anesthetics might have direct effects on tumor cells (Table 1). Tumor cells express VGSCs in a wide range of carcinomas, including breast, cervix, bowel, lung (small-cell, non-small-cell, and mesothelioma cancer), skin, ovarian and prostate cancer.2121 Catterall WA. Voltage-gated sodium channel: structure, function, and pathophysiology. J Physiol. 2012;590:2577-89. In vitro blocked of the neonatal splice variants in VGSC (Nvgsc 1.5) expressed in highly metastatic breast cancer cell lines (MDA-MB-231) was obtained with lidocaine, at clinical concentration.3333 Fraser SP, Diss JK, Chioni AM, et al. Voltage-gated sodium channel expression and potentiation of human breast cancer metastasis. Clin Cancer Res. 2005;11:5381-9. In vitro and in vivo studies show that lidocaine inhibits the migration and reduces the viability in all breast cancer cell lines, including high malignant cells such as triple-negative and HER2 positive.3434 Chamaraux-Tran TN, Mathelin C, Aprahamian M, 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. In this regard, Fraser et al. suggest that intra- and postoperative local anesthetics may reduce the ability of tumor cells to metastasize by blocking VGSC and thus inhibiting their motility and invasiveness.3535 Fraser S, Foo I, Djamgoz M. Local anaesthetic use in cancer surgery and disease recurrence: role of voltage-gated sodium channels?. Br J Anaesth. 2014;113:899-902. Independent actions of block of VGSC have been evidenced as possible antiproliferative properties. In vitro studies have shown that exposure to lidocaine inhibits Src protein tyrosine kinase in tumor cells, a protein involved in proliferation, migration, invasiveness and tumor metastasis.3636 Piegeler T, Votta-Velis EG, Liu G, et al. Antimetastatic potential of amide-linked local anesthetics: inhibition of lung adenocarcinoma cell migration and inflammatory Src signaling independent of sodium channel blockade. Anesthesiology. 2012;117:548-59. Moreover, it has been shown that both lidocaine and ropivacaine inhibit tumor cells the expression of Intercellular Adhesion Molecules (ICAM-1), which are synthesized at once in the metastasis process. Another antiproliferative effect of lidocaine is the induction of apoptosis in a culture of different tumor cells.3737 Chang Y, Liu CH, Chen M, et al. Local anesthetics induce apoptosis in human breast tumor cells. Anesth Analg. 2014;118:116-24.3939 Xing W, Chen DT, Pan JH, et al. Lidocaine induces apoptosis and suppresses tumor growth in human hepatocellular carcinoma cells in vitro and in a xenograft model in vivo. Anesthesiology. 2017;126:868-81. A research conducted by Lirk et al. showed that lidocaine interferes with the process of genetic regulation by altering DNA methylation in cancer cell cultures. Methylation is catalyzed by the enzyme DNA Methyltransferase (DNMT1), whose role is crucial in the cancer pathogenesis inhibiting the tumor growth. It is speculated that lidocaine might alter the methylation by inhibiting DNMT1. This was an additive effect in chemotherapy that was also shown using ropivacaine but not bupivacaine.4040 Lirk P, Berger R, Hollmann MW, et al. Lidocaine time- and dose-dependently demethylates deoxyribonucleic acid in breast cancer cell lines in vitro. Br J Anaesth. 2012;109:200-7. It is important to mention that opioids have the opposite effect, being involved in DNA hypermethylation.4141 Doehring A, Oertel BG, Sittl R, et al. Chronic opioid use is associated with increased DNA methylation correlating with increased clinical pain. Pain. 2013;154:15-23. Also, lidocaine would act by modulating the receptor for Epidermal Growth Factor (EGFR), impeding the mobility of tumor cells.4242 Sakaguchi M, Kuroda Y, Hirose M. The antiproliferative effect of lidocaine on human tongue cancer cells with inhibition of the activity of epidermal growth factor receptor. Anesth Analg. 2006;102:1103-7.,4343 Mammoto T, Higashiyama S, Mukai M, et al. Infiltration anesthetic lidocaine inhibits cancer cell invasion by modulating ectodomain shedding of heparin-binding epidermal growth factor-like growth factor (HB-EGF). J Cell Physiol. 2002;192:351-8. Eventually, a recent evidence has shown that tumor cells use Ca++ to mobilize through transient receptors type V6 (Transient Receptor Potential Cation Channel Subfamily V Member 6, TRPV-6). Lidocaine blocks TRPV-6 channels, decreasing intracellular calcium levels and thus inhibiting migration and invasion in cancer cells.4444 Jiang Y, Gou H, Zhu J, et al. Lidocaine inhibits the invasion and migration of TRPV6-expressing cancer cells by TRPV6 down-regulation. Oncol Lett. 2016;12:1164-70. These results, from different studies, have shown a multimolecular action of lidocaine on tumor cells, which could reduce the metastasis risk.4545 Piegeler T, Hollmann MW, Borgeat A, et al. Do amide local anesthetics play a therapeutic role in the perioperative management of cancer patients?. Int Anesthesiol Clin. 2016;54(4):e17-32.

