1
|
Verma A, Bharatiya P, Jaitak A, Nigam V, Monga V. Advances in the design, discovery, and optimization of aurora kinase inhibitors as anticancer agents. Expert Opin Drug Discov 2025; 20:475-497. [PMID: 40094219 DOI: 10.1080/17460441.2025.2481272] [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: 11/11/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
INTRODUCTION Aurora kinases (AKs) play key roles during carcinogenesis and show a close relationship with many cellular effects including mitotic entry, spindle assembly and chromosomal alignment biorientation. Indeed, elevated levels of AKs have been reported in several different tumor types, leading research scientists to investigate ways that we can target AKs for the purpose of developing new anticancer therapeutics. AREA COVERED This review examines the design, discovery, and development of Aurora kinase inhibitors (AKIs) as anticancer agents and delineates their roles in cancer progression or development. Various databases like PubMed, Scopus, Google scholar, SciFinder were used to search the relevant information. This article provides a comprehensive overview of recent advances in the medicinal chemistry of AKIs including the candidates under clinical development and list of patents filed. In addition, their mechanistic findings, SARs, and in silico studies have also been discussed to offer prospects in this field. EXPERT OPINION The integration of artificial intelligence and computational approaches is poised to accelerate the development of AKIs as anticancer agents. However, the associated challenges currently hindering its impact in drug development must be overcome before drugs can successfully translate from early drug development into clinical practice.
Collapse
Affiliation(s)
- Anubhav Verma
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Pradhuman Bharatiya
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Aashish Jaitak
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Vaibhav Nigam
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | - Vikramdeep Monga
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| |
Collapse
|
2
|
Das R, Chatterjee DR, Kapoor S, Vyas H, Shard A. Novel sulfonamides unveiled as potent anti-lung cancer agents via tumor pyruvate kinase M2 activation. RSC Med Chem 2024; 15:3070-3091. [PMID: 39309364 PMCID: PMC11411637 DOI: 10.1039/d4md00367e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 07/06/2024] [Indexed: 09/25/2024] Open
Abstract
This rational pursuit led to the identification of a novel sulfonamide derivative as a potent anti-lung cancer (LC) compound. Considering these results, we synthesized 38 novel sulfonamide derivatives with diverse skeletal structures. In vitro cytotoxicity assays revealed a potent and selective antiproliferative effect against A549 cells after evaluating a panel of cancer cell lines. Compound 9b has emerged as a potent activator of tumor pyruvate kinase M2 (PKM2), a protein known to play a critical role in LC. Apoptosis assays and cell cycle analysis demonstrated early apoptosis and G2 phase arrest. In silico studies demonstrated interactions between compound 9b and the activator binding site of PKM2. Surface plasmon resonance (SPR) experiments strongly indicated that 9b has a high affinity (K d of 1.378 nM) for PKM2. Furthermore, the increase in reactive oxygen species and decrease in lactate concentration suggested that compound 9b has significant anticancer effects. Notably, the increase in particle size following treatment with 9b suggested the tetramerization of PKM2. This work provides insights that might advance efforts to develop effective non-platinum anticancer agents.
Collapse
Affiliation(s)
- Rudradip Das
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce station Palaj, Gandhinagar Gujarat - 382355 India
| | - Deep Rohan Chatterjee
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce station Palaj, Gandhinagar Gujarat - 382355 India
| | - Saumya Kapoor
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce station Palaj, Gandhinagar Gujarat - 382355 India
| | - Het Vyas
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce station Palaj, Gandhinagar Gujarat - 382355 India
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A) Opposite Airforce station Palaj, Gandhinagar Gujarat - 382355 India
| |
Collapse
|
3
|
Kumar P, Bhalla A. Reaction Pattern and Mechanistic Aspects of Iodine and Iodine-Based Reagents in Selenylation of Aliphatic, Aromatic, and (Hetero)Cyclic Systems. Top Curr Chem (Cham) 2024; 382:12. [PMID: 38589598 DOI: 10.1007/s41061-024-00459-8] [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: 08/17/2023] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Organoselenium compounds have been the subject of extensive research since the discovery of the biologically active compound ebselen. Ebselen has recently been found to show activity against the main protease of the virus responsible for COVID-19. Other organoselenium compounds are also well-known for their diverse biological activities, with such compounds exhibiting interesting physical properties relevant to the fields of electronics, materials, and polymer chemistry. In addition, the incorporation of selenium into various organic molecules has garnered significant attention due to the potential of selenium to enhance the biological activity of these molecules, particularly in conjunction with bioactive heterocycles. Iodine and iodine-based reagents play a prominent role in the synthesis of organoselenium compounds, being valued for their cost-effectiveness, non-toxicity, and ease of handling. These reagents efficiently selenylate a broad range of organic substrates, encompassing alkenes, alkynes, and cyclic, aromatic, and heterocyclic molecules. They serve as catalysts, additives, inducers, and oxidizing agents, facilitating the introduction of different functional groups at alternate positions in the molecules, thereby allowing for regioselective and stereoselective approaches. Specific iodine reagents and their combinations can be tailored to follow the desired reaction pathways. Here, we present a comprehensive review of the progress in the selenylation of organic molecules using iodine reagents over the past decade, with a focus on reaction patterns, solvent effects, heating, microwave, and ultrasonic conditions. Detailed discussions on mechanistic aspects, such as electrophilic, nucleophilic, radical, electrochemical, and ring expansion reactions via selenylation, multiselenylation, and difunctionalization, are included. The review also highlights the formation of various cyclic, heterocyclic, and heteroarenes resulting from the in situ generation of selenium intermediates, encompassing cyclic ketones, cyclic ethers, cyclic lactones, selenophenes, chromones, pyrazolines, pyrrolidines, piperidines, indolines, oxazolines, isooxazolines, lactones, dihydrofurans, and isoxazolidines. To enhance the reader's interest, the review is structured into different sections covering the selenylation of aliphatic sp2/sp carbon and cyclic sp2 carbon, and then is further subdivided into various heterocyclic molecules.
Collapse
Affiliation(s)
- Pankaj Kumar
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, UT, 160014, India
| | - Aman Bhalla
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh, UT, 160014, India.
| |
Collapse
|
4
|
Yevale DB, Teraiya N, Lalwani TD, Ameta RK, Sangani CB. A novel class of pyrazole analogues as aurora kinase A inhibitor: design, synthesis, and anticancer evaluation. Bioorg Chem 2023; 141:106901. [PMID: 37797455 DOI: 10.1016/j.bioorg.2023.106901] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/04/2023] [Accepted: 09/29/2023] [Indexed: 10/07/2023]
Abstract
Pyrazole, as a small molecule, was discovered for higher cytotoxicity and affinity towards Aurora-A kinase. Based on these facts, a novel pyrazole substituted at the 4th position was designed, synthesized, and evaluated against MCF-7, MDA-MB-23, and Vero (non-cancerous kidney cell) cell lines. Compounds5hand5eexhibited greater cytotoxicity in the series against MCF-7 and MDA-MB-231, with GI50 values of 0.12 µM and 0.63 µM, respectively, as compared to Imatinib (GI50 values of 16.08 µM and 10.36 µM). All of the compounds displayed selective cytotoxicity against cancer cells but not on normal Vero cells, supporting the design strategy to be a selective anticancer agent. Furthermore, compounds 5h and 5e inhibited Aurora-A kinase with IC50 values of 0.78 µM (4.70-fold) and 1.12 µM (2.84-fold), respectively, as compared to alisertib (IC50 = 3.36 µM). In addition, compound 5h significantly arrested the cell cycle at G2/M (34.89 %, 5.56-fold) and the apoptotic phase (25.04 %, 11.81-fold) compared to the control. It also triggered an increase in early (7.43 %) and late (14.89 %) phase apoptosis compared to vehicle (0.235 and 0.36 %, respectively), causing 37.89-fold higher total apoptosis in the annexin-V assay. These data imply that Aurora-A kinase inhibition may be linked to apoptosis induction and cell cycle arrest. Furthermore, their higher docking score in the study confirmed evidence of Aurora-kinase suppression. It was observed that fluorine and imidazole increased the H-bond and lipophilic interactions with the binding residue. Also, the substitution of electron-rich and lipophilic groups increased hydrophobic interactions. Moreover, the three-atom linkage (CH2NHCH2) expanded compound 5h to fill the cavity. Based on current findings, it is concluded that compounds 5h and 5e with strong Aurora-A kinase suppression may be promising anticancer agents.
Collapse
Affiliation(s)
- Digambar B Yevale
- Department of Chemistry, Shri M.M Patel Institute of Sciences and Research, Kadi Sarva Vishwavidyalaya, Gandhinagar 382016, Gujarat, India; Piramal Pharma Limited, Plot No. 18, Pharmaceutical Special Economic Zone, Village-Matoda, Taluka-Sanand, Ahmedabad 382213, Gujarat, India
| | - Nishith Teraiya
- Department of Pharmaceutical Chemistry, K B Institute of Pharmaceutical Education and Research, Kadi Sarva Vishvavidhyalay, Gandhinagar, Gujarat 382023, India
| | - Twinkle D Lalwani
- Piramal Pharma Limited, Plot No. 18, Pharmaceutical Special Economic Zone, Village-Matoda, Taluka-Sanand, Ahmedabad 382213, Gujarat, India
| | - Rakesh Kumar Ameta
- Department of Chemistry, Shri M.M Patel Institute of Sciences and Research, Kadi Sarva Vishwavidyalaya, Gandhinagar 382016, Gujarat, India
| | - Chetan B Sangani
- Department of Chemistry, Shri M.M Patel Institute of Sciences and Research, Kadi Sarva Vishwavidyalaya, Gandhinagar 382016, Gujarat, India; Department of Chemistry, Government Science College, Gujarat University, Gandhinagar 382016, Gujarat, India.
| |
Collapse
|
5
|
Peytam F, Emamgholipour Z, Mousavi A, Moradi M, Foroumadi R, Firoozpour L, Divsalar F, Safavi M, Foroumadi A. Imidazopyridine-based kinase inhibitors as potential anticancer agents: A review. Bioorg Chem 2023; 140:106831. [PMID: 37683538 DOI: 10.1016/j.bioorg.2023.106831] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023]
Abstract
Considering the fundamental role of protein kinases in the mechanism of protein phosphorylation in critical cellular processes, their dysregulation, especially in cancers, has underscored their therapeutic relevance. Imidazopyridines represent versatile scaffolds found in abundant bioactive compounds. Given their structural features, imidazopyridines have possessed pivotal potency to interact with different protein kinases, inspiring researchers to carry out numerous structural variations. In this comprehensive review, we encompass an extensive survey of the design and biological evaluations of imidazopyridine-based small molecules as potential agents targeting diverse kinases for anticancer applications. We describe the structural elements critical to inhibitory potency, elucidating their key structure-activity relationships (SAR) and mode of actions, where available. We classify these compounds into two groups: Serine/threonine and Tyrosine inhibitors. By highlighting the promising role of imidazopyridines in kinase inhibition, we aim to facilitate the design and development of more effective, targeted compounds for cancer treatment.
