1
|
Meng YW, Liu JY. Pathological and pharmacological functions of the metabolites of polyunsaturated fatty acids mediated by cyclooxygenases, lipoxygenases, and cytochrome P450s in cancers. Pharmacol Ther 2024; 256:108612. [PMID: 38369063 DOI: 10.1016/j.pharmthera.2024.108612] [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: 10/30/2023] [Revised: 01/19/2024] [Accepted: 02/05/2024] [Indexed: 02/20/2024]
Abstract
Oxylipins have garnered increasing attention because they were consistently shown to play pathological and/or pharmacological roles in the development of multiple cancers. Oxylipins are the metabolites of polyunsaturated fatty acids via both enzymatic and nonenzymatic pathways. The enzymes mediating the metabolism of PUFAs include but not limited to lipoxygenases (LOXs), cyclooxygenases (COXs), and cytochrome P450s (CYPs) pathways, as well as the down-stream enzymes. Here, we systematically summarized the pleiotropic effects of oxylipins in different cancers through pathological and pharmacological aspects, with specific reference to the enzyme-mediated oxylipins. We discussed the specific roles of oxylipins on cancer onset, growth, invasion, and metastasis, as well as the expression changes in the associated metabolic enzymes and the associated underlying mechanisms. In addition, we also discussed the clinical application and potential of oxylipins and related metabolic enzymes as the targets for cancer prevention and treatment. We found the specific function of most oxylipins in cancers, especially the underlying mechanisms and clinic applications, deserves and needs further investigation. We believe that research on oxylipins will provide not only more therapeutic targets for various cancers but also dietary guidance for both cancer patients and healthy humans.
Collapse
Affiliation(s)
- Yi-Wen Meng
- CNTTI of the Institute of Life Sciences & Department of Anesthesia of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing 400016, China
| | - Jun-Yan Liu
- CNTTI of the Institute of Life Sciences & Department of Anesthesia of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing 400016, China; College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
2
|
Gomaa M, Gad W, Hussein D, Pottoo FH, Tawfeeq N, Alturki M, Alfahad D, Alanazi R, Salama I, Aziz M, Zahra A, Hanafy A. Sulfadiazine Exerts Potential Anticancer Effect in HepG2 and MCF7 Cells by Inhibiting TNFα, IL1b, COX-1, COX-2, 5-LOX Gene Expression: Evidence from In Vitro and Computational Studies. Pharmaceuticals (Basel) 2024; 17:189. [PMID: 38399404 PMCID: PMC10891904 DOI: 10.3390/ph17020189] [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: 01/02/2024] [Revised: 01/24/2024] [Accepted: 01/28/2024] [Indexed: 02/25/2024] Open
Abstract
Drug repurposing is a promising approach that has the potential to revolutionize the drug discovery and development process. By leveraging existing drugs, we can bring new treatments to patients more quickly and affordably. Anti-inflammatory drugs have been shown to target multiple pathways involved in cancer development and progression. This suggests that they may be more effective in treating cancer than drugs that target a single pathway. Cell viability was measured using the MTT assay. The expression of genes related to inflammation (TNFa, IL1b, COX-1, COX-2, and 5-LOX) was measured in HepG2, MCF7, and THLE-2 cells using qPCR. The levels of TNFα, IL1b, COX-1, COX-2, and 5-LOX were also measured in these cells using an ELISA kit. An enzyme binding assay revealed that sulfadiazine expressed weaker inhibitory activity against COX-2 (IC50 = 5.27 μM) in comparison with the COX-2 selective reference inhibitor celecoxib (COX-2 IC50 = 1.94 μM). However, a more balanced inhibitory effect was revealed for sulfadiazine against the COX/LOX pathway with greater affinity towards 5-LOX (IC50 = 19.1 μM) versus COX-1 (IC50 = 18.4 μM) as compared to celecoxib (5-LOX IC50 = 16.7 μM, and COX-1 IC50 = 5.9 μM). MTT assays revealed the IC50 values of 245.69 ± 4.1 µM and 215.68 ± 3.8 µM on HepG2 and MCF7 cell lines, respectively, compared to the standard drug cisplatin (66.92 ± 1.8 µM and 46.83 ± 1.3 µM, respectively). The anti-inflammatory effect of sulfadiazine was also depicted through its effect on the levels of inflammatory markers and inflammation-related genes (TNFα, IL1b, COX-1, COX-2, 5-LOX). Molecular simulation studies revealed key binding interactions that explain the difference in the activity profiles of sulfadiazine compared to celecoxib. The results suggest that sulfadiazine exhibited balanced inhibitory activity against the 5-LOX/COX-1 enzymes compared to the selective COX-2 inhibitor, celecoxib. These findings highlight the potential of sulfadiazine as a potential anticancer agent through balanced inhibitory activity against the COX/LOX pathway and reduction in the expression of inflammatory genes.
Collapse
Affiliation(s)
- Mohamed Gomaa
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (N.T.); (M.A.)
| | - Wael Gad
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 6860404, Egypt; (W.G.); (M.A.); (A.Z.); (A.H.)
| | - Dania Hussein
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Nada Tawfeeq
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (N.T.); (M.A.)
| | - Mansour Alturki
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (N.T.); (M.A.)
| | - Dhay Alfahad
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (D.A.); (R.A.)
| | - Razan Alanazi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (D.A.); (R.A.)
| | - Ismail Salama
- Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia 8366004, Egypt;
| | - Mostafa Aziz
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 6860404, Egypt; (W.G.); (M.A.); (A.Z.); (A.H.)
| | - Aboelnasr Zahra
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 6860404, Egypt; (W.G.); (M.A.); (A.Z.); (A.H.)
| | - Abeer Hanafy
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh 6860404, Egypt; (W.G.); (M.A.); (A.Z.); (A.H.)
| |
Collapse
|
3
|
Shokri S, Ayazi H, Tamjid M, Ghoreishi F, Shokri M, Badakhshannouri S, Naderi N, Daraei B, Mousavi Z, Davood A. Hybrid Analogues of Hydrazone and Phthalimide: Design, Synthesis, In vivo, In vitro, and In silico Evaluation as Analgesic Agents. Curr Comput Aided Drug Des 2024; 20:685-696. [PMID: 37194935 DOI: 10.2174/1573409919666230517121726] [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: 07/15/2022] [Revised: 12/10/2022] [Accepted: 01/04/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Based on the anti-inflammatory and analgesic activity of hydrazone and phthalimide, a new series of hybrid hydrazone and phthalimide pharmacophores was prepared and evaluated as analgesic agents. METHODS The designed ligands were synthesized by reaction of the appropriate aldehydes and 2- aminophthalimide. Analgesic, cyclooxygenase inhibitory, and cytostatic activity of prepared compounds were measured. RESULTS All the tested ligands demonstrated significant analgesic activity. Moreover, compounds 3i and 3h were the most potent ligands in the formalin and writhing tests, respectively. Compounds 3g, 3j, and 3l were the most COX-2 selective ligands and ligand 3e was the most potent COX inhibitor with a 0.79 of COX-2 selectivity ratio. The presence of electron-withdrawing moieties with hydrogen bonding ability at the meta position was found to affect the selectivity efficiently, in which compounds 3g, 3l, and 3k showed high COX-2 selectivity, and compound 3k was the most potent one. The cytostatic activity of selected ligands demonstrated that compounds 3e, 3f, 3h, 3k, and 3m showed good analgesic and COX inhibitory activity and were less toxic than the reference drug. CONCLUSION High therapeutic index of these ligands is one of the valuable advantages of these compounds.
