1
|
Huang R, Yu J, Zhang B, Li X, Liu H, Wang Y. Emerging COX-2 inhibitors-based nanotherapeutics for cancer diagnosis and treatment. Biomaterials 2025; 315:122954. [PMID: 39549439 DOI: 10.1016/j.biomaterials.2024.122954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/27/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
Increasing evidence has showed that tumorigenesis is closely linked to inflammation, regulated by multiple signaling pathways. Among these, the cyclooxygenase-2/prostaglandin E2 (COX-2/PGE2) axis plays a crucial role in the progression of both inflammation and cancer. Inhibiting the activity of COX-2 can reduce PGE2 secretion, thereby suppressing tumor growth. Therefore, COX-2 inhibitors are considered potential therapeutic agents for cancers. However, their clinical applications are greatly hindered by poor physicochemical properties and serious adverse effects. Fortunately, the advent of nanotechnology offers solutions to these limitations, enhancing drug delivery efficiency and mitigating adverse effects. Given the considerable progress in this area, it is timely to review emerging COX-2 inhibitors-based nanotherapeutics for cancer diagnosis and therapy. In this review, we first outline the various antineoplastic mechanisms of COX-2 inhibitors, then comprehensively summarize COX-2 inhibitors-based nanotherapeutics for cancer monotherapy, combination therapy, and diagnosis. Finally, we highlight and discuss future perspectives and challenges in the development of COX-2 inhibitors-based nanomedicine.
Collapse
Affiliation(s)
- Ruiping Huang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Baoyue Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Xin Li
- Department of Respiratory Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121001, PR China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China.
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China.
| |
Collapse
|
2
|
Qin X, Wang X, Yang C, Wang F, Fang T, Gu D, Guo Q, Meng Q, Liu W, Yang L. A potent dual inhibitor targeting COX-2 and HDAC of acute myeloid leukemia cells. Mol Divers 2024:10.1007/s11030-024-11000-w. [PMID: 39480610 DOI: 10.1007/s11030-024-11000-w] [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: 07/18/2024] [Accepted: 09/24/2024] [Indexed: 11/02/2024]
Abstract
Acute myeloid leukemia (AML) is an aggressive cancer with complex issues of drug resistance and a poor prognosis; thus, effective therapeutics is urgently needed for AML. In this study, we designed and synthesized dual cyclooxygenase-2 (COX-2) and histone deacetylase (HDAC) inhibitors, IMC-HA and IMC-OPD, and applied them for the treatment of AML. IMC-HA comprised a COX-2 inhibitor skeleton of indomethacin (IMC) and an HDAC inhibitor moiety of the hydroxamic group and was found to exhibit potent antiproliferative activity against AML cells (THP-1 and U937) and low cytotoxicity toward normal cells. Molecular docking simulations suggested that IMC-HA had a high binding affinity for HDAC and COX-2, with binding energies of -6.8 and -9.0 kcal/mol, respectively. Mechanistic studies revealed that IMC-HA induced apoptosis and G0/G1 phase arrest in AML cells, which were characterized by alterations in the expression of apoptotic and cell cycle-related proteins. Further study demonstrated that IMC-HA also inhibited the MEK/ERK signaling pathway in AML cells. Overall, we believe that IMC-HA could serve as a potent COX-2/HDAC dual inhibitor and improve the treatment of AML.
Collapse
Affiliation(s)
- Xiang Qin
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xueting Wang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Chunmei Yang
- Department of Radiology, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Fan Wang
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Tingting Fang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Didi Gu
- Department of Radiology, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qulian Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qiuyu Meng
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Interdisciplinary Research Academy (IRA), Zhejiang Shuren University, Hangzhou, 310015, China.
| | - Wenjun Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Lu Yang
- Department of Radiology, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
Saleem SM, Jabbar T, Imran MB, Noureen A, Sherazi TA, Afzal MS, Rab Nawaz HZ, Ramadan MF, Alkahtani AM, Alsuwat MA, Almubarak HA, Momenah MA, Naqvi SAR. Radiosynthesis and Preclinical Evaluation of [ 99mTc]Tc-Tigecycline Radiopharmaceutical to Diagnose Bacterial Infections. Pharmaceuticals (Basel) 2024; 17:1283. [PMID: 39458924 PMCID: PMC11510260 DOI: 10.3390/ph17101283] [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: 08/23/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES As a primary source of mortality and disability, bacterial infections continue to develop a severe threat to humanity. Nuclear medicine imaging (NMI) is known for its promising potential to diagnose deep-seated bacterial infections. This work aims to develop a new technetium-99m (99mTc) labeled tigecycline radiopharmaceutical as an infection imaging agent. METHODS Reduced 99mTc was used to make a coordinate complex with tigecycline at pH 7.7-7.9 at room temperature. Instantaneous thin-layer chromatography impregnated with silica gel (ITLC-SG) and ray detector equipped high-performance liquid chromatography (ray-HPLC) was performed to access the radiolabeling yield and radiochemical purity (RCP). RESULTS More than 91% labeling efficiency was achieved after 25 min of mild shaking of the reaction mixture. The radiolabeled complex was found intact up to 4 h in saline. Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli) infection-induced rats were used to record the biodistribution of the radiopharmaceutical and its target specificity; 2 h' post-injection biodistribution revealed a 2.39 ± 0.29 target/non-target (T/NT) ratio in the E. coli infection-induced animal model, while a 2.9 ± 0.31 T/NT value was recorded in the S. aureus bacterial infection-induced animal model. [99mTc]Tc-tigecycline scintigraphy was performed in healthy rabbits using a single photon emission computed tomography (SPECT) camera. Scintigrams showed normal kidney perfusion and excretion into the bladder. CONCLUSION In conclusion, the newly developed [99mTc]Tc-tigecycline radiopharmaceutical could be considered to diagnose broad-spectrum bacterial infections.
