1
|
Kumar D, Salahuddin, Mazumder A, Kumar R, Ahsan MJ, Yar MS, Abbussalam, Tyagi PK, Chaitanya MVNL. Pharmacological Evaluation of Bioisosterically Replaced and Triazole- Tethered Derivatives for Anticancer Therapy. Med Chem 2025; 21:264-293. [PMID: 40351067 DOI: 10.2174/0115734064320533240903062533] [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: 04/22/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 05/14/2025]
Abstract
Cancer has been the cause of the highest number of deaths in the human population despite the development and advancement in treatment therapies. The toxicity, drug resistance, and side effects of the current medicaments and therapies have left the void for more research and development. One of the possibilities to fill this void is by incorporating Triazole moieties within existing anticancer pharmacophores to develop new hybrid drugs with less toxicity and more potency. The placement of nitrogen in the triazole ring has endowed its characterization of being integrated with anticancer pharmacophores via bioisosteric replacement, click chemistry and organocatalyzed approaches. This review paper emphasizes the discussions from articles published from the early 2000s to the current 2020s about the triazole-based derivatives used in anticancer therapy, elaborating more on their chemical structures, target receptors or enzymes, mechanism of action, structure-activity relationships, different triazole-derived hybrid drugs under clinical and nonclinical trials, and recent advancements toward developing more potent and less toxic anticancer agents.
Collapse
Affiliation(s)
- Dipesh Kumar
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Plot no. 19, Knowledge Park-2, Greater Noida, 201306, Uttar Pradesh, India
| | - Salahuddin
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Plot no. 19, Knowledge Park-2, Greater Noida, 201306, Uttar Pradesh, India
| | - Avijit Mazumder
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Plot no. 19, Knowledge Park-2, Greater Noida, 201306, Uttar Pradesh, India
| | - Rajnish Kumar
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Plot no. 19, Knowledge Park-2, Greater Noida, 201306, Uttar Pradesh, India
| | - Mohamed Jawed Ahsan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jahangirabad Institute of Technology, Jahangirabad Fort, Jahangirabad, Barabanki Uttar Pradesh, 225203, India
| | - Mohammad Shahar Yar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard University, Hamdard Nagar, New Delhi, 110062, India
| | - Abbussalam
- Department of Physiology, Era's Lucknow Medical College and Hospital, Lucknow, 226003, India
| | - Pankaj Kumar Tyagi
- Department of Biotechnology, Noida Institute of Engineering and Technology, Plot No.19, Knowledge Park-2, Greater Noida, 201306, Uttar Pradesh, India
| | - M V N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| |
Collapse
|
2
|
Guan Q, Xing S, Wang L, Zhu J, Guo C, Xu C, Zhao Q, Wu Y, Chen Y, Sun H. Triazoles in Medicinal Chemistry: Physicochemical Properties, Bioisosterism, and Application. J Med Chem 2024; 67:7788-7824. [PMID: 38699796 DOI: 10.1021/acs.jmedchem.4c00652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Triazole demonstrates distinctive physicochemical properties, characterized by weak basicity, various dipole moments, and significant dual hydrogen bond acceptor and donor capabilities. These features are poised to play a pivotal role in drug-target interactions. The inherent polarity of triazole contributes to its lower logP, suggesting the potential improvement in water solubility. The metabolic stability of triazole adds additional value to drug discovery. Moreover, the metal-binding capacity of the nitrogen atom lone pair electrons of triazole has broad applications in the development of metal chelators and antifungal agents. This Perspective aims to underscore the unique physicochemical attributes of triazole and its application. A comparative analysis involving triazole isomers and other heterocycles provides guiding insights for the subsequent design of triazoles, with the hope of offering valuable considerations for designing other heterocycles in medicinal chemistry.
