1
|
Patharia MA, Raut SV, Dhotre BK, Pathan MA. Design, Synthesis of Some New N-(2-fluoro-4-morpholin-4-yl-phenyl)-Substituted-Benzamide Derivatives and Screening of Their Microbial Activities. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2020.1833047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Murtaza A. Patharia
- Research and Development, Navin Research and Innovation Centre, NFIL, Surat, Gujarat, India
| | - Santosh V. Raut
- Department of Chemistry, Maulana Azad College and Research Center, Aurangabad, Maharashtra, India
| | - Bharat K. Dhotre
- Department of Chemistry, Swami Vivekanand Sr. College, Mantha, Maharashtra, India
| | - Mohammad Arif Pathan
- Department of Chemistry, Maulana Azad College and Research Center, Aurangabad, Maharashtra, India
| |
Collapse
|
2
|
Asati V, Anant A, Mahapatra DK, Bharti SK. Recent Advances of PI3 Kinase Inhibitors: Structure Anticancer Activity Relationship Studies. Mini Rev Med Chem 2022; 22:MRMC-EPUB-120629. [PMID: 36471584 DOI: 10.2174/1389450123666220202154757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/27/2021] [Accepted: 12/01/2021] [Indexed: 11/22/2022]
Abstract
Phosphatidyl-inositol-3-kinase (PI3K) has emerged as a potential therapeutic target for the development of novel anticancer drugs. The dysregulation of PI3K has been associated with many human malignancies such as breast, colon, endometrial, brain, and prostate cancers. The PI3K kinases in their different isoforms namely α, β, δ, and γ, encode PIK3CA, PIK3CB, PIK3CD, and PIK3CG genes. Specific gene mutation or overexpression of the protein is responsible for therapeutic failure of current therapeutics. Recently, various PI3K signaling pathway inhibitors have been identified which showed promising therapeutic results by acting on specific isoforms of the kinase too. Several inhibitors containing medicinally privileged scaffolds like oxadiazole, pyrrolotriazine, quinazoline, quinazolinone, quinazoline-chalcone hybrids, quinazoline-sulfonamide, pyrazolochalcone, quinolone hydroxamic acid, benzofuropyridinone, imidazopyridine, benzoxazines, dibenzoxanthene, indoloderivatives, benzimidazole, and benzothiazine derivatives have been developed to target PI3K pathway and/or a specific isoform. The PI3K inhibitors which are under clinical trial studies include GDC-0032, INK1117 for PI3K-α, and AZD8186 for PI3K-β. This review primarily focuses on the structural insights and structure anticancer activity relationship studies of recent PI3K inhibitors including their clinical stages of development and therapeutic values.
Collapse
Affiliation(s)
- Vivek Asati
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Arjun Anant
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Debarshi Kar Mahapatra
- Department of Pharmaceutical Chemistry, Dadasaheb Balpande College of Pharmacy, Nagpur, Maharashtra, India
| | - Sanjay Kumar Bharti
- Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, Chhattisgarh, India
| |
Collapse
|
3
|
Bin H, Chen P, Wu M, Wang F, Lin G, Pan S, Liu J, Mu B, Nan J, Huang Q, Li L, Yang S. Discovery of a potent and highly selective inhibitor of ataxia telangiectasia mutated and Rad3-Related (ATR) kinase: Structural activity relationship and antitumor activity both in vitro and in vivo. Eur J Med Chem 2022; 232:114187. [DOI: 10.1016/j.ejmech.2022.114187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/28/2022] [Accepted: 02/04/2022] [Indexed: 11/26/2022]
|
4
|
Grover P, Bhardwaj M, Kapoor G, Mehta L, Ghai R, Nagarajan K. Advances on Quinazoline Based Congeners for Anticancer Potential. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825666210212121056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The heterocyclic compounds have a great significance in medicinal chemistry because
they have extensive biological activities. Cancer is globally the leading cause of death
and it is a challenge to develop appropriate treatment for the management of cancer. Continuous
efforts are being made to find a suitable medicinal agent for cancer therapy. Nitrogencontaining
heterocycles have received noteworthy attention due to their wide and distinctive
pharmacological activities. One of the most important nitrogen-containing heterocycles in
medicinal chemistry is ‘quinazoline’ that possesses a wide spectrum of biological properties.
This scaffold is an important pharmacophore and is considered a privileged structure. Various
substituted quinazolines displayed anticancer activity against different types of cancer. This
review highlights the recent advances in quinazoline based molecules as anticancer agents.
Several in-vitro and in-vivo models used along with the results are also included. A subpart briefing natural quinazoline
containing anticancer compounds is also incorporated in the review.
