1
|
Zhao L, Liu P, Sauvat A, Carnet Le Provost K, Liu J, Checcoli A, Pol J, Kepp O, Kroemer G, Bezu L. Dexmedetomidine induces immunogenic cancer cell death and sensitizes tumors to PD-1 blockade. J Immunother Cancer 2025; 13:e010714. [PMID: 40480656 PMCID: PMC12142037 DOI: 10.1136/jitc-2024-010714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 05/14/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Local anesthetics promote anticancer immune responses. A machine learning-based algorithm trained with information on the biological effects and molecular descriptors of analgesics, anesthetics, hypnotics and opioids predicted antitumor effects for dexmedetomidine (DEX). DEX is a sedative acting as an alpha2-adrenoceptor (ADRA2) agonist. Based on these premises, we investigated the putative antineoplastic effects of DEX. RESULTS In vitro, DEX promoted premortem stresses such as autophagy and partial endoplasmic reticulum stress with the phosphorylation of eukaryotic initiation factor 2 alpha and the inhibition of the splicing of X-box binding protein 1. DEX elicited the biomarkers of immunogenic cell death, including the release of ATP and high-mobility group box 1 protein, and the cell surface exposure of calreticulin, enhancing the engulfment of malignant cells by dendritic cells. In immunocompetent mice, DEX decreased the progression of colorectal cancers, fibrosarcomas, mammary carcinomas and melanomas, as it improved overall survival. These effects were inhibited by the ADRA2 antagonist yohimbine, suggesting that DEX mediates its anticancer effects at least in part on-target. Depending on the specific tumor model, DEX also enhanced the cytotoxic T cell/regulatory T cell ratio in the tumor bed and draining lymph nodes. Programmed cell death protein 1 blockade tended to improve DEX effects. After rechallenge with antigenically identical cells, no tumor appeared, indicating the formation of immunological memory. CONCLUSIONS These results confirm the machine learning-predicted anticancer activity of DEX. Beyond its utility as a sedative agent in oncological intensive care, DEX may improve anticancer immunosurveillance and sensitize tumors to immune checkpoint blockade.
Collapse
Affiliation(s)
- Liwei Zhao
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Peng Liu
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Allan Sauvat
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Killian Carnet Le Provost
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Jiani Liu
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Andrea Checcoli
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Jonathan Pol
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Oliver Kepp
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Guido Kroemer
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| | - Lucillia Bezu
- INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France
| |
Collapse
|
2
|
Dautelle J, Saint-Pol AL, l'Hospital AE, Degryse C, de Courson H, Biais M. Evaluation of early-death predictive factors and assessment of the benefits-risks ratio among patients with vertebral metastases needing surgery. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2025:10.1007/s00586-025-08803-2. [PMID: 40261393 DOI: 10.1007/s00586-025-08803-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/15/2025] [Accepted: 03/17/2025] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Preoperative survival prognostication is the main factor to consider when selecting patients who will benefit from metastatic spine surgery. Since patients' selection for surgery is not fully standardized, we aim to identify clinical, oncological and biological markers associated with poor post-operative prognosis. METHODS A retrospective analysis was conducted in 105 patients referred to a specialized center for surgery of spinal metastasis between October 2020 and January 2022. Detailed clinical and biological variables were collected. Univariate and multivariate analyses were performed to assess overall survival. RESULTS Among the 105 patients included, One-third died within six months. Multivariate survival analysis identified age, Karnofsky score, hyperalgesia and C-reactive protein (CRP) as independently associated with shorter survival. Patients with CRP values above 100 mg/L had a 57% mortality rate at three months. Hyperalgesic patients had a higher mortality rate at three months (p = 0.047), and preoperative use of strong opioids was significantly associated with lower rates of pain relief (p = 0.042). However, pre-operative neurological deficits did not significantly impact survival prognosis (p = 0.92) and were fully corrected by surgery in half of the cases. CONCLUSION Incorporating biological markers of inflammation, particularly CRP levels above 100 mg/L, into pre-operative multidisciplinary discussions could improve patient selection for surgery. Hyperalgesic presentation should weigh against surgery, unlike incomplete neurological deficits. This study pointed new clinical and biological markers for short term post-operative survival. Further studies are needed to develop accurate prognosis models to help clinical decision-making.
Collapse
Affiliation(s)
- Johanna Dautelle
- Department of Anesthesiology and Critical Care, Hôpital Pellegrin, Université de Bordeaux, Bordeaux, France.
- Service d'Anesthésie Orthopédique, Hôpital Pellegrin, Pl. Amélie Raba Léon, Bordeaux, 33000, France.
| | - Anne-Laure Saint-Pol
- Department of Anesthesiology and Critical Care, Hôpital Pellegrin, Université de Bordeaux, Bordeaux, France
| | - Aude Etienne l'Hospital
- Department of Anesthesiology and Critical Care, Hôpital Pellegrin, Université de Bordeaux, Bordeaux, France
| | - Cécile Degryse
- Department of Anesthesiology and Critical Care, Hôpital Pellegrin, Université de Bordeaux, Bordeaux, France
| | - Hugues de Courson
- Department of Anesthesiology and Critical Care, Hôpital Pellegrin, Université de Bordeaux, Bordeaux, France
| | - Matthieu Biais
- Department of Anesthesiology and Critical Care, Hôpital Pellegrin, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
3
|
Cui J, Ma N, Li X, Chen X, Zhang J, Zhang W, Li H. Morphine Contributes to Epithelial-Mesenchymal Transition in Triple-Negative Breast Cancer Cells by Blocking COX-2 Methylation via Regulating the miR-23a-3p/DNMT3A Feedback. Cell Biochem Biophys 2025:10.1007/s12013-025-01749-8. [PMID: 40227561 DOI: 10.1007/s12013-025-01749-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
To investigate the effects and mechanisms of morphine on epithelial-mesenchymal transformation (EMT) in triple-negative breast cancer (TNBC). The levels of miR-23a-3p, DNMT3A, and COX-2 in tumor tissues from metastatic TNBC patients treated with morphine were assessed using qRT-PCR. Functional assays assessed morphine's impact on TNBC cell malignancy. Dual luciferase reporter and RNA pull-down assays investigated the interaction between miR-23a-3p and DNMT3A. miR-23a-3p inhibitor and DNMT3A siRNA were transfected into TNBC cells. Protein expression was analyzed by Western blot. Methylation status of miR-23a-3p and COX-2 was assessed via methylation-specific PCR. Rescue experiments were performed to research whether morphine modulates EMT in TNBC through COX-2 methylation regulation via the miR-23a-3p/DNMT3A feedback loop. The effects of morphine on TNBC in nude mice xenotransplantation were studied. In metastatic TNBC patients treated with morphine, miR-23a-3p and COX-2 expression were elevated, and DNMT3A levels were reduced. In TNBC cells, morphine enhanced migration, invasion, and EMT, and suppressed apoptosis. It upregulated miR-23a-3p and COX-2; downregulated DNMT3A; and inhibited methylation of miR-23a-3p and COX-2. miR-23a-3p directly inhibited DNMT3A expression. In morphine-treated TNBC cells, silencing DNMT3A reduced methylation of miR-23a-3p and COX-2. miR-23a-3p inhibitor suppressed migration, invasion, and EMT, and promoted apoptosis; however, these effects were reversed by DNMT3A silencing. In vivo, morphine promoted tumor EMT and metastasis in TNBC; reduced miR-23a-3p and COX-2 methylation; and decreased DNMT3A expression. Morphine accelerated EMT in TNBC by inhibiting COX-2 methylation through the miR-23a-3p/DNMT3A loop.
