1
|
Wu Q, Ghosal K, Kana'an N, Roy S, Rashed N, Majumder R, Mandal M, Gao L, Farah S. On-demand imidazolidinyl urea-based tissue-like, self-healable, and antibacterial hydrogels for infectious wound care. Bioact Mater 2025; 44:116-130. [PMID: 39484021 PMCID: PMC11525126 DOI: 10.1016/j.bioactmat.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/15/2024] [Accepted: 10/01/2024] [Indexed: 11/03/2024] Open
Abstract
Bacterial wound infections are a growing challenge in healthcare, posing severe risks like systemic infection, organ failure, and sepsis, with projections predicting over 10 million deaths annually by 2050. Antibacterial hydrogels, with adaptable extracellular matrix-like features, are emerging as promising solutions for treating infectious wounds. However, the antibacterial properties of most of these hydrogels are largely attributed to extrinsic agents, and their mechanisms of action remain poorly understood. Herein we introduce for the first time, modified imidazolidinyl urea (IU) as the polymeric backbone for developing tissue-like antibacterial hydrogels. As-designed hydrogels behave tissue-like mechanical features, outstanding antifreeze behavior, and rapid self-healing capabilities. Molecular dynamics (MD) simulation and density functional theory (DFT) calculation were employed to well-understand the extent of H-bonding and metal-ligand coordination to finetune hydrogels' properties. In vitro studies suggest good biocompatibility of hydrogels against mouse fibroblasts & human skin, lung, and red blood cells, with potential wound healing capacity. Additionally, the hydrogels exhibit good 3D printability and remarkable antibacterial activity, attributed to concentration dependent ROS generation, oxidative stress induction, and subsequent disruption of bacterial membrane. On top of that, in vitro biofilm studies confirmed that developed hydrogels are effective in preventing biofilm formation. Therefore, these tissue-mimetic hydrogels present a promising and effective platform for accelerating wound healing while simultaneously controlling bacterial infections, offering hope for the future of wound care.
Collapse
Affiliation(s)
- Qi Wu
- The Laboratory for Advanced Functional/Medicinal Polymers & Smart Drug Delivery Technologies, The Wolfson Faculty of Chemical Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Krishanu Ghosal
- The Laboratory for Advanced Functional/Medicinal Polymers & Smart Drug Delivery Technologies, The Wolfson Faculty of Chemical Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Nadine Kana'an
- The Laboratory for Advanced Functional/Medicinal Polymers & Smart Drug Delivery Technologies, The Wolfson Faculty of Chemical Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Shounak Roy
- The Laboratory for Advanced Functional/Medicinal Polymers & Smart Drug Delivery Technologies, The Wolfson Faculty of Chemical Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Nagham Rashed
- The Laboratory for Advanced Functional/Medicinal Polymers & Smart Drug Delivery Technologies, The Wolfson Faculty of Chemical Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Ranabir Majumder
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, 721302, India
| | - Liang Gao
- Jinan Key Laboratory of High Performance Industrial Software, Jinan Institute of Supercomputing Technology, Jinan, 250000, China
| | - Shady Farah
- The Laboratory for Advanced Functional/Medicinal Polymers & Smart Drug Delivery Technologies, The Wolfson Faculty of Chemical Engineering, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
- The Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| |
Collapse
|
2
|
Chen Z, Luo R, Xu T, Wang L, Deng S, Wu J, Wang H, Lin Y, Bu M. Design, synthesis and antitumor effects of lupeol quaternary phosphonium salt derivatives. Bioorg Med Chem 2024; 113:117934. [PMID: 39369566 DOI: 10.1016/j.bmc.2024.117934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/08/2024]
Abstract
Lupeol is a natural pentacyclic triterpenoid with a wide range of biological activities. To improve the water solubility and targeting of lupeol, in the following study, we synthesized 27 lupeol derivatives in the first series by introducing lipophilic cations with lupeol as the lead compound. Through the screening of different cancer cells, we found that some of the derivatives showed better activity than cisplatin against human non-small cell lung cancer A549 cells, among which compound 6c was found to have an IC50 value of 1.83 μM and a selectivity index of 21.02 (IC50MRC-5/IC50A549) against A549 cells. To further improve the antiproliferative activity of the compounds, we replaced the ester linkage of the linker with a carbamate linkage and synthesized a second series of five lupeol derivatives which were screened for activity, among which compound 14f was found to have an IC50 value of 1.36 μM and a selectivity index of 15.60 (IC50MRC-5/IC50A549) against A549 cells. We further evaluated the bioactivity of compounds 6c and 14f and found that both compounds induced apoptosis in A549 cells, promoted an increase in intracellular reactive oxygen species and decrease in mitochondrial membrane potential, and inhibited the cell cycle in the S phase. Of the compounds, compound 14f showed stronger bioactivity than compound 6c. We then selected compound 14f for molecular-level Western blot evaluation and in vivo evaluation in the zebrafish xenograft A549 tumor cell model. Compound 14f was found to significantly downregulate Bcl-2 protein expression and upregulate Bax, Cyt C, cleaved caspase-9, and cleaved caspase-3 protein expression, and 14f was found to be able to inhibit the proliferation of A549 cells in the zebrafish xenograft model. The above results suggest that compound 14f has great potential in the development of antitumor drugs targeting mitochondria.
Collapse
Affiliation(s)
- Zongxing Chen
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Ran Luo
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Tianci Xu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Lu Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Siqi Deng
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Jiale Wu
- College of Pharmacy, Hainan University, Haikou 570228, Hainan, PR China
| | - Haijun Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Yu Lin
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China
| | - Ming Bu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, Heilongjiang, PR China.
| |
Collapse
|
3
|
Han Mİ, Gündüz MG, Ammara A, Supuran CT, Doğan ŞD. Tail-approach based design, synthesis, and molecular modeling of benzenesulfonamides carrying thiadiazole and urea moieties as novel carbonic anhydrase inhibitors. Arch Pharm (Weinheim) 2024; 357:e2400439. [PMID: 39079940 DOI: 10.1002/ardp.202400439] [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: 05/31/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 11/06/2024]
Abstract
We synthesized herein 16 compounds (SUT1-SUT16) as potential carbonic anhydrase (CA) inhibitors utilizing the tail-approach design. Based on this strategy, we connected benzenesulfonamide, the zinc-binding scaffold, to different urea moieties with the 1,3,4-thiadiazole ring as a linker. We obtained the target compounds by the reaction of 4-(5-amino-1,3,4-thiadiazol-2-yl)benzenesulfonamide with aryl isocyanates. Upon confirmation of their structures, the compounds were screened for their ability to inhibit the tumor-related human (h) isoforms human carbonic anhydrase (hCA) IX and XII, as well as the physiologically dominant hCA I and II. Most of the molecules demonstrated Ki values ≤ 10 nM with different selectivity profiles. The binding modes of SUT9, SUT10, and SUT5, the most effective inhibitors of hCA II, IX, and XII, respectively, were predicted by molecular docking. SUT16 (4-{5-[3-(naphthalen-1-yl)ureido]-1,3,4-thiadiazol-2-yl}benzenesulfonamide) was found to be the most selective inhibitor of the cancer-associated isoforms hCA IX and XII over the off-target isoforms, hCAI and II. The interaction dynamics and stability of SUT16 within hCA IX and XII were investigated by molecular dynamics simulations as well as dynophore analysis. Based on computational data, increased hydrophobic contacts and hydrogen bonds in the tail part of these molecules within hCA IX and XII were found as favorable interactions leading to effective inhibitors of cancer-related isoforms.
Collapse
Affiliation(s)
- M İhsan Han
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Miyase Gözde Gündüz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Andrea Ammara
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Firenze, Italy
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Firenze, Italy
| | - Şengül Dilem Doğan
- Department of Basic Sciences, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| |
Collapse
|
4
|
Rubbab Pasha A, Khan M, Khan A, Hussain J, Al-Rashida M, Islam T, Batool Z, Kashtoh H, Abdellattif MH, Al-Harrasi A, Shafiq Z, Schenone S. Synthesis, in vitro, and in silico study of novel pyridine based 1,3-diphenylurea derivatives as tyrosinase inhibitors. Bioorg Chem 2024; 152:107724. [PMID: 39167873 DOI: 10.1016/j.bioorg.2024.107724] [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: 07/22/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Tyrosinase inhibitors are studied in the cosmetics and pharmaceutical sectors as tyrosinase enzyme is involved in the biosynthesis and regulation of melanin, hence these inhibitors are beneficial for the management of melanogenesis and hyperpigmentation-related disorders. In the current work, a novel series of diphenyl urea derivatives containing a halo-pyridine moiety (5a-t) was synthesized via a multi-step synthesis. In vitro, tyrosinase inhibitory assay results showed that, except for two compounds, the derivatives were excellent inhibitors of human tyrosinase. The average IC50 value of the inhibitors (15.78 μM) is lower than that of kojic acid (17.3 μM) used as the reference compound, indicating that, on average, these molecules are more potent than the reference. Derivative 5a was identified as the most potent human tyrosinase inhibitor of the series, with an IC50 value of 3.5 ± 1.2 μM, approximately 5 times more potent than kojic acid. To get further insights into the nature of binding site interactions, molecular docking and molecular dynamics simulation studies were carried out. Moreover, the evaluation of in silico ADME properties showed a highly favorable profile for the synthesized compounds. These findings suggested that the further development of this class of compounds could be useful to get potent drug-like compounds that can target hyperpigmentation-related disorders.
Collapse
Affiliation(s)
- Anam Rubbab Pasha
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800, Pakistan; Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616, Birkat Al Mauz, Nizwa, Oman
| | - Majid Khan
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616, Birkat Al Mauz, Nizwa, Oman; Department of Biochemistry, Hazara University, Mansehra, Pakistan
| | - Ajmal Khan
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616, Birkat Al Mauz, Nizwa, Oman
| | - Javid Hussain
- Department of Biological Sciences and Chemistry, University of Nizwa, Oman
| | - Mariya Al-Rashida
- Department of Chemistry, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Talha Islam
- Department of Chemistry, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Zahra Batool
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Hamdy Kashtoh
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongbuk, Republic of Korea.
| | - Magda H Abdellattif
- Chemistry Department, College of Sciences, University College of Taraba, Taif University, Taif 21944, Saudi Arabia
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616, Birkat Al Mauz, Nizwa, Oman.
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Silvia Schenone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, Genoa 16132, Italy.
| |
Collapse
|
5
|
Rath S, Patel S, Choppella S, Menon P, Garain T, Banerjee S, Ravva MK, Sen S. Photolytic ortho-Selective Amino Pyridylation of Aryl Isocyanates with N-Amino Pyridinium Ylides for the Synthesis of N-Arylsulfonyl Ureas. J Org Chem 2024; 89:14770-14784. [PMID: 39373291 DOI: 10.1021/acs.joc.4c01408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Herein, we report an expedient synthesis of aryl sulfonyl ureas 4 and 5 from N-amino pyridinium ylides and aryl isocyanates. N-Aminopyridinium ylides 3 are synthesized via blue light-emitting diode irradiation of pyridine/isoquinoline and appropriate iminoiodinanes. The strategy involved a hitherto unknown carboamination of imine moieties (of aryl isocyanates) via a three-component reaction of pyridine derivatives/isoquinoline 1, N-aryl sulfonyl iminoiodinanes 2, and numerous aryl isocyanates at room temperature in 2-methyl tetrahydrofuran to afford the target compounds in moderate to excellent yields. N-Arylpyridinium ylides 3 (as intermediates) undergo a [3+2] cycloaddition with the aryl isocyanates followed by the aromatization of the pyridine/isoquinoline moiety to afford compounds 4. On the basis of the substitution pattern among the reactants, in some cases pyridine extrusion occurs during the reaction to afford depyridinylated aryl sulfonyl ureas 5. In general, isocyanates are used as dipolarophiles in [3+2] cycloaddition reactions. However, regioselective amino pyridylation of these species is a first. Control experiments and density functional theory calculations elucidate the reaction mechanism. The batch process of the protocol could be seamlessly transferred to the photoflow synthesis.
Collapse
Affiliation(s)
- Suchismita Rath
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Dadri, Chithera, Gautam Buddha Nagar, UP 201314, India
| | - Shreemad Patel
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Dadri, Chithera, Gautam Buddha Nagar, UP 201314, India
| | | | - Pranoy Menon
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Dadri, Chithera, Gautam Buddha Nagar, UP 201314, India
| | - Tanya Garain
- Molecular Biosciences, Middle Tennessee State University, Murfreesboro, Tennessee 37132-0001, United States
| | - Souvik Banerjee
- Molecular Biosciences, Middle Tennessee State University, Murfreesboro, Tennessee 37132-0001, United States
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, Tennessee 37132, United States
| | | | - Subhabrata Sen
- Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University, Dadri, Chithera, Gautam Buddha Nagar, UP 201314, India
| |
Collapse
|
6
|
Adeyemi OS, Johnson T, Maduakolam-Aniobi T, Kato K. Molecular modelling and experimental validation identified a new therapeutic inhibitor of toxoplasmosis. Comput Biol Med 2024; 183:109236. [PMID: 39378576 DOI: 10.1016/j.compbiomed.2024.109236] [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: 06/08/2024] [Revised: 09/02/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024]
Abstract
Toxoplasmosis is a widespread parasitic disease, caused by Toxoplasma gondii, that affects nearly one-third of the human population. The lack of effective treatments drives the demand for novel anti-toxoplasmosis therapeutic options. In the present study, we used computational approaches and experimental validation to identify therapeutic inhibitors of toxoplasmosis. Initially, using the structure of the co-crystallized ligand of T. gondii calcium-dependent protein kinase 1 (TgCDPK1), we retrieved 3000 compounds from the database of COCONUT (COlleCtion of Open Natural ProdUcTs). These compounds were docked against the crystal structure of TgCDPK1 on the Glide Ligand Docking panel of Maestro 12.5 (Schrödinger Suite 2020-3). Based on the docking scores, we assessed promising molecules for toxicity potential on the ProTox-II online server, while the ADME profiling was done on the SwissADME server. Following the computational studies, we selected nine promising compounds for experimental validation against T. gondii in vitro. Of the compounds, C4, C5, C6, and C8 exhibited dose-dependent anti-T. gondii action with EC50 values ranging from 3.3 to 120.2 μg/mL. Host toxicity profiling revealed differential cytotoxic action with a selectivity index (SI) of <1 for the compounds except C5, which had an SI of 1.8. To validate our screening assay, we used sulfadiazine, a standard drug for toxoplasmosis and showed that it inhibited parasite growth. Further experiments showed that C5, an imidazole-based natural compound, has strong but reversible anti-parasitic action that peaks within the first 8 h. In addition, C5 exhibited similar toxic tendencies towards T. gondii within (intracellular) and outside (extracellular) the host, suggesting it likely has a parasite target(s). C5 showed no effect on host invasion but strongly impeded parasite replication and growth, thereby affecting the T. gondii lytic cycle. Furthermore, C5 treatment raised the reactive oxygen species level, but this may be a secondary effect because augmentation with Trolox antioxidant failed to block C5 anti-T. gondii action. In addition, molecular dynamics simulations of C5 and TgCDPK1 complex revealed relative stability within 100 ns run time. Collectively, our findings support the potential of imidazole-based compounds as novel, alternative anti-parasitic agents.