Collapse
Affiliation(s)
- Parul Grover
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| | - Monika Bhardwaj
- Natural Product Chemistry Division, Indian Institute of Integrative Medicine, Canal Road, Jammu, 180001, India
| | - Garima Kapoor
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| | - Lovekesh Mehta
- Amity Institute of Pharmacy, Amity University, Noida, 201301, India
| | - Roma Ghai
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| | - K. Nagarajan
- KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, 201206, India
| |
Collapse
|
5
|
Murata Y, Kawamoto S, Fukuda K. Rocuronium Has a Suppressive Effect on Platelet Function via the P2Y12 Receptor Pathway In Vitro That Is Not Reversed by Sugammadex. Int J Mol Sci 2020; 21:ijms21176399. [PMID: 32899133 PMCID: PMC7504164 DOI: 10.3390/ijms21176399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/20/2020] [Accepted: 09/01/2020] [Indexed: 01/15/2023] Open
Abstract
Rocuronium is an aminosteroid nondepolarizing neuromuscular blocker that is widely used for anesthesia and intensive care. In this study, we investigated the effect of rocuronium on human platelet functions in vitro. The effects of rocuronium on platelet aggregation, P-selectin expression, and cyclic adenosine monophosphate (cAMP) levels in platelets were measured using an aggregometer, an enzyme immunoassay, and flow cytometry, respectively. Rocuronium inhibited ADP-induced platelet aggregation, P-selectin expression and suppression of cAMP production. These effects were not antagonized by equimolar sugammadex, a synthetic γ-cyclodextrin derivative that antagonizes rocuronium-induced muscle relaxation by encapsulating the rocuronium molecule. Morpholine, which constitutes a part of the rocuronium molecule but is not encapsulated by sugammadex, inhibited ADP-induced platelet aggregation. Vecuronium, which has a molecular structure similar to that of rocuronium but does not possess a morpholine ring, had no significant effect on ADP-induced platelet aggregation. These results indicate that rocuronium has a suppressive effect on platelet functions in vitro that is not reversed by sugammadex and suggest that this effect is mediated by blockade of the P2Y12 receptor signaling pathway via the morpholine ring of rocuronium.
Collapse
|
6
|
Moreira NM, Martelli LSR, de Julio KIR, Zukerman-Schpector J, Opatz T, Corrêa AG. Copper-Catalyzed One-Pot Synthesis of 3-( N
-Heteroarenyl)acrylonitriles through Radical Conjugated Addition of β-Nitrostyrene to Methylazaarenes. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Natália M. Moreira
- Centre of Excellence for Research in Sustainable Chemistry; Federal University of São Carlos; 13565-905 São Carlos SP Brazil
| | - Lorena S. R. Martelli
- Centre of Excellence for Research in Sustainable Chemistry; Federal University of São Carlos; 13565-905 São Carlos SP Brazil
| | - Kiyara I. R. de Julio
- Centre of Excellence for Research in Sustainable Chemistry; Federal University of São Carlos; 13565-905 São Carlos SP Brazil
| | | | - Till Opatz
- Department of Chemistry; Johannes Gutenberg-University; 55128 Mainz Germany
| | - Arlene G. Corrêa
- Centre of Excellence for Research in Sustainable Chemistry; Federal University of São Carlos; 13565-905 São Carlos SP Brazil
| |
Collapse
|
7
|
Jung YH, Yu J, Wen Z, Salmaso V, Karcz TP, Phung NB, Chen Z, Duca S, Bennett JM, Dudas S, Salvemini D, Gao ZG, Cook DN, Jacobson KA. Exploration of Alternative Scaffolds for P2Y 14 Receptor Antagonists Containing a Biaryl Core. J Med Chem 2020; 63:9563-9589. [PMID: 32787142 DOI: 10.1021/acs.jmedchem.0c00745] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Various heteroaryl and bicyclo-aliphatic analogues of zwitterionic biaryl P2Y14 receptor (P2Y14R) antagonists were synthesized, and affinity was measured in P2Y14R-expressing Chinese hamster ovary cells by flow cytometry. Given this series' low water solubility, various polyethylene glycol derivatives of the distally binding piperidin-4-yl moiety of moderate affinity were synthesized. Rotation of previously identified 1,2,3-triazole attached to the central m-benzoic acid core (25) provided moderate affinity but not indole and benzimidazole substitution of the aryl-triazole. The corresponding P2Y14R region is predicted by homology modeling as a deep, sterically limited hydrophobic pocket, with the outward pointing piperidine moiety being the most flexible. Bicyclic-substituted piperidine ring derivatives of naphthalene antagonist 1, e.g., quinuclidine 17 (MRS4608, IC50 ≈ 20 nM at hP2Y14R/mP2Y14R), or of triazole 2, preserved affinity. Potent antagonists 1, 7a, 17, and 23 (10 mg/kg) protected in an ovalbumin/Aspergillus mouse asthma model, and PEG conjugate 12 reduced chronic pain. Thus, we expanded P2Y14R antagonist structure-activity relationship, introducing diverse physical-chemical properties.