Collapse
Affiliation(s)
- Fariba Peytam
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Emamgholipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Mousavi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahfam Moradi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Roham Foroumadi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Divsalar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Alireza Foroumadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Břehová P, Řezníčková E, Škach K, Jorda R, Dejmek M, Vojáčková V, Šála M, Kovalová M, Dračínský M, Dolníková A, Strmeň T, Kinnertová M, Chalupský K, Dvořáková A, Gucký T, Mertlíková Kaiserová H, Klener P, Nencka R, Kryštof V. Inhibition of FLT3-ITD Kinase in Acute Myeloid Leukemia by New Imidazo[1,2- b]pyridazine Derivatives Identified by Scaffold Hopping. J Med Chem 2023; 66:11133-11157. [PMID: 37535845 PMCID: PMC10461230 DOI: 10.1021/acs.jmedchem.3c00575] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Indexed: 08/05/2023]
Abstract
FLT3 kinase is a potential drug target in acute myeloid leukemia (AML). Patients with FLT3 mutations typically have higher relapse rates and worse outcomes than patients without FLT3 mutations. In this study, we investigated the suitability of various heterocycles as central cores of FLT3 inhibitors, including thieno[3,2-d]pyrimidine, pyrazolo[1,5-a]pyrimidine, imidazo[4,5-b]pyridine, pyrido[4,3-d]pyrimidine, and imidazo[1,2-b]pyridazine. Our assays revealed a series of imidazo[1,2-b]pyridazines with high potency against FLT3. Compound 34f showed nanomolar inhibitory activity against recombinant FLT3-ITD and FLT3-D835Y (IC50 values 4 and 1 nM, respectively) as well as in the FLT3-ITD-positive AML cell lines MV4-11, MOLM-13, and MOLM-13 expressing the FLT3-ITD-D835Y mutant (GI50 values of 7, 9, and 4 nM, respectively). In contrast, FLT3-independent cell lines were much less sensitive. In vitro experiments confirmed suppression of FLT3 downstream signaling pathways. Finally, the treatment of MV4-11 xenograft-bearing mice with 34f at doses of 5 and 10 mg/kg markedly blocked tumor growth without any adverse effects.
Collapse
Affiliation(s)
- Petra Břehová
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Eva Řezníčková
- Department
of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech
Republic
| | - Kryštof Škach
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Radek Jorda
- Department
of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech
Republic
| | - Milan Dejmek
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Veronika Vojáčková
- Department
of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech
Republic
| | - Michal Šála
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Markéta Kovalová
- Department
of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech
Republic
| | - Martin Dračínský
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Alexandra Dolníková
- Institute
of Pathological Physiology, First Faculty of Medicine, Charles University, 12108 Prague, Czech Republic
| | - Timotej Strmeň
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Monika Kinnertová
- Department
of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech
Republic
| | - Karel Chalupský
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Alexandra Dvořáková
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Tomáš Gucký
- Department
of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech
Republic
| | - Helena Mertlíková Kaiserová
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Pavel Klener
- Institute
of Pathological Physiology, First Faculty of Medicine, Charles University, 12108 Prague, Czech Republic
| | - Radim Nencka
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 16000 Prague, Czech Republic
| | - Vladimír Kryštof
- Department
of Experimental Biology, Faculty of Science, Palacký University Olomouc, Šlechtitelů 27, 78371 Olomouc, Czech
Republic
- Institute
of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Hněvotínská 5, 77900 Olomouc, Czech Republic
| |
Collapse
|
7
|
Dupati P, Sapuri‐Butti AR, Musini A, Narayan Singh H. Synthesis of New 2‐(Pyridin‐4‐yl)‐1H‐imidazo[4,5‐
b
]pyridine Derivatives:
In vitro
Antibacterial and
In silico
Screening. ChemistrySelect 2023. [DOI: 10.1002/slct.202204774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Affiliation(s)
- Praveen Dupati
- Department of Chemistry Mahatma Gandhi University Nalgonda Telangana State - 508254 India
| | | | - Anjaneyulu Musini
- Centre for Biotechnology Institute of Science and Technology Jawaharlal Nehru Technological University Hyderabad Telangana State - 500085 India
| | - Himanshu Narayan Singh
- Department of Systems Biology Columbia University Irving Medical Centre 100032 New York USA
| |
Collapse
|
8
|
Khatun S, Singh A, Bader GN, Sofi FA. Imidazopyridine, a promising scaffold with potential medicinal applications and structural activity relationship (SAR): recent advances. J Biomol Struct Dyn 2022; 40:14279-14302. [PMID: 34779710 DOI: 10.1080/07391102.2021.1997818] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Imidazopyridine scaffold has gained tremendous importance over the past few decades. Imidazopyridines have been expeditiously used for the rationale design and development of novel synthetic analogs for various therapeutic disorders. A wide variety of imidazopyridine derivatives have been developed as potential anti-cancer, anti-diabetic, anti-tubercular, anti-microbial, anti-viral, anti-inflammatory, central nervous system (CNS) agents besides other chemotherapeutic agents. Imidazopyridine heterocyclic system acts as a key pharmacophore motif for the identification and optimization of lead structures to increase medicinal chemistry toolbox. The present review highlights the medicinal significances of imidazopyridines for their rationale development as lead molecules with improved therapeutic efficacies. This review further emphasis on the structure-activity relationships (SARs) of the various designed imidazopyridines to establish a relationship between the key structural features versus the biological activities.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Samima Khatun
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Punjab, India
| | - Abhinav Singh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Punjab, India
| | - Ghulam N Bader
- Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Srinagar, J & K, India
| | - Firdoos Ahmad Sofi
- Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Srinagar, J & K, India
| |
Collapse
|
9
|
Recent Advancement in Drug Design and Discovery of Pyrazole Biomolecules as Cancer and Inflammation Therapeutics. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248708. [PMID: 36557840 PMCID: PMC9780894 DOI: 10.3390/molecules27248708] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 11/27/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022]
Abstract
Pyrazole, an important pharmacophore and a privileged scaffold of immense significance, is a five-membered heterocyclic moiety with an extensive therapeutic profile, viz., anti-inflammatory, anti-microbial, anti-anxiety, anticancer, analgesic, antipyretic, etc. Due to the expansion of pyrazolecent red pharmacological molecules at a quicker pace, there is an urgent need to put emphasis on recent literature with hitherto available information to recognize the status of this scaffold for pharmaceutical research. The reported potential pyrazole-containing compounds are highlighted in the manuscript for the treatment of cancer and inflammation, and the results are mentioned in % inhibition of inflammation, % growth inhibition, IC50, etc. Pyrazole is an important heterocyclic moiety with a strong pharmacological profile, which may act as an important pharmacophore for the drug discovery process. In the struggle to cultivate suitable anti-inflammatory and anticancer agents, chemists have now focused on pyrazole biomolecules. This review conceals the recent expansion of pyrazole biomolecules as anti-inflammatory and anticancer agents with an aim to provide better correlation among different research going around the world.
Collapse
|
10
|
Mannem GR, Navudu R, Dubasi N, Mohammed MA, Bollikolla HB. Design, and Synthesis of Aryl Derivatives of Imidazopyridine‐Thiadiazoles as Possible Anticancer Agents. ChemistrySelect 2022. [DOI: 10.1002/slct.202200455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - Ramesh Navudu
- Department of Chemistry Acharya Nagarjuna University Nagar 522 510 Guntur AP-INDIA
| | | | - Mujahid Alam Mohammed
- Dept. of Chemistry College of Sciences King Khalid University, PO Box 9004 Abha 61413 Saudi Arabia
| | - Hari Babu Bollikolla
- Department of Chemistry Acharya Nagarjuna University Nagar 522 510 Guntur AP-INDIA
- Department of Nanotechnology Acharya Nagarjuna University Nagar 522 510 Guntur AP-INDIA
| |
Collapse
|
11
|
Kabi AK, Sravani S, Gujjarappa R, Garg A, Vodnala N, Tyagi U, Kaldhi D, Singh V, Gupta S, Malakar CC. Overview on Biological Activities of Pyrazole Derivatives. MATERIALS HORIZONS: FROM NATURE TO NANOMATERIALS 2022:229-306. [DOI: 10.1007/978-981-16-8399-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
12
|
Machado CB, DA Silva EL, Dias Nogueira BM, DA Silva JBS, DE Moraes Filho MO, Montenegro RC, DE Moraes MEA, Moreira-Nunes CA. The Relevance of Aurora Kinase Inhibition in Hematological Malignancies. CANCER DIAGNOSIS & PROGNOSIS 2021; 1:111-126. [PMID: 35399305 DOI: 10.21873/cdp.10016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 12/26/2022]
Abstract
Aurora kinases are a family of serine/threonine protein kinases that play a central role in eukaryotic cell division. Overexpression of aurora kinases in cancer and their role as major regulators of the cell cycle quickly inspired the idea that their inhibition might be a potential pathway when treating oncologic patients. Over the past couple of decades, the search for designing and testing of molecules capable of inhibiting aurora activities fueled many pre-clinical and clinical studies. In this study, data from the past 10 years of in vitro and in vivo investigations, as well as clinical trials, utilizing aurora kinase inhibitors as therapeutics for hematological malignancies were compiled and discussed, aiming to highlight potential uses of these inhibitors as a novel monotherapy model or alongside conventional chemotherapies. While there is still much to be elucidated, it is clear that these kinases play a key role in oncogenesis, and their manageable toxicity and potentially synergistic effects still render them a focus of interest for future investigations in combinatorial clinical trials.
