1
|
Patel P, Shah D, Bambharoliya T, Patel V, Patel M, Patel D, Bhavsar V, Padhiyar S, Patel B, Mahavar A, Patel R, Patel A. A Review on the Development of Novel Heterocycles as α-Glucosidase Inhibitors for the Treatment of Type-2 Diabetes Mellitus. Med Chem 2024; 20:503-536. [PMID: 38275074 DOI: 10.2174/0115734064264591231031065639] [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: 06/10/2023] [Revised: 07/30/2023] [Accepted: 09/08/2023] [Indexed: 01/27/2024]
Abstract
One of the most effective therapeutic decencies in the treatment of Type 2 Diabetes Mellitus is the inhibition of α-glucosidase enzyme, which is present at the brush border of the intestine and plays an important role in carbohydrate digestion to form mono-, di-, and polysaccharides. Acarbose, Voglibose, Miglitol, and Erniglitate have been well-known α-glucosidase inhibitors in science since 1990. However, the long synthetic route and side effects of these inhibitors forced the researchers to move their focus to innovate simple and small heterocyclic scaffolds that work as excellent α-glucosidase inhibitors. Moreover, they are also effective against the postprandial hyperglycemic condition in Type 2 Diabetes Mellitus. In this aspect, this review summarizes recent progress in the discovery and development of heterocyclic molecules that have been appraised to show outstanding inhibition of α-glucosidase to yield positive effects against diabetes.
Collapse
Affiliation(s)
- Prexa Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Gujarat, India
| | - Drashti Shah
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Gujarat, India
| | | | - Vidhi Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Gujarat, India
| | - Mehul Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Gujarat, India
| | - Dharti Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Gujarat, India
| | | | | | | | - Anjali Mahavar
- Chandaben Mohanbhai Patel Institute of Computer Application, Charotar University of Science and Technology, CHARUSAT-Campus, Changa, Gujarat, India
| | - Riddhisiddhi Patel
- Department of Pharmaceutical Science, Saurashtra University, Rajkot, Gujarat, India
| | - Ashish Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Gujarat, India
| |
Collapse
|
2
|
Rusinov VL, Sapozhnikova IM, Spasov AA, Chupakhin ON. Fused azoloazines with antidiabetic activity. Russ Chem Bull 2022. [DOI: 10.1007/s11172-022-3687-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
|
3
|
Langer P. Synthesis of Purines and Related Molecules by Cyclization Reactions of Heterocyclic Enamines. Synlett 2021. [DOI: 10.1055/s-0040-1719845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
AbstractA great variety of pharmacologically relevant fluorinated purine analogues are available by cyclization reactions of heterocyclic enamines with 1,3-dielectrophiles. The reactions usually proceed with excellent regioselectivities. As electrophiles, 1,3-diketones, enaminones or 3-chloro-2-en-1-ones were used. Other synthetic strategies are based on inverse-electron-demand Diels–Alder reactions of heterocyclic enamines with triazines. Purine analogues were further functionalized by transition-metal-catalyzed CH-coupling reactions or oxidative cyclizations, giving rise to more complex polycyclic products. Amidino-C-glycosides in their reactions with 1,3-dielectrophiles afforded pyrimidine-C-glycosides. Multicomponent reactions of heterocyclic enamines afforded complex products, including spirocyclic derivatives.1 Introduction2 1,3-Diketones3 Enaminones4 3-Chloro-2-en-1-ones5 Triazines6 Transition-Metal-Catalyzed Functionalizations7 Pyrimidine-C-Nucleosides8 Multicomponent Reactions9 Conclusions
Collapse
Affiliation(s)
- Peter Langer
- Institut für Chemie, Universität Rostock
- Leibniz-Institut für Katalyse e. V. an der Universität Rostock
| |
Collapse
|
4
|
Peytam F, Takalloobanafshi G, Saadattalab T, Norouzbahari M, Emamgholipour Z, Moghimi S, Firoozpour L, Bijanzadeh HR, Faramarzi MA, Mojtabavi S, Rashidi-Ranjbar P, Karima S, Pakraad R, Foroumadi A. Design, synthesis, molecular docking, and in vitro α-glucosidase inhibitory activities of novel 3-amino-2,4-diarylbenzo[4,5]imidazo[1,2-a]pyrimidines against yeast and rat α-glucosidase. Sci Rep 2021; 11:11911. [PMID: 34099819 PMCID: PMC8184976 DOI: 10.1038/s41598-021-91473-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 05/20/2021] [Indexed: 11/23/2022] Open
Abstract
