1
|
Rodrigues P, Bangali H, Hammoud A, Mustafa YF, Al-Hetty HRAK, Alkhafaji AT, Deorari MM, Al-Taee MM, Zabibah RS, Alsalamy A. COX 2-inhibitors; a thorough and updated survey into combinational therapies in cancers. Med Oncol 2024; 41:41. [PMID: 38165473 DOI: 10.1007/s12032-023-02256-7] [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: 10/11/2023] [Accepted: 11/15/2023] [Indexed: 01/03/2024]
Abstract
Cyclooxygenase (COX) enzymes are pivotal in inflammation and cancer development. COX-2, in particular, has been implicated in tumor growth, angiogenesis, and immune evasion. Recently, COX-2 inhibitors have arisen as potential therapeutic agents in cancer treatment. In addition, combining COX inhibitors with other treatment modalities has demonstrated the potential to improve therapeutic efficacy. This review aims to investigate the effects of COX inhibition, both alone and in combination with other methods, on signaling pathways and carcinogenesis in various cancers. In this study, a literature search of all major academic databases was conducted (PubMed, Scholar google), including the leading research on the mechanisms of COX-2, COX-2 inhibitors, monotherapy with COX-2 inhibitors, and combining COX-2-inhibitors with chemotherapeutic agents in tumors. The study encompasses preclinical and clinical evidence, highlighting the positive findings and the potential implications for clinical practice. According to preclinical studies, multiple signaling pathways implicated in tumor cell proliferation, survival, invasion, and metastasis can be suppressed by inhibiting COX. In addition, combining COX inhibitors with chemotherapy drugs, targeted therapies, immunotherapies, and miRNA-based approaches has enhanced anti-tumor activity. These results suggest that combination therapy has the potential to overcome resistance mechanisms and improve treatment outcomes. However, caution must be exercised when selecting and administering combination regimens. Not all combinations of COX-2 inhibitors with other drugs result in synergistic effects; some may even have unfavorable interactions. Therefore, personalized approaches that consider the specific characteristics of the cancer and the medications involved are crucial for optimizing therapeutic strategies. In conclusion, as monotherapy or combined with other methods, COX inhibition bears promise in modulating signaling pathways and inhibiting carcinogenesis in various cancers. Additional studies and well-designed clinical trials are required to completely elucidate the efficacy of COX inhibition and combination therapy in enhancing cancer treatment outcomes. This narrative review study provides a detailed summary of COX-2 monotherapy and combination targeted therapy in cancer treatment.
Collapse
Affiliation(s)
- Paul Rodrigues
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Asir-Abha, Kingdom of Saudi Arabia
| | - Harun Bangali
- Department of Computer Engineering, College of Computer Science, King Khalid University, Al-Faraa, Asir-Abha, Kingdom of Saudi Arabia
| | - Ahmad Hammoud
- Department of Medical and Technical Information Technology, Bauman Moscow State Technical University, Moscow, Russia.
- Department of Mathematics and Natural Sciences, Gulf University for Science and Technology, Mishref Campus, Mubarak Al-Abdullah, Kuwait.
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | | | | | - Maha Medha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Rahman S Zabibah
- College of Medical Technique, the Islamic University, Najaf, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, 66002, Iraq
| |
Collapse
|
2
|
Xu W, Yu H, Zhao R, Liang Y. Investigation of mitochondrial targeting ability of sydnones and sydnonimines and mitochondria-targeted delivery of celecoxib. Bioorg Med Chem Lett 2023; 81:129129. [PMID: 36634752 DOI: 10.1016/j.bmcl.2023.129129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/24/2022] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
Mitochondria are considered to be a promising target in cancer diagnosis and therapeutics. Recently, sydnone and sydnonimine, as mesoionic bioorthogonal reagents, have been used in cell labeling and drug delivery. Here we investigated the mitochondrial targeting ability of sydnones and sydnonimines for the first time. Experimental results show that sydnone and sydnonimine themselves have high mitochondrial distribution. However, the introduction of a phenyl group into the C4 position of sydnone dramatically decreases the mitochondrial affinity. In addition, we took advantage of mitochondrial targeting ability and click-and-release reaction of sydnonimine to evaluate anticancer activities of in-mitochondria delivery of celecoxib against HeLa and HepG2 cells, indicating that celecoxib-induced cancer cell death may not involve mitochondria-related pathway.