Collapse
Affiliation(s)
- Young-Hwan Jung
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Jinha Yu
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhiwei Wen
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Veronica Salmaso
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Tadeusz P Karcz
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, North Carolina 27709, United States.,Jagiellonian University, Kraków31-007, Poland
| | - Ngan B Phung
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhoumou Chen
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Boulevard, Saint Louis, Missouri 63104, United States
| | - Sierra Duca
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - John M Bennett
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Steven Dudas
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, 1402 South Grand Boulevard, Saint Louis, Missouri 63104, United States
| | - Zhan-Guo Gao
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Donald N Cook
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, North Carolina 27709, United States
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
8
|
Novak Jovanović I, Miličević A, Jadreško D, Hranjec M. Electrochemical oxidation of synthetic amino-substituted benzamides with potential antioxidant activity. J Electroanal Chem (Lausanne) 2020. [DOI: 10.1016/j.jelechem.2020.114244] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
9
|
Discovery of 2-(2-aminobenzo[d]thiazol-6-yl) benzo[d]oxazol-5-amine derivatives that regulated HPV relevant cellular pathway and prevented cervical cancer from abnormal proliferation. Eur J Med Chem 2020; 204:112556. [PMID: 32739649 DOI: 10.1016/j.ejmech.2020.112556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/19/2020] [Accepted: 06/07/2020] [Indexed: 12/24/2022]
Abstract
Human papillomavirus (HPV) is a well-established etiological factor for cervical cancer, and the expression of oncogenic protein E7 is crucial for carcinogenesis. Herein, virtual screening was performed and 2-(2-aminobenzo[d]thiazol-6-yl) benzo[d]oxazol-5-amine derivatives were designed, synthesized as antineoplastic agents, and evaluated for their anti-tumor activities. Among them, the most promising compound H1 showed specific anti-proliferation ability against HeLa cells (IC50 = 380 nM) as well as excellent inhibition of tumor growth in the HeLa xenograft model without inducing obvious side effects. It is interesting that compound H1 displayed significant inhibition against HPV18-positive cervical cell lines (HeLa) but not for HPV16-positive cervical cell lines (SiHa). Further study demonstrated that a low concentration of compound H1 could lead to a cell cycle blockage at the G1 phase and promote cell apoptosis slightly (8.77%). Compound H1 also exhibited transcription repression, especially those associated with the oncoprotein E7 cellular pathway like E7/Rb/E2F-1/DNMT1, which were essential in tumorigenesis. Proteomics analysis revealed that E7 might be degraded through E3 ubiquitin ligases, which aligned with decreasing expression of E7 following the treatment of compound H1. Taken together, it indicated that compound H1 could be a promising potential agent for cervical cancer treatment.
Collapse
|
10
|
Xin M, Wang HY, Zhang H, Shen Y, Zhang SQ. Synthesis and biological activity of new 2,4,6-trisubstituted triazines as potential phosphoinositide 3-kinase inhibitors. JOURNAL OF CHEMICAL RESEARCH 2020. [DOI: 10.1177/1747519820904844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Twenty-five novel 2,4,6-trisubstituted triazines were synthesized and biologically evaluated. Most of the compounds synthesized showed good antiproliferative activity against HCT-116 and MCF-7. Compounds B18 and B19 showed the best antiproliferative activity. Further study showed B18 and B19 inhibited four phosphoinositide 3-kinase isoforms and mammalian target of rapamycin with good potency. These results demonstrate that 2,4,6-trisubstituted triazines are potentially useful phosphoinositide 3-kinase inhibitors for the development of new anticancer drugs.