Collapse
Affiliation(s)
- Oluyomi Stephen Adeyemi
- Medicinal Biochemistry and Toxicology Laboratory, Department of Biochemistry, Bowen University, Iwo, 232101, Osun State, Nigeria; Laboratory of Sustainable Animal Environment Systems, Graduate School of Agricultural Sciences, Tohoku University, Japan
| | - Titilayo Johnson
- Department of Biochemistry, University of Jos, Jos, Plateau State, Nigeria
| | | | - Kentaro Kato
- Laboratory of Sustainable Animal Environment Systems, Graduate School of Agricultural Sciences, Tohoku University, Japan.
| |
Collapse
|
7
|
Mohamed MA, Elsaman T, Elderdery AY, Alsrhani A, Ghanem HB, Alruwaili MM, Hamza SMA, Mekki SEI, Alotaibi HA, Mills J. Unveiling the Anticancer Potential: Computational Exploration of Nitrogenated Derivatives of (+)-Pancratistatin as Topoisomerase I Inhibitors. Int J Mol Sci 2024; 25:10779. [PMID: 39409108 PMCID: PMC11476810 DOI: 10.3390/ijms251910779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Cancer poses a substantial global health challenge, driving the need for innovative therapeutic solutions that offer improved effectiveness and fewer side effects. Topoisomerase I (Topo I) has emerged as a validated molecular target in the pursuit of developing anticancer drugs due to its critical role in DNA replication and transcription. (+)-Pancratistatin (PST), a naturally occurring compound found in various Amaryllidaceae plants, exhibits promising anticancer properties by inhibiting Topo I activity. However, its clinical utility is hindered by issues related to limited chemical availability and aqueous solubility. To address these challenges, molecular modelling techniques, including virtual screening, molecular docking, molecular mechanics with generalised born and surface area solvation (MM-GBSA) calculations, and molecular dynamics simulations were utilised to evaluate the binding interactions and energetics of PST analogues with Topo I, comparing them with the well-known Topo I inhibitor, Camptothecin. Among the compounds screened for this study, nitrogenated analogues emerged as the most encouraging drug candidates, exhibiting improved binding affinities, favourable interactions with the active site of Topo I, and stability of the protein-ligand complex. Structural analysis pinpointed key molecular determinants responsible for the heightened potency of nitrogenated analogues, shedding light on essential structural modifications for increased activity. Moreover, in silico absorption, distribution, metabolism, excretion, and toxicity (ADMET) predictions highlighted favourable drug-like properties and reduced toxicity profiles for the most prominent nitrogenated analogues, further supporting their potential as effective anticancer agents. In summary, this screening study underscores the significance of nitrogenation in augmenting the anticancer efficacy of PST analogues targeting Topo I. The identified lead compounds exhibit significant potential for subsequent experimental validation and optimisation, thus facilitating the development of novel and efficacious anticancer therapeutics with enhanced pharmacological profiles.
Collapse
Affiliation(s)
- Magdi Awadalla Mohamed
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Tilal Elsaman
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Abozer Y. Elderdery
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia; (A.Y.E.); (A.A.); (H.B.G.)
| | - Abdullah Alsrhani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia; (A.Y.E.); (A.A.); (H.B.G.)
| | - Heba Bassiony Ghanem
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 42421, Saudi Arabia; (A.Y.E.); (A.A.); (H.B.G.)
| | - Majed Mowanes Alruwaili
- Nursing Administration & Education Department, College of Nursing, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Siddiqa M. A. Hamza
- Department of Pathology, College of Medicine, Umm Alqura University, Algunfudah 21912, Saudi Arabia;
| | | | | | - Jeremy Mills
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK;
| |
Collapse
|
8
|
Yücetepe S, Koçyiğit-Kaymakçıoğlu B, Yang X, Tabanca N, Tok F. Insecticidal effect of new synthesized chalcone derivatives on Caribbean fruit fly, Anastrepha suspensa. Z NATURFORSCH C 2024; 79:267-273. [PMID: 38695678 DOI: 10.1515/znc-2024-0005] [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: 01/09/2024] [Accepted: 04/18/2024] [Indexed: 09/08/2024]
Abstract
In this present study, new chalcone derivatives were synthesized from 4-aminoacetophenone, which were confirmed by spectroscopic methods. The toxic risks of chalcones to humans and the environment were investigated by a web-based platform called ADMETlab. With this program, the possible toxic effects of the compounds on liver, respiratory system, and eyes were evaluated. For the topical insecticidal activity, adult female Caribbean fruit fly, Anastrepha suspensa, was targeted. Results of the toxicity tests showed that chalcone derivatives are effective against female A. suspensa. Among the synthesized chalcones, 1-(4-cinnamoylphenyl)-3-(p-tolyl)urea (2) exhibited the greatest insecticidal activity, resulting in 73 % mortality at 100 µg/fly after 24 h, whereas other derivatives showed less than 30 % mortality. Our results demonstrate that insecticidal activity may be modulated by the presence of a certain phenyl ring in the structure of derivative 2 and, therefore, has potential for design of efficient chemicals for tephritid fruit fly management.
Collapse
Affiliation(s)
- Sümeyye Yücetepe
- 52982 Institute of Health Sciences, Marmara University , 34865, Istanbul, Türkiye
| | - Bedia Koçyiğit-Kaymakçıoğlu
- Department of Pharmaceutical Chemistry, 20420479 Faculty of Pharmacy, Biruni University , 34015, Istanbul, Türkiye
| | - Xiangbing Yang
- 2017123 United States Department of Agriculture-Agricultural Research Service (USDA-ARS) , Subtropical Horticulture Research Station (SHRS), 13601 Old Cutler Rd., Miami, FL 33158, USA
| | - Nurhayat Tabanca
- 2017123 United States Department of Agriculture-Agricultural Research Service (USDA-ARS) , Subtropical Horticulture Research Station (SHRS), 13601 Old Cutler Rd., Miami, FL 33158, USA
| | - Fatih Tok
- Department of Pharmaceutical Chemistry, 52982 Faculty of Pharmacy, Marmara University , 34854, Istanbul, Türkiye
| |
Collapse
|
9
|
Pasha AR, Ullah S, Khan A, Halim SA, Hussain J, Rehman T, Talib R, Alharthy RD, Kashtoh H, Abdellattif MH, Al-Harrasi A, Shafiq Z. Synthesis, in vitro and in silico study of novel 1,3-diphenylurea derived Schiff bases as competitive α-glucosidase inhibitors. RSC Adv 2024; 14:29288-29300. [PMID: 39285881 PMCID: PMC11403393 DOI: 10.1039/d4ra05767h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
Diabetes mellitus has become a major global health burden because of several related consequences, including heart disease, retinopathy, cataracts, metabolic syndrome, collapsed renal function, and blindness. In the recent study, thirty Schiff base derivatives of 1,3-diphenylurea were synthesized and their anti-diabetic activity was evaluated by targeting α-glucosidase. The compounds exhibited an overwhelming inhibitory potential for α-glucosidase with higher potency ranging from 2.49-37.16 μM. The most effective compound, 5h, showed competitive inhibition of α-glucosidase (K i = 3.96 ± 0.0048 μM) in the kinetic analysis and strong binding interactions with key residues α-glucosidase in docking analysis, indicating its potential for better glycemic control in diabetes patients.
Collapse
Affiliation(s)
- Anam Rubbab Pasha
- Institute of Chemical Sciences, Bahauddin Zakariya University Multan-60800 Pakistan
- Natural and Medical Sciences Research Centre, University of Nizwa P.O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| | - Saeed Ullah
- Natural and Medical Sciences Research Centre, University of Nizwa P.O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Centre, University of Nizwa P.O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
- Department of Chemical and Biological Engineering, College of Engineering, Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 Republic of Korea
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Centre, University of Nizwa P.O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| | - Javid Hussain
- Department of Biological Sciences and Chemistry, University of Nizwa Oman
| | - Tanzila Rehman
- Department of Chemistry, The Women University Multan-60000 Pakistan
| | - Rimsha Talib
- Institute of Chemical Sciences, Bahauddin Zakariya University Multan-60800 Pakistan
| | - Rima D Alharthy
- Department of Chemistry, Science & Arts College, Rabigh Branch, King Abdulaziz University Rabigh 21911 Saudi Arabia
| | - Hamdy Kashtoh
- Department of Biotechnology, Yeungnam University Gyeongsan 38541 Gyeongbuk Republic of Korea
| | - Magda H Abdellattif
- Chemistry Department, College of Sciences, University College of Taraba, Taif University Taif 21944 Saudi Arabia
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa P.O. Box 33, PC 616, Birkat Al Mauz Nizwa Sultanate of Oman
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University Multan-60800 Pakistan
| |
Collapse
|
10
|
Spieß P, Brześkiewicz J, Maulide N. Deprotective Lossen rearrangement: a direct and general transformation of Nms-amides to unsymmetrical ureas. Chem Sci 2024:d4sc04974h. [PMID: 39268216 PMCID: PMC11385062 DOI: 10.1039/d4sc04974h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Ureas stand out as potent pharmacophores in drug development, rendering them a prime focus for synthesis. Herein, we present an appealing entry point for urea synthesis from protected amines (Nms-amides) and relying on a Lossen-type rearrangement process as an elegant example of deprotective functionalisation. The method developed exhibits an exceptionally broad tolerance towards various protected amines, encompassing numerous drug derivatives, and delivers high reaction yields.
Collapse
Affiliation(s)
- Philipp Spieß
- Institute of Organic Chemistry, University of Vienna Währinger Strasse 38 1090 Vienna Austria
| | - Jakub Brześkiewicz
- Institute of Organic Chemistry, University of Vienna Währinger Strasse 38 1090 Vienna Austria
| | - Nuno Maulide
- Institute of Organic Chemistry, University of Vienna Währinger Strasse 38 1090 Vienna Austria
| |
Collapse
|
11
|
Gou W, Dai P, Wang M, Wang Y, Ma N, Zhou X, Xu Y, Zhang L, Li C. Synthesis of Diverse Ureas from Amines and CO 2 at Atmospheric Pressure and Room Temperature. J Org Chem 2024; 89:12498-12507. [PMID: 39180140 DOI: 10.1021/acs.joc.4c01474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/26/2024]
Abstract
A metal-free method is developed to perform the synthesis of urea derivatives utilizing CO2 as the C1 building block at atmospheric pressure and room temperature. In addition to diverse symmetric and dissymmetric ureas, benzimidazolones and quinazolinone can also be easily prepared using this protocol. Most importantly, the gram-scale preparation of fungicide pencycuron and antipsychotic drug pimavanserin proceeded smoothly under the mild conditions.
Collapse
Affiliation(s)
- Wenchang Gou
- School of Pharmaceutical Sciences, Guizhou Medical University, 550004 Guiyang, People's Republic of China
| | - Pinli Dai
- School of Pharmaceutical Sciences, Guizhou Medical University, 550004 Guiyang, People's Republic of China
| | - Mei Wang
- School of Pharmaceutical Sciences, Guizhou Medical University, 550004 Guiyang, People's Republic of China
| | - Yunhuan Wang
- School of Pharmaceutical Sciences, Guizhou Medical University, 550004 Guiyang, People's Republic of China
| | - Nana Ma
- School of Pharmaceutical Sciences, Guizhou Medical University, 550004 Guiyang, People's Republic of China
| | - Xuan Zhou
- School of Pharmaceutical Sciences, Guizhou Medical University, 550004 Guiyang, People's Republic of China
| | - Yingjian Xu
- GoldenKeys High-tech Materials Co., Ltd., Guian New Area, Guizhou 550025, People's Republic of China
| | - Lin Zhang
- School of Pharmaceutical Sciences, Guizhou Medical University, 550004 Guiyang, People's Republic of China
| | - Chun Li
- School of Pharmaceutical Sciences, Guizhou Medical University, 550004 Guiyang, People's Republic of China
| |
Collapse
|
12
|
Huang Z, Li L, Cheng B, Li D. Small molecules targeting HDAC6 for cancer treatment: Current progress and novel strategies. Biomed Pharmacother 2024; 178:117218. [PMID: 39084081 DOI: 10.1016/j.biopha.2024.117218] [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: 05/25/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
Histone deacetylase 6 (HDAC6) plays a crucial role in the initiation and progression of various cancers, as its overexpression is linked to tumor growth, invasion, migration, survival, apoptosis, and angiogenesis. Therefore, HDAC6 has emerged as an attractive target for anticancer drug discovery in the past decade. However, the development of conventional HDAC6 inhibitors has been hampered by their limited clinical efficacy, acquired resistance, and inability to inhibit non-enzymatic functions of HDAC6. To overcome these challenges, new strategies, such as dual-acting inhibitors, targeted protein degradation (TPD) technologies (including PROTACs, HyT), are essential to enhance the anticancer activity of HDAC6 inhibitors. In this review, we focus on the recent advances in the design and development of HDAC6 modulators, including isoform-selective HDAC6 inhibitors, HDAC6-based dual-target inhibitors, and targeted protein degraders (PROTACs, HyT), from the perspectives of rational design, pharmacodynamics, pharmacokinetics, and clinical status. Finally, we discuss the challenges and future directions for HDAC6-based drug discovery for cancer therapy.