Table 1
Direct effects of lidocaine on tumor cells.

Immunomodulatory effects of lidocaine

In the perioperative period, surgery induces immunosuppression, playing a critical role in the setting and growing of metastatic lesions.55 Kim J. Effects of surgery and anesthetic choice on immunosuppression and cancer recurrence. J Transl Med. 2018;16:8. Lidocaine has been shown to have analgesic, anti-inflammatory, and immunomodulatory properties, reducing neuroendocrine response caused by surgical stress.4646 Dunn LK, Durieux ME. Perioperative use of intravenous lidocaine. Anesthesiology. 2017;126:729-37. These actions involve the blocking of several channels, including VGSC, K+ channels, Ca++ channels, glycinergic system, G Protein-Coupled Receptors (GPCR) and N-Methyl-D-Aspartate receptors (NMDA).4747 Weinberg L, Peake B, Tan C, et al. Pharmacokinetics and pharmacodynamics of lignocaine: A review. World J Anesthesiol. 2015;4:17-29. The analgesic effect of intravenous administration is the result of increased acetylcholine levels in the cerebrospinal fluid, which causes downward inhibition, inhibition of glycine receptors, and increase the release of endogenous opioids. When lidocaine reaches the spinal cord, it reduces the post-synaptic depolarization mediated by NMDA and neurokinin receptors, thus modifying the pain response. NMDA blockade inhibits protein kinase C, thus reducing hyperalgesia and postoperative opioid tolerance. In animal models, lidocaine acts during the early stages of systemic inflammatory response, modulating the marginalization, adherence, and diapedesis of polymorphonuclear cells towards the site of the lesion, thus inhibiting the production of reactive oxygen species and the release of histamine. This immunomodulatory effect of the drug is achieved by blocking GPCR receptors since polymorphonuclear cells do not contain VGSCs. From an oncological point of view, lidocaine preserves the endothelial barrier function and cytotoxic function of NK cells.4444 Jiang Y, Gou H, Zhu J, et al. Lidocaine inhibits the invasion and migration of TRPV6-expressing cancer cells by TRPV6 down-regulation. Oncol Lett. 2016;12:1164-70. Concerning to endothelial barrier function, it is mainly regulated by the protein Src tyrosine kinase. Src is activated by inflammatory cytokines released due to surgical stress, such as TNFα, enabling the synthesis of Intracellular Adhesion Molecules-1 (ICAM-1), and initiating neutrophil transmigration and adhesion. It is known that tumor cells can synthesize ICAM-1, allowing the binding to a neutrophil, which results in extravasation and transmigration. Studies have shown that lidocaine preserves the integrity of the endothelial barrier by modulating the endothelial protein Src tyrosine kinase, which prevents the spread of tumor cells to the systemic circulation, a necessary step for the manifestation of metastasis. In vitro studies analyzing lung microvascular endothelial cells, incubated with TNFα, lidocaine or ropivacaine, have suggested that both local anesthetics would block the Receptor for TNFα (RTNFα), impeding the Src activation and the synthesis of ICAM.4848 Piegeler T, Votta-Velis EG, Liu G, et al. Antimetastatic potential of amide-linked local anesthetics: Inhibition of lung adenocarcinoma cell migration and inflammatory Src signaling independent of sodium channel blockade. Anesthesiology. 2012;117:548-59. Concerning NK cells, they are part of CMI, having a vital role in detecting and removing Circulating Tumor Cells (CTC) that may develop into micrometastasis. Surgical stress, pain, opioids, and volatile agents are involved in the decrease of cytolytic activity of NK cells. On the contrary, a recent study has shown that lidocaine at low concentrations might preserve the cytotoxic function of NK cells.4949 Ramirez MF, Tran P, Cata JP. The effect of clinically therapeutic plasma concentrations of lidocaine on natural killer cell cytotoxicity. Reg Anesth Pain Med. 2015;40:43-8. Ramirez et al. observed that lidocaine, at concentrations lower than those typically found during intravenous infusion, enhanced the cytotoxicity of NK cells against three different types of leukemia cell lines. The same group of investigators has shown that lidocaine in vitro has strong stimulatory effects on the killing activity of NK cells against pancreatic, ovarian and osteosarcoma cells. Finally, Wang et al. have shown that IV lidocaine might also preserve the balance of Th1/Th2 after radical hysterectomy for cervical cancer.5050 Wang HL, Yan HD, Liu YY, et al. Intraoperative intravenous lidocaine exerts a protective effect on cell-mediated immunity in patients undergoing radical hysterectomy. Mol Med Rep. 2015;12:7039-44. Perioperative intravenous lidocaine had a beneficial effect on CMI, and this was associated with the preservation of lymphocyte proliferation and attenuation of apoptosis, maintenance of the balance of Th1/Th2 cells and the decreased production of cytokines. Collectively, the clinical data suggested an enhanced effect of lidocaine on immunity and might support its clinical use during the perioperative period.

