1
|
Priyanka, Rani P, Ranolia A, Kiran, Joshi G, Singh S, Kumar R, Wahajuddin M, Kumar P, Singh D, Sindhu J. Design and synthesis of new 1,4-naphthoquinones appended sulfenylated thiazoles as cyclooxygenase II inhibitors: Exploring the utility in the development of anticancer agents. Bioorg Chem 2025; 161:108537. [PMID: 40334422 DOI: 10.1016/j.bioorg.2025.108537] [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: 01/22/2025] [Revised: 04/05/2025] [Accepted: 04/25/2025] [Indexed: 05/09/2025]
Abstract
Clinical evaluations revealed the direct role of chronic inflammation in cancer progression. The cyclooxygenase (COX) pathway is particularly important among the various pathways involved in inflammation. There are two COX isoforms: COX-1 and COX-2. The importance of drug discovery lies in selectively inhibiting COX-2, an enzyme expressed during inflammation, unlike COX-1, which is constitutively active. The inhibition of COX-1 is correlated further with gastric ulcers. However, recently approved COX-2 inhibitors have intricate cardiotoxicity, thus creating an utmost need for new COX-2 inhibitors. Considering this, in our present research, we rationally designed and synthesized a series (4a-4q) of 1,4-naphthoquinones appended sulfenylated thiazoles using molecular hybridization approach under metal-free conditions. These synthetics were explored for their anticancer potential against three cell lines, and the hits portraying anticancer effects were further tested against COX isoforms. Among all, compounds 4d and 4f were found to be potent anticancer leads, exhibiting selective inhibition of COX-2 and, concomitantly, the lipoxygenase (LOX) pathway, both of which share the same substrate, arachidonic acid. Both compounds were found to reduce oxidative stress and induce cancer cell death via apoptosis pathway. The experimental outcome was further corroborated using in silico techniques, including density field theory (DFT), Molecular docking, and dynamics.
Collapse
Affiliation(s)
- Priyanka
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Payal Rani
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Anju Ranolia
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Kiran
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India
| | - Gaurav Joshi
- Department of Pharmaceutical Sciences, Chauras Campus, HNB Garhwal University (A Central University), Srinagar, Uttarakhand 246174, India; Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, West Yorkshire BD7 1BD, United Kingdom.
| | - Snigdha Singh
- Department of Chemistry, University of Delhi, 110007, India
| | - Roshan Kumar
- Department of Microbiology, Central University of Punjab, VPO-Ghudda, Punjab 151401, India; Department of Microbiology, Graphic Era (Deemed to be University) Clement Town, Dehradun 248002, India
| | - Muhammad Wahajuddin
- Institute of Cancer Therapeutics, School of Pharmacy and Medical Sciences, University of Bradford, Bradford, West Yorkshire BD7 1BD, United Kingdom
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra 136119, India
| | - Devender Singh
- Department of Chemistry, Maharshi Dayanand University, Rohtak 124001, India
| | - Jayant Sindhu
- Department of Chemistry, COBS&H, CCS Haryana Agricultural University, Hisar 125004, India.
| |
Collapse
|
2
|
Singh SB, Kuniyal K, Rawat A, Bisht A, Shah V, Daverey A. Sophorolipids as anticancer agents: progress and challenges. Discov Oncol 2025; 16:507. [PMID: 40208440 PMCID: PMC11985733 DOI: 10.1007/s12672-025-02303-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
Sophorolipids (SLs) are considered effective biosurfactant for cancer treatment, which can efficiently inhibit the viability of various cancer types including breast, lung, liver, cervical and colon cancers. Their mechanism of action targets apoptosis and operates at the level of caspase enzymes, upregulation and downregulation of the B-cell lymphoma (Bcl)-family proteins, and changes in mitochondrial membrane permeability. The binding of SLs to the cancer cell receptors modulates the expression of Bax, APAF1, Bcl-2 and Bcl-x, and triggers the release of cytochrome c into the cytosol which further activates caspase-3/9 pathways leading to apoptosis. SLs also increase intracellular reactive oxygen species (ROS) level in cancer cells that activates pro-apoptotic JNK and p38 MAPK signaling pathways and induce apoptosis through the activation of caspase (3, 6 and 7) pathways. Recently, the integration of anticancer drugs like doxorubicin hydrochloride into SL based nanoparticles (SLNPs) enhanced stability, biocompatibility, bioavailability, pharmacokinetics and therapeutic efficacy. Besides, doxorubicin and resveratrol conjugated NPs induced apoptosis in resistant breast cancer cells by down-regulating the expression of Bcl-2, NF-kB and efflux transporters. However, several challenges exist regarding the stability of SLs under physiological conditions, targeting specific cancer cells, and their clinical applications. This study provides updated concepts on the formulations and properties of different types of SLs, their mechanism of anticancer action and applications in nanotechnology for targeted drug delivery system.
Collapse
Affiliation(s)
- Salam Bhopen Singh
- School of Biological Sciences, Doon University, Dehradun, 248012, Uttarakhand, India
| | - Kanupriya Kuniyal
- School of Environment and Natural Resources, Doon University, Dehradun, 248012, Uttarakhand, India
| | - Ananya Rawat
- School of Biological Sciences, Doon University, Dehradun, 248012, Uttarakhand, India
| | - Ananya Bisht
- School of Biological Sciences, Doon University, Dehradun, 248012, Uttarakhand, India
| | - Vijendra Shah
- School of Biological Sciences, Doon University, Dehradun, 248012, Uttarakhand, India
| | - Achlesh Daverey
- School of Biological Sciences, Doon University, Dehradun, 248012, Uttarakhand, India.
- School of Environment and Natural Resources, Doon University, Dehradun, 248012, Uttarakhand, India.
| |
Collapse
|
3
|
Sheehan BJ, Edwards B, Medrano IS, El-Saidi MA, Zaidan WR, El-Ezzi AA, Kuddus RH. Association between two single nucleotide polymorphisms of the Prostaglandin-Endoperoxide Synthase 1 and 2 genes and cell proliferative prostatic diseases in Lebanon. Oncotarget 2025; 16:262-272. [PMID: 40184332 PMCID: PMC11970937 DOI: 10.18632/oncotarget.28710] [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: 11/13/2024] [Accepted: 03/13/2025] [Indexed: 04/06/2025] Open
Abstract
The polymorphic genes PTGS1 and PTGS2 encode cyclooxygenases COX-1 and COX-2, respectively. Overexpression of these cyclooxygenases is linked to inflammation and neoplasms. This study investigated the potential association between the single nucleotide polymorphism (SNP) -842A>G (rs10306114) of the PTGS1 gene and SNP-765G>C (rs20417) of the PTGS2 gene with prostate cancer (PCa) and benign prostate hyperplasia (BPH). Blood leucocyte DNA from 56 healthy individuals, 61 individuals with PCa, and 51 individuals with BPH were genotyped using the PCR-RFLP method. Associations were inferred by calculating odds ratios (OR) and relative risks (RR) of genotype distributions and allele frequencies. The genotypes for both SNPs were in Hardy-Weinberg equilibrium for all groups. No significant association was observed between the A or G alleles or the AA, AG, or GG genotypes of the SNP-842A>G of the PTGS1 gene and prostatic diseases. However, the C allele of SNP-765G>C of the PTGS2 gene was significantly associated with an increased risk of BPH (OR = 2.30, p-value = 0.01). Differences in the ratios of GG/GC and GG/(GC+CC) genotypes also suggested a potential association between the C allele and PCa (p-value <0.1), and the combined affected (PCa+BPH) group (p-value <0.04). The small sample size and sampling from one ethnic group are limitations of this study.
Collapse
Affiliation(s)
- Brock J. Sheehan
- Department of Biology, Utah Valley University, Orem, UT 84058, USA
| | - Bryson Edwards
- Department of Biology, Utah Valley University, Orem, UT 84058, USA
| | | | - Mohammed A. El-Saidi
- Department of Strategic Management and Operations, Utah Valley University, Orem, UT 84058, USA
| | - Wissam R. Zaidan
- Radioimmunoassay Laboratory, Lebanese Atomic Energy Commission, Beirut, Lebanon
| | - Asmahan A. El-Ezzi
- Radioimmunoassay Laboratory, Lebanese Atomic Energy Commission, Beirut, Lebanon
- Department of Chemistry and Biochemistry, Lebanese University, Hadath, Lebanon
| | - Ruhul H. Kuddus
- Department of Biology, Utah Valley University, Orem, UT 84058, USA
| |
Collapse
|
4
|
Gajula SNR, Munjal V, Talari S, Rahman Z, Dandekar MP, Sonti R. The Influence of a Rat Model of Depression and Gastric Ulcer on the Pharmacokinetics of Encorafenib. Biopharm Drug Dispos 2025; 46:67-81. [PMID: 40202822 DOI: 10.1002/bdd.70006] [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: 03/09/2025] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
Encorafenib is used to treat melanoma and colorectal tumors. Depression and gastric ulcer conditions can impact various physiological processes, including drug metabolism and pharmacokinetics. This study investigated the effect of a rat model of depression and gastric ulcers on the pharmacokinetics (PK) of encorafenib using the developed LC-QqQ-MS method. The chronic unpredictable mild stress (CUMS) model of depression and the indomethacin-induced gastric ulcer model were utilized to investigate the effect of depression and gastric ulcers on the pharmacokinetics of orally administered encorafenib. The focus was on parameters such as maximum plasma concentration (Cmax), elimination half-life (t1/2), mean residence time (MRT), volume of distribution (Vd), area under the curve (AUC), and apparent clearance (CL/F). Rats with depression exhibited a significant increase in maximum plasma concentration (Cmax). In contrast, depression led to decreased t1/2 and MRT, implying alterations in the drug's absorption and clearance. On the one hand, rats with gastric ulcers displayed a significant decrease in Cmax, coupled with an extended time to reach maximum plasma concentration (Tmax), prolonged t1/2, MRT, and increased volume of distribution (Vd) of encorafenib. This preclinical study demonstrates that depression and gastric ulcers significantly impact the pharmacokinetics of encorafenib. These findings emphasize the importance of considering these disease conditions when designing encorafenib dosing regimens for optimal therapeutic outcomes in cancer patients.
Collapse
Affiliation(s)
- Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
- Department of Pharmaceutical Analysis, GITAM School of Pharmacy, GITAM (Deemed to be University), Visakhapatnam, India
| | - Vijay Munjal
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Sasikala Talari
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ziaur Rahman
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Manoj P Dandekar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Rajesh Sonti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
5
|
Carpio AR, Talubo ND, Tsai PW, Chen BY, Tayo LL. Berries as Nature's Therapeutics: Exploring the Potential of Vaccinium Metabolites in Gastric Cancer Treatment Through Computational Insights. Life (Basel) 2025; 15:406. [PMID: 40141751 PMCID: PMC11944152 DOI: 10.3390/life15030406] [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: 01/20/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Berries from the Vaccinium genus, known for their rich array of bioactive metabolites, are recognized for their antioxidant, anti-inflammatory, and anticancer properties. These compounds, including anthocyanins, flavonoids, and phenolic acids, have attracted significant attention for their potential health benefits, particularly in cancer prevention and treatment. Gastric cancer (GC), a leading cause of cancer-related deaths worldwide, remains challenging to treat, especially in its advanced stages. This study investigates the therapeutic potential of Vaccinium species in GC treatment using computational methods. RNA sequencing revealed upregulated genes associated with GC, while network pharmacology and molecular docking approaches identified strong interactions between cyanidin 3-O-glucoside (C3G), a key bioactive metabolite. Furthermore, molecular dynamics simulations of the HSP90AA1-C3G complex demonstrated stable binding and structural integrity, suggesting that C3G may inhibit HSP90AA1, a protein involved in cancer progression. These findings highlight the therapeutic potential of Vaccinium metabolites, offering a novel approach to GC treatment by targeting key molecular pathways. This research provides valuable insights into the role of berries as natural therapeutics, supporting their integration into future gastric cancer treatment strategies.
Collapse
Affiliation(s)
- Angelica Rachel Carpio
- School of Chemical, Biological, and Materials Engineering and Sciences, School of Graduate Studies, Mapúa University, Manila 1002, Philippines; (A.R.C.); (N.D.T.)
| | - Nicholas Dale Talubo
- School of Chemical, Biological, and Materials Engineering and Sciences, School of Graduate Studies, Mapúa University, Manila 1002, Philippines; (A.R.C.); (N.D.T.)
| | - Po-Wei Tsai
- Department of Food Science, National Taiwan Ocean University, Keelung 202, Taiwan;
| | - Bor-Yann Chen
- Department of Chemical and Materials Engineering, National I-Lan University, I-Lan 260, Taiwan
| | - Lemmuel L. Tayo
- Department of Biology, School of Health Sciences, Mapúa University, Makati 1200, Philippines
| |
Collapse
|
6
|
Mohanad M, Hamza HM, Bahnassy AA, Shaarawy S, Ahmed O, El-Mezayen HA, Ayad EG, Tahoun N, Abdellateif MS. Molecular profiling of breast cancer methylation pattern in triple negative versus non- triple negative breast cancer. Sci Rep 2025; 15:6894. [PMID: 40011499 PMCID: PMC11865568 DOI: 10.1038/s41598-025-90150-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
Epigenetic alterations, especially promotor methylation, have a significant impact on gene expression, molecular subtyping, prognosis, and outcome of breast cancer (BC). The methylation profile was assessed for 22 genes of the BC tissue using the EpiTect Methyl II PCR System in 40 triple-negative BC (TNBC) patients compared to 50 non-TNBC group. The data were corelated with the disease-free (DFS) and overall survival (OS) of the patients. Genes that were differentially hypermethylated in TNBC patients compared to those with non-TNBC included CCND2, CDKN2A, ESR1, CDH1, BRCA1, GSTP, RASSF1, SLIT2, MGMT, PTEN, TP73, and PRDM2. These panel achieved 95% sensitivity, 98% specificity, 97.44% positive predictive value (PPV), 94.23% negative predictive value (NPV), and AUC of 0.993. Hypermethylation of BRCA1, CDH1, CDKN2A, ESR1, GSTP, HIC1, MGMT, PRDM2, PTEN, PYCARDM, RASSF1M, THBS1, and TP73 associated significantly with worse OS and DFS in TNBC cohort. Meanwhile, CCNA1 and CDH1 hypermethylation demonstrated significant associations with poor DFS but did not show significant relationships with OS in TNBC patients. PTGS2 and TNFRSF10C methylation were associated with better DFS and OS rates in TNBC patients. On multivariate Cox regression, CCND2 and PTEN hypermethylation were independent predictors of DFS in the overall BC patients. The hypermethylation of BRCA1 and GSTP were independent predictors of DFS, while PTEN hypermethylation was an independent predictor of OS in the TNBC cohort. The identification of hypermethylated genes, such as BRCA1, CCND2, CDH1, ESR1, GSTP, RASSF1, SLIT2, MGMT, and PTEN may serve as potential biomarkers or therapeutic targets for TNBC.