Collapse
Affiliation(s)
- Caio Bezerra Machado
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Emerson Lucena DA Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Beatriz Maria Dias Nogueira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jean Breno Silveira DA Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Manoel Odorico DE Moraes Filho
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Caroline Aquino Moreira-Nunes
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
13
|
Quizartinib-resistant FLT3-ITD acute myeloid leukemia cells are sensitive to the FLT3-Aurora kinase inhibitor CCT241736. Blood Adv 2021; 4:1478-1491. [PMID: 32282883 DOI: 10.1182/bloodadvances.2019000986] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 02/11/2020] [Indexed: 12/17/2022] Open
Abstract
Internal tandem duplication of FLT3 (FLT3-ITD) is one of the most common somatic mutations in acute myeloid leukemia (AML); it causes constitutive activation of FLT3 kinase and is associated with high relapse rates and poor survival. Small-molecule inhibition of FLT3 represents an attractive therapeutic strategy for this subtype of AML, although resistance from secondary FLT3 tyrosine kinase domain (FLT3-TKD) mutations is an emerging clinical problem. CCT241736 is an orally bioavailable, selective, and potent dual inhibitor of FLT3 and Aurora kinases. FLT3-ITD+ cells with secondary FLT3-TKD mutations have high in vitro relative resistance to the FLT3 inhibitors quizartinib and sorafenib, but not to CCT241736. The mechanism of action of CCT241736 results in significant in vivo efficacy, with inhibition of tumor growth observed in efficacy studies in FLT3-ITD and FLT3-ITD-TKD human tumor xenograft models. The efficacy of CCT241736 was also confirmed in primary samples from AML patients, including those with quizartinib-resistant disease, which induces apoptosis through inhibition of both FLT3 and Aurora kinases. The unique combination of CCT241736 properties based on robust potency, dual selectivity, and significant in vivo activity indicate that CCT241736 is a bona fide clinical drug candidate for FLT3-ITD and TKD AML patients with resistance to current drugs.
Collapse
|
14
|
Wang Z, Cai J, Cheng J, Yang W, Zhu Y, Li H, Lu T, Chen Y, Lu S. FLT3 Inhibitors in Acute Myeloid Leukemia: Challenges and Recent Developments in Overcoming Resistance. J Med Chem 2021; 64:2878-2900. [PMID: 33719439 DOI: 10.1021/acs.jmedchem.0c01851] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene are often present in newly diagnosed acute myeloid leukemia (AML) patients with an incidence rate of approximately 30%. Recently, many FLT3 inhibitors have been developed and exhibit positive preclinical and clinical effects against AML. However, patients develop resistance soon after undergoing FLT3 inhibitor treatment, resulting in short durable responses and poor clinical effects. This review will discuss the main mechanisms of resistance to clinical FLT3 inhibitors and summarize the emerging strategies that are utilized to overcome drug resistance. Basically, medicinal chemistry efforts to develop new small-molecule FLT3 inhibitors offer a direct solution to this problem. Other potential strategies include the combination of FLT3 inhibitors with other therapies and the development of multitarget inhibitors. It is hoped that this review will provide inspiring insights into the discovery of new AML therapies that can eventually overcome the resistance to current FLT3 inhibitors.
Collapse
Affiliation(s)
- Zhijie Wang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jiongheng Cai
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Jie Cheng
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Wenqianzi Yang
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Yifan Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Hongmei Li
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Tao Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P.R. China
| | - Yadong Chen
- Laboratory of Molecular Design and Drug Discovery, China Pharmaceutical University, Nanjing, 211198, P.R. China
| | - Shuai Lu
- School of Science, China Pharmaceutical University, Nanjing 211198, P.R. China
| |
Collapse
|
15
|
Reddy JS, Chen CM, Coumar MS, Sun HY, Sun N, Hsieh HP. Development of a Robust Scale-Up Synthetic Route for BPR1K871: A Clinical Candidate for the Treatment of Acute Myeloid Leukemia and Solid Tumors. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.0c00515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Julakanti Satyanarayana Reddy
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan, ROC
| | - Chih-Ming Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan, ROC
| | - Mohane Selvaraj Coumar
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry 605014, India
| | - Hsu-Yi Sun
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan, ROC
| | - Na Sun
- China Gateway Pharmaceutical Development Co, Shanghai 201417, China
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan, ROC
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan, ROC
- Biomedical Translation Research Centre, Academia Sinica, Taipei 11520, Taiwan, ROC
| |
Collapse
|
16
|
Doganc F, Alp M, Karabay A, Koç A, Eren G, Göker H. Synthesis and Cytotoxicity of Some Imidazo[4,5‐
b
]pyridine Derivatives and Their Regioselective
N
‐Alkylation. ChemistrySelect 2021. [DOI: 10.1002/slct.202004584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Fatima Doganc
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy Ankara University 06100 Tandogan, Ankara Turkey
| | - Mehmet Alp
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy Ankara University 06100 Tandogan, Ankara Turkey
| | - Arzu Karabay
- Department of Biochemistry, Faculty of Pharmacy Ankara University 06100 Tandogan, Ankara Turkey
| | - Aslı Koç
- Department of Biochemistry, Faculty of Pharmacy Ankara University 06100 Tandogan, Ankara Turkey
| | - Gokcen Eren
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy Gazi University 06330 Etiler, Ankara Turkey
| | - Hakan Göker
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy Ankara University 06100 Tandogan, Ankara Turkey
| |
Collapse
|
17
|
Mologni L, Marzaro G, Redaelli S, Zambon A. Dual Kinase Targeting in Leukemia. Cancers (Basel) 2021; 13:E119. [PMID: 33401428 PMCID: PMC7796318 DOI: 10.3390/cancers13010119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Pharmacological cancer therapy is often based on the concurrent inhibition of different survival pathways to improve treatment outcomes and to reduce the risk of relapses. While this strategy is traditionally pursued only through the co-administration of several drugs, the recent development of multi-targeting drugs (i.e., compounds intrinsically able to simultaneously target several macromolecules involved in cancer onset) has had a dramatic impact on cancer treatment. This review focuses on the most recent developments in dual-kinase inhibitors used in acute myeloid leukemia (AML), chronic myelogenous leukemia (CML), and lymphoid tumors, giving details on preclinical studies as well as ongoing clinical trials. A brief overview of dual-targeting inhibitors (kinase/histone deacetylase (HDAC) and kinase/tubulin polymerization inhibitors) applied to leukemia is also given. Finally, the very recently developed Proteolysis Targeting Chimeras (PROTAC)-based kinase inhibitors are presented.
Collapse
Affiliation(s)
- Luca Mologni
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.M.); (S.R.)
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, via Marzolo 5, I-35131 Padova, Italy;
| | - Sara Redaelli
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (L.M.); (S.R.)
| | - Alfonso Zambon
- Department of Chemistry and Geological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
18
|
Li Y, Huang S, Li J, Li J, Ji X, Liu J, Chen L, Peng S, Zhang K. Access to 2-pyridinylamide and imidazopyridine from 2-fluoropyridine and amidine hydrochloride. Org Biomol Chem 2020; 18:9292-9299. [PMID: 33164006 DOI: 10.1039/d0ob01904f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Under catalyst-free conditions, an efficient method to synthesize 2-pyridinylamides has been developed, and the protocol uses inexpensive and readily available 2-fluoropyridine and amidine derivatives as the starting materials. Simultaneously, the copper-catalysed approach to imidazopyridine derivatives has been established with high chemoselectivity and regiospecificity. The results suggest that the nitrogen-heterocycles containing iodide substituents can also be compatible for the reaction via the cascade Ullmann-type coupling, and the nucleophilic substitution reaction provides the target products in a one-pot manner.
Collapse
Affiliation(s)
- Yibiao Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020 China.
| | - Shuo Huang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020 China.
| | - Jiaming Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020 China.
| | - Jian Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020 China.
| | - Xiaoliang Ji
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020 China.
| | - Jiasheng Liu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020 China.
| | - Lu Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020 China.
| | - Shiyong Peng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020 China.
| | - Kun Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, 529020 China.
| |
Collapse
|
19
|
Chojnacki K, Lindenblatt D, Wińska P, Wielechowska M, Toelzer C, Niefind K, Bretner M. Synthesis, biological properties and structural study of new halogenated azolo[4,5-b]pyridines as inhibitors of CK2 kinase. Bioorg Chem 2020; 106:104502. [PMID: 33317841 DOI: 10.1016/j.bioorg.2020.104502] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/06/2020] [Accepted: 11/19/2020] [Indexed: 11/26/2022]
Abstract
The new halogenated 1H-triazolo[4,5-b]pyridines and 1H-imidazo[4,5-b]pyridines were synthesised as analogues of known CK2 inhibitors: 4,5,6,7-tetrabromo-1H-benzotriazole (TBBt) and 4,5,6,7-tetrabromo-1H-benzimidazole (TBBi). Their influence on the activity of recombinant human CK2α, CK2α' and PIM1 kinases was determined. The most active inhibitors were di- and trihalogenated 1H-triazolo[4,5-b]pyridines (4a, 5a and 10a) with IC50 values 2.56, 3.82 and 3.26 μM respectively for CK2α. Furthermore, effect on viability of cancer cell lines MCF-7 (human breast adenocarcinoma) and CCRF-CEM (T lymphoblast leukemia) of all final compounds was evaluated. Finally, three crystal structures of complexes of CK2α1-335 with inhibitors 4a, 5a and 10a were obtained. In addition, new protocol was used to obtain high-resolution crystal structures of CK2α'Cys336Ser in complex with four inhibitors (4a, 5a, 5b, 10a).