Collapse
Affiliation(s)
- Shahla Shokri
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Tehran Islamic Azad Medical Sciences University, Tehran, Iran
| | - Hoda Ayazi
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Tehran Islamic Azad Medical Sciences University, Tehran, Iran
| | - Mohsen Tamjid
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ghoreishi
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Shokri
- Department of Toxicology and Pharmacology, Faculty of Pharmaceutical Sciences, Tehran Islamic Azad Medical Sciences University, Tehran, Iran
| | - Sogol Badakhshannouri
- Department of Toxicology and Pharmacology, Faculty of Pharmaceutical Sciences, Tehran Islamic Azad Medical Sciences University, Tehran, Iran
| | - Nima Naderi
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Daraei
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Mousavi
- Department of Toxicology and Pharmacology, Faculty of Pharmaceutical Sciences, Tehran Islamic Azad Medical Sciences University, Tehran, Iran
| | - Asghar Davood
- Department of Medicinal Chemistry, Faculty of Pharmaceutical Sciences, Tehran Islamic Azad Medical Sciences University, Tehran, Iran
| |
Collapse
|
4
|
Wang Z, Wang T, Chen X, Cheng J, Wang L. Pterostilbene regulates cell proliferation and apoptosis in non-small-cell lung cancer via targeting COX-2. Biotechnol Appl Biochem 2023; 70:106-119. [PMID: 35231150 DOI: 10.1002/bab.2332] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/05/2022] [Indexed: 11/11/2022]
Abstract
Non-small-cell lung cancer (NSCLC), occupying a great proportion of lung cancer, threatens the health of patients, and the cyclooxygenase-2 (COX-2) expression is found to be upregulated in lung cancer. Pterostilbene (PTE) is perceived as a novel method for clinical therapy due to its high performance. However, the mechanism underlying and the interaction between PTE and COX-2 remain vague. We simulated radiation circumstances and transfected cells with the interference of PTE and COX-2. Our results showed that radiation or PTE treatment alone restrained cell proliferation and viability while stimulating cell apoptosis, and the above properties were strengthened when the two were in combination. The COX-2 expression was promoted by radiation but was reduced by PTE. PTE reversed the effects of radiation on the COX-2 expression. COX-2 knockdown suppressed COX-2 expression and proliferation and enhanced apoptosis of cells suffering radiation, while COX-2 overexpression reversed the inhibition of PTE. Our study suggested PTE regulated NSCLC cell proliferation and apoptosis via targeting COX-2, which might shed a light on cancer therapy.
Collapse
Affiliation(s)
- Zhimin Wang
- Department of Integrated Chinese and Western Medicine, Taizhou Central Hospital (Taizhou University Hospital), Jiaojiang District, Taizhou City, China
| | - Tingting Wang
- Department of Integrated Chinese and Western Medicine, Taizhou Central Hospital (Taizhou University Hospital), Jiaojiang District, Taizhou City, China
| | - Xu Chen
- Department of Integrated Chinese and Western Medicine, Taizhou Central Hospital (Taizhou University Hospital), Jiaojiang District, Taizhou City, China
| | - Jing Cheng
- Department of Integrated Chinese and Western Medicine, Taizhou Central Hospital (Taizhou University Hospital), Jiaojiang District, Taizhou City, China
| | - Lijuan Wang
- Respiratory and Critical Care Medicine Department, Taizhou Central Hospital (Taizhou University Hospital), Jiaojiang District, Taizhou City, China
| |
Collapse
|
5
|
Vieira TC, Oliveira EA, dos Santos BJ, Souza FR, Veloso ES, Nunes CB, Del Puerto HL, Cassali GD. COX-2 expression in mammary invasive micropapillary carcinoma is associated with prognostic factors and acts as a potential therapeutic target in comparative oncology. Front Vet Sci 2022; 9:983110. [PMID: 36172611 PMCID: PMC9510711 DOI: 10.3389/fvets.2022.983110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022] Open
Abstract
Pure human and canine mammary invasive micropapillary carcinoma is a rare malignant epithelial tumor accounting for 0.9 to 2% of all invasive mammary carcinomas and present a high rate of lymphatic invasion and metastasis, with unfavorable prognosis. Surgery and chemotherapy are standard treatments for almost all mammary cancer in both species, as well as hormonal and target therapies available for human patients. However, depending on the patient's clinical staging, satisfactory therapeutic results for invasive micropapillary carcinoma are a challenge due to its high capacity of invasion and metastasis. Cyclooxygenase-2 (COX-2) isoform is an important enzyme stimulated by cytokines, growth factors and oncogenes activation to synthetizes prostaglandins in inflammatory process. COX-2 overexpression is associated with angiogenesis and invasion and contributes to cancer development, disease progression, tumor recurrence and regional lymph node metastasis in human and canine mammary carcinomas. This enzyme can be targeted by non-steroidal anti-inflammatory drugs and its inhibition can reduce tumor growth and metastasis in several cancer types. Given the similarity between both species, the present study aims to elucidate the involvement of COX-2 mRNA and protein expression in canine (cIMPC) and human (hIMPC) pure invasive mammary micropapillary carcinoma, with clinicopathological and survival data. Twenty-nine cases of cIMPC and 17 cases of hIMPC were analyzed regarding histologic type, grade, age, tumor size, lymph node condition, extracapsular extension, inflammatory infiltrate and immunophenotype. When available, information on adjuvant treatment, recurrence, metastasis and overall survival were collected. The present study demonstrated COX-2 protein expression in 65.5% of cIMPC and 92.3% of hIMPC, and an association with more advanced histological grades in bitches and higher Ki67 in women. COX-2 mRNA expression was significantly higher in cIMPC than in hIMPC, and its expression was not associated with COX-2 protein expression in both species. COX-2 mRNA expression was associated with negative-ER hIMPC as well as higher Ki67. cIMPC demonstrated proportional early development, more regional metastasis, and a prevalence of negative estrogen receptor, than hIMPC. This is the first time COX-2 expression is associated with negative prognostic factors in both cIMPC and hIMPC, besides the overexpression of COX-2 protein in such unfavorable histological type, which suggests that COX-2 can act as a potential target in IMPC.
Collapse
Affiliation(s)
- Thaynan Cunha Vieira
- Laboratory of Comparative Oncology, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Evelyn Ane Oliveira
- Laboratory of Comparative Oncology, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Bárbara Jaime dos Santos
- Laboratory of Breast Pathology, Medical School, Department of Pathological Anatomy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Rezende Souza
- Laboratory of Comparative Oncology, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Emerson Soares Veloso
- Laboratory of Cellular Behavior, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Cristiana Buzelin Nunes
- Laboratory of Breast Pathology, Medical School, Department of Pathological Anatomy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Helen Lima Del Puerto
- Laboratory of Cellular Behavior, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanni Dantas Cassali
- Laboratory of Comparative Oncology, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Geovanni Dantas Cassali
| |
Collapse
|
6
|
Rahimifard M, Bagheri Z, Hadjighassem M, Jaktaji RP, Behroodi E, Haghi-Aminjan H, Movahed MA, Latifi H, Hosseindoost S, Zarghi A, Pourahmad J. Investigation of anti-cancer effects of new pyrazino[1,2-a]benzimidazole derivatives on human glioblastoma cells through 2D in vitro model and 3D-printed microfluidic device. Life Sci 2022; 302:120505. [PMID: 35358594 DOI: 10.1016/j.lfs.2022.120505] [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: 01/08/2022] [Revised: 03/18/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022]
Abstract
AIMS Recent studies show targeted therapy of new pyrazino[1,2-a]benzimidazole derivatives with COX-II inhibitory effects on different cancer cells. This study aimed to investigate 2D cell culture and 3D spheroid formation of glioblastoma multiforme (GBM) cells using a microfluidic device after exposure to these compounds. MAIN METHODS After isolating astrocytes from human GBM samples, IC50 of 2,6-dimethyl pyrazino[1,2-a]benzimidazole (L1) and 3,4,5-trimethoxy pyrazino[1,2-a]benzimidazole (L2) were determined as 13 μM and 85 μM, respectively. Then, in all experiments, cells were exposed to subtoxic concentrations of L1 (6.5 μM) and L2 (42.5 μM), which were ½IC50. In the following, in two phases, cell cycle, migration, and gene expression through 2D cell culture and tumor spheroid formation ability using a 3D-printed microfluidic chip were assessed. KEY FINDINGS The obtained results showed that both compounds have positive effects in reducing G2/M cell population and GBM cell migration. Furthermore, real-time gene expression data showed that L1 and L2 significantly impact the upregulation of P21 and P53 and down-regulation of cyclin D1, MMP2, and MMP9. On the other hand, GBM spheroids exposed to L1 and L2 become smaller with fewer live cells. SIGNIFICANCE Our data on human isolated astrocyte cells in 2D and 3D cell culture conditions showed that L1 and L2 compounds could reduce GBM cells' invasion by controlling gene expressions associated with migration and proliferation. Moreover, designing microfluidic platform and related cell culture protocols facilitates the broad screening of 3D multicellular tumor spheroids derived from GBM tumor biopsies and provides effective drug development for brain gliomas.