Collapse
Affiliation(s)
- Marwa Mohanad
- Biochemistry Department, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology, 6th October, Egypt
| | - Hager M Hamza
- Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Abeer A Bahnassy
- Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Sabry Shaarawy
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Ola Ahmed
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Hatem A El-Mezayen
- Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Eman G Ayad
- Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Neveen Tahoun
- Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
7
|
Wahnou H, Limami Y, Duval RE, Ismail B, Léger DY, Sol V, Liagre B. Photodynamic anti-cancer therapy and arachidonic acid metabolism: State of the art in 2024. ANNALES PHARMACEUTIQUES FRANÇAISES 2025:S0003-4509(25)00042-2. [PMID: 40020873 DOI: 10.1016/j.pharma.2025.02.010] [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: 12/22/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Photodynamic therapy (PDT) has emerged as a promising and evolving modality in cancer treatment leveraging light-sensitive compounds known as photosensitizers to selectively induce cell death in malignant tissues through the generation of reactive oxygen species (ROS). This review delves into the intricate mechanisms of PDT highlighting the pivotal role of photosensitizers and the resultant oxidative stress that damages cancer cells. It explores the versatile applications of PDT across various cancer types alongside the advantages and limitations inherent to this therapy. Recent technological advancements including improved photosensitizers and novel light delivery systems are also discussed. Additionally the review examines the critical role of arachidonic acid (AA) metabolism in cancer progression detailing the cyclooxygenase, lipoxygenase and cytochrome P450 pathways and their contributions to tumor biology. By elucidating the interplay between PDT and AA metabolism the review underscores the potential of targeting AA metabolic pathways to enhance PDT efficacy. Finally it provides clinical and translational perspectives highlighting ongoing research and future directions aimed at optimizing PDT for improved cancer treatment outcomes.
Collapse
Affiliation(s)
- Hicham Wahnou
- Laboratory of Immunology and Biodiversity, Faculty of Sciences Ain Chock, Hassan II University, BP2693 Maarif, Casablanca, Morocco.
| | - Youness Limami
- Institute of Health Sciences, Hassan First University, Settat, Morocco.
| | | | - Bassel Ismail
- College of Health and Medical Technology, Medical Laboratories Technology Department, Alayen Iraqi University, Thi-Qar 64001, Iraq.
| | - David Yannick Léger
- Université de Limoges, LABCiS UR 22722, faculté de Pharmacie, 87000 Limoges, France.
| | - Vincent Sol
- Université de Limoges, LABCiS UR 22722, faculté de Pharmacie, 87000 Limoges, France.
| | - Bertrand Liagre
- Université de Limoges, LABCiS UR 22722, faculté de Pharmacie, 87000 Limoges, France.
| |
Collapse
|
8
|
Zhang G, Xu Y, Zhou A, Yu Y, Ning X, Bao H. Bioengineered NanoAid synergistically targets inflammatory pro-tumor processes to advance glioblastoma chemotherapy. NANOSCALE 2025; 17:2753-2768. [PMID: 39831463 DOI: 10.1039/d4nr04557b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Through transcriptomic analysis of patient-derived glioblastoma tissues, we identify an overactivation of inflammatory pathways that contribute to the development of a tumor-promoting microenvironment and therapeutic resistance. To address this critical mechanism, we present NanoAid, a biomimetic nanoplatform designed to target inflammatory pro-tumor processes to advance glioblastoma chemotherapy. NanoAid employs macrophage-membrane-liposome hybrids to optimize the delivery of COX-2 inhibitor parecoxib and paclitaxel. By inheriting macrophage characteristics, NanoAid not only efficiently traverses the blood-brain barrier and precisely accumulates within tumors but also enhances cancer cell uptake, thereby improving overall anticancer efficacy. Notably, the combination of parecoxib and paclitaxel effectively disrupts inflammatory pro-tumor processes while inducing a synergistic effect that inhibits tumor growth, overcomes therapeutic resistance, and minimizes adverse effects. This results in substantial tumor growth inhibition and extends the median survival of tumor-bearing mice. Thus, our study bridges clinical insights with fundamental research, potentially revolutionizing tumor therapy paradigms.
Collapse
Affiliation(s)
- Gui Zhang
- The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Yongle Yu
- Medical College of Guangxi University, Nanning 530004, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, China.
| | - Hongguang Bao
- Department of Anaesthesiology, Perioperative and Pain Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 211101, China.
| |
Collapse
|
9
|
Gaiaschi L, Casali C, Stabile A, D'Amico S, Ravera M, Gabano E, Galluzzo A, Favaron C, Gola F, De Luca F, Pellegatta S, Bottone MG. DNA Damage Repair in Glioblastoma: A Novel Approach to Combat Drug Resistance. Cell Prolif 2025:e13815. [PMID: 39866010 DOI: 10.1111/cpr.13815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/18/2024] [Accepted: 01/15/2025] [Indexed: 01/28/2025] Open
Abstract
Due to the lack of effective therapeutic approach, glioblastoma (GBM) remains one of the most malignant brain tumour. By in vitro investigations on primary GBM stem cells, we highlighted one of the underlying mechanisms of drug resistance to alkylating agents, the DNA damage responses. Here, flow cytometric analysis and viability and repopulation assays were used to assess the long-term cytotoxic effect induced by the administration of a fourth-generation platinum prodrug, the (OC-6-44)-acetatodiamminedichlorido(2-(2-propynyl)octanoato) platinum(IV) named Pt(IV)Ac-POA, in comparison to the most widely used Cisplatin. The immunofluorescence studies revealed changing pathways involved in the DNA damage response mechanisms in response to the two chemotherapies, suggesting in particular the role of Poly (ADP-Ribose) polymerases in the onset of resistance to Cisplatin-induced cytotoxicity. Thus, this research provides a proof of concept for how the use of a prodrug which allows the co-administration of Cisplatin and an Histone DeACetylase inhibitors, could suppress DNA repair mechanisms, suggesting a novel effective approach in GBM treatment.
Collapse
Affiliation(s)
- Ludovica Gaiaschi
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Claudio Casali
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Andrea Stabile
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Sharon D'Amico
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Mauro Ravera
- Department of Sciences and Technological Innovation (DiSIT), University of Piemonte Orientale "A. Avogadro", Alessandria, Italy
| | - Elisabetta Gabano
- Department of Sustainable Development and Ecological Transition, University of Piemonte Orientale, Vercelli, Italy
| | - Andrea Galluzzo
- Unit of Immunotherapy of Brain Tumors, Fondazione IRCCS Istituto Neurologico "C. Besta", Milan, Italy
| | - Cristina Favaron
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Federica Gola
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Fabrizio De Luca
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Serena Pellegatta
- Unit of Immunotherapy of Brain Tumors, Fondazione IRCCS Istituto Neurologico "C. Besta", Milan, Italy
| | | |
Collapse
|
10
|
Kassab AE, Gedawy EM. Repurposing of Indomethacin and Naproxen as anticancer agents: progress from 2017 to present. RSC Adv 2024; 14:40031-40057. [PMID: 39717807 PMCID: PMC11664213 DOI: 10.1039/d4ra07581a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
Inflammation is strongly linked to cancer and is essential for the growth and development of tumors. Targeting inflammation and the mediators involved in the inflammatory process could therefore provide a suitable method for cancer prevention and therapy. Numerous studies have shown that inflammation can predispose tumors. Non-steroidal anti-inflammatory drugs (NSAIDs) can affect the tumor microenvironment through increasing apoptosis and chemo-sensitivity while decreasing cell migration. Since the development of novel drugs requires a significant amount of money and time and poses a significant challenge for drug discovery, there has been a recent increase in interest in drug repositioning or repurposing. The growing body of research suggests that drug repurposing is essential for the quicker and less expensive development of anticancer therapies. In order to set the course for potential future repositioning of NSAIDs for clinical deployment in the treatment of cancer, the antiproliferative activity of derivatives of Indomethacin and Naproxen as well as their mechanism of action and structural activity relationships (SARs) published in the time frame from 2017 to 2024 are summarized in this review.
Collapse
Affiliation(s)
- Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University Kasr El-Aini Street, P. O. Box 11562 Cairo Egypt +2023635140 +2023639307
| | - Ehab M Gedawy
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University Kasr El-Aini Street, P. O. Box 11562 Cairo Egypt +2023635140 +2023639307
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC) Badr City, P. O. Box 11829 Cairo Egypt
| |
Collapse
|
11
|
Hamdi A, Tawfik SS, Ali AR, Ewes WA, Haikal A, El-Azab AS, Bakheit AH, Hefnawy MM, Ghabbour HA, Abdel-Aziz AAM. Harnessing potential COX-2 engagement for boosting anticancer activity of substituted 2-mercapto-4(3H)-quinazolinones with promising EGFR/VEGFR-2 inhibitory activities. Bioorg Chem 2024; 153:107951. [PMID: 39541892 DOI: 10.1016/j.bioorg.2024.107951] [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: 09/09/2024] [Revised: 10/25/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024]
Abstract
We designed and synthesized new quinazolinone-tethered phenyl thiourea/thiadiazole derivatives 4-26. Based on their structural characteristics, these compounds were proposed to have a multi-target mode of action for their anticancer activities. Using the MTT assay method, antiproliferative effects were assessed against three human cancer cell lines (HEPG-2, MCF-7, and HCT-116). In vitro assessment for enzymatic inhibitory activity of the most active compounds 4, 9 and 20 was done for EGFR, VEGFR-2 and COX-2 as potential targets. The screened compounds showed low micromolar IC50 inhibitory effects against the three targets. Compound 9 demonstrated similar EGFR/VEGFR-2 inhibitory effect to the control drugs and potential inhibitory activity for COX-2 enzyme. In MCF-7 cells, the most active analog 9 caused 41.02% total apoptosis, and arrested the cell cycle at the G2/M phase. Taken as a whole, the findings of this study provide significant new understandings into the relationship between COX inhibition and cancer therapy. Furthermore, the outcomes showcased the encouraging efficacy of these compounds with a multi-target mechanism, making them excellent choices for additional research and development into possible anticancer drug.
Collapse
Affiliation(s)
- Abdelrahman Hamdi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Samar S Tawfik
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed R Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Wafaa A Ewes
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Abdullah Haikal
- Department of Pharmacognosy, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Adel S El-Azab
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P. O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ahmed H Bakheit
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P. O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohamed M Hefnawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P. O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Hazem A Ghabbour
- School of Health and Biomedical Sciences, RMIT University, Melbourne 3083, Australia
| | - Alaa A-M Abdel-Aziz
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P. O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
12
|
Singh MT, Thaggikuppe Krishnamurthy P, Magham SV. Harnessing the synergistic potential of NK1R antagonists and selective COX-2 inhibitors for simultaneous targeting of TNBC cells and cancer stem cells. J Drug Target 2024; 32:258-269. [PMID: 38252517 DOI: 10.1080/1061186x.2024.2309568] [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: 09/26/2023] [Accepted: 01/11/2024] [Indexed: 01/24/2024]
Abstract
Triple-negative breast cancer (TNBC) lacks the expression of oestrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), rendering it unresponsive to endocrine therapy and HER2 targeted treatments. Though certain chemotherapeutics targeting the cell cycle have shown efficacy to a certain extent, the presence of chemotherapy-resistant cancer stem cells (CSCs) presents a significant challenge in tackling TNBC. Multiple lines of evidence suggest the upregulation of neuropeptide Substance P (SP), its NK-1 receptor (NK1R) and the Cyclooxygenase-2 (COX-2) enzyme in TNBC patients. Upregulation of the SP/NK1R system and COX-2 influences major signalling pathways involved in cell proliferation, growth, survival, angiogenesis, inflammation, metastasis and stem cell activity. The simultaneous activation and crosstalk between the pathways activated by SP/NK1R and COX-2 consequently increase the levels of key regulators of self-renewal pathways in CSCs, promoting stemness. The combination therapy with NK1R antagonists and COX-2 inhibitors can simultaneously target TNBC cells and CSCs, thereby enhancing treatment efficacy and reducing the risk of recurrence and relapse. This review discusses the rationale for combining NK1R antagonists and COX-2 inhibitors for the better management of TNBC and a novel strategy to deliver drug cargo precisely to the tumour site to address the challenges associated with off-target binding.