Collapse
Affiliation(s)
- Jian Cui
- Department of Anaesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Nina Ma
- Department of Anaesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Xiaohui Li
- Department of Anaesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Xuexin Chen
- Department of Anaesthesiology and Perioperative Medicine, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Junxia Zhang
- Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Wenjuan Zhang
- Ningxia Medical University, Yinchuan, Ningxia Province, China
| | - Hong Li
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Province, China.
| |
Collapse
|
4
|
Bezu L, Forget P, Hollmann MW, Parat MO, Piegeler T. Potential influence of different peri-operative analgesic regimens on tumour biology and outcome after oncologic surgery: A narrative review. Eur J Anaesthesiol 2025; 42:233-243. [PMID: 39743967 DOI: 10.1097/eja.0000000000002118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The management of peri-operative pain is one of the pillars of anaesthesia and is of particular importance in patients undergoing surgery for solid malignant tumours. Amongst several options, the most commonly employed analgesic regimens involve opioids, NSAIDs and regional anaesthesia techniques with different local anaesthetics. In recent years, several research reports have tried to establish a connection between peri-operative anaesthesia care and outcome after cancer surgery. Experimental studies have indicated that certain pain management substances may influence cancer progression, mainly by modifying the tumour's response to surgical stress and peri-operative inflammation. However, these promising in-vitro and in-vivo data have yet to be confirmed by randomised clinical trials. The reason for this might lie with the nature of tumour biology itself, and in the diversity of patient and tumour phenotypes. In a translational approach, future research should therefore concentrate on patient and tumour-related factors or biomarkers, which might either influence the tumour and its microenvironment or predict potential responses to interventions, including the choice of the analgesic. This might not only be relevant for the daily practice of clinical anaesthesia, but would also be of great importance for patients undergoing cancer surgery, who might be able to receive an individualised anaesthetic regimen based on their phenotypic profile.
Collapse
Affiliation(s)
- Lucillia Bezu
- From the Département d'Anesthésie, Chirurgie et Interventionnel (LB), U1138 Metabolism, Cancer and Immunity, Gustave Roussy, Villejuif, France (LB), Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA (LB), Aberdeen Centre for Arthritis and Musculoskeletal Health (Epidemiology Group), Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition (PF), Anaesthesia department, NHS Grampian, Aberdeen, UK (PF), IMAGINE UR UM 103, Montpellier University, Anesthesia Critical Care, Emergency and Pain Medicine Division, Nîmes University Hospital, Nîmes, France (PF), Pain and Opioids after Surgery (PANDOS) European Society of Anaesthesiology and Intensive Care (ID ESAIC_RG_PAND) Research Group, Brussels, Belgium (PF), Department of Anaesthesiology, Amsterdam UMC, Amsterdam, The Netherlands (MWH), School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba Qld, Australia (M-OP), Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany (TP), EuroPeriscope, ESAIC Onco-Anaesthesiology Research Group, Brussels, Belgium (TP, LB, PF, MWH)
| | | | | | | | | |
Collapse
|
5
|
Bezu L, Rahmani LS, Buggy DJ. The effect of the type of anaesthesia on long-term outcomes after cancer resection surgery: a narrative review. Anaesthesia 2025; 80:179-187. [PMID: 39777733 DOI: 10.1111/anae.16464] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND The peri-operative period may create a biological environment conducive to cancer cell survival and dissemination. Microscopic residual tumours (micrometastases) can be dislodged even with excellent surgical technique. At the same time, the stress response from surgery can temporarily impair immune function and activate inflammatory processes, increasing the risk of tumour proliferation. METHODS This narrative review investigates how peri-operative anaesthetic and analgesic interventions may influence cancer recurrence and metastasis by exploring evidence from both experimental and clinical studies. A comprehensive review of the literature was conducted to identify relevant preclinical and clinical studies published. RESULTS While surgery remains the best curative option for many cancers, metastasis remains the leading cause of death. Numerous studies have suggested that different anaesthetic drugs and techniques could impact cancer outcomes including volatile anaesthetic agents; total intravenous anaesthesia with propofol; local anaesthetics; regional anaesthesia; and opioids. This review focuses on these five commonly used anaesthetic approaches and evaluates their potential impact on cancer progression. DISCUSSION There is a complex interplay between anaesthetic and analgesic techniques and cancer outcomes. Despite promising data from laboratory experimental models, the balance of available clinical trials indicates an equivalent influence of all evaluated anaesthetic techniques on long-term oncologic outcomes, except, possibly, for peritumoral or intraperitoneal local anaesthetic infiltration.
Collapse
Affiliation(s)
- Lucillia Bezu
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group ESA_IC_RG_EP, Brussels, Belgium
- Département d'Anesthésie, Chirurgie et Interventionnel, Gustave Roussy, France
| | - Lua S Rahmani
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group ESA_IC_RG_EP, Brussels, Belgium
- Department of Anaesthesiology, Mater University Hospital, School of Medicine, University College Dublin, Ireland
| | - Donal J Buggy
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group ESA_IC_RG_EP, Brussels, Belgium
- Department of Anaesthesiology, Mater University Hospital, School of Medicine, University College Dublin, Ireland
| |
Collapse
|
6
|
Wu Y, Yuan X, Zhang Y, Ma F, Zhao W, Sun X, Ma X, Chen Y. Sialidase NEU3 silencing inhibits angiogenesis of EA.hy926 cells by regulating Wnt/β-catenin signaling pathway. Biochem Biophys Res Commun 2025; 742:151098. [PMID: 39672004 DOI: 10.1016/j.bbrc.2024.151098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/15/2024]
Abstract
Angiogenesis significantly drives tumor progression, and the functions of vascular endothelial cells are influenced by various factors. Tumor cells are characterized by abnormal sialylation, and their dynamic balance depends on sialyltransferases and sialidases. NEU3 is a plasma membrane-associated sialidase, vital for the regulation of cell surface sialylation. Our study revealed that, NEU3 is the most abundantly expressed among the four sialidase subtypes in EA.hy926 cells. Silencing NEU3 expression resulted in cell apoptosis and reduced proliferation, highlighting its crucial function in the regulation of cell activity. Subsequent experiments using transwell and tube formation assays demonstrated that the inhibition of NEU3 expression suppressed cell migration and angiogenesis. RNA sequencing analysis further elucidated that altering NEU3 expression in EA.hy926 cells impacts the Wnt/β-Catenin signaling pathway and c-Myc levels, thereby modulating cellular survival and migration capacity and exerting a regulatory effect on angiogenesis. These findings suggest that targeting NEU3 in the vascular endothelium may represent a promising strategy for anti-angiogenic therapy in tumors.