Collapse
Affiliation(s)
- Syeda Marab Saleem
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38040, Pakistan
| | - Tania Jabbar
- Punjab Institute of Nuclear Medicine, Faisalabad 38040, Pakistan
| | | | - Asma Noureen
- Department of Zoology, Ghazi University, Dera Ghazi Khan 03222, Pakistan
| | - Tauqir A. Sherazi
- Department of Chemistry, COMSAT University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | | | - Hafiza Zahra Rab Nawaz
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38040, Pakistan
| | - Mohamed Fawzy Ramadan
- Department of Clinical Nutrition, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Abdullah M. Alkahtani
- Department of Microbiology & Clinical Parasitology, College of Medicine, King Khalid University, Abha 61421, Saudi Arabia
| | - Meshari A. Alsuwat
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif 21944, Saudi Arabia
| | - Hassan Ali Almubarak
- Assistant Professor Nuclear Medicine, Division of Radiology, Department of Medicine, College of Medicine and Surgery, King Khalid University, Abha 61421, Saudi Arabia
| | - Maha Abdullah Momenah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Syed Ali Raza Naqvi
- Department of Chemistry, Government College University Faisalabad, Faisalabad 38040, Pakistan
| |
Collapse
|
4
|
Begum SY, Imran PM, Kubaib A, Yassin MT, Al-Otibi FO, Selvakumaran M, Basha AA, Sulthanudeen S. Unveiling multifunctional inhibitors: holistic spectral, electronic and molecular characterization, coupled with biological profiling of substituted pyridine derivatives against LD transpeptidase, heme oxygenase and PPAR gamma. RSC Adv 2024; 14:29896-29909. [PMID: 39301237 PMCID: PMC11411632 DOI: 10.1039/d4ra04217d] [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: 06/08/2024] [Accepted: 08/14/2024] [Indexed: 09/22/2024] Open
Abstract
This study delves into the therapeutic potential of a molecule, 3-substituted phenyl-1-(pyridine-4-carbonyl)-1H-pyrazole-4-carboxylic acid (PPP), for antimicrobial, antioxidant and anti-diabetic activities. The research encompasses design, synthesis, molecular docking and biological screening of related pyrazole carboxylic acid derivatives. Spectral studies confirmed the structures and molecular mechanics with DFT calculations provided insights into molecular properties and interactions. Quantum chemical descriptors were employed to assess the stability while NBO analysis predicted reactivity, ELF and LOL methods identified electron density. Non-covalent interactions were characterized using RDG and IRI, while the Multiwfn tool was used to evaluate intra and intermolecular aspects. Docking studies elucidated potential therapeutic efficacy against specific protein targets.
Collapse
Affiliation(s)
- Shaik Yasmin Begum
- Department of Chemistry, Islamiah College (Autonomous) Vaniyambadi 635752 Tamilnadu India
- Thiruvalluvar University Serkkadu Vellore 632 115 Tamilnadu India
| | - Predhanekar Mohamed Imran
- Department of Chemistry, Islamiah College (Autonomous) Vaniyambadi 635752 Tamilnadu India
- Thiruvalluvar University Serkkadu Vellore 632 115 Tamilnadu India
| | - Attar Kubaib
- Department of Chemistry, Islamiah College (Autonomous) Vaniyambadi 635752 Tamilnadu India
- Thiruvalluvar University Serkkadu Vellore 632 115 Tamilnadu India
| | - Mohamed Taha Yassin
- Department of Botany and Microbiology, College of Science, King Saud University P. O. Box 2455 Riyadh 11451 Saudi Arabia
| | - Fatimah O Al-Otibi
- Department of Botany and Microbiology, College of Science, King Saud University P. O. Box 2455 Riyadh 11451 Saudi Arabia
| | - M Selvakumaran
- Department of Chemistry, Islamiah College (Autonomous) Vaniyambadi 635752 Tamilnadu India
- Thiruvalluvar University Serkkadu Vellore 632 115 Tamilnadu India
| | - A Aathif Basha
- Department of Physics, Islamiah College (Autonomous) Vaniyambadi - 635752 Tamilnadu India
- Thiruvalluvar University Serkkadu Vellore 632 115 Tamilnadu India
| | - S Sulthanudeen
- Department of Chemistry, Islamiah College (Autonomous) Vaniyambadi 635752 Tamilnadu India