In an attempt to find novel, potent α-glucosidase inhibitors, a library of poly-substituted 3-amino-2,4-diarylbenzo[4,5]imidazo[1,2-a]pyrimidines 3a-ag have been synthesized through heating a mixture of 2-aminobenzimidazoles 1 and α-azidochalcone 2 under the mild conditions. This efficient, facile protocol has been resulted into the desirable compounds with a wide substrate scope in good to excellent yields. Afterwards, their inhibitory activities against yeast α-glucosidase enzyme were investigated. Showing IC50 values ranging from 16.4 ± 0.36 µM to 297.0 ± 1.2 µM confirmed their excellent potency to inhibit α-glucosidase which encouraged us to perform further studies on α-glucosidase enzymes obtained from rat as a mammal source. Among various synthesized 3-amino-2,4-diarylbenzo[4,5]imidazo[1,2-a]pyrimidines, compound 3k exhibited the highest potency against both Saccharomyces cerevisiae α-glucosidase (IC50 = 16.4 ± 0.36 μM) and rat small intestine α-glucosidase (IC50 = 45.0 ± 8.2 μM). Moreover, the role of amine moiety on the observed activity was studied through substituting with chlorine and hydrogen resulted into a considerable deterioration on the inhibitory activity. Kinetic study and molecular docking study have confirmed the in-vitro results.
Collapse
Affiliation(s)
- Fariba Peytam
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | | | - Toktam Saadattalab
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Norouzbahari
- Faculty of Medicine, Eastern Mediterranean University, via Mersin 10, Famagusta, Northern Cyprus, Turkey
| | - Zahra Emamgholipour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Setareh Moghimi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Loghman Firoozpour
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Bijanzadeh
- Department of Environmental Sciences, Faculty of Natural Resources and Marine Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Mojtabavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Roya Pakraad
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Alireza Foroumadi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Rational Design, Synthesis, In Vitro, and In Silico Studies of Dihydropyrimidinone Derivatives as β-Glucuronidase Inhibitors. J CHEM-NY 2021. [DOI: 10.1155/2021/6664756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In the current study, a series of dihydropyrimidinone derivatives were rationally designed as β-glucuronidase inhibitors. These designed compounds were successfully synthesized and characterized through various spectroscopic techniques such as IR, 1H-NMR, 13C-NMR, and EI-MS. A structure-activity relationship (SAR) of synthesized derivatives to inhibit β-glucuronidase was also established. In vitro biological evaluations revealed that 4i as the most potent compound in this series has an IC50 value of 31.52 ± 2.54 μM compared to the standard D-saccharic acid 1,4-lactone (IC50 = 41.32 ± 1.82 µM). Also, molecular docking and dynamics studies of the most potent compound are performed to evaluate interactions between the active compound and binding site.
Collapse
|
6
|
Sari S, Barut B, Özel A, Saraç S. Discovery of potent α-glucosidase inhibitors through structure-based virtual screening of an in-house azole collection. Chem Biol Drug Des 2020; 97:701-710. [PMID: 33107197 DOI: 10.1111/cbdd.13805] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/01/2020] [Accepted: 10/17/2020] [Indexed: 12/21/2022]
Abstract
Diabetes mellitus, a chronic disorder characterized by hyperglycemia, is considered a pandemic of modern times. α-Glucosidase inhibitors emerged as a promising class of antidiabetic drugs with better tolerability compared with its alternatives. Azoles, although widely preferred in drug design, have scarcely been investigated for their potential against α-glucosidase. In this study, we evaluated α-glucosidase inhibitory effects 20 azole derivatives selected out of an in-house collection via structure-based virtual screening (VS) with consensus scoring approach. Seven compounds were identified with better IC50 values than acarbose (IC50 = 68.18 ± 1.01 µM), a well-known α-glucosidase inhibitor drug, which meant 35% success for our VS methodology. Compound 52, 54, 56, 59, and 81 proved highly potent with IC50 values in the range of 40-60 µM. According to the enzyme kinetics study, four of them were competitive, 56 was non-competitive inhibitor. Structure-activity relationships, quantum mechanical, and docking analyses showed that azole rings at ionized state may be key to the potency observed for the active compounds and modifications to shift the balance between the neutral and ionized states further to the latter could yield more potent derivatives.