Collapse
Affiliation(s)
- Mahban Rahimifard
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Bagheri
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University G.C., Tehran, Iran
| | - Mahmoudreza Hadjighassem
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ebrahim Behroodi
- Laser and Plasma Research Institute, Shahid Beheshti University G.C., Tehran, Iran
| | - Hamed Haghi-Aminjan
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mahsa Azami Movahed
- Department of Medicinal and Pharmaceutical Chemistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Latifi
- Laser and Plasma Research Institute, Shahid Beheshti University G.C., Tehran, Iran
| | - Saereh Hosseindoost
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Afshin Zarghi
- Department of Medicinal and Pharmaceutical Chemistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Jalal Pourahmad
- Department of Toxicology and Pharmacology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Buddeberg BS, Seeberger MD. Anesthesia and Oncology: Friend or Foe? Front Oncol 2022; 12:802210. [PMID: 35359377 PMCID: PMC8963958 DOI: 10.3389/fonc.2022.802210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/17/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer is a leading cause of death, and surgery is an important treatment modality. Laboratory research and retrospective studies have raised the suspicion that the choice of anesthetics for cancer surgery might affect the course of cancerous disease. The aim of this review is to provide a critical overview of the current state of knowledge. Inhalational anesthesia with volatiles or total intravenous anesthesia (TIVA) with propofol are the two most commonly used anesthetic techniques. Most data comparing volatile anesthetics with TIVA is from either in vitro or retrospective studies. Although conflicting, data shows a trend towards favoring propofol. Opioids are commonly used in anesthesia. Data on potential effects of opioids on growth and recurrence of cancer are scarce and conflicting. Preclinical studies have shown that opioids stimulate cancer growth through the µ-opioid receptor. Opioids also act as immunosuppressants and, therefore, have the potential to facilitate metastatic spread. However, the finding of an adverse effect of opioids on tumor growth and cancer recurrence by some retrospective studies has not been confirmed by prospective studies. Regional anesthesia has not been found to have a beneficial effect on the outcome of surgically treated cancer patients, but prospective studies are scarce. Local anesthetics might have a beneficial effect, as observed in animal and in vitro studies. However, prospective clinical studies strongly question such an effect. Blood products, which may be needed during extensive cancer surgery suppress the immune system, and data strongly suggest a negative impact on cancer recurrence. The potential effects of other commonly used anesthetic agents on the outcome of cancer patients have not been sufficiently studied for drawing valid conclusions. In conclusion, laboratory data and most retrospective studies suggest a potential advantage of TIVA over inhalational anesthesia on the outcome of surgical cancer patients, but prospective, randomized studies are missing. Given the state of weak scientific evidence, TIVA may be used as the preferred type of anesthesia unless there is an individual contraindication against it. Studies on the effects of other drugs frequently used in anesthesia are limited in number and quality, and have found conflicting results.
Collapse
Affiliation(s)
- Bigna S. Buddeberg
- Clinic for Anesthesia, Intermediate Care, Prehospital Emergency Medicine and Pain Therapy, University Hospital Basel, Basel, Switzerland
- Medical School, University of Basel, Basel, Switzerland
| | - Manfred D. Seeberger
- Medical School, University of Basel, Basel, Switzerland
- *Correspondence: Manfred D. Seeberger,
| |
Collapse
|
8
|
Bouchet A, Le Clec'h C, Rogalev L, Le Duc G, Pelletier L. Meloxicam can Potentiate the Therapeutic Effects of Synchrotron Microbeam Radiation Therapy on High-Grade Glioma Bearing Rats. Radiat Res 2022; 197:655-661. [PMID: 35245385 DOI: 10.1667/rade-21-00107.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/24/2022] [Indexed: 11/03/2022]
Abstract
The microbeam radiation therapy (MRT), a spatially micro-fractionated synchrotron radiotherapy, leads to better control of incurable high-grade glioma than that obtained upon homogeneous radiotherapy. We evaluated the effect of meloxicam, a non-steroidal anti-inflammatory drug (NSAID), to increase the MRT response. Survival of rats bearing intracranial 9L gliosarcoma treated with meloxicam and/or MRT (400 Gy, 50 μm-wide microbeams, 200 μm spacing) was monitored. Tumor growth was assessed on histological tissue sections and COX-2 transcriptomic expression was studied 1 to 25 days after radiotherapy. Meloxicam significantly extended the median survival of microbeam-irradiated rats (from +10.5 to +20 days). Dual treatment led to last survivors until D90 (D39 for the MRT group) and to tumor 9.5 times smaller than MRT alone. No significant modification of COX-2 expression was induced by MRT in normal and tumor tissues. The meloxicam reinforced the anti-tumor effect of MRT for glioma treatment. Although the mechanisms of interaction between meloxicam and MRT remain to be elucidated, the addition of this NSAID, easily implemented as a supplement to water for example, is a very favorable therapeutic regimen since it doubled the survival benefit compared to MRT alone.
Collapse
Affiliation(s)
- Audrey Bouchet
- INSERM U1296 "Radiation: Defense, Health Environment", Centre Léon-Bérard, 28 Rue Laennec, 69008 Lyon, France.,Biomedical Beamline, European Synchrotron Radiation Facility, BP220, F38043 Grenoble cedex
| | - Céline Le Clec'h
- Biomedical Beamline, European Synchrotron Radiation Facility, BP220, F38043 Grenoble cedex
| | - Léonid Rogalev
- Biomedical Beamline, European Synchrotron Radiation Facility, BP220, F38043 Grenoble cedex
| | - Géraldine Le Duc
- Biomedical Beamline, European Synchrotron Radiation Facility, BP220, F38043 Grenoble cedex
| | - Laurent Pelletier
- Grenoble University Hospital, BP217, F-38043 Grenoble cedex.,INSERM U836, Team Nanomedicine and brain, 6 Rue Fortuné Ferrini, F38706 La Tronche
| |
Collapse
|
9
|
Wu K, Peng X, Chen M, Li Y, Tang G, Peng J, Peng Y, Cao X. Recent progress of research on anti‐tumor agents using benzimidazole as the structure unit. Chem Biol Drug Des 2022; 99:736-757. [DOI: 10.1111/cbdd.14022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/10/2022] [Accepted: 01/15/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Kaiyue Wu
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Xiaoyu Peng
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Miaojia Chen
- Department of Pharmacy the first People's Hospital Pingjiang Yueyang Hunan China
| | - Yang Li
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Junmei Peng
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| | - Yuanyuan Peng
- School of Electrical and Automation Engineering East China Jiaotong University Nanchang 330000 China
| | - Xuan Cao
- Institute of Pharmacy and Pharmacology Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study College of Pharmacy Hengyang Medical School University of South China Hengyang China
| |
Collapse
|
10
|
Shen Z, Chen M, Luo F, Xu H, Zhang P, Lin J, Kang M. Identification of Key Genes and Pathways Associated With Paclitaxel Resistance in Esophageal Squamous Cell Carcinoma Based on Bioinformatics Analysis. Front Genet 2021; 12:671639. [PMID: 34456964 PMCID: PMC8386171 DOI: 10.3389/fgene.2021.671639] [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: 02/24/2021] [Accepted: 07/02/2021] [Indexed: 01/07/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) ranks as the fourth leading cause of cancer-related death in China. Although paclitaxel has been shown to be effective in treating ESCC, the prolonged use of this chemical will lead to paclitaxel resistance. In order to uncover genes and pathways driving paclitaxel resistance in the progression of ESCC, bioinformatics analyses were performed based on The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database including GSE86099 and GSE161533. Differential expression analysis was performed in TCGA data and two GEO datasets to obtain differentially expressed genes (DEGs). Based on GSE161533, weighted gene co-expression network analysis (WGCNA) was conducted to identify the key modules associated with ESCC tumor status. The DEGs common to the two GEO datasets and the genes in the key modules were intersected to obtain the paclitaxel resistance-specific or non-paclitaxel resistance-specific genes, which were subjected to subsequent least absolute shrinkage and selection operator (LASSO) feature selection, whereby paclitaxel resistance-specific or non-paclitaxel resistance-specific key genes were selected. Ten machine learning models were used to validate the biological significance of these key genes; the potential therapeutic drugs for paclitaxel resistance-specific genes were also predicted. As a result, we identified 24 paclitaxel resistance-specific genes and 18 non-paclitaxel resistance-specific genes. The ESCC machine classifiers based on the key genes achieved a relatively high AUC value in the cross-validation and in an independent test set, GSE164158. A total of 207 drugs (such as bevacizumab) were predicted to be alternative therapeutics for ESCC patients with paclitaxel resistance. These results might shed light on the in-depth research of paclitaxel resistance in the context of ESCC progression.