Collapse
Affiliation(s)
- Minhang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Hui-Yan Wang
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Hao Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - Ying Shen
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an, P.R. China
| |
Collapse
|
11
|
Morpholine as ubiquitous pharmacophore in medicinal chemistry: Deep insight into the structure-activity relationship (SAR). Bioorg Chem 2020; 96:103578. [PMID: 31978684 DOI: 10.1016/j.bioorg.2020.103578] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/09/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022]
Abstract
Morpholine is a versatile moiety, a privileged pharmacophore and an outstanding heterocyclic motif with wide ranges of pharmacological activities due to different mechanisms of action. The ability of morpholine to enhance the potency of the molecule through molecular interactions with the target protein (kinases) or to modulate the pharmacokinetic properties propelled medicinal chemists and researchers to synthesize morpholine ring by the efficient ways and to incorporate this moiety to develop various lead compounds with diverse therapeutic activities. The present review primarily focused on discussing the most promising synthetic leads containing morpholine ring along with structure-activity relationship (SAR) to reveal the active pharmacophores accountable for anticancer, anti-inflammatory, antiviral, anticonvulsant, antihyperlipidemic, antioxidant, antimicrobial and antileishmanial activity. This review outlines some of the recent effective chemical synthesis for morpholine ring. The review also highlighted the metabolic liability of some clinical drugs containing this nucleus and various researches on modified morpholine to enhance the metabolic stability of drugs as well. Drugs bearing morpholine ring and those under clinical trials are also mentioned with the role of morpholine and their mechanism of action. This review will provide the necessary knowledge base to the medicinal chemists in making strategic structural changes in designing morpholine derivatives.
Collapse
|
12
|
Wang R, Yu S, Zhao X, Chen Y, Yang B, Wu T, Hao C, Zhao D, Cheng M. Design, synthesis, biological evaluation and molecular docking study of novel thieno[3,2-d]pyrimidine derivatives as potent FAK inhibitors. Eur J Med Chem 2019; 188:112024. [PMID: 31923858 DOI: 10.1016/j.ejmech.2019.112024] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 12/13/2019] [Accepted: 12/29/2019] [Indexed: 10/25/2022]
Abstract
A series of 2,7-disubstituted-thieno[3,2-d]pyrimidine derivatives were designed, synthesized and evaluated as novel focal adhesion kinase (FAK) inhibitors. The novel 2,7-disubstituted-thieno[3,2-d]pyrimidine scaffold has been designed as a new kinase inhibitor platform that mimics the bioactive conformation of the well-known diaminopyrimidine motif. Most of the compounds potently suppressed the enzymatic activities of FAK and potently inhibited the proliferation of U-87MG, A-549 and MDA-MB-231 cancer cell lines. Among these derivatives, the optimized compound 26f potently inhibited the enzyme (IC50 = 28.2 nM) and displayed stronger potency than TAE-226 in U-87MG, A-549 and MDA-MB-231 cells, with IC50 values of 0.16, 0.27, and 0.19 μM, respectively. Compound 26f also exhibited relatively less cytotoxicity (IC50 = 3.32 μM) toward a normal human cell line, HK2. According to the flow cytometry results, compound 26f induced the apoptosis of MDA-MB-231 cells in a dose-dependent manner and effectively arrested MDA-MB-231 cells in G0/G1 phase. Further investigations revealed that compound 26f potently suppressed the migration of MDA-MB-231 cells. Collectively, these data support the further development of compound 26f as a lead compound for FAK-targeted anticancer drug discovery.
Collapse
Affiliation(s)
- Ruifeng Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Sijia Yu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xiangxin Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Yixuan Chen
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China; The School of Life Science and Biopharmaceutical, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Bowen Yang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Tianxiao Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Chenzhou Hao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Dongmei Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| |
Collapse
|
13
|
Phosphatidylinositol 3 kinase (PI3K) inhibitors as new weapon to combat cancer. Eur J Med Chem 2019; 183:111718. [DOI: 10.1016/j.ejmech.2019.111718] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022]
|
14
|
Bollini M, Bruno AM, Niño ME, Casal JJ, Sasiambarrena LD, Valdez DAG, Battini L, Puente VR, Lombardo ME. Synthesis, 2D-QSAR Studies and Biological Evaluation of Quinazoline Derivatives as Potent Anti-Trypanosoma cruzi Agents. Med Chem 2019; 15:265-276. [PMID: 30295191 DOI: 10.2174/1573406414666181005145042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 07/24/2018] [Accepted: 08/23/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Chagas disease affects about 7 million people worldwide. Only two drugs are currently available for the treatment for this parasite disease, namely, benznidazol (Bzn) and nifurtimox (Nfx). Both drugs have limited curative power in the chronic phase of the disease. Therefore, continuous research is an urgent need so as to discover novel therapeutic alternatives. OBJECTIVE The development of safer and more efficient therapeutic anti-T. cruzi drugs continues to be a major goal in trypanocidal chemotherapy. METHOD Synthesis, 2D-QSAR and drug-like physicochemical properties of a set of quinazolinone and quinazoline derivatives were studied as trypanocidal agents. All compounds were screened in vitro against Trypanosoma cruzi (Tulahuen strain, Tul 2 stock) epimastigotes and bloodstream trypomastigotes. RESULTS Out of 34 compounds synthesized and tested, six compounds (5a, 5b, 9b, 9h, 13f and 13p) displayed significant activity against both epimastigotes and tripomastigotes, without exerting toxicity on Vero cells. CONCLUSION The antiprotozoal activity of these quinazolinone and quinazoline derivatives represents an interesting starting point for a medicinal chemistry program aiming at the development of novel chemotherapies for Chagas disease.