Collapse
Affiliation(s)
- Yilun Wu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xin Yuan
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yi Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fang Ma
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wei Zhao
- Affiliated Hospital of Chengdu University, Chengdu, Sichuan, 637000, China
| | - Xinrui Sun
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xue Ma
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yingjiao Chen
- Office for West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, 610021, China.
| |
Collapse
|
7
|
Zhu Y, Gong Y, Wang Y, Jiang Z, Yao Y, Miao X, Wang S, Zhang Y, Cao J. Flurbiprofen axetil is involved in basal-like breast cancer metastasis via suppressing the MEK/ERK signaling pathway. Cell Biol Int 2025; 49:68-78. [PMID: 39364685 DOI: 10.1002/cbin.12251] [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: 10/22/2023] [Revised: 07/12/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024]
Abstract
Flurbiprofen axetil is commonly utilized in clinical practice as one of the nonsteroidal anti-inflammatory drugs (NSAIDs) and is included in multimodal analgesia regimens postbreast cancer surgery. Numerous NSAIDs have been studied for their potential to both promote and inhibit cancer. Given the variability in their effects on tumors, further investigation into the specific role of flurbiprofen axetil is warranted. Therefore, the primary objective of this study was to assess the impact of flurbiprofen axetil on basal-like breast cancer (BLBC) metastasis and elucidate the underlying molecular mechanisms involved. The BLBC metastasis mouse model was established by caudal vein injection of tumor cells. The lung metastasis of breast cancer in mice and the effect of flurbiprofen axetil were assessed by in vivo bioluminescence imaging, hematoxylin and eosin staining and immunohistochemistry. In vitro, the results of flurbiprofen axetil on the proliferation, migration, and invasion of MDA-MB-231 human breast cancer cells and BT-549 human breast cancer cells were assessed by colony formation assay and transwell assay. The effects of flurbiprofen axetil on several tumor metastasis-related signaling pathway proteins were examined by western blot, and the reversal extent of the flurbiprofen axetil effect by Ro 67-7476 (ERK phosphorylation agonist) was detected by transwell assay. The results showed that flurbiprofen axetil significantly inhibited BLBC lung metastasis in mice. Flurbiprofen axetil similarly inhibited breast cancer cell migration and invasion in vitro but did not affect their proliferation. Mechanistic investigations have revealed that flurbiprofen axetil exerts a noteworthy inhibitory influence on the MEK/ERK pathway while exhibiting no significant alteration in the expression of other pathway proteins intricately associated with epithelial-mesenchymal transition. In conclusion, the inhibitory effect of flurbiprofen axetil on BLBC metastasis is characterized by its selectivity in targeting the MEK/ERK signaling pathway rather than exerting a broad impact on the global signaling pathway.
Collapse
Affiliation(s)
- Yalin Zhu
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, China
- Changhai Hospital, Faculty of Anesthesiology, Naval Medical University, Shanghai, China
- Department of Anesthesiology, Naval Hospital of Eastern Theater, Zhoushan, China
| | - Yi Gong
- Department of Respiratory Diseases and Critical Medicine, Quzhou Hospital Affiliated to Wenzhou Medical University, Quzhou, Zhejiang, China
- Department of Respiratory Diseases and Critical Medicine, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Yifei Wang
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, China
| | - Zhengyu Jiang
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, China
- Changhai Hospital, Faculty of Anesthesiology, Naval Medical University, Shanghai, China
| | - Ying Yao
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, China
| | - Xiaoyong Miao
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, China
| | - Shuoer Wang
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Zhang
- Changhai Hospital, Faculty of Anesthesiology, Naval Medical University, Shanghai, China
| | - Jianping Cao
- Department of Anesthesiology, Naval Medical Center, Naval Medical University, Shanghai, China
| |
Collapse
|
8
|
Koochaki R, Amini E, Zarehossini S, Zareh D, Haftcheshmeh SM, Jha SK, Kesharwani P, Shakeri A, Sahebkar A. Alkaloids in Cancer therapy: Targeting the tumor microenvironment and metastasis signaling pathways. Fitoterapia 2024; 179:106222. [PMID: 39343104 DOI: 10.1016/j.fitote.2024.106222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024]
Abstract
The use of phytomedicine in cancer therapy is a growing field of research that takes use of the medicinal properties of plant-derived compounds. Under the domain of cancer therapy and management, alkaloids, a prominent group of natural compounds, have showed significant potential. Alkaloids often affect a wide range of essential cellular mechanisms involved in cancer progression. These multi-targeting capabilities, can give significant advantages to alkaloids in overcoming resistance mechanisms. For example, berberine, an alkaloid found in Berberis species, is widely reported to induce apoptosis by activating caspases and regulating apoptotic pathways. Notably, alkaloids like as quinine have showed promise in inhibiting the formation of new blood vessels required for tumor growth. In addition, alkaloids have shown anti-proliferative and anticancer properties mostly via modulating key signaling pathways involved in metastasis, including those regulating epithelial-mesenchymal transition. This work provides a comprehensive overview of naturally occurring alkaloids that exhibit anticancer properties, with a specific emphasis on their underlying molecular mechanisms of action. Furthermore, many methods to modify previously reported difficult physicochemical properties using nanocarriers in order to enhance its systemic bioavailability have been discussed as well. This study also includes information on newly discovered alkaloids that are now being studied in clinical trials for their potential use in cancer treatment. Further, we have also briefly mentioned on the application of high-throughput screening and molecular dynamics simulation for acceleration on the identification of potent alkaloids based compounds to target and treat cancer.
Collapse
Affiliation(s)
- Raoufeh Koochaki
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Elaheh Amini
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Sara Zarehossini
- Department of Cell & Molecular Biology (genetic), Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Danial Zareh
- Department of Cell & Molecular Biology (genetic), Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Abolfazl Shakeri
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran..
| |
Collapse
|
9
|
Wang R, Li S, Wang B, Wang G, Zheng H. Impact of opioids and mu-opioid receptors on oncologic metastasis. Am J Cancer Res 2024; 14:4236-4247. [PMID: 39417177 PMCID: PMC11477826 DOI: 10.62347/scls3277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/22/2024] [Indexed: 10/19/2024] Open
Abstract
Opioids are the most effective and widely used treatments for acute and chronic pain in patients with cancer. This review focuses on the impact of opioids and mu-opioid receptors (MORs) on the stages of oncologic metastasis. Studies have shown that opioids can facilitate tumor progression and are related to a poor prognosis in patients with cancer. As the primary receptor for opioids, MORs play a significant role in regulating malignant tumor transformation and are involved in processes, such as proliferation, angiogenesis, epithelial-mesenchymal transition (EMT), circulating tumor cells (CTCs) and the tumor microenvironment (TME). While clinical trials have investigated the relationship between opioids and patient prognosis, further research is needed to clarify the relationship between opioids, MORs and metastasis.