Collapse
Affiliation(s)
- Ziqian Huang
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, PR China
| | - Ling Li
- The Eighth Affiliated Hospital Sun Yat-sen University, 3025 Shennan Middle Road, Shenzhen 518000, China.
| | - Binbin Cheng
- School of Medicine, Hubei Polytechnic University, Huangshi 435003, China.
| | - Deping Li
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, PR China.
| |
Collapse
|
13
|
Kline GM, Madrazo N, Cole CM, Pannikkat M, Bollong MJ, Rosarda JD, Kelly JW, Wiseman RL. Metabolically activated proteostasis regulators that protect against erastin-induced ferroptosis. RSC Chem Biol 2024; 5:866-876. [PMID: 39211477 PMCID: PMC11353103 DOI: 10.1039/d4cb00027g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 07/04/2024] [Indexed: 09/04/2024] Open
Abstract
We previously showed that the proteostasis regulator compound AA147 (N-(2-hydroxy-5-methylphenyl)benzenepropanamide) potently protects against neurotoxic insults, such as glutamate-induced oxytosis. Though AA147 is a selective activator of the ATF6 arm of the unfolded protein response in non-neuronal cells, AA147-dependent protection against glutamate toxicity in cells of neuronal origin is primarily mediated through activation of the NRF2 oxidative stress response. AA147 activates NRF2 through a mechanism involving metabolic activation of AA147 by endoplasmic reticulum (ER) oxidases, affording an AA147-based quinone methide that covalently targets the NRF2 repressor protein KEAP1. Previous results show that the 2-amino-p-cresol A-ring of AA147 is required for NRF2 activation, while the phenyl B-ring of AA147 is amenable to modification. Here we explore whether the protease-sensitive amide linker between the A- and B-rings of this molecule can be modified to retain NRF2 activation. We show that replacement of the amide linker of AA147 with a carbamate linker retains NRF2 activation in neuronal cells and improves protection against neurotoxic insults, including glutamate-induced oxytosis and erastin-induced ferroptosis. Moreover, we demonstrate that inclusion of this carbamate linker facilitates identification of next-generation AA147 analogs with improved cellular tolerance and activity in disease-relevant assays.
Collapse
Affiliation(s)
- Gabriel M Kline
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
| | - Nicole Madrazo
- Department of Molecular and Cellular Biology, The Scripps Research Institute La Jolla CA 92037 USA
| | - Christian M Cole
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
| | - Meera Pannikkat
- Department of Molecular and Cellular Biology, The Scripps Research Institute La Jolla CA 92037 USA
| | - Michael J Bollong
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
| | - Jessica D Rosarda
- Department of Molecular and Cellular Biology, The Scripps Research Institute La Jolla CA 92037 USA
- Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences Bethesda MD 20814 USA
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute La Jolla CA 92037 USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute La Jolla CA 92037 USA
| | - R Luke Wiseman
- Department of Molecular and Cellular Biology, The Scripps Research Institute La Jolla CA 92037 USA
| |
Collapse
|
14
|
Tian S, Zhao Y, Deng S, Hou L, Song J, Wang M, Bu M. Lupeol-3-carbamate Derivatives: Synthesis and Biological Evaluation as Potential Antitumor Agents. Molecules 2024; 29:3990. [PMID: 39274838 PMCID: PMC11396318 DOI: 10.3390/molecules29173990] [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: 07/23/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
In the following study, a series of new lupeol-3-carbamate derivatives were synthesized, and the structures of all the newly derived compounds were characterized. The new compounds were screened to determine their anti-proliferative activity against human lung cancer cell line A549, human liver cancer cell line HepG2, and human breast cancer cell line MCF-7. Most of the compounds were found to show better anti-proliferative activity in vitro than lupeol. Among them, obvious anti-proliferation activity (IC50 = 5.39~9.43 μM) was exhibited by compound 3i against all three tumor cell lines. In addition, a salt reaction was performed on compound 3k (IC50 = 13.98 μM) and it was observed that the anti-proliferative activity and water solubility of compound 3k·CH3I (IC50 = 3.13 μM), were significantly enhanced subsequent to the salt formation process. The preliminary mechanistic studies demonstrated that apoptosis in HepG2 cells was induced by compound 3k·CH3I through the inhibition of the PI3K/AKT/mTOR pathway. In conclusion, a series of new lupeol-3-carbamate derivatives were synthesized via the structural modification of the C-3 site of lupeol, thus laying a theoretical foundation for the design of this new anticancer drug.
Collapse
Affiliation(s)
- Shuang Tian
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
| | - Yinxu Zhao
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
| | - Siqi Deng
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
| | - Liman Hou
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
| | - Juan Song
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
| | - Ming Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
| | - Ming Bu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China
| |
Collapse
|
15
|
Zhu W, Liu C, Xi K, Li A, Shen LA, Li Y, Jia M, He Y, Chen G, Liu C, Chen Y, Chen K, Sun F, Zhang D, Duan C, Wang H, Wang D, Zhao Y, Meng X, Zhu D. Discovery of Novel 1-Phenylpiperidine Urea-Containing Derivatives Inhibiting β-Catenin/BCL9 Interaction and Exerting Antitumor Efficacy through the Activation of Antigen Presentation of cDC1 Cells. J Med Chem 2024; 67:12485-12520. [PMID: 38912577 DOI: 10.1021/acs.jmedchem.3c02079] [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: 06/25/2024]
Abstract
Aberrant activation of the Wnt/β-catenin signaling is associated with tumor development, and blocking β-catenin/BCL9 is a novel strategy for oncogenic Wnt/β-catenin signaling. Herein, we presented two novel β-catenin variations and exposed conformational dynamics in several β-catenin crystal structures at the BCL9 binding site. Furthermore, we identified a class of novel urea-containing compounds targeting β-catenin/BCL9 interaction. Notably, the binding modalities of inhibitors were greatly affected by the conformational dynamics of β-catenin. Among them, 28 had a strong affinity for β-catenin (Kd = 82 nM), the most potent inhibitor reported. In addition, 13 and 35 not only activate T cells but also promote the antigen presentation of cDC1, showing robust antitumor efficacy in the CT26 model. Collectively, our study demonstrated a series of potent small-molecule inhibitors targeting β-catenin/BCL9, which can enhance antigen presentation and activate cDC1 cells, delivering a potential strategy for boosting innate and adaptive immunity to overcome immunotherapy resistance.
Collapse
Affiliation(s)
- Wenhua Zhu
- Anhui University of Chinese Medicine, Hefei 230012, China
- Yangtze Delta Drug Advanced Research Institute and Yangtze Delta Pharmaceutical College, Nantong 226133, China
| | - Cuiting Liu
- Anhui University of Chinese Medicine, Hefei 230012, China
- Yangtze Delta Drug Advanced Research Institute and Yangtze Delta Pharmaceutical College, Nantong 226133, China
| | - Kang Xi
- Anhui University of Chinese Medicine, Hefei 230012, China
- Yangtze Delta Drug Advanced Research Institute and Yangtze Delta Pharmaceutical College, Nantong 226133, China
| | - Anqi Li
- School of Basic Medical Sciences, Fudan University, Shanghai 201210, China
| | - Li-An Shen
- School of Basic Medical Sciences, Fudan University, Shanghai 201210, China
| | - Yana Li
- Anhui University of Chinese Medicine, Hefei 230012, China
- Yangtze Delta Drug Advanced Research Institute and Yangtze Delta Pharmaceutical College, Nantong 226133, China
| | - Miaomiao Jia
- Anhui University of Chinese Medicine, Hefei 230012, China
- Yangtze Delta Drug Advanced Research Institute and Yangtze Delta Pharmaceutical College, Nantong 226133, China
| | - Yangbo He
- Anhui University of Chinese Medicine, Hefei 230012, China
- Yangtze Delta Drug Advanced Research Institute and Yangtze Delta Pharmaceutical College, Nantong 226133, China
| | - Gang Chen
- Anhui University of Chinese Medicine, Hefei 230012, China
- Yangtze Delta Drug Advanced Research Institute and Yangtze Delta Pharmaceutical College, Nantong 226133, China
| | - Chenglong Liu
- School of Basic Medical Sciences, Fudan University, Shanghai 201210, China
| | - Yangqiang Chen
- Anhui University of Chinese Medicine, Hefei 230012, China
- Yangtze Delta Drug Advanced Research Institute and Yangtze Delta Pharmaceutical College, Nantong 226133, China
| | - Kai Chen
- Shanghai Jiao Tong University, Shanghai 201210, China
| | - Fan Sun
- Shanghai Jiao Tong University, Shanghai 201210, China
| | - Daizhou Zhang
- Shandong Academy of Pharmaceutical Science, Jinan 250101, China
| | - Chonggang Duan
- Shandong Academy of Pharmaceutical Science, Jinan 250101, China
| | - Heng Wang
- School of Basic Medical Sciences, Fudan University, Shanghai 201210, China
| | | | - Yujun Zhao
- State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd., Shanghai 201203, China
| | - Xiangjing Meng
- Shandong Academy of Pharmaceutical Science, Jinan 250101, China
| | - Di Zhu
- School of Basic Medical Sciences, Fudan University, Shanghai 201210, China
| |
Collapse
|
16
|
Xu J, Niu Y, Lin BL. Monomer-Recyclable Polyester from CO 2 and 1,3-Butadiene. Macromol Rapid Commun 2024; 45:e2400163. [PMID: 38690806 DOI: 10.1002/marc.202400163] [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: 03/19/2024] [Revised: 04/22/2024] [Indexed: 05/03/2024]
Abstract
Synthesis of monomer-recyclable polyesters solely from CO2 and bulk olefins holds great potential in significantly reducing CO2 emissions and addressing the issue of plastic pollution. Due to the kinetic disadvantage of direct copolymerization of CO2 and bulk olefins compared to homopolymerization of bulk olefins, considerable research attention has been devoted to synthesis of polyester via the ring-opening polymerization (ROP) of a six-membered disubstituted lactone intermediate, 1,2-ethylidene-6-vinyl-tetrahydro-2H-pyran-2-one (𝜹-L), obtained from telomerization of CO2 and 1,3-butadiene. However, the conjugate olefin on the six-membered ring of 𝜹-L leads to serious Michael addition side reactions. Thus, the selective ROP of 𝜹-L, which can precisely control the repeating unit for the production of polyesters potentially amenable to efficient monomer recycling, remains an unresolved challenge. Herein, the first example of selective ROP of 𝜹-L is reported using a combination of organobase and N,N'-Bis[3,5-bis(trifluoromethyl)phenyl]urea as the catalytic system. Systematic modifications of the substituent of the urea show that the presence of electron-deficient 3,5-bis(trifluoromethyl)-phenyl groups is the key to the extraordinary selectivity of ring opening over Michael addition. Efficient monomer recovery of oligo(𝜹-L) is also achieved under mild catalytic conditions.
Collapse
Affiliation(s)
- Jialin Xu
- School of Physical Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Yuxuan Niu
- School of Physical Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Bo-Lin Lin
- School of Physical Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| |
Collapse
|
17
|
Pallabothula VSK, Abdalrahman NT, Mori M, Fekri AH, Janďourek O, Konečná K, Paterová P, Novák M, Dudášová-Hatoková P, Štěrbová-Kovaříková P, Castellano C, Meneghetti F, Villa S, Kuneš J, Juhás M, Zitko J. A hit expansion of 3-benzamidopyrazine-2-carboxamide: Toward inhibitors of prolyl-tRNA synthetase with antimycobacterial activity. Arch Pharm (Weinheim) 2024; 357:e2400171. [PMID: 38710636 DOI: 10.1002/ardp.202400171] [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: 03/05/2024] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 05/08/2024]
Abstract
This study presents an exploration of the chemical space around derivatives of 3-benzamidopyrazine-2-carboxamides, previously identified as potent antimycobacterial compounds with predicted binding to mycobacterial prolyl-transfer RNA synthetase. New urea derivatives (Series-1) were generally inactive, probably due to their preference for cis-trans conformation (confirmed by density functional theory calculations and experimentally by nuclear overhauser effect spectroscopy NMR). Series-2 (3-benzamidopyrazine-2-carboxamides with disubstituted benzene ring) demonstrated that substituents larger than fluorine are not tolerated in the ortho position of the benzene ring. This series brought two new compounds (21: R = 2-F, 4-Cl and 22: R = 2-F, 4-Br) with in vitro activity against Mycobacterium tuberculosis H37Rv as well as multidrug-resistant clinical isolates, with minimum inhibitory concentration ranging from 6.25 to 25 μg/mL. The lactone-type derivatives 4H-pyrazino[2,3-d][1,3]oxazin-4-ones (Series-3) were inactive, but solvent stability studies of compound 29 indicated that they might be developed to usable lactone prodrugs of inhibitors of mycobacterial aspartate decarboxylase (PanD).
Collapse
Affiliation(s)
| | | | - Matteo Mori
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Amir Hossein Fekri
- Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Ondřej Janďourek
- Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Klára Konečná
- Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Pavla Paterová
- Department of Clinical Microbiology, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Martin Novák
- Biomedical Research Centre, University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | | | | | | | | | - Stefania Villa
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Jiří Kuneš
- Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| | - Martin Juhás
- Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
- Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Jan Zitko
- Faculty of Pharmacy in Hradec Králové, Charles University, Hradec Králové, Czech Republic
| |
Collapse
|
18
|
Sabale P, Sayyad N, Ali A, Sabale V, Kaleem M, Asar TO, Ali A, Mujtaba MA, Anwer MK. Design, synthesis, molecular docking and in vitro anticancer activities of 1-(4-(benzamido)phenyl)-3-arylurea derivatives. RSC Adv 2024; 14:23785-23795. [PMID: 39077323 PMCID: PMC11284930 DOI: 10.1039/d4ra02882a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024] Open
Abstract
In both premenopausal and postmenopausal women, oestrogens play a critical role in the development of breast cancer. Aromatase is an enzyme that catalyses the final step in the biosynthesis of estrogen and has emerged as a promising target for therapeutic intervention. This study aimed to design and evaluate novel 1-(4-(benzamido)phenyl)-3-arylurea derivatives as potential aromatase inhibitors. Through molecular docking, promising leads were identified and synthesized. Spectroscopic techniques confirmed their structural integrity. Cytotoxicity against various cancer cell lines was assessed using MTT assay. Docking investigations against the aromatase enzyme (3s7s) elucidated binding interactions and energies. Compound 6g, exhibiting a binding energy of -8.6 kcal mol-1 and interacting with ALA306 and THR310 residues, showed the most promising activity. It demonstrated GI50 values ranging from 14.46 μM, 13.97 μM, 11.35 μM, 11.58 μM, and 15.77 μM against A-498, NCI-H23, MDAMB-231, MCF-7, and A-549 respectively. Lastly, the physicochemical, and ADMET properties of the compound were predicted. These findings highlight the potential of 1-(4-(benzamido)phenyl)-3-arylureas as a new class of antitumor agents targeting aromatase. Their versatility and superior activity compared to standard chemotherapeutic agents, like doxorubicin, warrant further investigation for the development of broader-spectrum anticancer drugs.