Collapse
Affiliation(s)
- Mariela Bollini
- Laboratorio de Quimica Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)- CONICET, Godoy Cruz 2390, C1425FQD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Ana M Bruno
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - María E Niño
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Juan J Casal
- Laboratorio de Quimica Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)- CONICET, Godoy Cruz 2390, C1425FQD, Ciudad Autonoma de Buenos Aires, Argentina.,Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Leandro D Sasiambarrena
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Damián A G Valdez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquimica, Departamento de Quimica Organica, Junin 956, C1113AAD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Leandro Battini
- Laboratorio de Quimica Medicinal, Centro de Investigaciones en Bionanociencias (CIBION)- CONICET, Godoy Cruz 2390, C1425FQD, Ciudad Autonoma de Buenos Aires, Argentina
| | - Vanesa R Puente
- Centro de Investigaciones Sobre Porfirinas y Porfirias (CIPYP, UBA-CONICET), Hospital de Clinicas Jose de San Martin, Avenida Cordoba 2351, C1120AAR, Ciudad Autonoma de Buenos Aires, Argentina
| | - María E Lombardo
- Centro de Investigaciones Sobre Porfirinas y Porfirias (CIPYP, UBA-CONICET), Hospital de Clinicas Jose de San Martin, Avenida Cordoba 2351, C1120AAR, Ciudad Autonoma de Buenos Aires, Argentina
| |
Collapse
|
15
|
Martinez-Quiroz M, Aguilar-Martinez XE, Oropeza-Guzman MT, Valdez R, Lopez-Maldonado EA. Evaluation of N-Alkyl-bis- o-aminobenzamide Receptors for the Determination and Separation of Metal Ions by Fluorescence, UV-Visible Spectrometry and Zeta Potential. Molecules 2019; 24:molecules24091737. [PMID: 31060217 PMCID: PMC6539274 DOI: 10.3390/molecules24091737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/13/2019] [Accepted: 04/13/2019] [Indexed: 11/16/2022] Open
Abstract
This paper presents the synthesis and evaluation of physicochemical behavior of a new series of N-alkyl-bis-o-aminobenzamides (BOABs) in aqueous solution. The study was targeted to the complexing capacity of five metal ions (Fe2+, Cu2+, Cd2+, Hg2+ and Pb2+) of environmental concern as the medullar principle of a liquid phase sensor for its application in the determination of these metal ions due to its versatility of use. Molecular fluorescence, UV-visible and Zeta potential were measured for five BOABs and the effect of alkyl groups with different central chain length (n = 3, 4, 6, 8 and 10) on physicochemical performance determined. The results have shown that these derivatives present higher sensibility and selectivity for Cu2+ even in the presence of the other metal ions. An additional application test was done adding a pectin (0.1 wt %) solution to the BOAB-Cu+2 complex to obtain a precipitate (flocs) as a potential selective separation process of Cu from aqueous solution. The solid was then lyophilized and analyzed by SEM-EDS, the images showed spheric forms containing Cu+2 with diameter of approximately of 8 μm and 30 wt %.
Collapse
Affiliation(s)
- Marisela Martinez-Quiroz
- CETYS Universidad, Centro de Innovación y Diseño, Escuela de Ingenieria Av. CETYS Universidad No. 4 Fracc. El Lago, Tijuana, B.C. CP 22210, México.