- Thiruvalluvar University Serkkadu Vellore 632 115 Tamilnadu India
| |
Collapse
|
5
|
Pimenta J, Prada J, Pires I, Cotovio M. Cyclooxygenase-2 (COX-2) Expression in Equine Melanocytic Tumors. Vet Sci 2024; 11:77. [PMID: 38393095 PMCID: PMC10891553 DOI: 10.3390/vetsci11020077] [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: 01/08/2024] [Revised: 01/27/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Equine melanocytic tumors are common and have an unusual benign behavior with low invasiveness and metastatic rates. However, tumoral mass growth is usually a concern that can have life-threatening consequences. COX-2 is related to oncogenesis, promoting neoplastic cell proliferation, invasion, and metastasis. The aim of this study was to evaluate the immunohistochemical expression of COX-2 in equine melanocytic tumors. Through extension and intensity of labeling, 39 melanocytomas and 38 melanomas were evaluated. Of the malignant tumors, 13.2% were negative and 63.2% presented a low COX-2 expression. Only 6 malignant tumors presented >50% of labeled cells, 18 malignant and 8 benign had an expression between 21 and 50%, 8 malignant and 3 benign tumors had an expression between 6 and 20%, 1 malignant tumor had an expression between 1 and 5%, and 5 malignant and 28 benign tumors had no expression. Malignant tumors showed higher COX-2 expression than did benign tumors, with statistically significant differences. The low levels of COX-2 may be one of the molecular reasons for the presence of expansive mass growth instead of the invasive pattern of other species, which is related to high COX-2 levels.
Collapse
Affiliation(s)
- José Pimenta
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- CIVG—Vasco da Gama Research Center, EUVG—Vasco da Gama University School, 3020-210 Coimbra, Portugal
| | - Justina Prada
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Isabel Pires
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Mário Cotovio
- CECAV—Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; (J.P.); (I.P.); (M.C.)
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 5000-801 Vila Real, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Campo Grande 376, 1749-024 Lisbon, Portugal
| |
Collapse
|
6
|
Karthikeyan S, Thirunarayanan A, Shano LB, Hemamalini A, Sundaramoorthy A, Mangaiyarkarasi R, Abu N, Ganesan S, Chinnathambi S, Pandian GN. Chalcone derivatives' interaction with human serum albumin and cyclooxygenase-2. RSC Adv 2024; 14:2835-2849. [PMID: 38234869 PMCID: PMC10792617 DOI: 10.1039/d3ra07438b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/30/2023] [Indexed: 01/19/2024] Open
Abstract
Chalcone derivatives are an extremely valuable class of compounds, primarily due to the keto-ethylenic group, CO-CH[double bond, length as m-dash]CH-, they contain. Moreover, the presence of a reactive α,β-unsaturated carbonyl group confers upon them a broad range of pharmacological properties. Recent developments in heterocyclic chemistry have led to the synthesis of chalcone derivatives, which have been biologically investigated for their activity against certain diseases. In this study, we investigated the binding of new chalcone derivatives with COX-2 (cyclooxygenase-2) and HSA (Human Serum Albumin) using spectroscopic and molecular modeling studies. COX-2 is commonly found in cancer and plays a role in the production of prostaglandin E (2), which can help tumors grow by binding to receptors. HSA is the most abundant protein in blood plasma, and it transports various compounds, including hormones and fatty acids. The conformation of chalcone derivatives in the HSA complex system was established through fluorescence steady and excited state spectroscopy techniques and FTIR analyses. To gain a more comprehensive understanding, molecular docking, and dynamics were conducted on the target protein (COX-2) and transport protein (HSA). In addition, we conducted density-functional theory (DFT) and single-point DFT to understand intermolecular interaction in protein active sites.