Collapse
Affiliation(s)
- Prafulla Sabale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University Mahatma Jyotiba Fuley Shaikshanik Parisar Nagpur-440033 India +919158537050
| | - Nusrat Sayyad
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University Mahatma Jyotiba Fuley Shaikshanik Parisar Nagpur-440033 India +919158537050
| | - Abuzer Ali
- Department of Pharmacognosy, College of Pharmacy, Taif University P.O. Box 11099 Taif 21944 Saudi Arabia
| | - Vidya Sabale
- Department of Pharmaceutics, Dadasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University Nagpur Maharashtra 440037 India
| | - Mohammed Kaleem
- Department of Pharmacology, Dadasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University Nagpur Maharashtra 440037 India
| | - Turky Omar Asar
- Department of Biology, College of Science and Arts at Alkamil, University of Jeddah Saudi Arabia
| | - Amena Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University P.O. Box 11099 Taif 21944 Saudi Arabia
| | - Md Ali Mujtaba
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University Arar Saudi Arabia
| | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University P.O. Box 173 Al-Kharj 11942 Saudi Arabia
| |
Collapse
|
19
|
Wang Z, He Y, Wang F, Wang Y, Luo H, Wu J, Yang J. Green and efficient synthesis of dibenzyl cyanamides and ureas with cyanamide as a block. RSC Adv 2024; 14:23693-23698. [PMID: 39077314 PMCID: PMC11284761 DOI: 10.1039/d4ra04286g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/16/2024] [Indexed: 07/31/2024] Open
Abstract
A method for the two-step synthesis of dibenzyl cyanamide and dibenzyl urea via cyanamide is presented. This approach is both efficient and environmentally friendly. Various N,N-dibenzyl ureas could be obtained by reactions of N,N-dibenzyl cyanamides and N,N-dibenzyl cyanamides as intermediates formed from cyanamide. In the absence of metal, ligand and hydrogen peroxide as the oxidant, products with moderate yields have been obtained under mild conditions. Key features include the use of widely available and easily handled cyanamide sources as starting materials.
Collapse
Affiliation(s)
- Zhongjie Wang
- State Key Laboratory of High-Efficiency Utilization of Coal and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University Yinchuan 750021 China
| | - Yu He
- State Key Laboratory of High-Efficiency Utilization of Coal and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University Yinchuan 750021 China
| | - Fang Wang
- State Key Laboratory of High-Efficiency Utilization of Coal and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University Yinchuan 750021 China
| | - Yan Wang
- State Key Laboratory of High-Efficiency Utilization of Coal and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University Yinchuan 750021 China
| | - Hui Luo
- State Key Laboratory of High-Efficiency Utilization of Coal and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University Yinchuan 750021 China
| | - Jianglong Wu
- School of Chemistry and Chemical Engineering, Ningxia Normal University Guyuan 756000 China
| | - Jinhui Yang
- State Key Laboratory of High-Efficiency Utilization of Coal and Green Chemical Engineering, School of Chemistry and Chemical Engineering, Ningxia University Yinchuan 750021 China
| |
Collapse
|
20
|
Foster DJ, Dunnavant K, Shrader CW, LoPresti M, Seay S, Kharel Y, Brown AM, Huang T, Lynch KR, Santos WL. Discovery of Potent, Orally Bioavailable Sphingosine-1-Phosphate Transporter (Spns2) Inhibitors. J Med Chem 2024; 67:11273-11295. [PMID: 38952222 PMCID: PMC11247503 DOI: 10.1021/acs.jmedchem.4c00879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Targeting the S1P pathway has resulted in the development of S1P1 receptor modulators for the treatment of multiple sclerosis and ulcerative colitis. We hypothesize that targeting an upstream node of the S1P pathway may provide an improved adverse event profile. In this report, we performed a structure-activity relationship study focusing on the benzoxazole scaffold in SLB1122168, which lead to the discovery of 11i (SLF80821178) as a potent inhibitor of S1P release from HeLa cells (IC50: 51 ± 3 nM). Administration of SLF80821178 to mice induced ∼50% reduction in circulating lymphocyte counts, recapitulating the lymphopenia characteristic of Spns2 null animals. Molecular modeling studies suggest that SLF80821178 binds Spns2 in its occluded inward-facing state and forms hydrogen bonds with Asn112 and Ser211 and π stacking with Phe234. Taken together, SLF80821178 can serve as a scaffold for future inhibitor development and represents a chemical tool to study the therapeutic implication of inhibiting Spns2.
Collapse
Affiliation(s)
- Daniel J Foster
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Kyle Dunnavant
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Christopher W Shrader
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Marion LoPresti
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Sarah Seay
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Anne M Brown
- Department of Biochemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Tao Huang
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Kevin R Lynch
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Webster L Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
21
|
Zhang Q, Soulère L, Queneau Y. Amide bioisosteric replacement in the design and synthesis of quorum sensing modulators. Eur J Med Chem 2024; 273:116525. [PMID: 38801798 DOI: 10.1016/j.ejmech.2024.116525] [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: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
The prevention or control of bacterial infections requires continuous search for novel approaches among which bacterial quorum sensing inhibition is considered as a complementary antibacterial strategy. Quorum sensing, used by many different bacteria, functions through a cell-to-cell communication mechanism relying on chemical signals, referred to as autoinducers, such as N-acyl homoserine lactones (AHLs) which are the most common chemical signals in this system. Designing analogs of these autoinducers is one of the possible ways to interfere with quorum sensing. Since bioisosteres are powerful tools in medicinal chemistry, targeting analogs of AHLs or other signal molecules and mimics of known QS modulators built on amide bond bioisosteres is a relevant strategy in molecular design and synthetic routes. This review highlights the application of amide bond bioisosteric replacement in the design and synthesis of novel quorum sensing inhibitors.
Collapse
Affiliation(s)
- Qiang Zhang
- Hubei Key Laboratory of Purification and Application of Plant Anti-cancer Active Ingredients, Hubei University of Education, 129 Second Gaoxin Road, Wuhan 430205, China
| | - Laurent Soulère
- INSA Lyon, CNRS, Universite Claude Bernard Lyon 1, UMR 5246, ICBMS, Bât. E. Lederer, 1 rue Victor Grignard, F-69622, Villeurbanne, France
| | - Yves Queneau
- INSA Lyon, CNRS, Universite Claude Bernard Lyon 1, UMR 5246, ICBMS, Bât. E. Lederer, 1 rue Victor Grignard, F-69622, Villeurbanne, France.
| |
Collapse
|
22
|
Sroor FM, Soliman AAF, Youssef EM, Abdelraof M, El-Sayed AF. Green, facile synthesis and evaluation of unsymmetrical carbamide derivatives as antimicrobial and anticancer agents with mechanistic insights. Sci Rep 2024; 14:15441. [PMID: 38965246 PMCID: PMC11224357 DOI: 10.1038/s41598-024-65308-6] [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: 11/09/2023] [Accepted: 06/19/2024] [Indexed: 07/06/2024] Open
Abstract
A very practical method for the synthesis of unsymmetrical carbamide derivatives in good to excellent yield was presented, without the need for any catalyst and at room temperature. Using a facile and robust protocol, fifteen unsymmetrical carbamide derivatives (9-23) bearing different aliphatic amine moieties were designed and synthesized by the reaction of secondary aliphatic amines with isocyanate derivatives in the presence of acetonitrile as an appropriate solvent in good to excellent yields. Trusted instruments like IR, mass spectrometry, NMR spectra, and elemental analyses were employed to validate the purity and chemical structures of the synthesized compounds. All the synthesized compounds were tested as antimicrobial agents against some clinically bacterial pathogens such as Salmonella typhimurium, Bacillus subtilis, Pseudomonas aeruginosa, Staphylococcus aureus and Candida albicans. Compounds 15, 16, 17, 19 and 22 showed potent antimicrobial activity with promising MIC values compared to the positive controls. Moreover, compounds 15 and 22 provide a potent lipid peroxidation (LPO) of the bacterial cell wall. On the other hand, we investigated the anti-proliferative activity of compounds 9-23 against selected human cancerous cell lines of breast (MCF-7), colon (HCT-116), and lung (A549) relative to healthy noncancerous control skin fibroblast cells (BJ-1). The mechanism of their cytotoxic activity has been also examined by immunoassaying the levels of key anti- and pro-apoptotic protein markers. The results of MTT assay revealed that compounds 10, 13, 21, 22 and 23 possessed highly cytotoxic effects. Out of these, three synthesized compounds 13, 21 and 22 showed cytotoxicity with IC50 values (13, IC50 = 62.4 ± 0.128 and 22, IC50 = 91.6 ± 0.112 µM, respectively, on MCF-7), (13, IC50 = 43.5 ± 0.15 and 21, IC50 = 38.5 ± 0.17 µM, respectively, on HCT-116). Cell cycle and apoptosis/necrosis assays demonstrated that compounds 13 and 22 induced S and G2/M phase cell cycle arrest in MCF-7 cells, while only compound 13 had this effect on HCT-116 cells. Furthermore, compound 13 exhibited the greatest potency in inducing apoptosis in both cell lines compared to compounds 21 and 22. Docking studies indicated that compounds 10, 13, 21 and 23 could potentially inhibit enzymes and exert promising antimicrobial effects, as evidenced by their lower binding energies and various types of interactions observed at the active sites of key enzymes such as Sterol 14-demethylase of C. albicans, Dihydropteroate synthase of S. aureus, LasR of P. aeruginosa, Glucosamine-6-phosphate synthase of K. pneumenia and Gyrase B of B. subtilis. Moreover, 13, 21, and 22 demonstrated minimal binding energy and favorable affinity towards the active pocket of anticancer receptor proteins, including CDK2, EGFR, Erα, Topoisomerase II and VEGFFR. Physicochemical properties, drug-likeness, and ADME (absorption, distribution, metabolism, excretion, and toxicity) parameters of the selected compounds were also computed.
Collapse
Affiliation(s)
- Farid M Sroor
- Organometallic and Organometalloid Chemistry Department, National Research Centre, Cairo, 12622, Egypt.
| | - Ahmed A F Soliman
- Pharmacognosy Department, National Research Centre, Dokki, 12622, Egypt
| | | | - Mohamed Abdelraof
- Microbial Chemistry Department, Biotechnology Research Institute, National Research Centre, Giza, Egypt
| | - Ahmed F El-Sayed
- Microbial Genetics Department, Biotechnology Research Institute, National Research Centre, Giza, Egypt
- Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| |
Collapse
|
23
|
Ngoc Toan V, Son Hai D, Thi Kim Van H, Minh Tri N, Ngoc Toan D, Thi Thanh Mai N, Dinh Thanh N. Design, synthesis, inhibitory activity, and molecular simulations study for d-glucose-conjugated thioureas containing pyrimidine ring as multitarget inhibitors against α-amylase, α-glucosidase, DDP-4, and PTP1B in Type 2 diabetes mellitus. RSC Med Chem 2024:d4md00334a. [PMID: 39185455 PMCID: PMC11342126 DOI: 10.1039/d4md00334a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 06/24/2024] [Indexed: 08/27/2024] Open
Abstract
A series of tetra-O-acetyl-α-d-glucopyranosyl thioureas 8a-l of substituted 2-aminopyrimidines 4a-l have been designed and synthesized. The latter were prepared from corresponding chalcones 3a-l of p-bromoacetophenone and appropriate substituted benzaldehydes by their reaction with guanidine. The target thiourea compounds 8a-l exhibited significant inhibitory activity in vitro against enzymes that were related to type 2 diabetes mellitus, including α-amylase, α-glucosidase, DPP-4, and PTP1B. Amongst these thioureas, compound 8k with an ortho-methoxy group was the most potential enzyme inhibitor against α-amylase with an IC50 value of 9.72 ± 0.34 μM. Its meta-isomer 8j was the strongest inhibitor against α-glucosidase with IC50 = 9.73 ± 0.72 μM. In the inhibition against DPP-4, compound 8f with a para-bromo substituent exhibited the strongest activity with an IC50 value of 2.53 ± 0.03 nM. In the inhibition against PTP1B, compound 8h with a para-isopropyl substituent had the strongest inhibitory activity with an IC50 value of 2.74 ± 0.03 μM. The enzyme kinetics of the most active compounds, including 8j, 8f and 8h against α-glucosidase, DPP-4, and PTP1B, respectively, were studied. The obtained results showed that 8j was a competitive α-glucosidase inhibitor with an inhibitory constant K I value of 9.31 μM. Compound 8f was a non-competitive inhibitor for DDP-4 with an inhibitory constant K I value of 12.57 μM. Compound 8h was also a non-competitive inhibitor for DDP-4 with an inhibitory constant K I value of 12.41 μM. The cytotoxicity of the most active compounds, including 8f and 8k (against α-amylase), 8i and 8j (against α-glucosidase), 8a, 8f, and 8g (against DPP-4), and 8d, 8f, and 8h (against PTP1B) was screened. The obtained cytotoxicity showed that all tested inhibitors were noncytotoxic to human normal cell line 3T3. Induced fit docking simulations of all synthesized compounds 8a-l were performed on four enzymes 4W93 (for α-amylase), 3TOP (for α-glucosidase), 3W2T (for DPP-4), and 1NNY (for PTP1B). Key interactions of each of these ligands with residues in the active pocket of each studied enzyme have been shown.
Collapse
Affiliation(s)
- Vu Ngoc Toan
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi) 19 Le Thanh Tong, Hoan Kiem Hanoi Vietnam
- Academy of Military Science and Technology, Ministry of Defence, Institute of New Technology 17 Hoang Sam, Cau Giay Hanoi Vietnam
| | - Do Son Hai
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi) 19 Le Thanh Tong, Hoan Kiem Hanoi Vietnam
- Ministry of Public Security of Vietnam, Institute of Science and Technology 47 Pham Van Dong, Cau Giay Hanoi Vietnam
| | - Hoang Thi Kim Van
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi) 19 Le Thanh Tong, Hoan Kiem Hanoi Vietnam
- Faculty of Chemical Technology, Viet Tri University of Industry Tien Kien, Lam Thao Phu Tho Vietnam
| | - Nguyen Minh Tri
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi) 19 Le Thanh Tong, Hoan Kiem Hanoi Vietnam
- Academy of Military Science and Technology, Ministry of Defence, Institute of New Technology 17 Hoang Sam, Cau Giay Hanoi Vietnam
| | - Duong Ngoc Toan
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi) 19 Le Thanh Tong, Hoan Kiem Hanoi Vietnam
- Faculty of Chemistry, Thai Nguyen University of Education 20 Luong Ngoc Quyen Thai Nguyen Vietnam
| | - Nguyen Thi Thanh Mai
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi) 19 Le Thanh Tong, Hoan Kiem Hanoi Vietnam
- Faculty of Chemical Technology, Ha Noi University of Industry 298 Cau Dien Road, North Tu Liem Hanoi Vietnam
| | - Nguyen Dinh Thanh
- Faculty of Chemistry, University of Science (Vietnam National University, Hanoi) 19 Le Thanh Tong, Hoan Kiem Hanoi Vietnam
| |
Collapse
|
24
|
Bonnemaire CM, Windhorst AD, Orru R, Ruijter E, Vugts DJ. [11C]CO2 BOP fixation with amines to access 11C-labeled ureas for PET imaging. J Labelled Comp Radiopharm 2024; 67:201-210. [PMID: 38073118 DOI: 10.1002/jlcr.4075] [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: 08/18/2023] [Revised: 10/11/2023] [Accepted: 11/03/2023] [Indexed: 05/16/2024]
Abstract
Carbon-11 (11C) is a widely used radionuclide for positron emission tomography (PET) owing to the omnipresence of carbon atoms in organic molecules. While its half-life of 20.4 min is ideal for imaging and dosimetry, it also limits the synthetic possibilities. As such, the development of fast and easy, high-yielding synthesis methods is crucial for the application of 11C-labeled tracers in humans. In this study, we present a novel and efficient method for the reaction of [11C]CO2 with amine precursors using benzotriazole-1-yl-oxy-tris-(dimethylamino)-phosphonium hexafluorophosphate (BOP) to access 11C-labeled ureas. Our method is extremely fast as it only requires transfer of [11C]CO2 into a solution with precursor and BOP at room temperature, where it reacts momentary into the desired 11C-labeled urea. This simple procedure makes it possible to radiolabel urea directly from [11C]CO2 without the need for advanced equipment, making the method applicable for all laboratories where [11C]CO2 is available. We synthesized a small series of aliphatic symmetrical and non-symmetrical 11C-labeled ureas using this method, and achieved good to excellent yields. The novelty of our study lies in the fact that peptide coupling reagent BOP is used for the first time in radiochemistry to activate [11C]CO2, facilitating its reaction with amines to obtain 11C-labeled ureas.