Collapse
Affiliation(s)
- K Chojnacki
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland.
| | - D Lindenblatt
- Department für Chemie, Institut für Biochemie, Universtät zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany
| | - P Wińska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - M Wielechowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - C Toelzer
- Department für Chemie, Institut für Biochemie, Universtät zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany
| | - K Niefind
- Department für Chemie, Institut für Biochemie, Universtät zu Köln, Zülpicher Straße 47, D-50674 Köln, Germany
| | - M Bretner
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| |
Collapse
|
20
|
Malik HS, Bilal A, Ullah R, Iqbal M, Khan S, Ahmed I, Krohn K, Saleem RSZ, Hussain H, Faisal A. Natural and Semisynthetic Chalcones as Dual FLT3 and Microtubule Polymerization Inhibitors. JOURNAL OF NATURAL PRODUCTS 2020; 83:3111-3121. [PMID: 32975953 DOI: 10.1021/acs.jnatprod.0c00699] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Activating mutations in FLT3 receptor tyrosine kinase are found in a third of acute myeloid leukemia (AML) patients and are associated with disease relapse and a poor prognosis. The majority of these mutations are internal tandem duplications (ITDs) in the juxtamembrane domain of FLT3, which have been validated as a therapeutic target. The clinical success of selective inhibitors targeting oncogenic FLT3, however, has been limited due to the acquisition of drug resistance. Herein the identification of a dual FLT3/microtubule polymerization inhibitor, chalcone 4 (2'-allyloxy-4,4'-dimethoxychalcone), is reported through screening of 15 related chalcones for differential antiproliferative activity in leukemia cell lines dependent on FLT3-ITD (MV-4-11) or BCR-ABL (K562) oncogenes and by subsequent screening for mitotic inducers in the HCT116 cell line. Three natural chalcones (1-3) were found to be differentially more potent toward the MV-4-11 (FLT3-ITD) cell line compared to the K562 (BCR-ABL) cell line. Notably, the new semisynthetic chalcone 4, which is a 2'-O-allyl analogue of the natural chalcone 3, was found to be more potent toward the FLT3-ITD+ cell line and inhibited FLT3 signaling in FLT3-dependent cells. An in vitro kinase assay confirmed that chalcone 4 directly inhibited FLT3. Moreover, chalcone 4 induced mitotic arrest in these cells and inhibited tubulin polymerization in both cellular and biochemical assays. Treatment of MV-4-11 cells with this inhibitor for 24 and 48 h resulted in apoptotic cell death. Finally, chalcone 4 was able to overcome TKD mutation-mediated acquired resistance to FLT3 inhibitors in a MOLM-13 cell line expressing FLT3-ITD with the D835Y mutation. Chalcone 4 is, therefore, a promising lead for the discovery of dual-target FLT3 inhibitors.
Collapse
Affiliation(s)
- Haleema Sadia Malik
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Aishah Bilal
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Rahim Ullah
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Maheen Iqbal
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Sardraz Khan
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Ishtiaq Ahmed
- Department of Chemistry, University of Paderborn, Warburger Straße 100, 33098 Paderborn, Germany
| | - Karsten Krohn
- Department of Chemistry, University of Paderborn, Warburger Straße 100, 33098 Paderborn, Germany
| | - Rahman Shah Zaib Saleem
- Department of Chemistry and Chemical Engineering, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - Hidayat Hussain
- Department of Chemistry, University of Paderborn, Warburger Straße 100, 33098 Paderborn, Germany
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Weinberg 3, D-06120 Halle (Saale), Germany
| | - Amir Faisal
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
21
|
Liu M, Ju X, Zou J, Shi J, Jia G. Recent researches for dual Aurora target inhibitors in antitumor field. Eur J Med Chem 2020; 203:112498. [PMID: 32693295 DOI: 10.1016/j.ejmech.2020.112498] [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: 03/21/2020] [Revised: 05/05/2020] [Accepted: 05/28/2020] [Indexed: 11/17/2022]
Abstract
Non-infectious and chronic diseases such as malignant tumors are now one of the main causes of human death. Its occurrence is a multi-factor, multi-step complex process with biological characteristics such as cell differentiation, abnormal proliferation, uncontrolled growth, and metastasis. It has been found that a variety of human malignant tumors are accompanied by over-expression and proliferation of Aurora kinase, which causes abnormalities in the mitotic process and is related to the instability of the genome that causes tumors. Therefore, the use of Aurora kinase inhibitors to target tumors is becoming a research hotspot. However, in cancer, because of the complexity of signal transduction system and the participation of different proteins and enzymes, the anticancer effect of selective single-target drugs is limited. After inhibiting one pathway, signal molecules can be conducted through other pathways, resulting in poor therapeutic effect of single-target drug treatment. Multi-target drugs can solve this problem very well. It can regulate the various links that cause disease at the same time without completely eliminating the relationship between the signal transmission systems, and it is not easy to cause drug resistance. Currently, studies have shown that Aurora dual-target inhibitors generated with the co-inhibition of Aurora and another target (such as CDK, PLK, JAK2, etc.) have better therapeutic effects on tumors. In this paper, we reviewed the studies of dual Aurora inhibitors that have been discovered in recent years.
Collapse
Affiliation(s)
- Maoyu Liu
- The Ministry of Education Key Laboratory of Standardization of Chinese Herbal Medicines of Ministry, State Key Laboratory Breeding Base of Systematic Research, Development and Utilization of Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xueming Ju
- Department of Ultrasound, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jing Zou
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
22
|
Fernandes ÍA, Braga Resende D, Ramalho TC, Kuca K, da Cunha EFF. Theoretical Studies Aimed at Finding FLT3 Inhibitors and a Promising Compound and Molecular Pattern with Dual Aurora B/FLT3 Activity. Molecules 2020; 25:molecules25071726. [PMID: 32283751 PMCID: PMC7181172 DOI: 10.3390/molecules25071726] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 01/16/2023] Open
Abstract
FLT3 and dual Aurora B/FLT3 inhibitors have shown relevance in the search for promising new anticancer compounds, mainly for acute myeloid leukemia (AML). This study was designed to investigate the interactions between human FLT3 in the kinase domain with several indolin-2-one derivatives, structurally similar to Sunitinib. Molegro Virtual Docker (MVD) software was utilized in docking analyses. The predicted model of the training group, considering nineteen amino acid residues, performed in Chemoface, achieved an R2 of 0.82, suggesting that the binding conformations of the ligands with FLT3 are reasonable, and the data can be used to predict the interaction energy of other FLT3 inhibitors with similar molecular patterns. The MolDock Score for energy for compound 1 showed more stable interaction energy (-233.25 kcal mol-1) than the other inhibitors studied, while Sunitinib presented as one of the least stable (-160.94 kcal mol-1). Compounds IAF70, IAF72, IAF75, IAF80, IAF84, and IAF88 can be highlighted as promising derivatives for synthesis and biological evaluation against FLT3. Furthermore, IAF79 can be considered to be a promising dual Aurora B/FLT3 inhibitor, and its molecular pattern can be exploited synthetically to search for new indolin-2-one derivatives that may become drugs used in the treatment of cancers, including AML.
Collapse
Affiliation(s)
- Ítalo Antônio Fernandes
- Department of Chemistry, Federal University of Lavras, P.O. Box 3037, Lavras-MG 37200-000, Brazil; (I.A.F.); (T.C.R.)
| | - Déborah Braga Resende
- Department of Veterinary Medicine, Federal University of Lavras, P.O. Box 3037, Lavras-MG 37200-000, Brazil;
| | - Teodorico Castro Ramalho
- Department of Chemistry, Federal University of Lavras, P.O. Box 3037, Lavras-MG 37200-000, Brazil; (I.A.F.); (T.C.R.)
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Faculty of Science, Department of Chemistry, University of Hradec Kralove, Rokitanskeho 62, 500 03 Hradec Kralove, Czech Republic
- Correspondence: (K.K.); (E.F.F.d.C.)
| | - Elaine Fontes Ferreira da Cunha
- Department of Chemistry, Federal University of Lavras, P.O. Box 3037, Lavras-MG 37200-000, Brazil; (I.A.F.); (T.C.R.)
- Correspondence: (K.K.); (E.F.F.d.C.)
| |
Collapse
|
23
|
IODVA1, a guanidinobenzimidazole derivative, targets Rac activity and Ras-driven cancer models. PLoS One 2020; 15:e0229801. [PMID: 32163428 PMCID: PMC7067412 DOI: 10.1371/journal.pone.0229801] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/13/2020] [Indexed: 12/17/2022] Open
Abstract
We report the synthesis and preliminary characterization of IODVA1, a potent small molecule that is active in xenograft mouse models of Ras-driven lung and breast cancers. In an effort to inhibit oncogenic Ras signaling, we combined in silico screening with inhibition of proliferation and colony formation of Ras-driven cells. NSC124205 fulfilled all criteria. HPLC analysis revealed that NSC124205 was a mixture of at least three compounds, from which IODVA1 was determined to be the active component. IODVA1 decreased 2D and 3D cell proliferation, cell spreading and ruffle and lamellipodia formation through downregulation of Rac activity. IODVA1 significantly impaired xenograft tumor growth of Ras-driven cancer cells with no observable toxicity. Immuno-histochemistry analysis of tumor sections suggests that cell death occurs by increased apoptosis. Our data suggest that IODVA1 targets Rac signaling to induce death of Ras-transformed cells. Therefore, IODVA1 holds promise as an anti-tumor therapeutic agent.
Collapse
|
24
|
Fan C, Zhong T, Yang H, Yang Y, Wang D, Yang X, Xu Y, Fan Y. Design, synthesis, biological evaluation of 6-(2-amino-1H-benzo[d]imidazole-6-yl)quinazolin-4(3H)-one derivatives as novel anticancer agents with Aurora kinase inhibition. Eur J Med Chem 2020; 190:112108. [PMID: 32058239 DOI: 10.1016/j.ejmech.2020.112108] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/11/2020] [Accepted: 01/28/2020] [Indexed: 02/07/2023]
Abstract
Aurora A kinase, a member of the Aurora kinase family, is frequently overexpressed in various human cancers. In addition, Overexpression of Aurora A kinase is associated with drug resistance and poor prognosis in many cancers including breast cancer. Therefore, Aurora A kinase has been considered as an attractive anticancer target for the treatment of human cancers. Herein, A series of 6-(2-amino-1H-benzo[d]imidazole-6-yl)quinazolin-4(3H)-one derivatives were designed, synthesized, and evaluated as Aurora A kinase inhibitors. The cell-based cytotoxicity assays showed that compound 16h was the most potent cytotoxic agent against all tested cancer cells and had a lower IC50 value than ENMD-2076 against MDA-MB-231 cells. Meanwhile, Aurora A kinase assay and Western blot analysis showed that 16h inhibited Aurora A kinase with an IC50 value of 21.94 nM and suppressed the phosphorylation of Histone H3 on Ser10 and Aurora A kinase on Thr288, which were consistent with the activation of Aurora A kinase. Accordingly, 16h caused aberrant mitotic phenotypes and obvious G2/M phase arrest in MDA-MB-231 cells and induced caspase-dependent apoptosis in MDA-MB-231 cells. These results demonstrated that 16h is a potential candidate for the development of anticancer agents targeting Aurora A kinase.