Collapse
Affiliation(s)
- Wenyuan Xu
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Hongzhe Yu
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ruohan Zhao
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yong Liang
- State Key Laboratory of Coordination Chemistry, Jiangsu Key Laboratory of Advanced Organic Materials, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
3
|
Ramos-Inza S, Ruberte AC, Sanmartín C, Sharma AK, Plano D. NSAIDs: Old Acquaintance in the Pipeline for Cancer Treatment and Prevention─Structural Modulation, Mechanisms of Action, and Bright Future. J Med Chem 2021; 64:16380-16421. [PMID: 34784195 DOI: 10.1021/acs.jmedchem.1c01460] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The limitations of current chemotherapeutic drugs are still a major issue in cancer treatment. Thus, targeted multimodal therapeutic approaches need to be strategically developed to successfully control tumor growth and prevent metastatic burden. Inflammation has long been recognized as a hallmark of cancer and plays a key role in the tumorigenesis and progression of the disease. Several epidemiological, clinical, and preclinical studies have shown that traditional nonsteroidal anti-inflammatory drugs (NSAIDs) exhibit anticancer activities. This Perspective reports the most recent outcomes for the treatment and prevention of different types of cancers for several NSAIDs alone or in combination with current chemotherapeutic drugs. Furthermore, an extensive review of the most promising structural modifications is reported, such as phospho, H2S, and NO releasing-, selenium-, metal complex-, and natural product-NSAIDs, among others. We also provide a perspective about the new strategies used to obtain more efficient NSAID- or NSAID derivative- formulations for targeted delivery.
Collapse
Affiliation(s)
- Sandra Ramos-Inza
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Ana Carolina Ruberte
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Arun K Sharma
- Department of Pharmacology, Penn State Cancer Institute, CH72, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Daniel Plano
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| |
Collapse
|
4
|
Ahmed KS, Liu S, Mao J, Zhang J, Qiu L. Dual-Functional Peptide Driven Liposome Codelivery System for Efficient Treatment of Doxorubicin-Resistant Breast Cancer. Drug Des Devel Ther 2021; 15:3223-3239. [PMID: 34349500 PMCID: PMC8326382 DOI: 10.2147/dddt.s317454] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The active-targeted drug delivery systems had attracted more and more attention to efficiently overcome multidrug resistance (MDR) in cancer treatments. The aim of the work was to develop a multifunctional nano-structured liposomal system for co-delivery of doxorubicin hydrochloride (DOX) and celecoxib (CEL) to overcome doxorubicin resistance in breast cancer. METHODS A functional hybrid peptide (MTS-R8H3) with unique cellular penetrability, endo-lysosomal escape and mitochondrial targeting ability was successfully synthesized using solid phase synthesis technology. The peptide modified targeted liposomes (DOX/CEL-MTS-R8H3 lipo) for co-delivery of DOX and CEL were formulated to overcome the chemoresistance in MCF/ADR cells. RESULTS DOX/CEL-MTS-R8H3 lipo showed nanosized shape and displayed high stability for one month. The cytotoxicity effect of the co-delivery of DOX and CEL through peptide modified liposomes had remarkable treatment efficacy on killing MCF/ADR cells. Targeted liposome exhibited greater cellular entry ability about 5.72-fold stronger than DOX solution. Moreover, as compared with unmodified liposomes, the presence of MTS-R8H3 peptide entity on liposome surface enhanced the mitochondrial-targeting ability and achieved effective reactive oxygen species (ROS) production with significant inhibition of P-gp efflux activity. CONCLUSION The study suggested that the DOX/CEL-MTS-R8H3 lipo is a promising strategy for overcoming drug resistance in breast cancer treatments with high targeting inhibition efficiency.