Collapse
Affiliation(s)
- Coralie M Bonnemaire
- Radiology and Nuclear Medicine(s), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Radiology and Nuclear Medicine(s), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Romano Orru
- Bio-based Organic Chemistry, Aachen-Maastricht Institute for Biobased Materials (AMIBM), Maastricht University, Geleen, The Netherlands
| | - Eelco Ruijter
- Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecular and Life Sciences (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Danielle J Vugts
- Radiology and Nuclear Medicine(s), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Hipólito J, Meyrelles R, Maryasin B, Alves LG, Martins AM. Reactions of Heteroallenes with Salan-based Ti(IV) Complexes: A Joint Experimental and Computational Study. Chem Asian J 2024; 19:e202400165. [PMID: 38403858 DOI: 10.1002/asia.202400165] [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: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
The reaction of Ti(NMe2)4 with the salan ligand precursor H2N2O2H2 led to the formation of [(L*)Ti(NHMe2)2] (L*=N2O2 4-) that forms [(H2N2O2)TiCl2] upon reaction with two equiv. of Me3SiCl. [(L*)Ti(py)2] was obtained from the reaction of [Ti(NtBu)Cl2(py)3] with the sodium salt H2N2O2Na2. Treatment of [(L*)Ti(NHMe2)2] with two equiv. of tBuNCO led to the insertion of the isocyanate molecules into the Ti-Nsalan bonds with the formation of [{L*(N(tBu)CO)2}Ti]. Conversely, the reaction of [(H2N2O2)Ti(OiPr)2] with two equiv. of tBuNCO led to the insertion of one isocyanate molecule into a Ti-Nsalan bond with the formation of [{(HN2O2)(N(tBu)CO)}Ti(OiPr)]. Computational studies were performed to gain insight into the reactivity of isocyanates with salan-based Ti(IV) complexes.
Collapse
Affiliation(s)
- Joana Hipólito
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisboa, Portugal
| | - Ricardo Meyrelles
- Institute of Organic Chemistry, University of Vienna, Währinger Strasse 38, 1090, Vienna, Austria
- Institute of Theoretical Chemistry, University of Vienna, Währinger Strasse 17, 1090, Vienna, Austria
- Doctoral School in Chemistry, University of Vienna, Währinger Strasse 42, 1090, Vienna, Austria
| | - Boris Maryasin
- Institute of Organic Chemistry, University of Vienna, Währinger Strasse 38, 1090, Vienna, Austria
- Institute of Theoretical Chemistry, University of Vienna, Währinger Strasse 17, 1090, Vienna, Austria
| | - Luis G Alves
- Centro de Química Estrutural - Institute of Molecular Sciences, Associação do Instituto Superior Técnico para a Investigação e Desenvolvimento, Av. António José de Almeida Lisboa, n°12, 1000-043, Lisboa, Portugal
| | - Ana M Martins
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisboa, Portugal
| |
Collapse
|
26
|
Yin F, Qu L, Chen Y, Luo Z, Kong L, Wang X. Stereoselective Synthesis of β, γ-Fused Bicyclic γ-Ureasultams via an Intramolecular Mannich and aza-Michael Addition Cascade. Chemistry 2024; 30:e202400438. [PMID: 38470414 DOI: 10.1002/chem.202400438] [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: 01/31/2024] [Revised: 03/12/2024] [Accepted: 03/12/2024] [Indexed: 03/13/2024]
Abstract
A novel approach has been developed for the synthesis of bicyclic β, γ-fused bicyclic γ-ureasultams containing two consecutive chiral centers through an intramolecular Mannich and aza-Michael addition cascade of alkenyl sulfamides. The straightforward practical procedure and readily available starting materials enable the synthesis of variously substituted ureasultams. In addition, bicyclic γ-ureasultams is a class of potential biotin analogues.
Collapse
Affiliation(s)
- Fucheng Yin
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, People's Republic of China
| | - Lailiang Qu
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, People's Republic of China
- Xinyang Normal University, Xinyang, 464000, People's Republic of China
| | - Yifan Chen
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, People's Republic of China
| | - Zhongwen Luo
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, People's Republic of China
| | - Lingyi Kong
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, People's Republic of China
| | - Xiaobing Wang
- China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, People's Republic of China
| |
Collapse
|
27
|
Duan L, Chu C, Huang X, Yao H, Wen J, Chen R, Wang C, Tu Y, Lv Q, Pan Q, Xu S. Rational design and synthesis of 2,4-dichloro-6-methyl pyrimidine derivatives as potential selective EGFR T790M/L858R inhibitors for the treatment of non-small cell lung cancer. Arch Pharm (Weinheim) 2024; 357:e2300736. [PMID: 38381049 DOI: 10.1002/ardp.202300736] [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: 12/14/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/22/2024]
Abstract
Many patients with non-small cell lung cancer (NSCLC) initially benefit from epidermal growth factor receptor (EGFR) targeted therapy. Unfortunately, varying degrees of resistance or side effects eventually develop. Overcoming and preventing the resistance and side effects of EGFR inhibitors has become a hot topic of research today. Based on the previous studies on AZD-9291, we designed and synthesized two series of 2,4-dichloro-6-methylpyrimidine derivatives, 19 compounds in total, as potential inhibitors of the EGFR kinase. The most promising compound, L-18, showed better inhibitory activity (81.9%) and selectivity against EGFRT790M/L858R kinase. In addition, L-18 showed strong antiproliferative activity against H1975 cells with an IC50 value of 0.65 ± 0.06 μM and no toxicity to normal cells (LO-2). L-18 was able to dose-dependently induce the apoptosis of H1975 cells and produced a cell-cycle-blocking effect, and it can also dose-dependently inhibit the migration and invasion of H1975 cells. L-18 also showed in vivo anticancer efficacy in H1975 cells xenograft mice. We also performed a series of in vivo and in vitro toxicological evaluations of compound L-18, which did not cause obvious injury in mice during administration. These results suggest that L-18 may be a promising drug candidate that warrants further investigation.
Collapse
Affiliation(s)
- Lei Duan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Cilong Chu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Xiaoling Huang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Huizhi Yao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Jie Wen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Rui Chen
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Caolin Wang
- School of Pharmacy, East China University of Science & Technology, Shanghai, China
| | - Yuanbiao Tu
- Cancer Research Center, Jangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Qiaoli Lv
- Jiangxi Key Laboratory of Translational Cancer Research, Jiangxi Cancer Hospital, Nanchang, Jiangxi, People's Republic of China
| | - Qingshan Pan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Science & Technology Normal University, Nanchang, Jiangxi, China
| |
Collapse
|
28
|
Pavić K, Poje G, Pessanha de Carvalho L, Tandarić T, Marinović M, Fontinha D, Held J, Prudêncio M, Piantanida I, Vianello R, Krošl Knežević I, Perković I, Rajić Z. Discovery of harmiprims, harmine-primaquine hybrids, as potent and selective anticancer and antimalarial compounds. Bioorg Med Chem 2024; 105:117734. [PMID: 38677112 DOI: 10.1016/j.bmc.2024.117734] [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: 03/17/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Although cancer and malaria are not etiologically nor pathophysiologically connected, due to their similarities successful repurposing of antimalarial drugs for cancer and vice-versa is known and used in clinical settings and drug research and discovery. With the growing resistance of cancer cells and Plasmodium to the known drugs, there is an urgent need to discover new chemotypes and enrich anticancer and antimalarial drug portfolios. In this paper, we present the design and synthesis of harmiprims, hybrids composed of harmine, an alkaloid of the β-carboline type bearing anticancer and antiplasmodial activities, and primaquine, 8-aminoquinoline antimalarial drug with low antiproliferative activity, covalently bound via triazole or urea. Evaluation of their antiproliferative activities in vitro revealed that N-9 substituted triazole-type harmiprime was the most selective compound against MCF-7, whereas C1-substituted ureido-type hybrid was the most active compound against all cell lines tested. On the other hand, dimeric harmiprime was not toxic at all. Although spectrophotometric studies and thermal denaturation experiments indicated binding of harmiprims to the ds-DNA groove, cell localization showed that harmiprims do not enter cell nucleus nor mitochondria, thus no inhibition of DNA-related processes can be expected. Cell cycle analysis revealed that C1-substituted ureido-type hybrid induced a G1 arrest and reduced the number of cells in the S phase after 24 h, persisting at 48 h, albeit with a less significant increase in G1, possibly due to adaptive cellular responses. In contrast, N-9 substituted triazole-type harmiprime exhibited less pronounced effects on the cell cycle, particularly after 48 h, which is consistent with its moderate activity against the MCF-7 cell line. On the other hand, screening of their antiplasmodial activities against the erythrocytic, hepatic, and gametocytic stages of the Plasmodium life cycle showed that dimeric harmiprime exerts powerful triple-stage antiplasmodial activity, while computational analysis showed its binding within the ATP binding site of PfHsp90.
Collapse
Affiliation(s)
- Kristina Pavić
- University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10000 Zagreb, Croatia.
| | - Goran Poje
- University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10000 Zagreb, Croatia
| | | | - Tana Tandarić
- Department of Cell and Molecular Biology, Uppsala University, 75124 Uppsala, Sweden; Rudjer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Marina Marinović
- University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10000 Zagreb, Croatia
| | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Jana Held
- University of Tübingen, Institute of Tropical Medicine, Wilhelmstraße 27, 72074 Tübingen, Germany; German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal
| | - Ivo Piantanida
- Rudjer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Robert Vianello
- Rudjer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | | | - Ivana Perković
- University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10000 Zagreb, Croatia
| | - Zrinka Rajić
- University of Zagreb Faculty of Pharmacy and Biochemistry, A. Kovačića 1, 10000 Zagreb, Croatia.
| |
Collapse
|
29
|
Luo Y, Zhai B, Li M, Zhou W, Yang J, Shu Y, Fang Y. Self-adhesive, surface adaptive, regenerable SERS substrates for in-situ detection of urea on bio-surfaces. J Colloid Interface Sci 2024; 660:513-521. [PMID: 38262178 DOI: 10.1016/j.jcis.2024.01.068] [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: 11/08/2023] [Revised: 12/18/2023] [Accepted: 01/10/2024] [Indexed: 01/25/2024]
Abstract
Wearable SERS substrates have gained substantial attention for health monitoring and other applications. Current designs often rely on conventional polymer substrates, leading to discomfort and complexity due to the need of additional adhesive layers. To address the issues, we fabricate a flexible, uniform, ultrathin, transparent and porous SERS substrate via depositing Ag nanoparticles (AgNPs) onto the CdS nanowires (CdSNWs) grown on the surface of a prepared nanofilm (AgNPs-CdSNWs/nanofilm). Unlike the wearable SERS substrates reported in literature, the one presented in this work is self-adhesive to a variety of surfaces, which simplifies structure, enhances comfort and improves performance. Importantly, the new SERS substrate as developed is highly stable and reusable. Artificial sample tests revealed that the substrate showed a great enhancement factor (EF) of 4.2 × 107 and achieved a remarkable detection limit (DL) of 1.0 × 10-14 M for rhodamine 6G (R6G), which are among the highest records observed in wearable SERS substrates reported in literature. Moreover, the substrate enables at real-time and in-situ reliable monitoring of urea dynamics in human sweat and plant leaves, indicating its applicability for health analysis and in precision agriculture.
Collapse
Affiliation(s)
- Yan Luo
- Key Laboratory of Applied Surface and Colloid Chemistry of Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Binbin Zhai
- Key Laboratory of Applied Surface and Colloid Chemistry of Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Min Li
- Key Laboratory of Applied Surface and Colloid Chemistry of Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Wenjingli Zhou
- Key Laboratory of Applied Surface and Colloid Chemistry of Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China
| | - Jinglun Yang
- Department of Materials Science and Engineering, City University of Hong Kong, Hong Kong SAR 999077, China
| | - Yuanhong Shu
- Key Laboratory of Applied Surface and Colloid Chemistry of Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
| | - Yu Fang
- Key Laboratory of Applied Surface and Colloid Chemistry of Ministry of Education, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
30
|
Liu Y, Zhou Q, Huo Y, Sun X, Hu J. Recent advances in developing modified C14 side chain pleuromutilins as novel antibacterial agents. Eur J Med Chem 2024; 269:116313. [PMID: 38503168 DOI: 10.1016/j.ejmech.2024.116313] [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: 12/25/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/21/2024]
Abstract
Owing to the increasing resistance to most existing antimicrobial drugs, research has shifted towards developing novel antimicrobial agents with mechanisms of action distinct from those of current clinical options. Pleuromutilins are antibiotics known for their distinct mechanism of action, inhibiting bacterial protein synthesis by binding to the peptidyl transferase center of the ribosome. Recent studies have revealed that pleuromutilin derivatives can disrupt bacterial cell membranes, thereby enhancing antibacterial efficacy. Both marketed pleuromutilin derivatives and those in clinical trials have been developed by structurally modifying the pleuromutilin C14 side chain to improve their antimicrobial activity. Therefore, this review aims to review advancement in the chemical structural characteristics, antibacterial activities, and structure-activity relationship studies of pleuromutilins, specifically focusing on modifications made to the C14 side chain in recent years. These findings provide a valuable reference for future research and development of pleuromutilins.