Collapse
Affiliation(s)
- Madhu Tanya Singh
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Sai Varshini Magham
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| |
Collapse
|
13
|
Li J, Yang D, Lin L, Yu L, Chen L, Lu K, Lan J, Zeng Y, Xu Y. Important functions and molecular mechanisms of aquaporins family on respiratory diseases: potential translational values. J Cancer 2024; 15:6073-6085. [PMID: 39440058 PMCID: PMC11493008 DOI: 10.7150/jca.98829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/25/2024] [Indexed: 10/25/2024] Open
Abstract
Aquaporins (AQPs) are a subgroup of small transmembrane transporters that are distributed in various types of tissues, including the lung, kidney, heart and central nervous system. It is evident that respiratory diseases represent a significant global health concern, with a considerable number of deaths occurring worldwide. Recent researches have demonstrated that AQPs play a pivotal role in respiratory diseases, including chronic obstructive pulmonary disease (COPD), asthma, acute respiratory distress syndrome (ARDS), and particularly non-small cell lung cancer (NSCLC). In the context of NSCLC, the overexpression of AQP1, AQP3, AQP4, and AQP5 has been demonstrated to facilitate tumor angiogenesis, as well as the proliferation, migration, and invasiveness of tumor cells. This review concisely explores the role of AQP family on respiratory diseases, to assess their clinical and translational significance for understanding molecular pathogenesis. However, the potential translation of AQPs biomarkers into clinical applications is promising and the understanding of the precise mechanisms influencing respiratory diseases is still ongoing. Addressing the challenges and outlining the future perspectives in AQPs development is essential for clinical progress in a concise manner.
Collapse
Affiliation(s)
- Jinshan Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Dongyong Yang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Lanlan Lin
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Liying Yu
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Luyang Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Kaiqiang Lu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Jieli Lan
- Clinical Research Unit, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Yiming Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
| | - Yuan Xu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Clinical Research Center of Interventional Respirology, Quanzhou, Fujian Province, 362000, China
- School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, 350000, China
| |
Collapse
|
14
|
Ahn SY, Kim KA, Lee S, Kim KH. Potential skin anti-aging effects of main phenolic compounds, tremulacin and tremuloidin from Salix chaenomeloides leaves on TNF-α-stimulated human dermal fibroblasts. Chem Biol Interact 2024; 402:111192. [PMID: 39127184 DOI: 10.1016/j.cbi.2024.111192] [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: 05/08/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
The genus Salix spp. has long been recognized as a healing herb for its use in treating fever, inflammation, and pain relief, as well as a food source for its nutritional value. In this study, we aimed to explore the potential bioactive natural products in the leaves of Salix chaenomeloides, commonly known as Korean pussy willow, for their protective effects against skin damage, including aging. Utilizing LC/MS-guided chemical analysis of the ethanol extract of S. chaenomeloides leaves, with a focus on major compounds, we successfully isolated two main phenolic compounds, tremulacin (1) and tremuloidin (2). Subsequently, we investigated the protective effects of tremulacin (1) and tremuloidin (2) in TNF-α-stimulated human dermal fibroblasts (HDFs). The results revealed that both tremulacin (1) and tremuloidin (2) inhibited TNF-α-stimulation-induced ROS, suppressed matrix metalloproteinase-1 (MMP-1) expression, and enhanced collagen secretion. This implies that both tremulacin (1) and tremuloidin (2) hold promise as preventive agents against photoaging-induced skin aging. Furthermore, we assessed the activity of mitogen-activated protein kinases (MAPKs), cyclooxygenase-2 (COX-2), and heme oxygenase 1 (HO-1) to elucidate the mechanism of photoaging inhibition by tremuloidin (2), which exhibited superior efficacy. We found that tremuloidin (2) inhibited ERK and p38 phosphorylation and notably suppressed COX-2 expression while significantly upregulating HO-1 expression. These findings suggest potent anti-inflammatory and antioxidant properties of tremuloidin (2), positioning it as a potential candidate for combating photoaging-induced skin aging.
Collapse
Affiliation(s)
- Si-Young Ahn
- Department of Life Science, College of Bio-Nano Technology, Gachon University, Seongnam, 13120, Republic of Korea
| | - Kyung Ah Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sullim Lee
- Department of Life Science, College of Bio-Nano Technology, Gachon University, Seongnam, 13120, Republic of Korea.
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
15
|
Lv Y, Wang C, Liu R, Wu S, Chen J, Zheng X, Jiang T, Chen L. NUP37 promotes the proliferation and invasion of glioma cells through DNMT1-mediated methylation. Cell Death Discov 2024; 10:373. [PMID: 39174498 PMCID: PMC11341718 DOI: 10.1038/s41420-024-02138-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024] Open
Abstract
Nuclear regulation has potential in cancer therapy, with the nuclear pore complex (NPC) serving as a critical channel between the nucleus and cytoplasm, playing a role in regulating various biological processes and cancer. DNA methylation, an epigenetic modification mediated by DNA methyltransferases (DNMTs), influences gene expression and cell differentiation, and is crucial for the development and progression of tumor cells. Gliomas are the most common primary brain tumors, with glioblastoma being particularly aggressive, characterized by invasiveness, migration capability, and resistance to conventional treatments, resulting in poor prognosis. Our study revealed that the expression level of NUP37 affects the proliferation and invasion of glioma cells, and that the overexpression of DNMT1 can alleviate the adverse effects caused by NUP37 depletion. These findings suggest that NUP37 promotes the proliferation and invasion of glioma cells through its interaction with DNMT1.
Collapse
Affiliation(s)
- Yongqiang Lv
- Department of Neurosurgery, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Chaolian Wang
- Department of Neurosurgery, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Ruoyu Liu
- Department of Neurosurgery, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Junjun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Tianwei Jiang
- Department of Neurosurgery, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
| |
Collapse
|
16
|
Tang B, Guo M, Zhai Y, Zhang K, Ni K, Zhang Y, Huang L. Human esophageal cancer stem-like cells escape the cytotoxicity of natural killer cells via down-regulation of ULBP-1. J Transl Med 2024; 22:737. [PMID: 39103915 PMCID: PMC11301968 DOI: 10.1186/s12967-024-05549-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Cancer stem-like cells (CSCs) play an important role in initiation and progression of aggressive cancers, including esophageal cancer. Natural killer (NK) cells are key effector lymphocytes of innate immunity that directly attack a wide variety of cancer cells. NK cell-based therapy may provide a new treatment option for targeting CSCs. In this study, we aimed to investigate the sensitivity of human esophageal CSCs to NK cell-mediated cytotoxicity. METHODS CSCs were enriched from human esophageal squamous cell carcinoma cell lines via sphere formation culture. Human NK cells were selectively expanded from the peripheral blood of healthy donors. qRT-PCR, flow cytometry and ELISA assays were performed to examine RNA expression and protein levels, respectively. CFSE-labeled target cells were co-cultured with human activated NK cells to detect the cytotoxicity of NK cells by flow cytometry. RESULTS We observed that esophageal CSCs were more resistant to NK cell-mediated cytotoxicity compared with adherent counterparts. Consistently, esophageal CSCs showed down-regulated expression of ULBP-1, a ligand for NK cells stimulatory receptor NKG2D. Knockdown of ULBP-1 resulted in significant inhibition of NK cell cytotoxicity against esophageal CSCs, whereas ULBP-1 overexpression led to the opposite effect. Finally, the pro-differentiation agent all-trans retinoic acid was found to enhance the sensitivity of esophageal CSCs to NK cell cytotoxicity. CONCLUSIONS This study reveals that esophageal CSCs are more resistant to NK cells through down-regulation of ULBP-1 and provides a promising approach to promote the activity of NK cells targeting esophageal CSCs.
Collapse
Affiliation(s)
- Bo Tang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengxing Guo
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yujia Zhai
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kai Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaiyuan Ni
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lan Huang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
17
|
Ma ZY, Ding XJ, Zhu ZZ, Chen Q, Wang DB, Qiao X, Xu JY. Pt(iv) derivatives of cisplatin and oxaliplatin bearing an EMT-related TMEM16A/COX-2-selective dual inhibitor against colorectal cancer cells HCT116. RSC Med Chem 2024:d4md00327f. [PMID: 39185449 PMCID: PMC11342162 DOI: 10.1039/d4md00327f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/30/2024] [Indexed: 08/27/2024] Open
Abstract
Colorectal cancer represents the over-expression of TMEM16A and COX-2, offering a promising therapeutic strategy. Two Pt(iv) conjugates derived from Pt(ii) drug (cisplatin or oxaliplatin) and niflumic acid, complexes 1 and 2, were designed and prepared to exert the positive impact of multiple biological targets of DNA/TMEM16A/COX-2 against colorectal cancer. Complex 2 afforded higher cytotoxicity than 1 and the combination of an intermediate of oxidized oxaliplatin and NFA against cancer cells A549, HeLa, MCF-7, and HCT116. Especially for colorectal cancer cells HCT116, 2 was significantly more toxic (22-fold) and selective to cancer cells against normal HUVEC cells (4-fold) than first-line oxaliplatin. The outstanding anticancer activity of 2 is partly attributed to its dramatic increase in cellular uptake, DNA damage, and apoptosis. Mechanistic studies indicated that 2 inhibited HCT116 cell metastasis by triggering TMEM16A, COX-2, and their downstream signaling pathways, including EGFR, STAT3, E-cadherin and N-cadherin.
Collapse
Affiliation(s)
- Zhong-Ying Ma
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University Tianjin 300070 China
| | - Xiao-Jing Ding
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University Tianjin 300070 China
| | - Zhen-Zhen Zhu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University Tianjin 300070 China
| | - Qian Chen
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University Tianjin 300070 China
| | - Dong-Bo Wang
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University Tianjin 300070 China
| | - Xin Qiao
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University Tianjin 300070 China
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University Tianjin 300070 China
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Tianjin Medical University Tianjin 300070 China
| |
Collapse
|
18
|
Fang J, Gerschel P, Singh K, Apfel UP, Suntharalingam K. Cobalt(III)-Macrocyclic Scaffolds with Anti-Cancer Stem Cell Activity. Molecules 2024; 29:2743. [PMID: 38930809 PMCID: PMC11206342 DOI: 10.3390/molecules29122743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Cobalt(III) compounds with tetradentate ligands have been widely employed to deliver cytotoxic and imaging agents into cells. A large body of work has focused on using cobalt(III)-cyclam scaffolds for this purpose. Here, we investigate the cytotoxic properties of cobalt(III) complexes containing 14-membered macrocycles related to cyclam. A breast cancer stem cell (CSC) in vitro model was used to gauge efficacy. Specifically, [Co(1,4,7,11-tetraazacyclotetradecane)Cl2]+ (1) and [Co(1-oxa-4,8,12-triazacyclotetradecane)Cl2]+ (2) were synthesised and characterised, and their breast CSC activity was determined. The cobalt(III) complexes 1 and 2 displayed micromolar potency towards bulk breast cancer cells and breast CSCs grown in monolayers. Notably, 1 and 2 displayed selective potency towards breast CSCs over bulk breast cancer cells (up to 4.5-fold), which was similar to salinomycin (an established breast CSC-selective agent). The cobalt(III) complexes 1 and 2 were also able to inhibit mammosphere formation at low micromolar doses (with respect to size and number). The mammopshere inhibitory effect of 2 was similar to that of salinomycin. Our studies show that cobalt(III) complexes with 1,4,7,11-tetraazacyclotetradecane and 1-oxa-4,8,12-triazacyclotetradecane macrocycles could be useful starting points for the development of new cobalt-based delivery systems that can transport cytotoxic and imaging agents into breast CSCs.
Collapse
Affiliation(s)
- Jiaxin Fang
- School of Chemistry, University of Leicester, Leicester LE1 7RH, UK; (J.F.); (K.S.)
| | - Philipp Gerschel
- Inorganic Chemistry I, Ruhr-Universität Bochum, Universitätsstr. 150, 44801 Bochum, Germany;
| | - Kuldip Singh
- School of Chemistry, University of Leicester, Leicester LE1 7RH, UK; (J.F.); (K.S.)
| | - Ulf-Peter Apfel
- Inorganic Chemistry I, Ruhr-Universität Bochum, Universitätsstr. 150, 44801 Bochum, Germany;
- Fraunhofer UMSICHT, Osterfelder Str. 3, 46047 Oberhausen, Germany
| | | |
Collapse
|
19
|
Antonucci L, Karin M. The Past and Future of Inflammation as a Target to Cancer Prevention. Cancer Prev Res (Phila) 2024; 17:141-155. [PMID: 38271694 PMCID: PMC10987280 DOI: 10.1158/1940-6207.capr-23-0423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/21/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024]
Abstract
Inflammation is an essential defense mechanism in which innate immune cells are coordinately activated on encounter of harmful stimuli, including pathogens, tissue injury, and toxic compounds and metabolites to neutralize and eliminate the instigator and initiate healing and regeneration. Properly terminated inflammation is vital to health, but uncontrolled runaway inflammation that becomes chronic begets a variety of inflammatory and metabolic diseases and increases cancer risk. Making damaged tissues behave as "wounds that do not heal" and sustaining the production of growth factors whose physiologic function is tissue healing, chronic inflammation accelerates cancer emergence from premalignant lesions. In 1863, Rudolf Virchow, a leading German pathologist, suggested a possible association between inflammation and tumor formation, but it took another 140 years to fully elucidate and appreciate the tumorigenic role of inflammation. Key findings outlined molecular events in the inflammatory cascade that promote cancer onset and progression and enabled a better appreciation of when and where inflammation should be inhibited. These efforts triggered ongoing research work to discover and develop inflammation-reducing chemopreventive strategies for decreasing cancer risk and incidence.
Collapse
Affiliation(s)
- Laura Antonucci
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine; La Jolla, CA 92093, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine; La Jolla, CA 92093, USA
| |
Collapse
|
20
|
Li Y, Fang J, Singh K, Ortu F, Suntharalingam K. An immunogenic anti-cancer stem cell bi-nuclear copper(II)-flufenamic acid complex. Dalton Trans 2024; 53:6410-6415. [PMID: 38501501 DOI: 10.1039/d4dt00384e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
An asymmetric bi-nuclear copper(II) complex with both cytotoxic and immunogenic activity towards breast cancer stem cells (CSCs) is reported. The bi-nuclear copper(II) complex comprises of two copper(II) centres bound to flufenamic acid and 3,4,7,8-tetramethyl-1,10-phenanthroline. The bi-nuclear copper(II) complex exhibits sub-micromolar potency towards breast CSCs grown in monolayers and three-dimensional cultures. Remarkably, the bi-nuclear copper(II) complex is up to 25-fold more potent toward breast CSC mammospheres than salinomycin (a gold standard anti-breast CSC agent) and cisplatin (a clinically administered metallodrug). Mechanistic studies showed that the bi-nuclear copper(II) complex readily enters breast CSCs, elevates intracellular reactive oxygen species levels, induces apoptosis, and promotes damage-associated molecular pattern release. The latter triggers phagocytosis of breast CSCs by macrophages. As far as we are aware, this is the first report of a bi-nuclear copper(II) complex to induce engulfment of breast CSCs by immune cells.