Collapse
Affiliation(s)
- Qianwen Guan
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Lei Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Jiawei Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Can Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Chunlei Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Qun Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Yulan Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
3
|
Osman AMA, Arabi AA. Average Electron Density: A Quantitative Tool for Evaluating Non-Classical Bioisosteres of Amides. ACS OMEGA 2024; 9:13172-13182. [PMID: 38524460 PMCID: PMC10955596 DOI: 10.1021/acsomega.3c09732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/12/2024] [Accepted: 02/07/2024] [Indexed: 03/26/2024]
Abstract
Bioisosterism is strategically used in drug design to enhance the pharmacokinetic and pharmacodynamic properties of therapeutic molecules. The average electron density (AED) tool has been used in several studies to quantify similarities among nonclassical bioisosteres of carboxylic acid. In this study, the AED tool is used to quantify the similarities among nonclassical bioisosteres of an amide group. In particular, amide-to-1,2,3-triazole bioisosterism is considered. To evaluate the AED differences exhibited by isomers of nonclassical bioisosteres, both isomers of amide (cis and trans) and 1,2,3-triazole (1,4 and 1,5 disubstituted moieity) were considered. The amide and 1,2,3-triazole bioisosteric moieties were capped with various R groups (R= methyl, hydrogen, and chloro) to account for changes in their environment. Amide-to-triazole bioisosteric substitutions were then explored in a more realistic environment, that is, within the FDA-approved anticancer imatinib drug. The AED tool effectively identified similarities between substantially different moieties, 1,2,3-triazole and amide, showing AED differences of no more than 4%. The AED tool was also proven to be useful in evaluating the contribution of various factors affecting triazole-amide bioisosterism including isomerism and changes in their environment. The AED values of each bioisostere were transferable within a maximum difference of 2.6%, irrespective of the change in environment. The 1,4- and 1,5-disubstituted isomers of 1,2,3-triazole have AED values that differ by less than unity, 0.52%. Similarly, the AED values of the cis- and trans-amide isomers differ by only 1.31%. Overall, the AED quantitative tool not only replicated experimental observations regarding similarities in bioisosteres, but also explained and quantified each contributing factor. This demonstrates the extended utility of the AED tool from nonclassical carboxylic acid bioisosteres to amide equivalents.On the contrary, electrostatic potential maps, usually used in the literature to qualitatively evaluate bioisosterism, were not similar for the 1,2,3-triazole and amide bioisosteres, under different environments. Overall, the AED tool proves to be powerful in quantitatively evaluating and predicting bioisosterism across diverse moieties considering structural and environmental variations, making it valuable in drug design.
Collapse
Affiliation(s)
- Alaa MA Osman
- College of Medicine and Health
Sciences, Department of Biochemistry and Molecular Biology, United Arab Emirates University, AlAin P.O. Box: 15551, United Arab Emirates
| | - Alya A. Arabi
- College of Medicine and Health
Sciences, Department of Biochemistry and Molecular Biology, United Arab Emirates University, AlAin P.O. Box: 15551, United Arab Emirates
| |
Collapse
|
4
|
Mohseniabbasabadi T, Behboodyzad F, Abolhasani Zadeh F, Balali E. Vismodegib anticancer drug: Analyzing electronic and structural features and examining biological activities. MAIN GROUP CHEMISTRY 2021. [DOI: 10.3233/mgc-210160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Vismodegib (Vis) is an anticancer drug, in which its electronic and structural features were examined in this work. To this aim, the chlorine atoms of original Vis model were substituted by other fluorine, bromine, and iodine halogen atoms yielding F-Vis, Br-Vis, and I-Vis in addition to the original Cl-Vis model. The models were optimized by performing quantum chemical calculations and their interactions with the smoothened (SMO) target were examined by performing molecular docking simulations. The results indicated that the stabilized structures of halogenated Vis models were achievable and their features indicated the dominant role of halogen atoms for their participation in interactions with other substances. Based on the obtained results, Br-Vis model was seen suitable for participating in interaction with the SMO target even better than the original Vis model. The hypothesis of this work was affirmed by employing the in silico approach for analyzing the features of singular ligands and for evaluating their biological functions.