Collapse
Affiliation(s)
- Yue Liu
- Shandong Second Medical University, No.7166 Baotong Road, Weifang, 261053, PR China
| | - Qinjiang Zhou
- Shandong Second Medical University, No.7166 Baotong Road, Weifang, 261053, PR China
| | - Yiwen Huo
- Shandong Second Medical University, No.7166 Baotong Road, Weifang, 261053, PR China
| | - Xiujuan Sun
- Shandong Second Medical University, No.7166 Baotong Road, Weifang, 261053, PR China
| | - Jinxing Hu
- Shandong Second Medical University, No.7166 Baotong Road, Weifang, 261053, PR China.
| |
Collapse
|
31
|
Berlin CB, Sharma E, Kozlowski MC. Quantification of Hydrogen-Bond-Donating Ability of Biologically Relevant Compounds. J Org Chem 2024; 89:4684-4690. [PMID: 38483838 PMCID: PMC11305090 DOI: 10.1021/acs.joc.3c02939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Hydrogen bonding is a key factor in the design of ligands for biological binding, including drug targets. Our group previously developed a method for experimentally assessing the hydrogen-bond-donating ability of an analyte using UV-vis titrations with a colorimetric sensor. Using this method, 79 new titrations were performed on weak hydrogen-bond donors, with a focus on heterocycles and pharmaceutically relevant motifs. The hydrogen-bond donating abilities of drug compounds and the substructures of drug compounds were also measured. These titrations will be used to build a database of hydrogen-bond donors.
Collapse
Affiliation(s)
- Cameron B. Berlin
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6223, United States
| | - Eesha Sharma
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6223, United States
| | - Marisa C. Kozlowski
- Department of Chemistry, Roy and Diana Vagelos Laboratories, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6223, United States
| |
Collapse
|
32
|
Xu Y, Lv J, Kong C, Liu Y, Wang K, Tang Z, Chen X. Introducing urea into tirapazamine derivatives to enhance anticancer therapy. Natl Sci Rev 2024; 11:nwae038. [PMID: 38440219 PMCID: PMC10911816 DOI: 10.1093/nsr/nwae038] [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: 09/25/2023] [Revised: 12/20/2023] [Accepted: 01/25/2024] [Indexed: 03/06/2024] Open
Abstract
Tirapazamine (TPZ) has been approved for multiple clinical trials relying on its excellent anticancer potential. However, as a typical hypoxia-activated prodrug (HAP), TPZ did not exhibit survival advantages in Phase III clinical trials when used in combination therapy due to the insufficient hypoxia levels in patients' tumors. In this study, to improve the therapeutic effects of TPZ, we first introduced urea to synthesize a series of urea-containing derivatives of TPZ. All urea-containing TPZ derivatives showed increased hypoxic cytotoxicity (9.51-30.85-fold) compared with TPZ, while maintaining hypoxic selectivity. TPZP, one of these derivatives, showed 20-fold higher cytotoxicity than TPZ while maintaining a similar hypoxic cytotoxicity ratio. To highly efficiently deliver TPZP to the tumors and reduce its side effects on healthy tissues, we further prepared TPZP into a nanodrug with fibrin-targeting ability: FT11-TPZP-NPs. CA4-NPs, a vascular disrupting agent, was used to increase the fibrin level within tumors and exacerbate tumor hypoxia. By being combined with CA4-NPs, FT11-TPZP-NPs can accumulate in the hypoxia-aggravated tumors and activate sufficiently to kill tumor cells. After a single-dose treatment, FT11-TPZP-NPs + CA4-NPs showed a high inhibition rate of 98.1% against CT26 tumor models with an initial volume of ∼480 mm3 and four out of six tumors were completely eliminated; it thereby exerted a significant antitumor effect. This study provides a new strategy for improving the therapeutic effect of TPZ and other HAPs in anticancer therapy.
Collapse
Affiliation(s)
- Yajun Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jianlin Lv
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Chaoying Kong
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Ya Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Kun Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
33
|
Cai B, Xu Y, Luo R, Lu K, Wang Y, Zheng L, Zhang Y, Yin L, Tu L, Luo W, Zheng L, Zhang F, Lv X, Tang Q, Liang G, Chen L. Discovery of a doublecortin-like kinase 1 inhibitor to prevent inflammatory responses in acute lung injury. Bioorg Chem 2024; 145:107215. [PMID: 38394920 DOI: 10.1016/j.bioorg.2024.107215] [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: 12/24/2023] [Revised: 02/07/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024]
Abstract
Doublecortin-like kinase 1 (DCLK1) is a microtubule-associated protein kinase involved in neurogenesis and human cancer. Recent studies have revealed a novel functional role for DCLK1 in inflammatory signaling, thus positioning it as a novel target kinase for respiratory inflammatory disease treatment. In this study, we designed and synthesized a series of NVP-TAE684-based derivatives as novel anti-inflammatory agents targeting DCLK1. Bio-layer interferometry binding screening and kinase assays of the NVP-TAE684 derivatives led to the discovery of an effective DCLK1 inhibitor (a24), with an IC50 of 179.7 nM. Compound a24 effectively inhibited lipopolysaccharide (LPS)-induced inflammation in macrophages with higher potency than the lead compound. Mechanistically, compound a24 inhibited LPS-induced inflammation by inhibiting DCLK1-mediated IKKβ phosphorylation. Furthermore, compound a24 showed in vivo anti-inflammatory activity in an LPS-challenged acute lung injury model. These findings suggest that compound a24 may serve as a novel candidate for the development of DCLK1 inhibitors and a potential therapeutic agent for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Binhao Cai
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ying Xu
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China
| | - Ruixiang Luo
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China
| | - Kongqin Lu
- Schol of Basic Medicine, Inner Mongolia Medical University, Hohhot 010059, China
| | - Yuhan Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China
| | - Lei Zheng
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China
| | - Yawen Zhang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China
| | - Lina Yin
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China
| | - Linglan Tu
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lulu Zheng
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang 310000, China
| | - Fengzhi Zhang
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China
| | - Xinting Lv
- School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China
| | - Qidong Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Guang Liang
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China; Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Lingfeng Chen
- Department of Pharmacy and Institute of Inflammation, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; School of Pharmacy, Hangzhou Medical College, Hangzhou 310012 Zhejiang, China.
| |
Collapse
|
34
|
Whitely C, Winburn H, Li Y. Synthesis and Identification of Heterobivalent Anticancer Compounds Containing Urea and 5-Arylidene-2-Thiohydantoin Motifs. ChemistrySelect 2024; 9:e202304688. [PMID: 38585446 PMCID: PMC10993625 DOI: 10.1002/slct.202304688] [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: 11/20/2023] [Accepted: 02/27/2024] [Indexed: 04/09/2024]
Abstract
Urea and thiohydantoin are among the important privileged structures for drug discovery. We have developed a synthetic approach to the high-throughput synthesis of the heterobivalent compounds containing both urea and 5-arylidene-2-thiohydantoin functional groups. This synthetic methodology was applied to the synthesis of a mixture-based library containing a total of 5280 compounds in a positional scanning format. The library was screened for its antiproliferative activity against cancer cells using a tetrazolium dye (MTT) based assay. Deconvolution of the library identified six hit compounds exhibiting moderate inhibitory potency against cancer cell proliferation.
Collapse
Affiliation(s)
- Chelsi Whitely
- Department of Drug Discovery & Biomedical Sciences, University of South Carolina, 517 Sumter St., CLS 617, Columbia, SC 29208
| | - Haley Winburn
- Department of Drug Discovery & Biomedical Sciences, University of South Carolina, 517 Sumter St., CLS 617, Columbia, SC 29208
| | - Yangmei Li
- Department of Drug Discovery & Biomedical Sciences, University of South Carolina, 517 Sumter St., CLS 617, Columbia, SC 29208
| |
Collapse
|
35
|
Canale V, Skiba-Kurek I, Klesiewicz K, Papież M, Ropek M, Pomierny B, Piska K, Koczurkiewicz-Adamczyk P, Empel J, Karczewska E, Zajdel P. Improving Activity of New Arylurea Agents against Multidrug-Resistant and Biofilm-Producing Staphylococcus epidermidis. ACS Med Chem Lett 2024; 15:369-375. [PMID: 38505856 PMCID: PMC10945555 DOI: 10.1021/acsmedchemlett.3c00536] [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: 11/28/2023] [Revised: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 03/21/2024] Open
Abstract
Multidrug-resistant (MDR) strains of Staphylococcus epidermidis (S. epidermidis), prevalent in hospital environments, contribute to increased morbidity and mortality, especially among newborns, posing a critical concern for neonatal sepsis. In response to the pressing demand for novel antibacterial therapies, we present findings from synthetic chemistry and structure-activity relationship studies focused on arylsulfonamide/arylurea derivatives of aryloxy[1-(thien-2-yl)propyl]piperidines. Through bioisosteric replacement of the sulfonamide fragment with a urea moiety, compound 25 was identified, demonstrating potent bacteriostatic activity against clinical multidrug-resistant S. epidermidis strains (MIC50 and MIC90 = 1.6 and 3.125 μg/mL). Importantly, it showed activity against linezolid-resistant strains and exhibited selectivity over mammalian cells. Compound 25 displayed antibiofilm-forming properties against clinical S. epidermidis strains and demonstrated the capacity to eliminate existing biofilm layers. Additionally, it induced complete depolarization of the bacterial membrane in clinical S. epidermidis strains. In light of these findings, targeting bacterial cell membranes with compound 25 emerges as a promising strategy in the fight against multidrug-resistant S. epidermidis strains.
Collapse
Affiliation(s)
- Vittorio Canale
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Iwona Skiba-Kurek
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Karolina Klesiewicz
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Monika Papież
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Marlena Ropek
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Bartosz Pomierny
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Kamil Piska
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | | | - Joanna Empel
- Department
of Epidemiology and Clinical Microbiology, National Medicines Institute, 30/34 Chełmska Street, 00-725 Warsaw, Poland
| | - Elżbieta Karczewska
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| | - Paweł Zajdel
- Faculty
of Pharmacy Jagiellonian University Medical College, 9 Medyczna Str., 30-688 Kraków, Poland
| |
Collapse
|
36
|
Espinoza-Chávez RM, Oliveira Rezende Júnior CD, de Souza ML, Pauli I, Valli M, Gomes Ferreira LL, Chelucci RC, Michelan-Duarte S, Krogh R, Romualdo da Silva FB, Cruz FC, de Oliveira AS, Andricopulo AD, Dias LC. Structure-activity relationships of novel N-imidazoylpiperazines with potent anti- Trypanosoma cruzi activity. Future Med Chem 2024; 16:253-269. [PMID: 38193294 DOI: 10.4155/fmc-2023-0185] [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: 06/27/2023] [Accepted: 12/06/2023] [Indexed: 01/10/2024] Open
Abstract
Background: Chagas disease is caused by the parasite Trypanosoma cruzi, and the lack of effective and safe treatments makes identifying new classes of compounds with anti-T. cruzi activity of paramount importance. Methods: Hit-to-lead exploration of a metabolically stable N-imidazoylpiperazine was performed. Results: Compound 2, a piperazine derivative active against T. cruzi, was selected to perform the hit-to-lead exploration, which involved the design, synthesis and biological evaluation of 39 new derivatives. Conclusion: Compounds 6e and 10a were identified as optimized compounds with low micromolar in vitro activity, low cytotoxicity and suitable preliminary absorption, distribution, metabolism and excretion and physicochemical properties. Both compounds reduced parasitemia in mouse models of Chagas disease, providing a promising opportunity for further exploration of new antichagasic compounds.
Collapse
Affiliation(s)
- Rocío Marisol Espinoza-Chávez
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, State University of Campinas, Campinas-SP, 13084-971, Brazil
| | - Celso de Oliveira Rezende Júnior
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, State University of Campinas, Campinas-SP, 13084-971, Brazil
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia-MG, 38400-902, Brazil
| | - Mariana Laureano de Souza
- Laboratory of Medicinal & Computational Chemistry, São Carlos Institute of Physics, University of São Paulo, São Carlos-SP, 13563-120, Brazil
| | - Ivani Pauli
- Laboratory of Medicinal & Computational Chemistry, São Carlos Institute of Physics, University of São Paulo, São Carlos-SP, 13563-120, Brazil
| | - Marilia Valli
- Laboratory of Medicinal & Computational Chemistry, São Carlos Institute of Physics, University of São Paulo, São Carlos-SP, 13563-120, Brazil
| | - Leonardo Luiz Gomes Ferreira
- Laboratory of Medicinal & Computational Chemistry, São Carlos Institute of Physics, University of São Paulo, São Carlos-SP, 13563-120, Brazil
| | - Rafael Consolin Chelucci
- Laboratory of Medicinal & Computational Chemistry, São Carlos Institute of Physics, University of São Paulo, São Carlos-SP, 13563-120, Brazil
| | - Simone Michelan-Duarte
- Laboratory of Medicinal & Computational Chemistry, São Carlos Institute of Physics, University of São Paulo, São Carlos-SP, 13563-120, Brazil
| | - Renata Krogh
- Laboratory of Medicinal & Computational Chemistry, São Carlos Institute of Physics, University of São Paulo, São Carlos-SP, 13563-120, Brazil
| | | | - Fábio Cardoso Cruz
- Department of Pharmacology, Federal University of São Paulo - UNIFESP, São Paulo-SP, 04023-062, Brazil
| | - Aldo Sena de Oliveira
- Department of Exact Sciences & Education, Federal University of Santa Catarina, Campus of Blumenau, Santa Catarina-SC, 89036-256, Brazil
| | - Adriano Defini Andricopulo
- Laboratory of Medicinal & Computational Chemistry, São Carlos Institute of Physics, University of São Paulo, São Carlos-SP, 13563-120, Brazil
| | - Luiz Carlos Dias
- Laboratory of Synthetic Organic Chemistry, Institute of Chemistry, State University of Campinas, Campinas-SP, 13084-971, Brazil
| |
Collapse
|
37
|
Eldehna WM, Al-Ansary GH, Al-Warhi T, Jaballah MY, Elaasser M, Rashed M. Identification of novel ureido benzothiophenes as dual VEGFR-2/EGFR anticancer agents. Bioorg Chem 2024; 143:107037. [PMID: 38134521 DOI: 10.1016/j.bioorg.2023.107037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
Presently, dual-targeting by a single small molecule stands out as a fruitful cancer-fighting strategy. Joining the global effort to fight cancer, a leading cause of death worldwide, we report in this study a novel set for benzothiophene-based aryl urea derivatives as potential anti-proliferative candidates endowed with dual VEGFR-2/EGFR inhibitory activities. The prepared ureido benzothiophenes 6a-r have been evaluated for their anticancer action on a panel of tumor cell lines, namely PanC-1, MCF-7, and HepG2 cells. Most newly synthesized benzo[b]thiophene ureas disclosed effective cytotoxic activities against the examined cancer cell lines. In particular, compound 6q, with an appended 4-trifluoromethoxy group on the terminal phenyl ring, exhibited the most significant cytotoxic activity in MCF-7 with IC50 3.86 ± 0.72 ug/mL; IC50 of 3.65 ± 0.18 ug/ml in PanC-1 cell line and an IC50 of 4.78 ± 0.06 ug/ml in HepG2. After that, derivatives that exhibited the most potent cytotoxic activities (6g, 6j, 6q, and 6r) were further evaluated as VEGFR-2 and EGFR inhibitors. Fortunately, they displayed low nanomolar IC50 values against both enzymes, where compound 6q emerged to possess superior inhibitory effects towards both EGFR and VEGFR-2 with IC50 46.6 nM and 11.3 nM simultaneously compared to the reference medications Erlotinib and Sorafenib, respectively. The docked structure of 6q within the catalytic region of VEGFR-2 and EGFR kinases was acquired and studied so that we could investigate potential binding mechanisms for the target ureido benzothiophenes. Hence, the benzothiophene-based aryl urea scaffold has great potential for advancing the development of highly effective dual inhibitors targeting both EGFR and VEGFR-2, which can serve as effective candidates for anticancer therapy.