Collapse
Affiliation(s)
- Chengcheng Fan
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China; Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ting Zhong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Huarong Yang
- The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Ying Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Daoping Wang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Xiaosheng Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China
| | - Yongnan Xu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yanhua Fan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang 550014, China.
| |
Collapse
|
25
|
Wood FL, Shepherd S, Hayes A, Liu M, Grira K, Mok Y, Atrash B, Faisal A, Bavetsias V, Linardopoulos S, Blagg J, Raynaud FI. Metabolism of the dual FLT-3/Aurora kinase inhibitor CCT241736 in preclinical and human in vitro models: Implication for the choice of toxicology species. Eur J Pharm Sci 2019; 139:104899. [PMID: 30953752 PMCID: PMC6892276 DOI: 10.1016/j.ejps.2019.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/26/2019] [Accepted: 04/02/2019] [Indexed: 02/02/2023]
Abstract
CCT241736 is a dual fms-like tyrosine kinase 3 (FLT3)/Aurora kinase inhibitor in development for the treatment of acute myeloid leukaemia. The successful development of any new drug relies on adequate safety testing including preclinical toxicology studies. Selection of an appropriate preclinical species requires a thorough understanding of the compound's metabolic clearance and pathways, as well as other pharmacokinetic and pharmacodynamic considerations. In addition, elucidation of the metabolising enzymes in human facilitates improved clinical prediction based on population pharmacokinetics and can inform drug-drug interaction studies. Intrinsic clearance (CLint) determination and metabolite profiling of CCT241736 in human and four preclinical species (dog, minipig, rat and mouse) was undertaken in cryopreserved hepatocytes and liver microsomes. Recombinant human cytochrome P450 bactosomes (rCYP) were utilised to provide reaction phenotyping data and support prediction of metabolic pathways. CCT241736 exhibited low CLint in both hepatocytes and liver microsomes of human, dog, minipig and rat, but considerably higher CLint in mouse. CYP3A4 and CYP3A5 were identified as the major enzymes responsible for biotransformation of CCT241736 in human, exclusively forming five out of seven metabolites. Minipig showed greatest similarity to human with regard to both overall metabolic profile and abundance of specific metabolites relative to parent compound, and is therefore proposed as the most appropriate toxicological species. The greatest disparity was observed between human and dog. Based on metabolic profile, either mouse or rat is a suitable rodent species for toxicology studies.
Collapse
Affiliation(s)
- Francesca L Wood
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Sam Shepherd
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Angela Hayes
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Manjuan Liu
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Katia Grira
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Yi Mok
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Butrus Atrash
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Amir Faisal
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Vassilios Bavetsias
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Spiros Linardopoulos
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Florence I Raynaud
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
26
|
Casari I, Domenichini A, Sestito S, Capone E, Sala G, Rapposelli S, Falasca M. Dual PDK1/Aurora Kinase A Inhibitors Reduce Pancreatic Cancer Cell Proliferation and Colony Formation. Cancers (Basel) 2019; 11:cancers11111695. [PMID: 31683659 PMCID: PMC6896057 DOI: 10.3390/cancers11111695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/26/2019] [Accepted: 10/28/2019] [Indexed: 01/01/2023] Open
Abstract
Deregulation of different intracellular signaling pathways is a common feature in cancer. Numerous studies indicate that persistent activation of the phosphoinositide 3-kinase (PI3K) pathway is often observed in cancer cells. 3-phosphoinositide dependent protein kinase-1 (PDK1), a transducer protein that functions downstream of PI3K, is responsible for the regulation of cell proliferation and migration and it also has been found to play a key role in different cancers, pancreatic and breast cancer amongst others. As PI3K is being described to be aberrantly expressed in several cancer types, designing inhibitors targeting various downstream molecules of PI3K has been the focus of anticancer agent development for a long time. In particular, dual inhibitory drugs targeting key signaling molecules in the PI3K pathway have attracted the attention of scientists. Several drugs have progressed to clinical trials, with limited success due to toxicity and bioavailability concerns. Very few anticancer drugs targeting the PI3K pathway have been approved for clinical use and their efficacy is particularly limited towards certain tumors such as pancreatic cancer. Here, we tested two drugs displaying dual inhibitory activity towards PDK1 and Aurora kinase A in a panel of pancreatic cancer cell lines and in two in vivo models of pancreatic cancer. Our data show that both inhibitors are able to impair cell proliferation and clonogenic potential in pancreatic cancer cells. However, the limited activity of both compounds in vivo indicates that further optimization of the pharmacokinetics properties is required.
Collapse
Affiliation(s)
- Ilaria Casari
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia.
| | - Alice Domenichini
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia.
| | - Simona Sestito
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy.
| | - Emily Capone
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University "G. d'Annunzio" di Chieti-Pescara, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy.
| | - Gianluca Sala
- Dipartimento di Scienze Mediche, Orali e Biotecnologiche, University "G. d'Annunzio" di Chieti-Pescara, Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy.
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy.
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Australia.
| |
Collapse
|
27
|
Yuan T, Qi B, Jiang Z, Dong W, Zhong L, Bai L, Tong R, Yu J, Shi J. Dual FLT3 inhibitors: Against the drug resistance of acute myeloid leukemia in recent decade. Eur J Med Chem 2019; 178:468-483. [PMID: 31207462 DOI: 10.1016/j.ejmech.2019.06.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/16/2019] [Accepted: 06/02/2019] [Indexed: 01/18/2023]
Abstract
Acute myeloid leukemia (AML) is a malignant disease characterized by abnormal growth and differentiation of hematopoietic stem cells. Although the pathogenesis has not been fully elucidated, many specific gene mutations have been found in AML. Fms-like tyrosine kinase 3 (FLT3) is recognized as a drug target for the treatment of AML, and the activation mutations of FLT3 were found in about 30% of AML patients. Targeted inhibition of FLT3 receptor tyrosine kinase has shown promising results in the treatment of FLT3 mutation AML. Unfortunately, the therapeutic effects of FLT3 tyrosine kinase inhibitors used as AML monotherapy are usually accompanied by the high risk of resistance development within a few months after treatment. FLT3 dual inhibitors were generated with the co-inhibition of FLT3 and another target, such as CDK4, JAK2, MEK, Mer, Pim, etc., to solve the problems mentioned above. As a result, the therapeutic effect of the drug is significantly improved, while the toxic and side effects are reduced. Besides, the life quality of AML patients with FLT3 mutation has been effectively improved. In this paper, we reviewed the studies of dual FLT3 inhibitors that have been discovered in recent years for the treatment of AML.
Collapse
Affiliation(s)
- Ting Yuan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Baowen Qi
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Zhongliang Jiang
- Miller School of Medicine, University of Miami, Miami, Florida, 33136, USA
| | - Wenjuan Dong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lei Zhong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lan Bai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jiying Yu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
28
|
Meyers J, Chessum NEA, Ali S, Mok NY, Wilding B, Pasqua AE, Rowlands M, Tucker MJ, Evans LE, Rye CS, O’Fee L, Le Bihan YV, Burke R, Carter M, Workman P, Blagg J, Brown N, van Montfort RLM, Jones K, Cheeseman MD. Privileged Structures and Polypharmacology within and between Protein Families. ACS Med Chem Lett 2018; 9:1199-1204. [PMID: 30613326 PMCID: PMC6295861 DOI: 10.1021/acsmedchemlett.8b00364] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/16/2018] [Indexed: 12/31/2022] Open
Abstract
Polypharmacology is often a key contributor to the efficacy of a drug, but is also a potential risk. We investigated two hits discovered via a cell-based phenotypic screen, the CDK9 inhibitor CCT250006 (1) and the pirin ligand CCT245232 (2), to establish methodology to elucidate their secondary protein targets. Using computational pocket-based analysis, we discovered intrafamily polypharmacology for our kinase inhibitor, despite little overall sequence identity. The interfamily polypharmacology of 2 with B-Raf was used to discover a novel pirin ligand from a very small but privileged compound library despite no apparent ligand or binding site similarity. Our data demonstrates that in areas of drug discovery where intrafamily polypharmacology is often an issue, ligand dissimilarity cannot necessarily be used to assume different off-target profiles and that understanding interfamily polypharmacology will be important in the future to reduce the risk of idiopathic toxicity and in the design of screening libraries.
Collapse
Affiliation(s)
- Joshua Meyers
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Nicola E. A. Chessum
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Salyha Ali
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - N. Yi Mok
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Birgit Wilding
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - A. Elisa Pasqua
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Martin Rowlands
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Michael J. Tucker
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Lindsay E. Evans
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Carl S. Rye
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Lisa O’Fee
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Yann-Vaï Le Bihan
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Rosemary Burke
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Michael Carter
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul Workman
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Julian Blagg
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Nathan Brown
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Rob L. M. van Montfort
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Keith Jones
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Matthew D. Cheeseman
- Cancer
Research UK Cancer Therapeutics Unit and Division of Structural Biology, The Institute of Cancer Research, London SW7 3RP, United Kingdom
| |
Collapse
|
29
|
Uras IZ, Maurer B, Nebenfuehr S, Zojer M, Valent P, Sexl V. Therapeutic Vulnerabilities in FLT3-Mutant AML Unmasked by Palbociclib. Int J Mol Sci 2018; 19:ijms19123987. [PMID: 30544932 PMCID: PMC6321303 DOI: 10.3390/ijms19123987] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/07/2018] [Indexed: 12/25/2022] Open
Abstract
While significant progress has been made in the treatment of acute myeloid leukemia (AML), not all patients can be cured. Mutated in about 1/3 of de novo AML, the FLT3 receptor tyrosine kinase is an attractive target for drug development, activating mutations of the FLT3 map to the juxtamembrane domain (internal tandem duplications, ITD) or the tyrosine kinase domain (TKD), most frequently at codon D835. While small molecule tyrosine kinase inhibitors (TKI) effectively target ITD mutant forms, those on the TKD are not responsive. Moreover, FLT3 inhibition fails to induce a persistent response in patients due to mutational resistance. More potent compounds with broader inhibitory effects on multiple FLT3 mutations are highly desirable. We describe a critical role of CDK6 in the survival of FLT3+ AML cells as palbociclib induced apoptosis not only in FLT3–ITD+ cells but also in FLT3–D835Y+ cells. Antineoplastic effects were also seen in primary patient-derived cells and in a xenograft model, where therapy effectively suppressed tumor formation in vivo at clinically relevant concentrations. In cells with FLT3–ITD or -TKD mutations, the CDK6 protein not only affects cell cycle progression but also transcriptionally regulates oncogenic kinases mediating intrinsic drug resistance, including AURORA and AKT—a feature not shared by its homolog CDK4. While AKT and AURORA kinase inhibitors have significant therapeutic potential in AML, single agent activity has not been proven overly effective. We describe synergistic combination effects when applying these drugs together with palbociclib which could be readily translated to patients with AML bearing FLT3–ITD or –TKD mutations. Targeting synergistically acting vulnerabilities, with CDK6 being the common denominator, may represent a promising strategy to improve AML patient responses and to reduce the incidence of selection of resistance-inducing mutations.