Collapse
Affiliation(s)
- Subramani Karthikeyan
- Centre for Healthcare Advancement, Innovation and Research, Vellore Institute of Technology Chennai 600 127 India
| | - Ayyavu Thirunarayanan
- Department of Chemical Engineering Biotechnology and Materials, FCFM, University of Chile Av. Beauchef 851 Santiago Chile
| | - Leon Bernet Shano
- Division of Physics, School of Advanced Sciences, Vellore Insititute of Technology (VIT) Chennai Campus Vandalur-Kelambakkam Road Tamil Nadu 600127 India
| | - Arasappan Hemamalini
- Department of Chemistry, Government College of Engineering Srirangam Sethurapatti Thiruchirappalli Tamil Nadu India
| | | | | | - Norhidayah Abu
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Kubang Kerian 16150 Kelantan Malaysia
| | | | - Shanmugavel Chinnathambi
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University Kyoto 616-8510 Japan
| | - Ganesh N Pandian
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University Kyoto 616-8510 Japan
| |
Collapse
|
7
|
Shams A, Alzahrani AA, Ayash TA, Tamur S, Al-Mourgi M. The Multifaceted Roles of Myrrha in the Treatment of Breast Cancer: Potential Therapeutic Targets and Promises. Integr Cancer Ther 2024; 23:15347354241309659. [PMID: 39707884 DOI: 10.1177/15347354241309659] [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] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Breast cancer is a critical threat to human health, and effective targeted agents showing lower systemic toxicity are still lacking. Therefore, exploring new potent therapeutic candidates with a broader safety margin is warranted. Alternative medicine, which has historically been used in traditional Chinese medicine, has played an increasingly prominent role in this area of research. This study introduces Commiphora myrrha (or myrrh) as a potential therapeutic candidate for treating breast cancer patients. Myrrh bioactive extracts have been used traditionally for decades to treat numerous medical disorders, including cancers, specifically breast cancer. Nonetheless, myrrh's precise rudimentary mechanisms of action in regulating genes involved in breast cancer evolution and progression remain elusive. PURPOSE Herein, we use a network pharmacology platform to identify the potential genes targeted by myrrh-active molecules in breast cancer. METHOD The identified targets' expression profiles were determined at the mRNA and protein levels using The Breast Cancer Gene-Expression Miner v5.0 (bcGen-ExMiner v5.0) and The Human Protein Atlas datasets, respectively. A gene signature composed of the specifically designated genes was constructed, and its association with different breast cancer molecular subtypes was investigated through the Gene expression-based Outcome for Breast Cancer (GOBO) online tool. The protein mapping relationship between potential myrrh targets and their partner proteins during breast cancer development was screened and constructed through the STRING and ShinyGO databases. In addition, the Kaplan-Meier plots (KM-plot) prognostic tool was applied to assess the survival rate associated with the expression of the current gene signature in different human cancers, including breast cancer. RESULTS Combining the results of network pharmacology with other bioinformatics databases suggests that myrrh's active components exert anti-cancer effects by regulating genes involved in breast cancer pathogenesis, particularly PTGS2, EGFR, ESR2, MMP2, and JUN. An individual evaluation of the expression profiles of these genes at both mRNA and protein levels reveals that a high expression profile of each gene is associated with breast cancer advancement. Moreover, the GOBO analysis shows an elevated expression profile of the PTGS2/ESR2/EGFR/JUN/MMP2 genes' signature in the most aggressive breast cancer subtype (Basal) in breast tumor samples and breast cancer cell lines. Furthermore, the STRING protein interaction network and the KEGG analyses indicate that myrrh exerts therapeutic effects on breast cancer by regulating several biological processes such as cell proliferation, cell migration, apoptosis, and various signaling pathways, including TNF, PI3K-Akt, NF-κB, and MAPK. Consistently, breast cancer patients with high expression of this genes' signature display poor survival outcomes. CONCLUSIONS The present study is the first attempt to explore the biological involvement of myrrh-targeted genes during breast cancer development. Therefore, suppressing the effects of the intended genes' signature using myrrh extracts would provide encouraging results in blocking breast cancer tumorigenesis. Thus, our findings provide conclusive evidence and deepen the current understanding of the molecular role of myrrh in the treatment of breast cancer, further supporting its clinical application.
Collapse
Affiliation(s)
- Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, Taif, Saudi Arabia
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif, Saudi Arabia
- High Altitude Research Center, Taif University, Taif, Saudi Arabia
| | | | - Taghreed A Ayash
- Department of General Science, Ibnsina International Medical College, Jeddah, Saudi Arabia
- Research and Innovation Central lab, Chair of Research and Innovation Central Lab, Ibnsina International Medical College, Jeddah, Saudi Arabia
| | - Shadi Tamur
- Department of Pediatric, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Majed Al-Mourgi
- Department of Surgery, College of Medicine, Taif University, Taif, Saudi Arabia
| |
Collapse
|
8
|
Alam MM, Alsenani NI, Abdelhamid AA, Ahmad A, Baothman OA, Hosawi SA, Altayeb H, Nadeem MS, Ahmad V, Nazreen S, Elhenawy AA. New paracetamol hybrids as anticancer and COX-2 inhibitors: Synthesis, biological evaluation and docking studies. Arch Pharm (Weinheim) 2024; 357:e2300340. [PMID: 37880869 DOI: 10.1002/ardp.202300340] [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: 06/23/2023] [Revised: 08/31/2023] [Accepted: 09/28/2023] [Indexed: 10/27/2023]
Abstract
Drug repurposing is an emerging field in drug development that has provided many successful drugs. In the current study, paracetamol, a known antipyretic and analgesic agent, was chemically modified to generate paracetamol derivatives as anticancer and anticyclooxygenase-2 (COX-2) agents. Compound 11 bearing a fluoro group was the best cytotoxic candidate with half-maximal inhibitory concentration (IC50 ) values ranging from 1.51 to 6.31 μM and anti-COX-2 activity with IC50 = 0.29 μM, compared to the standard drugs, doxorubicin and celecoxib. The cell cycle and apoptosis studies revealed that compound 11 possesses the ability to induce cell cycle arrest in the S phase and apoptosis in colon Huh-7 cells. These results were strongly supported by docking studies, which showed strong interactions with the amino acids of the COX-2 protein, and in silico pharmacokinetic predictions were found to be favorable for these newly synthesized paracetamol derivatives. It can be concluded that compound 11 could block cell growth and proliferation by inhibiting the COX-2 enzyme in cancer therapy.