Collapse
Affiliation(s)
- Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt.
| | - Ghada H Al-Ansary
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo 11566, Egypt
| | - Tarfah Al-Warhi
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Maiy Y Jaballah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo 11566, Egypt
| | - Mahmoud Elaasser
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo 11651, Egypt
| | - Mahmoud Rashed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| |
Collapse
|
38
|
Acharya A, Yadav M, Nagpure M, Kumaresan S, Guchhait SK. Molecular medicinal insights into scaffold hopping-based drug discovery success. Drug Discov Today 2024; 29:103845. [PMID: 38013043 DOI: 10.1016/j.drudis.2023.103845] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2023]
Abstract
In both academia and the pharmaceutical industry, innovative hypotheses, methodologies and technologies that can shorten the drug research and development, leading to higher success rates, are vital. In this review, we demonstrate how innovative variations of the scaffold-hopping strategy have been used to create new druggable molecular spaces, drugs, clinical candidates, preclinical candidates, and bioactive agents. We also analyze molecular modulations that enabled improvements of the pharmacodynamic (PD), physiochemical, and pharmacokinetic (PK) properties (P3 properties) of the drugs resulting from these scaffold-hopping strategies.
Collapse
Affiliation(s)
- Ayan Acharya
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Mukul Yadav
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Mithilesh Nagpure
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Sanathanalaxmi Kumaresan
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India; National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India
| | - Sankar K Guchhait
- National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
39
|
Olgen S, Kaleli SNB, Karaca BT, Demirel UU, Bristow HK. Synthesis and Anticancer Activity of Novel Indole Derivatives as Dual EGFR/SRC Kinase Inhibitors. Curr Med Chem 2024; 31:3798-3817. [PMID: 37365789 DOI: 10.2174/0929867330666230626143911] [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: 03/28/2023] [Revised: 05/18/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Recent studies showed that the cooperation between c-SRC and EGFR is responsible for more aggressive phenotype in diverse tumors, including glioblastomas and carcinomas of the colon, breast, and lung. Studies show that combination of SRC and EGFR inhibitors can induce apoptosis and delay the acquired resistance to chemotherapy. Therefore, such combination may lead to a new therapeutic strategy for the treatment of EGFR-mutant lung cancer. Osimertinib was developed as a third-generation EGFR-TKI to combat the toxicity of EGFR mutant inhibitors. Due to the resistance and adverse reaction of osimertinib and other kinase inhibitors, 12 novel compounds structurally similar to osimertinib were designed and synthesized. METHODS Compounds were synthesized by developing novel original synthesis methods and receptor interactions were evaluated through a molecular docking study. To evaluate their inhibitory activities against EGFR and SRC kinase, in vitro enzyme assays were used. Anticancer potencies were determined using lung, breast, prostate (A549, MCF6, PC3) cancer cell lines. Compounds were also tested against normal (HEK293) cell line to evaluate their cyctotoxic effects. RESULTS Although, none of compounds showed stronger inhibition compared to osimertinib in the EGFR enzyme inhibition studies, compound 16 showed the highest efficacy with an IC50 of 1.026 μM. It also presented potent activity against SRC kinase with an IC50 of 0.002 μM. Among the tested compounds, the urea containing derivatives 6-11 exhibited a strong inhibition profile (80.12-89.68%) against SRC kinase in comparison to the reference compound dasatinib (93.26%). Most of the compounds caused more than 50% of cell death in breast, lung and prostate cancer cell lines and weak toxicity for normal cells in comparison to reference compounds osimertinib, dasatinib and cisplatin. Compound 16 showed strong cytotoxicity on lung and prostate cancer cells. Treatment of prostate cancer cell lines with the most active compound, 16, significantly increased the caspase-3 (8-fold), caspase-8 (6-fold) and Bax (5.7-fold) levels and decreased the Bcl-2 level (2.3-fold) compared to the control group. These findings revealed that the compound 16 strongly induces apoptosis in the prostate cancer cell lines. CONCLUSION Overall kinase inhibition, cytotoxicity and apoptosis assays presented that compound 16 has dual inhibitory activity against SRC and EGFR kinases while maintaining low toxicity against normal cells. Other compounds also showed considerable activity profiles in kinase and cell culture assays.
Collapse
Affiliation(s)
- Sureyya Olgen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, 34010, Zeytinburnu, Istanbul, Turkey
| | - Sevde Nur Biltekin Kaleli
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Istanbul Medipol University, 34815 Beykoz-Istanbul, Turkey
| | - Banu Taktak Karaca
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Istanbul Atlas University, İstanbul, Turkey
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Beykoz-Istanbul, Turkey
| | - Ural U Demirel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Altınbaş University, Istanbul, Turkey
| | - Hacer Karatas Bristow
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810 Beykoz-Istanbul, Turkey
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Istanbul Medipol University, 34815 Beykoz-Istanbul, Turkey
- C. Eugene Bennett Department of Chemistry, West Virgina University, 26506 Morgantown, West Virginia, USA
| |
Collapse
|
40
|
Henry C, Kruell JA, Wilson RM, Chang CF, Woo CM, Koehler AN. A Versatile Isocyanate-Mediated Strategy for Appending Chemical Tags onto Drug-Like Small Molecules. Bioconjug Chem 2023; 34:2181-2186. [PMID: 38052453 PMCID: PMC10739574 DOI: 10.1021/acs.bioconjchem.3c00352] [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: 08/06/2023] [Revised: 10/09/2023] [Accepted: 10/23/2023] [Indexed: 12/07/2023]
Abstract
Target identification studies are a major hurdle in probe and drug discovery pipelines due to the need to chemically modify small molecules of interest, which can be time intensive and have low throughput. Here, we describe a versatile and scalable method for attaching chemical moieties to a small molecule, isocyanate-mediated chemical tagging (IMCT). By preparation of a template resin with an isocyanate capture group and a cleavable linker, nucleophilic groups on small molecules can be modified with an enforced one-to-one stoichiometry. We demonstrate a small molecule substrate scope that includes primary and secondary amines, thiols, phenols, benzyl alcohols, and primary alcohols. Cheminformatic analyses predict that IMCT is reactive with more than 25% of lead-like compounds in publicly available databases. To demonstrate that the method can produce biologically active molecules, we generated FKBP12 photoaffinity labeling (PAL) compounds with a wide range of affinities and showed that purified and crude cleavage products can bind to and label FKBP12. This method could be used to rapidly modify small molecules for many applications, including the synthesis of PAL probes, fluorescence polarization probes, pull-down probes, and degraders.
Collapse
Affiliation(s)
- Catherine
C. Henry
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- MIT
Center for Precision Cancer Medicine, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02142, United States
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Jasmin A. Kruell
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- MIT
Center for Precision Cancer Medicine, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02142, United States
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Robert M. Wilson
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- MIT
Center for Precision Cancer Medicine, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02142, United States
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Chia-Fu Chang
- Chemistry
and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M. Woo
- Chemistry
and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Angela N. Koehler
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- MIT
Center for Precision Cancer Medicine, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02142, United States
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Biological Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
41
|
Pavić K, Poje G, Carvalho LPD, Held J, Rajić Z. Synthesis, antiproliferative and antiplasmodial evaluation of new chloroquine and mefloquine-based harmiquins. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2023; 73:537-558. [PMID: 38147482 DOI: 10.2478/acph-2023-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/09/2023] [Indexed: 12/28/2023]
Abstract
Here we present the synthesis and evaluation of the biological activity of new hybrid compounds, ureido-type (UT) harmiquins, based on chloroquine (CQ) or mefloquine (MQ) scaffolds and β-carboline alkaloid harmine against cancer cell lines and Plasmodium falciparum. The hybrids were prepared from the corresponding amines by 1,1'-carbonyldiimidazole (CDI)-mediated synthesis. In vitro evaluation of the biological activity of the title compounds revealed two hit compounds. Testing of the antiproliferative activity of the new UT harmiquins, and previously prepared triazole-(TT) and amide-type (AT) CQ-based harmiquins, against a panel of human cell lines, revealed TT harmiquine 16 as the most promising compound, as it showed pronounced and selective activity against the tumor cell line HepG2 (IC 50 = 5.48 ± 3.35 μmol L-1). Screening of the antiplasmodial activities of UT harmiquins against erythrocytic stages of the Plasmodium life cycle identified CQ-based UT harmiquine 12 as a novel antiplasmodial hit because it displayed low IC 50 values in the submicromolar range against CQ-sensitive and resistant strains (IC 50 0.06 ± 0.01, and 0.19 ± 0.02 μmol L-1, respectively), and exhibited high selectivity against Plasmodium, compared to mammalian cells (SI = 92).
Collapse
Affiliation(s)
- Kristina Pavić
- 1University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medicinal Chemistry 10 000 Zagreb, Croatia
| | - Goran Poje
- 1University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medicinal Chemistry 10 000 Zagreb, Croatia
| | | | - Jana Held
- 2University of Tübingen, Institute of Tropical Medicine, 72074, Tübingen Germany
- 3German Center for Infection Research (DZIF), 72074, Tübingen Germany
| | - Zrinka Rajić
- 1University of Zagreb Faculty of Pharmacy and Biochemistry Department of Medicinal Chemistry 10 000 Zagreb, Croatia
| |
Collapse
|
42
|
Kumar P, Bhalla A. Isothiocyanates ( in situ) and sulfonyl chlorides in water for N-functionalization of bicyclic amidines: access to N-alkylated γ-/ω-lactam derivatized thiourea and sulfonamides. Org Biomol Chem 2023; 21:8868-8874. [PMID: 37888837 DOI: 10.1039/d3ob01584j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Herein, we showcase the potential of isothiocyanates generated in situ and aryl sulfonyl chlorides as electrophiles in water for N-functionalization of bicyclic amidines (DBN and DBU). This strategy provides complementary access to a range of thiouredosulfides, sulfonamides, aroylthioureas and amides derivativatized with distal γ- and ω-lactams. A novel sulfonyl chloride mediated formation of β-uredo sulfides has been achieved from β-isothiocyanato sulfides, removing the requirement for the harsh synthesis of unstable isocyanates. Mechanistic studies suggest a radical mechanism for the difunctionalization of alkenes, the efficacy of H2O in the ring opening of bicyclic amidines, and an oxygen source along with sulfonyl chloride as desulfurization agents for thiourea to afford urea derivatives.
Collapse
Affiliation(s)
- Pankaj Kumar
- Department of Chemistry and Centre of Advance Studies in Chemistry, Panjab University, Chandigarh, 160014, UT, India.
| | - Aman Bhalla
- Department of Chemistry and Centre of Advance Studies in Chemistry, Panjab University, Chandigarh, 160014, UT, India.
| |
Collapse
|
43
|
García A, Torres-Ruiz S, Vila L, Villarroel-Vicente C, Bernabeu Á, Eroles P, Cabedo N, Cortes D. Synthesis of 2-aminopropyl benzopyran derivatives as potential agents against triple-negative breast cancer. RSC Med Chem 2023; 14:2327-2341. [PMID: 38020071 PMCID: PMC10650959 DOI: 10.1039/d3md00385j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/05/2023] [Indexed: 12/01/2023] Open
Abstract
Synthesis of three series of 2-aminopropyl derivatives containing a benzopyran nucleus was performed to evaluate their performance against triple-negative breast cancer cell lines (MDA-MB-231 and MDA-MB-436) and normal breast epithelial cells (MCF10A). For the three series, the cytotoxic activity was as follows: N-methylated derivatives (tertiary amines) 5b, 6b, and 7b > secondary amine benzopyrans 5, 6, and 7 > quaternary amine salts 5c, 6c, and 7c > free phenolic derivatives 5a, 6a, and 7a. The structure-activity relationship showed the importance of the presence of an amine group and a p-fluorobenzyloxy substituent in the chromanol ring (IC50 values from 1.5 μM to 58.4 μM). In addition, 5a, 5b, 6a, and 7b displayed slight selectivity towards tumor cells. Compounds 5, 5a, 5b, 6, 6a, 6c, 7, and 7b showed apoptotic/necrotic effects due to, at least in part, an increase in reactive oxygen species generation, whereas 5b, 5c, 6b, 7a, and 7c caused cell cycle arrest in the G1 phase. Further cell-based mechanistic studies revealed that 5a, 6a, and 7b, which were the most promising compounds, downregulated the expression of Bcl-2, while 5b downregulated the expression of cyclins CCND1 and CCND2. Therefore, 2-aminopropyl benzopyran derivatives emerge as new hits and potential leads for developing useful agents against breast cancer.