Collapse
Affiliation(s)
- Iris Z Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Barbara Maurer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| | - Peter Valent
- Ludwig Boltzmann Institute for Hematology & Oncology, Medical University of Vienna, 1090 Vienna, Austria.
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria.
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria.
| |
Collapse
|
30
|
Padmaja RD, Devi C. V, Mukku N, Chanda K, Maiti B. Rapid Construction of an Imidazo[4,5- b]pyridine Skeleton from 2-Chloro-3-nitropyridine via Tandem Reaction in H 2O-IPA Medium. ACS OMEGA 2018; 3:4583-4590. [PMID: 31458681 PMCID: PMC6641603 DOI: 10.1021/acsomega.8b00426] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/13/2018] [Indexed: 05/05/2023]
Abstract
A highly efficient, clean, and simple procedure for the synthesis of a privilege imidazo[4,5-b]pyridine scaffold from 2-chloro-3-nitropyridine in combination with environmentally benign H2O-IPA as a green solvent is presented. The scope of the novel method has been demonstrated through the tandem sequence of SNAr reaction with substituted primary amines followed by the in situ nitro group reduction and subsequent heteroannulation with substituted aromatic aldehydes to obtain functionalized imidazo[4,5-b]pyridines with only one chromatographic purification step. The synthesis pathway appears to be green, simple, and superior compared with other already reported procedures, with the high abundance of reagents and great ability in expanding the structural diversity.
Collapse
|
31
|
Hsu YC, Coumar MS, Wang WC, Shiao HY, Ke YY, Lin WH, Kuo CC, Chang CW, Kuo FM, Chen PY, Wang SY, Li AS, Chen CH, Kuo PC, Chen CP, Wu MH, Huang CL, Yen KJ, Chang YI, Hsu JTA, Chen CT, Yeh TK, Song JS, Shih C, Hsieh HP. Discovery of BPR1K871, a quinazoline based, multi-kinase inhibitor for the treatment of AML and solid tumors: Rational design, synthesis, in vitro and in vivo evaluation. Oncotarget 2018; 7:86239-86256. [PMID: 27863392 PMCID: PMC5349910 DOI: 10.18632/oncotarget.13369] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/07/2016] [Indexed: 12/15/2022] Open
Abstract
The design and synthesis of a quinazoline-based, multi-kinase inhibitor for the treatment of acute myeloid leukemia (AML) and other malignancies is reported. Based on the previously reported furanopyrimidine 3, quinazoline core containing lead 4 was synthesized and found to impart dual FLT3/AURKA inhibition (IC50 = 127/5 nM), as well as improved physicochemical properties. A detailed structure-activity relationship study of the lead 4 allowed FLT3 and AURKA inhibition to be finely tuned, resulting in AURKA selective (5 and 7; 100-fold selective over FLT3), FLT3 selective (13; 30-fold selective over AURKA) and dual FLT3/AURKA selective (BPR1K871; IC50 = 19/22 nM) agents. BPR1K871 showed potent anti-proliferative activities in MOLM-13 and MV4-11 AML cells (EC50 ∼ 5 nM). Moreover, kinase profiling and cell-line profiling revealed BPR1K871 to be a potential multi-kinase inhibitor. Functional studies using western blot and DNA content analysis in MV4-11 and HCT-116 cell lines revealed FLT3 and AURKA/B target modulation inside the cells. In vivo efficacy in AML xenograft models (MOLM-13 and MV4-11), as well as in solid tumor models (COLO205 and Mia-PaCa2), led to the selection of BPR1K871 as a preclinical development candidate for anti-cancer therapy. Further detailed studies could help to investigate the full potential of BPR1K871 as a multi-kinase inhibitor.
Collapse
Affiliation(s)
- Yung Chang Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Mohane Selvaraj Coumar
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Kalapet, Puducherry, India
| | - Wen-Chieh Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Hui-Yi Shiao
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Yi-Yu Ke
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Wen-Hsing Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chun-Wei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Fu-Ming Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Pei-Yi Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Sing-Yi Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - An-Siou Li
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chun-Hwa Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Po-Chu Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Ching-Ping Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Ming-Hsine Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chen-Lung Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Kuei-Jung Yen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Yun-I Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - John T-A Hsu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Jen-Shin Song
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Chuan Shih
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan, ROC.,Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan, ROC
| |
Collapse
|
32
|
Wang Y, Zhi Y, Jin Q, Lu S, Lin G, Yuan H, Yang T, Wang Z, Yao C, Ling J, Guo H, Li T, Jin J, Li B, Zhang L, Chen Y, Lu T. Discovery of 4-((7H-Pyrrolo[2,3-d]pyrimidin-4-yl)amino)-N-(4-((4-methylpiperazin-1-yl)methyl)phenyl)-1H-pyrazole-3-carboxamide (FN-1501), an FLT3- and CDK-Kinase Inhibitor with Potentially High Efficiency against Acute Myelocytic Leukemia. J Med Chem 2018; 61:1499-1518. [DOI: 10.1021/acs.jmedchem.7b01261] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Yue Wang
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Yanle Zhi
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Qiaomei Jin
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Shuai Lu
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Guowu Lin
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Haoliang Yuan
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Taotao Yang
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Zhanwei Wang
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Chao Yao
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Jun Ling
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Hao Guo
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Tonghui Li
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Jianlin Jin
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Baoquan Li
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Li Zhang
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Yadong Chen
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Tao Lu
- School
of Sciences and ‡State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| |
Collapse
|
33
|
Karrouchi K, Radi S, Ramli Y, Taoufik J, Mabkhot YN, Al-Aizari FA, Ansar M. Synthesis and Pharmacological Activities of Pyrazole Derivatives: A Review. Molecules 2018; 23:molecules23010134. [PMID: 29329257 PMCID: PMC6017056 DOI: 10.3390/molecules23010134] [Citation(s) in RCA: 489] [Impact Index Per Article: 69.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/03/2018] [Accepted: 01/05/2018] [Indexed: 12/31/2022] Open
Abstract
Pyrazole and its derivatives are considered a pharmacologically important active scaffold that possesses almost all types of pharmacological activities. The presence of this nucleus in pharmacological agents of diverse therapeutic categories such as celecoxib, a potent anti-inflammatory, the antipsychotic CDPPB, the anti-obesity drug rimonabant, difenamizole, an analgesic, betazole, a H2-receptor agonist and the antidepressant agent fezolamide have proved the pharmacological potential of the pyrazole moiety. Owing to this diversity in the biological field, this nucleus has attracted the attention of many researchers to study its skeleton chemically and biologically. This review highlights the different synthesis methods and the pharmacological properties of pyrazole derivatives. Studies on the synthesis and biological activity of pyrazole derivatives developed by many scientists around the globe are reported.
Collapse
Affiliation(s)
- Khalid Karrouchi
- Medicinal Chemistry Laboratory, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco.
- LCAE, Department of Chemistry, Faculty of Sciences, University Mohamed I, 60000 Oujda, Morocco.
- Physicochemical service, Drugs Quality Control Laboratory, Division of Drugs and Pharmacy, Ministry of Health, 10100 Rabat, Morocco.
| | - Smaail Radi
- LCAE, Department of Chemistry, Faculty of Sciences, University Mohamed I, 60000 Oujda, Morocco.
| | - Youssef Ramli
- Medicinal Chemistry Laboratory, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco.
| | - Jamal Taoufik
- Medicinal Chemistry Laboratory, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco.
| | - Yahia N Mabkhot
- Department of Chemistry, Faculty of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - Faiz A Al-Aizari
- Department of Chemistry, Faculty of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - M'hammed Ansar
- Medicinal Chemistry Laboratory, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco.
| |
Collapse
|
34
|
Karrouchi K, Radi S, Ramli Y, Taoufik J, Mabkhot YN, Al-Aizari FA, Ansar M. Synthesis and Pharmacological Activities of Pyrazole Derivatives: A Review. Molecules 2018. [PMID: 29329257 DOI: 10.3390/molecules23010134k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
Pyrazole and its derivatives are considered a pharmacologically important active scaffold that possesses almost all types of pharmacological activities. The presence of this nucleus in pharmacological agents of diverse therapeutic categories such as celecoxib, a potent anti-inflammatory, the antipsychotic CDPPB, the anti-obesity drug rimonabant, difenamizole, an analgesic, betazole, a H2-receptor agonist and the antidepressant agent fezolamide have proved the pharmacological potential of the pyrazole moiety. Owing to this diversity in the biological field, this nucleus has attracted the attention of many researchers to study its skeleton chemically and biologically. This review highlights the different synthesis methods and the pharmacological properties of pyrazole derivatives. Studies on the synthesis and biological activity of pyrazole derivatives developed by many scientists around the globe are reported.
Collapse
Affiliation(s)
- Khalid Karrouchi
- Medicinal Chemistry Laboratory, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco.
- LCAE, Department of Chemistry, Faculty of Sciences, University Mohamed I, 60000 Oujda, Morocco.
- Physicochemical service, Drugs Quality Control Laboratory, Division of Drugs and Pharmacy, Ministry of Health, 10100 Rabat, Morocco.
| | - Smaail Radi
- LCAE, Department of Chemistry, Faculty of Sciences, University Mohamed I, 60000 Oujda, Morocco.
| | - Youssef Ramli
- Medicinal Chemistry Laboratory, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco.
| | - Jamal Taoufik
- Medicinal Chemistry Laboratory, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco.
| | - Yahia N Mabkhot
- Department of Chemistry, Faculty of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - Faiz A Al-Aizari
- Department of Chemistry, Faculty of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia.
| | - M'hammed Ansar
- Medicinal Chemistry Laboratory, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco.
| |
Collapse
|
35
|
Nisha, Bhargava G, Kumar Y. Copper(I)-Catalyzed Regioselective C-H Amination of N
-Pyridyl Imines Using Azidotrimethylsilane and TBHP: A One-Pot, Domino Approach to Substituted Imidazo[4, 5-b]pyridines. ChemistrySelect 2017. [DOI: 10.1002/slct.201700969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Nisha
- Department of Chemistry; Faculty of Physical Sciences; SGT University; Gurugram Haryana-122505 India
| | - Gaurav Bhargava
- Department of Chemical Sciences; IKG Punjab Technical University; Kapurthala Punjab-144603 India
| | - Yogesh Kumar
- Department of Chemical Sciences; IKG Punjab Technical University; Kapurthala Punjab-144603 India
- Sphaerapharma Pvt. Ltd.; Manesar, Haryana India-122051 India
| |
Collapse
|
36
|
Vormoor B, Veal GJ, Griffin MJ, Boddy AV, Irving J, Minto L, Case M, Banerji U, Swales KE, Tall JR, Moore AS, Toguchi M, Acton G, Dyer K, Schwab C, Harrison CJ, Grainger JD, Lancaster D, Kearns P, Hargrave D, Vormoor J. A phase I/II trial of AT9283, a selective inhibitor of aurora kinase in children with relapsed or refractory acute leukemia: challenges to run early phase clinical trials for children with leukemia. Pediatr Blood Cancer 2017; 64. [PMID: 27905678 DOI: 10.1002/pbc.26351] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 10/07/2016] [Indexed: 01/21/2023]
Abstract
Aurora kinases regulate mitosis and are commonly overexpressed in leukemia. This phase I/IIa study of AT9283, a multikinase inhibitor, was designed to identify maximal tolerated doses, safety, pharmacokinetics, and pharmacodynamic activity in children with relapsed/refractory acute leukemia. The trial suffered from poor recruitment and terminated early, therefore failing to identify its primary endpoints. AT9283 caused tolerable toxicity, but failed to show clinical responses. Future trials should be based on robust preclinical data that provide an indication of which patients may benefit from the experimental agent, and recruitment should be improved through international collaborations and early combination with established treatment strategies.