- Tecnologico Nacional de México, Instituto Tecnológico de Tijuana, Blvd. Alberto Limón Padilla s/n, Mesa de Otay, Tijuana, B.C. CP 22500, México.
| | - Xiomara E Aguilar-Martinez
- Tecnologico Nacional de México, Instituto Tecnológico de Tijuana, Blvd. Alberto Limón Padilla s/n, Mesa de Otay, Tijuana, B.C. CP 22500, México.
| | - Mercedes T Oropeza-Guzman
- Tecnologico Nacional de México, Instituto Tecnológico de Tijuana, Blvd. Alberto Limón Padilla s/n, Mesa de Otay, Tijuana, B.C. CP 22500, México.
| | - Ricardo Valdez
- Centro de Nanociencias y Nanotecnología CNyN-UNAM, Km 107 Carretera Tijuana-Ensenada, Ensenada, B.C. CP 22860, México.
| | - Eduardo A Lopez-Maldonado
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Tijuana, B.C. CP 22390, México.
| |
Collapse
|
16
|
Garces AE, Stocks MJ. Class 1 PI3K Clinical Candidates and Recent Inhibitor Design Strategies: A Medicinal Chemistry Perspective. J Med Chem 2018; 62:4815-4850. [DOI: 10.1021/acs.jmedchem.8b01492] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Aimie E. Garces
- Centre for Biomolecular Sciences, University Park Nottingham, Nottingham NG7 2RD, U.K
| | - Michael J. Stocks
- Centre for Biomolecular Sciences, University Park Nottingham, Nottingham NG7 2RD, U.K
| |
Collapse
|
17
|
Discovery of 2-(aminopyrimidin-5-yl)-4-(morpholin-4-yl)-6- substituted triazine as PI3K and BRAF dual inhibitor. Future Med Chem 2018; 10:2445-2455. [PMID: 30325235 DOI: 10.4155/fmc-2018-0145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
AIM The discovery and development of novel agents simultaneously targeting PI3K/AKT/mammalian target of rapamycin and Ras/RAF/MEK, two signaling pathways, are urgent to improve the curative effect of kinase inhibitors and overcome acquired resistance. METHODS/RESULTS In the present study, 2-(2-aminopyrimidin-5-yl)-4-(morpholin-4-yl)-6-(N-cyclopropyl-N- (1-benzoylpiperidin-4-yl))triazines/pyrimidines were designed as PI3K and BRAF dual inhibitors. The synthesized 20 compounds exhibited potent antiproliferative effects in vitro against HCT116, A375, MCF-7, Colo205, A549 and LOVO cancer cell lines. The tested compounds A6, A7, A9 and A11 remarkably displayed inhibitory activities toward both PI3Kα and BRAFV600E. CONCLUSION These results indicated that our design compounds can serve as potent PI3Kα and BRAFV600E dual inhibitors and effective antiproliferative agents, which can be further optimized to discover more potent PI3Kα and BRAFV600E dual inhibitors.
Collapse
|
18
|
Design, synthesis, bioactivity, and computational studies of some morpholine-clubbed coumarinyl acetamide and cinnamide derivatives. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2018. [DOI: 10.1007/s13738-018-1324-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
19
|
Shagufta, Ahmad I. An insight into the therapeutic potential of quinazoline derivatives as anticancer agents. MEDCHEMCOMM 2017; 8:871-885. [PMID: 30108803 PMCID: PMC6072504 DOI: 10.1039/c7md00097a] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/05/2017] [Indexed: 12/20/2022]
Abstract
Cancer is one of the major causes of worldwide human mortality. A wide range of cytotoxic drugs are available on the market, and several compounds are in different phases of clinical trials. Many studies suggest that these cytotoxic molecules are also associated with different types of adverse side effects; therefore researchers around the globe are involved in the development of more efficient and safer anticancer drugs. In recent years, quinazoline and its derivatives have been considered as a novel class of cancer chemotherapeutic agents that show promising activity against different tumors. The aim of this article is to comprehensively review and highlight the recent developments concerning the anticancer activity of quinazoline derivatives as well as offer perspectives on the development of novel quinazoline derivatives as anticancer agents in the near future.