Collapse
Affiliation(s)
- Tahereh Mohseniabbasabadi
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Farnoosh Behboodyzad
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Ebrahim Balali
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
5
|
Lutter FH, Jouffroy M. Facile Conversion of Molecularly Complex (Hetero)aryl Carboxylic Acids into Alkynes for Accelerated SAR Exploration. Chemistry 2021; 27:14816-14820. [PMID: 34460121 DOI: 10.1002/chem.202102130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Indexed: 11/10/2022]
Abstract
1,2,3-Triazoles are well-established bioisosteres for amides, often installed as a result of structure-activity-relationship (SAR) exploration. A straightforward approach to assess the effect of the replacement of an amide by a triazole would start from the carboxylic acid and the amine used for the formation of a given amide and convert them into the corresponding alkyne and azide for cyclization by copper-catalyzed alkyne-azide cycloaddition (CuAAC). Herein, we report a functional-group-tolerant and operationally simple decarbonylative alkynylation that allows the conversion of complex (hetero)aryl carboxylic acids into alkynes. Furthermore, the utility of this method was demonstrated in the preparation of a triazolo analog of the commercial drug moclobemide. Lastly, mechanistic investigations using labeled carboxylic acid derivatives clearly show the decarbonylative nature of this transformation.
Collapse
Affiliation(s)
- Ferdinand H Lutter
- Chemical Process R&D, Discovery Process Research, Janssen Pharmaceutica N.V., Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Matthieu Jouffroy
- Chemical Process R&D, Discovery Process Research, Janssen Pharmaceutica N.V., Turnhoutseweg 30, 2340, Beerse, Belgium
| |
Collapse
|
6
|
Bonandi E, Mori M, Infante P, Basili I, Di Marcotullio L, Calcaterra A, Catti F, Botta B, Passarella D. Design and Synthesis of New Withaferin A Inspired Hedgehog Pathway Inhibitors. Chemistry 2021; 27:8350-8357. [PMID: 33811701 PMCID: PMC8251939 DOI: 10.1002/chem.202100315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Indexed: 12/28/2022]
Abstract
Withanolides constitute a well-known family of plant-based alkaloids characterised by widespread biological properties, including the ability of interfering with Hedgehog (Hh) signalling pathway. Following our interest in natural products and in anticancer compounds, we report here the synthesis of a new class of Hh signalling pathway inhibitors, inspired by withaferin A, the first isolated member of withanolides. The decoration of our scaffolds was rationally supported by in silico studies, while functional evaluation revealed promising candidates, confirming once again the importance of natural products as inspiration source for the discovery of novel bioactive compounds. A stereoselective approach, based on Brown chemistry, allowed the obtainment and the functional evaluation of the enantiopure hit compounds.
Collapse
Affiliation(s)
- Elisa Bonandi
- Department of Chemistry, Università degli Studi di MilanoVia Golgi 1920133MilanItaly
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy Università degli Studi di SienaVia Aldo Moro 253100SienaItaly
| | - Paola Infante
- Istituto Italiano di TecnologiaViale Regina Elena 29100161RomeItaly
| | - Irene Basili
- Department of Molecular MedicineUniversity La Sapienza, RomaViale Regina Elena 29100161RomaItaly
| | - Lucia Di Marcotullio
- Department of Molecular MedicineUniversity La Sapienza, RomaViale Regina Elena 29100161RomaItaly
- Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci BolognettiDepartment of Molecular MedicineUniversity La SapienzaViale Regina Elena 29100161RomaItaly
| | - Andrea Calcaterra
- Department of Chemistry and Technology of DrugsUniversity La Sapienza, RomaPiazzale Aldo Moro 500185RomeItaly
| | - Federica Catti
- Arkansas State UniversityCampus Querétaro Carretera Estatal 100, km 17.5. C.P.76270 Municipio de ColónQuerétaroMéxico
| | - Bruno Botta
- Department of Chemistry and Technology of DrugsUniversity La Sapienza, RomaPiazzale Aldo Moro 500185RomeItaly
| | - Daniele Passarella
- Department of Chemistry, Università degli Studi di MilanoVia Golgi 1920133MilanItaly
| |
Collapse
|
7
|
Ghirga F, Quaglio D, Mori M, Cammarone S, Iazzetti A, Goggiamani A, Ingallina C, Botta B, Calcaterra A. A unique high-diversity natural product collection as a reservoir of new therapeutic leads. Org Chem Front 2021. [DOI: 10.1039/d0qo01210f] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We review the successful application of computer-aided methods to screen a unique and high-diversity in house collection library composed of around 1000 individual natural products.