Collapse
Affiliation(s)
- Zhimin Shen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingduan Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Fei Luo
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hui Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jihong Lin
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
Khafaga AF, Shamma RN, Abdeen A, Barakat AM, Noreldin AE, Elzoghby AO, Sallam MA. Celecoxib repurposing in cancer therapy: molecular mechanisms and nanomedicine-based delivery technologies. Nanomedicine (Lond) 2021; 16:1691-1712. [PMID: 34264123 DOI: 10.2217/nnm-2021-0086] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
While cancer remains a significant global health problem, advances in cancer biology, deep understanding of its underlaying mechanism and identification of specific molecular targets allowed the development of new therapeutic options. Drug repurposing poses several advantages as reduced cost and better safety compared with new compounds development. COX-2 inhibitors are one of the most promising drug classes for repurposing in cancer therapy. In this review, we provide an overview of the detailed mechanism and rationale of COX-2 inhibitors as anticancer agents and we highlight the most promising research efforts on nanotechnological approaches to enhance COX-2 inhibitors delivery with special focus on celecoxib as the most widely studied agent for chemoprevention or combined with chemotherapeutic and herbal drugs for combating various cancers.
Collapse
Affiliation(s)
- Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, 22758, Egypt
| | - Rehab N Shamma
- Department of Pharmaceutics & Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed Abdeen
- Department of Forensic Medicine & Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh, 13736, Egypt
| | | | - Ahmed E Noreldin
- Department of Histology & Cytology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22516, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.,Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| | - Marwa A Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
| |
Collapse
|
12
|
van Gisbergen MW, Zwilling E, Dubois LJ. Metabolic Rewiring in Radiation Oncology Toward Improving the Therapeutic Ratio. Front Oncol 2021; 11:653621. [PMID: 34041023 PMCID: PMC8143268 DOI: 10.3389/fonc.2021.653621] [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: 01/14/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
To meet the anabolic demands of the proliferative potential of tumor cells, malignant cells tend to rewire their metabolic pathways. Although different types of malignant cells share this phenomenon, there is a large intracellular variability how these metabolic patterns are altered. Fortunately, differences in metabolic patterns between normal tissue and malignant cells can be exploited to increase the therapeutic ratio. Modulation of cellular metabolism to improve treatment outcome is an emerging field proposing a variety of promising strategies in primary tumor and metastatic lesion treatment. These strategies, capable of either sensitizing or protecting tissues, target either tumor or normal tissue and are often focused on modulating of tissue oxygenation, hypoxia-inducible factor (HIF) stabilization, glucose metabolism, mitochondrial function and the redox balance. Several compounds or therapies are still in under (pre-)clinical development, while others are already used in clinical practice. Here, we describe different strategies from bench to bedside to optimize the therapeutic ratio through modulation of the cellular metabolism. This review gives an overview of the current state on development and the mechanism of action of modulators affecting cellular metabolism with the aim to improve the radiotherapy response on tumors or to protect the normal tissue and therefore contribute to an improved therapeutic ratio.
Collapse
Affiliation(s)
- Marike W van Gisbergen
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands.,Department of Dermatology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Emma Zwilling
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
13
|
Huang H, Huang Y, Chen Y, Luo Z, Zhang Z, Sun R, Wan Z, Sun J, Lu B, Zhang L, Hu J, Li S. A novel immunochemotherapy based on targeting of cyclooxygenase and induction of immunogenic cell death. Biomaterials 2021; 270:120708. [PMID: 33578254 PMCID: PMC7910750 DOI: 10.1016/j.biomaterials.2021.120708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 02/08/2023]
Abstract
Cyclooxygenase (COX) plays a crucial role in the "inflammogenesis of cancer", which leads to tumor progression, metastasis, and immunotherapy resistance. Therefore, reducing "inflammogenesis" by COX inhibition may be a key perspective for cancer therapy. However, the role of tumor-derived COX in the actions of COX inhibitors remains incompletely understood. In this study, applying "old drug new tricks" to repurpose 5-aminosalicylic acid (5-ASA), a COX inhibitor, we examined the effect of 5-ASA, alone or in combination with doxorubicin (DOX), in several cancer cell lines with different levels of COX expression. To facilitate the evaluation of the combination effect on tumors in vivo, a new micellar carrier based on PEG-b-PNHS polymer-conjugated 5-ASA (PASA) was developed to enhance codelivery of 5-ASA and DOX. Folate was also introduced to the polymer (folate-PEG-NH2-conjugated PASA (FASA)) to further improve delivery to tumors via targeting both tumor cells and tumor macrophages. An unprecedented high DOX loading capacity of 42.28% was achieved through various mechanisms of carrier/drug interactions. FASA was highly effective in targeting to and in inhibiting the growth of both 4T1.2 and CT26 tumors in BALB/c mice. However, FASA was more effective in CT26 tumor that has a high level of COX expression. Codelivery of DOX via PASA and FASA led to a further improvement in antitumor activity. Mechanistic studies suggest that inhibition of COX in vivo led to a more active tumor immune microenvironment. Interestingly, treatment with FASA led to upregulation of PD-1 on T cells, likely due to repressing the inhibitory effect of prostaglandin E2 (PGE2) on PD-1 expression on T cells. Combination of FASA/DOX with anti-PD-1 antibody led to a drastic improvement in the overall antitumor activity including regression of some established tumors at a suboptimal dose of FASA/DOX. Our data suggest that FASA/DOX may represent a new and effective immunochemotherapy for various types of cancers, particularly those cancers with high levels of COX expression.
Collapse
Affiliation(s)
- Haozhe Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Yuang Chen
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Zhangyi Luo
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Ziqian Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Runzi Sun
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Zhuoya Wan
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Jingjing Sun
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Binfeng Lu
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Jing Hu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, 15261, USA; University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
14
|
Celecoxib induces apoptosis through Akt inhibition in 5-fluorouracil-resistant gastric cancer cells. Toxicol Res 2021; 37:25-33. [PMID: 33489855 DOI: 10.1007/s43188-020-00044-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/10/2020] [Accepted: 03/04/2020] [Indexed: 10/24/2022] Open
Abstract
Gastric cancer is the fifth leading cause of cancer and a global public health problem. 5-Fluorouracil (5-FU) is the primary drug chosen for the treatment of advanced gastric cancer, but acquired cancer drug resistance limits its effectiveness and clinical use. Proliferation assays showed that a gastric carcinoma cell line, AGS and 5-FU-resistant AGS cells (AGS FR) treated with 3-100 μM 5-FU for 48 h or 72 h showed different sensitivities to 5-FU. Immunoblot assay demonstrated that AGS FR cells expressed more COX-2 and PGE2-cognated receptor EP2 than AGS cells. AGS FR cells considerably produced PGE2 than AGS upon stimulation with 5-FU. These results suggest that COX-2 expression is associated with 5-FU resistance. Unlike AGS FR cells, AGS cells showed increased levels of both cleaved caspase-3 and Bax following 5-FU treatment. Treatment of cells with the COX-2 selective inhibitor celecoxib induced cell death of AGS FR cells in a time- and concentration-dependent manner. FACS analysis showed that celecoxib at high doses caused apoptotic cell death, demonstrating a concentration-dependent increase in the cell populations undergoing early apoptosis and late apoptosis. This apoptotic induction was strongly supported by the expression profiles of apoptosis- and survival-associated proteins in response to celecoxib; pro-apoptotic cellular proteins increased while expressions of COX-2 and p-Akt were downregulated in a concentration-dependent manner. An increase in PTEN expression was accompanied with downregulation of p-Akt. Based on the data that downregulation of COX-2 was correlated with the concentrations of celecoxib, COX-2 may play a key role in celecoxib-induced cell death of AGS FR cells. Butaprost, the EP2 agonist, promoted proliferative activity of AGS FR cells in a concentration-dependent manner compared with AGS cells. In cells exposed to butaprost, expressions of COX-2 and p-Akt were increased in a concentration-dependent manner with concomitantly reduced PTEN levels. Taken together, 5-FU-resistance in gastric cancer is correlated with COX-2 expression, and therefore the selective inhibition of COX-2 leads to suppression of cell proliferation of AGS FR cells. Modulation of COX-2 expression and its catalytic activity may be a potential therapeutic strategy to overcome 5-FU-resistant gastric cancer.