Clinical application of lidocaine

Lidocaine can be used in different scenarios in the anesthetic practice, such as local infiltration, regional blocks (epidural, spinal, peripheral nerves), as well as an intravenous infusion (IV) in the context of multimodal analgesia.1414 Soto G, Naranjo Gonzalez M, Calero F. Intravenous lidocaine infusion. Rev Esp Anestesiol Reanim. 2018;65:269-74. Recently, Brown et al. introduced the concept of multimodal general anesthesia, combined different antinociceptive agents and monitoring continuously levels of antinociception and unconsciousness.5151 Brown EN, Pavone KJ, Naranjo M. Multimodal general anesthesia: theory and practice. Anesth Analg. 2018;127:1246-58. Regarding IV lidocaine infusion for multimodal analgesia, Chamaraux et al.5252 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. 2017;4:235. hypothesized that the administration in oncological surgery could be beneficial due to its antimetastatic effects on tumor cells. The IV lidocaine infusion decreases the exposure to opioids and volatile agents,5353 Calero F, Pignolo F, Soto G. Effect of intravenous lidocaine infusion on sevoflurane and fentanyl consumption, hemodynamic response and ventricular repolarization. Rev Argent Anestesiol. 2016;74:49-56. anesthetic drugs that suppress cell-mediated immunity, promote proliferation and angiogenesis.5454 Sherwin A, Buggy DJ. The effect of anaesthetic and analgesic technique on oncological outcomes. Curr Anesthesiol Rep. 2018;8:411. Opioids are associated with immunosuppression by modulating the humoral and cell response, and direct actions on µ receptors expressed in tumor and endothelial cells.5555 Singleton PA, Moss J. Effect of perioperative opioids on cancer recurrence: a hypothesis. Future Oncol. 2010;6:1237-42. Regarding volatile agents, some animal experiments indicate that both the number and the incidence of metastasis in cancer experimental models are increased5656 Hooijmans CR, Geessink FJ, Ritskes-Hoitinga M, et al. A systematic review of the modifying effect of anaesthetic drugs on metastasis in animal models for cancer. PLoS One. 2016;11:e0156152. because of the stimulation of the Hypoxia-Inducible Factor α (HIF-α),5757 Zhao H, Iwasaki M, Yang J, et al. Hypoxia-inducible factor-1: A possible link between inhalational anesthetics and tumor progression?. Act Anaesth Taiwan. 2014;52:70-6. which provides cryoprotection and a greater survival to the tumor cell.5858 Liao D, Johnson RS. Hypoxia: a key regulator of angiogenesis in cancer. Cancer Metastasis Rev. 2007;26:281-90. Volatile agents modify the immune response during the perioperative period in vitro and animal models,5959 Evans D, Fowler-Williams C, Ma D. Is volatile anesthesia during cancer surgery likely to increase the metastatic risk?. Int Anesth Clin. 2016;54:92-107.,6060 Elena G, Amerio N, Ferrero P, et al. Effects of repetitive sevoflurane anaesthesia on immune response, select biochemical parameters and organ histology in mice. Lab Animals. 