Collapse
Affiliation(s)
- Yue Li
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK.
| | - Jiaxin Fang
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK.
| | - Kuldip Singh
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK.
| | - Fabrizio Ortu
- School of Chemistry, University of Leicester, Leicester, LE1 7RH, UK.
| | | |
Collapse
|
21
|
Fang J, Orobator ON, Olelewe C, Passeri G, Singh K, Awuah SG, Suntharalingam K. A Breast Cancer Stem Active Cobalt(III)-Cyclam Complex Containing Flufenamic Acid with Immunogenic Potential. Angew Chem Int Ed Engl 2024; 63:e202317940. [PMID: 38063406 PMCID: PMC10952489 DOI: 10.1002/anie.202317940] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Indexed: 12/31/2023]
Abstract
The cytotoxic and immunogenic-activating properties of a cobalt(III)-cyclam complex bearing the non-steroidal anti-inflammatory drug, flufenamic acid is reported within the context of anti-cancer stem cell (CSC) drug discovery. The cobalt(III)-cyclam complex 1 displays sub-micromolar potency towards breast CSCs grown in monolayers, 24-fold and 31-fold greater than salinomycin (an established anti-breast CSC agent) and cisplatin (an anticancer metallopharmaceutical), respectively. Strikingly, the cobalt(III)-cyclam complex 1 is 69-fold and 50-fold more potent than salinomycin and cisplatin towards three-dimensionally cultured breast CSC mammospheres. Mechanistic studies reveal that 1 induces DNA damage, inhibits cyclooxygenase-2 expression, and prompts caspase-dependent apoptosis. Breast CSCs treated with 1 exhibit damage-associated molecular patterns characteristic of immunogenic cell death and are phagocytosed by macrophages. As far as we are aware, 1 is the first cobalt complex of any oxidation state or geometry to display both cytotoxic and immunogenic-activating effects on breast CSCs.
Collapse
Affiliation(s)
- Jiaxin Fang
- School of ChemistryUniversity of LeicesterLeicesterUK
| | | | | | | | - Kuldip Singh
- School of ChemistryUniversity of LeicesterLeicesterUK
| | - Samuel G. Awuah
- Department of ChemistryUniversity of KentuckyLexingtonKYUSA
- Department of Pharmaceutical SciencesUniversity of KentuckyLexingtonKYUSA
| | | |
Collapse
|
22
|
Patterson C, Hazime KS, Zelenay S, Davis DM. Prostaglandin E₂ impacts multiple stages of the natural killer cell antitumor immune response. Eur J Immunol 2024; 54:e2350635. [PMID: 38059519 DOI: 10.1002/eji.202350635] [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: 06/29/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/08/2023]
Abstract
Tumor immune escape is a major factor contributing to cancer progression and unresponsiveness to cancer therapies. Tumors can produce prostaglandin E2 (PGE2 ), an inflammatory mediator that directly acts on Natural killer (NK) cells to inhibit antitumor immunity. However, precisely how PGE2 influences NK cell tumor-restraining functions remains unclear. Here, we report that following PGE₂ treatment, human NK cells exhibited altered expression of specific activating receptors and a reduced ability to degranulate and kill cancer targets. Transcriptional analysis uncovered that PGE₂ also differentially modulated the expression of chemokine receptors by NK cells, inhibiting CXCR3 but increasing CXCR4. Consistent with this, PGE₂-treated NK cells exhibited decreased migration to CXCL10 but increased ability to migrate toward CXCL12. Using live cell imaging, we showed that in the presence of PGE2 , NK cells were slower and less likely to kill cancer target cells following conjugation. Imaging the sequential stages of NK cell killing revealed that PGE₂ impaired NK cell polarization, but not the re-organization of synaptic actin or the release of perforin itself. Together, these findings demonstrate that PGE₂ affects multiple but select NK cell functions. Understanding how cancer cells subvert NK cells is necessary to more effectively harness the cancer-inhibitory function of NK cells in treatments.
Collapse
Affiliation(s)
- Chloe Patterson
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
| | - Khodor S Hazime
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, United Kingdom
| | - Santiago Zelenay
- The Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, United Kingdom
- Cancer Inflammation and Immunity Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Daniel M Davis
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, South Kensington, London, United Kingdom
| |
Collapse
|
23
|
Kassab AE, Gedawy EM. Recent Advancements in Refashioning of NSAIDs and their Derivatives as Anticancer Candidates. Curr Pharm Des 2024; 30:1217-1239. [PMID: 38584541 DOI: 10.2174/0113816128304230240327044201] [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: 01/13/2024] [Revised: 03/02/2024] [Accepted: 03/09/2024] [Indexed: 04/09/2024]
Abstract
Inflammation is critical to the formation and development of tumors and is closely associated with cancer. Therefore, addressing inflammation and the mediators that contribute to the inflammatory process may be a useful strategy for both cancer prevention and treatment. Tumor predisposition can be attributed to inflammation. It has been demonstrated that NSAIDs can modify the tumor microenvironment by enhancing apoptosis and chemosensitivity and reducing cell migration. There has been a recent rise in interest in drug repositioning or repurposing because the development of innovative medications is expensive, timeconsuming, and presents a considerable obstacle to drug discovery. Repurposing drugs is crucial for the quicker and less expensive development of anticancer medicines, according to an increasing amount of research. This review summarizes the antiproliferative activity of derivatives of NSAIDs such as Diclofenac, Etodolac, Celecoxib, Ibuprofen, Tolmetin, and Sulindac, published between 2017 and 2023. Their mechanism of action and structural activity relationships (SARs) were also discussed to set the path for potential future repositioning of NSAIDs for clinical deployment in the treatment of cancer.
Collapse
Affiliation(s)
- Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Ehab M Gedawy
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, P.O. Box 11562, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC), Badr City, Cairo, P.O. Box 11829, Egypt
| |
Collapse
|
24
|
Richter S, Steenblock C, Fischer A, Lemm S, Ziegler CG, Bechmann N, Nölting S, Pietzsch J, Ullrich M. Improving susceptibility of neuroendocrine tumors to radionuclide therapies: personalized approaches towards complementary treatments. Theranostics 2024; 14:17-32. [PMID: 38164150 PMCID: PMC10750207 DOI: 10.7150/thno.87345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/30/2023] [Indexed: 01/03/2024] Open
Abstract
Radionuclide therapies are an important tool for the management of patients with neuroendocrine neoplasms (NENs). Especially [131I]MIBG and [177Lu]Lu-DOTA-TATE are routinely used for the treatment of a subset of NENs, including pheochromocytomas, paragangliomas and gastroenteropancreatic tumors. Some patients suffering from other forms of NENs, such as medullary thyroid carcinoma or neuroblastoma, were shown to respond to radionuclide therapy; however, no general recommendations exist. Although [131I]MIBG and [177Lu]Lu-DOTA-TATE can delay disease progression and improve quality of life, complete remissions are achieved rarely. Hence, better individually tailored combination regimes are required. This review summarizes currently applied radionuclide therapies in the context of NENs and informs about recent advances in the development of theranostic agents that might enable targeting subgroups of NENs that previously did not respond to [131I]MIBG or [177Lu]Lu-DOTA-TATE. Moreover, molecular pathways involved in NEN tumorigenesis and progression that mediate features of radioresistance and are particularly related to the stemness of cancer cells are discussed. Pharmacological inhibition of such pathways might result in radiosensitization or general complementary antitumor effects in patients with certain genetic, transcriptomic, or metabolic characteristics. Finally, we provide an overview of approved targeted agents that might be beneficial in combination with radionuclide therapies in the context of a personalized molecular profiling approach.
Collapse
Affiliation(s)
- Susan Richter
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Charlotte Steenblock
- Department of Internal Medicine III, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Alessa Fischer
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), and University of Zurich (UZH), Zurich, Switzerland
| | - Sandy Lemm
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Dresden, Germany
| | - Christian G. Ziegler
- Department of Internal Medicine III, University Clinic Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- University Hospital Würzburg, Division of Endocrinology and Diabetes, Würzburg, Germany
| | - Nicole Bechmann
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Svenja Nölting
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), and University of Zurich (UZH), Zurich, Switzerland
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Dresden, Germany
| | - Martin Ullrich
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
25
|
Ayaz M, Alam A, Zainab, Assad M, Javed A, Islam MS, Rafiq H, Ali M, Ahmad W, Khan A, Latif A, Al-Harrasi A, Ahmad M. Biooriented Synthesis of Ibuprofen-Clubbed Novel Bis-Schiff Base Derivatives as Potential Hits for Malignant Glioma: In Vitro Anticancer Activity and In Silico Approach. ACS OMEGA 2023; 8:49228-49243. [PMID: 38173864 PMCID: PMC10764114 DOI: 10.1021/acsomega.3c07216] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/18/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
This research work is based on the synthesis of bis-Schiff base derivatives of the commercially available ibuprofen drug in outstanding yields through multistep reactions. Structures of the synthesized compounds were confirmed by the help of modern spectroscopic techniques including high-resolution electrospray ionization mass spectrometry (HR-ESI-MS), 1H NMR, and 13C NMR. The synthesized compounds were evaluated for their anticancer activity using a normal human embryonic kidney HEK293 cell and U87-malignant glioma (ATCC-HTB-14) as a cancer cell line. All of the synthesized compounds among the series exhibited excellent to less antiproliferative activity having IC50 values ranging from 5.75 ± 0.43 to 150.45 ± 0.20 μM. Among them, compound 5e (IC50 = 5.75 ± 0.43 μM) was found as the most potent antiprolifarative agent, while 5f, 5b, 5a, 5n, 5r, 5s, 5g, 5q, 5i, and 5j exhibited good activity with IC50 values from 24.17 ± 0.46 to 43.71 ± 0.07 μM. These findings suggest that these cells (HEK293) are less cytotoxic to the activities of compounds and increase the cancer cell death in brain, while the lower cytotoxicity of the potent compounds in noncancerous cells suggests that these derivatives will provide promising treatment for patients suffering from brain cancer. The results of the docking study exposed a promising affinity of the active compounds toward casein kinase-2 enzyme, which shows green signal for cancer treatment.
Collapse
Affiliation(s)
- Muhammad Ayaz
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Aftab Alam
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Zainab
- College
of Chemistry and Materials Science, Hebei
Normal University, Shijiazhuang 050024, China
| | - Mohammad Assad
- Department
of Biochemistry, Abdul Wali Khan University
Mardan, Khyber
Pakhtunkhwa 23200, Pakistan
| | - Aneela Javed
- Molecular
Immunology Laboratory, Department of Healthcare Biotechnology Atta-Ur-Rahman
School of Applied Biosciences, National
University of Sciences and Technology, H-12 Campus, Islamabad 44000, Pakistan
| | - Mohammad Shahidul Islam
- Department
of Chemistry, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Huma Rafiq
- Molecular
Immunology Laboratory, Department of Healthcare Biotechnology Atta-Ur-Rahman
School of Applied Biosciences, National
University of Sciences and Technology, H-12 Campus, Islamabad 44000, Pakistan
| | - Mumtaz Ali
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Waqar Ahmad
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Ajmal Khan
- Natural and
Medical Sciences Research Center, University
of Nizwa, P.O. Box 33, Birkat Al Mauz, PC 616 Nizwa, Sultanate of Oman
| | - Abdul Latif
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| | - Ahmed Al-Harrasi
- Natural and
Medical Sciences Research Center, University
of Nizwa, P.O. Box 33, Birkat Al Mauz, PC 616 Nizwa, Sultanate of Oman
| | - Manzoor Ahmad
- Department
of Chemistry, University of Malakand, Dir Lower, Khyber Pakhtunkhwa 18800, Pakistan
| |
Collapse
|
26
|
Aliabadi A, Khanniri E, Mahboubi-Rabbani M, Bayanati M. Dual COX-2/15-LOX inhibitors: A new avenue in the prevention of cancer. Eur J Med Chem 2023; 261:115866. [PMID: 37862815 DOI: 10.1016/j.ejmech.2023.115866] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/07/2023] [Accepted: 10/09/2023] [Indexed: 10/22/2023]
Abstract
Dual cyclooxygenase 2/15-lipoxygenase inhibitors constitute a valuable alternative to classical non-steroidal anti-inflammatory drugs (NSAIDs) and selective COX-2 (cyclooxygenase-2) inhibitors for the treatment of inflammatory diseases, as well as preventing the cancer. Indeed, these latter present diverse side effects, which are reduced or absent in dual-acting agents. In this review, COX-2 and 15-LOX (15-lipoxygenase) pathways are first described in order to highlight the therapeutic interest of designing such compounds. Various structural families of dual inhibitors are illustrated. This study discloses various structural families of dual 15-LOX/COX-2 inhibitors, thus pave the way to design potentially-active anticancer agents with balanced dual inhibition of these enzymes.