Collapse
|
15
|
Wang Y, Yin L. LINC00461 Promoted Endometrial Carcinoma Growth and Migration by Targeting MicroRNA-219-5p/Cyclooxygenase-2 Signaling Axis. Cell Transplant 2021; 30:963689721989616. [PMID: 33573388 PMCID: PMC7885031 DOI: 10.1177/0963689721989616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/10/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Endometrial carcinoma (EC) ranks as the most common female genital cancer in developed countries. Lately, more and more long noncoding RNAs (lncRNAs) have been identified as vital regulators in numerous physiological and pathological processes, including EC. However, the expression pattern and precise functions of different lncRNAs in EC remain unclear. In this study, we reported LINC00461 was upregulated in EC patient tissues and cell lines. In addition, LINC00461 knockdown could remarkably suppress cell proliferation, cell cycle progression, cell migration, and promote cell apoptosis in EC cells. We discovered LINC00461 could sponge microRNA-219-5p (miR-219-5p) and suppress its expression, thereby upregulating expression level of miR-219-5p's target, cyclooxygenase-2 (COX-2). In vivo animal models, LINC00461 knockdown inhibited tumor growth by increasing miR-219-5p level and reducing COX-2 expression, thus confirming LINC00461 functions as an oncogene in EC. In this study, a novel regulatory role of LINC00461/miR-219-5p/COX-2 axis was systematically investigated in context of EC, with the aim to provide promising intervention targets for EC therapy from bench to clinic. [Formula: see text].
Collapse
Affiliation(s)
- Yu Wang
- Department of Obstetrics & Gynecology, Shengjing Hospital of China Medical University, Liaoning Province, PR China
| | - Lili Yin
- Department of Obstetrics & Gynecology, Shengjing Hospital of China Medical University, Liaoning Province, PR China
| |
Collapse
|
16
|
Azmoonfar R, Amini P, Saffar H, Motevaseli E, Khodamoradi E, Shabeeb D, Musa AE, Najafi M. Celecoxib A Selective COX-2 Inhibitor Mitigates Fibrosis but not Pneumonitis Following Lung Irradiation: A Histopathological Study. CURRENT DRUG THERAPY 2020. [DOI: 10.2174/1574885514666191119124739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Lung is one of the radiosensitive and late responding organs, and is an
important target for ionizing radiation. Radiation-induced pneumonitis and fibrosis are major consequences
of lung exposure to a high dose of radiation and pose threats to the lives of exposed people.
Mitigation of lung injury following an accidental radiation event or for patients with lung cancer
is one of the most interesting issues in radiobiology. In the current study, we aimed to determine
whether celecoxib, the most common cyclooxygenase-2 (COX-2) inhibitor, is able to mitigate
pneumonitis and fibrosis following lung irradiation or not.
Materials and methods:
20 male mice were assigned to 4 groups: control, celecoxib treatment,
radiation, and radiation plus celecoxib. Irradiation was performed with a dose of 18 Gy cobalt-60
(60Co) gamma rays. Celecoxib treatment (50 mg/kg) started 24 h after irradiation and continued four
times per week for 4 weeks.
Results:
Irradiation of lung led to remarkable infiltration of macrophages, lymphocytes, mast cells
and neutrophils. Also, a mild increase in fibrosis markers including accumulation of collagen, and
alveolar and vascular thickening, was observed. Post-exposure treatment with celecoxib was able to
mitigate fibrosis as well as alveolar and vascular changes, however, it was unable to mitigate pneumonitis
markers.
Conclusion:
Celecoxib showed that it may have an anti-fibrosis effect following exposure of mice
lung to radiation, although it was unable to prevent pneumonitis.
Collapse
Affiliation(s)
- Rasoul Azmoonfar
- Radiology Department, Faculty of Paramedical, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Hana Saffar
- Clinical and Anatomical Pathologist at Tehran University of Medical Science, Imam Khomeini Hospital Complex, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed Eleojo Musa
- Research Center for Molecular and Cellular Imaging, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
17
|
Tołoczko-Iwaniuk N, Dziemiańczyk-Pakieła D, Nowaszewska BK, Celińska-Janowicz K, Miltyk W. Celecoxib in Cancer Therapy and Prevention - Review. Curr Drug Targets 2020; 20:302-315. [PMID: 30073924 DOI: 10.2174/1389450119666180803121737] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/04/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND OBJECTIVES It is generally accepted that inflammatory cells found in the tumor microenvironment are involved in the neoplastic process, promoting cell proliferation, survival, and migration. Therefore, administering anti-inflammatory medication in cancer therapy seems to be justified. A potential pathway associated with the aforementioned issue is cyclooxygenase-2 inhibition, particularly as the overexpression of this enzyme has been proven to occur in cancer tissues and is also associated with a poor prognosis in several types of human malignancies. Celecoxib, a COX-2 selective inhibitor, has been utilized for over 20 years, particularly as an anti-inflammatory, analgesic and antipyretic medication. However, to date, its antineoplastic properties have not been sufficiently investigated. In recent years, the number of research studies on the antineoplastic effects of celecoxib has increased considerably. The vast majority of publications refers to preclinical studies attempting to elucidate its mechanisms of action. Clinical trials concerning celecoxib have focused primarily on the treatment of cancers of the colon, breast, lung, prostate, stomach, head and neck, as well as premalignant lesions such as familial adenoma polyposis. In this review article authors attempt to summarise the latest research which has elucidated celecoxib use in the treatment and prevention of cancer. CONCLUSION Both preclinical and clinical studies have demonstrated promising results of the role of celecoxib in the treatment and prevention of cancer - the best outcome was observed in colon, breast, prostate and head and neck cancers. However, more clinical trials providing real evidence-based clinical advances of celecoxib use are needed.
Collapse
Affiliation(s)
- Natalia Tołoczko-Iwaniuk
- Department of Pharmaceutical Analysis, Medical University of Bialystok, Mickiewicza 2D Street, 15-222 Bialystok, Poland
| | - Dorota Dziemiańczyk-Pakieła
- Department of Maxillofacial and Plastic Surgery, Medical University of Bialystok, Skłodowskiej-Curie 24A, 15-404 Bialystok, Poland
| | - Beata Klaudia Nowaszewska
- Department of Maxillofacial and Plastic Surgery, Medical University of Bialystok, Skłodowskiej-Curie 24A, 15-404 Bialystok, Poland
| | - Katarzyna Celińska-Janowicz
- Department of Pharmaceutical Analysis, Medical University of Bialystok, Mickiewicza 2D Street, 15-222 Bialystok, Poland
| | - Wojciech Miltyk
- Department of Pharmaceutical Analysis, Medical University of Bialystok, Mickiewicza 2D Street, 15-222 Bialystok, Poland
| |
Collapse
|
18
|
Anoopkumar-Dukie S, Conere T, Houston A, King L, Christie D, McDermott C, Allshire A. The COX-2 inhibitor NS398 selectively sensitizes hypoxic HeLa cells to ionising radiation by mechanisms both dependent and independent of COX-2. Prostaglandins Other Lipid Mediat 2020; 148:106422. [PMID: 32004752 DOI: 10.1016/j.prostaglandins.2020.106422] [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] [Received: 07/15/2019] [Revised: 11/24/2019] [Accepted: 01/24/2020] [Indexed: 02/06/2023]
Abstract
It is widely accepted that the hypoxic nature of solid tumors contribute to their resistance to radiation therapy. There is increasing evidence that cyclooxygenase-2 (COX-2) contributes to increased resistance of tumors to radiation therapy. Several studies demonstrate that combination of COX-2 selective inhibitors with radiation therapy selectively enhances radio responsiveness of tumor cells. However, the majority of these studies utilised suprapharmacological concentrations under normoxic conditions only. Furthermore, the mechanism by which these agents act remain largely unclear. Therefore, the aim of this study was to determine the impact of COX-2 selective inhibitors on both normoxic and hypoxic radiosensitivity in vitro and the mechanisms underlying this. Because of the close, reciprocal relationship between COX-2 and p53 we investigated their contribution to radioresistance. To achieve this we exposed HeLa, MCF-7 and MeWo cells to the COX-2 selective inhibitor, NS398 (10μM). NS398 (10μM) selectively sensitized hypoxic HeLa and MCF-7 but not MeWo cells to ionising radiation (5 Gy). Furthermore, while knockdown of COX-2 with siRNA did not affect either normoxic radiosensitivity in HeLa cells, the radiosensitisation observed with NS398 was lost suggesting both COX-2 dependent and independent mechanisms. We also show that ionising radiation at 5 Gy results in phosphorylation of p53 at serine 15, a key phosphorylation site for p53-mediated apoptosis, and that hypoxia attenuates this phosphorylation. Attenuated phosphorylation of p53 under hypoxic conditions may therefore contribute to hypoxic radioresistance. We also show that NS398 selectively phosphorylates p53 under hypoxic conditions following irradiation at 5 Gy. p53 phosphorylation could be an underlying mechanism by which this agent and other COX-2 inhibitors sensitize tumors to radiation therapy.