Collapse
Affiliation(s)
- Kamel S Ahmed
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 19623, Egypt
| | - Shenhuan Liu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
| | - Jing Mao
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
| | - Jie Zhang
- The Jiaxing Key Laboratory of Oncological Photodynamic Therapy and the Targeted Drug Research, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, 314001, People’s Republic of China
| | - Lipeng Qiu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
| |
Collapse
|
5
|
Watson J, Ninh MK, Ashford S, Cornett EM, Kaye AD, Urits I, Viswanath O. Anesthesia Medications and Interaction with Chemotherapeutic Agents. Oncol Ther 2021; 9:121-138. [PMID: 33861416 PMCID: PMC8140172 DOI: 10.1007/s40487-021-00149-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/13/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer is now a leading health concern worldwide. In an effort to provide these patients with adequate care, coordination between anesthesiologists and surgeons is crucial. In cancer-related treatment, it is very clear that radio-chemotherapy and medical procedures are important. There are some obstacles to anesthesia when dealing with cancer treatment, such as physiological disturbances, tumor-related symptoms, and toxicity in traditional chemotherapy treatment. Therefore, it is important that a multisystemic, multidisciplinary and patient-centered approach is used to preserve perioperative homeostasis and immune function integrity. Adding adjuvants can help increase patient safety and satisfaction and improve clinical efficacy. Correctly paired anesthetic procedures and medications will reduce perioperative inflammatory and immune changes that could potentially contribute to improved results for future cancer patients. Further research into best practice strategies is required which will help to enhance the acute and long-term effects of cancer care in clinical practice.
Collapse
Affiliation(s)
- Jeremy Watson
- LSU Health Shreveport, 1501 Kings Hwy, Shreveport, LA 71103 USA
| | - Michael K. Ninh
- LSU Health Shreveport, 1501 Kings Hwy, Shreveport, LA 71103 USA
| | - Scott Ashford
- LSU Health Shreveport, 1501 Kings Hwy, Shreveport, LA 71103 USA
| | - Elyse M. Cornett
- Department of Anesthesiology, LSU Health Shreveport, 1501 Kings Highway, P.O. Box 33932, Shreveport, LA 71103 USA
| | - Alan David Kaye
- Departments of Anesthesiology and Pharmacology, Toxicology, and Neurosciences, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71103 USA
| | - Ivan Urits
- Department of Anesthesia, Critical Care, and Pain Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215 USA
| | - Omar Viswanath
- Department of Anesthesiology, Louisiana State University Shreveport, Shreveport, LA USA
- Valley Pain Consultants – Envision Physician Services, Phoenix, AZ USA
- Department of Anesthesiology, University of Arizona College of Medicine-Phoenix, Phoenix, AZ USA
- Department of Anesthesiology, Creighton University School of Medicine, Omaha, NE USA
| |
Collapse
|
6
|
Srivastava S, Dewangan J, Mishra S, Divakar A, Chaturvedi S, Wahajuddin M, Kumar S, Rath SK. Piperine and Celecoxib synergistically inhibit colon cancer cell proliferation via modulating Wnt/β-catenin signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 84:153484. [PMID: 33667839 DOI: 10.1016/j.phymed.2021.153484] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Celecoxib (CXB), a selective COX-2 inhibitor NSAID, has exhibited prominent anti-proliferative potential against numerous cancers. However, its low bioavailability and long term exposure related cardiovascular side effects, limit its clinical application. In order to overcome these limitations, natural bioactive compounds with lower toxicity profile are used in combination with therapeutic drugs. Therfore, in this study Piperine (PIP), a natural chemo-preventive agent possessing drug bioavailability enhancing properties, was considered to be used in combination with low doses of CXB. PURPOSE We hypothesized that the combination of PIP with CXB will have a synergistic anti-proliferative effect on colon cancer cells. STUDY DESIGN The potency of PIP and CXB alone and in combination was evaluated in HT-29 human colon adenocarcinoma cells and mechanism of growth inhibition was investigated by analyzing the players in apoptotic and Wnt/β-catenin signaling pathways. METHODS The effect of PIP on the oral bioavailability of CXB in mice was investigated using HPLC analysis. The study investigated the synergistic anti-proliferative effect of CXB and PIP on HT-29 cells and IEC-6 non-tumorigenic rat intestinal epithelial cells by SRB cell viability assay. Further, the cellular and molecular mechanism(s) involved in the anti-proliferative combinatorial effect was extensively explored in HT-29 cells by flow cytometry and western blotting. The in vivo efficacy of this combination was studied in CT26.WT tumor syngeneic Balb/c mice model. RESULTS PIP as a bioenhancer increased the oral bioavailability of CXB (129%). The IC50 of CXB and PIP were evaluated to select doses for combination treatment of HT-29 cells. The drug combinations having combination index (CI) less than 1 were screened using CompuSyn software. These combinations were significantly cytotoxic to HT-29 cells but IEC-6 were least effected. Further, the mechanism behind CXB and PIP mediated cell death was explored. The co-treatment led to reactive oxygen species generation, mitochondrial dysfunction, caspase activation and enhanced apoptosis in HT-29 cells. Additionally, the combination treatment synergistically modulated Wnt/β-catenin pathway, downregulated the stemness markers and boosted therapeutic response in CT26 syngeneic Balb/c mice. CONCLUSION The outcomes of the study suggests that combining CXB and PIP offers a novel approach for the treatment of colon cancer.