Collapse
Affiliation(s)
- Runjia Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan, Shandong, China
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| | - Bomin Wang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan, Shandong, China
| | - Gongming Wang
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinan, Shandong, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China
| |
Collapse
|
10
|
Zhu X, Yi H, Gu J, Liu S, Hayashi K, Ikegami D, Pardo M, Toborek M, Roy S, Li H, Levitt RC, Hao S. Sirtuin 3 Mediated by Spinal cMyc-Enhancer of Zeste Homology 2 Pathway Plays an Important Role in Human Immunodeficiency Virus-Related Neuropathic Pain Model. Anesth Analg 2024; 139:647-659. [PMID: 38446700 DOI: 10.1213/ane.0000000000006873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
BACKGROUND Clinical data demonstrate that chronic use of opioid analgesics increases neuropathic pain in people living with human immunodeficiency virus (HIV). Therefore, it is important to elucidate the molecular mechanisms of HIV-related chronic pain. In this study, we investigated the role of the transcription factor cMyc, epigenetic writer enhancer of zeste homology 2 (EZH2), and sirtuin 3 (Sirt3) pathway in HIV glycoprotein gp120 with morphine (gp120M)-induced neuropathic pain in rats. METHODS Neuropathic pain was induced by intrathecal administration of recombinant gp120 with morphine. Mechanical withdrawal threshold was measured using von Frey filaments, and thermal latency using the hotplate test. Spinal expression of cMyc, EZH2, and Sirt3 were measured using Western blots. Antinociceptive effects of intrathecal administration of antisense oligodeoxynucleotide against cMyc, a selective inhibitor of EZH2, or recombinant Sirt3 were tested. RESULTS In the spinal dorsal horn, gp120M upregulated expression of cMyc (ratio of gp120M versus control, 1.68 ± 0.08 vs 1.00 ± 0.14, P = .0132) and EZH2 (ratio of gp120M versus control, 1.76 ± 0.05 vs 1.00 ± 0.16, P = .006), and downregulated Sirt3 (ratio of control versus gp120M, 1.00 ± 0.13 vs 0.43 ± 0.10, P = .0069) compared to control. Treatment with intrathecal antisense oligodeoxynucleotide against cMyc, GSK126 (EZH2 selective inhibitor), or recombinant Sirt3 reduced mechanical allodynia and thermal hyperalgesia in this gp120M pain model. Knockdown of cMyc reduced spinal EZH2 expression in gp120M treated rats. Chromatin immunoprecipitation (ChIP) assay showed that enrichment of cMyc binding to the ezh2 gene promoter region was increased in the gp120M-treated rat spinal dorsal horn, and that intrathecal administration of antisense ODN against cMyc (AS-cMyc) reversed the increased enrichment of cMyc. Enrichment of trimethylation of histone 3 on lysine residue 27 (H3K27me3; an epigenetic mark associated with the downregulation of gene expression) binding to the sirt3 gene promoter region was upregulated in the gp120M-treated rat spinal dorsal horn; that intrathecal GSK126 reversed the increased enrichment of H3K27me3 in the sirt3 gene promoter. Luciferase reporter assay demonstrated that cMyc mediated ezh2 gene transcription at the ezh2 gene promoter region, and that H3K27me3 silenced sirt3 gene transcription at the gene promoter region. CONCLUSION These results demonstrated that spinal Sirt3 decrease in gp120M-induced neuropathic pain was mediated by cMyc-EZH2/H3K27me3 activity in an epigenetic manner. This study provided new insight into the mechanisms of neuropathic pain in HIV patients with chronic opioids.
Collapse
Affiliation(s)
- Xun Zhu
- From the Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
- Department of Anesthesiology, the 6th Affiliated Hospital of Guangzhou Medical University and Qing Yuan People's Hospital, Qingyuan, Guangdong Province, China
| | - Hyun Yi
- From the Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Jun Gu
- From the Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Shue Liu
- From the Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Kentaro Hayashi
- From the Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Daigo Ikegami
- From the Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Marta Pardo
- From the Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Sabita Roy
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Heng Li
- Department of Anesthesiology, the 6th Affiliated Hospital of Guangzhou Medical University and Qing Yuan People's Hospital, Qingyuan, Guangdong Province, China
| | - Roy C Levitt
- From the Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida
- John T. MacDonald Foundation, Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida
| | - Shuanglin Hao
- From the Department of Anesthesiology, University of Miami Miller School of Medicine, Miami, Florida
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida
- Miami VA Healthcare System, Miami, Florida
| |
Collapse
|
11
|
Kuoch C, Bezu L. Impact of Surgical and Anesthetic Procedures after Colorectal Cancer Surgery: A Propensity Score-Matched Cohort Study (The PROCOL Study). MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1362. [PMID: 39202644 PMCID: PMC11356255 DOI: 10.3390/medicina60081362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/10/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024]
Abstract
Background: Surgical inflammatory pain decreases the innate and adaptive immune antitumor response and favors residual circulating tumor cells. Objectives: This study investigated whether minimally invasive surgeries (laparoscopic and robotic procedures), which are less painful and inflammatory, improved oncological outcomes after colorectal resection compared to laparotomy. Methods: This research was a single-center propensity score-matched study involving patients who underwent colectomy and rectum resection from July 2017 to December 2019. Results: Seventy-four laparotomies and 211 minimally invasive procedures were included. Minimally invasive procedures were associated with less blood loss (0 mL vs. 75 mL, p < 0.001), shorter length of stay (8 days vs. 12 days, p < 0.001), and fewer complications at 3 months (11.8% vs. 29.4%, p = 0.02) compared to laparotomies. No difference in overall survival (OS) and recurrence-free survival (RFS) at 3 years between groups was observed. Univariate Cox regression analyses demonstrated that age and ASA > 3 can negatively impact OS, while adjuvant chemotherapy can positively influence OS. pT3-T4 stage and postoperative pain could negatively influence RFS. Multivariate Cox regression analyses concluded that age (HR 1.08, p < 0.01) and epidural analgesia (HR 0.12, p = 0.03) were predictors for OS. Lidocaine infusion (HR 0.39, p = 0.04) was a positive predictor for RFS. Conclusions: Minimally invasive procedures reduce postoperative complications and shorten the length of hospital stay compared to major surgeries without improving prognosis. However, the administration of local anesthetics through neuraxial anesthesia or intravenous infusion could improve survival and decrease the occurrence of relapses.