Collapse
Affiliation(s)
- Shailendra Anoopkumar-Dukie
- School of Pharmacy and Pharmacology, Griffith University, Queensland, Australia; Quality Use of Medicines Network, Griffith University, Queensland, Australia.
| | - Tom Conere
- Department of Medical Physics, Cork University Hospital, Wilton, Cork, Ireland
| | - Aileen Houston
- Department of Medicine, University College Cork, Cork, Ireland
| | - Liam King
- School of Pharmacy and Pharmacology, Griffith University, Queensland, Australia
| | | | - Catherine McDermott
- Centre for Urology Research, Bond University, Gold Coast, Queensland, Australia
| | - Ashley Allshire
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| |
Collapse
|
19
|
Kefayat A, Ghahremani F, Safavi A, Hajiaghababa A, Moshtaghian J. C-phycocyanin: a natural product with radiosensitizing property for enhancement of colon cancer radiation therapy efficacy through inhibition of COX-2 expression. Sci Rep 2019; 9:19161. [PMID: 31844085 PMCID: PMC6915779 DOI: 10.1038/s41598-019-55605-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022] Open
Abstract
Different chemical and nanomaterial agents have been introduced for radiosensitizing purposes. However, many researchers believe these agents are far away from clinical application due to side effects and limited knowledge about their behavior in the human body. In this study, C-phycocyanin (C-PC) was used as a natural radiosensitizer for enhancement of radiation therapy (RT) efficacy. C-PC treatment's effect on the COX-2 expression of cancer cells was investigated by flow cytometry, western blot, qRT-PCR analyses in vitro and in vivo. Subsequently, the radiosensitizing effect of C-PC treatment was investigated by MTT and clonogenic cell survival assays for CT-26, DLD-1, HT-29 colon cancer cell lines and the CRL-1831 as normal colonic cells. In addition, the C-PC treatment effect on the radiation therapy efficacy was evaluated according to CT-26 tumor's growth progression and immunohistochemistry analyses of Ki-67 labeling index. C-PC treatment (200 µg/mL) could significantly enhance the radiation therapy efficacy in vitro and in vivo. Synergistic interaction was detected at C-PC and radiation beams co-treatment based on Chou and Talalay formula (combination index <1), especially at 200 µg/mL C-PC and 6 Gy radiation dosages. The acquired DEF of C-PC treatment was 1.39, 1.4, 1.63, and 1.05 for CT-26, DLD-1, HT-29, and CRL-1831 cells, respectively. Also, C-PC + RT treated mice exhibited 35.2% lower mean tumors' volume and about 6 days more survival time in comparison with the RT group (P < 0.05). In addition, C-PC + RT group exhibited 54% lower Ki-67 index in comparison with the RT group. Therefore, C-PC can exhibit high radiosensitizing effects. However, the potential cardiovascular risks of C-PC as a COX-2 inhibitor should be evaluated with extensive preclinical testing before developing this agent for clinical trials.
Collapse
Affiliation(s)
- Amirhosein Kefayat
- Department of Oncology, Cancer Prevention Research Center, Isfahan University of Medical Sciences, Isfahan, 81746-73461, Iran
| | - Fatemeh Ghahremani
- Department of Medical Physics and Radiotherapy, Arak University of Medical Sciences, Arak, 38481-76941, Iran.
| | - Ashkan Safavi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Jamal Moshtaghian
- Division of Cell and Molecular Biology, Department of Biology, Faculty of Science, University of Isfahan, Isfahan, Iran
| |
Collapse
|
20
|
Mortezaee K, Najafi M, Farhood B, Ahmadi A, Potes Y, Shabeeb D, Musa AE. Modulation of apoptosis by melatonin for improving cancer treatment efficiency: An updated review. Life Sci 2019; 228:228-241. [DOI: 10.1016/j.lfs.2019.05.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022]
|
21
|
Semenok D, Medvedev J, Giassafaki LP, Lavdas I, Vizirianakis IS, Eleftheriou P, Gavalas A, Petrou A, Geronikaki A. 4,5-Diaryl 3( 2H)Furanones: Anti-Inflammatory Activity and Influence on Cancer Growth. Molecules 2019; 24:molecules24091751. [PMID: 31064095 PMCID: PMC6539231 DOI: 10.3390/molecules24091751] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 04/24/2019] [Accepted: 04/30/2019] [Indexed: 11/22/2022] Open
Abstract
Apart from their anti-inflammatory action, COX inhibitors have gathered the interest of many scientists due to their potential use for the treatment and prevention of cancer. It has been shown that cyclooxygenase inhibitors restrict cancer cell growth and are able to interact with known antitumor drugs, enhancing their in vitro and in vivo cytotoxicity. The permutation of hydrophilic and hydrophobic aryl groups in COX inhibitors leads to cardinal changes in the biological activity of the compounds. In the present study, thirteen heterocyclic coxib-like 4,5-diarylfuran-3(2H)-ones and their annelated derivatives—phenanthro[9,10-b]furan-3-ones—were synthesized and studied for anti-inflammatory and COX-1/2 inhibitory action and for their cytotoxic activity on the breast cancer (MCF-7) and squamous cell carcinoma (HSC-3) cell lines. The F-derivative of the –SOMe substituted furan-3(2H)-ones exhibited the best activity (COX-1 IC50 = 2.8 μM, anti-inflammatory activity (by carrageenan paw edema model) of 54% (dose 0.01 mmol/kg), and MCF-7 and HSC-3 cytotoxicity with IC50 values of 10 μM and 7.5 μM, respectively). A cytotoxic effect related to the COX-1 inhibitory action was observed and a synergistic effect with the anti-neoplastic drugs gefitinib and 5-fluorouracil was found. A phenanthrene derivative exhibited the best synergistic effect with gefitinib.
Collapse
Affiliation(s)
- Dmitrii Semenok
- Skolkovo Institute of Science and Technology, Skolkovo Innovation Center, 3 Nobel Street, 143026 Moscow, Russia.
- Moscow Institute of Physics and Technology, 9 Institutsky lane, 141700 Dolgoprudny, Russia.
| | - Jury Medvedev
- Saint-Petersburg State University, Institute of Chemistry, Universitetskiy Prospekt, 26, 198504 Petergof, Russia.
| | - Lefki-P Giassafaki
- Department of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Iason Lavdas
- Department of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Ioannis S Vizirianakis
- Department of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Phaedra Eleftheriou
- Department of Medical Laboratory Studies, School of Health and Medical Care, Alexander Technological Educational Institute of Thessaloniki, 57400 Thessaloniki, Greece.
| | - Antonis Gavalas
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Anthi Petrou
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| | - Athina Geronikaki
- Department of Pharmaceutical Chemistry, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece.
| |
Collapse
|
22
|
Banarouei N, Davood A, Shafaroodi H, Saeedi G, Shafiee A. N-arylmethylideneaminophthalimide: Design, Synthesis and Evaluation as Analgesic and Anti-inflammatory Agents. Mini Rev Med Chem 2019; 19:679-687. [DOI: 10.2174/1389557518666180424101009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 10/19/2017] [Accepted: 04/09/2018] [Indexed: 11/22/2022]
Abstract
Background and Objective:
N-aryl derivatives of phthalimide and 4-nitro phthalimide have
demonstrated cyclooxygenase inhibitory activity. Also, they possess excellent analgesic and antiinflammatory
activity. In this work, a new series of N-arylmethylideneamino derivatives of
phthalimide and 4-nitro phthalimide were designed and synthesized.
Methods:
The designed compounds were synthesized by condensation of the appropriate aldehyde and
N-aminophthalimide in ethanol at room temperature at PH around 3. Their analgesic and antiinflammatory
activity were evaluated by acetic acid-induced pain test and carrageenan-induced paw
edema test in mice and rats, respectively.
Results and Conclusion::
The details of the synthesis and chemical characterization of the analogs are
described. In vivo screening showed compounds 3a, 3b, 3f and 3h were the most potent analgesic
compounds. In addition, compounds 3a, 3c, 3d, 3e and 3j indicated comparable anti-inflammatory activity
to indomethacin as a reference drug.