Collapse
Affiliation(s)
- Francesca Ghirga
- Center For Life Nano Science@Sapienza
- Istituto Italiano di Tecnologia
- 00161 Rome
- Italy
| | - Deborah Quaglio
- Department of Chemistry and Technology of Drugs
- “Department of Excellence 2018–2022”
- The Sapienza University of Rome
- 00185 Rome
- Italy
| | - Mattia Mori
- Department of Biotechnology
- Chemistry and Pharmacy
- “Department of Excellence 2018–2022”
- University of Siena
- 53100 Siena
| | - Silvia Cammarone
- Department of Chemistry and Technology of Drugs
- “Department of Excellence 2018–2022”
- The Sapienza University of Rome
- 00185 Rome
- Italy
| | - Antonia Iazzetti
- Department of Chemistry and Technology of Drugs
- “Department of Excellence 2018–2022”
- The Sapienza University of Rome
- 00185 Rome
- Italy
| | - Antonella Goggiamani
- Department of Chemistry and Technology of Drugs
- “Department of Excellence 2018–2022”
- The Sapienza University of Rome
- 00185 Rome
- Italy
| | - Cinzia Ingallina
- Department of Chemistry and Technology of Drugs
- “Department of Excellence 2018–2022”
- The Sapienza University of Rome
- 00185 Rome
- Italy
| | - Bruno Botta
- Department of Chemistry and Technology of Drugs
- “Department of Excellence 2018–2022”
- The Sapienza University of Rome
- 00185 Rome
- Italy
| | - Andrea Calcaterra
- Department of Chemistry and Technology of Drugs
- “Department of Excellence 2018–2022”
- The Sapienza University of Rome
- 00185 Rome
- Italy
| |
Collapse
|
8
|
Kumari S, Carmona AV, Tiwari AK, Trippier PC. Amide Bond Bioisosteres: Strategies, Synthesis, and Successes. J Med Chem 2020; 63:12290-12358. [PMID: 32686940 DOI: 10.1021/acs.jmedchem.0c00530] [Citation(s) in RCA: 295] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The amide functional group plays a key role in the composition of biomolecules, including many clinically approved drugs. Bioisosterism is widely employed in the rational modification of lead compounds, being used to increase potency, enhance selectivity, improve pharmacokinetic properties, eliminate toxicity, and acquire novel chemical space to secure intellectual property. The introduction of a bioisostere leads to structural changes in molecular size, shape, electronic distribution, polarity, pKa, dipole or polarizability, which can be either favorable or detrimental to biological activity. This approach has opened up new avenues in drug design and development resulting in more efficient drug candidates introduced onto the market as well as in the clinical pipeline. Herein, we review the strategic decisions in selecting an amide bioisostere (the why), synthetic routes to each (the how), and success stories of each bioisostere (the implementation) to provide a comprehensive overview of this important toolbox for medicinal chemists.