Collapse
Affiliation(s)
- Sonal Srivastava
- Genotoxicity Laboratory, Division of Toxicology and Experimental Medicine, Lucknow 226031, Uttar Pradesh, India
| | - Jayant Dewangan
- Genotoxicity Laboratory, Division of Toxicology and Experimental Medicine, Lucknow 226031, Uttar Pradesh, India
| | - Sakshi Mishra
- Genotoxicity Laboratory, Division of Toxicology and Experimental Medicine, Lucknow 226031, Uttar Pradesh, India
| | - Aman Divakar
- Pharmacokinetics and Metabolism Division, CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| | - Swati Chaturvedi
- Pharmacokinetics and Metabolism Division, CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| | - Muhammad Wahajuddin
- Pharmacokinetics and Metabolism Division, CSIR-Central Drug Research Institute, Lucknow 226031, Uttar Pradesh, India
| | - Sadan Kumar
- Immunotoxicology laboratory Division of Toxicology and Experimental Medicine, Lucknow 226031, Uttar Pradesh, India
| | - Srikanta Kumar Rath
- Genotoxicity Laboratory, Division of Toxicology and Experimental Medicine, Lucknow 226031, Uttar Pradesh, India.
| |
Collapse
|
7
|
High ROS Production by Celecoxib and Enhanced Sensitivity for Death Ligand-Induced Apoptosis in Cutaneous SCC Cell Lines. Int J Mol Sci 2021; 22:ijms22073622. [PMID: 33807213 PMCID: PMC8036359 DOI: 10.3390/ijms22073622] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/24/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
Incidence of cutaneous squamous cell carcinoma (cSCC) and actinic keratosis has increased worldwide, and non-steroidal anti-inflammatory drugs as celecoxib are considered for treatment. We show here strong anti-proliferative effects of celecoxib in four cSCC cell lines, while apoptosis and cell viability largely remained unaffected. Impeded apoptosis was overcome in combinations with agonistic CD95 antibody or TNF-related apoptosis-inducing ligand (TRAIL), resulting in up to 60% apoptosis and almost complete loss of cell viability. Proapoptotic caspase cascades were activated, and apoptosis was suppressed by caspase inhibition. TRAIL receptor (DR5) and proapoptotic Bcl-2 proteins (Puma and Bad) were upregulated, while anti-apoptotic factors (survivin, XIAP, cFLIP, Mcl-1, and Bcl-w) were downregulated. Strongly elevated levels of reactive oxygen species (ROS) turned out as particularly characteristic for celecoxib, appearing already after 2 h. ROS production alone was not sufficient for apoptosis induction but may play a critical role in sensitizing cancer cells for apoptosis and therapy. Thus, the full therapeutic potential of celecoxib may be better used in combinations with death ligands. Furthermore, the immune response against cSCC/AK may be improved by celecoxib, and combinations with checkpoint inhibitors, recently approved for the treatment of cSCC, may be considered.