2003;37:193-203. mainly affecting the cytokines release. IV lidocaine infusion (bolus of 1.5 mg.kg−1 followed by 2 mg.kg−1.h−1) is the most widely used and best-described dosage. When lidocaine is administered at this range of infusion, plasma concentrations of approximately 2 ug.mL−1 are obtained. Plasma concentrations of more than 5 ug.mL−1 are considered toxic. However, adverse effects and toxicity are extremely rare in controlled infusions.4646 Dunn LK, Durieux ME. Perioperative use of intravenous lidocaine. Anesthesiology. 2017;126:729-37. In a recent study, Greenwood et al measured the plasma lidocaine concentration of 32 patients at 30 minutes, 6 hours and 12 hours after starting IV lidocaine infusion for analgesia after major colorectal surgery. Patients received a bolus of 1.5 mg.kg−1 followed by a continuous infusion of lidocaine at 3 mL.hr 1 (60 mg.hr−1) or 6 mL.hr−1 (120 mg.hr−1). The overall mean plasma lidocaine concentration was 4.0 µg.mL−1 (range 0.6–12.3 µg.mL−1). There were no adverse events or reports of symptoms of local anesthetic toxicity.6161 Greenwood E, Nimmo S, Paterson H, et al. Intravenous lidocaine infusion as a component of multimodal analgesia for colorectal surgery measurement of plasma levels. Periop Med. 2019;8:1. Although there is strong evidence from in vitro studies suggesting a protective effect on cancer recurrence, further preclinical and clinical studies are needed to support its role in oncological surgery. Consensus guidelines have recently recommended the use of standardized endpoints in investigation of the perioperative management of cancer patients to develop future clinical recommendations.6262 Buggy DJ, Freeman J, Johnson MZ, et al. Systematic review and consensus definitions for standardised endpoints in perioperative medicine: postoperative cancer outcomes. Br J Anaesth. 2018;121:38-44.

Conclusion

Functionally active VGSCs are expressed in many metastatic cancer cells. In epithelial tumor cells, VGSCs are part of cellular structures named invadopodia, involved in cancer progression. This functional expression is an integral element of the metastatic process in many different solid tumors. For this reason, VGSCs can be targeted to decrease migration, invasion and metastasis. Lidocaine acts by blocking VGSCs and other receptors, and it is used in different scenarios in anesthetic practice, including anesthesia. IV lidocaine as part of the perioperative anesthesia regimen would be of major interest for anesthesiologists, as it might bear the potential to reduce the risk of cancer recurrence or progression patients undergoing cancer surgery. Despite these encouraging studies, they need to be replicated in humans, in the context of oncological surgery. While the concept that anesthetic or analgesic techniques might affect cancer outcomes, there is currently insufficient evidence to support any change in clinical practice.

Acknowledgements

The authors gratefully acknowledge Juan P Cata, MD, MD Anderson Cancer Center, The University of Texas for a critical revision.

References

  • 1
    Jemal A, Bray F, Center MM, et al. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.
  • 2
    Exadaktylos AK, Buggy DJ, Moriarty DC, et al. Can anesthetic technique for primary breast cancer surgery affect recurrence or metastasis?. Anesthesiology. 2006;105:660-4.
  • 3
    Buggy DJ, Borgeat A, Cata J, et al. Consensus statement from the BJA workshop on cancer and anaesthesia. Br J Anaesth. 2015;114(1):2-3.
  • 4
    Heaney A, Buggy DJ. Can anaesthetic and analgesic techniquesaffect cancer recurrence or metastasis? Br J Anaesth. 2012;109.S1:i17-i28.
  • 5
    Kim J. Effects of surgery and anesthetic choice on immunosuppression and cancer recurrence. J Transl Med. 2018;16:8.
  • 6
    Lee BM, Cata JP. Impact of anesthesia on cancer recurrence. Rev Esp Anestesiol Reanim. 2015;62:570-5.
  • 7
    Gottschalk A, Sharma S, Ford J, et al. The role of the perioperative period in recurrence after cancer surgery. Anesth Analg. 2010;110:1636-43.
  • 8
    McCausland K, Martin N, Missair A. Anaesthetic technique and cancer recurrence: current understanding. OA Anaesth. 2014;18:1-7.
  • 9
    Besson P, Driffort V, Bon E, et al. How do voltage. gated sodium channels enhance migration and invasiveness in cancer cells?. Biochim Biophys. 2015;1848:2493-501.
  • 10
    Brackenbury WJ. Voltage-gated sodium channels and metastatic disease. Channels (Austin). 2012;6:352-61.
  • 11
    Fife M, McCarroll JA, Kavallaris M. Movers and shakers: cell cytoskeleton in cancer metastasis. Br J Pharm. 2014;171:5507-23.
  • 12
    Sekandarzad MW, Van Zundert AAJ, Lirk PB, et al. Perioperative anesthesia care and tumor progression. Anesth Analg. 2017;124:1697-708.
  • 13
    Weinberg L, Peake B, Tan C, et al. Pharmacokinetics and pharmacodynamics of lignocaine: a review. World J Anesthesiol. 2015;4:17-29.
  • 14
    Soto G, Naranjo Gonzalez M, Calero F. Intravenous lidocaine infusion. Rev Esp Anestesiol Reanim. 2018;65:269-74.
  • 15
    Ramírez MF, Huitink JM, Cata JP. Perioperative clinical interventions that modify the immune response in cancer patients. Op J Anesth. 2013;3:133-9.
  • 16
    Missair A, Cata JP, Votta-Velis G, et al. Impact of perioperative pain management on cancer recurrence: an ASRA/ESRA special article. Reg Anesth Pain Med. 2019;44:13-28.
  • 17
    Cassinello F, Prieto I, del Olmo M, et al. Cancer surgery: how may anesthesia influence outcome?. J Clin Anesth. 2015;27:262-72.
  • 18
    Gupta GP, Massague J. Cancer metastasis: building a framework. Cell. 2006;127:679-95.
  • 19
    Friedl P, Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell. 2011;147:992-1009.
  • 20
    Catterall WA. From ionic currents to molecular mechanisms: the structure and function of voltage-gated sodium channels. Neuron. 2000;26:13-25.
  • 21
    Catterall WA. Voltage-gated sodium channel: structure, function, and pathophysiology. J Physiol. 2012;590:2577-89.
  • 22
    Black JA, Waxman SG. Non-canonical roles of voltage-gated sodium channels. Neuron. 2013;80:280-91.
  • 23
    Roger S, Gillet L, LeGuennec JY, et al. Voltage-gated sodium channels and cancer: is excitability their primary role?. Front Pharmacol. 2015;6:152.
  • 24
    Lambert AW, Pattabiraman DR, Weinberg RA. Emerging biological principles of metastasis. Cell. 2017;168:670-91.
  • 25
    Jobe N, Rösel D, Tolde O, et al. Complex 3D models to study drug targeting of invadopodias. Clin Cancer Drugs. 2014;1:85-9.
  • 26
    Linder S. The matrix corroded: podosomes and invadopodias in extracellular matrix degradation. Trends Cell Biol. 2007;17:107-17.
  • 27
    Eckert MA, Yang J. Targeting invadopodia to block breast cancer metastasis. Oncotarget. 2011;2:562-8.
  • 28
    Brisson L, Driffort V, Benoist L, et al. Nav1.5 Na+ channels allosterically regulate the NHE-1 exchanger and promote the activity of breast cancer cell invadopodia. J Cell Sci. 2013;126:4835-42.
  • 29
    Brackenbury WJ, Chioni AM, Diss JK, et al. The neonatal splice variant of Nav1.5 potentiates in vitro invasive behavior of MDA-MB-231 human breast cancer cells. Breast Cancer Res Treat. 2007;101:149-60.
  • 30
    Djamgoz MBA. Biophysics of cancer: cellular excitability (“CELEX”) hypothesis of metastasis. J Clin Exp Oncol. 2014.S1:005.
  • 31
    Martin F, Ufodiama C, Watt I, et al. Therapeutic value of voltage-gated sodium channel inhibitors in breast, colorectal, and prostate cancer: a systematic review. Front Pharmacol. 2015;6:273.
  • 32
    Koltai T. Voltage-gated sodium channel as a target for metastatic risk reduction with re-purposed drugs. F1000Res. 2015;4:297.
  • 33
    Fraser SP, Diss JK, Chioni AM, et al. Voltage-gated sodium channel expression and potentiation of human breast cancer metastasis. Clin Cancer Res. 2005;11:5381-9.
  • 34
    Chamaraux-Tran TN, Mathelin C, Aprahamian M, 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.
  • 35
    Fraser S, Foo I, Djamgoz M. Local anaesthetic use in cancer surgery and disease recurrence: role of voltage-gated sodium channels?. Br J Anaesth. 2014;113:899-902.
  • 36
    Piegeler T, Votta-Velis EG, Liu G, et al. Antimetastatic potential of amide-linked local anesthetics: inhibition of lung adenocarcinoma cell migration and inflammatory Src signaling independent of sodium channel blockade. Anesthesiology. 2012;117:548-59.
  • 37
    Chang Y, Liu CH, Chen M, et al. Local anesthetics induce apoptosis in human breast tumor cells. Anesth Analg. 2014;118:116-24.
  • 38
    Wang HW, Wang LY, Jiang L, et al. Amide-linked local anesthetics induce apoptosis in human non-small cell lung cancer. J Thorac Dis. 2016;8:2748-57.
  • 39
    Xing W, Chen DT, Pan JH, et al. Lidocaine induces apoptosis and suppresses tumor growth in human hepatocellular carcinoma cells in vitro and in a xenograft model in vivo. Anesthesiology. 2017;126:868-81.
  • 40
    Lirk P, Berger R, Hollmann MW, et al. Lidocaine time- and dose-dependently demethylates deoxyribonucleic acid in breast cancer cell lines in vitro. Br J Anaesth. 2012;109:200-7.
  • 41
    Doehring A, Oertel BG, Sittl R, et al. Chronic opioid use is associated with increased DNA methylation correlating with increased clinical pain. Pain. 2013;154:15-23.
  • 42
    Sakaguchi M, Kuroda Y, Hirose M. The antiproliferative effect of lidocaine on human tongue cancer cells with inhibition of the activity of epidermal growth factor receptor. Anesth Analg. 2006;102:1103-7.
  • 43
    Mammoto T, Higashiyama S, Mukai M, et al. Infiltration anesthetic lidocaine inhibits cancer cell invasion by modulating ectodomain shedding of heparin-binding epidermal growth factor-like growth factor (HB-EGF). J Cell Physiol. 2002;192:351-8.
  • 44
    Jiang Y, Gou H, Zhu J, et al. Lidocaine inhibits the invasion and migration of TRPV6-expressing cancer cells by TRPV6 down-regulation. Oncol Lett. 2016;12:1164-70.
  • 45
    Piegeler T, Hollmann MW, Borgeat A, et al. Do amide local anesthetics play a therapeutic role in the perioperative management of cancer patients?. Int Anesthesiol Clin. 2016;54(4):e17-32.
  • 46
    Dunn LK, Durieux ME. Perioperative use of intravenous lidocaine. Anesthesiology. 2017;126:729-37.
  • 47
    Weinberg L, Peake B, Tan C, et al. Pharmacokinetics and pharmacodynamics of lignocaine: A review. World J Anesthesiol. 2015;4:17-29.
  • 48
    Piegeler T, Votta-Velis EG, Liu G, et al. Antimetastatic potential of amide-linked local anesthetics: Inhibition of lung adenocarcinoma cell migration and inflammatory Src signaling independent of sodium channel blockade. Anesthesiology. 2012;117:548-59.
  • 49
    Ramirez MF, Tran P, Cata JP. The effect of clinically therapeutic plasma concentrations of lidocaine on natural killer cell cytotoxicity. Reg Anesth Pain Med. 2015;40:43-8.
  • 50
    Wang HL, Yan HD, Liu YY, et al. Intraoperative intravenous lidocaine exerts a protective effect on cell-mediated immunity in patients undergoing radical hysterectomy. Mol Med Rep. 2015;12:7039-44.
  • 51
    Brown EN, Pavone KJ, Naranjo M. Multimodal general anesthesia: theory and practice. Anesth Analg. 2018;127:1246-58.
  • 52
    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. 2017;4:235.
  • 53
    Calero F, Pignolo F, Soto G. Effect of intravenous lidocaine infusion on sevoflurane and fentanyl consumption, hemodynamic response and ventricular repolarization. Rev Argent Anestesiol. 2016;74:49-56.
  • 54
    Sherwin A, Buggy DJ. The effect of anaesthetic and analgesic technique on oncological outcomes. Curr Anesthesiol Rep. 2018;8:411.
  • 55
    Singleton PA, Moss J. Effect of perioperative opioids on cancer recurrence: a hypothesis. Future Oncol. 2010;6:1237-42.
  • 56
    Hooijmans CR, Geessink FJ, Ritskes-Hoitinga M, et al. A systematic review of the modifying effect of anaesthetic drugs on metastasis in animal models for cancer. PLoS One. 2016;11:e0156152.
  • 57
    Zhao H, Iwasaki M, Yang J, et al. Hypoxia-inducible factor-1: A possible link between inhalational anesthetics and tumor progression?. Act Anaesth Taiwan. 2014;52:70-6.
  • 58
    Liao D, Johnson RS. Hypoxia: a key regulator of angiogenesis in cancer. Cancer Metastasis Rev. 2007;26:281-90.
  • 59
    Evans D, Fowler-Williams C, Ma D. Is volatile anesthesia during cancer surgery likely to increase the metastatic risk?. Int Anesth Clin. 2016;54:92-107.
  • 60
    Elena G, Amerio N, Ferrero P, et al. Effects of repetitive sevoflurane anaesthesia on immune response, select biochemical parameters and organ histology in mice. Lab Animals. 2003;37:193-203.
  • 61
    Greenwood E, Nimmo S, Paterson H, et al. Intravenous lidocaine infusion as a component of multimodal analgesia for colorectal surgery measurement of plasma levels. Periop Med. 2019;8:1.
  • 62
    Buggy DJ, Freeman J, Johnson MZ, et al. Systematic review and consensus definitions for standardised endpoints in perioperative medicine: postoperative cancer outcomes. Br J Anaesth. 2018;121:38-44.

Publication Dates

  • Publication in this collection
    23 Oct 2020
  • Date of issue
    Sep-Oct 2020

History

  • Received
    26 May 2019
  • Accepted
    17 Apr 2020
Sociedade Brasileira de Anestesiologia R. Professor Alfredo Gomes, 36, 22251-080 Botafogo RJ Brasil, Tel: +55 21 2537-8100, Fax: +55 21 2537-8188 - Campinas - SP - Brazil
E-mail: bjan@sbahq.org