Collapse
Affiliation(s)
- Mohammad Mahboob Alam
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Nawaf I Alsenani
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Antar A Abdelhamid
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
- Department of Chemistry, Faculty of Science, Sohag University, Sohag, Egypt
| | - Abrar Ahmad
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Othman A Baothman
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Salman A Hosawi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Hisham Altayeb
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Mohammad Shahid Nadeem
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Varish Ahmad
- Department of Health Information Technology, Faculty of Applied Studies, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Syed Nazreen
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
| | - Ahmed A Elhenawy
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, Kingdom of Saudi Arabia
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, Cairo, Egypt
| |
Collapse
|
9
|
Rohilla S, Goyal G, Berwal P, Mathur N. A Review on Indole-triazole Molecular Hybrids as a Leading Edge in Drug Discovery: Current Landscape and Future Perspectives. Curr Top Med Chem 2024; 24:1557-1588. [PMID: 38766822 DOI: 10.2174/0115680266307132240509065351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 05/22/2024]
Abstract
Molecular hybridization is a rational design strategy used to create new ligands or prototypes by identifying and combining specific pharmacophoric subunits from the molecular structures of two or more known bioactive derivatives. Molecular hybridization is a valuable technique in drug discovery, enabling the modulation of unwanted side effects and the creation of potential dual-acting drugs that combine the effects of multiple therapeutic agents. Indole-triazole conjugates have emerged as promising candidates for new drug development. The indole and triazole moieties can be linked through various synthetic strategies, such as click chemistry or other coupling reactions, to generate a library of diverse compounds for biological screening. The achievable structural diversity with indole-triazole conjugates offers avenues to optimize their pharmacokinetic and pharmacodynamic attributes, amplifying their therapeutic efficacy. Researchers have extensively tailored both indole and triazole frameworks with diverse modifications to comprehend their impact on the drug's pharmacokinetic and pharmacodynamic characteristics. The current review article endeavours to explore and discuss various research strategies to design indoletriazole hybrids and elucidate their significance in a variety of pathological conditions. The insights provided herein are anticipated to be beneficial for the researchers and will likely encourage further exploration in this field.
Collapse
Affiliation(s)
- Suman Rohilla
- Department of Pharmaceutical Chemistry, SGT College of Pharmacy, Shree Guru Gobind Singh Tricentenary University, Gurugram, Haryana, India
| | - Garima Goyal
- Department of Pharmaceutical Chemistry, SGT College of Pharmacy, Shree Guru Gobind Singh Tricentenary University, Gurugram, Haryana, India
| | - Paras Berwal
- Department of Pharmaceutical Chemistry, SGT College of Pharmacy, Shree Guru Gobind Singh Tricentenary University, Gurugram, Haryana, India
| | - Nancy Mathur
- Department of Pharmaceutical Chemistry, SGT College of Pharmacy, Shree Guru Gobind Singh Tricentenary University, Gurugram, Haryana, India
| |
Collapse
|
10
|
Une S, Nakata R, Nonaka K, Akiyama J. Antiproliferative and anti-inflammatory effects of fractionated crude lectins from boiled kidney beans (Phaseolus vulgaris). J Food Sci 2024; 89:671-683. [PMID: 37983860 DOI: 10.1111/1750-3841.16847] [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: 10/05/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023]
Abstract
In this study, we investigated the biological profile of lectins isolated from raw and boiled Japanese red Kintoki beans (red kidney beans [RKB]; Phaseolus vulgaris). Lectins in beans showing agglutination activity were retained after heating. Raw and boiled RKB lectins were fractionated using carboxymethyl- and diethylaminoethyl-Sepharose, respectively. Boiled RKB lectins were evaluated for carbohydrate specificity as well as cytokine-inducing and antiproliferative activities against cancer cells and compared with raw RKB lectins. Raw RKB lectins showed specificity for thyroglobulin and fetuin, whereas boiled lectins showed specificity for N-acetylneuraminic acid. Raw RKB lectins showed low resistance to proteases and tolerated temperatures greater than 80°C for 5 min. Notably, lectins from raw and boiled beans showed antiproliferative activity against five types of cancer cells B16, LM8, HeLa, HepG2, and Colo 679. In particular, lectins from raw beans exhibited a significantly stronger activity than those from boiled beans. Anti-inflammatory effects were notably observed in crude extracts from raw and boiled beans. Specifically, lectins fractionated from boiled beans markedly inhibited the expression of tumor necrosis factor-α and interleukin-6. Overall, our results showed that RKB lectins from boiled beans exert anti-inflammatory and anticancer effects and could be developed as potential chemopreventive agents. PRACTICAL APPLICATION: Japanese red kidney beans (RKB) are cultivated in numerous parts of the temperate zone and consumed in many countries. Lectins from boiled beans exhibited anticancer activity, similar to lectins from raw beans. Additionally, crude and fractionated lectins from boiled beans showed anti-inflammatory activity. Thus, boiled RKB lectins have the potential to be used as a bioactive protein for medical research and could be developed as anticancer agents.