Collapse
|
8
|
Monteleone NJ, Lutz CS. miR-708-5p targets oncogenic prostaglandin E2 production to suppress a pro-tumorigenic phenotype in lung cancer cells. Oncotarget 2020; 11:2464-2483. [PMID: 32655834 PMCID: PMC7335672 DOI: 10.18632/oncotarget.27614] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
Many cancers maintain an inflammatory microenvironment to promote their growth. Lung cancer is of particular importance, as it is the deadliest cancer worldwide. One inflammatory pathway commonly dysregulated in cancer is the metabolism of arachidonic acid (AA) by Cyclooxygenase-2 (COX-2) and microsomal Prostaglandin E Synthase 1 (mPGES-1) into Prostaglandin E2 (PGE2). While researchers have identified PGE2's pro-tumorigenic functions, the mechanisms governing overexpression of COX-2 and mPGES-1 are incompletely understood. MicroRNAs (miRNAs) are important post-transcriptional regulators commonly dysregulated in cancer. Interestingly, miR-708-5p (miR-708) is predicted to target both COX-2 and mPGES-1. In this study, we show that high miR-708 expression is associated with survival rates in lung squamous cell carcinoma patients. miR-708 also represses PGE2 production by suppressing both COX-2 and mPGES-1 expression in lung cancer cells. miR-708 regulation of COX-2 and mPGES-1 is mediated through targeting of their 3' untranslated regions (UTRs). Moreover, miR-708 decreases proliferation, survival, and migration of lung cancer cells, which can be partially attributed to miR-708's inhibition of PGE2 signaling. Lastly, we identify novel miR-708 predicted targets and possible regulators of miR-708 expression in lung cancer. Collectively, these data demonstrate that dysregulated miR-708 expression contributes to exacerbated PGE2 production, leading to an enhanced pro-tumorigenic phenotype in lung cancer cells.
Collapse
Affiliation(s)
- Nicholas J. Monteleone
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical & Health Sciences, New Jersey Medical School, School of Graduate Studies, Newark, NJ 07103, USA
| | - Carol S. Lutz
- Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers Biomedical & Health Sciences, New Jersey Medical School, School of Graduate Studies, Newark, NJ 07103, USA
| |
Collapse
|
9
|
Li S, Jiang M, Wang L, Yu S. Combined chemotherapy with cyclooxygenase-2 (COX-2) inhibitors in treating human cancers: Recent advancement. Biomed Pharmacother 2020; 129:110389. [PMID: 32540642 DOI: 10.1016/j.biopha.2020.110389] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/28/2020] [Accepted: 06/07/2020] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy with a single chemotherapeutic agent or a combined chemotherapeutic regimen is the clinically standardized treatment for almost all human cancers. Upregulated expression of cyclooxygenase (COX)-2, also known as prostaglandin-endoperoxide synthase (PTGS), is associated with human carcinogenesis and cancer progression and COX-2 inhibitors show antitumor activity in different human cancers. Thus, a combination of chemotherapeutic agents with COX-2 inhibitors has been shown to improve therapeutic effects on human cancers. This review discusses and summarizes recent advances in cancer control and treatment using various antineoplastic drugs combined with COX-2 inhibitors. These combinations showed synergistic antitumor effects. At the gene level, COX-2 inhibitors can reduce inflammatory factors thereby regulating macrophage recruitment for activating the antitumor immune microenvironment; downregulating vascular endothelial growth factor (VEGF) to inhibit tumor angiogenesis; and inhibiting the PI3K/Akt signaling pathway to induce tumor cell apoptosis. In addition, such a combination can reduce toxicity and chemoresistance and enhance radiosensitivity, although COX-2 inhibitors-related cardiotoxicity may potentially affect its use. Further in-depth investigation of these drug combinations is needed to maximize antitumor efficacy and minimize the side effects.