Collapse
Affiliation(s)
- Nasimossadat Banarouei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Asghar Davood
- Department of Medicinal Chemistry, Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Shafaroodi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Saeedi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Abbas Shafiee
- Department of Medicinal Chemistry, Faculty of Pharmacy and Pharmaceutical Research Center, Tehran University of Medical Sciences, Iran
| |
Collapse
|
23
|
Kim W, Son B, Lee S, Do H, Youn B. Targeting the enzymes involved in arachidonic acid metabolism to improve radiotherapy. Cancer Metastasis Rev 2018; 37:213-225. [DOI: 10.1007/s10555-018-9742-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
24
|
Zhong J, Yu X, Dong X, Lu H, Zhou W, Li L, Li Z, Sun P, Shi X. Therapeutic role of meloxicam targeting secretory clusterin-mediated invasion in hepatocellular carcinoma cells. Oncol Lett 2018; 15:7191-7199. [PMID: 29731881 PMCID: PMC5920948 DOI: 10.3892/ol.2018.8186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 02/28/2018] [Indexed: 12/23/2022] Open
Abstract
Recurrence and metastasis are the two leading causes of poor prognosis in patients with hepatocellular carcinoma (HCC). Secreted clusterin (sCLU) is a stress-induced chaperone that is overexpressed in HCC. However, the precise molecular mechanisms of sCLU in HCC invasion and migration are largely unknown. In the present study, it was indicated that downregulation of sCLU significantly alleviated invasiveness whereas overexpression of sCLU notably enhanced the number of invasive cells via mediating the expression level of MMP-2 and E-cadherin in Bel-7402 and SMMC-7721 cells. Furthermore, as an important mediator of invasiveness, sCLU may be responsible for proliferation and invasion suppression induced by meloxicam (a selective inhibitor of cyclooxygenase-2) in HCC cells. The combination of meloxicam and CLU shRNA significantly decreased invasion in HCC cells in vitro. Furthermore, it was observed that overexpression of sCLU significantly potentiated expression of p-AKT and MMP-2. However, downregulation of sCLU by CLU shRNA alleviated the extent of p-AKT. These results suggest the targeting of sCLU may be a novel therapeutic strategy against invasion and migration in HCC.
Collapse
Affiliation(s)
- Jingtao Zhong
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Xiaoming Yu
- Department of Ophthalmology, Shandong Jiaotong Hospital, Jinan, Shandong 250031, P.R. China
| | - Xiaofeng Dong
- Department of Hepatobiliary Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Hong Lu
- Department of Radiology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, Shandong 250117, P.R. China
| | - Wuyuan Zhou
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Lei Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Zhongchao Li
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Pengfei Sun
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| | - Xuetao Shi
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
25
|
Sekandarzad MW, van Zundert AAJ, Lirk PB, Doornebal CW, Hollmann MW. Perioperative Anesthesia Care and Tumor Progression. Anesth Analg 2017; 124:1697-1708. [PMID: 27828796 DOI: 10.1213/ane.0000000000001652] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This narrative review discusses the most recent up-to-date findings focused on the currently available "best clinical practice" regarding perioperative anesthesia care bundle factors and their effect on tumor progression. The main objective is to critically appraise the current literature on local anesthetics, regional outcome studies, opioids, and nonsteroidal anti-inflammatory drugs (NSAIDs) and their ability to decrease recurrence in patients undergoing cancer surgery. A brief discussion of additional topical perioperative factors relevant to the anesthesiologist including volatile and intravenous anesthetics, perioperative stress and anxiety, nutrition, and immune stimulation is included. The results of several recently published systematic reviews looking at the association between cancer recurrences and regional anesthesia have yielded inconclusive data and provide insufficient evidence regarding a definitive benefit of regional anesthesia. Basic science data suggests an anti tumor effect induced by local anesthetics. New refined animal models show that opioids can safely be used for perioperative pain management. Preliminary evidence suggests that NSAIDs should be an essential part of multimodal analgesia. Volatile anesthetics have been shown to increase tumor formation, whereas preclinical and emerging clinical data from propofol indicate tumor protective qualities. The perioperative period in the cancer patient represents a unique environment where surgically mediated stress response leads to immune suppression. Regional anesthesia techniques when indicated in combination with multimodal analgesia that include NSAIDs, opioids, and local anesthetics to prevent the pathophysiologic effects of pain and neuroendocrine stress response should be viewed as an essential part of balanced anesthesia.
Collapse
Affiliation(s)
- Mir W Sekandarzad
- From the *Department of Anaesthesia and Perioperative Medicine, Royal Brisbane and Women's Hospital, The University of Queensland, Herston-Brisbane, Queensland, Australia; and †Division of Anesthesiology, Intensive Care, Emergency Medicine, Pain Therapy and Palliative Care, University Medical Center Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | |
Collapse
|
26
|
Prathyusha AMVN, Raghu G, Bramhachari PV. HIF-1α: Its Role in Metastasis of Oesophageal Malignancy. ROLE OF TRANSCRIPTION FACTORS IN GASTROINTESTINAL MALIGNANCIES 2017:73-89. [DOI: 10.1007/978-981-10-6728-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
27
|
Rahmanian N, Hosseinimehr SJ, Khalaj A. The paradox role of caspase cascade in ionizing radiation therapy. J Biomed Sci 2016; 23:88. [PMID: 27923354 PMCID: PMC5142153 DOI: 10.1186/s12929-016-0306-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/30/2016] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy alone or in combination with chemotherapy/surgery is widely used for treatment of cancers. It reduces tumor growth and prevents metastasis. While ionizing radiation activates caspase cascade resulted in apoptosis in cancer cells, it also stimulates tumor cell re-population that leads to reduce the effectiveness of the radiation therapy. This review describes the mechanisms for paradox role of caspase cascade in cancer therapy and discusses the logical and practical strategies for improvement the therapeutic index of radiotherapy through enhancement of radiosensitivity and decreasing the rate of tumor recurrence.
Collapse
Affiliation(s)
- Najmeh Rahmanian
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Khalaj
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Li T, Zhong J, Dong X, Xiu P, Wang F, Wei H, Wang X, Xu Z, Liu F, Sun X, Li J. Meloxicam suppresses hepatocellular carcinoma cell proliferation and migration by targeting COX-2/PGE2-regulated activation of the β-catenin signaling pathway. Oncol Rep 2016; 35:3614-3622. [PMID: 27109804 DOI: 10.3892/or.2016.4764] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/17/2016] [Indexed: 11/05/2022] Open
Abstract
Recurrence and metastasis are the two leading causes of poor prognosis of hepatocellular carcinoma (HCC) patients. Cyclooxygenase (COX)-2 is overexpressed in many types of cancers including HCC and promotes its metastasis. Meloxicam is a selective COX-2 inhibitor that has been reported to exert an anti-proliferation and invasion/migration response in various tumors. In this study, we examined the role of meloxicam on HCC cell proliferation and migration and explored the molecular mechanisms underlying this effect. We found that meloxicam inhibited HCC cell proliferation and had a cell cycle arrest effect in human HCC cells. Furthermore, meloxicam suppressed the ability of HCC cells expressing higher levels of COX-2 and prostaglandin E2 (PGE2) to migration via potentiating expression of E-cadherin and alleviating expression of matrix metalloproteinase (MMP)-2 and -9. COX-2/PGE2 has been considered to activate the β-catenin signaling pathway which promotes cancer cell migration. We found that treatment with PGE2 significantly enhanced nuclear accumulation of β-catenin and the activation of GSK3β which could be reversed by meloxicam in HCC cells. We also observed that HCC cell migration and upregulation of the level of MMP-2/9 and downregulation of E-cadherin induced by PGE2 were suppressed by FH535, an inhibitor of β-catenin. Taken together, these findings provide a new treatment strategy against HCC proliferation and migration.