Collapse
Affiliation(s)
- B Vormoor
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
- Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - G J Veal
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - M J Griffin
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - A V Boddy
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - J Irving
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - L Minto
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - M Case
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - U Banerji
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, U.K
- The Royal Marsden Hospital, Sutton, UK
| | - K E Swales
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, U.K
| | - J R Tall
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, U.K
| | - A S Moore
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, U.K
- The Royal Marsden Hospital, Sutton, UK
| | - M Toguchi
- Astex Pharmaceuticals, Cambridge Science Park, Cambridge, UK
| | - G Acton
- Cancer Research UK Centre for Drug Development, London, UK
| | - K Dyer
- Cancer Research UK Centre for Drug Development, London, UK
| | - C Schwab
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - C J Harrison
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - J D Grainger
- Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Faculty of Medical & Human Sciences, University of Manchester, Manchester, UK
| | | | - P Kearns
- Institute of Cancer and Genomic Medicine, University of Birmingham, Birmingham, UK
| | - D Hargrave
- Great Ormond Street Hospital for Children, London, UK
| | - J Vormoor
- Newcastle Cancer Centre at the Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
- Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
37
|
Lomov DA, Lyashchuk SN, Abramyants MG. Design and synthesis of imidazo[4,5-c]pyridine derivatives as promising Aurora kinase A (AURKA) inhibitors. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2017. [DOI: 10.1134/s1070428016120198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
A Simple Precursor for Highly Functionalized Fused Imidazo[4,5-b]pyridines and Imidazo[4,5-b]-1,8-naphthyridine. Molecules 2016; 21:molecules21121646. [PMID: 27916964 PMCID: PMC6273580 DOI: 10.3390/molecules21121646] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 12/02/2022] Open
Abstract
1-alkyl aryl-5-amino-4-(cyanoformimidoyl)imidazoles 4 were reacted with malononitrile and 2-amino-1,1,3-propenetricarbonitrile under mild experimental conditions, which led to 5-amino-3-(substituted benzyl)-6,7-dicyano-3H-imidazo[4,5-b]pyridines 5 and 6,8-diamino-3-(4-substituted benzyl)-3H-imidazo[4,5-b]-1,8-naphthyridine-7,9-dicarbonitrile 6, respectively, when the reaction was carried out in the absence of a base, or to 5,7-diamino-3-(4-alkyl aryl)-3H-imidazo[4,5-b]pyridine-6-carbonitrile 8, and 6,8,9-triamino-3-(4-substitutedbenzyl)-3H-imidazo[4,5-b]-1,8-naphthyridine-7-carbonitrile 10 in the presence of 1,8-diazabicyclo(5.4.0)undec-7-ene (DBU). Both reactions evolved from an adduct formed by nucleophilic attack of the malononitrile anion or 2-amino-1,1,3-propenetricarbonitrile anion to the carbon of the cyanoformimidoyl substituent. In the case of the malononitrile anion, a 5-amino-1-alkyl aryl-4-(1-amino-2,2-dicyanovinyl)imidazole 7 was isolated when this reaction was carried out in the presence of DBU. The structure of compound 7 was confirmed by spectroscopic methods, and cyclized intramolecularly to 8 by heating in ethanol/triethyl amine.
Collapse
|
39
|
The Intersection of Structural and Chemical Biology - An Essential Synergy. Cell Chem Biol 2016; 23:173-182. [PMID: 26933743 DOI: 10.1016/j.chembiol.2015.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 12/22/2022]
Abstract
The continual improvement in our ability to generate high resolution structural models of biological molecules has stimulated and supported innovative chemical biology projects that target increasingly challenging ligand interaction sites. In this review we outline some of the recent developments in chemical biology and rational ligand design and show selected examples that illustrate the synergy between these research areas.
Collapse
|
40
|
Algamal ZY, Lee MH, Al-Fakih AM, Aziz M. High-dimensional QSAR modelling using penalized linear regression model with L1/2-norm. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2016; 27:703-719. [PMID: 27628959 DOI: 10.1080/1062936x.2016.1228696] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 08/22/2016] [Indexed: 06/06/2023]
Abstract
In high-dimensional quantitative structure-activity relationship (QSAR) modelling, penalization methods have been a popular choice to simultaneously address molecular descriptor selection and QSAR model estimation. In this study, a penalized linear regression model with L1/2-norm is proposed. Furthermore, the local linear approximation algorithm is utilized to avoid the non-convexity of the proposed method. The potential applicability of the proposed method is tested on several benchmark data sets. Compared with other commonly used penalized methods, the proposed method can not only obtain the best predictive ability, but also provide an easily interpretable QSAR model. In addition, it is noteworthy that the results obtained in terms of applicability domain and Y-randomization test provide an efficient and a robust QSAR model. It is evident from the results that the proposed method may possibly be a promising penalized method in the field of computational chemistry research, especially when the number of molecular descriptors exceeds the number of compounds.
Collapse
Affiliation(s)
- Z Y Algamal
- a Department of Mathematical Sciences , Universiti Teknologi Malaysia , Johor , Malaysia
| | - M H Lee
- a Department of Mathematical Sciences , Universiti Teknologi Malaysia , Johor , Malaysia
| | - A M Al-Fakih
- b Department of Chemistry , Universiti Teknologi Malaysia , Johor , Malaysia
| | - M Aziz
- b Department of Chemistry , Universiti Teknologi Malaysia , Johor , Malaysia
| |
Collapse
|
41
|
The therapeutic voyage of pyrazole and its analogs: A review. Eur J Med Chem 2016; 120:170-201. [DOI: 10.1016/j.ejmech.2016.04.077] [Citation(s) in RCA: 262] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 04/25/2016] [Accepted: 04/28/2016] [Indexed: 02/05/2023]
|
42
|
Gurden MD, Anderhub SJ, Faisal A, Linardopoulos S. Aurora B prevents premature removal of spindle assembly checkpoint proteins from the kinetochore: A key role for Aurora B in mitosis. Oncotarget 2016; 9:19525-19542. [PMID: 29731963 PMCID: PMC5929406 DOI: 10.18632/oncotarget.10657] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 06/01/2016] [Indexed: 12/21/2022] Open
Abstract
Accurate chromosome segregation is dependent on the spindle assembly checkpoint (SAC). In current models, the key direct role of Aurora B in the SAC has been suggested to be to promote rapid kinetochore localisation of MPS1, allowing MPS1 to generate the checkpoint signal. However, Aurora B is also thought to play an indirect role in the SAC through the destabilisation of kinetochore-microtubule (KT-MT) attachments. Here, we demonstrate that Aurora B activity is not required for the kinetochore recruitment of the majority of SAC proteins. More importantly, we show that the primary role of Aurora B in the SAC is to prevent the premature removal of SAC proteins from the kinetochore, which is strictly dependent on KT-MT interactions. Moreover, in the presence of KT-MT interactions, Aurora B inhibition silences a persistent SAC induced by tethering MPS1 to the kinetochore. This explains the highly synergistic interaction between Aurora B and MPS1 inhibitors to override the SAC, which is lost when cells are pre-arrested in nocodazole. Furthermore, we show that Aurora B and MPS1 inhibitors synergistically kill a panel of breast and colon cancer cell lines, including cells that are otherwise insensitive to Aurora B inhibitors alone. These data demonstrate that the major role of Aurora B in SAC is to prevent the removal of SAC proteins from tensionless kinetochores, thus inhibiting premature SAC silencing, and highlights a therapeutic strategy through combination of Aurora B and MPS1 inhibitors.
Collapse
Affiliation(s)
- Mark D Gurden
- Breast Cancer Now, Division of Breast Cancer Research, The Institute of Cancer Research, London, United Kingdom
| | - Simon J Anderhub
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom.,Present address: Phenex Pharmaceuticals, Ludwigshafen am Rhein, Germany
| | - Amir Faisal
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom.,Present address: Lahore University of Management Sciences, D.H.A. Lahore Cantt, Lahore, Pakistan
| | - Spiros Linardopoulos
- Breast Cancer Now, Division of Breast Cancer Research, The Institute of Cancer Research, London, United Kingdom.,Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
43
|
Bavetsias V, Linardopoulos S. Aurora Kinase Inhibitors: Current Status and Outlook. Front Oncol 2015; 5:278. [PMID: 26734566 PMCID: PMC4685048 DOI: 10.3389/fonc.2015.00278] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/27/2015] [Indexed: 11/24/2022] Open
Abstract
The Aurora kinase family comprises of cell cycle-regulated serine/threonine kinases important for mitosis. Their activity and protein expression are cell cycle regulated, peaking during mitosis to orchestrate important mitotic processes including centrosome maturation, chromosome alignment, chromosome segregation, and cytokinesis. In humans, the Aurora kinase family consists of three members; Aurora-A, Aurora-B, and Aurora-C, which each share a conserved C-terminal catalytic domain but differ in their sub-cellular localization, substrate specificity, and function during mitosis. In addition, Aurora-A and Aurora-B have been found to be overexpressed in a wide variety of human tumors. These observations led to a number of programs among academic and pharmaceutical organizations to discovering small molecule Aurora kinase inhibitors as anti-cancer drugs. This review will summarize the known Aurora kinase inhibitors currently in the clinic, and discuss the current and future directions.