Collapse
Affiliation(s)
- Céline Kuoch
- Département d’Anesthésie, Chirurgie et Interventionnel, Gustave Roussy, FR-94805 Villejuif, France
| | - Lucillia Bezu
- Département d’Anesthésie, Chirurgie et Interventionnel, Gustave Roussy, FR-94805 Villejuif, France
- U1138 Metabolomics and Cell Biology Platforms, Gustave Roussy, Université Paris Saclay, FR-94805 Villejuif, France
- EuroPeriscope Group, ESA-IC, Onco-Anesthesiology Research Group, B-1000 Brussels, Belgium
| |
Collapse
|
12
|
Bezu L, Akçal Öksüz D, Bell M, Buggy D, Diaz-Cambronero O, Enlund M, Forget P, Gupta A, Hollmann MW, Ionescu D, Kirac I, Ma D, Mokini Z, Piegeler T, Pranzitelli G, Smith L, The EuroPeriscope Group. Perioperative Immunosuppressive Factors during Cancer Surgery: An Updated Review. Cancers (Basel) 2024; 16:2304. [PMID: 39001366 PMCID: PMC11240822 DOI: 10.3390/cancers16132304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
Surgical excision of the primary tumor represents the most frequent and curative procedure for solid malignancies. Compelling evidence suggests that, despite its beneficial effects, surgery may impair immunosurveillance by triggering an immunosuppressive inflammatory stress response and favor recurrence by stimulating minimal residual disease. In addition, many factors interfere with the immune effectors before and after cancer procedures, such as malnutrition, anemia, or subsequent transfusion. Thus, the perioperative period plays a key role in determining oncological outcomes and represents a short phase to circumvent anesthetic and surgical deleterious factors by supporting the immune system through the use of synergistic pharmacological and non-pharmacological approaches. In line with this, accumulating studies indicate that anesthetic agents could drive both protumor or antitumor signaling pathways during or after cancer surgery. While preclinical investigations focusing on anesthetics' impact on the behavior of cancer cells are quite convincing, limited clinical trials studying the consequences on survival and recurrences remain inconclusive. Herein, we highlight the main factors occurring during the perioperative period of cancer surgery and their potential impact on immunomodulation and cancer progression. We also discuss patient management prior to and during surgery, taking into consideration the latest advances in the literature.
Collapse
Affiliation(s)
- Lucillia Bezu
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Département d'Anesthésie, Chirurgie et Interventionnel, Gustave Roussy, 94805 Villejuif, France
- U1138 Metabolism, Cancer and Immunity, Gustave Roussy, 94805 Villejuif, France
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Dilara Akçal Öksüz
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Clinic for Anesthesiology, Intensive Care, Emergency Medicine, Pain Therapy and Palliative Medicine, Marienhaus Klinikum Hetzelstift, 67434 Neustadt an der Weinstrasse, Germany
- ESAIC Mentorship Program, BE-1000 Brussels, Belgium
| | - Max Bell
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Department of Perioperative Medicine and Intensive Care (PMI), Karolinska University Hospital, Solna, 17176 Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institute, 17176 Stockholm, Sweden
| | - Donal Buggy
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Division of Anaesthesiology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland
- School of Medicine, University College, D04 V1W8 Dublin, Ireland
| | - Oscar Diaz-Cambronero
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Department of Anesthesiology, Hospital Universitario y Politécnico la Fe, 46026 Valencia, Spain
- Perioperative Medicine Research, Health Research Institute Hospital la Fe, 46026 Valencia, Spain
- Faculty of Medicine, Department of Surgery, University of Valencia, 46010 Valencia, Spain
| | - Mats Enlund
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Center for Clinical Research, Uppsala University, SE-72189 Västerås, Sweden
- Department of Anesthesia & Intensive Care, Västmanland Hospital, SE-72189 Västerås, Sweden
| | - Patrice Forget
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Aberdeen Centre for Arthritis and Musculoskeletal Health (Epidemiology Group), Institute of Applied Health Sciences, Epidemiology Group, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZN, UK
- Department of Anaesthesia, NHS Grampian, University of Aberdeen, Aberdeen AB25 2ZN, UK
- Pain and Opioids after Surgery (PANDOS) ESAIC Research Group, European Society of Anaesthesiology and Intensive Care, 1000 Brussels, Belgium
- IMAGINE UR UM 103, Anesthesia Critical Care, Emergency and Pain Medicine Division, Nîmes University Hospital, Montpellier University, 30900 Nîmes, France
| | - Anil Gupta
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Department of Physiology and Pharmacology, Karolinska Institute, 17176 Stockholm, Sweden
| | - Markus W Hollmann
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Department of Anesthesiology, Amsterdam UMC, 1100 DD Amsterdam, The Netherlands
| | - Daniela Ionescu
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Department of Anesthesia and Intensive Care, University of Medicine and Pharmacy "Iuliu Hatieganu", 400012 Cluj-Napoca, Romania
- Outcome Research Consortium, Cleveland, OH 44195, USA
| | - Iva Kirac
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Genetic Counselling Unit, University Hospital for Tumors, Sestre Milosrdnice University Hospital Centre, 10000 Zagreb, Croatia
| | - Daqing Ma
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London SW10 9NH, UK
- Department of Anesthesiology, Perioperative and Systems Medicine Laboratory, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Zhirajr Mokini
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- ESAIC Mentorship Program, BE-1000 Brussels, Belgium
- Clinique du Pays de Seine, 77590 Bois le Roi, France
| | - Tobias Piegeler
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Department of Anesthesiology and Intensive Care, University of Leipzig Medical Center, 04275 Leipzig, Germany
| | - Giuseppe Pranzitelli
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Department of Anesthesiology and Intensive Care, San Timoteo Hospital, 86039 Termoli, Italy
| | - Laura Smith
- EuroPeriscope, ESA-IC Onco-Anaesthesiology Research Group, B-1000 Brussels, Belgium
- Department of Anaesthesia, NHS Grampian, University of Aberdeen, Aberdeen AB25 2ZN, UK
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZN, UK
| | | |
Collapse
|
13
|
Coutinho LL, Femino EL, Gonzalez AL, Moffat RL, Heinz WF, Cheng RYS, Lockett SJ, Rangel MC, Ridnour LA, Wink DA. NOS2 and COX-2 Co-Expression Promotes Cancer Progression: A Potential Target for Developing Agents to Prevent or Treat Highly Aggressive Breast Cancer. Int J Mol Sci 2024; 25:6103. [PMID: 38892290 PMCID: PMC11173351 DOI: 10.3390/ijms25116103] [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: 02/28/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Nitric oxide (NO) and reactive nitrogen species (RNS) exert profound biological impacts dictated by their chemistry. Understanding their spatial distribution is essential for deciphering their roles in diverse biological processes. This review establishes a framework for the chemical biology of NO and RNS, exploring their dynamic reactions within the context of cancer. Concentration-dependent signaling reveals distinctive processes in cancer, with three levels of NO influencing oncogenic properties. In this context, NO plays a crucial role in cancer cell proliferation, metastasis, chemotherapy resistance, and immune suppression. Increased NOS2 expression correlates with poor survival across different tumors, including breast cancer. Additionally, NOS2 can crosstalk with the proinflammatory enzyme cyclooxygenase-2 (COX-2) to promote cancer progression. NOS2 and COX-2 co-expression establishes a positive feed-forward loop, driving immunosuppression and metastasis in estrogen receptor-negative (ER-) breast cancer. Spatial evaluation of NOS2 and COX-2 reveals orthogonal expression, suggesting the unique roles of these niches in the tumor microenvironment (TME). NOS2 and COX2 niche formation requires IFN-γ and cytokine-releasing cells. These niches contribute to poor clinical outcomes, emphasizing their role in cancer progression. Strategies to target these markers include direct inhibition, involving pan-inhibitors and selective inhibitors, as well as indirect approaches targeting their induction or downstream effectors. Compounds from cruciferous vegetables are potential candidates for NOS2 and COX-2 inhibition offering therapeutic applications. Thus, understanding the chemical biology of NO and RNS, their spatial distribution, and their implications in cancer progression provides valuable insights for developing targeted therapies and preventive strategies.