Collapse
Affiliation(s)
- Shikha Kumari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Angelica V Carmona
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, Ohio 43614, United States
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States.,UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
9
|
Sahin ID, Christodoulou MS, Guzelcan EA, Koyas A, Karaca C, Passarella D, Cetin-Atalay R. A small library of chalcones induce liver cancer cell death through Akt phosphorylation inhibition. Sci Rep 2020; 10:11814. [PMID: 32678233 PMCID: PMC7367369 DOI: 10.1038/s41598-020-68775-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 06/26/2020] [Indexed: 01/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) ranks as the fifth most common and the second deadliest cancer worldwide. HCC is extremely resistant to the conventional chemotherapeutics. Hence, it is vital to develop new treatment options. Chalcones were previously shown to have anticancer activities in other cancer types. In this study, 11 chalcones along with quercetin, papaverin, catechin, Sorafenib and 5FU were analyzed for their bioactivities on 6 HCC cell lines and on dental pulp stem cells (DPSC) which differentiates into hepatocytes, and is used as a model for untransformed control cells. 3 of the chalcones (1, 9 and 11) were selected for further investigation due to their high cytotoxicity against liver cancer cells and compared to the other clinically established compounds. Chalcones did not show significant bioactivity (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\hbox {IC}_{50}>20\upmu \hbox {M}$$\end{document}IC50>20μM) on dental pulp stem cells. Cell cycle analysis revealed that these 3 chalcone-molecules induced SubG1/G1 arrest. Akt protein phosphorylation was inhibited by these molecules in PTEN deficient, drug resistant, mesenchymal like Mahlavu cells leading to the activation of p21 and the inhibition of NF\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$ \kappa $$\end{document}κB-p65 transcription factor. Hence the chalcones induced apoptotic cell death pathway through NF\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$ \kappa $$\end{document}κB-p65 inhibition. On the other hand, these molecules triggered p21 dependent activation of Rb protein and thereby inhibition of cell cycle and cell growth in liver cancer cells. Involvement of PI3K/Akt pathway hyperactivation was previously described in survival of liver cancer cells as carcinogenic event. Therefore, our results indicated that these chalcones can be considered as candidates for liver cancer therapeutics particularly when PI3K/Akt pathway involved in tumor development.
Collapse
Affiliation(s)
| | - Michael S Christodoulou
- DISFARM, Sezione di Chimica Generale e Organica "A. Marchesini" Universitádegli Studi di Milano, via Venezian 21, 20133, Milano, Italy
| | - Ece Akhan Guzelcan
- CanSyL, Graduate School of Informatics, Middle East Technical University, 06800, Ankara, Turkey
| | - Altay Koyas
- CanSyL, Graduate School of Informatics, Middle East Technical University, 06800, Ankara, Turkey
| | - Cigdem Karaca
- Faculty of Dentistry, Department of Oral and Maxillofacial Surgery, Hacettepe University, 06230, Ankara, Turkey
| | - Daniele Passarella
- Department of Chemistry, Universitá degli Studi di Milano, Via Golgi 19, 20133, Milano, Italy
| | - Rengul Cetin-Atalay
- CanSyL, Graduate School of Informatics, Middle East Technical University, 06800, Ankara, Turkey
| |
Collapse
|
10
|
Corsini E, Facchetti G, Esposito S, Maddalon A, Rimoldi I, Christodoulou MS. Antiproliferative effects of chalcones on T cell acute lymphoblastic leukemia-derived cells: Role of PKCβ. Arch Pharm (Weinheim) 2020; 353:e2000062. [PMID: 32394529 DOI: 10.1002/ardp.202000062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/18/2020] [Accepted: 04/22/2020] [Indexed: 12/16/2022]
Abstract
In this study, a series of 20 chalcone derivatives was synthesized, and their antiproliferative activity was tested against the human T cell acute lymphoblastic leukemia-derived cell line, CCRF-CEM. On the basis of the structural features of the most active compounds, a new library of chalcone derivatives, according to the structure-activity relationship design, was synthesized, and their antiproliferative activity was tested against the same cancer cell line. Furthermore, four of these derivatives (compounds 3, 4, 8, 28), based on lower IC50 values (between 6.1 and 8.9 μM), were selected for further investigation regarding the modulation of the protein expression of RACK1 (receptor for activated C kinase), protein kinase C (PKC)α and PKCβ, and their action on the cell cycle level. The cell cycle analysis indicated a block in the G0/G1 phase for all four compounds, with a statistically significant decrease in the percentage of cells in the S phase, with no indication of apoptosis (sub-G0/G1 phase). Compounds 4 and 8 showed a statistically significant reduction in the expression of PKCα and an increase in PKCβ, which together with the demonstration of an antiproliferative role of PKCβ, as assessed by treating cells with a selective PKCβ activator, indicated that the observed antiproliferative effect is likely to be mediated through PKCβ induction.