Collapse
Affiliation(s)
- Vassilios Bavetsias
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research , London , UK
| | - Spiros Linardopoulos
- Cancer Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK; Breast Cancer Now, Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| |
Collapse
|
44
|
Bavetsias V, Pérez-Fuertes Y, McIntyre PJ, Atrash B, Kosmopoulou M, O'Fee L, Burke R, Sun C, Faisal A, Bush K, Avery S, Henley A, Raynaud FI, Linardopoulos S, Bayliss R, Blagg J. 7-(Pyrazol-4-yl)-3H-imidazo[4,5-b]pyridine-based derivatives for kinase inhibition: Co-crystallisation studies with Aurora-A reveal distinct differences in the orientation of the pyrazole N1-substituent. Bioorg Med Chem Lett 2015; 25:4203-9. [PMID: 26296477 PMCID: PMC4577729 DOI: 10.1016/j.bmcl.2015.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 12/12/2022]
Abstract
Introduction of a 1-benzyl-1H-pyrazol-4-yl moiety at C7 of the imidazo[4,5-b]pyridine scaffold provided 7a which inhibited a range of kinases including Aurora-A. Modification of the benzyl group in 7a, and subsequent co-crystallisation of the resulting analogues with Aurora-A indicated distinct differences in binding mode dependent upon the pyrazole N-substituent. Compounds 7a and 14d interact with the P-loop whereas 14a and 14b engage with Thr217 in the post-hinge region. These crystallographic insights provide options for the design of compounds interacting with the DFG motif or with Thr217.
Collapse
Affiliation(s)
- Vassilios Bavetsias
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom.
| | - Yolanda Pérez-Fuertes
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Patrick J McIntyre
- University of Leicester, Department of Biochemistry, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | - Butrus Atrash
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Magda Kosmopoulou
- Division of Structural Biology, The Institute of Cancer Research, London, United Kingdom
| | - Lisa O'Fee
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Rosemary Burke
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Chongbo Sun
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Amir Faisal
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Katherine Bush
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Sian Avery
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Alan Henley
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Florence I Raynaud
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom
| | - Spiros Linardopoulos
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom; Breakthrough Breast Cancer Research Centre at The Institute of Cancer Research, London, United Kingdom
| | - Richard Bayliss
- University of Leicester, Department of Biochemistry, Lancaster Road, Leicester LE1 9HN, United Kingdom; Division of Structural Biology, The Institute of Cancer Research, London, United Kingdom.
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
45
|
Synthesis and potent cytotoxicity of some novel imidazopyridine derivatives against MCF-7 human breast adenocarcinoma cell line. Chem Heterocycl Compd (N Y) 2015. [DOI: 10.1007/s10593-015-1765-7] [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]
|
46
|
Rosenberg AJ, Ahmed I, Wilson RJ, Williams TM, Kaminsky L, Clark DA. An Improved Synthesis of Imidazo[4,5-b]pyridines and Imidazo[4,5-b]pyrazines by Palladium Catalyzed Amidation using Xantphos in a 1,4-Dioxane:tert-Amyl Alcohol Solvent System. Adv Synth Catal 2014. [DOI: 10.1002/adsc.201400465] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
47
|
Novel acylureidoindolin-2-one derivatives as dual Aurora B/FLT3 inhibitors for the treatment of acute myeloid leukemia. Eur J Med Chem 2014; 85:268-88. [PMID: 25089810 DOI: 10.1016/j.ejmech.2014.07.108] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/29/2014] [Accepted: 07/29/2014] [Indexed: 12/22/2022]
Abstract
A series of 6-acylureido derivatives containing a 3-(pyrrol-2-ylmethylidene)indolin-2-one scaffold were synthesized as potential dual Aurora B/FLT3 inhibitors by replacing the 6-arylureido moiety in 6-arylureidoindolin-2-one-based multi-kinase inhibitors. (Z)-N-(2-(pyrrolidin-1-yl)ethyl)-5-((6-(3-(2-fluoro-4-methoxybenzoyl)ureido)-2-oxoindolin-3-ylidene)methyl)-2,4-dimethyl-1H-pyrrole-3-carboxamide (54) was identified as a dual Aurora B/FLT3 inhibitor (IC50 = 0.4 nM and 0.5 nM, respectively). Compound 54 also exhibited potent cytotoxicity with single-digit nanomolar IC50 values against the FLT3 mutant-associated human acute myeloid leukemia (AML) cell lines MV4-11 (FLT3-ITD) and MOLM-13 (FLT3-ITD). Compound 54 also specifically induced extrinsic apoptosis by inhibiting the phosphorylation of the Aurora B and FLT3 pathways in MOLM-13 cells. Compound 54 had a moderate pharmacokinetic profile. The mesylate salt of 54 efficiently inhibited tumor growth and reduced the mortality of BALB/c nude mice (subcutaneous xenograft model) that had been implanted with AML MOLM-13 cells. Compound 54 is more potent than sunitinib not only against FLT3-WT AML cells but also active against sunitinib-resistant FLT3-ITD AML cells. This study demonstrates the significance of dual Aurora B/FLT3 inhibitors for the development of potential agents to treat AML.
Collapse
|
48
|
Chang Hsu Y, Ke YY, Shiao HY, Lee CC, Lin WH, Chen CH, Yen KJ, Hsu JTA, Chang C, Hsieh HP. Facile identification of dual FLT3-Aurora A inhibitors: a computer-guided drug design approach. ChemMedChem 2014; 9:953-61. [PMID: 24665000 DOI: 10.1002/cmdc.201300571] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Indexed: 12/22/2022]
Abstract
Computer-guided drug design is a powerful tool for drug discovery. Herein we disclose the use of this approach for the discovery of dual FMS-like receptor tyrosine kinase-3 (FLT3)-Aurora A inhibitors against cancer. An Aurora hit compound was selected as a starting point, from which 288 virtual molecules were screened. Subsequently, some of these were synthesized and evaluated for their capacity to inhibit FLT3 and Aurora kinase A. To further enhance FLT3 inhibition, structure-activity relationship studies of the lead compound were conducted through a simplification strategy and bioisosteric replacement, followed by the use of computer-guided drug design to prioritize molecules bearing a variety of different terminal groups in terms of favorable binding energy. Selected compounds were then synthesized, and their bioactivity was evaluated. Of these, one novel inhibitor was found to exhibit excellent inhibition of FLT3 and Aurora kinase A and exert a dramatic antiproliferative effect on MOLM-13 and MV4-11 cells, with an IC50 value of 7 nM. Accordingly, it is considered a highly promising candidate for further development.
Collapse
Affiliation(s)
- Yung Chang Hsu
- Institute of Biotechnology & Pharmaceutical Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 350, Taiwan (ROC)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kumar BS, Dhakshinamoorthy A, Pitchumani K. K10 montmorillonite clays as environmentally benign catalysts for organic reactions. Catal Sci Technol 2014. [DOI: 10.1039/c4cy00112e] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This perspective summarizes the catalytic activity of K10 montmorillonite as a multifunctional catalyst for organic reactions.
Collapse
Affiliation(s)
| | - Amarajothi Dhakshinamoorthy
- School of Chemistry
- Madurai Kamaraj University
- Madurai 625021, India
- Centre for Green Chemistry Processes
- School of Chemistry
| | - Kasi Pitchumani
- School of Chemistry
- Madurai Kamaraj University
- Madurai 625021, India
- Centre for Green Chemistry Processes
- School of Chemistry
| |
Collapse
|
50
|
Bavetsias V, Faisal A, Crumpler S, Brown N, Kosmopoulou M, Joshi A, Atrash B, Pérez-Fuertes Y, Schmitt JA, Boxall KJ, Burke R, Sun C, Avery S, Bush K, Henley A, Raynaud FI, Workman P, Bayliss R, Linardopoulos S, Blagg J. Aurora isoform selectivity: design and synthesis of imidazo[4,5-b]pyridine derivatives as highly selective inhibitors of Aurora-A kinase in cells. J Med Chem 2013; 56:9122-35. [PMID: 24195668 PMCID: PMC3848336 DOI: 10.1021/jm401115g] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Indexed: 12/12/2022]
Abstract
Aurora-A differs from Aurora-B/C at three positions in the ATP-binding pocket (L215, T217, and R220). Exploiting these differences, crystal structures of ligand-Aurora protein interactions formed the basis of a design principle for imidazo[4,5-b]pyridine-derived Aurora-A-selective inhibitors. Guided by a computational modeling approach, appropriate C7-imidazo[4,5-b]pyridine derivatization led to the discovery of highly selective inhibitors, such as compound 28c, of Aurora-A over Aurora-B. In HCT116 human colon carcinoma cells, 28c and 40f inhibited the Aurora-A L215R and R220K mutants with IC50 values similar to those seen for the Aurora-A wild type. However, the Aurora-A T217E mutant was significantly less sensitive to inhibition by 28c and 40f compared to the Aurora-A wild type, suggesting that the T217 residue plays a critical role in governing the observed isoform selectivity for Aurora-A inhibition. These compounds are useful small-molecule chemical tools to further explore the function of Aurora-A in cells.
Collapse
Affiliation(s)
- Vassilios Bavetsias
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Amir Faisal
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Simon Crumpler
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Nathan Brown
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Magda Kosmopoulou
- Division
of Structural Biology, The Institute of
Cancer Research, Chester Beatty Laboratories, London SW3 6JB, United
Kingdom
| | - Amar Joshi
- Department
of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, United
Kingdom
| | - Butrus Atrash
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Yolanda Pérez-Fuertes
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Jessica A. Schmitt
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Katherine J. Boxall
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Rosemary Burke
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Chongbo Sun
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Sian Avery
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Katherine Bush
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Alan Henley
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Florence I. Raynaud
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Paul Workman
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| | - Richard Bayliss
- Division
of Structural Biology, The Institute of
Cancer Research, Chester Beatty Laboratories, London SW3 6JB, United
Kingdom
- Department
of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, United
Kingdom
| | - Spiros Linardopoulos
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
- The
Breakthrough Breast Cancer Research Centre, Division of Breast Cancer
Research, The Institute of Cancer Research, London SW3 6JB, United Kingdom
| | - Julian Blagg
- Cancer
Research UK Cancer Therapeutics Unit, Division of Cancer Therapeutics, The Institute of Cancer Research, London SM2 5NG, United Kingdom
| |
Collapse
|