Collapse
Affiliation(s)
- Tao Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Jingtao Zhong
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Xiaofeng Dong
- Department of Hepatobiliary Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530021, P.R. China
| | - Peng Xiu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Fuhai Wang
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Honglong Wei
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Xin Wang
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Zongzhen Xu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Feng Liu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Xueying Sun
- The Hepatosplenic Surgery Center, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jie Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
29
|
Sahu RP, Harrison KA, Weyerbacher J, Murphy RC, Konger RL, Garrett JE, Chin-Sinex HJ, Johnston ME, Dynlacht JR, Mendonca M, McMullen K, Li G, Spandau DF, Travers JB. Radiation therapy generates platelet-activating factor agonists. Oncotarget 2016; 7:20788-20800. [PMID: 26959112 PMCID: PMC4991492 DOI: 10.18632/oncotarget.7878] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/06/2016] [Indexed: 01/22/2023] Open
Abstract
Pro-oxidative stressors can suppress host immunity due to their ability to generate oxidized lipid agonists of the platelet-activating factor-receptor (PAF-R). As radiation therapy also induces reactive oxygen species, the present studies were designed to define whether ionizing radiation could generate PAF-R agonists and if these lipids could subvert host immunity. We demonstrate that radiation exposure of multiple tumor cell lines in-vitro, tumors in-vivo, and human subjects undergoing radiation therapy for skin tumors all generate PAF-R agonists. Structural characterization of radiation-induced PAF-R agonistic activity revealed PAF and multiple oxidized glycerophosphocholines that are produced non-enzymatically. In a murine melanoma tumor model, irradiation of one tumor augmented the growth of the other (non-treated) tumor in a PAF-R-dependent process blocked by a cyclooxygenase-2 inhibitor. These results indicate a novel pathway by which PAF-R agonists produced as a byproduct of radiation therapy could result in tumor treatment failure, and offer important insights into potential therapeutic strategies that could improve the overall antitumor effectiveness of radiation therapy regimens.
Collapse
Affiliation(s)
- Ravi P. Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, USA
| | - Kathleen A. Harrison
- Department of Pharmacology, University of Colorado Health Sciences Center, Aurora, CO, USA
| | - Jonathan Weyerbacher
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Robert C. Murphy
- Department of Pharmacology, University of Colorado Health Sciences Center, Aurora, CO, USA
| | - Raymond L. Konger
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joy Elizabeth Garrett
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Helen Jan Chin-Sinex
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Joseph R. Dynlacht
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Marc Mendonca
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kevin McMullen
- Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gengxin Li
- Department of Biostatistics, Wright State University, Dayton, OH, USA
| | - Dan F. Spandau
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jeffrey B. Travers
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH, USA
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN, USA
- The Dayton V.A. Medical Center, Dayton, OH, USA
| |
Collapse
|
30
|
Zhong J, Xiu P, Dong X, Wang F, Wei H, Wang X, Xu Z, Liu F, Li T, Wang Y, Li J. Meloxicam combined with sorafenib synergistically inhibits tumor growth of human hepatocellular carcinoma cells via ER stress-related apoptosis. Oncol Rep 2015; 34:2142-50. [PMID: 26252057 DOI: 10.3892/or.2015.4181] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 07/10/2015] [Indexed: 11/05/2022] Open
Abstract
Sorafenib (SOR) is a promising treatment for advanced hepatocellular carcinoma (HCC). However, the precise mechanisms of toxicity and drug resistance have not been fully explored and new strategies are urgently needed for HCC therapy. Meloxicam (MEL) is a selective cyclooxygenase-2 (COX-2) inhibitor which elicits antitumor effects in human HCC cells. In the present study, we investigated the interaction between MEL and SOR in human SMMC‑7721 cells and the role endoplasmic reticulum (ER) stress exerts in the combination of SOR with MEL treatment-induced cytotoxicity. Our results revealed that the combination treatment synergistically inhibited cell proliferation and enhanced apoptosis. Furthermore, the combination treatment enhanced ER stress-related molecules which involved in SMMC-7721 cell apoptosis. GRP78 knockdown by siRNA or co-treatment with MG132 significantly increased this combination treatment-induced apoptosis. In addition, we found that the combination treatment suppressed tumor growth by way of activation of ER stress in in vivo models. We concluded that the combination of SOR with MEL treatment-induced ER stress, and eventually apoptosis in human SMMC-7721 cells. Knockdown of GRP78 using siRNA or proteosome inhibitor enhanced the cytotoxicity of the combination of SOR with MEL-treatment in SMMC-7721 cells. These findings provided a new potential treatment strategy against HCC.
Collapse
Affiliation(s)
- Jingtao Zhong
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Peng Xiu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Xiaofeng Dong
- Department of General Surgery, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, P.R. China
| | - Fuhai Wang
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Honglong Wei
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Xin Wang
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Zongzhen Xu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Feng Liu
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Tao Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yong Wang
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Jie Li
- Department of General Surgery, Qianfoshan Hospital, Shandong University, Jinan, Shandong, P.R. China
| |
Collapse
|
31
|
Akutsu Y, Matsubara H. Chemoradiotherapy and surgery for T4 esophageal cancer in Japan. Surg Today 2015; 45:1360-5. [PMID: 25583206 DOI: 10.1007/s00595-015-1116-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 12/23/2014] [Indexed: 01/06/2023]
Abstract
Esophageal cancer is thought to be the most malignant neoplasm due to its biological aggressiveness. The most effective treatment modality for esophageal cancer, particularly T4 esophageal cancer, is chemoradiotherapy (CRT). Some T4 patients show long-term survival after receiving CRT, suggesting that even T4 esophageal cancer can be cured with this modality. Although surgery is performed after CRT in some T4 cases, its prognostic benefit is controversial. In this paper, we review the status of CRT and conversion surgery as well as the development of new regimens and discuss the future prospects of CRT in the treatment of T4 esophageal cancer in Japan.
Collapse
Affiliation(s)
- Yasunori Akutsu
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuoku, Chiba, 260-8670, Japan.
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, 1-8-1, Inohana, Chuoku, Chiba, 260-8670, Japan
| |
Collapse
|
32
|
Zeng L, Zhen Y, Chen Y, Zou L, Zhang Y, Hu F, Feng J, Shen J, Wei B. Naringin inhibits growth and induces apoptosis by a mechanism dependent on reduced activation of NF‑κB/COX‑2‑caspase-1 pathway in HeLa cervical cancer cells. Int J Oncol 2014; 45:1929-36. [PMID: 25174821 DOI: 10.3892/ijo.2014.2617] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 07/01/2014] [Indexed: 11/05/2022] Open
Abstract
Naringin (NRG), a bioflavonoid found in citrus fruit extracts, has been pharmacologically evaluated as a potential anticancer agent. This study confirmed a novel mechanism of the anticancer effects of NRG in the human cervical cancer HeLa cell line (HeLa cells). Exposure of HeLa cells to NRG resulted in growth inhibition, as evidenced by a decrease in cell viability. In addition, NRG treatment induced apoptosis, as indicated by the increased apoptotic percentage and the cleaved caspase-3 expression. Importantly, exposure of the cells to NRG attenuated the expression levels of phosphorylated (p) nuclear factor κB (NF-κB) p65 subunit, cyclooxygenase-2 (COX-2) and cysteinyl aspartate proteinase-1 (caspase-1). Treatment with PDTC (an inhibitor of NF-κB) or NS-398 (an inhibitor of COX-2) or SC-3069 (an inhibitor of caspase-1) markedly induced growth inhibition and apoptosis. Treatment with PDTC or NS-398 also reduced caspase-1 expression. Interestingly, PDTC treatment blocked the expression of COX-2 and NS-398 reduced the p-NF-κB p65 expression. Taken together, this study provides novel evidence that NRG induces growth inhibition and apoptosis by inhibiting the NF-κB/COX-2-caspase-1 pathway and that a positive interaction between NF-κB and COX-2 pathway contributes to the growth and antiapoptotic effect in HeLa cells.
Collapse
Affiliation(s)
- Lan Zeng
- Department of Gynecology, Liwan Chinese Traditional Medicine Hospital, Guangzhou, Guangdong 510140, P.R. China
| | - Yulan Zhen
- Department of Oncology, The Affiliated Hospital, Guangdong Medical College, Zhanjiang 524001, P.R. China
| | - Yiming Chen
- Department of Gynecology, Liwan Chinese Traditional Medicine Hospital, Guangzhou, Guangdong 510140, P.R. China
| | - Lin Zou
- Department of Reproductive Medical Center, The Affiliated Hospital, Guangdong Medical College, Zhanjiang 524001, P.R. China
| | - Ying Zhang
- Department of Oncology, The Affiliated Hospital, Guangdong Medical College, Zhanjiang 524001, P.R. China
| | - Fen Hu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jianqiang Feng
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jianhua Shen
- Department of Gynecology, Liwan Chinese Traditional Medicine Hospital, Guangzhou, Guangdong 510140, P.R. China
| | - Bing Wei
- Department of Reproductive Medical Center, The Affiliated Hospital, Guangdong Medical College, Zhanjiang 524001, P.R. China
| |
Collapse
|