Collapse
Affiliation(s)
- Shagufta
- Department of Mathematics and Natural Sciences , School of Arts and Sciences , American University of Ras Al Khaimah , Ras Al Khaimah , United Arab Emirates . ;
| | - Irshad Ahmad
- Department of Mathematics and Natural Sciences , School of Arts and Sciences , American University of Ras Al Khaimah , Ras Al Khaimah , United Arab Emirates . ;
| |
Collapse
|
20
|
Xin M, Hei YY, Zhang H, Shen Y, Zhang SQ. Design and synthesis of novel 6-aryl substituted 4-anilinequinazoline derivatives as potential PI3Kδ inhibitors. Bioorg Med Chem Lett 2017; 27:1972-1977. [DOI: 10.1016/j.bmcl.2017.03.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/05/2017] [Accepted: 03/09/2017] [Indexed: 01/22/2023]
|
21
|
Mao B, Gao S, Weng Y, Zhang L, Zhang L. Design, synthesis, and biological evaluation of imidazo[1,2-b]pyridazine derivatives as mTOR inhibitors. Eur J Med Chem 2017; 129:135-150. [PMID: 28235701 DOI: 10.1016/j.ejmech.2017.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 12/18/2022]
Abstract
ATP-competitive mTOR inhibitors have been studied as potential antitumor agents. Based on the structure-activity relationship of known mTOR inhibitors, a series of novel imidazo[1,2-b]pyridazine derivatives were synthesized and characterized. The anti-proliferative activities of these compounds were evaluated by SRB assay against six human cancer cell lines. Imidazo[1,2-b]pyridazine diaryl urea derivatives A15-A24 exhibited significant anti-proliferative activity especially against non-small cell lung cancer A549 and H460 with IC50 values ranging from 0.02 μM to 20.7 μM. Among them, compounds A17 and A18 showed mTOR inhibitory activity with IC50 of 0.067 μM and 0.062 μM, respectively. A more detailed analysis of compounds A17 and A18 showed that they induced G1-phase cell cycle arrest and suppressed the phosphorylation of AKT and S6 at cellular level. Moreover, obvious anticancer effect of A17 in vivo was observed in established nude mice A549 xenograft model.
Collapse
Affiliation(s)
- Beibei Mao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Shanyun Gao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Yiran Weng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China.
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, PR China
| |
Collapse
|
22
|
Synthesis and antitumor activity evaluation of 4,6-disubstituted quinazoline derivatives as novel PI3K inhibitors. Bioorg Med Chem Lett 2016; 26:4408-4413. [DOI: 10.1016/j.bmcl.2016.08.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 07/16/2016] [Accepted: 08/06/2016] [Indexed: 02/06/2023]
|
23
|
Khan I, Zaib S, Batool S, Abbas N, Ashraf Z, Iqbal J, Saeed A. Quinazolines and quinazolinones as ubiquitous structural fragments in medicinal chemistry: An update on the development of synthetic methods and pharmacological diversification. Bioorg Med Chem 2016; 24:2361-2381. [PMID: 27112448 DOI: 10.1016/j.bmc.2016.03.031] [Citation(s) in RCA: 186] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/16/2016] [Accepted: 03/18/2016] [Indexed: 12/17/2022]
Abstract
Nitrogen-rich heterocycles, particularly quinazolines and quinazolinones, represent a unique class of diversified frameworks displaying a broad spectrum of biological functions. Over the past several years, intensive medicinal chemistry efforts have generated numerous structurally functionalized quinazoline and quinazolinone derivatives. Interest in expanding the biological effects, demonstrated by these motifs, is growing exponentially, as indicated by the large number of publications reporting the easy accessibility of these skeletons in addition to the diverse nature of synthetic as well as biological applications. Therefore, the main focus of the present review is to provide an ample but condensed overview on various synthetic approaches providing access to quinazoline and quinazolinone compounds with multifaceted biological activities. Furthermore, mechanistic insights, synthetic utilization, structure-activity relationships and molecular modeling inputs for the potent derivatives have also been discussed.
Collapse
Affiliation(s)
- Imtiaz Khan
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| | - Sumera Zaib
- Department of Biochemistry, Hazara University, Garden Campus, Mansehra, Pakistan; Centre for Advanced Drug Research, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Sadaf Batool
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad 44000, Pakistan
| | - Naeem Abbas
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Zaman Ashraf
- Department of Chemistry, Allama Iqbal Open University, Islamabad 44000, Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
24
|
Peng W, Tu ZC, Long ZJ, Liu Q, Lu G. Discovery of 2-(2-aminopyrimidin-5-yl)-4-morpholino-N-(pyridin-3-yl)quinazolin-7-amines as novel PI3K/mTOR inhibitors and anticancer agents. Eur J Med Chem 2015; 108:644-654. [PMID: 26731167 DOI: 10.1016/j.ejmech.2015.11.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/19/2015] [Accepted: 11/21/2015] [Indexed: 10/22/2022]
Abstract
In this study, a series of novel 7 or 8-substituted 4-morpholine-quinazoline derivatives was designed and synthesized. Their PI3Kα inhibitory activities, antiproliferative activities against seven cancer cell lines, namely, PC-3, DU145, MCF-7, BT474, SK-BR-3, U937 and A431, were evaluated in vitro. Compound 17f proved to be a potential drug candidate with high PI3Kα inhibition activity (IC50 = 4.2 nM) and good antiproliferative activity. Compound 17f was also tested for its inhibitory activities against other kinases, such as PI3Kβ, PI3Kγ, PI3Kδ and mTOR, its effects on p-Akt (S473) and cell cycle. These results suggested that compound 17f could significantly inhibit the PI3K/Akt/mTOR pathway as a potent PI3K inhibitor and anticancer agent.