Collapse
Affiliation(s)
- Ainhoa García
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Sandra Torres-Ruiz
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Laura Vila
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Carlos Villarroel-Vicente
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Álvaro Bernabeu
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
| | - Pilar Eroles
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
- Department of Physiology, University of Valencia 46010 Valencia Spain
- Center for Biomedical Network Research on Cancer (CIBERONC) 28019 Madrid Spain
| | - Nuria Cabedo
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Diego Cortes
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
| |
Collapse
|
44
|
Inoue T, Ota M, Amijima Y, Takahashi H, Hamada S, Nakamura S, Kobayashi Y, Sasamori T, Furuta T. Dual Chalcogen-Bonding Interactions for the Conformational Control of Urea. Chemistry 2023; 29:e202302139. [PMID: 37507838 DOI: 10.1002/chem.202302139] [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: 07/05/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 07/30/2023]
Abstract
Dual chalcogen-bonding interactions is proposed as a novel means for the conformational control of urea derivatives. The formation of a chalcogen-bonding interaction at both sides of the urea carbonyl group was unambiguously confirmed by X-ray diffraction as well as computational studies including non-covalent interaction (NCI) plot index analysis, quantum theory of atoms in molecules (QTAIM) analysis, and natural bond orbital (NBO) analysis via DFT calculations. By virtue of this dual interaction, urea derivatives that bear chalcogen atoms (X=S and Se) adopt a planar structure via the carbonyl oxygen (O) with an X⋅⋅⋅O⋅⋅⋅X arrangement on the same side of the molecule. The rigidity of the conformational lock was evaluated using the molecular arrangement in the crystal and the rotational barrier of benzochalcogenophene ring, which indicated a stronger conformational lock in benzoselenophene than in benzothiophene urea derivatives. Furthermore, the acidity of the urea derivatives increases according to the Lewis-acidic properties of the chalcogen-bonding interactions, whereby benzoselenophene urea is more acidic than benzothiophene urea. Tweezer-shaped urea derivatives were prepared, and their stereostructure proved the viability of the conformational control for defining the location of the substituents on the urea framework.
Collapse
Affiliation(s)
- Takumi Inoue
- Department of Pharmaceutical Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Moe Ota
- Department of Pharmaceutical Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Yui Amijima
- Department of Pharmaceutical Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Haru Takahashi
- Department of Pharmaceutical Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Shohei Hamada
- Department of Pharmaceutical Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Seikou Nakamura
- Department of Pharmacognosy, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Yusuke Kobayashi
- Department of Pharmaceutical Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, 607-8414, Japan
| | - Takahiro Sasamori
- Department of Chemistry Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8571, Japan
| | - Takumi Furuta
- Department of Pharmaceutical Chemistry, Kyoto Pharmaceutical University, Yamashina-ku, Kyoto, 607-8414, Japan
| |
Collapse
|
45
|
Milani G, Budriesi R, Tavazzani E, Cavalluzzi MM, Mattioli LB, Miniero DV, Delre P, Belviso BD, Denegri M, Cuocci C, Rotondo NP, De Palma A, Gualdani R, Caliandro R, Mangiatordi GF, Kumawat A, Camilloni C, Priori S, Lentini G. hERG stereoselective modulation by mexiletine-derived ureas: Molecular docking study, synthesis, and biological evaluation. Arch Pharm (Weinheim) 2023; 356:e2300116. [PMID: 37460390 DOI: 10.1002/ardp.202300116] [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: 02/24/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 10/06/2023]
Abstract
Long QT syndrome (LQTS) is a disorder of cardiac electrophysiology resulting in life-threatening arrhythmias; nowadays, only a few drugs are available for the management of LQTS. Focusing our attention on LQT2, one of the most common subtypes of LQTS caused by mutations in the human ether-à-go-go-related gene (hERG), in the present work, the stereoselectivity of the recently discovered mexiletine-derived urea 8 was investigated on the hERG potassium channel. According to preliminary in silico predictions, in vitro studies revealed a stereoselective behavior, with the meso form showing the greatest hERG opening activity. In addition, functional studies on guinea pig isolated left atria, aorta, and ileum demonstrated that 8 does not present any cardiac or intestinal liability in our ex vivo studies. Due to its overall profile, (R,S)-8 paves the way for the design and development of a new series of compounds potentially useful in the treatment of both congenital and drug-induced forms of LQTS.
Collapse
Affiliation(s)
- Gualtiero Milani
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Roberta Budriesi
- Department of Pharmacy and Biotechnology, Food Chemistry and Nutraceutical Lab, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | | | | | - Laura Beatrice Mattioli
- Department of Pharmacy and Biotechnology, Food Chemistry and Nutraceutical Lab, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Daniela Valeria Miniero
- Department of Biosciences, Biotechnologies, and Environment, University Aldo Moro of Bari, Bari, Italy
| | - Pietro Delre
- Chemistry Department, University of Bari Aldo Moro, Bari, Italy
- CNR-Institute of Crystallography, Bari, Italy
| | | | | | | | - Natalie Paola Rotondo
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa De Palma
- Department of Biosciences, Biotechnologies, and Environment, University Aldo Moro of Bari, Bari, Italy
| | - Roberta Gualdani
- Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | | | | | - Amit Kumawat
- Department of Biosciences, University of Milan, Milano, Italy
| | - Carlo Camilloni
- Department of Biosciences, University of Milan, Milano, Italy
| | - Silvia Priori
- ICS-Maugeri IRCCS, Pavia, Italy
- Molecular Cardiology, Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Giovanni Lentini
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
46
|
Sahoo SK, Ommi O, Maddipatla S, Singh P, Ahmad MN, Kaul G, Nanduri S, Dasgupta A, Chopra S, Yaddanapudi VM. Isoxazole carboxylic acid methyl ester-based urea and thiourea derivatives as promising antitubercular agents. Mol Divers 2023; 27:2037-2052. [PMID: 36282413 PMCID: PMC9592870 DOI: 10.1007/s11030-022-10543-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/28/2022] [Indexed: 11/01/2022]
Abstract
In our continued efforts to find potential chemotherapeutics active against drug-resistant (DR) Mycobacterium tuberculosis (Mtb), causative agent of Tuberculosis (TB) and to curb the current burdensome treatment regimen, herein we describe the synthesis and biological evaluation of urea and thiourea variants of 5-phenyl-3-isoxazolecarboxylic acid methyl esters as promising anti-TB agent. Majority of the tested compounds displayed potent in vitro activity not only against drug-susceptible (DS) Mtb H37Rv but also against drug-resistant (DR) Mtb. Cell viability test against Vero cells deemed these compounds devoid of significant toxicity. 3,4-Dichlorophenyl derivative (MIC 0.25 µg/mL) and 4-chlorophenyl congener (MIC 1 µg/mL) among urea and thiourea libraries respectively exhibited optimum potency. Lead optimization resulted in the identification of 1,4-linked analogue of 3,4-dichlorophenyl urea derivative demonstrating improved selectivity. Further, in silico study complemented with previously proposed prodrug like attributes of isoxazole esters. Taken together, this molecular hybridization approach presents a new chemotype having potential to be translated into an alternate anti-Mtb agent.
Collapse
Affiliation(s)
- Santosh Kumar Sahoo
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Ojaswitha Ommi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Sarvan Maddipatla
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Priti Singh
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Mohammad Naiyaz Ahmad
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Grace Kaul
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Arunava Dasgupta
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India.
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Sidharth Chopra
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India.
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
47
|
El-Atawy MA, Alsubaie MS, Alazmi ML, Hamed EA, Hanna DH, Ahmed HA, Omar AZ. Synthesis, Characterization, and Anticancer Activity of New N,N'-Diarylthiourea Derivative against Breast Cancer Cells. Molecules 2023; 28:6420. [PMID: 37687250 PMCID: PMC10490226 DOI: 10.3390/molecules28176420] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/21/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The goal of the current study was to prepare two new homologous series of N,N'-diarylurea and N,N'-diarylthiourea derivatives to investigate the therapeutic effects of these derivatives on the methodologies of inhibition directed on human MCF-7 cancer cells. The molecular structures of the prepared derivatives were successfully revealed through elemental analyses, 1H-NMR, 13C-NMR and FT-IR spectroscopy. The cytotoxic results showed that Diarylthiourea (compound 4) was the most effective in suppressing MCF-7 cell growth when compared to all other prepared derivatives, with the most effective IC50 value (338.33 ± 1.52 µM) after an incubation period of 24 h and no cytotoxic effects on normal human lung cells (wi38 cells). Using the annexin V/PI and comet tests, respectively, treated MCF-7 cells with this IC50 value of the Diarylthiourea 4 compound displayed a considerable increase in early and late apoptotic cells, as well as an intense comet nucleus in comparison to control cells. An arrest of the cell cycle in the S phase was observed via flow cytometry in MCF-7 cells treated with the Diarylthiourea 4 compound, suggesting the onset of apoptosis. Additionally, ELISA research showed that caspase-3 was upregulated in MCF-7 cells treated with compound 4 compared to control cells, suggesting that DNA damage induced by compound 4 may initiate an intrinsic apoptotic pathway and activate caspase-3. These results contributed to recognizing that the successfully prepared Diarylthiourea 4 compound inhibited the proliferation of MCF-7 cancer cells by arresting the S cell cycle and caspase-3 activation via an intrinsic apoptotic route. These results, however, need to be verified through in vivo studies utilizing an animal model.
Collapse
Affiliation(s)
- Mohamed A. El-Atawy
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
- Chemistry Department, Faculty of Science, Taibah University, Yanbu 46423, Saudi Arabia
| | - Mai S. Alsubaie
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
| | - Mohammed L. Alazmi
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
| | - Ezzat A. Hamed
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
| | - Demiana H. Hanna
- Department of Chemistry, Faculty of Science, Cairo University, Giza 12613, Egypt;
| | - Hoda A. Ahmed
- Department of Chemistry, Faculty of Science, Cairo University, Giza 12613, Egypt;
| | - Alaa Z. Omar
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426 Ibrahemia, Alexandria 21321, Egypt; (M.A.E.-A.); (A.Z.O.)
| |
Collapse
|
48
|
Jin J, Cui Z, Lv C, Peng X, Yan Z, Song Y, Cao Y, Zhou W, Wang E, Chen X, Kang D, Hu L, Wang J. Design, synthesis, and biological evaluation of a series of indolone derivatives as novel FLT3 inhibitors for the treatment of acute myeloid leukemia. Bioorg Chem 2023; 138:106645. [PMID: 37327602 DOI: 10.1016/j.bioorg.2023.106645] [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: 11/23/2022] [Revised: 04/24/2023] [Accepted: 05/29/2023] [Indexed: 06/18/2023]
Abstract
FLT3-ITD mutant has been extensively studied as a drug discovery target for acute myeloid leukemia. Based on our previous discovered FLT3 inhibitor (2), a series of urea group based indolone derivatives were designed, synthesized, and biological evaluated as novel FLT3 inhibitors for the treatment of FLT3-ITD positive AML. Among them, compound LC-3 exhibited potent inhibitory effects against FLT3 (IC50 = 8.4 nM) and significantly inhibited the proliferation of FLT3-ITD positive AML cells MV-4-11 (IC50 = 5.3 nM). In the cellular context, LC-3 strongly inhibited FLT3-mediated signaling pathways and induced cellular apoptosis by arresting cell cycle in G1 phase. In the in vivo studies, LC-3 significantly suppressed the tumor growth on MV-4-11 xenograft models (10 mg/kg/day, TGI = 92.16%) without exhibiting obvious toxicity. These results suggested that compound LC-3 might be a potential drug candidate for FLT3-ITD positive AML.
Collapse
Affiliation(s)
- Jiaming Jin
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zhenzhen Cui
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Cheng Lv
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Xuemei Peng
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zhiqi Yan
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yi Song
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yu Cao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Wenyi Zhou
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Enpeng Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Xufan Chen
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Di Kang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Junwei Wang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
49
|
Sepehri S, Khedmati M. An overview of the privileged synthetic heterocycles as urease enzyme inhibitors: Structure-activity relationship. Arch Pharm (Weinheim) 2023; 356:e2300252. [PMID: 37401193 DOI: 10.1002/ardp.202300252] [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: 05/08/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 07/05/2023]
Abstract
Urease is a metalloenzyme including two Ni2+ ions, found in some plants, bacteria, fungi, microorganisms, invertebrate animals, and animal tissues. Urease acts as a significant virulence factor, mainly in catheter blockage and infective urolithiasis as well as in the pathogenesis of gastric infection. Therefore, studies on urease lead to novel synthetic inhibitors. In this review, the synthesis and antiurease activities of a collection of privileged synthetic heterocycles such as (thio)barbiturate, (thio)urea, dihydropyrimidine, and triazol derivatives were described and discussed according to structure-activity relationship findings in search of the best moieties and substituents that are answerable for encouraging the desired activity even more potent than the standard. It was found that linking substituted phenyl and benzyl rings to the heterocycles led to potent urease inhibitors.
Collapse
Affiliation(s)
- Saghi Sepehri
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mina Khedmati
- Students Research Committee, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
50
|
Dar MA. Implications of the Pore Size of Graphitic Carbon Nitride Monolayers on the Selectivity of Dual-Boron Atom Catalysts for the Reduction of N 2 to Urea and Ammonia: A Computational Investigation. Inorg Chem 2023; 62:13672-13679. [PMID: 37555942 DOI: 10.1021/acs.inorgchem.3c02316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
The formation of urea by electrocatalytic means remains a great challenge due to the lack of a suitable catalyst that is capable of not only activating inert N2 and CO2 molecules but also circumventing the complexity associated with subsequent reaction steps leading to urea formation. Herein, by means of comprehensive density functional theory simulations, we investigate the catalytic activity of highly stable transition-metal-free dual-boron atom-doped graphitic carbon-nitride monolayers with different pore sizes toward urea production under ambient conditions. As per the results, dual boron atoms impregnated in g-C2N and g-C6N6 monolayers with large pore diameters can successfully activate the N2 molecule and lead to the spontaneous formation of the *NCO*N intermediate, which is the most crucial step for urea formation via direct coupling of N2 and CO2. Interestingly, the B2@g-C2N and B2@g-C6N6 favor urea production with low limiting potentials of -1.11 and -1.18 V compared to very high limiting potentials of -1.71 and -1.88 V, respectively, for ammonia synthesis, leading to an almost 100% Faradaic efficiency for urea formation over ammonia. The dual-boron doping in g-C3N4 with a smaller pore size depicts comparatively weaker N2 adsorption than g-C2N and g-C6N6 counterparts. Further, B2@g-C3N4 prefers ammonia formation at a very low limiting potential of -0.40 V compared to a very high limiting potential of -2.11 V for urea formation. Thus, our findings clearly highlight the critical role played by the pore size of carbon-nitride monolayers in tuning the reactivity and catalytic activity of dual-boron atom catalysts toward urea formation in a selective manner, thereby providing valuable guidance in exploring other highly efficient urea catalysts.
Collapse
Affiliation(s)
- Manzoor Ahmad Dar
- Department of Chemistry, Islamic University of Science and Technology, Awantipora 192122, Kashmir, India
| |
Collapse
|