Collapse
Affiliation(s)
- Leandro L. Coutinho
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
- Center for Translational Research in Oncology, ICESP/HC, Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, SP, Brazil;
| | - Elise L. Femino
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| | - Ana L. Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| | - Rebecca L. Moffat
- Optical Microscopy and Analysis Laboratory, Office of Science and Technology Resources, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA;
| | - William F. Heinz
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (W.F.H.); (S.J.L.)
| | - Robert Y. S. Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| | - Stephen J. Lockett
- Optical Microscopy and Analysis Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (W.F.H.); (S.J.L.)
| | - M. Cristina Rangel
- Center for Translational Research in Oncology, ICESP/HC, Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo 01246-000, SP, Brazil;
| | - Lisa A. Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| | - David A. Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA; (L.L.C.); (E.L.F.); (A.L.G.); (R.Y.S.C.)
| |
Collapse
|
14
|
Carnet Le Provost K, Kepp O, Kroemer G, Bezu L. Trial watch: beta-blockers in cancer therapy. Oncoimmunology 2023; 12:2284486. [PMID: 38126031 PMCID: PMC10732641 DOI: 10.1080/2162402x.2023.2284486] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
Compelling evidence supports the hypothesis that stress negatively impacts cancer development and prognosis. Irrespective of its physical, biological or psychological source, stress triggers a physiological response that is mediated by the hypothalamic-pituitary-adrenal axis and the sympathetic adrenal medullary axis. The resulting release of glucocorticoids and catecholamines into the systemic circulation leads to neuroendocrine and metabolic adaptations that can affect immune homeostasis and immunosurveillance, thus impairing the detection and eradication of malignant cells. Moreover, catecholamines directly act on β-adrenoreceptors present on tumor cells, thereby stimulating survival, proliferation, and migration of nascent neoplasms. Numerous preclinical studies have shown that blocking adrenergic receptors slows tumor growth, suggesting potential clinical benefits of using β-blockers in cancer therapy. Much of these positive effects of β-blockade are mediated by improved immunosurveillance. The present trial watch summarizes current knowledge from preclinical and clinical studies investigating the anticancer effects of β-blockers either as standalone agents or in combination with conventional antineoplastic treatments or immunotherapy.
Collapse
Affiliation(s)
- Killian Carnet Le Provost
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, 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, Institut Universitaire de France, 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, Institut Universitaire de France, 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, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Université Paris Saclay, Villejuif, France
- Gustave Roussy, Département d’anesthésie, Chirurgie et Interventionnel, Villejuif, France
| |
Collapse
|
15
|
Gao FY, Li XT, Xu K, Wang RT, Guan XX. c-MYC mediates the crosstalk between breast cancer cells and tumor microenvironment. Cell Commun Signal 2023; 21:28. [PMID: 36721232 PMCID: PMC9887805 DOI: 10.1186/s12964-023-01043-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 01/08/2023] [Indexed: 02/01/2023] Open
Abstract
The MYC oncogenic family is dysregulated in diverse tumors which is generally linked to the poor prognosis of tumors. The members in MYC family are transcription factors which are responsible for the regulation of various genes expression. Among them, c-MYC is closely related to the progression of tumors. Furthermore, c-MYC aberrations is tightly associated with the prevalence of breast cancer. Tumor microenvironment (TME) is composed of many different types of cellular and non-cellular factors, mainly including cancer-associated fibroblasts, tumor-associated macrophages, vascular endothelial cells, myeloid-derived suppressor cells and immune cells, all of which can affect the diagnosis, prognosis, and therapeutic efficacy of breast cancer. Importantly, the biological processes occurred in TME, such as angiogenesis, immune evasion, invasion, migration, and the recruition of stromal and tumor-infiltrating cells are under the modulation of c-MYC. These findings indicated that c-MYC serves as a critical regulator of TME. Here, we aimed to summarize and review the relevant research, thus to clarify c-MYC is a key mediator between breast cancer cells and TME. Video Abstract.
Collapse
Affiliation(s)
- Fang-yan Gao
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Xin-tong Li
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Kun Xu
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Run-tian Wang
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Xiao-xiang Guan
- grid.412676.00000 0004 1799 0784Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| |
Collapse
|
16
|
Bezu L, Kepp O, Kroemer G. Impact of local anesthetics on epigenetics in cancer. Front Oncol 2022; 12:849895. [PMID: 36110954 PMCID: PMC9468863 DOI: 10.3389/fonc.2022.849895] [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: 01/06/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Defective silencing of tumor suppressor genes through epigenetic alterations contributes to oncogenesis by perturbing cell cycle regulation, DNA repair or cell death mechanisms. Reversal of such epigenetic changes including DNA hypermethylation provides a promising anticancer strategy. Until now, the nucleoside derivatives 5-azacytidine and decitabine are the sole DNA methyltransferase (DNMT) inhibitors approved by the FDA for the treatment of specific hematological cancers. Nevertheless, due to their nucleoside structure, these inhibitors directly incorporate into DNA, which leads to severe side effects and compromises genomic stability. Much emphasis has been placed on the development of less toxic epigenetic modifiers. Recently, several preclinical studies demonstrated the potent epigenetic effects of local anesthetics, which are routinely used during primary tumor resection to relief surgical pain. These non-nucleoside molecules inhibit DNMT activity, affect the expression of micro-RNAs and repress histone acetylation, thus exerting cytotoxic effects on malignant cells. The in-depth mechanistic comprehension of these epigenetic effects might promote the use of local anesthetics as anticancer drugs.