Collapse
Affiliation(s)
- Ali Aliabadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elham Khanniri
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahboubi-Rabbani
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maryam Bayanati
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Kobayashi A, Hiroyama Y, Mamiya T, Oikawa M, Konishi T. The COX-2/PGE2 Response Pathway Upregulates Radioresistance in A549 Human Lung Cancer Cells through Radiation-Induced Bystander Signaling. BIOLOGY 2023; 12:1368. [PMID: 37997966 PMCID: PMC10669009 DOI: 10.3390/biology12111368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023]
Abstract
This study aimed to determine the mechanism underlying the modulation of radiosensitivity in cancer cells by the radiation-induced bystander effect (RIBE). We hypothesized that the RIBE mediates cyclooxygenase-2 (COX-2) and its metabolite prostaglandin E2 (PGE2) in elevating radioresistance in unirradiated cells. In this study, we used the SPICE-QST microbeam irradiation system to target 0.07-0.7% cells by 3.4-MeV proton microbeam in the cell culture sample, such that most cells in the dish became bystander cells. Twenty-four hours after irradiation, we observed COX-2 protein upregulation in microbeam-irradiated cells compared to that of controls. Additionally, 0.29% of the microbeam-irradiated cells exhibited increased cell survival and a reduced micronucleus rate against X-ray irradiation compared to that of non-microbeam irradiated cells. The radioresistance response was diminished in both cell groups with the hemichannel inhibitor and in COX-2-knockout cells under cell-to-cell contact and sparsely distributed conditions. The results indicate that the RIBE upregulates the cell radioresistance through COX-2/PGE2 intercellular responses, thereby contributing to issues, such as the risk of cancer recurrence.
Collapse
Affiliation(s)
- Alisa Kobayashi
- Single Cell Radiation Biology Team, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inageku, Chiba 263-8555, Japan
- Radiation Effect Research Group, Department of Accelerator and Medical Physics, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inageku, Chiba 263-8555, Japan
| | - Yota Hiroyama
- Single Cell Radiation Biology Team, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inageku, Chiba 263-8555, Japan
- Department of Radiological Technology, Graduate School of Health Sciences, Hirosaki University, 66-1 Hon-cho, Hirosaki-shi, Aomori 036-8564, Japan
- Department of Radiology, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba 260-8677, Japan
| | - Taisei Mamiya
- Single Cell Radiation Biology Team, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inageku, Chiba 263-8555, Japan
- Graduate School of Science, Rikkyo (St. Paul’s) University, 3-34-1 Nishi-Ikebukuro, Toshima-ku, Tokyo 171-8501, Japan
| | - Masakazu Oikawa
- Electrostatic Accelerator Operation Section, Department of Accelerator and Medical Physics, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inageku, Chiba 263-8555, Japan
| | - Teruaki Konishi
- Single Cell Radiation Biology Team, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inageku, Chiba 263-8555, Japan
- Department of Radiological Technology, Graduate School of Health Sciences, Hirosaki University, 66-1 Hon-cho, Hirosaki-shi, Aomori 036-8564, Japan
- Graduate School of Science, Rikkyo (St. Paul’s) University, 3-34-1 Nishi-Ikebukuro, Toshima-ku, Tokyo 171-8501, Japan
| |
Collapse
|
28
|
Ahsan H, Malik SI, Shah FA, El-Serehy HA, Ullah A, Shah ZA. Celecoxib Suppresses NF-κB p65 (RelA) and TNFα Expression Signaling in Glioblastoma. J Clin Med 2023; 12:6683. [PMID: 37892820 PMCID: PMC10607796 DOI: 10.3390/jcm12206683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/13/2023] [Accepted: 08/03/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) harbors significant genetic heterogeneity, high infiltrative capacity, and patterns of relapse following many therapies. The expression of nuclear factor kappa-B (NF-κB p65 (RelA)) and signaling pathways is constitutively activated in GBM through inflammatory stimulation such as tumor necrosis factor-alpha (TNFα), cell invasion, motility, abnormal physiological stimuli, and inducible chemoresistance. However, the underlying anti-tumor and anti-proliferative mechanisms of NF-κB p65 (RelA) and TNFα are still poorly defined. This study aimed to investigate the expression profiling of NF-κB p65 (RelA) and TNFα as well as the effectiveness of celecoxib along with temozolomide (TMZ) in reducing the growth of the human GBM cell line SF-767. METHODS genome-wide expression profiling, enrichment analysis, immune infiltration, quantitative expression, and the Microculture Tetrazolium Test (MTT) proliferation assay were performed to appraise the effects of celecoxib and TMZ. RESULTS demonstrated the upregulation of NF-κB p65 (RelA) and TNFα and celecoxib reduced the viability of the human glioblastoma cell line SF-767, cell proliferation, and NF-κB p65 (RelA) and TNFα expression in a dose-dependent manner. Overall, these findings demonstrate for the first time how celecoxib therapy could mitigate the invasive characteristics of the human GBM cell line SF-767 by inhibiting the NF-κB mediated stimulation of the inflammatory cascade. CONCLUSION based on current findings, we propose that celecoxib as a drug candidate in combination with temozolomide might dampen the transcriptional and enzymatic activities associated with the aggressiveness of GBM and reduce the expression of GBM-associated NF-κB p65 (RelA) and TNFα inflammatory genes expression.
Collapse
Affiliation(s)
- Hina Ahsan
- Department of Bioinformatics and Biosciences, Faculty of Health and Life Sciences, Capital University of Science and Technology (CUST), Islamabad 44000, Pakistan;
- Riphah Institute of Pharmaceutical Sciences Islamabad, Riphah International University, Islamabad 44000, Pakistan
| | - Shaukat Iqbal Malik
- Department of Bioinformatics and Biosciences, Faculty of Health and Life Sciences, Capital University of Science and Technology (CUST), Islamabad 44000, Pakistan;
| | - Fawad Ali Shah
- Swat College of Pharmaceutical Sciences, Swat 19200, Pakistan;
| | - Hamed A. El-Serehy
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Amin Ullah
- Department of Health and Biological Sciences, Abasyn University Peshawar, Peshawar 25000, Pakistan;
- Institute of Pathology, University Hospital of Cologne, 50923 Cologne, Germany
| | - Zafar Abbas Shah
- Department of Bioinformatics, Hazara University, Mansehra 21120, Pakistan
| |
Collapse
|
29
|
Zeiz A, Kawtharani R, Elmasri M, Khawaja G, Hamade E, Habib A, Ayoub AJ, Abarbri M, El-Dakdouki MH. Molecular properties prediction, anticancer and anti-inflammatory activities of some pyrimido[1,2-b]pyridazin-2-one derivatives. BIOIMPACTS : BI 2023; 14:27688. [PMID: 38505674 PMCID: PMC10945296 DOI: 10.34172/bi.2023.27688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 08/08/2023] [Accepted: 08/23/2023] [Indexed: 03/21/2024]
Abstract
Introduction The anticancer and anti-inflammatory activities of a novel series of eleven pyrimido[1,2-b]pyridazin-2-one analogues substituted at position 7 were assessed in the current study. Methods The physicochemical characteristics were studied using MolSoft software. The antiproliferative activity was investigated by MTT cell viability assay, and cell cycle analysis elucidated the antiproliferative mechanism of action. Western blot analysis examined the expression levels of key pro-apoptotic (Bax, p53) and pro-survival (Bcl-2) proteins. The anti-inflammatory activity was assessed by measuring the production levels of nitric oxide in RAW264.7 cells, and the expression levels of COX-2 enzyme in LPS-activated THP-1 cells. In addition, the gene expression of various pro-inflammatory cytokines (IL-6, IL-8, IL-1β, TNF-α) and chemokines (CCL2, CXCL1, CXCL2, CXCL3) was assessed by RT-qPCR. Results Compound 1 bearing a chlorine substituent displayed the highest cytotoxic activity against HCT-116 and MCF-7 cancer cells where IC50 values of 49.35 ± 2.685 and 69.32 ± 3.186 µM, respectively, were achieved. Compound 1 increased the expression of pro-apoptotic proteins p53 and Bax while reducing the expression of pro-survival protein Bcl-2. Cell cycle analysis revealed that compound 1 arrested cell cycle at the G0/G1 phase. Anti-inflammatory assessments revealed that compound 1 displayed the strongest inhibitory activity on NO production with IC50 of 29.94 ± 2.24 µM, and down-regulated the expression of COX-2. Compound 1 also induced a statistically significant decrease in the gene expression of various cytokines and chemokines. Conclusion These findings showed that the pyrimidine derivative 1 displayed potent anti-inflammatory and anticancer properties in vitro, and can be selected as a lead compound for further investigation.
Collapse
Affiliation(s)
- Ali Zeiz
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Debbieh, Lebanon
| | - Ranin Kawtharani
- Laboratory of Medicinal Chemistry and Natural Products, Lebanese University, Faculty of Science-I, Beirut, Lebanon
| | - Mirvat Elmasri
- Department of Chemistry and Biochemistry, Faculty of Science-I, Lebanese University, Beirut, Lebanon
| | - Ghada Khawaja
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Debbieh, Lebanon
| | - Eva Hamade
- Department of Chemistry and Biochemistry, Faculty of Science-I, Lebanese University, Beirut, Lebanon
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Aida Habib
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha 2713, Qatar
| | - Abeer J. Ayoub
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Mohamed Abarbri
- Laboratoire de Physico-Chimie des Matériaux et des Electrolytes pour l'Energie (PCM2E)., EA 6299. Avenue Monge Faculté des Sciences, Parc de Grandmont, 37200 Tours, France
| | | |
Collapse
|
30
|
Lu G, Wang X, Cheng M, Wang S, Ma K. The multifaceted mechanisms of ellagic acid in the treatment of tumors: State-of-the-art. Biomed Pharmacother 2023; 165:115132. [PMID: 37423169 DOI: 10.1016/j.biopha.2023.115132] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023] Open
Abstract
Ellagic acid (EA) is a kind of polyphenol compound extracted from a variety of herbs, such as paeoniae paeoniae, raspberry, Chebule, walnut kernel, myrrh, loquat leaf, pomegranate bark, quisquite, and fairy herb. It has anti-tumor, anti-oxidation, anti-inflammatory, anti-mutation, anti-bacterial, anti-allergic and multiple pharmacological properties. Studies have shown its anti-tumor effect in gastric cancer, liver cancer, pancreatic cancer, breast cancer, colorectal cancer, lung cancer and other malignant tumors, mainly through inducing tumor cell apoptosis, inhibiting tumor cell proliferation, inhibiting tumor cell metastasis and invasion, inducing autophagy, affecting tumor metabolic reprogramming and other forms of anti-tumor efficacy. Its molecular mechanism is mainly reflected in inhibiting the proliferation of tumor cells through VEGFR-2 signaling pathway, Notch signaling pathway, PKC signaling pathway and COX-2 signaling pathway. PI3K/Akt signaling pathway, JNK (cJun) signaling pathway, mitochondrial pathway, Bcl-2 / Bax signaling pathway, TGF-β/Smad3 signaling pathway induced apoptosis of tumor cells and blocked EMT process and MMP SDF1α/CXCR4 signaling pathway inhibits the metastasis and invasion of tumor cells, induces autophagy and affects tumor metabolic reprogramming to produce anti-tumor effects. At present, the analysis of the anti-tumor mechanism of ellagic acid is slightly lacking, so this study comprehensively searched the literature on the anti-tumor mechanism of ellagic acid in various databases, reviewed the research progress of the anti-tumor effect and mechanism of ellagic acid, in order to provide reference and theoretical basis for the further development and application of ellagic acid.
Collapse
Affiliation(s)
- Guangying Lu
- Shandong Co-Innovation Center of Classic Traditional Chinese Medicine Formula, Shandong University of Traditional Chinese Medicine, China
| | - Xuezhen Wang
- Tianjin University of Traditional Chinese Medicine, China
| | - Ming Cheng
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, China
| | - Shijun Wang
- Shandong Co-Innovation Center of Classic Traditional Chinese Medicine Formula, Shandong University of Traditional Chinese Medicine, China.
| | - Ke Ma
- Shandong Co-Innovation Center of Classic Traditional Chinese Medicine Formula, Shandong University of Traditional Chinese Medicine, China.
| |
Collapse
|
31
|
Chebaro Z, Abdallah R, Badran A, Hamade K, Hijazi A, Maresca M, Mesmar JE, Baydoun E. Study of the antioxidant and anti-pancreatic cancer activities of Anchusa strigosa aqueous extracts obtained by maceration and ultrasonic extraction techniques. Front Pharmacol 2023; 14:1201969. [PMID: 37593172 PMCID: PMC10427766 DOI: 10.3389/fphar.2023.1201969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy and a leading cause of cancer-related deaths worldwide. Moreover, the incidence and mortality rates for pancreatic cancer are projected to keep increasing. A major challenge in the treatment of pancreatic cancer is the lack of effective screening approaches, which contributes to its poor prognosis, indicating the need for new treatment regimens and alternative therapies, such as herbal medicine. The medicinal plant A. strigosa, which is widely distributed in the Eastern Mediterranean region, is a short prickly plant from the Boraginaceae family that has been widely used in traditional medicine for treating various diseases. Nevertheless, its effect on human pancreatic cancer remains poorly investigated. In the present study, we screened the phytochemical content of Anchusa strigosa aqueous extracts obtained by maceration and ultrasound-assisted methods (ASM and ASU, respectively) and evaluated their antioxidant effects. We also investigated their anticancer effects and possible underlying mechanisms. The results show that both extracts were rich in bioactive molecules, with slight differences in their composition. Both extracts exhibited remarkable antioxidant potential and potent radical-scavenging activity in vitro. Additionally, non-cytotoxic concentrations of both extracts attenuated cell proliferation in a time- and concentration-dependent manner, which was associated with a decrease in the proliferation marker Ki67 and an induction of the intrinsic apoptotic pathway. Furthermore, the extracts increased the aggregation of pancreatic cancer cells and reduced their migratory potential, with a concomitant downregulation of integrin β1. Finally, we showed that the ASM extract caused a significant decrease in the levels of COX-2, an enzyme that has been linked to inflammation, carcinogenesis, tumor progression, and metastasis. Taken together, our findings provide evidence that A. strigosa extracts, particularly the extract obtained using the maceration method, have a potential anticancer effect and may represent a new resource for the design of novel drugs against pancreatic cancer.