Collapse
Affiliation(s)
- Suat Sari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Burak Barut
- Department of Biochemistry, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkey
| | - Arzu Özel
- Department of Biochemistry, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkey.,Drug and Pharmaceutical Technology Application and Research Center, Karadeniz Technical University, Trabzon, Turkey
| | - Selma Saraç
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
7
|
Awolade P, Cele N, Kerru N, Gummidi L, Oluwakemi E, Singh P. Therapeutic significance of β-glucuronidase activity and its inhibitors: A review. Eur J Med Chem 2020; 187:111921. [PMID: 31835168 PMCID: PMC7111419 DOI: 10.1016/j.ejmech.2019.111921] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 01/02/2023]
Abstract
The emergence of disease and dearth of effective pharmacological agents on most therapeutic fronts, constitutes a major threat to global public health and man's existence. Consequently, this has created an exigency in the search for new drugs with improved clinical utility or means of potentiating available ones. To this end, accumulating empirical evidence supports molecular target therapy as a plausible egress and, β-glucuronidase (βGLU) - a lysosomal acid hydrolase responsible for the catalytic deconjugation of β-d-glucuronides has emerged as a viable molecular target for several therapeutic applications. The enzyme's activity level in body fluids is also deemed a potential biomarker for the diagnosis of some pathological conditions. Moreover, due to its role in colon carcinogenesis and certain drug-induced dose-limiting toxicities, the development of potent inhibitors of βGLU in human intestinal microbiota has aroused increased attention over the years. Nevertheless, although our literature survey revealed both natural products and synthetic scaffolds as potential inhibitors of the enzyme, only few of these have found clinical utility, albeit with moderate to poor pharmacokinetic profile. Hence, in this review we present a compendium of exploits in the present millennium directed towards the inhibition of βGLU. The aim is to proffer a platform on which new scaffolds can be modelled for improved βGLU inhibitory potency and the development of new therapeutic agents in consequential.
Collapse
Affiliation(s)
- Paul Awolade
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Nosipho Cele
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Nagaraju Kerru
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Lalitha Gummidi
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Ebenezer Oluwakemi
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa
| | - Parvesh Singh
- School of Chemistry and Physics, University of KwaZulu-Natal, P/Bag X54001, Westville, Durban, South Africa.
| |
Collapse
|
8
|
Discovery of novel pyrido-pyrrolidine hybrid compounds as alpha-glucosidase inhibitors and alternative agent for control of type 1 diabetes. Eur J Med Chem 2020; 188:112034. [DOI: 10.1016/j.ejmech.2020.112034] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/17/2019] [Accepted: 01/02/2020] [Indexed: 12/13/2022]
|
9
|
Dhameja M, Gupta P. Synthetic heterocyclic candidates as promising α-glucosidase inhibitors: An overview. Eur J Med Chem 2019; 176:343-377. [DOI: 10.1016/j.ejmech.2019.04.025] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/19/2019] [Accepted: 04/10/2019] [Indexed: 01/18/2023]
|
10
|
Yan M, Zhao J, Sun D, Sun W, Zhang B, Deng W, Zhang D, Wang L. Synthesis and effect on SMMC-7721 cells of new benzo[ c , d ]indole rhodanine complex merocyanines as PDT photosensitizers. Tetrahedron 2017. [DOI: 10.1016/j.tet.2017.04.060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
11
|
Liu Z, Ma S. Recent Advances in Synthetic α-Glucosidase Inhibitors. ChemMedChem 2017; 12:819-829. [PMID: 28498640 DOI: 10.1002/cmdc.201700216] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/08/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Zhiyang Liu
- Department of Medicinal Chemistry; Key Laboratory of Chemical Biology (Ministry of Education); School of Pharmaceutical Sciences; Shandong University; 44 West Culture Road Jinan 250012 P.R. China
| | - Shutao Ma
- Department of Medicinal Chemistry; Key Laboratory of Chemical Biology (Ministry of Education); School of Pharmaceutical Sciences; Shandong University; 44 West Culture Road Jinan 250012 P.R. China
| |
Collapse
|