Collapse
Affiliation(s)
- Satsuki Une
- Faculty of Education, Kagawa University, Takamatsu, Kagawa, Japan
| | - Rieko Nakata
- Faculty of human Life and Environment, Nara Women's University, Nara, Japan
| | - Koji Nonaka
- Faculty of Health Science, Naragakuen University, Sangho-cho, Nara, Japan
| | - Junich Akiyama
- Department of Physical Therapy, Kibi International University, Takahasi, Okayama, Japan
| |
Collapse
|
11
|
Ridnour LA, Cheng RYS, Heinz WF, Pore M, Gonzalez AL, Femino EL, Moffat R, Wink AL, Imtiaz F, Coutinho L, Butcher D, Edmondson EF, Rangel MC, Wong STC, Lipkowitz S, Glynn S, Vitek MP, McVicar DW, Li X, Anderson SK, Paolocci N, Hewitt SM, Ambs S, Billiar TR, Chang JC, Lockett SJ, Wink DA. Spatial analysis of NOS2 and COX2 interaction with T-effector cells reveals immunosuppressive landscapes associated with poor outcome in ER- breast cancer patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572867. [PMID: 38187660 PMCID: PMC10769421 DOI: 10.1101/2023.12.21.572867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Multiple immunosuppressive mechanisms exist in the tumor microenvironment that drive poor outcomes and decrease treatment efficacy. The co-expression of NOS2 and COX2 is a strong predictor of poor prognosis in ER- breast cancer and other malignancies. Together, they generate pro-oncogenic signals that drive metastasis, drug resistance, cancer stemness, and immune suppression. Using an ER- breast cancer patient cohort, we found that the spatial expression patterns of NOS2 and COX2 with CD3+CD8+PD1- T effector (Teff) cells formed a tumor immune landscape that correlated with poor outcome. NOS2 was primarily associated with the tumor-immune interface, whereas COX2 was associated with immune desert regions of the tumor lacking Teff cells. A higher ratio of NOS2 or COX2 to Teff was highly correlated with poor outcomes. Spatial analysis revealed that regional clustering of NOS2 and COX2 was associated with stromal-restricted Teff, while only COX2 was predominant in immune deserts. Examination of other immunosuppressive elements, such as PDL1/PD1, Treg, B7H4, and IDO1, revealed that PDL1/PD1, Treg, and IDO1 were primarily associated with restricted Teff, whereas B7H4 and COX2 were found in tumor immune deserts. Regardless of the survival outcome, other leukocytes, such as CD4 T cells and macrophages, were primarily in stromal lymphoid aggregates. Finally, in a 4T1 model, COX2 inhibition led to a massive cell infiltration, thus validating the hypothesis that COX2 is an essential component of the Teff exclusion process and, thus, tumor evasion. Our study indicates that NOS2/COX2 expression plays a central role in tumor immunosuppression. Our findings indicate that new strategies combining clinically available NOS2/COX2 inhibitors with various forms of immune therapy may open a new avenue for the treatment of aggressive ER-breast cancers.
Collapse
Affiliation(s)
- Lisa A Ridnour
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Robert Y S Cheng
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - William F Heinz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Milind Pore
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research
| | - Ana L Gonzalez
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Elise L Femino
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Rebecca Moffat
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Adelaide L Wink
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Fatima Imtiaz
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - Leandro Coutinho
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
- Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Donna Butcher
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
| | - Elijah F Edmondson
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
| | - M Cristina Rangel
- Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Sharon Glynn
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
| | | | - Daniel W McVicar
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
| | - Stephen K Anderson
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
- Basic Science Program, Frederick National Laboratory for Cancer Research
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins University, and Department of Biomedical Sciences, University of Padova, Italy
- Laboratory of Pathology CCR, NCI, NIH
| | | | - Stefan Ambs
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Timothy R Billiar
- Mary and Ron Neal Cancer Center, Houston Methodist Hospital and Weill Cornell Medicine, Houston, TX
| | - Jenny C Chang
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
- Imaging Mass Cytometry Frederick National Laboratory for Cancer Research
- Faculdade de Medicina da Universidade de São Paulo and Comprehensive Center for Precision Oncology, Universidade de São Paulo, São Paulo, SP, Brazil
- Molecular Histopathology Laboratories, Leidos Biomedical Research Inc. for the National Cancer Institute
- Houston Methodist Weill Cornell Medical College, Houston TX
- Women's Malignancies Branch, CCR, NCI, NIH
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, University of Galway, Galway, Ireland
- (Mike Duke)
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA
- Basic Science Program, Frederick National Laboratory for Cancer Research
- Division of Cardiology, Department of Medicine, Johns Hopkins University, and Department of Biomedical Sciences, University of Padova, Italy
- Laboratory of Pathology CCR, NCI, NIH
- Laboratory of Human Carcinogenesis, CCR, NCI, NIH, Bethesda, MD