Collapse
Affiliation(s)
- Ziad Chebaro
- Platforme de Recherche et D’analyse en Sciences de L’environnement (EDST-PRASE), Beirut, Lebanon
| | - Rola Abdallah
- Department of Biology, American University of Beirut, Beirut, Lebanon
| | - Adnan Badran
- Department of Nutrition, University of Petra, Amman, Jordan
| | - Kamar Hamade
- UMRT INRE 1158 BioEcoAgro, Laboratorie BIOPI, University of Picardie Jules Verne, Amiens, France
| | - Akram Hijazi
- Platforme de Recherche et D’analyse en Sciences de L’environnement (EDST-PRASE), Beirut, Lebanon
| | - Marc Maresca
- Aix-Marseille Univ, CNRS, Centrale Marseille, iSM2, Marseille, France
| | | | - Elias Baydoun
- Department of Biology, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
32
|
Şenol H, Çağman Z, Gençoğlu Katmerlikaya T, Sinan Tokalı F. New Anthranilic Acid Hydrazones as Fenamate Isosteres: Synthesis, Characterization, Molecular Docking, Dynamics & in Silico ADME, in Vitro Anti-Inflammatory and Anticancer Activity Studies. Chem Biodivers 2023; 20:e202300773. [PMID: 37384873 DOI: 10.1002/cbdv.202300773] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/15/2023] [Accepted: 06/29/2023] [Indexed: 07/01/2023]
Abstract
In this study, twenty new anthranilic acid hydrazones 6-9 (a-e) were synthesized and their structures were characterized by Fourier-transform Infrared (FT-IR), Nuclear Magnetic Resonance (1 H-NMR - 13 C-NMR), and High-resolution Mass Spectroscopy (HR-MS). The inhibitory effects of the compounds against COX-II were evaluated. IC50 values of the compounds were found in the range of >200-0.32 μM and compounds 6e, 8d, 8e, 9b, 9c, and 9e were determined to be the most effective inhibitors. Cytotoxic effects of the most potent compounds were investigated against human hepatoblastoma (Hep-G2) and human healthy embryonic kidney (Hek-293) cell lines. Doxorubicin (IC50 : 8.68±0.16 μM for Hep-G2, 55.29±0.56 μM for Hek-293) was used as standard. 8e is the most active compound, with low IC50 against Hep-G2 (4.80±0.04 μM), high against Hek-293 (159.30±3.12), and high selectivity (33.15). Finally, molecular docking and dynamics studies were performed to understand ligand-protein interactions between the most potent compounds and COX II, Epidermal Growth Factor Receptor (EGFR), and Transforming Growth Factor beta II (TGF-βII). The docking scores were calculated in the range of -10.609--6.705 kcal/mol for COX-II, -8.652--7.743 kcal/mol for EGFR, and -10.708--8.596 kcal/mol for TGF-βII.
Collapse
Affiliation(s)
- Halil Şenol
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, Fatih, 34093, Istanbul, Türkiye
| | - Zeynep Çağman
- Bezmialem Vakif University, Faculty of Pharmacy, Department of Biochemistry, Fatih, 34093, Istanbul, Türkiye
| | - Tuğba Gençoğlu Katmerlikaya
- Bezmialem Vakif University, Institute of Health Sciences, Department of Biotechnology, 34093, İstanbul, Türkiye
| | - Feyzi Sinan Tokalı
- Kafkas University, Kars Vocational School, Department of Material and Material Processing Technologies, 36100, Kars, Türkiye
| |
Collapse
|
33
|
Kciuk M, Alam M, Ali N, Rashid S, Głowacka P, Sundaraj R, Celik I, Yahya EB, Dubey A, Zerroug E, Kontek R. Epigallocatechin-3-Gallate Therapeutic Potential in Cancer: Mechanism of Action and Clinical Implications. Molecules 2023; 28:5246. [PMID: 37446908 PMCID: PMC10343677 DOI: 10.3390/molecules28135246] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Cellular signaling pathways involved in the maintenance of the equilibrium between cell proliferation and apoptosis have emerged as rational targets that can be exploited in the prevention and treatment of cancer. Epigallocatechin-3-gallate (EGCG) is the most abundant phenolic compound found in green tea. It has been shown to regulate multiple crucial cellular signaling pathways, including those mediated by EGFR, JAK-STAT, MAPKs, NF-κB, PI3K-AKT-mTOR, and others. Deregulation of the abovementioned pathways is involved in the pathophysiology of cancer. It has been demonstrated that EGCG may exert anti-proliferative, anti-inflammatory, and apoptosis-inducing effects or induce epigenetic changes. Furthermore, preclinical and clinical studies suggest that EGCG may be used in the treatment of numerous disorders, including cancer. This review aims to summarize the existing knowledge regarding the biological properties of EGCG, especially in the context of cancer treatment and prophylaxis.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (M.K.); (R.K.)
- Doctoral School of Exact and Natural Sciences, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
| | - Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Pola Głowacka
- Department of Medical Biochemistry, Medical University of Lodz, Mazowiecka 6/8, 90-001 Lodz, Poland;
- Doctoral School of Medical University of Lodz, Hallera 1 Square, 90-700 Lodz, Poland
| | - Rajamanikandan Sundaraj
- Department of Biochemistry, Centre for Drug Discovery, Karpagam Academy of Higher Education, Coimbatore 641021, India;
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri 38280, Turkey;
| | - Esam Bashir Yahya
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia;
| | - Amit Dubey
- Computational Chemistry and Drug Discovery Division, Quanta Calculus, Greater Noida 201310, India;
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospital, Chennai 600077, India
| | - Enfale Zerroug
- LMCE Laboratory, Group of Computational and Pharmaceutical Chemistry, University of Biskra, Biskra 07000, Algeria;
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland; (M.K.); (R.K.)
| |
Collapse
|
34
|
Yu Z, Peng Y, Gao J, Zhou M, Shi L, Zhao F, Wang C, Tian X, Feng L, Huo X, Zhang B, Liu M, Fang D, Ma X. The p23 co-chaperone is a succinate-activated COX-2 transcription factor in lung adenocarcinoma tumorigenesis. SCIENCE ADVANCES 2023; 9:eade0387. [PMID: 37390202 PMCID: PMC10313168 DOI: 10.1126/sciadv.ade0387] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 05/30/2023] [Indexed: 07/02/2023]
Abstract
P23, historically known as a heat shock protein 90 (HSP90) co-chaperone, exerts some of its critical functions in an HSP90-independent manner, particularly when it translocates into the nucleus. The molecular nature underlying how this HSP90-independent p23 function is achieved remains as a biological mystery. Here, we found that p23 is a previously unidentified transcription factor of COX-2, and its nuclear localization predicts the poor clinical outcomes. Intratumor succinate promotes p23 succinylation at K7, K33, and K79, which drives its nuclear translocation for COX-2 transcription and consequently fascinates tumor growth. We then identified M16 as a potent p23 succinylation inhibitor from 1.6 million compounds through a combined virtual and biological screening. M16 inhibited p23 succinylation and nuclear translocation, attenuated COX-2 transcription in a p23-dependent manner, and markedly suppressed tumor growth. Therefore, our study defines p23 as a succinate-activated transcription factor in tumor progression and provides a rationale for inhibiting p23 succinylation as an anticancer chemotherapy.
Collapse
Affiliation(s)
- Zhenlong Yu
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Yulin Peng
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Jian Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
- School of Medicine, Anhui University of Science and Technology, Huainan 232001, China
| | - Meirong Zhou
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Lei Shi
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Feng Zhao
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Chao Wang
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Xiangge Tian
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Lei Feng
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Xiaokui Huo
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Baojing Zhang
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| | - Min Liu
- Neurology Department, Dalian University Affiliated Xinhua Hospital, Dalian 116021, China
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Xiaochi Ma
- College of Pharmacy, the Second Affiliated Hospital, Dalian Medical University, Dalian 116000, China
| |
Collapse
|
35
|
Ganai SA, Rajamanikandan S, Shah BA, Lone A, Arwa F, Malik FA. Comparative structural study of selective and non-selective NSAIDs against the enzyme cyclooxygenase-2 through real-time molecular dynamics linked to post-dynamics MM-GBSA and e-pharmacophores mapping. J Mol Model 2023; 29:192. [PMID: 37256432 DOI: 10.1007/s00894-023-05603-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 05/23/2023] [Indexed: 06/01/2023]
Abstract
BACKGROUND Inflammation-provoked disorders including cancer are arbitrated by cyclooxygenase-2 (COX-2). Celecoxib and niflumic acid are among the potent and selective inhibitors of this enzyme while aspirin (acetylsalicylic acid) and sodium salicylate are its non-selective and lesser potent inhibitors. Despite these proven studies, the comparative structural study of these selective and non-selective molecules at atomistic scale in complex state with COX-2 that may answer this differential inhibitory behavior has not been accomplished spotlighting the imperative need of additional research in this area. Thus, this study was framed to provide a strong explanation for the enigma of higher inhibitory activity of celecoxib-niflumic acid duo in comparison to aspirin and sodium salicylate towards COX-2. METHODS A contemporary approach including advanced molecular docking against COX2, molecular dynamics of receptor-ligand complexes, simulation-trajectory-backed MMGBSA for different time points, radius of gyration (Rg) calculations, and e-pharmacophores approach was employed to attain a rational conclusion. RESULTS Our findings demonstrated the higher binding affinity of celecoxib and niflumic acid over aspirin and sodium salicylate against COX-2. Although both selective and non-selective COX-2 inhibitors manifested nearly the same stability in the active site of this enzyme but the e-pharmocophoric features found in the case of selective inhibitors scored over non-selective ones. Thus, our findings excluded the differential stability to be the cause of stronger potency of selective inhibitors but attributed their potency to greater number of complementary features present in these inhibitors against the active site of inflammation engendering COX-2.
Collapse
Affiliation(s)
- Shabir Ahmad Ganai
- Research Centre for Residue and Quality Analysis, FoH, SKUAST-Kashmir, Shalimar, Srinagar, 190025, Jammu & Kashmir, India.
| | - Sundararaj Rajamanikandan
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India
- Centre for Drug Discovery, Karpagam Academy of Higher Education, Coimbatore, 641021, Tamil Nadu, India
| | - Basit Amin Shah
- Department of Biotechnology, University of Kashmir, Srinagar, 190006, Jammu & Kashmir, India
| | - Asif Lone
- Department of Biochemistry, Deshbandhu College, University of Delhi, 110019, New Delhi, India
| | - Faieza Arwa
- Department of Veterinary Physiology, SKUAST-J-180009, Jammu, Jammu & Kashmir, India
| | - Firdose Ahmad Malik
- College of Temperate Sericulture, SKUAST-Kashmir, Mirgund, 193121, Jammu & Kashmir, India
| |
Collapse
|
36
|
Liu X, Wenisch D, Dahlke P, Jordan PM, Jakupec MA, Kowol CR, Liebing P, Werz O, Keppler BK, Weigand W. Multi-action platinum(IV) prodrugs conjugated with COX-inhibiting NSAIDs. Eur J Med Chem 2023; 257:115515. [PMID: 37295160 DOI: 10.1016/j.ejmech.2023.115515] [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: 03/23/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
In the last decades, inflammation has been recognized as being closely connected to cancer, and joint strategies encompassing chemotherapeutic and anti-inflammatory agents have been extensively studied. In this work, a series of novel cisplatin and oxaliplatin-based Pt(IV) complexes comprising non-steroidal anti-inflammatory drugs (NSAIDs) and their carboxyl ester analogues as axial moieties were synthesized. Several of the cisplatin-based Pt(IV) complexes 22-30 showed increased cytotoxicity in the human cancer cell lines CH1/PA-1, SW480 and A549 compared to the Pt(II) drug. For the most potent complex 26, comprising two aceclofenac (AFC) moieties, the formation of Pt(II)-9-methylguanine (9-MeG) adducts after activation with ascorbic acid (AsA) was proven. Additionally, a significant inhibition of cyclooxygenase (COX) activity and prostaglandin E2 (PGE2) production was observed, as well as increased cellular accumulation, depolarization of mitochondrial membranes, and strong proapoptotic potencies in SW480 cells. Overall, these systematic effects shown in vitro confer 26 as a potential anticancer agent combined with anti-inflammatory properties.
Collapse
Affiliation(s)
- Xiao Liu
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller Universität Jena, Humboldt Str. 8, 07743, Jena, Germany
| | - Dominik Wenisch
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Strasse 42, 1090, Vienna, Austria
| | - Philipp Dahlke
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Paul M Jordan
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743, Jena, Germany
| | - Michael A Jakupec
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Strasse 42, 1090, Vienna, Austria; Research Cluster 'Translational Cancer Therapy Research', University of Vienna, Währinger Strasse 42, 1090, Vienna, Austria
| | - Christian R Kowol
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Strasse 42, 1090, Vienna, Austria; Research Cluster 'Translational Cancer Therapy Research', University of Vienna, Währinger Strasse 42, 1090, Vienna, Austria
| | - Phil Liebing
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller Universität Jena, Humboldt Str. 8, 07743, Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Philosophenweg 14, D-07743, Jena, Germany.
| | - Bernhard K Keppler
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Währinger Strasse 42, 1090, Vienna, Austria; Research Cluster 'Translational Cancer Therapy Research', University of Vienna, Währinger Strasse 42, 1090, Vienna, Austria.
| | - Wolfgang Weigand
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller Universität Jena, Humboldt Str. 8, 07743, Jena, Germany.
| |
Collapse
|
37
|
Tacchini M, Sacchetti G, Guerrini A, Paganetto G. Mycochemicals against Cancer Stem Cells. Toxins (Basel) 2023; 15:360. [PMID: 37368660 DOI: 10.3390/toxins15060360] [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: 03/23/2023] [Revised: 05/08/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Since ancient times, mushrooms have been considered valuable allies of human well-being both from a dietary and medicinal point of view. Their essential role in several traditional medicines is explained today by the discovery of the plethora of biomolecules that have shown proven efficacy for treating various diseases, including cancer. Numerous studies have already been conducted to explore the antitumoural properties of mushroom extracts against cancer. Still, very few have reported the anticancer properties of mushroom polysaccharides and mycochemicals against the specific population of cancer stem cells (CSCs). In this context, β-glucans are relevant in modulating immunological surveillance against this subpopulation of cancer cells within tumours. Small molecules, less studied despite their spread and assortment, could exhibit the same importance. In this review, we discuss several pieces of evidence of the association between β-glucans and small mycochemicals in modulating biological mechanisms which are proven to be involved with CSCs development. Experimental evidence and an in silico approach are evaluated with the hope of contributing to future strategies aimed at the direct study of the action of these mycochemicals on this subpopulation of cancer cells.