Collapse
Affiliation(s)
- Lucillia Bezu
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, 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
- *Correspondence: Lucillia Bezu, ; Guido Kroemer,
| | - Oliver Kepp
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université de Paris, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Institut Universitaire de France, 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, Institut Universitaire de France, 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
- *Correspondence: Lucillia Bezu, ; Guido Kroemer,
| |
Collapse
|
17
|
Abujbarah SM, Jogerst K, Kosiorek HE, Ahmad S, Cronin PA, Casey W, Craner R, Rebecca A, Pockaj BA. Postoperative Hematomas in the Era of Outpatient Mastectomy: Is Ketorolac Really to Blame? Ann Surg Oncol 2022; 29:6395-6403. [PMID: 35849298 DOI: 10.1245/s10434-022-12141-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/16/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Enhanced recovery after surgery (ERAS) protocols following mastectomy with or without implant-based breast reconstruction (IBBR) include ketorolac for multimodal perioperative analgesia. There are concerns that ketorolac could be associated with increased risk of postoperative hematoma formation. METHODS Retrospective review of patients undergoing mastectomy with or without IBBR between January 2013 and December 2019 at a single institution. Patients received 15 mg, 30 mg, or no ketorolac depending on ERAS protocol adherence, patient characteristics, and surgeon preference. Clinically significant hematoma was defined as requiring surgical intervention on day of surgery or postoperative day 1. Patients were compared by demographics, surgical characteristics, ketorolac dose, and hematoma prevalence. Univariable and multivariable logistic regression evaluated hematoma formation odds. RESULTS Eight hundred patients met inclusion criteria: 477 received ketorolac. Those who received ketorolac were younger, had lower ASA scores, were more likely to have bilateral procedures and undergo concomitant IBBR, had longer operative times, were less likely to take antiplatelet or anticoagulation medications, had higher PACU pain scores, and had higher incidence of hematomas requiring surgical intervention. Of the cohort, 4.4% had clinically significant hematomas. The 15 mg and 30 mg ketorolac groups had similar prevalence (6.0% vs 5.8%, p = 0.95). On univariable regression, there were increased odds of hematoma formation in patients who were younger, had bilateral procedures, had longer OR times, and who received ketorolac. On multivariable regression, none of the prior variables remained significant. CONCLUSION After accounting for associations with longer operative times, concomitant IBBR, and bilateral procedures, ketorolac administration did not remain an independent risk factor for hematoma formation.
Collapse
Affiliation(s)
- Sami M Abujbarah
- Mayo Clinic Alix School of Medicine, Arizona Campus, Scottsdale, AZ, USA
| | - Kristen Jogerst
- Department of General Surgery, Division of Surgical Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Heidi E Kosiorek
- Department of Research-Biostatistics, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Sarwat Ahmad
- Department of General Surgery, Division of Surgical Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Patricia A Cronin
- Department of General Surgery, Division of Surgical Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - William Casey
- Department of Surgery, Division of Plastic Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Ryan Craner
- Department of Anesthesiology, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Alanna Rebecca
- Department of Surgery, Division of Plastic Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| | - Barbara A Pockaj
- Department of General Surgery, Division of Surgical Oncology, Mayo Clinic Arizona, Phoenix, AZ, USA.
| |
Collapse
|
18
|
Role of Anti-Angiogenic Factors in the Pathogenesis of Breast Cancer: A Review of Therapeutic Potential. Pathol Res Pract 2022; 236:153956. [DOI: 10.1016/j.prp.2022.153956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 11/23/2022]
|
19
|
Bezu L, Wu Chuang A, Sauvat A, Humeau J, Xie W, Cerrato G, Liu P, Zhao L, Zhang S, Le Naour J, Pol J, van Endert P, Kepp O, Barlesi F, Kroemer G. Local anesthetics elicit immune-dependent anticancer effects. J Immunother Cancer 2022; 10:e004151. [PMID: 35483744 PMCID: PMC9052055 DOI: 10.1136/jitc-2021-004151] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Retrospective clinical trials reported a reduced local relapse rate, as well as improved overall survival after injection of local anesthetics during cancer surgery. Here, we investigated the anticancer effects of six local anesthetics used in clinical practice. RESULTS In vitro, local anesthetics induced signs of cancer cell stress including inhibition of oxidative phosphorylation, and induction of autophagy as well as endoplasmic reticulum (ER) stress characterized by the splicing of X-box binding protein 1 (XBP1s) mRNA, cleavage of activating transcription factor 6 (ATF6), phosphorylation of eIF2α and subsequent upregulation of activating transcription factor 4 (ATF4). Both eIF2α phosphorylation and autophagy required the ER stress-relevant eukaryotic translation initiation factor 2 alpha kinase 3 (EIF2AK3, best known as PERK). Local anesthetics also activated two hallmarks of immunogenic cell death, namely, the release of ATP and high-mobility group box 1 protein (HMGB1), yet failed to cause the translocation of calreticulin (CALR) from the ER to the plasma membrane. In vivo, locally injected anesthetics decreased tumor growth and improved survival in several models of tumors established in immunocompetent mice. Systemic immunotherapy with PD-1 blockade or intratumoral injection of recombinant CALR protein, increased the antitumor effects of local anesthetics. Local anesthetics failed to induce antitumor effects in immunodeficient mice or against cancers unable to activate ER stress or autophagy due to the knockout of EIF2AK3/PERK or ATG5, respectively. Uncoupling agents that inhibit oxidative phosphorylation and induce autophagy and ER stress mimicked the immune-dependent antitumor effects of local anesthetics. CONCLUSION Altogether, these results indicate that local anesthetics induce a therapeutically relevant pattern of immunogenic stress responses in cancer cells.
Collapse
Affiliation(s)
- Lucillia Bezu
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Anesthesiology Department, Gustave Roussy Cancer Campus, Villejuif, France
| | - Alejandra Wu Chuang
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Allan Sauvat
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Juliette Humeau
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, Québec, Canada
- Department of Medicine, Université de Montréal, Montreal, Québec, Canada
| | - Wei Xie
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Giulia Cerrato
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Peng Liu
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Liwei Zhao
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Shuai Zhang
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Julie Le Naour
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Jonathan Pol
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Peter van Endert
- Institut National de la Santé et de la Recherche Médicale, Unité 1151, Université de Paris Cité, Centre National de la Recherche Scientifique, UMR 8253, Paris, France
| | - Oliver Kepp
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Fabrice Barlesi
- Gustave Roussy, Villejuif, France
- Aix Marseille University, INSERM, CNRS, CRCM, Marseille, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM UMR1138, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Pôle de Biologie, Hôpital européen Georges Pompidou, AP-HP, Paris, France
| |
Collapse
|
20
|
Kim R, Kawai A, Wakisaka M, Kin T. Current Status and Prospects of Anesthesia and Breast Cancer: Does Anesthetic Technique Affect Recurrence and Survival Rates in Breast Cancer Surgery? Front Oncol 2022; 12:795864. [PMID: 35223475 PMCID: PMC8864113 DOI: 10.3389/fonc.2022.795864] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/17/2022] [Indexed: 01/13/2023] Open
Abstract
The relationship between the anesthetic technique and cancer recurrence has not yet been clarified in cancer surgery. Surgical stress and inhalation anesthesia suppress cell-mediated immunity (CMI), whereas intravenous (IV) anesthesia with propofol and regional anesthesia (RA) are known to be protective for CMI. Surgical stress, general anesthesia (GA) with inhalation anesthesia and opioids contribute to perioperative immunosuppression and may increase cancer recurrence and decrease survival. Surgical stress and GA activate the hypothalamic-pituitary-adrenal axis and release neuroendocrine mediators such as cortisol, catecholamines, and prostaglandin E2, which may reduce host defense immunity and promote distant metastasis. On the other hand, IV anesthesia with propofol and RA with paravertebral block or epidural anesthesia can weaken surgical stress and GA-induced immunosuppression and protect the host defense immunity. IV anesthesia with propofol and RA or in combination with GA may reduce cancer recurrence and improve patient survival compared to GA alone. We review the current status of the relationship between anesthesia and breast cancer recurrence using retrospective and prospective studies conducted with animal models and clinical samples, and discuss the future prospects for reducing breast cancer recurrence and improving survival rates in breast cancer surgery.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | - Ami Kawai
- Department of Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | - Megumi Wakisaka
- Department of Breast Surgery, Hiroshima Mark Clinic, Hiroshima, Japan
| | - Takanori Kin
- Department of Breast Surgery, Hiroshima City Hospital, Hiroshima, Japan
| |
Collapse
|
21
|
Zhang H, Zhou D, Gu J, Qu M, Guo K, Chen W, Miao C. Targeting the mu-Opioid Receptor for Cancer Treatment. Curr Oncol Rep 2021; 23:111. [PMID: 34342720 DOI: 10.1007/s11912-021-01107-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW Opioids are still the most effective and widely used treatments for acute and chronic pain in cancer patients. This review focuses on the impact of opioids and mu-opioid receptors (MOR) on tumor progression and providing new ideas for targeting the MOR in cancer treatment. RECENT FINDINGS Studies estimated that opioids facilitate tumor progression and are related to the worse prognosis in cancer patients. As the primary receptor of opioids, MOR is involved in the regulation of malignant transformation of tumors and participating in proliferation, invasion, metastasis, and angiogenesis. MOR may be a new molecular marker of malignant tumors and thus become a new target for cancer therapy, which may be beneficial to the outcomes of cancer patients.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
| | - Di Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
| | - Jiahui Gu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
| | - Wankun Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China. .,Fudan Zhangjiang Institute, Shanghai, 201203, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, 180# Feng-Lin Road, Shanghai, 200032, China.