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
- Mary and Ron Neal Cancer Center, Houston Methodist Hospital and Weill Cornell Medicine, Houston, TX
| | - Stephen J Lockett
- Optical Microscopy and Analysis Laboratory, Frederick National Laboratory for Cancer Research; Leidos Biomedical Research Inc. for the National Cancer Institute, Frederick, MD
| | - David A Wink
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD
| |
Collapse
|
12
|
Kang Q, Zheng K, Jiang GM, Li YK, Liang YB, Geng Q, Qian CH, Wang QB, He ZY, Huang SQ, Yang C, Li J, Li YH, Ke Y. Brucine suppresses proliferation and promotes apoptosis of human cholangiacarcinoma cells via the inhibition of COX2 expression. J Cancer 2023; 14:2700-2706. [PMID: 37779869 PMCID: PMC10539398 DOI: 10.7150/jca.87514] [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: 06/27/2023] [Accepted: 08/22/2023] [Indexed: 10/03/2023] Open
Abstract
Aims The aim of this study was to investigate the anti-tumor efficacy of brucine on intrahepatic cholangiocarcinoma (ICC). Methods ICC QBC939 cells were treated with brucine, cell viability, cell cycle and apoptosis were analyzed using CCK-8 and flow cytometry. The expression of COX-2 and apoptosis related proteins Casp3, Bax and Bcl-2 were detected by Western blot analysis. QBC939 cells were subcutaneously transplanted into nude mice and the mice were injected with brucine intraperitoneally. The expression of Ki67, COX-2 and apoptosis related proteins were detected by immunohistochemical staining and Western blot analysis. Results Brucine significantly inhibited the proliferation and cell cycle progression while promoted the apoptosis of QBC939 cells. The expression of the apoptotic proteins Casp3 and Bax was upregulated, while the expression of Bcl-2 and COX-2 was downregulated in QBC939 cells with brucine treatment. Moreover, the overexpression of COX-2 could antagonize the effects of brucine on QBC939 cells. In vivo, brucine inhibited subcutaneous tumor formation in nude mice, and the expression of Ki67, COX-2 and Bcl-2 decreased while the expression of Casp3 and Bax increased in tumor tissues from nude mice with brucine treatment. Conclusions Brucine can significantly inhibit the progression of cholangiocarcinoma in vitro and in vivo, and the mechanism may be related to the inhibition of COX-2 expression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yue-Hua Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| | - Yang Ke
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, China
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Mfotie Njoya E, Ndemangou B, Akinyelu J, Munvera AM, Chukwuma CI, Mkounga P, Mashele SS, Makhafola TJ, McGaw LJ. In vitro antiproliferative, anti-inflammatory effects and molecular docking studies of natural compounds isolated from Sarcocephalus pobeguinii (Hua ex Pobég). Front Pharmacol 2023; 14:1205414. [PMID: 37416061 PMCID: PMC10320002 DOI: 10.3389/fphar.2023.1205414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Background: Sarcocephalus pobeguinii (Hua ex Pobég) is used in folk medicine to treat oxidative-stress related diseases, thereby warranting the investigation of its anticancer and anti-inflammatory properties. In our previous study, the leaf extract of S. pobeguinii induced significant cytotoxic effect against several cancerous cells with high selectivity indexes towards non-cancerous cells. Aim: The current study aims to isolate natural compounds from S. pobeguinii, and to evaluate their cytotoxicity, selectivity and anti-inflammatory effects as well as searching for potential target proteins of bioactive compounds. Methods: Natural compounds were isolated from leaf, fruit and bark extracts of S. pobeguinii and their chemical structures were elucidated using appropriate spectroscopic methods. The antiproliferative effect of isolated compounds was determined on four human cancerous cells (MCF-7, HepG2, Caco-2 and A549 cells) and non-cancerous Vero cells. Additionally, the anti-inflammatory activity of these compounds was determined by evaluating the nitric oxide (NO) production inhibitory potential and the 15-lipoxygenase (15-LOX) inhibitory activity. Furthermore, molecular docking studies were carried out on six putative target proteins found in common signaling pathways of inflammation and cancer. Results: Hederagenin (2), quinovic acid 3-O-[α-D-quinovopyranoside] (6) and quinovic acid 3-O-[β-D-quinovopyranoside] (9) exhibited significant cytotoxic effect against all cancerous cells, and they induced apoptosis in MCF-7 cells by increasing caspase-3/-7 activity. (6) showed the highest efficacy against all cancerous cells with poor selectivity (except for A549 cells) towards non-cancerous Vero cells; while (2) showed the highest selectivity warranting its potential safety as a chemotherapeutic agent. Moreover, (6) and (9) significantly inhibited NO production in LPS-stimulated RAW 264.7 cells which could mainly be attributed to their high cytotoxic effect. Besides, the mixture nauclealatifoline G and naucleofficine D (1), hederagenin (2) and chletric acid (3) were active against 15-LOX as compared to quercetin. Docking results showed that JAK2 and COX-2, with the highest binding scores, are the potential molecular targets involved in the antiproliferative and anti-inflammatory effects of bioactive compounds. Conclusion: Overall, hederagenin (2), which selectively killed cancer cells with additional anti-inflammatory effect, is the most prominent lead compound which may be further investigated as a drug candidate to tackle cancer progression.