Collapse
Affiliation(s)
- Massimo Tacchini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Gianni Sacchetti
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Alessandra Guerrini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Guglielmo Paganetto
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
38
|
Rashid G, Khan NA, Elsori D, Rehman A, Tanzeelah, Ahmad H, Maryam H, Rais A, Usmani MS, Babker AM, Kamal MA, Hafez W. Non-steroidal anti-inflammatory drugs and biomarkers: A new paradigm in colorectal cancer. Front Med (Lausanne) 2023; 10:1130710. [PMID: 36950511 PMCID: PMC10025514 DOI: 10.3389/fmed.2023.1130710] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Colorectal cancer is a sporadic, hereditary, or familial based disease in its origin, caused due to diverse set of mutations in large intestinal epithelial cells. Colorectal cancer (CRC) is a common and deadly disease that accounts for the 4th worldwide highly variable malignancy. For the early detection of CRC, the most common predictive biomarker found endogenously are KRAS and ctDNA/cfDNA along with SEPT9 methylated DNA. Early detection and screening for CRC are necessary and multiple methods can be employed to screen and perform early diagnosis of CRC. Colonoscopy, an invasive method is most prevalent for diagnosing CRC or confirming the positive result as compared to other screening methods whereas several non-invasive techniques such as molecular analysis of breath, urine, blood, and stool can also be performed for early detection. Interestingly, widely used medicines known as non-steroidal anti-inflammatory drugs (NSAIDs) to reduce pain and inflammation have reported chemopreventive impact on gastrointestinal malignancies, especially CRC in several epidemiological and preclinical types of research. NSAID acts by inhibiting two cyclooxygenase enzymes, thereby preventing the synthesis of prostaglandins (PGs) and causing NSAID-induced apoptosis and growth inhibition in CRC cells. This review paper majorly focuses on the diversity of natural and synthetic biomarkers and various techniques for the early detection of CRC. An approach toward current advancement in CRC detection techniques and the role of NSAIDs in CRC chemoprevention has been explored systematically. Several prominent governing mechanisms of the anti-cancer effects of NSAIDs and their synergistic effect with statins for an effective chemopreventive measure have also been discussed in this review paper.
Collapse
Affiliation(s)
- Gowhar Rashid
- Department of Amity Medical School, Amity University, Gurugram, India
| | - Nihad Ashraf Khan
- Department of Biosciences, Jamia Millia Islamia, Central University, New Delhi, India
| | - Deena Elsori
- Faculty of Resillience, Deans Office Rabdan Academy, Abu Dhabi, United Arab Emirates
| | - Andleeb Rehman
- Department of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| | - Tanzeelah
- Department of Biochemistry, University of Kashmir, Srinagar, India
| | - Haleema Ahmad
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh, India
| | - Humaira Maryam
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh, India
| | - Amaan Rais
- Department of Biochemistry, Faculty of Life Sciences, AMU, Aligarh, India
| | - Mohd Salik Usmani
- The Department of Surgery, Faculty of Medicine, JNMCH, AMU, Uttar Pradesh, India
| | - Asaad Ma Babker
- Department of Medical Laboratory Sciences, Gulf Medical University, Ajman, United Arab Emirates
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Wael Hafez
- Department of Internal Medicine, NMC Royal Hospital, Abu Dhabi, United Arab Emirates
- The Medical Research Division, Department of Internal Medicine, The National Research Center, Ad Doqi, Egypt
| |
Collapse
|
39
|
Mahboubi-Rabbani M, Abbasi M, Zarghi A. Natural-Derived COX-2 Inhibitors as Anticancer Drugs: A Review of their Structural Diversity and Mechanism of Action. Anticancer Agents Med Chem 2023; 23:15-36. [PMID: 35638275 DOI: 10.2174/1389450123666220516153915] [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: 01/12/2022] [Revised: 02/07/2022] [Accepted: 03/01/2022] [Indexed: 02/08/2023]
Abstract
Cyclooxygenase-2 (COX-2) is a key-type enzyme playing a crucial role in cancer development, making it a target of high interest for drug designers. In the last two decades, numerous selective COX-2 inhibitors have been approved for various clinical conditions. However, data from clinical trials propose that the prolonged use of COX-2 inhibitors is associated with life-threatening cardiovascular side effects. The data indicate that a slight structural modification can help develop COX-2 selective inhibitors with comparative efficacy and limited side effects. In this regard, secondary metabolites from natural sources offer great hope for developing novel COX-2 inhibitors with potential anticancer activity. In recent years, various nature-derived organic scaffolds are being explored as leads for developing new COX-2 inhibitors. The current review attempts to highlight the COX-2 inhibition activity of some naturally occurring secondary metabolites, concerning their capacity to inhibit COX-1 and COX-2 enzymes and inhibit cancer development, aiming to establish a structure-activity relationship.
Collapse
Affiliation(s)
- Mohammad Mahboubi-Rabbani
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Abbasi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afshin Zarghi
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
Kuroe T, Watanabe R, Morisue R, Miyazaki S, Kojima M, Murata SC, Nakai T, Taki T, Sakashita S, Sakamoto N, Matsubara N, Masuda H, Ushiku T, Ishii G. Dirty necrosis in renal cell carcinoma is associated with NETosis and systemic inflammation. Cancer Med 2023; 12:4557-4567. [PMID: 36127822 PMCID: PMC9972113 DOI: 10.1002/cam4.5249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 08/24/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022] Open
Abstract
AIM Dirty necrosis (DN) in renal cell carcinoma (RCC) is morphologically characterized by abundant neutrophil infiltration and has significant potential as an unfavorable prognostic indicator. This study aimed to analyze the pathological and biological features of DN. MATERIALS AND METHODS A total of 81 RCC tumors, including 33 cases of DN and 48 cases of tumor necrosis without DN features (ghost necrosis [GN]), were enrolled in this study. We compared the number of neutrophils; the activation of cell death pathways, including ferroptosis, NETosis, and apoptosis; the rate of epithelial-mesenchymal transition (EMT); and proliferation status using immunohistochemistry. We further assessed the effect of the necrosis type on systemic inflammation. RESULTS DN tumors had a significantly higher number of neutrophils in both areas around the necrotic foci and far from the necrotic foci. Ferroptosis status did not differ between DN and GN; however, DN tumors had significantly larger areas exhibiting cell detritus with neutrophil extracellular traps (NETs) detected by citrullinated histone H3 (citH3) than GN tumors. DN tumors also had more apoptotic cells within areas around the necrotic foci. There was no significant difference between the EMT and proliferation status between DN and GN groups. Systemic inflammation markers including C-reactive protein (CRP), CRP-to-albumin ratio (CRP/Alb), platelet-to-lymphocyte ratio (PLR), and hemoglobin were significantly higher in patients with DN. In addition, some of these inflammation markers (CRP/Alb and PLR) significantly decreased after surgery. CONCLUSIONS DN in RCC is characterized by NETs production and systemic inflammation.
Collapse
Affiliation(s)
- Takashi Kuroe
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.,Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Reiko Watanabe
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Ryo Morisue
- Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan.,Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Saori Miyazaki
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Motohiro Kojima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.,Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Shawhay Charles Murata
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.,Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Tokiko Nakai
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Tetsuro Taki
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Shingo Sakashita
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.,Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Naoya Sakamoto
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.,Division of Pathology, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Nobuaki Matsubara
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Hitoshi Masuda
- Department of Urology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Genichiro Ishii
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.,Division of Innovative Pathology and Laboratory Medicine, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| |
Collapse
|
41
|
Cicek B, Hacimuftuoglu A, Kuzucu M, Cetin A, Yeni Y, Genc S, Yildirim S, Bolat I, Kantarci M, Gul M, Hayme S, Matthaios D, Vageli DP, Doukas SG, Tsatsakis A, Taghizadehghalehjoughi A. Sorafenib Alleviates Inflammatory Signaling of Tumor Microenvironment in Precancerous Lung Injuries. Pharmaceuticals (Basel) 2023; 16:221. [PMID: 37259369 PMCID: PMC9963576 DOI: 10.3390/ph16020221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 09/16/2024] Open
Abstract
According to population-based studies, lung cancer is the prominent reason for cancer-related mortality worldwide in males and is also rising in females at an alarming rate. Sorafenib (SOR), which is approved for the treatment of hepatocellular carcinoma and renal cell carcinoma, is a multitargeted protein kinase inhibitor. Additionally, SOR is the subject of interest for preclinical and clinical trials in lung cancer. This study was designed to assess in vivo the possible effects of sorafenib (SOR) in diethylnitrosamine (DEN)-induced lung carcinogenesis and examine its probable mechanisms of action. A total of 30 adult male rats were divided into three groups (1) control, (2) DEN, and (3) DEN + SOR. The chemical induction of lung carcinogenesis was performed by injection of DEN intraperitoneally at 150 mg/kg once a week for two weeks. The DEN-administered rats were co-treated with SOR of 10 mg/kg by oral gavage for 42 alternate days. Serum and lung tissue samples were analyzed to determine SRY-box transcription factor 2 (SOX-2) levels. The tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) levels were measured in lung tissue supernatants. Lung sections were analyzed for cyclooxygenase-2 (COX-2) and c-Jun N-terminal kinase (JNK) histopathologically. In addition, cyclooxygenase-2 (COX-2) and c-Jun N-terminal kinase (JNK) were analyzed by immunohistochemistry and immunofluorescence methods, respectively. SOR reduced the level of SOX-2 that maintenance of cancer stemness and tumorigenicity, and TNF-α and IL-1β levels. Histopathological analysis demonstrated widespread inflammatory cell infiltration, disorganized alveolar structure, hyperemia in the vessels, and thickened alveolar walls in DEN-induced rats. The damage was markedly reduced upon SOR treatment. Further, immunohistochemical and immunofluorescence analysis also revealed increased expression of COX-2 and JNK expression in DEN-intoxicated rats. However, SOR treatment alleviated the expression of these inflammatory markers in DEN-induced lung carcinogenesis. These findings suggested that SOR inhibits DEN-induced lung precancerous lesions through decreased inflammation with concomitant in reduced SOX-2 levels, which enables the maintenance of cancer stem cell properties.
Collapse
Affiliation(s)
- Betul Cicek
- Faculty of Medicine, Department of Physiology, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
| | - Ahmet Hacimuftuoglu
- Faculty of Medicine, Department of Medical Pharmacology, Ataturk University, Erzurum 25240, Turkey
| | - Mehmet Kuzucu
- Faculty of Arts and Sciences, Department of Biology, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
| | - Ahmet Cetin
- Department of Biology, Graduate School of Natural and Applied Sciences, Erzincan Binali Yildirim University, 24100 Erzincan, Turkey
| | - Yesim Yeni
- Faculty of Medicine, Department of Medical Pharmacology, Malatya Turgut Ozal University, Malatya 44210, Turkey
| | - Sidika Genc
- Faculty of Medicine, Department of Medical Pharmacology, Bilecik Seyh Edebali University, Bilecik 11230, Turkey
| | - Serkan Yildirim
- Faculty of Veterinary, Department of Pathology, Ataturk University, Erzurum 25240, Turkey
| | - Ismail Bolat
- Faculty of Veterinary, Department of Pathology, Ataturk University, Erzurum 25240, Turkey
| | - Mecit Kantarci
- Faculty of Medicine, Department of Radiology, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
- Faculty of Medicine, Department of Radiology, Ataturk University, Erzurum 25240, Turkey
| | - Mustafa Gul
- Faculty of Medicine, Department of Physiology, Ataturk University, Erzurum 25240, Turkey
| | - Serhat Hayme
- Faculty of Medicine, Department of Biostatistics, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
| | | | - Dimitra P. Vageli
- Yale Larynx Laboratory, Department of Surgery (Otololaryngology), Yale School of Medicine, Yale University, New Havan, CT 06510, USA
| | - Sotirios G. Doukas
- Department of Internal Medicine, Division of Gastroenterology, Rutgers/Saint Peter’s University Hospital, New Brunswick, NJ 08901, USA
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Ali Taghizadehghalehjoughi
- Faculty of Medicine, Department of Medical Pharmacology, Bilecik Seyh Edebali University, Bilecik 11230, Turkey
| |
Collapse
|
42
|
Solid-Phase Parallel Synthesis of Dual Histone Deacetylase-Cyclooxygenase Inhibitors. Molecules 2023; 28:molecules28031061. [PMID: 36770730 PMCID: PMC9920637 DOI: 10.3390/molecules28031061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/10/2023] [Accepted: 01/15/2023] [Indexed: 01/21/2023] Open
Abstract
Multi-target drugs (MTDs) are emerging alternatives to combination therapies. Since both histone deacetylases (HDACs) and cyclooxygenase-2 (COX-2) are known to be overexpressed in several cancer types, we herein report the design, synthesis, and biological evaluation of a library of dual HDAC-COX inhibitors. The designed compounds were synthesized via an efficient parallel synthesis approach using preloaded solid-phase resins. Biological in vitro assays demonstrated that several of the synthesized compounds possess pronounced inhibitory activities against HDAC and COX isoforms. The membrane permeability and inhibition of cellular HDAC activity of selected compounds were confirmed by whole-cell HDAC inhibition assays and immunoblot experiments. The most promising dual inhibitors, C3 and C4, evoked antiproliferative effects in the low micromolar concentration range and caused a significant increase in apoptotic cells. In contrast to previous reports, the simultaneous inhibition of HDAC and COX activity by dual HDAC-COX inhibitors or combination treatments with vorinostat and celecoxib did not result in additive or synergistic anticancer activities.