Collapse
Affiliation(s)
- Emanuela Corsini
- Laboratory of Toxicology, Dipartimento di Scienze Politiche ed Ambientali, Università degli Studi di Milano, Milano, Italy
| | - Giorgio Facchetti
- DISFARM, Sezione di Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milano, Italy
| | - Sara Esposito
- Laboratory of Toxicology, Dipartimento di Scienze Politiche ed Ambientali, Università degli Studi di Milano, Milano, Italy
| | - Ambra Maddalon
- Laboratory of Toxicology, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milano, Italy
| | - Isabella Rimoldi
- DISFARM, Sezione di Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milano, Italy
| | - Michael S Christodoulou
- DISFARM, Sezione di Chimica Generale e Organica "A. Marchesini", Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
11
|
Novel 3,3-disubstituted oxindole derivatives. Synthesis and evaluation of the anti-proliferative activity. Bioorg Med Chem Lett 2019; 30:126845. [PMID: 31831381 DOI: 10.1016/j.bmcl.2019.126845] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 12/14/2022]
Abstract
3,3-Disubstituted oxindole derivatives bearing a nitrogen atom at the C-3 position have been synthesized starting from 3-alkyl oxindole through a metal free pathway. These derivatives have been tested in five human tumor cell lines (PC3, MCF7, SW620, MiaPaca2 and A375) and on primary cells (PBMCs) from healthy donors providing compound 6d showing a strong anticancer effect in all cancer lines on the low micromolar range.
Collapse
|
12
|
Xu Z, Zhao SJ, Liu Y. 1,2,3-Triazole-containing hybrids as potential anticancer agents: Current developments, action mechanisms and structure-activity relationships. Eur J Med Chem 2019; 183:111700. [PMID: 31546197 DOI: 10.1016/j.ejmech.2019.111700] [Citation(s) in RCA: 291] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/08/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022]
Abstract
Anticancer agents are critical for the cancer treatment, but side effects and the drug resistance associated with the currently used anticancer agents create an urgent need to explore novel drugs with low side effects and high efficacy. 1,2,3-Triazole is privileged building block in the discovery of new anticancer agents, and some of its derivatives have already been applied in clinics or under clinical trials for fighting against cancers. Hybrid molecules occupy an important position in cancer control, and hybridization of 1,2,3-triazole framework with other anticancer pharmacophores may provide valuable therapeutic intervention for the treatment of cancer, especially drug-resistant cancer. This review emphasizes the recent advances in 1,2,3-triazole-containing hybrids with anticancer potential, covering articles published between 2015 and 2019, and the structure-activity relationships, together with mechanisms of action are also discussed.
Collapse
Affiliation(s)
- Zhi Xu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China.
| | - Shi-Jia Zhao
- Wuhan University of Science and Technology, Wuhan, PR China
| | - Yi Liu
- Wuhan University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
13
|
Doiron JE, Le CA, Ody BK, Brace JB, Post SJ, Thacker NL, Hill HM, Breton GW, Mulder MJ, Chang S, Bridges TM, Tang L, