1
|
Luo L, Cheng Y, Wang H, Li L, Niu H, Yang Y, Zhou Q, He J, Xu J. Lidocaine-A Promising Candidate for the Treatment of Cancer-Induced Bone Pain: A Narrative Review. Adv Ther 2025; 42:2587-2605. [PMID: 40232625 DOI: 10.1007/s12325-025-03192-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 03/27/2025] [Indexed: 04/16/2025]
Abstract
Pain is one of the most common symptoms in patients with cancer, with cancer-induced bone pain (CIBP) significantly affecting their quality of life. Opioids are commonly used as first-line treatments for cancer pain, but their use requires caution due to non-mechanistic analgesia and significant side effects. As a result, there is a need for new non-opioid drugs that target cancer pain through specific mechanisms. Recent studies on the anticancer effects of lidocaine have highlighted its potential benefits in both treating cancer and alleviating cancer-induced pain. This article discusses the mechanism of action and clinical applications of lidocaine in cancer pain management, and suggests new treatment approaches for patients with CIBP.
Collapse
Affiliation(s)
- Lihan Luo
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yuqi Cheng
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Hanxi Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Li Li
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Hanyun Niu
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yuzhu Yang
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Qianqian Zhou
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Jiannan He
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| | - Jianhong Xu
- Department of Anesthesiology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China.
| |
Collapse
|
2
|
Cai Y, Li Q, Banga AK, Wesselmann U, Zhao C. Tetrodotoxin Delivery Pen Safely Uses Potent Natural Neurotoxin to Manage Severe Cutaneous Pain. Adv Healthc Mater 2025; 14:e2401549. [PMID: 39981822 PMCID: PMC11975472 DOI: 10.1002/adhm.202401549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 12/23/2024] [Indexed: 02/22/2025]
Abstract
Clinically available therapies often inadequately address severe chronic cutaneous pain due to short anesthetic duration, insufficient intensity, or side effects. This study introduces a pen device delivering tetrodotoxin (TTX), a potent neurotoxin targeting nerve voltage-gated sodium channels, as a safe and effective topical anesthetic to treat severe chronic cutaneous pain. Chemical permeation enhancers, such as sodium dodecyl sulfate (SDS) and limonene (LIM), are incorporated to enhance TTX skin permeability. The device ensures precise TTX dosing down to the nanogram level, essential to avoid TTX overdose. In rats, the pen device treatment produces TTX-dose-dependent anesthetic effectiveness. An administration of 900 ng of TTX with SDS and LIM to the rat back skin produces a 393.25% increase (measurement limit) in the nociceptive skin pressure threshold, and the hypoalgesia lasts for 11.25 h, outperforming bupivacaine (28 µg), of which are 25.24% and under 1 h. Moreover, the pen device provides on-demand therapy for multiple treatments, consistently achieving prolonged anesthesia over ten sessions (1 treatment per day) without noted toxicity. Furthermore, a single topical administration of 16 µg of TTX exhibits no TTX-related toxicity in rats. The TTX delivery pen paves the way for clinical trials, offering a promising solution for severe cutaneous pain.
Collapse
Affiliation(s)
- Yuhao Cai
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Qi Li
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Ajay K Banga
- Center for Drug Delivery Research, Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, 30341, USA
| | - Ursula Wesselmann
- Departments of Anesthesiology and Perioperative Medicine/Division of Pain Medicine, Neurology and Psychology, and Consortium for Neuroengineering and Brain-Computer Interfaces, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Chao Zhao
- Department of Chemical and Biological Engineering, University of Alabama, Tuscaloosa, AL, 35487, USA
- Center for Convergent Biosciences and Medicine, University of Alabama, Tuscaloosa, AL, 35487, USA
| |
Collapse
|
3
|
Lv R, Wang Y, Sun J, Kang Y, Mou C, Chen Y, Feng Z, Xue X. The effect of lidocaine infusion in oncologic surgery: A bibliometric analysis based on CiteSpace. Medicine (Baltimore) 2024; 103:e40980. [PMID: 39705410 PMCID: PMC11666215 DOI: 10.1097/md.0000000000040980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/20/2024] [Accepted: 11/27/2024] [Indexed: 12/22/2024] Open
Abstract
BACKGROUND Over the past 2 decades, lidocaine's application in oncologic surgery has received significant attention. It has potential antitumor effects and acts as a chemosensitizer. The aim of this study is to analyze the process and frontiers of lidocaine application in oncologic surgery over the past 20 years. METHODS A bibliometric analysis was performed and CiteSpace software was used to conduct metrology, co-occurrence, and cluster analysis. Articles retrieved from the Web of Science database from January 1, 2004 to May 27, 2024. Inclusion criteria comprised peer-reviewed original articles or reviews on lidocaine and cancer, excluding conference abstracts, corrigenda, repeated publications, and unrelated articles. RESULTS A total of 956 articles were included in this study. Two hundred seventeen were selected for detailed analysis. The annual publication count showed an overall increasing trend, peaking in 2022. The United States emerged as the leading country in terms of publication frequency and centrality. Major research themes included lidocaine's antiproliferative effects, enhancement of chemotherapy efficacy, and various administration methods. CONCLUSION Based on the above results, we draw a conclusion that the relationship between lidocaine and cancer has garnered increasing attention, with research in this area rapidly developing. Lidocaine exhibits significant antitumor effects and potential as a chemosensitizer, enhancing the efficacy of traditional chemotherapy. These findings underscore the importance of further research to fully elucidate lidocaine's mechanisms and its potential clinical applications in oncology.
Collapse
Affiliation(s)
- Ranxu Lv
- Jinshan District Central Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yong Wang
- Shanghai Jinshan District Hospital of Integrated Traditional and Western Medicine, Shanghai, China
| | - Jing Sun
- Jinshan District Central Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yuqing Kang
- Jinshan District Central Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Chunyan Mou
- Jinshan District Central Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yujing Chen
- Jinshan District Central Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Zhaoming Feng
- Jinshan District Central Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Xing Xue
- Jinshan District Central Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| |
Collapse
|
4
|
Huang X, Yi P, Gou W, Zhang R, Wu C, Liu L, He Y, Jiang X, Feng J. Neddylation signaling inactivation by tetracaine hydrochloride suppresses cell proliferation and alleviates vemurafenib-resistance of melanoma. Cell Biol Toxicol 2024; 40:81. [PMID: 39297891 PMCID: PMC11413085 DOI: 10.1007/s10565-024-09916-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 09/04/2024] [Indexed: 09/21/2024]
Abstract
Tetracaine, a local anesthetic, exhibits potent cytotoxic effects on multiple cancer; however, the precise underlying mechanisms of its anti-cancer activity remain uncertain. The anti-cancer activity of tetracaine was found to be the most effective among commonly used local anesthetics in this study. After tetracaine treatment, the differentially expressed genes in melanoma cells were identified by the RNAseq technique and enriched in the lysosome signaling pathway, cullin family protein binding, and proteasome signaling pathway through Kyoto Encyclopedia of Genes and Genomes. Additionally, the ubiquitin-like neddylation signaling pathway, which is hyperactivated in melanoma, could be abrogated due to decreased NAE2 expression after tetracaine treatment. The neddylation of the pro-oncogenic Survivin, which enhances its stability, was significantly reduced following treatment with tetracaine. The activation of neddylation signaling by NEDD8 overexpression could reduce the antitumor efficacy of tetracaine in vivo and in vitro. Furthermore, vemurafenib-resistant melanoma cells showed higher level of neddylation, and potential substrate proteins undergoing neddylation modification were identified through immunoprecipitation and mass spectrometry. The tetracaine treatment could reduce drug resistance via neddylation signaling pathway inactivation in melanoma cells. These findings demonstrate that tetracaine effectively inhibits cell proliferation and alleviates vemurafenib resistance in melanoma by suppressing the neddylation signaling pathway, providing a promising avenue for controlling cancer progression.
Collapse
Affiliation(s)
- Xiang Huang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Department of Anesthesiology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou Province, China
| | - Peng Yi
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Wanrong Gou
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Ran Zhang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Chunlin Wu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Yijing He
- Laboratory of Nervous System Disease and Brain Functions, Clinical Research Institute, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Xian Jiang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Department of Anesthesiology, Luzhou People's Hospital, Luzhou, 646000, Sichuan Province, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
| |
Collapse
|
5
|
Yoo YC, Kim NY, Shin S, Yang Y, Jun JH, Oh JE, Kim MH. Anti-Proliferative Effects of Lidocaine as an Autophagy Inducer in Bladder Cancer via Intravesical Instillation: In Vitro and Xenograft Mouse Model Experiments. Cancers (Basel) 2024; 16:1267. [PMID: 38610945 PMCID: PMC11010986 DOI: 10.3390/cancers16071267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/14/2024] Open
Abstract
Lidocaine exerts potential anti-tumor effects on various cancer cell lines, and its intravesical instillation is considered safer than intravenous administration for bladder cancer. However, the mechanisms underlying its anti-tumor effects have not been fully elucidated. Here, we aimed to elucidate the anti-tumor molecular mechanisms of lidocaine in bladder cancer cells and a xenograft model to substantiate the efficacy of its intravesical administration. We investigated the anti-proliferative and autophagyinducing activities of lidocaine in Nara Bladder Tumor No. 2 (NBT-II) rat bladder carcinoma cells using cell viability, flow cytometry, a wound healing assay, and western blotting. We also established a xenograft mouse model of bladder cancer, and cancer growth was examined using in vivo bioluminescence imaging. Lidocaine decreased cell viability, induced G0/G1 phase cell cycle arrest, and inhibited cell migration partially via glycogen synthase kinase (GSK) 3β phosphorylation. Moreover, a combination of lidocaine and SB216763 (a GSK3β inhibitor) suppressed autophagy-related protein expression. Bafilomycin-A1 with lidocaine significantly enhanced microtubule-associated protein 1A/1B-light chain (LC3B) expression; however, it decreased LC3B expression in combination with 3-methyladenine compared to lidocaine alone. In the xenograft mouse model, the bladder cancer volume was reduced by lidocaine. Overall, lidocaine exerts anti-proliferative effects on bladder cancer via an autophagy-inducing mechanism.
Collapse
Affiliation(s)
- Young Chul Yoo
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; (Y.C.Y.); (N.-Y.K.); (S.S.)
| | - Na-Young Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; (Y.C.Y.); (N.-Y.K.); (S.S.)
| | - Seokyung Shin
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; (Y.C.Y.); (N.-Y.K.); (S.S.)
| | - Yunil Yang
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Gangnam Severance Hospital, Eonju-ro 211, Gangnam-gu, Seoul 06273, Republic of Korea;
| | - Ji Hae Jun
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea;
| | - Ju Eun Oh
- Anesthesia and Pain Research Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea;
| | - Myoung Hwa Kim
- Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute, Yonsei University College of Medicine, Gangnam Severance Hospital, Eonju-ro 211, Gangnam-gu, Seoul 06273, Republic of Korea;
| |
Collapse
|
6
|
Carnet Le Provost K, Kepp O, Kroemer G, Bezu L. Trial watch: local anesthetics in cancer therapy. Oncoimmunology 2024; 13:2308940. [PMID: 38504848 PMCID: PMC10950281 DOI: 10.1080/2162402x.2024.2308940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
Preclinical evidence indicates potent antitumor properties of local anesthetics. Numerous underlying mechanisms explaining such anticancer effects have been identified, suggesting direct cytotoxic as well as indirect immunemediated effects that together reduce the proliferative, invasive and migratory potential of malignant cells. Although some retrospective and correlative studies support these findings, prospective randomized controlled trials have not yet fully confirmed the antineoplastic activity of local anesthetics, likely due to the intricate methodology required for mitigating confounding factors. This trial watch aims at compiling all published preclinical and clinical research, along with completed and ongoing trials, that explore the potential antitumor effects of local anesthetics.
Collapse
Affiliation(s)
- Killian Carnet Le Provost
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Oliver Kepp
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Guido Kroemer
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Pôle de Biologie, Hôpital européen Georges Pompidou, AP-HP, Paris, France
| | - Lucillia Bezu
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, Centre de Recherche des Cordeliers, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Université Paris Saclay, Villejuif, France
- Gustave Roussy, Département Anesthésie, Chirurgie et Interventionnel, Villejuif, France
| |
Collapse
|
7
|
Priščáková P, Svoboda M, Feketová Z, Hutník J, Repiská V, Gbelcová H, Gergely L. Syncytin-1, syncytin-2 and suppressyn in human health and disease. J Mol Med (Berl) 2023; 101:1527-1542. [PMID: 37855856 PMCID: PMC10697888 DOI: 10.1007/s00109-023-02385-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023]
Abstract
In this review, we summarized the results of experimental and clinical studies about three human endogenous retroviruses and their products-syncytin-1, syncytin-2, and suppressyn in human physiology and pathophysiology. We summed up the described connection with various pathological processes and diseases, mainly with pregnancy-induced hypertensive diseases such as preeclampsia, oncogenesis, gestational trophoblastic disease, and multiple sclerosis. Supposed mechanisms of action and the potential of clinical applications are also described.
Collapse
Affiliation(s)
- Petra Priščáková
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, Bratislava, 811 08, Slovak Republic
| | - Michal Svoboda
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, Bratislava, 811 08, Slovak Republic
| | - Zuzana Feketová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, Bratislava, 811 08, Slovak Republic
| | - Juraj Hutník
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, Bratislava, 811 08, Slovak Republic
| | - Vanda Repiská
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, Bratislava, 811 08, Slovak Republic
| | - Helena Gbelcová
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, Bratislava, 811 08, Slovak Republic
| | - Lajos Gergely
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University Bratislava, Sasinkova 4, Bratislava, 811 08, Slovak Republic.
| |
Collapse
|
8
|
Debel W, Ramadhan A, Vanpeteghem C, Forsyth RG. Does the Choice of Anaesthesia Affect Cancer? A Molecular Crosstalk between Theory and Practice. Cancers (Basel) 2022; 15:cancers15010209. [PMID: 36612205 PMCID: PMC9818147 DOI: 10.3390/cancers15010209] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
In recent years, there has been an increasing scientific interest in the interaction between anaesthesia and cancer development. Retrospective studies show that the choice of anaesthetics may influence cancer outcome and cancer recurrence; however, these studies show contradictory results. Recently, some large randomized clinical trials have been completed, yet they show no significant effect of anaesthetics on cancer outcomes. In this scoping review, we compiled a body of in vivo and in vitro studies with the goal of evaluating the biological effects of anaesthetics on cancer cells in comparison to clinical effects as described in recent studies. It was found that sevoflurane, propofol, opioids and lidocaine are likely to display direct biological effects on cancer cells; however, significant effects are only found in studies with exposure to high concentrations of anaesthetics and/or during longer exposure times. When compared to clinical data, these differences in exposure and dose-effect relation, as well as tissue selectivity, population selection and unclear anaesthetic dosing protocols might explain the lack of outcome.
Collapse
Affiliation(s)
- Wiebrecht Debel
- Department of Anesthesiology, University Hospital Ghent, 9000 Ghent, Belgium
| | - Ali Ramadhan
- Department of Pathological Anatomy, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium
| | | | - Ramses G. Forsyth
- Department of Pathological Anatomy, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium
- Laboratorium for Experimental Pathology (EXPA), Vrije Universiteit Brussel, 1090 Brussels, Belgium
- Correspondence: ; Tel.: +32-(2)-4775084
| |
Collapse
|
9
|
Haraguchi-Suzuki K, Kawabata-Iwakawa R, Suzuki T, Suto T, Takazawa T, Saito S. Local anesthetic lidocaine-inducible gene, growth differentiation factor-15 suppresses the growth of cancer cell lines. Sci Rep 2022; 12:14520. [PMID: 36008442 PMCID: PMC9411556 DOI: 10.1038/s41598-022-18572-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022] Open
Abstract
Administration of local anesthetics, such as lidocaine, in the perioperative period improves outcomes of cancer patients. However, its precise mechanism is still unresolved. The growth of human cancer cell lines, including HeLa cells, are suppressed by lidocaine treatment. We identified that growth differentiation factor-15 (GDF-15) was commonly upregulated in lidocaine-treated cancer cell lines. GDF-15 is a divergent member of the transforming growth factor-β (TGF-β) superfamily and it is produced as an unprocessed pro-protein form and then cleaved to generate a mature form. In lidocaine-treated HeLa cells, increased production of GDF-15 in the endoplasmic reticulum (ER) was observed and unprocessed pro-protein form of GDF-15 was secreted extracellularly. Further, lidocaine induced apoptosis and apoptosis-inducible Tribbles homologue 3 (TRIB3) was also commonly upregulated in lidocaine-treated cancer cell lines. In addition, transcription factor C/EBP homologous protein (CHOP), which is a positive regulator of not only GDF-15 but TRIB3 was also induced by lidocaine. Lidocaine-induced growth suppression and apoptosis was suppressed by knockdown of GDF-15 or TRIB3 expression by small interference RNA (siRNA). These observations suggest that lidocaine suppresses the growth of cancer cells through increasing GDF-15 and TRIB3 expression, suggesting its potential application as cancer therapy.
Collapse
Affiliation(s)
- Keiko Haraguchi-Suzuki
- Intensive Care Unit, Gunma University Hospital, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan.
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Initiative for Advanced Research, Gunma University, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Toru Suzuki
- Laboratory for Immunogenetics, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Takashi Suto
- Department of Anesthesiology, Gunma University Hospital, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Tomonori Takazawa
- Intensive Care Unit, Gunma University Hospital, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Shigeru Saito
- Department of Anesthesiology, Gunma University Hospital, 3-39-15, Showa-machi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
10
|
Luan T, Li Y, Sun L, Xu S, Wang H, Wang J, Li C. Systemic immune effects of anesthetics and their intracellular targets in tumors. Front Med (Lausanne) 2022; 9:810189. [PMID: 35966857 PMCID: PMC9365985 DOI: 10.3389/fmed.2022.810189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 07/06/2022] [Indexed: 11/30/2022] Open
Abstract
According to the result released by the World Health Organization (WHO), non-communicable diseases have occupied four of the top 10 current causes for death in the world. Cancer is one of the significant factors that trigger complications and deaths; more than 80% cancer patients require surgical or palliative treatment. In this case, anesthetic treatment is indispensable. Since cancer is a heterogeneous disease, various types of interventions can activate oncogenes or mutate tumor suppressor genes. More and more researchers believe that anesthetics have a certain effect on the long-term recurrence and metastasis of tumors, but it is still controversial whether they promote or inhibit the progression of cancer. On this basis, a series of retrospective or prospective randomized clinical trials have been conducted, but it seems to be difficult to reach a conclusion within 5 years or longer. This article focuses on the effects of anesthetic drugs on immune function and cancer and reviews their latest targets on the tumor cells, in order to provide a theoretical basis for optimizing the selection of anesthetic drugs, exploring therapeutic targets, and improving the prognosis of cancer patients.
Collapse
Affiliation(s)
- Ting Luan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, China
| | - Yi Li
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Lihui Sun
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Zhongke Jianlan Medical Research Institute, Beijing, China
| | - Siqi Xu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Zhongke Jianlan Medical Research Institute, Beijing, China
| | - Haifeng Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, China
- *Correspondence: Haifeng Wang,
| | - Jiansong Wang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, China
- Jiansong Wang,
| | - Chong Li
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Zhongke Jianlan Medical Research Institute, Beijing, China
- Chong Li,
| |
Collapse
|
11
|
Brogi E, Forfori F. Anesthesia and cancer recurrence: an overview. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE (ONLINE) 2022; 2:33. [PMID: 37386584 DOI: 10.1186/s44158-022-00060-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/05/2022] [Indexed: 07/01/2023]
Abstract
Several perioperative factors are responsible for the dysregulation or suppression of the immune system with a possible impact on cancer cell growth and the development of new metastasis. These factors have the potential to directly suppress the immune system and activate hypothalamic-pituitary-adrenal axis and the sympathetic nervous system with a consequent further immunosuppressive effect.Anesthetics and analgesics used during the perioperative period may modulate the innate and adaptive immune system, inflammatory system, and angiogenesis, with a possible impact on cancer recurrence and long-term outcome. Even if the current data are controversial and contrasting, it is crucial to increase awareness about this topic among healthcare professionals for a future better and conscious choice of anesthetic techniques.In this article, we aimed to provide an overview regarding the relationship between anesthesia and cancer recurrence. We reviewed the effects of surgery, perioperative factors, and anesthetic agents on tumor cell survival and tumor recurrence.
Collapse
Affiliation(s)
- Etrusca Brogi
- Department of Anesthesia and Intensive Care, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy.
| | - Francesco Forfori
- Department of Anesthesia and Intensive Care, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| |
Collapse
|
12
|
Hao J, Zhang W, Huang Z. Bupivacaine modulates the apoptosis and ferroptosis in bladder cancer via phosphatidylinositol 3-kinase (PI3K)/AKT pathway. Bioengineered 2022; 13:6794-6806. [PMID: 35246010 PMCID: PMC9278971 DOI: 10.1080/21655979.2022.2036909] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The study aimed to explore the effects of local anesthetic bupivacaine on bladder cancer cells in vivo and in vitro. The cytotoxicity was detected by MTT assay. Apoptosis was measured by Hoechst 33342 staining and TUNEL. The contents of Fe2+, Malondialdehyde (MDA), Glutathione (GSH) and reactive oxygen species (ROS) were evaluated by the corresponding kit. Mitochondrial membrane potential was assessed by JC-1 kit. HE staining, TUNEL and immunohistochemistry were used to detect the xenografted tumors. Protein expression was estimated by Western blot. Bupivacaine significantly inhibited the activity of T24 cells and 5637 cells at 0.25-16 mM. Bupivacaine promoted cell apoptosis with increased concentration. bupivacaine inhibited the expression of Bcl-2 and increased the expression of Bax and cytochrome C. Moreover, bupivacaine amplified the level of Fe2+ and ROS, and restrained the expression of cystine/glutamic acid reverse transporter (xCT) and glutathione peroxidase 4 (GPX4). Further results showed that bupivacaine decreased mitochondrial membrane potential, reduced GSH, and increased MDA levels. Besides, bupivacaine attenuated the phosphorylation of PI3K, Akt, and mTOR. In addition, bupivacaine suppressed the growth of xenografted tumors, induced apoptosis and ferroptosis, and inhibited the activity of PI3K/AKT signaling pathway in xenografted tumors. Bupivacaine could induce apoptosis and ferroptosis by inhibiting PI3K/Akt signaling pathway in bladder cancer cells.
Collapse
Affiliation(s)
- Jianli Hao
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Weiqing Zhang
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Zeqing Huang
- Department of Anesthesiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| |
Collapse
|
13
|
Ma X, Yan W, He N. Lidocaine attenuates hypoxia/reoxygenation‑induced inflammation, apoptosis and ferroptosis in lung epithelial cells by regulating the p38 MAPK pathway. Mol Med Rep 2022; 25:150. [PMID: 35244190 PMCID: PMC8941375 DOI: 10.3892/mmr.2022.12666] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 11/22/2021] [Indexed: 11/12/2022] Open
Abstract
Lung ischemia-reperfusion (I/R) injury poses a serious threat to human health, worldwide. The current study aimed to determine the role of lidocaine in A549 cells, in addition to the involvement of the p38 MAPK pathway. Oxygen deprivation/reoxygenation-induced A549 cells were utilized to simulate I/R injury in vitro. Cell viability and apoptosis were detected using MTT and TUNEL assays, respectively. The levels of IL-6, IL-8, TNF-α, malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase, iron and reactive oxygen species (ROS) were measured using corresponding commercial kits. The corresponding protein expression levels were also measured using western blotting. Moreover, a monolayer cell paracellular permeability assay was performed to determine the permeability of A549 cells. The results demonstrated that, whilst lidocaine had no influence on untreated A549 cells, it significantly increased the viability of hypoxia/reoxygenation (H/R)-induced A549 cells. A549 cell apoptosis and the release of inflammatory cytokines in the H/R group were decreased after the addition of lidocaine. When compared with the H/R group, increased MDA level and decreased SOD level were observed in H/R-induced A549 cells following lidocaine treatment. In addition, the permeability of H/R-induced A549 cells was markedly decreased following lidocaine treatment. Compared with the H/R group, the expression levels of tight junction and ferroptosis-related proteins were significantly upregulated by lidocaine, whereas the expression of transferrin was downregulated. However, p79350, an agonist of p38, reversed the effects of lidocaine on H/R-induced A549 cells. In conclusion, lidocaine exerted a protective role in HR-induced lung epithelial cell injury, which may serve as a potential agent for the treatment of patients with lung I/R injury.
Collapse
Affiliation(s)
- Xiaojun Ma
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Weihua Yan
- Department of Pathology, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Na He
- Department of Anesthesiology, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010000, P.R. China
| |
Collapse
|
14
|
Liu T, Jiang F, Yu LY, Wu YY. Lidocaine represses proliferation and cisplatin resistance in cutaneous squamous cell carcinoma via miR-30c/SIRT1 regulation. Bioengineered 2022; 13:6359-6370. [PMID: 35212616 PMCID: PMC8974189 DOI: 10.1080/21655979.2022.2031419] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
This study aimed to determine the effects of lidocaine on cell proliferation and cisplatin resistance in A431 human cutaneous squamous cell carcinoma (cSCC) cells and elucidate the underlying mechanism. Cell proliferation, colony numbers, and cisplatin resistance were determined in A431 or cisplatin-resistant A431 (A431-R) cells that were first transfected with miR-30c-inhibitor or miR-30c-mimic, respectively, and then treated with different concentrations of lidocaine, cisplatin, or both. The expression levels of miR-30c and Sirtuin 1 (SIRT1) in A431 and A431-R cells were determined by quantitative real-time polymerase chain reaction and Western blotting. Lidocaine suppressed A431 cell proliferation and cisplatin resistance in a dose- and time-dependent manner via the miR-30c/SIRT1 pathway. MiR-30c overexpression also suppressed cell proliferation and cisplatin resistance in A431 cells by directly targeting and downregulating SIRT1, thus enhancing the protective effects of lidocaine. Conversely, SIRT1 upregulation or miR-30c inhibition antagonized the inhibitory effects of lidocaine. Our results suggest that lidocaine may suppress the progression of cSCC by activating the miR-30c/SIRT1 pathway, indicating its promising potential as a treatment strategy for cSCC.
Collapse
Affiliation(s)
- Tao Liu
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Fei Jiang
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Li-Yuan Yu
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - You-Yang Wu
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| |
Collapse
|
15
|
Santander Ballestín S, Lanuza Bardaji A, Marco Continente C, Luesma Bartolomé MJ. Antitumor Anesthetic Strategy in the Perioperatory Period of the Oncological Patient: A Review. Front Med (Lausanne) 2022; 9:799355. [PMID: 35252243 PMCID: PMC8894666 DOI: 10.3389/fmed.2022.799355] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
The stress response triggered by the surgical aggression and the transient immunosuppression produced by anesthetic agents stimulate the inadvertent dispersion of neoplastic cells and, paradoxically, tumor progression during the perioperative period. Anesthetic agents and techniques, in relation to metastatic development, are investigated for their impact on long-term survival. Scientific evidence indicates that inhaled anesthetics and opioids benefit immunosuppression, cell proliferation, and angiogenesis, providing the ideal microenvironment for tumor progression. The likely benefit of reducing their use, or even replacing them as much as possible with anesthetic techniques that protect patients from the metastatic process, is still being investigated. The possibility of using "immunoprotective" or "antitumor" anesthetic techniques would represent a turning point in clinical practice. Through understanding of pharmacological mechanisms of anesthetics and their effects on tumor cells, new perioperative approaches emerge with the aim of halting and controlling metastatic development. Epidural anesthesia and propofol have been shown to maintain immune activity and reduce catecholaminergic and inflammatory responses, considering the protective techniques against tumor spread. The current data generate hypotheses about the influence of anesthesia on metastatic development, although prospective trials that determinate causality are necessary to make changes in clinical practice.
Collapse
Affiliation(s)
- Sonia Santander Ballestín
- Department of Pharmacology, Physiology and Legal and Forensic Medicine, Faculty of Medicine, University of Zaragoza, Zaragoza, Spain
| | | | | | | |
Collapse
|
16
|
Wu Chuang A, Kepp O, Kroemer G, Bezu L. Direct Cytotoxic and Indirect, Immune-Mediated Effects of Local Anesthetics Against Cancer. Front Oncol 2022; 11:821785. [PMID: 35096626 PMCID: PMC8796204 DOI: 10.3389/fonc.2021.821785] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/27/2021] [Indexed: 12/15/2022] Open
Abstract
Local anesthetics are frequently employed during surgery in order to control peri- and postoperative pain. Retrospective studies have revealed an unexpected correlation between increased long-term survival and the use of local anesthetics during oncological surgery. This effect of local anesthetics might rely on direct cytotoxic effects on malignant cells or on indirect, immune-mediated effects. It is tempting to speculate, yet needs to be formally proven, that the combination of local anesthetics with oncological surgery and conventional anticancer therapy would offer an opportunity to control residual cancer cells. This review summarizes findings from fundamental research together with clinical data on the use of local anesthetics as anticancer standalone drugs or their combination with conventional treatments. We suggest that a better comprehension of the anticancer effects of local anesthetics at the preclinical and clinical levels may broadly improve the surgical treatment of cancer.
Collapse
Affiliation(s)
- Alejandra Wu Chuang
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Oliver Kepp
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
| | - Guido Kroemer
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France.,Pôle de Biologie, Hôpital européen Georges Pompidou, AP-HP, Paris, France
| | - Lucillia Bezu
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France.,Service d'anesthésie, Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
17
|
Yan Y, He W, Chen Y, Li Q, Pan J, Yuan Y, Zeng W, Chen D, Xing W. Comprehensive Analysis to Identify the Encoded Gens of Sodium Channels as a Prognostic Biomarker in Hepatocellular Carcinoma. Front Genet 2022; 12:802067. [PMID: 35126466 PMCID: PMC8815461 DOI: 10.3389/fgene.2021.802067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/30/2021] [Indexed: 12/24/2022] Open
Abstract
The SCN family as the encoded gens of sodium channels has been proven to participate in development of cancers including hepatocellular carcinoma (HCC), but the prognostic value of the SCN family is unclear. The results of the UALCAN database had showed that SCN2A/4A/5A/8A mRNA were highly expressed in tumour tissues, while SCN1A/7A/11A mRNA were expressed at low levels (p < 0.05), furthermore, the expression of SCN4A and SCN7A had the similar levels in microarray analysis result. The pan-tumour analysis showed that SCN7A expression was stably lower in tumours than SCN4A expression by TIMER. Both SCN4A and SCN7A were related to tumour grade, nodal metastatic status, histological subtype, patient race, individual cancer stages and TP53 mutation status to varying degrees. The Kaplan–Meier plotter demonstrated that high SCN4A mRNA expression was correlated with better overall survival (OS), disease-specific survival (DSS) and progression-free survival (PFS) and that high expression of SCN7A mRNA was associated with better OS; however, in Asians, higher SCN4A was correlated with better OS and DSS, and higher SCN7A was well correlated with better OS, recurrence-free survival (RFS), DSS and PFS. Analysis of data from cBioPortal showed that mutation of SCN7A was related to RFS and PFS. The protein expression of SCN4A and SCN7A had been detected by Immunohistochemistry. Univariate survival analysis revealed that high SCN7A protein expression was significantly linked to better OS (p = 0.001) and RFS (p = 0.003). Moreover, SCN7A displayed as an independent prognostic factor by multivariate analysis. In addition, a lower methylation level indicated a poor outcome. Pathway and functional enrichment analysis predicted a relationship between SCN7A and the PI3K pathway. In conclusion, there are significant and stable changes in SCN4A and SCN7A expression in HCC. SCN7A expression has better prognostic value and might participate in HCC progression.
Collapse
Affiliation(s)
- Yan Yan
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Anesthesiology, Huizhou Municipal Central Hospital, Huizhou, China
| | - Wen He
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yonghua Chen
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Qiang Li
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jiahao Pan
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yunfei Yuan
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Weian Zeng
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- *Correspondence: Weian Zeng, ; Dongtai Chen, ; Wei Xing,
| | - Dongtai Chen
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- *Correspondence: Weian Zeng, ; Dongtai Chen, ; Wei Xing,
| | - Wei Xing
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- *Correspondence: Weian Zeng, ; Dongtai Chen, ; Wei Xing,
| |
Collapse
|
18
|
Teng X, Liu Y, Wang L, Wang G. Lidocaine exerts anticancer activity in bladder cancer by targeting isoprenylcysteine carboxylmethyltransferase ( ICMT). Transl Androl Urol 2022; 10:4219-4230. [PMID: 34984187 PMCID: PMC8661257 DOI: 10.21037/tau-21-893] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022] Open
Abstract
Background Bladder cancer is one of the most common malignant tumors among humans and has a high mortality. Clinically, lidocaine is the most commonly used local anesthetic, which can inhibit the proliferation of bladder cancer cells; however, its downstream specific molecular mechanisms are unclear. Methods The SwissTarget and TargetNet databases were used to analyze the target of lidocaine. The online public cancer transcriptome database UALCAN was used to analyze the up-regulated genes in The Cancer Genome Atlas Urothelial Bladder Carcinoma (TCGA-BLCA) data collection, and the intersection of the 2 was used to obtain the core target. The only target, isoprenylcysteine carboxylmethyltransferase (ICMT), was obtained by combining the correlation between the target and the clinical information of bladder cancer and the Kaplan-Meier (K-M) survival curve. Then, UMUC3 and T24 cells were selected as research vectors in vitro. Cell proliferation, cell cycle, and apoptosis were detected by cell counting kit-8, colony formation, flow cytometry, and western blotting. Results Network pharmacology analysis showed that ICMT might be one of the targets of lidocaine, and the expression level of ICMT was closely related to the clinical phenotype of bladder cancer. Lidocaine treatment (4 and 8 mM) significantly inhibited the proliferation of UMUC3 and T24 cells, promoted apoptosis, and significantly inhibited the mass and volume of xenograft tumors. In vitro experiments showed that ICMT promoted the proliferation of UMUC3 and T24 cells. Lidocaine inhibited the expression of ICMT in UMUC3 and T24 cells in a concentration and time-dependent manner, and inhibited cell proliferation by down-regulating ICMT expression. Conclusions Lidocaine exerts anti-tumor effect by down-regulating the expression of ICMT in bladder cancer.
Collapse
Affiliation(s)
- Xiaodan Teng
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yang Liu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Liping Wang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guonian Wang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
19
|
Zhang Y, Jing Y, Pan R, Ding K, Chen R, Meng Q. Mechanisms of Cancer Inhibition by Local Anesthetics. Front Pharmacol 2021; 12:770694. [PMID: 34950031 PMCID: PMC8688799 DOI: 10.3389/fphar.2021.770694] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 11/15/2021] [Indexed: 01/17/2023] Open
Abstract
The use of local anesthetics during surgical treatment of cancer patients is an important part of perioperative analgesia. In recent years, it has been showed that local anesthetics can directly or indirectly affect the progression of tumors. In vitro and in vivo studies have demonstrated that local anesthetics reduced cancer recurrence. The etiology of this effect is likely multifactorial. Numerous mechanisms were proposed based on the local anesthetic used and the type of cancer. Mechanisms center on NaV1.5 channels, Ras homolog gene family member A, cell cycle, endothelial growth factor receptor, calcium Influx, microRNA and mitochondrial, in combination with hyperthermia and transient receptor potential melastatin 7 channels. Local anesthetics significantly decrease the proliferation of cancers, including ovarian, breast, prostate, thyroid, colon, glioma, and histiocytic lymphoma cell cancers, by activating cell death signaling and decreasing survival pathways. We also summarized clinical evidence and randomized trial data to confirm that local anesthetics inhibited tumor progression.
Collapse
Affiliation(s)
- Yiguo Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yixin Jing
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rui Pan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingtao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesiology, East Hospital, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
20
|
Voltage-Gated Sodium Channels as Potential Biomarkers and Therapeutic Targets for Epithelial Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13215437. [PMID: 34771603 PMCID: PMC8582439 DOI: 10.3390/cancers13215437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Voltage-gated sodium channels are membrane proteins that change conformation in response to depolarization of the membrane potential, allowing sodium ions to flow into cells. While voltage-gated sodium channels are normally studied in terms of neuron impulses and skeletal or cardiac muscle contraction, abnormal ion channel expression is a feature of many cancer cells. The aim of our study was to assess the expression of voltage-gated sodium channels in ovarian cancer cells. We found that ovarian cancer cells generally express lower levels of voltage-gated sodium channels than normal cells and that two voltage-gated sodium channels, SCN8A and SCN1B, were prognostic biomarkers for ovarian cancer overall survival. In vitro studies suggested that drugs that block voltage-gated sodium channels, such as certain anti-epileptic drugs and local anesthetics, might sensitize ovarian cancer cells to chemotherapy. These findings suggest that voltage-gated sodium channels may be interesting targets for ovarian cancer therapy. Abstract Abnormal ion channel expression distinguishes several types of carcinoma. Here, we explore the relationship between voltage-gated sodium channels (VGSC) and epithelial ovarian cancer (EOC). We find that EOC cell lines express most VGSC, but at lower levels than fallopian tube secretory epithelial cells (the cells of origin for most EOC) or control fibroblasts. Among patient tumor samples, lower SCN8A expression was associated with improved overall survival (OS) (median 111 vs. 52 months; HR 2.04 95% CI: 1.21–3.44; p = 0.007), while lower SCN1B expression was associated with poorer OS (median 45 vs. 56 months; HR 0.69 95% CI 0.54–0.87; p = 0.002). VGSC blockade using either anti-epileptic drugs or local anesthetics (LA) decreased the proliferation of cancer cells. LA increased cell line sensitivity to platinum and taxane chemotherapies. While lidocaine had similar additive effects with chemotherapy among EOC cells and fibroblasts, bupivacaine showed a more pronounced impact on EOC than fibroblasts when combined with either carboplatin (ΔAUC −37% vs. −16%, p = 0.003) or paclitaxel (ΔAUC −37% vs. −22%, p = 0.02). Together, these data suggest VGSC are prognostic biomarkers in EOC and may inform new targets for therapy.
Collapse
|
21
|
Wall TP, Buggy DJ. Perioperative Intravenous Lidocaine and Metastatic Cancer Recurrence - A Narrative Review. Front Oncol 2021; 11:688896. [PMID: 34408981 PMCID: PMC8365881 DOI: 10.3389/fonc.2021.688896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer is a major global health problem and the second leading cause of death worldwide. When detected early, surgery provides a potentially curative intervention for many solid organ tumours. Unfortunately, cancer frequently recurs postoperatively. Evidence from laboratory and retrospective clinical studies suggests that the choice of anaesthetic and analgesic agents used perioperatively may influence the activity of residual cancer cells and thus affect subsequent recurrence risk. The amide local anaesthetic lidocaine has a well-established role in perioperative therapeutics, whether used systemically as an analgesic agent or in the provision of regional anaesthesia. Under laboratory conditions, lidocaine has been shown to inhibit cancer cell behaviour and exerts beneficial effects on components of the inflammatory and immune responses which are known to affect cancer biology. These findings raise the possibility that lidocaine administered perioperatively as a safe and inexpensive intravenous infusion may provide significant benefits in terms of long term cancer outcomes. However, despite the volume of promising laboratory data, robust prospective clinical evidence supporting beneficial anti-cancer effects of perioperative lidocaine treatment is lacking, although trials are planned to address this. This review provides a state of the art summary of the current knowledge base and recent advances regarding perioperative lidocaine therapy, its biological effects and influence on postoperative cancer outcomes.
Collapse
Affiliation(s)
- Thomas P Wall
- Department of Anaesthesiology, Mater Misericordiae University Hospital, School of Medicine, University College Dublin, Dublin, Ireland.,EU COST Action 15204, Euro-Periscope, Brussels, Belgium
| | - Donal J Buggy
- Department of Anaesthesiology, Mater Misericordiae University Hospital, School of Medicine, University College Dublin, Dublin, Ireland.,EU COST Action 15204, Euro-Periscope, Brussels, Belgium.,Outcomes Research, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
22
|
Wang L, Guo W, Guan H, Yan N, Cai X, Zhu L. Local anesthetic bupivacaine inhibits proliferation and metastasis of hepatocellular carcinoma cells via suppressing PI3K/Akt and MAPK signaling. J Biochem Mol Toxicol 2021; 35:e22871. [PMID: 34338398 DOI: 10.1002/jbt.22871] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 07/04/2021] [Accepted: 07/19/2021] [Indexed: 12/16/2022]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death worldwide. Retrospective studies suggest that using local/regional anesthetic (LA/RA) is associated with better outcomes in primary HCC patients. In this study, we evaluated the effects of LA/RA bupivacaine in HCC cells and the underlying molecular mechanisms. The biological functions of bupivacaine in HCC cells were evaluated by transcriptome RNA sequencing, cell viability assay, bromodeoxyuridine incorporation assay, colony formation assay, flow cytometry, western blot, wound healing assay, transwell cell migration assay, tumor xenograft formation, and lung metastasis assay. Bupivacaine suppressed proliferation and induced apoptosis of HepG2 and SNU-449 cells in a time- and dose-dependent manner. Bupivacaine treatment also decreased colony formation, migration, and invasion of HepG2 and SNU-449 cells. In mouse models, bupivacaine repressed tumor xenograft growth and lung metastasis of HepG2 cells. Transcriptome sequencing of HepG2 cells suggested that PI3K/Akt and MAPK signaling pathways were suppressed by bupivacaine treatment. In western blot analysis, bupivacaine reduced the expression of total and phosphorylated Akt, mTOR, and MAPK. Furthermore, reactivated PI3K/Akt and MAPK signaling by EGF or NRG1 partially reversed the effects of bupivacaine on cell growth, colony formation, and invasion of HCC cells. Local anesthetic bupivacaine suppressed proliferation, migration and invasion, and induced apoptosis of HCC cells. Our results provided novel insights into the local anesthetic bupivacaine in the therapy of HCC patients.
Collapse
Affiliation(s)
- Lei Wang
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Weijia Guo
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hongman Guan
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ni Yan
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaolan Cai
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lili Zhu
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
23
|
Sun M, Huang S, Gao Y. Lidocaine inhibits the proliferation and metastasis of epithelial ovarian cancer through the Wnt/β-catenin pathway. Transl Cancer Res 2021; 10:3479-3490. [PMID: 35116652 PMCID: PMC8799064 DOI: 10.21037/tcr-21-1047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Lidocaine, an amide local anesthetic, has recently been found to have anticancer action in various cancer cells. However, the role of lidocaine in epithelial ovarian cancer (EOC) remains largely unknown. In the present study, we investigated how lidocaine regulates the progression of EOC. METHODS Real-time polymerase chain reaction was used to examine the expression of Snail, Wnt, β-catenin, E-cadherin, vimentin, matrix metalloproteinase (MMP)-7, MMP-9, and vascular endothelial growth factor in lidocaine-treated cells. Cell proliferation assays, cell apoptosis assays, and cell migration assays were employed to verify the function of lidocaine in EOC cells. Cell proliferation and cell migration assays were employed to verify the function of Wnt/β-catenin signaling in lidocaine-treated EOC cells together with Wnt-overexpressing plasmids or inhibitor NVP-XAV939. RESULTS Lidocaine could inhibit proliferation, migration, and invasion, and induce apoptosis in ovarian cancer cells lines in a dose-dependent manner. Wnt/β-catenin signaling was involved in the suppression of epithelial-mesenchymal transition progression of ovarian cancer cells, which resulted in the downregulation of Snail and vimentin, as well as the upregulation of E-cadherin. Furthermore, overexpressed Wnt could reverse the carcinostatic effect of lidocaine, while Wnt inhibitor XAV-939 synergistically enhanced the antitumor effect of lidocaine. CONCLUSIONS Mechanistically, lidocaine could inhibit the proliferation and metastasis of EOC by the Wnt/β-catenin pathway to regulate the progression of EOC.
Collapse
Affiliation(s)
- Mei Sun
- Department of Anesthesiology, the Affiliated Nanjing Jiangbei Hospital of Nantong University, Nanjing, China
| | - Saisai Huang
- Department of Anesthesiology, the Affiliated Hospital of Nantong University, Nantong, China
| | - Yongtao Gao
- Department of Anesthesiology, the Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
24
|
Zhang C, Xie C, Lu Y. Local Anesthetic Lidocaine and Cancer: Insight Into Tumor Progression and Recurrence. Front Oncol 2021; 11:669746. [PMID: 34249706 PMCID: PMC8264592 DOI: 10.3389/fonc.2021.669746] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 06/03/2021] [Indexed: 12/11/2022] Open
Abstract
Cancer is a leading contributor to deaths worldwide. Surgery is the primary treatment for resectable cancers. Nonetheless, it also results in inflammatory response, angiogenesis, and stimulated metastasis. Local anesthetic lidocaine can directly and indirectly effect different cancers. The direct mechanisms are inhibiting proliferation and inducing apoptosis via regulating PI3K/AKT/mTOR and caspase-dependent Bax/Bcl2 signaling pathways or repressing cytoskeleton formation. Repression invasion, migration, and angiogenesis through influencing the activation of TNFα-dependent, Src-induced AKT/NO/ICAM and VEGF/PI3K/AKT signaling pathways. Moreover, the indirect influences are immune regulation, anti-inflammation, and postoperative pain relief. This review summarizes the latest evidence that revealed potential clinical benefits of lidocaine in cancer treatment to explore the probable molecular mechanisms and the appropriate dose.
Collapse
Affiliation(s)
- Caihui Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cuiyu Xie
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yao Lu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Ambulatory Surgery Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
25
|
Wang Z, Zhang J, Wen Y, Wang P, Fan L. Bupivacaine inhibits the malignant biological behavior of oral squamous cell carcinoma cells by inhibiting the activation of ERK1/2 and STAT3. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:839. [PMID: 34164473 PMCID: PMC8184410 DOI: 10.21037/atm-21-1414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Background Oral squamous cell carcinoma (OSCC) is an aggressive malignant tumor. Bupivacaine (Bupi), a local anesthetic drug, has been shown to display anti-tumor activity against a variety of tumors. Methods We selected OSCC CAL-27 cells as the in vitro model. Cell toxicity, proliferation, apoptosis, and stemness were conducted, respectively. The protein levels of Ki67, PCNA, caspase-3, caspase-9, survivin, SOX2, NANOG, OCT4, STAT3, p-STAT3, ERK1/2, and p-ERK1/2 were evaluated by western blotting. Male BALB/c nude mice xenograft model was used to evaluate the effect of Bupi on tumor growth in vivo. Results Compared with the control group, Bupi (0.2, 0.5, or 1 µm) significantly decreased the cell viability and the proliferation of CAL-27 cells. Meanwhile, Bupi significantly promoted apoptosis of CAL-27 cells compared with the control group. Additionally, Bupi inhibited the stemness of CAL-27 cells which was evidenced by a sphere formation assay. Bupi decreased the phosphorylation level of STAT3 and ERK1/2 in a dose-dependent manner. The addition of interferon-γ (IFN-γ, 20 ng/mL) in the experiment verified the role of Bupi on STAT3 and ERK1/2 signaling. In vivo, Bupi (40 µmol/kg) obviously suppressed the weight and size of the xenograft tumor, the number of apoptotic cells and Ki67+ decreased. Also, Bupi treatment inhibited the expression of stem-like marker proteins. Conclusions Bupi could be used as an anticancer drug against the growth and stemness ability of OSCC. The underlying mechanism may be due to down-regulation of STAT3 and ERK1/2 signaling. This study provides a new insight for the application of Bupi.
Collapse
Affiliation(s)
- Zhongchao Wang
- Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, China.,Department of Periodontics & Oral Medicine, The Affiliated Hospital of Stomatology of Southwest Medical University, Luzhou, China
| | - Jie Zhang
- Medical Administration Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yongmei Wen
- Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, China.,Department of Prosthodontics, The Affiliated Hospital of Stomatology of Southwest Medical University, Luzhou, China
| | - Pin Wang
- Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, China.,Department of Prosthodontics, The Affiliated Hospital of Stomatology of Southwest Medical University, Luzhou, China
| | - Liyuan Fan
- Oral & Maxillofacial Reconstruction and Regeneration Laboratory, Southwest Medical University, Luzhou, China.,Department of Prosthodontics, The Affiliated Hospital of Stomatology of Southwest Medical University, Luzhou, China
| |
Collapse
|
26
|
Gu JH, Liu CC, Xie JL, Ma B, Cui SM, Yang GZ, He SC. The Local Anesthetic Bupivacaine Inhibits the Progression of Non-Small Cell Lung Cancer by Inducing Autophagy Through Akt/mTOR Signaling. Front Oncol 2021; 11:616445. [PMID: 33777755 PMCID: PMC7991299 DOI: 10.3389/fonc.2021.616445] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/14/2021] [Indexed: 12/24/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a prevalent malignancy with high mortality and poor prognosis. Bupivacaine serves as a widely used local anesthetic and presents potential anti-tumor activity. Nevertheless, the function of bupivacaine in the NSCLC development remains elusive. Here, we tried to investigate the impact of bupivacaine on the NSCLC progression. Significantly, we revealed that bupivacaine was able to reduce the proliferation and induce the apoptosis of NSCLC cells. Bupivacaine could attenuate the invasion and migration in the cells. Mechanically, the treatment of bupivacaine increased the expression ratio of light chain 3B-II (LC3B-II)/LC3B-I and the expression of Beclin-1 in the NSCLC cells. Meanwhile, the expression of the autophagic adaptor protein p62 was decreased by bupivacaine treatment in the cells. The treatment of bupivacaine attenuated the phosphorylation of AKT and mTOR in the NSCLC cells. The AKT activator SC79 and autophagy inhibitor 3-methyladenine (3-MA) reversed the bupivacaine-inhibited phosphorylation of AKT and mTOR and bupivacaine-induced autophagy, as well as the bupivacaine-attenuated NSCLC progression in the cells. Bupivacaine could inhibit the tumor growth in vivo. In conclusion, we discovered that the local anesthetic bupivacaine inhibited the progression of NSCLC by inducing autophagy through Akt/mTOR signaling. Our finding provides new insights into the mechanism by which bupivacaine attenuates the development of NSCLC. Bupivacaine may serve as a potential anti-tumor candidate for the therapeutic strategy of NSCLC.
Collapse
Affiliation(s)
- Jian-Hua Gu
- Department of Anesthesia, Jinan People's Hospital, Jinan, China
| | - Cui-Cui Liu
- Department of Anesthesia, Jinan People's Hospital, Jinan, China
| | - Jin-Lan Xie
- Department of Anesthesia, Jinan People's Hospital, Jinan, China
| | - Bin Ma
- Department of Anesthesia, Jinan People's Hospital, Jinan, China
| | - Shao-Min Cui
- Department of Anesthesia, Jinan People's Hospital, Jinan, China
| | - Guang-Zhu Yang
- Department of Anesthesia, Jinan People's Hospital, Jinan, China
| | - Shun-Cheng He
- Department of Anesthesia, Jinan People's Hospital, Jinan, China
| |
Collapse
|
27
|
Neurotoxic Effects of Local Anesthetics on Developing Motor Neurons in a Rat Model. J Clin Med 2021; 10:jcm10050901. [PMID: 33668828 PMCID: PMC7956179 DOI: 10.3390/jcm10050901] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 11/16/2022] Open
Abstract
Neurotoxic effects of local anesthetics (LAs) on developing motor neurons have not been documented. We investigated the neurotoxic effects of LAs on developing motor neurons in terms of cell viability, cytotoxicity, reactive oxygen species (ROS), and apoptosis. Embryonic spinal cord motor neurons were isolated from Sprague-Dawley rat fetuses and exposed to one of the three LAs-lidocaine, bupivacaine, or ropivacaine-at concentrations of 1, 10, 100, or 1000 µM. The exposure duration was set to 1 or 24 h. The neurotoxic effects of LAs were determined by evaluating the following: cell viability, cytotoxicity, ROS production, and apoptosis. In the 1-h exposure group, the motor neurons exposed to lidocaine and bupivacaine had reduced cell viability and increased cytotoxicity, ROS, and apoptosis in a concentration-dependent manner. Lidocaine showed the highest toxicity, followed by bupivacaine. In the 24-h exposure group, all three LAs showed significant effects (decreased cell viability and increased cytotoxicity, ROS, and apoptosis) on the motor neurons in a concentration-dependent manner. The neurotoxic effects of lidocaine were greater than those of bupivacaine and ropivacaine. Ropivacaine appeared to have the least effect on motor neurons. This study identified the neurotoxic effects of lidocaine and bupivacaine on developing spinal cord motor neurons.
Collapse
|
28
|
Local Anesthetics and Recurrence after Cancer Surgery-What's New? A Narrative Review. J Clin Med 2021; 10:jcm10040719. [PMID: 33670434 PMCID: PMC7918400 DOI: 10.3390/jcm10040719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/27/2021] [Accepted: 02/08/2021] [Indexed: 12/16/2022] Open
Abstract
The perioperative use of regional anesthesia and local anesthetics is part of almost every anesthesiologist’s daily clinical practice. Retrospective analyses and results from experimental studies pointed towards a potential beneficial effect of the local anesthetics regarding outcome—i.e., overall and/or recurrence-free survival—in patients undergoing cancer surgery. The perioperative period, where the anesthesiologist is responsible for the patients, might be crucial for the further course of the disease, as circulating tumor cells (shed from the primary tumor into the patient’s bloodstream) might form new micro-metastases independent of complete tumor removal. Due to their strong anti-inflammatory properties, local anesthetics might have a certain impact on these circulating tumor cells, either via direct or indirect measures, for example via blunting the inflammatory stress response as induced by the surgical stimulus. This narrative review highlights the foundation of these principles, features recent experimental and clinical data and provides an outlook regarding current and potential future research activities.
Collapse
|
29
|
Viability and intracellular nitric oxide generation in the umbilical cord blood CD34 +CD133 - and CD34 +CD133 + cell populations exposed to local anaesthetics. Cent Eur J Immunol 2021; 45:369-376. [PMID: 33658886 PMCID: PMC7882401 DOI: 10.5114/ceji.2020.103410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/09/2019] [Indexed: 11/17/2022] Open
Abstract
Local anesthetics (LAs) are capable of influencing cell viability in systemic immunity and may also modify metabolism of those present in umbilical cord blood (UCB) following obstetric neuraxial analgesia and anaesthesia. Data regarding UCB immature cells, important for the neonate and critical for putative UCB transplantations, are lacking. LAs are capable of stimulating intracellular nitric oxide (NO) in human neutrophils; no information is available concerning newly perpetuated cells and its potential association with viability. The study aimed at assessing the LAs influence on the cell viability and intracellular NO production by UCB CD34+CD133– and CD34+ CD133+ cell populations. Mononuclear cells separated from UCB samples (n = 19) were incubated with bupivacaine (0.0005, 0.005, 1 mM), lidocaine (0.002, 0.02, 4 mM), and ropivacaine (0.0007, 0.007, 1.4 mM) for 4 h. Flow cytometry was applied for the assessment of cell viability and intracellular NO generation in CD34+CD133– and CD34+CD133+ cell populations using annexinV/7-AAD and DAF-2DA stainings, respectively. CD34+CD133+ cells showed less pronounced late apoptosis and necrosis as compared to CD34+CD133-population. Intracellular NO generation was comparable between both cell populations studied. LAs neither influenced cell viability nor changed NO production in either population. LAs do not interfere with viability and intracellular NO generation in the UCB CD34+CD133– and CD34+CD133+ cell populations.
Collapse
|
30
|
Grandhi RK, Perona B. Mechanisms of Action by Which Local Anesthetics Reduce Cancer Recurrence: A Systematic Review. PAIN MEDICINE 2021; 21:401-414. [PMID: 31282958 DOI: 10.1093/pm/pnz139] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Surgery in concert with anesthesia is a key part of the management of advanced-stage cancers. Anesthetic agents such as opioids and volatile anesthetics have been shown to promote recurrence in preclinical models, whereas some animal models have shown that the use of lidocaine may be beneficial in reducing cancer recurrence. The purpose of this article is to review the current literature to highlight the mechanisms of action by which local anesthetics are thought to reduce cancer recurrence. METHODS A systematic review was conducted using the PubMed (1966 to 2018) electronic database. Search terms included "lidocaine," "ropivicaine," "procaine," "bupivicaine," "mepivicaine," "metastasis," "cancer recurrence," "angiogenesis," and "local anesthetics" in various combinations. The search yielded 146 total abstracts for initial review, 20 of which met criteria for inclusion. Theories for lidocaine's effect on cancer recurrence were recorded. All studies were reviewed using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses checklist. RESULTS Numerous mechanisms were proposed based on the local anesthetic used and the type of cancer. Mechanisms include those that are centered on endothelial growth factor receptor, voltage-gated sodium and calcium channels, transient receptor melanoplastin 7, hyperthermia, cell cycle, and demyelination. CONCLUSIONS In vivo models suggest that local anesthetic administration leads to reduced cancer recurrence. The etiology of this effect is likely multifactorial through both inhibition of certain pathways and direct induction of apoptosis, a decrease in tumor migration, and an association with cell cycle-mediated and DNA-mediated effects. Additional research is required to further define the clinical implications.
Collapse
Affiliation(s)
- Ravi K Grandhi
- Department of Anesthesiology, Maimonides Medical Center, Brooklyn, New York
| | - Barbara Perona
- Department of Anesthesiology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
31
|
Mirshahidi S, Shields TG, de Necochea-Campion R, Yuan X, Janjua A, Williams NL, Mirshahidi HR, Reeves ME, Duerksen-Hughes P, Zuckerman LM. Bupivacaine and Lidocaine Induce Apoptosis in Osteosarcoma Tumor Cells. Clin Orthop Relat Res 2021; 479:180-194. [PMID: 33009230 PMCID: PMC7899706 DOI: 10.1097/corr.0000000000001510] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 09/01/2020] [Indexed: 01/31/2023]
Abstract
BACKGROUND Osteosarcoma is the most common type of bone cancer in adolescents. There have been no significant improvements in outcomes since chemotherapy was first introduced. Bupivacaine and lidocaine have been shown to be toxic to certain malignancies. This study evaluates the effect of these medications on two osteosarcoma cell lines. QUESTIONS/PURPOSES (1) Does incubation of osteosarcoma cells with bupivacaine or lidocaine result in cell death? (2) Does this result from an apoptotic mechanism? (3) Is a specific apoptotic pathway implicated? METHODS Two cell lines were chosen to account for the inherent heterogeneity of osteosarcoma. UMR-108 is a transplantable cell line that has been used in multiple studies as a primary tumor. MNNG/HOS has a high metastatic rate in vivo. Both cell lines were exposed bupivacaine (0.27, 0.54, 1.08, 2.16, 4.33 and 8.66 mM) and lidocaine (0.66, 1.33, 5.33, 10.66, 21.32 and 42.64 mM) for 24 hours, 48 hours, and 72 hours. These concentrations were determined by preliminary experiments that found the median effective dose was 1.4 mM for bupivacaine and 7.0 mM for lidocaine in both cell lines. Microculture tetrazolium and colony formation assay determined whether cell death occurred. Apoptosis induction was evaluated by phase-contrast micrographs, flow cytometry, DNA fragmentation and reactive oxygen species (ROS). The underlying pathways were analyzed by protein electrophoresis and Western blot. All testing was performed in triplicate and compared with pH-adjusted controls. Quantitative results were analyzed without blinding. RESULTS Both medications caused cell death in a dose- and time-dependent manner. Exposure to bupivacaine for 24 hours reduced viability of UMR-108 cells by 6 ± 0.75% (95% CI 2.9 to 9.11; p = 0.01) at 1.08 mM and 89.67 ± 1.5% (95% CI 82.2 to 95.5; p < 0.001) at 2.16 mM. Under the same conditions, MNNG/HOS viability was decreased in a similar fashion. After 24 hours, the viability of UMR-108 and MNNG/HOS cells exposed to 5.33 mM of lidocaine decreased by 25.33 ± 8.3% (95% CI 2.1 to 48.49; p = 0.03) and 39.33 ± 3.19% (95% CI 30.46 to 48.21; p < 0.001), respectively, and by 90.67 ± 0.66% (95% CI 88.82 to 92.52; p < 0.001) and 81.6 ± 0.47% (95% CI 79.69 to 82.31; p < 0.001) at 10.66 mM, respectively. After 72 hours, the viability of both cell lines was further reduced. Cell death was consistent with apoptosis based on cell morphology, total number of apoptotic cells and DNA fragmentation. The percentage increase of apoptotic UMR-108 and MNNG/HOS cells confirmed by Annexin-V positivity compared with controls was 21.3 ± 2.82 (95% CI 16.25 to 26.48; p < 0.001) and 21.23 ± 3.23% (95% CI 12.2 to 30.2; p = 0.003) for bupivacaine at 1.08 mM and 25.15 ± 4.38 (95% CI 12.9 to 37.3; p = 0.004) and 9.11 ± 1.74 (95% CI 4.35 to 13.87; p = 0.006) for lidocaine at 5.33 mM. The intrinsic apoptotic pathway was involved as the expression of Bcl-2 and survivin were down-regulated, and Bax, cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase-1 were increased. ROS production increased in the UMR-108 cells but was decreased in the MNNG/HOS cells. CONCLUSION These findings provide a basis for evaluating these medications in the in vivo setting. Studies should be performed in small animals to determine if clinically relevant doses have a similar effect in vivo. In humans, biopsies could be performed with standard doses of these medications to see if there is a difference in biopsy tract contamination on definitive resection. CLINICAL RELEVANCE Bupivacaine and lidocaine could potentially be used for their ability to induce and enhance apoptosis in local osteosarcoma treatment. Outcome data when these medications are used routinely during osteosarcoma treatment can be evaluated compared with controls. Further small animal studies should be performed to determine if injection into the tumor, isolated limb perfusion, or other modalities of treatment are viable.
Collapse
Affiliation(s)
- Saied Mirshahidi
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Troy G Shields
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Rosalia de Necochea-Campion
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Xiangpeng Yuan
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Ata Janjua
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Nadine L Williams
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Hamid R Mirshahidi
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Mark E Reeves
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Penelope Duerksen-Hughes
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Lee M Zuckerman
- S. Mirshahidi, R. de Necochea-Campion, A. Janjua, Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Medicine and Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA
- T. G. Shields, N. L. Williams, Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA
- X. Yuan, Department of Otolaryngology-Head and Neck Surgery, Loma Linda University Medical Center, Loma Linda, CA, USA Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
- H. R. Mirshahidi Department of Medical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- M. E. Reeves Department of Surgical Oncology, Loma Linda University Medical Center, Loma Linda, CA, USA
- P. Duerksen-Hughes, Department of Biochemistry, Loma Linda University Medical Center, Loma Linda, CA, USA
- L. M. Zuckerman, Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
32
|
Bruse LM. CORR Insights®: Bupivacaine and Lidocaine Induce Apoptosis in Osteosarcoma Tumor Cells. Clin Orthop Relat Res 2021; 479:195-197. [PMID: 33284231 PMCID: PMC7899593 DOI: 10.1097/corr.0000000000001566] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 10/12/2020] [Indexed: 01/31/2023]
Affiliation(s)
- Laura Marie Bruse
- L. M. Bruse, Orthopaedic Surgeon, Beautiful Bones Orthopaedics, Henderson, NV, USA
| |
Collapse
|
33
|
Woo MS, Park J, Ok SH, Park M, Sohn JT, Cho MS, Shin IW, Kim YA. The proper concentrations of dextrose and lidocaine in regenerative injection therapy: in vitro study. Korean J Pain 2021; 34:19-26. [PMID: 33380564 PMCID: PMC7783851 DOI: 10.3344/kjp.2021.34.1.19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/18/2020] [Accepted: 10/07/2020] [Indexed: 12/17/2022] Open
Abstract
Background Prolotherapy is a proliferation therapy as an alternative medicine. A combination of dextrose solution and lidocaine is usually used in prolotherapy. The concentrations of dextrose and lidocaine used in the clinical field are very high (dextrose 10%-25%, lidocaine 0.075%-1%). Several studies show about 1% dextrose and more than 0.2% lidocaine induced cell death in various cell types. We investigated the effects of low concentrations of dextrose and lidocaine in fibroblasts and suggest the optimal range of concentrations of dextrose and lidocaine in prolotherapy. Methods Various concentrations of dextrose and lidocaine were treated in NIH-3T3. Viability was examined with trypan blue exclusion assay and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Migration assay was performed for measuring the motile activity. Extracellular signal-regulated kinase (Erk) activation and protein expression of collagen I and α-smooth muscle actin (α-SMA) were determined with western blot analysis. Results The cell viability was decreased in concentrations of more than 5% dextrose and 0.1% lidocaine. However, in the concentrations 1% dextrose (D1) and 0.01% lidocaine (L0.01), fibroblasts proliferated mildly. The ability of migration in fibroblast was increased in the D1, L0.01, and D1 + L0.01 groups sequentially. D1 and L0.01 increased Erk activation and the expression of collagen I and α-SMA and D1 + L0.01 further increased. The inhibition of Erk activation suppressed fibroblast proliferation and the synthesis of collagen I. Conclusions D1, L0.01, and the combination of D1 and L0.01 induced fibroblast proliferation and increased collagen I synthesis via Erk activation.
Collapse
Affiliation(s)
- Min Seok Woo
- Department of Convergence Medical Science, Gyeongsang National University, Jinju, Korea
| | - Jiyoung Park
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - Seong-Ho Ok
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon, Korea.,Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Miyeong Park
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon, Korea
| | - Ju-Tae Sohn
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, Jinju, Korea.,Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Man Seok Cho
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Il-Woo Shin
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Hospital, Jinju, Korea.,Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Korea
| | - Yeon A Kim
- Department of Anesthesiology and Pain Medicine, Gyeongsang National University Changwon Hospital, Changwon, Korea.,Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Korea
| |
Collapse
|
34
|
Liu R, Wu M, Xu G, Ju L, Xiao J, Zhong W, He X, Yang Y. Ropivacaine inhibits proliferation, migration, and invasion while inducing apoptosis of glioma cells by regulating the SNHG16/miR-424-5p axis. Open Life Sci 2020; 15:988-999. [PMID: 33817285 PMCID: PMC7874551 DOI: 10.1515/biol-2020-0108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Regional anesthesia has anti-proliferative and pro-apoptotic effects in various cancers. Therefore, the purpose of this study was to investigate the effects of ropivacaine on the proliferation, migration, invasion, and apoptosis of glioma cells in vitro. METHODS Under ropivacaine stimulation conditions, proliferation, apoptosis, migration, and invasion of glioma cells were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazol-3-ium bromide (MTT), flow cytometry, and transwell assays, respectively. Western blot assay was employed to measure the protein expression levels in glioma cells. The expression levels of small nucleolar RNA host gene 16 (SNHG16) and miR-424-5p were assessed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The interaction relationship between SNHG16 and miR-424-5p was predicted and confirmed using a bioinformatics database and dual-luciferase reporter, RNA immunoprecipitation (RIP) and RNA pull-down assays. RESULTS After treatment with ropivacaine, proliferation, migration, and invasion were repressed while apoptosis was enhanced in glioma cells in a dose-depended manner. In addition, ropivacaine impeded SNHG16 expression in glioma cells. Importantly, overexpression of SNHG16 abolished the ropivacaine-induced effects on glioma cells. Analogously, knockdown of miR-424-5p counteracted the function of ropivacaine in glioma cells. We also found that SNHG16 bound to miR-424-5p and negatively regulated miR-424-5p expression in glioma cells. The rescue experiments indicated that ropivacaine might regulate glioma progression by targeting the SNHG16/miR-424-5p axis. CONCLUSION Our findings revealed the anti-tumor effects of ropivacaine in glioma by targeting the SNHG16/miR-424-5p axis. These data might extend the understanding of regulatory mechanisms by which ropivacaine could suppress glioma development.
Collapse
Affiliation(s)
- Rong Liu
- Department of Anesthesiology, The 908th Hospital of Chinese PLA Logistical Support Force, No.4, Hudong Road, Yuehu District, Yingtan 335000, Jiangxi, China
| | - Min Wu
- Department of Anesthesiology, The 908th Hospital of Chinese PLA Logistical Support Force, No.4, Hudong Road, Yuehu District, Yingtan 335000, Jiangxi, China
| | - Guiju Xu
- Department of Anesthesiology, The 908th Hospital of Chinese PLA Logistical Support Force, No.4, Hudong Road, Yuehu District, Yingtan 335000, Jiangxi, China
| | - Lu Ju
- Department of Anesthesiology, The 908th Hospital of Chinese PLA Logistical Support Force, No.4, Hudong Road, Yuehu District, Yingtan 335000, Jiangxi, China
| | - Jinhui Xiao
- Department of Anesthesiology, The 908th Hospital of Chinese PLA Logistical Support Force, No.4, Hudong Road, Yuehu District, Yingtan 335000, Jiangxi, China
| | - Wei Zhong
- Department of Internal Medicine, Ruijin Hospital of traditional Chinese Medicine, Ruijin, Jiangxi, 342500, China
| | - Xiao He
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Gannan Medical University, No.128, Jinling Road, Golden Development Zone, Ganzhou, Jiangxi, 341000, China
| | - Yan Yang
- Department of Anesthesiology, Ruijin Maternal and Child Health Hospital, Ruijin, Jiangxi, 342500, China
| |
Collapse
|
35
|
Chen D, Yan Y, Xie J, Pan J, Chen Y, Li Q, Yuan Y, Zeng W, Xing W. Amide-type local anesthetics may suppress tumor cell proliferation and sensitize Human Hepatocellular Carcinoma Cells to Cisplatin via upregulation of RASSF1A expression and demethylation. J Cancer 2020; 11:7312-7319. [PMID: 33193895 PMCID: PMC7646167 DOI: 10.7150/jca.46630] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/10/2020] [Indexed: 12/18/2022] Open
Abstract
Background: It has been reported that local anesthetics are toxic to various types of cells. Furthermore, several local anesthetics have been confirmed to exert demethylation effects and regulate the proliferation of human cancer cells. Our previous findings suggest that lidocaine may exert potential antitumor activity and enhance the sensitivity of cisplatin to hepatocellular carcinoma in vitro and in vivo. A recent study proved that lidocaine sensitizes breast cancer cells to cisplatin via upregulation of RASSF1A, a promotor of tumor suppressive gene (TSG) demethylation. We sought to determine whether amide-type local anesthetics (lidocaine, ropivacaine and bupivacaine) exert growth-inhibitory effects on human hepatoma cells and to determine whether amide-type local anesthetics sensitize human hepatoma cells to cisplatin-mediated cytotoxicity via upregulation of RASSF1A expression. Methods: Human hepatoma cell lines HepG2 and BEL-7402 were incubated with lidocaine, ropivacaine and bupivacaine. The viability of local anesthetic-treated cells with or without cisplatin was investigated. Further, we evaluated RASSF1A expression after treatment of HepG2 and BEL-7402 cells with three local anesthetics and determined the influence of RASSF1A expression on the toxicity of cisplatin to these cells. Results: The viability of HepG2 and BEL-7402 cells was significantly decreased by treatment with amide-type local anesthetics (lidocaine, ropivacaine and bupivacaine). In these cells, the combination treatment with cisplatin and local anesthetics exhibited a stronger reduction in viability. Lidocaine, ropivacaine and bupivacaine promoted a significant increase in RASSF1A expression and a decrease in RASSF1A methylation. The combined treatment with both local anesthetics and cisplatin resulted in a significantly lower level of HepG2 and BEL-7402 cell viability than that with singular local anesthetics or cisplatin treatment. Moreover, local anesthetics enhanced the cytotoxicity of cisplatin against HepG2 and BEL-7402 cells, accompanied by an increase in RASSF1A expression. Conclusions: These data indicated that amide-type local anesthetics (lidocaine, ropivacaine and bupivacaine) have growth-inhibitory and demethylation effects in human hepatoma cells. We also found that these amide local anesthetics may enhance the cytotoxicity of cisplatin in human hepatocellular carcinoma cells possibly via upregulation of RASSF1A expression and demethylation.
Collapse
Affiliation(s)
- Dongtai Chen
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yan Yan
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.,Department of Anesthesiology, Huizhou Municipal Central Hospital, Huizhou 516001, China
| | - Jingdun Xie
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jiahao Pan
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yonghua Chen
- Department of Anesthesiology, Peking University Shenzhen Hospital, Shenzhen 518000, China
| | - Qiang Li
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yunfei Yuan
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Weian Zeng
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Wei Xing
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
36
|
Fan X, Wang D, Chen X, Wang R. Effects of Anesthesia on Postoperative Recurrence and Metastasis of Malignant Tumors. Cancer Manag Res 2020; 12:7619-7633. [PMID: 32922072 PMCID: PMC7457832 DOI: 10.2147/cmar.s265529] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/04/2020] [Indexed: 01/17/2023] Open
Abstract
It is difficult to control the recurrence and metastasis of malignant tumors; furthermore, anesthesia is considered one of the main influencing factors. There has been increasing clinical attention on the effects of anesthetic drugs and methods on postoperative tumor growth and metastasis. We reviewed the effects of anesthesia on tumor recurrence and metastasis; specifically, the effects of anesthetic agents, anesthesia methods, and related factors during the perioperative period on the tumor growth and metastasis were analyzed. This study can provide reference standards for rational anesthesia formulations and cancer-related pain analgesia protocols for surgical procedures in patients with malignant tumors. Moreover, it contributes toward an experimental basis for the improvement and development of novel anesthetic agents and methods.
Collapse
Affiliation(s)
- Xiaoqing Fan
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui 230001, People's Republic of China.,Department of Anesthesiology, Anhui Provincial Hospital, Hefei 230001, Anhui, People's Republic of China
| | - Delong Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui 230001, People's Republic of China.,Department of Anesthesiology, Anhui Provincial Hospital, Hefei 230001, Anhui, People's Republic of China
| | - Xueran Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, People's Republic of China.,Department of Molecular Pathology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, People's Republic of China
| | - Ruiting Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, Anhui 230001, People's Republic of China.,Department of Anesthesiology, Anhui Provincial Hospital, Hefei 230001, Anhui, People's Republic of China
| |
Collapse
|
37
|
Levobupivacaine inhibits proliferation and promotes apoptosis of breast cancer cells by suppressing the PI3K/Akt/mTOR signalling pathway. BMC Res Notes 2020; 13:386. [PMID: 32807213 PMCID: PMC7430121 DOI: 10.1186/s13104-020-05191-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/16/2020] [Indexed: 12/16/2022] Open
Abstract
Objective This study aimed to test the hypothesis that levobupivacaine has anti-tumour effects on breast cancer cells. Results Colony formation and transwell assay were used to determine breast cancer cells proliferation. Flow Cytometry (annexin V and PI staining) was used to investigate breast cancer cells apoptosis. The effects of levobupivacaine on cellular signalling and molecular response were studied with Quantitative Polymerase Chain Reaction and western blot. Induction of apoptosis was confirmed by cell viability, morphological changes showed cell shrinkage, rounding, and detachments from plates. The results of the western blot and Quantitative Polymerase Chain Reaction indicated activation of active caspase-3 and inhibition of FOXO1. The results of the flow Cytometry confirmed that levobupivacaine inhibited breast cancer cell proliferation and enhanced apoptosis of breast cancer cells. Quantitative Polymerase Chain Reaction and Western blot analysis showed increased p21 and decreased cyclin D. Quantitative Polymerase Chain Reaction and western blot analysis showed that levobupivacaine significantly increased Bax expression, accompanied by a significant decreased Bcl-2 expression and inhibition of PI3K/Akt/mTOR signalling pathway. These findings suggested that levobupivacaine inhibits proliferation and promotes breast cancer cells apoptosis in vitro.
Collapse
|
38
|
Çömez MS, Borazan Y, Özgür T, İşler CT, Cellat M, Güvenç M, Altuğ ME. Effects of Dexamethasone on Bupivacaine-Induced Peripheral Nerve Injection Injury in the Rat Sciatic Model. J INVEST SURG 2020; 34:1339-1347. [PMID: 32781870 DOI: 10.1080/08941939.2020.1805053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION The aim of this study was to investigate the effect of perineural dexamethasone against intraneural bupivacaine. MATERIAL AND METHODS Rats were divided into 9 groups with 6 animals in each group; Group 1 (Intraneural saline 600 µL-2ndday), Group 2 (Intraneural saline 600 µL-7th day), Group 3 (Intraneural saline 600 µL + perineural dexamethasone 0.5 mg/kg-2nd day), Group 4 (Intraneural saline 600 µL + perineural dexamethasone 0.5 mg/kg-7th day), Group 5 (Intraneural bupivacaine 10 mg/kg-2nd day), Group 6 (Intranueral bupivacaine 10 mg/kg-7th day), Group 7 (Intraneural bupivacaine 10 mg/kg + perineurald exam ethasone 0.5 mg/kg-2nd day), Group 8 (Intraneural bupivacaine 10 mg/kg + perineural dexamethasone 0.5 mg/kg-7th day), Group 9 (Control group). At the end of the application period, histopathological and immunohistochemical examinations were analyzed. RESULTS AND CONCLUSION It was observed that caspase 3 levels significantly increased in the 5th and 6th groups compared to the 1st and 2nd groups (p < 0.01). However, in the 7th and 8th groups, these levels were similar with 1st and 2nd groups. While a significant decrease in S 100 levels was detected in group 6 (p < 0.05), a significant increase occurred in Group 8 and reached the same levels as Group 2. According to histopathological evaluation, edema, vacuolization and myelin degeneration were significantly increased in groups 5 and 6 (p < 0.05). However, in the 8th group, the mentioned data showed a significant decrease and reached the same levels as group 2. As a result, perineural dexamethasone was found to have protective effects against intraneural bupivacaine induced sciatic nerve damage.
Collapse
Affiliation(s)
- Mehmet Selim Çömez
- Department of Anesthesiology and Reanimation Tayfur Ata Sökmen Faculty of Medicine, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Yakup Borazan
- Department of Anhestesiology and Reanimation, Training and Research Hospital, Adiyaman University, Adıyaman, Turkey
| | - Tümay Özgür
- Department of Pathology, Tayfur Ata Sökmen Faculty of Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
| | - Cafer Tayer İşler
- Department of Surgery, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
| | - Mustafa Cellat
- Department of Physiology, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
| | - Mehmet Güvenç
- Department of Physiology, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
| | - Muhammed Enes Altuğ
- Department of Surgery, Faculty of Veterinary Medicine, Hatay Mustafa Kemal University, Antakya, Turkey
| |
Collapse
|
39
|
Muñoz-Leyva F, Cubillos J, Chin KJ. Managing rebound pain after regional anesthesia. Korean J Anesthesiol 2020; 73:372-383. [PMID: 32773724 PMCID: PMC7533186 DOI: 10.4097/kja.20436] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Rebound pain after regional anesthesia can be defined as transient acute postoperative pain that ensues following resolution of sensory blockade, and is clinically significant, either with regard to the intensity of pain or the impact on psychological well-being, quality of recovery, and activities of daily living. Current evidence suggests that it represents an unmasking of the expected nociceptive response in the absence of adequate systemic analgesia, rather than an exaggerated hyperalgesic phenomenon induced by local anesthetic neural blockade. In the majority of patients, it does not appear to significantly impact cumulative postoperative opioid consumption, quality of recovery, or patient satisfaction, and is not associated with longer-term sequelae such as persistent post-surgical pain. Nevertheless, it must be considered whenever regional anesthesia is incorporated into perioperative management. Strategies to mitigate the impact of rebound pain include routine prescribing of a systemic multimodal analgesic regimen, as well as patient education on appropriate expectations regarding block offset and expected surgical pain, and timely initiation of analgesic medication. Prolonging the duration of action of regional anesthesia with continuous catheter techniques or local anesthetic adjuncts may also help alleviate rebound pain, although further research is required to confirm this.
Collapse
Affiliation(s)
- Felipe Muñoz-Leyva
- Department of Anesthesiology and Pain Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Javier Cubillos
- Department of Anesthesia and Perioperative Medicine, University Hospital, London Health Sciences Center, Western University, London, ON, Canada
| | - Ki Jinn Chin
- Department of Anesthesiology and Pain Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
Zuckerman LM, Frames WL, Mirshahidi HR, Williams NL, Shields TG, Otoukesh S, Mirshahidi S. Antiproliferative effect of bupivacaine on patient-derived sarcoma cells. Mol Clin Oncol 2020; 13:7. [PMID: 32754321 PMCID: PMC7393627 DOI: 10.3892/mco.2020.2077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Sarcomas are rare tumors with limited treatment options. Although chemotherapy is standard for certain subtypes, overall survival has not improved in several decades. Bupivacaine has been shown to induce apoptosis and prevent cell growth in multiple different types of malignancies but has not been studied in sarcoma. The current study evaluated the effects of bupivacaine on multiple patient-derived sarcoma cells and a commercial sarcoma cell line. Multiple patient-derived sarcoma cell subtypes and a commercial synovial cell sarcoma cell line were exposed to bupivacaine for different durations and at different concentrations. The patient-derived cells included a high-grade conventional osteosarcoma, a high-grade undifferentiated pleomorphic sarcoma of bone, and a high-grade synovial sarcoma. Flow cytometry and an MTT assay were used to evaluate whether a treatment effect was observed. Treatment of all the subtypes of sarcomas in this study with bupivacaine demonstrated a time- and dose-dependent increase in apoptosis and decrease in cell viability. A cell viability assay demonstrated that the IC50 was between 0.04 and 0.05% and that the treatment effect occurred at clinically relevant doses in vitro. Bupivacaine was toxic to both the patient-derived cells and the commercial cell line at doses commonly used in the clinical setting. These findings provide a foundation for further in vivo studies to evaluate whether these effects will translate to the clinical setting. Although further research is necessary, bupivacaine shows promise as not only an adjunct for pain management but as a treatment modality for sarcoma.
Collapse
Affiliation(s)
- Lee M Zuckerman
- Department of Surgery, Division of Orthopaedic Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - William L Frames
- Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | - Hamid R Mirshahidi
- Division of Hematology and Oncology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | - Nadine L Williams
- Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | - Troy G Shields
- Department of Orthopaedic Surgery, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | - Salman Otoukesh
- Division of Hematology and Oncology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| | - Saied Mirshahidi
- Loma Linda Cancer Center Biospecimen Laboratory, Loma Linda University Medical Center, Loma Linda, CA 92354, USA
| |
Collapse
|
41
|
Wang X, Yao Y, Gao J. Sevoflurane inhibits growth factor-induced angiogenesis through suppressing Rac1/paxillin/FAK and Ras/Akt/mTOR. Future Oncol 2020; 16:1619-1627. [PMID: 32479124 DOI: 10.2217/fon-2020-0221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: We investigated the direct effects of sevoflurane on angiogenesis and a variety of tumor cells. Materials & methods: The antiangiogenic activity of sevoflurane was determined using angiogenesis and biochemical assays. Results: Sevoflurane at low doses inhibits capillary network formation. Sevoflurane inhibited VEGF- and bFGF-stimulated migration, adhesion and growth in endothelial cells and induced apoptosis. Sevoflurane only at high doses inhibited growth and migration of tumor cells, suggesting differential effects of sevoflurane between endothelial and tumor cells. Mechanistically, sevoflurane decreased growth factors-induced Ras and Rac1 activation, and suppressed Ras and Rac1 signaling. Conclusion: We demonstrate the antiangiogenic effects of sevoflurane and provide preclinical evidence into the potential mechanisms by which sevoflurane may negatively affect cancer growth and metastasis.
Collapse
Affiliation(s)
- Xuesong Wang
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts & Science, Xiangyang, Hubei Province 441021, PR China
| | - Yao Yao
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts & Science, Xiangyang, Hubei Province 441021, PR China
| | - Jin Gao
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts & Science, Xiangyang, Hubei Province 441021, PR China
| |
Collapse
|
42
|
Zhu G, Zhang L, Dan J, Zhu Q. Differential effects and mechanisms of local anesthetics on esophageal carcinoma cell migration, growth, survival and chemosensitivity. BMC Anesthesiol 2020; 20:126. [PMID: 32450791 PMCID: PMC7249391 DOI: 10.1186/s12871-020-01039-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/17/2020] [Indexed: 12/29/2022] Open
Abstract
Background Retrospective analysis and pre-clinical studies suggest that local anesthetics have anti-tumoral effects. However, the association between cancer recurrence and the use of local anesthesia is inconclusive and most reports are based on single local anesthetic results. Methods The biological effects (growth, migration and survival) of four common local anesthetics on esophageal carcinoma cells were compared. Biochemical assays on molecules involved in cell migration and proliferation were analyzed. Results Ropivacaine and bupivacaine significantly inhibited esophageal carcinoma cell migration, at clinically relevant micromolar concentrations. Mepivacaine and lidocaine showed less potent cell migration inhibition than ropivacaine or bupivacaine. All four local anesthetics inhibited cell proliferation. Of note, the effective concentration of anti-proliferative activities requires higher doses. At millimolar concentrations of these local anesthetics, cell apoptosis was moderately affected. Drug combination analysis demonstrated that two of four local anesthetics augmented chemotherapeutic drugs in inhibiting migration. However, all four local anesthetics significantly augmented chemotherapeutic drugs in inhibiting growth and inducing apoptosis. The anti-growth and anti-survival effects of four local anesthetics were attributed to mitochondrial dysfunction and oxidative damage. The anti-migratory effect of local anesthetics is likely through decreasing Rac1 activity. Conclusions Our work demonstrates the differential effects and proposes the mechanisms of local anesthetics on esophageal carcinoma cell migration, growth, survival and chemosensitivity.
Collapse
Affiliation(s)
- Guanhua Zhu
- Department of Anesthesiology, Jingzhou Central Hospital, Jingzhou, Hubei Province, China
| | - Ling Zhang
- Department of Anesthesiology, Jingzhou Central Hospital, Jingzhou, Hubei Province, China
| | - Jiapeng Dan
- Department of Anesthesiology, Jingzhou Central Hospital, Jingzhou, Hubei Province, China
| | - Qiong Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Yangtze University, Hangkong Road 8, Jingzhou, 434020, Hubei Province, China.
| |
Collapse
|
43
|
Qin A, Liu Q, Wang J. Ropivacaine inhibits proliferation, invasion, migration and promotes apoptosis of papillary thyroid cancer cells via regulating ITGA2 expression. Drug Dev Res 2020; 81:700-707. [PMID: 32314406 DOI: 10.1002/ddr.21671] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/13/2020] [Accepted: 03/14/2020] [Indexed: 12/24/2022]
Abstract
The present study aimed to investigate the roles of Ropivacaine in papillary thyroid cancer (PTC) and identify the possible mechanisms. The expression of integrin alpha-2 (ITGA2) in TC cell lines was tested using Western blotting and RT-qPCR. Subsequently, the level of ITGA2 in human PTC cell line (TPC-1) was measured following intervention with a series of concentrations of Ropivacaine. Then, cell counting kit-8 (CCK-8) assay and colony formation assay were executed for detecting proliferation of cells after transfection with ITGA2 pcDNA3.1. The expression of proliferation-related protein was determined by Western blotting. Additionally, the abilities of TPC-1 cell invasion and migration were examined using Transwell assay and scratch wound healing assay. Apoptosis of TPC-1 cells was analyzed using TUNEL assay and the expressions of apoptosis-related proteins were tested via West blotting. The results suggested that ITGA2 was highly expressed in TC cell lines, especially in TPC-1 cells. Ropivacaine decreased the expression of ITGA2 in a dose-dependent manner. Moreover, after treatment with Ropivacaine, cell proliferation was inhibited accompanied by changes of proliferation-related protein expressions, which was reversed following co-transfection with ITGA2 pcDNA3.1. Furthermore, Ropivacaine concentration-dependently suppressed invasion and migration of TPC-1 cells, whereas these inhibitory effects were attenuated after ITGA2 overexpression. Furthermore, apoptosis was promoted, coupled with a decrease of Bcl-2 expression and increases of Bax, cleaved caspase-3 and cleaved caspase-9 expression, in Ropivacaine-treated TPC-1 cells, which was restored following ITGA2 overexpression. These findings demonstrated that Ropivacaine could suppress proliferation, invasion, migration, and accelerate apoptosis of PTC cells via regulating ITGA2 expression.