Collapse
Affiliation(s)
- Wei Peng
- Institute of Drug Synthesis and Pharmaceutical Process, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | - Zheng-Chao Tu
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, PR China
| | - Zi-Jie Long
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, PR China
| | - Quentin Liu
- State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou 510060, PR China
| | - Gui Lu
- Institute of Drug Synthesis and Pharmaceutical Process, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China; Institute of Human Virology, Sun Yat-sen University, Guangzhou 510080, PR China.
| |
Collapse
|
25
|
Synthesis and antitumor activity evaluation of PI3K inhibitors containing 3-substituted quinazolin-4(3H)-one moiety. Bioorg Med Chem 2015; 23:7765-76. [PMID: 26652969 DOI: 10.1016/j.bmc.2015.11.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 11/17/2015] [Accepted: 11/20/2015] [Indexed: 10/22/2022]
Abstract
In present study, a series of N-(2-methoxy-5-(3-substituted quinazolin-4(3H)-one-6-yl)-pyridin-3-yl)phenylsulfonamide were synthesized. Their antiproliferative activities in vitro were evaluated via MTT assay against HCT116 and MCF-7 cancer cell lines. The SAR of title compounds was discussed. The compounds (S)-C5 and (S)-C8 displayed potent inhibitory activity against PI3Ks and mTOR, especially against PI3Kα. In addition, compound (S)-C5 can efficaciously inhibit tumor growth in a mice S-180 model. These findings suggest that our designed compounds can serve as potent PI3K inhibitors and effective anticancer agents.
Collapse
|
26
|
Wang XM, Mao S, Cao L, Xie XX, Xin MH, Lian JF, Cao YX, Zhang SQ. Modification of N -(6-(2-methoxy-3-(4-fluorophenylsulfonamido)pyridin-5-yl)-[1,2,4]triazolo[1,5- a ]pyridin-2-yl)acetamide as PI3Ks inhibitor by replacement of the acetamide group with alkylurea. Bioorg Med Chem 2015. [DOI: 10.1016/j.bmc.2015.07.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
27
|
Xie XX, Li H, Wang J, Mao S, Xin MH, Lu SM, Mei QB, Zhang SQ. Synthesis and anticancer effects evaluation of 1-alkyl-3-(6-(2-methoxy-3-sulfonylaminopyridin-5-yl)benzo[d]thiazol-2-yl)urea as anticancer agents with low toxicity. Bioorg Med Chem 2015; 23:6477-85. [PMID: 26321603 DOI: 10.1016/j.bmc.2015.08.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/12/2015] [Accepted: 08/12/2015] [Indexed: 01/01/2023]
Abstract
As a PI3K and mTOR dual inhibitor, N-(2-chloro-5-(2-acetylaminobenzo[d]thiazol-6-yl)pyridin-3-yl)-4-fluorophenylsulfonamide displays toxicity when orally administrated. In the present study, alkylurea moiety replaced the acetamide group in the compound and a series of 1-alkyl-3-(6-(2,3-disubstituted pyridin-5-yl)benzo[d]thiazol-2-yl)urea derivatives were synthesized. The antiproliferative activities of the synthesized compounds in vitro were evaluated against HCT116, MCF-7, U87 MG and A549 cell lines. The compounds with potent antiproliferative activity were tested for their acute oral toxicity and inhibitory activity against PI3Ks and mTORC1. The results indicate that the compound attached a 2-(dialkylamino)ethylurea moiety at the 2-positeion of benzothiazole can retain the antiproliferative activity and inhibitory activity against PI3K and mTOR. In addition, their acute oral toxicity reduced dramatically. Moreover, compound 2f can effectively inhibit tumor growth in a mice S180 homograft model. These findings suggest that 1-(2-dialkylaminoethyl)-3-(6-(2-methoxy-3-sulfonylaminopyridin-5-yl)benzo[d]thiazol-2-yl)urea derivatives can serve as potent PI3K inhibitors and anticancer agents with low toxicity.
Collapse
Affiliation(s)
- Xiao-Xiao Xie
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Huan Li
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Juan Wang
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, PR China
| | - Shuai Mao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Min-Hang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - She-Min Lu
- Department of Genetics and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Qi-Bing Mei
- Center for Pharmacological Evaluation and Research, Shanghai Institute of Pharmaceutical Industry, Shanghai 200437, PR China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|