Collapse
Affiliation(s)
- Emmanuel Mfotie Njoya
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, South Africa
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaound, Cameroon
| | - Brigitte Ndemangou
- University Institute of Technology of Wood Technology, Mbalmayo, Cameroon
| | - Jude Akinyelu
- Department of Biochemistry, Federal University Oye-Ekiti, Oye, Nigeria
| | - Aristide M. Munvera
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaound, Cameroon
| | - Chika. I. Chukwuma
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, South Africa
| | - Pierre Mkounga
- Department of Organic Chemistry, Faculty of Science, University of Yaoundé I, Yaound, Cameroon
| | - Samson S. Mashele
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, South Africa
| | - Tshepiso J. Makhafola
- Centre for Quality of Health and Living, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, South Africa
| | - Lyndy J. McGaw
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
16
|
Coskun GP, Ozhan Y, Dobričić V, Bošković J, Reis R, Sipahi H, Sahin Z, Demirayak S. Discovery of Novel Thiophene/Hydrazones: In Vitro and In Silico Studies against Pancreatic Cancer. Pharmaceutics 2023; 15:1441. [PMID: 37242684 PMCID: PMC10223062 DOI: 10.3390/pharmaceutics15051441] [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: 03/31/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Cancer is the disease with the highest mortality. Drug studies contribute to promising treatments; however there is an urgent need for selective drug candidates. Pancreatic cancer is difficult to treat and the cancer progresses rapidly. Unfortunately, current treatments are ineffective. In this study, ten new diarylthiophene-2-carbohydrazide derivatives were synthesized and evaluated for their pharmacological activity. The 2D and 3D anticancer activity studies suggested the compounds 7a, 7d, and 7f were promising. Among these, 7f (4.86 µM) showed the best 2D inhibitory activity against PaCa-2 cells. Compounds 7a, 7d and 7f were also tested for their cytotoxic effects on healthy cell line but only compound 7d showed selectivity. Compounds 7a, 7d, and 7f showed the best 3D cell line inhibitory effect according to spheroid diameters. The compounds were screened for their COX-2 and 5-LOX inhibitory activity. For COX-2, the best IC50 value was observed for 7c (10.13 µM) and all compounds showed significantly lower inhibition compared to standard. In the 5-LOX inhibition study, compounds 7a (3.78 µM), 7c (2.60 µM), 7e (3.3 µM), and 7f (2.94 µM) demonstrated influential activity compared to standard. Regarding molecular docking studies, binding mode of compounds 7c, 7e, and 7f to the 5-LOX enzyme were non-redox or redox types, but not the iron-binding type. As dual inhibitors of 5-LOX and pancreatic cancer cell line, 7a and 7f were identified as the most promising compounds.
Collapse
Affiliation(s)
- Goknil Pelin Coskun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| | - Yagmur Ozhan
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Yeditepe University, 34755 Istanbul, Turkey; (Y.O.); (H.S.)
| | - Vladimir Dobričić
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (V.D.)
| | - Jelena Bošković
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, 11221 Belgrade, Serbia; (V.D.)
| | - Rengin Reis
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Hande Sipahi
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Yeditepe University, 34755 Istanbul, Turkey; (Y.O.); (H.S.)
| | - Zafer Sahin
- Department of Pharmaceutical Chemistry, Hamidiye Faculty of Pharmacy, University of Health Sciences, 34668 Istanbul, Turkey;
| | - Seref Demirayak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kocaeli Health and Technology University, 41090 Kocaeli, Turkey;
| |
Collapse
|
17
|
Li S, Chen T, Liu J, Zhang H, Li J, Wang Z, Shang G. PROTACs: Novel tools for improving immunotherapy in cancer. Cancer Lett 2023; 560:216128. [PMID: 36933781 DOI: 10.1016/j.canlet.2023.216128] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Posttranslational modifications (PTMs), such as phosphorylation, methylation, ubiquitination, and acetylation, are important in governing protein expression levels. Proteolysis targeting chimeras (PROTACs) are novel structures designed to target a protein of interest (POI) for ubiquitination and degradation, leading to the selective reduction in the expression levels of the POI. PROTACs have exhibited great promise due to their ability to target undruggable proteins, including several transcription factors. Recently, PROTACs have been characterized to improve anticancer immunotherapy via the regulation of specific proteins. In this review, we describe how the PROTACs target several molecules, including HDAC6, IDO1, EGFR, FoxM1, PD-L1, SHP2, HPK1, BCL-xL, BET proteins, NAMPT, and COX-1/2, to regulate immunotherapy in human cancers. PROTACs may provide potential treatment benefits by enhancing immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Shizhe Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| | - Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| | - Jinxin Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| | - He Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| | - Jiatong Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| | - Zhiwei Wang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Anhui, 233030, China; The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| | - Guanning Shang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China.
| |
Collapse
|