Collapse
Affiliation(s)
- Aichun Qin
- Department of Anesthesiology, Hubei Cancer Hospital, Wuhan, China
| | - Qiong Liu
- Otolaryngological Department, General Hospital of the Central Theater of the People's Liberation Army of China, Wuhan, China
| | - Jingfang Wang
- Department of Anesthesiology, Zhejiang Sian International Hospital, Jiaxing, China
| |
Collapse
|
44
|
Zhu Q, Zhu G, Xu W, Dan J, Xia R, Liu W. Bupivacaine inhibits angiogenesis through oxidative stress‐dependent inhibition of Akt/mTOR and activation of AMPK. Fundam Clin Pharmacol 2020; 34:581-590. [PMID: 32145095 DOI: 10.1111/fcp.12554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/19/2020] [Accepted: 03/04/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Qiong Zhu
- Department of Anesthesiology The First Affiliated Hospital of Yangtze University Jingzhou Hubei Province China
| | - Guanhua Zhu
- Department of Anesthesiology Jingzhou Central Hospital Jingzhou Hubei Province China
| | - Wei Xu
- Department of Anesthesiology The First Affiliated Hospital of Yangtze University Jingzhou Hubei Province China
| | - Jiapeng Dan
- Department of Anesthesiology Jingzhou Central Hospital Jingzhou Hubei Province China
| | - Rui Xia
- College of Clinical Medicine Yangtze University Jingzhou Hubei Province China
| | - Weiwei Liu
- College of Clinical Medicine Yangtze University Jingzhou Hubei Province China
| |
Collapse
|
45
|
de Solorzano IO, Prieto M, Mendoza G, Sebastian V, Arruebo M. Triggered drug release from hybrid thermoresponsive nanoparticles using near infrared light. Nanomedicine (Lond) 2020; 15:219-234. [DOI: 10.2217/nnm-2019-0270] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim: Developing hybrid poly(N-isopropylacrylamide)-based nanogels decorated with plasmonic hollow gold nanoparticles for on-demand drug delivery and their physico-chemical characterization, bupivacaine loading and release ability upon light irradiation, and in vitro cell viability. Materials & methods: Hollow gold nanoparticles were prepared by galvanic replacement reaction; poly(N-isopropylacrylamide)-based nanogels were synthesized via precipitation polymerization and their electrostatic coupling was accomplished using poly(allylamine hydrochloride) as cationic polyelectrolyte linker. Results & conclusion: Colloidal stability of the resulted hybrid nanovectors was demonstrated under physiological conditions together with their fast response and excellent heating efficiency after light stimulation, indicating their potential use as triggered drug-delivery vectors. Moreover, their influence on cell metabolism and cell cycle under subcytotoxic doses were studied showing excellent cytocompatibility.
Collapse
Affiliation(s)
- Isabel Ortiz de Solorzano
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018-Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials & Nanomedicine, CIBER-BBN, 28029-Madrid, Spain
| | - Martin Prieto
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018-Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Gracia Mendoza
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018-Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Victor Sebastian
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018-Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials & Nanomedicine, CIBER-BBN, 28029-Madrid, Spain
| | - Manuel Arruebo
- Department of Chemical Engineering, Aragon Institute of Nanoscience (INA), University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018-Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Networking Research Center on Bioengineering, Biomaterials & Nanomedicine, CIBER-BBN, 28029-Madrid, Spain
| |
Collapse
|
46
|
Hsu YC, Lee JJ, Chien MN, Chen MJ, Leung CH, Cheng SP. Is papillary thyroid microcarcinoma a biologically different disease? A propensity score-matched analysis. J Surg Oncol 2019; 120:1023-1030. [PMID: 31407354 DOI: 10.1002/jso.25670] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/04/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Papillary thyroid microcarcinoma exhibits an indolent clinical course and could be a candidate for active surveillance in the appropriate setting. It remains unknown whether papillary microcarcinoma is biologically different from larger papillary carcinoma >1 cm. METHODS We analyzed clinicopathological information and transcriptome data of papillary thyroid cancer samples from The Cancer Genome Atlas. Propensity-score matching was used to construct a matched cohort consisting of 29 microcarcinomas and 58 carcinomas. Principal component analysis and unsupervised hierarchical cluster analysis were carried out to investigate the similarity of gene expression profiles. RESULTS After adjustment for differences in baseline clinicopathological and genetic factors, transcriptome could be grouped mainly on the basis of tumor class (BRAF-like vs RAS-like) and tumor size (microcarcinoma vs carcinoma). The gene set enrichment analysis showed that extracellular matrix-associated pathways were enriched in the MSigDB database. CONCLUSION Papillary thyroid microcarcinomas display a distinct gene expression pattern different from the corresponding carcinomas. We hypothesize that tumor microenvironment may play a role in the microcarcinoma/carcinoma phenotypic divergence.
Collapse
Affiliation(s)
- Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Jie-Jen Lee
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Ming-Nan Chien
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Ming-Jen Chen
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Hsiang Leung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
47
|
Jin H, Yu J. Lidocaine protects H9c2 cells from hypoxia-induced injury through regulation of the MAPK/ERK/NF-κB signaling pathway. Exp Ther Med 2019; 18:4125-4131. [PMID: 31641386 DOI: 10.3892/etm.2019.8055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 08/16/2019] [Indexed: 01/09/2023] Open
Abstract
The aim of the present study was to investigate the effect of Lidocaine on hypoxia-induced injury in cardiomyoblasts whilst exploring the associated molecular mechanism. In the present study, hypoxia was induced in H9c2 cells to establish an in vitro model of myocardial infarction. The cells were treated with lidocaine (0.5, 1, 5, 10 mM) for 48 h under hypoxic conditions. Cell viability and apoptosis levels were determined by MTT assay and flow cytometry, and ELISA was used to measure the levels of inflammatory cytokines released. A creatine kinase isoenzyme/cardiac troponin I detection kit was used to show that lidocaine significantly reduced hypoxia-induced cardiac troponin 1 and creatine kinase-muscle/brain release in a dose-dependent manner. Mitochondrial viability staining suggested that lidocaine significantly enhanced mitochondrial viability under hypoxic conditions. Lidocaine also significantly reduced hypoxia-induced apoptosis and increased H9c2 viability in a dose-dependent manner. Additionally, under hypoxic conditions, lidocaine dose-dependently promoted Bcl-2 expression, while decreasing Bax and caspase-3 expression in H9c2 cells. ELISA and reverse transcription quantitative PCR were used to detect the levels of tumor necrosis factor (TNF-α), interleukin (IL)-1β and IL-6 released by H9c2 cells. Results showed that lidocaine markedly reduced the hypoxia-induced expression levels of IL-1β, TNF-α and IL-6 in a dose-dependent manner. In addition, protein levels of phosphorylated (p)-ERK1/2 and NF-κB p-p65 were analyzed by western blotting, and results indicated that lidocaine significantly increased the protein levels of p-ERK1/2 and decreased the protein level of NF-κB p-p65 in a dose-dependent manner under hypoxic conditions. These data suggested that lidocaine might protect cardiomyoblasts from hypoxia-induced injury via activation of the mitogen activated protein kinase/ERK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Haibin Jin
- Department of Cardiology, Tianjin Hospital, Tianjin 300211, P.R. China
| | - Jin Yu
- Department of Anesthesiology, People's Liberation Army 951 Hospital, Korla, Xinjiang 841000, P.R. China
| |
Collapse
|
48
|
Wall TP, Crowley PD, Sherwin A, Foley AG, Buggy DJ. Effects of Lidocaine and Src Inhibition on Metastasis in a Murine Model of Breast Cancer Surgery. Cancers (Basel) 2019; 11:cancers11101414. [PMID: 31546727 PMCID: PMC6826872 DOI: 10.3390/cancers11101414] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer recurs in 20% of patients following intended curative resection. In vitro data indicates that amide local anaesthetics, including lidocaine, inhibit cancer cell metastasis by inhibiting the tyrosine kinase enzyme Src. In a murine breast cancer surgery model, systemic lidocaine reduces postoperative pulmonary metastases. We investigated whether the additional administration of bosutinib (a known Src inhibitor) influences lidocaine’s observed beneficial effect in this in vivo model. Female BALB/c mice (n = 95) were inoculated with 25,000 4T1 cells into the mammary fad pad and after 7 days the resulting tumours were excised under sevoflurane anaesthesia. Experimental animals were randomized to one of four treatments administered intravenously prior to excision: lidocaine, bosutinib, both lidocaine and bosutinib in combination, or saline. Animals were euthanized 14 days post-surgery and lung and liver metastatic colonies were evaluated. Post-mortem serum was analysed for MMP-2 and MMP-9, pro-metastatic enzymes whose expression is influenced by the Src pathway. Lidocaine reduced lung, but not liver metastatic colonies versus sevoflurane alone (p = 0.041), but bosutinib alone had no metastasis-inhibiting effect. When combined with lidocaine, bosutinib reversed the anti-metastatic effect observed with lidocaine on sevoflurane anaesthesia. Only lidocaine alone reduced MMP-2 versus sevoflurane (p = 0.044). Both bosutinib (p = 0.001) and bosutinib/lidocaine combined (p = 0.001) reduced MMP-9 versus sevoflurane, whereas lidocaine alone did not. In a murine surgical breast cancer model, the anti-metastatic effects of lidocaine under sevoflurane anaesthesia are abolished by the Src inhibitor bosutinib, and lidocaine reduces serum MMP-2. These results suggest that lidocaine may act, at least partly, via an inhibitory effect on MMP-2 expression to reduce pulmonary metastasis, but whether this is due to an effect on Src or via another pathway remains unclear.
Collapse
Affiliation(s)
- Thomas P Wall
- Department of Anaesthesiology & Perioperative Medicine, Mater University Hospital, School of Medicine, University College Dublin, D07 KH4C Dublin, Ireland.
- Conway Institute for Biomolecular and Biomedical Research, School of Medicine University College Dublin, D04 V1W8 Dublin, Ireland.
| | - Peter D Crowley
- Conway Institute for Biomolecular and Biomedical Research, School of Medicine University College Dublin, D04 V1W8 Dublin, Ireland.
| | - Aislinn Sherwin
- Department of Anaesthesiology & Perioperative Medicine, Mater University Hospital, School of Medicine, University College Dublin, D07 KH4C Dublin, Ireland.
| | - Andrew G Foley
- Berand Neuropharmacology Ltd., NovaUCD, D04 V1W8 Dublin, Ireland.
| | - Donal J Buggy
- Department of Anaesthesiology & Perioperative Medicine, Mater University Hospital, School of Medicine, University College Dublin, D07 KH4C Dublin, Ireland.
- Conway Institute for Biomolecular and Biomedical Research, School of Medicine University College Dublin, D04 V1W8 Dublin, Ireland.
- Outcomes Research, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
49
|
Castelli V, Piroli A, Marinangeli F, d'Angelo M, Benedetti E, Ippoliti R, Zis P, Varrassi G, Giordano A, Paladini A, Cimini A. Local anesthetics counteract cell proliferation and migration of human triple-negative breast cancer and melanoma cells. J Cell Physiol 2019; 235:3474-3484. [PMID: 31541469 DOI: 10.1002/jcp.29236] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/10/2019] [Indexed: 12/21/2022]
Abstract
In different retrospective studies, a protective role of regional anesthetics in reducing cancer recurrence after surgery was indicated. Accordingly, it has been previously demonstrated a protective effect of anesthetics in breast cancer cells and in other types of cancer. On the other hand, how anesthetics influence cancer needs in-depth investigations. For this purpose, two different human cancer cell lines, MDA-MB-231, triple-negative breast cancer, and A375, melanoma, were used in this study. By means of Western blotting and immunofluorescence and terminal deoxynucleotidyl transferase dUTP nick end labeling analyses, the signal transduction pathways activated by the anesthetics, such as ropivacaine and levobupivacaine, were analyzed. The data obtained demonstrated that both anesthetics are able to counteract cell proliferation by positively modulating cell death signaling and by decreasing cell proliferation and survival pathways.
Collapse
Affiliation(s)
- Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alba Piroli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Franco Marinangeli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Panagiotis Zis
- Academic Department of Neurosciences, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.,Medical School, University of Cyprus, Nicosia, Cyprus
| | - Giustino Varrassi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Antonio Giordano
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, Pennsylvania.,Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Antonella Paladini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
50
|
Baiardo Redaelli M, Zangrillo A, Gregorc V, Ciceri F, Dagna L, Tshomba Y, Navalesi P, Landoni G. How to obtain severe hypoglycemia without causing brain or cardiac damage. Med Hypotheses 2019; 130:109276. [DOI: 10.1016/j.mehy.2019.109276] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/03/2019] [Accepted: 06/10/2019] [Indexed: 12/17/2022]
|