Collapse
Affiliation(s)
- Shuangshuang Li
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Min Jiang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Lu Wang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China.
| | - Shuwen Yu
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Department of Pharmacy, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250013, China.
| |
Collapse
|
10
|
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
|
11
|
Mármol I, Castellnou P, Alvarez R, Gimeno MC, Rodríguez-Yoldi MJ, Cerrada E. Alkynyl Gold(I) complexes derived from 3-hydroxyflavones as multi-targeted drugs against colon cancer. Eur J Med Chem 2019; 183:111661. [PMID: 31546196 DOI: 10.1016/j.ejmech.2019.111661] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/20/2019] [Accepted: 08/29/2019] [Indexed: 01/26/2023]
Abstract
The design of multi-targeted drugs has gained considerable interest in the last decade thanks to their advantages in the treatment of different diseases, including cancer. The simultaneous inhibition of selected targets from cancerous cells to induce their death represents an attractive objective for the medicinal chemist in order to enhance the efficiency of chemotherapy. In the present work, several alkynyl gold(I) phosphane complexes derived from 3-hydroxyflavones active against three human cancer cell lines, colorectal adenocarcinoma Caco-2/TC7, breast adenocarcinoma MCF-7 and hepatocellular carcinoma HepG2, have been synthesized and characterized. Moreover, these compounds display high selective index values towards differentiated Caco-2 cells, which are considered as a model of non-cancerous cells. The antiproliferative effect of the most active complexes [Au(L2b)PPh3] (3b) and [Au(L2c)PTA] (4c) on Caco-2 cells, seems to be mediated by the inhibition of the enzyme cyclooxygenase-1/2 and alteration of the activities of the redox enzymes thioredoxin reductase and glutathione reductase. Both complexes triggered cell death by apoptosis, alterations in cell cycle progression and increased of ROS production. These results provide support for the suggestion that multi-targeting approach involving the interaction with cyclooxygenase-1/2 and the redox enzymes that increases ROS production, enhances cell death in vitro. All these results indicate that complexes [Au(L2b)PPh3] and [Au(L2c)PTA] are promising antiproliferative agents for further anticancer drug development.
Collapse
Affiliation(s)
- Inés Mármol
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., 50009, Zaragoza, Spain; Departamento de Farmacología y Fisiología, Unidad de Fisiología, Universidad de Zaragoza, CIBERobn, IIS Aragón, IA2, 50013, Zaragoza, Spain
| | - Pilar Castellnou
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., 50009, Zaragoza, Spain
| | - Raquel Alvarez
- Departamento de Farmacología y Fisiología, Unidad de Fisiología, Universidad de Zaragoza, CIBERobn, IIS Aragón, IA2, 50013, Zaragoza, Spain
| | - M Concepción Gimeno
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., 50009, Zaragoza, Spain
| | - M Jesús Rodríguez-Yoldi
- Departamento de Farmacología y Fisiología, Unidad de Fisiología, Universidad de Zaragoza, CIBERobn, IIS Aragón, IA2, 50013, Zaragoza, Spain.
| | - Elena Cerrada
- Departamento de Química Inorgánica, Instituto de Síntesis Química y Catálisis Homogénea-ISQCH, Universidad de Zaragoza-C.S.I.C., 50009, Zaragoza, Spain.
| |
Collapse
|
12
|
Emerging Perspective: Role of Increased ROS and Redox Imbalance in Skin Carcinogenesis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8127362. [PMID: 31636809 PMCID: PMC6766104 DOI: 10.1155/2019/8127362] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/25/2019] [Accepted: 07/31/2019] [Indexed: 02/08/2023]
Abstract
Strategies to battle malignant tumors have always been a dynamic research endeavour. Although various vehicles (e.g., chemotherapeutic therapy, radiotherapy, surgical resection, etc.) are used for skin cancer management, they mostly remain unsatisfactory due to the complex mechanism of carcinogenesis. Increasing evidence indicates that redox imbalance and aberrant reactive oxygen species (ROS) are closely implicated in the oncogenesis of skin cancer. When ROS production goes beyond their clearance, excessive or accumulated ROS could disrupt redox balance, induce oxidative stress, and activate the altered ROS signals. These would damage cellular DNA, proteins, and lipids, further leading to gene mutation, cell hyperproliferation, and fatal lesions in cells that contribute to carcinogenesis in the skin. It has been known that ROS-mediated skin carcinogenesis involves multiple ways, including modulating related signaling pathways, changing cell metabolism, and causing the instability of the genome and epigenome. Nevertheless, the exact role of ROS in skin cancer has not been thoroughly elucidated. In spite of ROS inducing skin carcinogenesis, toxic-dose ROS could trigger cell death/apoptosis and, therefore, may be an efficient therapeutic tool to battle skin cancer. Considering the dual role of ROS in the carcinogenesis and treatment of skin cancer, it would be essential to clarify the relationship between ROS and skin cancer. Thus, in this review, we get the related data together to seek the connection between ROS and skin carcinogenesis. Besides, strategies basing on ROS to fight skin cancer are discussed.
Collapse
|
13
|
Buzharevski A, Paskas S, Sárosi MB, Laube M, Lönnecke P, Neumann W, Mijatovic S, Maksimovic-Ivanic D, Pietzsch J, Hey-Hawkins E. Carboranyl Analogues of Celecoxib with Potent Cytostatic Activity against Human Melanoma and Colon Cancer Cell Lines. ChemMedChem 2019; 14:315-321. [PMID: 30602073 DOI: 10.1002/cmdc.201800685] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/14/2018] [Indexed: 12/13/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most common way of treating inflammatory disorders. Their widespread use helped reveal their other modes of action as pharmaceuticals, such as a profound effect on various cancers. Celecoxib has proven to be a very prominent member of this group with cytostatic activities. On the other hand, the highly dynamic field of drug design is constantly searching for new ways of modifying known structures to obtain more powerful and less harmful drugs. A very interesting development is the implementation of carboranes in pharmacologically active structures, mostly as phenyl mimetics. Herein we report the synthesis of three carborane-containing derivatives of the COX-2-selective NSAID celecoxib. The new compounds proved to have promising cytostatic potential against various melanoma and colorectal adenocarcinoma cell lines. Inhibited proliferation accompanied by caspase-independent apoptotic cell death was found to be the main cause of decreased cell viability upon treatment with the most efficient celecoxib analogue, 3 b (4-[5-(1,7-dicarba-closo-dodecaboranyl)-3-trifluoromethyl-1H-pyrazol-1-yl]-1-methylsulfonylbenzene).
Collapse
Affiliation(s)
- Antonio Buzharevski
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Svetlana Paskas
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", Belgrade University, Belgrade, Serbia
| | - Menyhárt-Botond Sárosi
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Markus Laube
- Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Peter Lönnecke
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Wilma Neumann
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| | - Sanja Mijatovic
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", Belgrade University, Belgrade, Serbia
| | - Danijela Maksimovic-Ivanic
- Department of Immunology, Institute for Biological Research "Sinisa Stankovic", Belgrade University, Belgrade, Serbia
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstrasse 4, 01062, Dresden, Germany
| | - Evamarie Hey-Hawkins
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103, Leipzig, Germany
| |
Collapse
|
14
|
Ni H, Hu S, Chen X, Liu Y, Ni T, Cheng L. Tra2β silencing suppresses cell proliferation in laryngeal squamous cell carcinoma via inhibiting PI3K/AKT signaling. Laryngoscope 2018; 129:E318-E328. [PMID: 30597574 DOI: 10.1002/lary.27716] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Hao‐Sheng Ni
- Department of OtorhinolaryngologyFirst Affiliated Hospital of Nanjing Medical University Nanjing China
- Department of OtorhinolaryngologyAffiliated Hospital of Nantong University Nantong China
| | - Song‐Qun Hu
- Department of OtorhinolaryngologyFirst Affiliated Hospital of Nanjing Medical University Nanjing China
- Department of OtorhinolaryngologyAffiliated Hospital of Nantong University Nantong China
| | - Xi Chen
- Department of OtorhinolaryngologyFirst Affiliated Hospital of Nanjing Medical University Nanjing China
| | - Yi‐Fei Liu
- Department of PathologyAffiliated Hospital of Nantong University Nantong China
| | - Ting‐Ting Ni
- Department of OncologyNantong Tumor Hospital Nantong China
| | - Lei Cheng
- Department of OtorhinolaryngologyFirst Affiliated Hospital of Nanjing Medical University Nanjing China
| |
Collapse
|
15
|
Reactive Oxygen Species-Mediated Tumor Microenvironment Transformation: The Mechanism of Radioresistant Gastric Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5801209. [PMID: 29770167 PMCID: PMC5892229 DOI: 10.1155/2018/5801209] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 01/30/2018] [Accepted: 02/26/2018] [Indexed: 01/01/2023]
Abstract
Radioresistance is one of the primary causes responsible for therapeutic failure and recurrence of cancer. It is well documented that reactive oxygen species (ROS) contribute to the initiation and development of gastric cancer (GC), and the levels of ROS are significantly increased in patients with GC accompanied with abnormal expressions of multiple inflammatory factors. It is also well documented that ROS can activate cancer cells and inflammatory cells, stimulating the release of a variety of inflammatory cytokines, which subsequently mediates the tumor microenvironment (TME) and promotes cancer stem cell (CSC) maintenance as well as renewal and epithelial-mesenchymal transition (EMT), ultimately resulting in radioresistance and recurrence of GC.
Collapse
|
16
|
Gomes BRB, Firmino M, Jorge JS, Ferreira MLO, Rodovalho TM, Weis SN, Souza GEP, Morais PC, Sousa MV, Souza PEN, Veiga-Souza FH. Increase of reactive oxygen species in different tissues during lipopolysaccharide-induced fever and antipyresis: an electron paramagnetic resonance study. Free Radic Res 2018; 52:351-361. [PMID: 29308684 DOI: 10.1080/10715762.2018.1425549] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Fever is a regulated increase in body temperature and a component of the acute-phase response, triggered mainly after the invasion of pathogens in the body. Reactive oxygen species (ROS) are generated during the physiological and pathological processes, and can act as both signalling molecules as well as promoters of oxidative stress. Male Wistar rats, pretreated with oral doses of acetaminophen, celecoxib, dipyrone, or ibuprofen 30 min before an intravenous lipopolysaccharide (LPS) or sterile saline injection, showed a reduced febrile response in all animals tested. The formation of ROS in the fresh blood, liver, brown adipose tissue (BAT), and hypothalamus of febrile and antipyretic-treated animals was assessed by electron paramagnetic resonance using the spin probe 1-hydroxy-3-methoxycarbonyl-2,2,5,5-tetramethylpyrrolidine (CMH). While the CM• concentrations remained unaltered in the blood samples examined 5 h after the induction of fever, we found increased CM• levels in the liver (in µM, saline: 290 ± 42; LPS: 512 ± 34), BAT (in µM, saline: 509 ± 79, LPS: 855 ± 79), and hypothalamus (in µM, saline: 292 ± 35; LPS: 467 ± 8) at the same time point. Importantly, none of the antipyretics were seen to alter the CM• accumulation profile. Data from this study suggest that there is an increased formation of ROS in the different tissues during fever, which may cause oxidative stress, and that the antipyretics tested do not interfere with ROS production.
Collapse
Affiliation(s)
- Bruna R B Gomes
- a Department of Cell Biology, Laboratory of Protein Chemistry and Biochemistry , Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Marina Firmino
- a Department of Cell Biology, Laboratory of Protein Chemistry and Biochemistry , Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Jardeson S Jorge
- b School of Ceilandia , University of Brasilia , Brasília , Brazil
| | | | | | - Simone N Weis
- a Department of Cell Biology, Laboratory of Protein Chemistry and Biochemistry , Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Gloria E P Souza
- c Laboratory of Pharmacology, School of Pharmaceutical Sciences of Ribeirão Preto , University of São Paulo , Ribeirão Preto , Brazil
| | - Paulo C Morais
- d School of Chemistry and Chemical Engineering , Anhui University , Hefei , China.,e Laboratory of Electron Paramagnetic Resonance , Institute of Physics, University of Brasilia , Brasília , Brazil
| | - Marcelo V Sousa
- a Department of Cell Biology, Laboratory of Protein Chemistry and Biochemistry , Institute of Biology, University of Brasilia , Brasília , Brazil
| | - Paulo E N Souza
- e Laboratory of Electron Paramagnetic Resonance , Institute of Physics, University of Brasilia , Brasília , Brazil
| | - Fabiane H Veiga-Souza
- a Department of Cell Biology, Laboratory of Protein Chemistry and Biochemistry , Institute of Biology, University of Brasilia , Brasília , Brazil.,b School of Ceilandia , University of Brasilia , Brasília , Brazil
| |
Collapse
|