| |
Collapse
|
22
|
Li L, Zhao X, Xiang B, Tang X. Ketorolac following Mastectomy: Is There an Increased Risk of Reoperation? Ann Surg Oncol 2021; 28:775-776. [PMID: 33903999 DOI: 10.1245/s10434-021-10071-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 02/05/2023]
Affiliation(s)
- Lang Li
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan, People's Republic of China
| | - Xue Zhao
- Department of Obstetrics and Gynaecology Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Bo Xiang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Xueyang Tang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
23
|
Xue C, Chen W, Yuan A, Chen C, Li S, Chen K, Zhao Y, Xiao T, Shao G, Zou Y, Zheng D. Dezocine, An Opioid Analgesic, Exerts Antitumor Effects in Triple-Negative Breast Cancer by Targeting Nicotinamide Phosphoribosyltransferase. Front Pharmacol 2021; 12:600296. [PMID: 33912035 PMCID: PMC8072669 DOI: 10.3389/fphar.2021.600296] [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: 08/29/2020] [Accepted: 02/22/2021] [Indexed: 01/15/2023] Open
Abstract
Opioids are a potential adjuvant treatment for certain cancers; while they are primarily used to relieve chronic pain, these drugs may also affect cancer progression and recurrence. Dezocine is one opioid commonly used in China, but its effects on cancer cells are unknown. Here, we demonstrated the inhibitory effect of dezocine on triple-negative breast cancer (TNBC) cells, and determined the underlying molecular mechanism. We found that dezocine suppressed cell proliferation, migration and invasion, and induced apoptosis in TNBC cells. Xenograft models demonstrated the inhibitory effects of dezocine treatment on TNBC tumor growth in vivo. The anticancer effects of dezocine were independent of opioid receptors, which are not highly expressed by normal breast or breast cancer tissues. A pull-down assay and LC-MS/MS analysis indicated that dezocine directly targets NAMPT: computer modeling verified that the free energy of dezocine kinetically bound into the pocket of NAMPT was −17.4 kcal/mol. Consequently, dezocine treatment inhibited NAMPT enzyme activity, resulting in cellular NAD abolishment. We confirmed the dezocine-induced inhibition of cell proliferation by both NAMPT knockdown and upon treatment with the inhibitor FK866. Our results suggest that both dezocine and NAMPT might represent novel therapeutic targets for TNBC.
Collapse
Affiliation(s)
- Chenyang Xue
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, School of Pharmaceutical Sciences, Health Science Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Wei Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, School of Pharmaceutical Sciences, Health Science Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Aiwu Yuan
- Department of Anesthesiology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China
| | - Cheng Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, School of Pharmaceutical Sciences, Health Science Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Shuaihu Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, School of Pharmaceutical Sciences, Health Science Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Kai Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Yang Zhao
- Department of Anesthesiology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China
| | - Tian Xiao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, School of Pharmaceutical Sciences, Health Science Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Genze Shao
- Department of Cell Biology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yongdong Zou
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, School of Pharmaceutical Sciences, Health Science Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Duo Zheng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, School of Pharmaceutical Sciences, Health Science Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| |
Collapse
|
24
|
Ponferrada AR, Orriach JLG, Manso AM, Haro ES, Molina SR, Heredia AF, Lopez MB, Mañas JC. Anaesthesia and cancer: can anaesthetic drugs modify gene expression? Ecancermedicalscience 2020; 14:1080. [PMID: 32863874 PMCID: PMC7434501 DOI: 10.3332/ecancer.2020.1080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Indexed: 01/21/2023] Open
Abstract
Cancer remains a primary cause of morbidity and mortality worldwide, and its incidence continues to increase. The most common cause of death in cancer patients is tumour recurrence. Surgery is the gold standard in the treatment of most tumours. However, cancer surgery can lead to the release of tumour cells into the systemic circulation. Surgical stress and several perioperative factors have been suggested to boost tumour growth, thereby increasing the risk of metastatic recurrence. Preclinical and clinical studies suggest that anaesthetics and adjuvants administered during the perioperative period may impact cancer recurrence and survival. This document summarises the current evidence regarding the effects of anaesthetic drugs and analgesic techniques on the immune system, systemic inflammatory response and tumour cells, as well as their impact on cancer recurrence.
Collapse
Affiliation(s)
- Aida Raigon Ponferrada
- Institute of Biomedical Research in Malaga [IBIMA], Malaga 29010, Spain
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga 29010, Spain
| | - Jose Luis Guerrero Orriach
- Institute of Biomedical Research in Malaga [IBIMA], Malaga 29010, Spain
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga 29010, Spain
- Department of Pharmacology and Pediatrics, School of Medicine, University of Malaga, Malaga 29010, Spain
- Member of COST Action 15204
| | - Alfredo Malo Manso
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga 29010, Spain
| | - Enrique Sepúlveda Haro
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga 29010, Spain
| | - Salvador Romero Molina
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga 29010, Spain
| | - Ana Fontaneda Heredia
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga 29010, Spain
| | - Manolo Baena Lopez
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga 29010, Spain
| | - Jose Cruz Mañas
- Department of Anaesthesiology, Virgen de la Victoria University Hospital, Malaga 29010, Spain
| |
Collapse
|