Collapse
|
43
|
Johnson A, Olelewe C, Kim JH, Northcote-Smith J, Mertens RT, Passeri G, Singh K, Awuah SG, Suntharalingam K. The anti-breast cancer stem cell properties of gold(i)-non-steroidal anti-inflammatory drug complexes. Chem Sci 2023; 14:557-565. [PMID: 36741517 PMCID: PMC9847679 DOI: 10.1039/d2sc04707a] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/10/2022] [Indexed: 12/14/2022] Open
Abstract
The anti-breast cancer stem cell (CSC) properties of a series of gold(i) complexes comprising various non-steroidal anti-inflammatory drugs (NSAIDs) and triphenylphosphine 1-8 are reported. The most effective gold(i)-NSAID complex 1, containing indomethacin, exhibits greater potency for breast CSCs than bulk breast cancer cells (up to 80-fold). Furthermore, 1 reduces mammosphere viability to a better extent than a panel of clinically used breast cancer drugs and salinomycin, an established anti-breast CSC agent. Mechanistic studies suggest 1-induced breast CSC death results from breast CSC entry, cytoplasm localisation, an increase in intracellular reactive oxygen species levels, cyclooxygenase-2 downregulation and inhibition, and apoptosis. Remarkably, 1 also significantly inhibits tumour growth in a murine metastatic triple-negative breast cancer model. To the best of our knowledge, 1 is the first gold complex of any geometry or oxidation state to demonstrate anti-breast CSC properties.
Collapse
Affiliation(s)
- Alice Johnson
- School of Chemistry, University of LeicesterLeicesterUK,Biomolecular Sciences Research Centre, Sheffield Hallam UniversitySheffieldUK
| | - Chibuzor Olelewe
- Department of Chemistry, University of KentuckyLexingtonKentuckyUSA
| | - Jong Hyun Kim
- Department of Chemistry, University of KentuckyLexingtonKentuckyUSA
| | | | - R. Tyler Mertens
- Department of Chemistry, University of KentuckyLexingtonKentuckyUSA
| | | | - Kuldip Singh
- School of Chemistry, University of LeicesterLeicesterUK
| | - Samuel G. Awuah
- Department of Chemistry, University of KentuckyLexingtonKentuckyUSA,Department of Pharmaceutical Sciences, University of KentuckyLexingtonKentuckyUSA
| | | |
Collapse
|
44
|
Mirjafary Z, Mohammad Karbasi M, Hesamzadeh P, Shaker HR, Amiri A, Saeidian H. Novel 1,2,3-Triazole-Based Benzothiazole Derivatives: Efficient Synthesis, DFT, Molecular Docking, and ADMET Studies. Molecules 2022; 27:8555. [PMID: 36500647 PMCID: PMC9740823 DOI: 10.3390/molecules27238555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
A new series of 1,2,3-triazole derivatives 5a-f based on benzothiazole were synthesized by the 1,3-dipolar cycloaddition reaction of S-propargyl mercaptobenzothiazole and α-halo ester/amide in moderate to good yields (47-75%). The structure of all products was characterized by 1H NMR, 13C NMR, and CHN elemental data. This protocol is easy and green and proceeds under mild and green reaction conditions with available starting materials. The structural and electronic analysis and 1H and 13C chemical shifts of the characterized structure of 5e were also calculated by applying the B3LYP/6-31 + G(d, p) level of density functional theory (DFT) method. In the final section, all the synthesized compounds were evaluated for their anti-inflammatory activity by biochemical COX-2 inhibition, antifungal inhibition with CYP51, anti-tuberculosis target protein ENR, DPRE1, pks13, and Thymidylate kinase by molecular docking studies. The ADMET analysis of the molecules 5a-f revealed that 5d and 5a are the most-promising drug-like molecules out of the six synthesized molecules.
Collapse
Affiliation(s)
- Zohreh Mirjafary
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran 14515-775, Iran
| | - Mahdieh Mohammad Karbasi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran 14515-775, Iran
| | - Parsa Hesamzadeh
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran 14515-775, Iran
| | - Hamid Reza Shaker
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran 14515-775, Iran
| | - Asghar Amiri
- Department of Science, Payame Noor University (PNU), Tehran P.O. Box 19395-4697, Iran
| | - Hamid Saeidian
- Department of Science, Payame Noor University (PNU), Tehran P.O. Box 19395-4697, Iran
| |
Collapse
|
45
|
Mohsin NUA, Aslam S, Ahmad M, Irfan M, Al-Hussain SA, Zaki MEA. Cyclooxygenase-2 (COX-2) as a Target of Anticancer Agents: A Review of Novel Synthesized Scaffolds Having Anticancer and COX-2 Inhibitory Potentialities. Pharmaceuticals (Basel) 2022; 15:ph15121471. [PMID: 36558921 PMCID: PMC9783503 DOI: 10.3390/ph15121471] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/19/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a serious threat to human beings and is the second-largest cause of death all over the globe. Chemotherapy is one of the most common treatments for cancer; however, drug resistance and severe adverse effects are major problems associated with anticancer therapy. New compounds with multi-target inhibitory properties are targeted to surmount these challenges. Cyclooxygenase-2 (COX-2) is overexpressed in cancers of the pancreas, breast, colorectal, stomach, and lung carcinoma. Therefore, COX-2 is considered a significant target for the synthesis of new anticancer agents. This review discusses the biological activity of recently prepared dual anticancer and COX-2 inhibitory agents. The most important intermolecular interactions with the COX-2 enzyme have also been presented. Analysis of these agents in the active area of the COX-2 enzyme could guide the introduction of new lead compounds with extreme selectivity and minor side effects.
Collapse
Affiliation(s)
- Noor ul Amin Mohsin
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sana Aslam
- Department of Chemistry, Government College Women University, Faisalabad 38000, Pakistan
| | - Matloob Ahmad
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
- Correspondence: (M.A.); (M.E.A.Z.)
| | - Muhammad Irfan
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
- Correspondence: (M.A.); (M.E.A.Z.)
| |
Collapse
|
46
|
The Molecular and Cellular Strategies of Glioblastoma and Non-Small-Cell Lung Cancer Cells Conferring Radioresistance. Int J Mol Sci 2022; 23:ijms232113577. [PMID: 36362359 PMCID: PMC9656305 DOI: 10.3390/ijms232113577] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Ionizing radiation (IR) has been shown to play a crucial role in the treatment of glioblastoma (GBM; grade IV) and non-small-cell lung cancer (NSCLC). Nevertheless, recent studies have indicated that radiotherapy can offer only palliation owing to the radioresistance of GBM and NSCLC. Therefore, delineating the major radioresistance mechanisms may provide novel therapeutic approaches to sensitize these diseases to IR and improve patient outcomes. This review provides insights into the molecular and cellular mechanisms underlying GBM and NSCLC radioresistance, where it sheds light on the role played by cancer stem cells (CSCs), as well as discusses comprehensively how the cellular dormancy/non-proliferating state and polyploidy impact on their survival and relapse post-IR exposure.
Collapse
|
47
|
Ho CH, Chen ML, Huang HL, Lai CJ, Liu CH, Chuu CP, Lin YH. Active Targeting of P-Selectin by Fucoidan Modulates the Molecular Profiling of Metastasis in Docetaxel-Resistant Prostate Cancer. Mar Drugs 2022; 20:md20090542. [PMID: 36135731 PMCID: PMC9500773 DOI: 10.3390/md20090542] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 12/17/2022] Open
Abstract
The standard of care for prostate cancer (PCa) is androgen deprivation therapy (ADT). Although hormone-sensitive PCa is curable by ADT, most conditions progress to castration-resistant prostate cancer (CRPCa) and metastatic CRPCa (mCRPCa). Front-line docetaxel has been administered to patients with CRPCa and mCRPCa. Nevertheless, docetaxel resistance after half a year of therapy has emerged as an urgent clinical concern in patients with CRPCa and mCRPCa. We verified the mechanism by which docetaxel-resistant PCa cells (DU/DX50) exhibited significant cell migration and expression of malignant tumor-related proteins. Our study shows that the biological activity of fucoidan has an important application for docetaxel-resistant PCa cells, inhibiting IL-1R by binding to P-selectin and reducing the expression levels of NF-κB p50 and Cox2 in this metastasis-inhibiting signaling pathway. Furthermore, the combined treatment of fucoidan and docetaxel showed significant anticancer and synergistic effects on the viability of DU/DX50 cells, which is relevant for overcoming the current limitations and improving treatment outcomes. Overall, fucoidan-based combination chemotherapy may exert beneficial effects and facilitate the treatment of docetaxel-resistant PCa.
Collapse
Affiliation(s)
- Chang-Hsun Ho
- Department of Anesthesiology, Show Chwan Memorial Hospital, Changhua 50008, Taiwan
| | - Mei-Lin Chen
- Department of Pharmacy, Cheng Hsin General Hospital, Taipei 11220, Taiwan
| | - Hau-Lun Huang
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chih-Jen Lai
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chih-Hsin Liu
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Chih-Pin Chuu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Yu-Hsin Lin
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Medical Device Innovation and Translation Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Correspondence: ; Tel.: +886-2-28267000 (ext. 7932)
| |
Collapse
|
48
|
Design, synthesis and mechanistic studies of novel imidazo[1,2-a]pyridines as anticancer agents. Bioorg Chem 2022; 128:106042. [PMID: 35878430 DOI: 10.1016/j.bioorg.2022.106042] [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: 03/20/2022] [Revised: 05/16/2022] [Accepted: 07/17/2022] [Indexed: 11/20/2022]
Abstract
Herein, the design, synthesis and mechanistic study of five series of imidazo[1,2-a]pyridines 8a-d, 9a-f, 11a-c, 12a-d and 14a-d as anticancer agents were discussed. The cytotoxicity of imidazo[1,2-a]pyridine derivatives was screened against NCI 60 cancer cell lines. The cytotoxicity of compounds 8b, 8c, 9e and 9f was then evaluated against leukemia K-562 cancer cell line and normal lung fibroblasts (WI38). The hydrazone derivatives 8b and 8c exhibited significant cytotoxic activities against the leukemia K-562 cancer cell line with good safety margins (IC50 = 2.91 µM, SI = 8.32 and IC50 = 1.09 µM, SI = 10.54, respectively). In addition, compounds 8b, 8c, 9e and 9f were tested for their EGFR and COX-2 inhibitory activities. The hydrazone derivatives 8b and 8c were the most active EGFR inhibitors with IC50 values of 0.123 and 0.072 µM, respectively. Compound 8c selectively inhibited COX-2 (IC50 = 1.09 µM, SI = 13.78). Moreover, the potential of compound 8c to induce apoptosis in leukemia K-562 cell line was determined. Compound 8c showed a pre-G1 apoptosis and a growth arrest of leukemia K-562 cell line at G1 phase of cell cycle. Also, compound 8c was able to induce caspase-3 overexpression (6.98 folds), if compared to control. Finally, molecular docking studies and physicochemical properties calculation of compounds 8b, 8c, 9e and 9f were carried out to explain the biological data and to predict bioavailability of the most active compounds.
Collapse
|
49
|
Moslehi M, Moazamiyanfar R, Dakkali MS, Rezaei S, Rastegar-Pouyani N, Jafarzadeh E, Mouludi K, Khodamoradi E, Taeb S, Najafi M. Modulation of the immune system by melatonin; implications for cancer therapy. Int Immunopharmacol 2022; 108:108890. [PMID: 35623297 DOI: 10.1016/j.intimp.2022.108890] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/07/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022]
Abstract
Immune system interactions within the tumour have a key role in the resistance or sensitization of cancer cells to anti-cancer agents. On the other hand, activation of the immune system in normal tissues following chemotherapy or radiotherapy is associated with acute and late effects such as inflammation and fibrosis. Some immune responses can reduce the efficiency of anti-cancer therapy and also promote normal tissue toxicity. Modulation of immune responses can boost the efficiency of anti-tumour therapy and alleviate normal tissue toxicity. Melatonin is a natural body agent that has shown promising results for modulating tumour response to therapy and also alleviating normal tissue toxicity. This review tries to focus on the immunomodulatory actions of melatonin in both tumour and normal tissues. We will explain how anti-cancer drugs may cause toxicity for normal tissues and how tumours can adapt themselves to ionizing radiation and anti-cancer drugs. Then, cellular and molecular mechanisms of immunoregulatory effects of melatonin alone or combined with other anti-cancer agents will be discussed.
Collapse
Affiliation(s)
- Masoud Moslehi
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Moazamiyanfar
- Department of Medical Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Sepideh Rezaei
- Department of Chemistry, University of Houston, 3585 Cullen Blvd., Fleming Bldg. Rm 112, Houston, TX 77204-5003, USA
| | - Nima Rastegar-Pouyani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Emad Jafarzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Kave Mouludi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ehsan Khodamoradi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran; Medical Biotechnology Research Center, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran; Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
50
|
Antioxidant and cytotoxic activities of selected salicylidene imines: experimental and computational study. Mol Divers 2022; 26:3115-3128. [PMID: 35147861 DOI: 10.1007/s11030-021-10370-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 12/15/2021] [Indexed: 10/19/2022]
Abstract
Selected salicylidene imines were evaluated for their antioxidant and cytotoxic potentials. Several of them exerted potent scavenging capacity towards ABTS radical and hydrogen peroxide. The insight into the preferable antioxidative mechanism was reached employing density functional theory. In the absence of free radicals, the SPLET mechanism is dominant in polar surroundings, while HAT is prevailing in a non-polar environment. The results obtained for the reactions of the most active compounds with some medically relevant radicals pointed out competition between HAT and SPLET mechanisms. The assessment of their cytotoxic properties revealed inhibition of ER-a human breast adenocarcinoma cells or estrogen-independent prostate cancer cells. Molecular docking study with the cyclooxygenase (COX) 2 enzyme was performed to examine the most probable bioactive conformations and possible interactions between the tested derivatives and COX-2 binding pocket.
Collapse
|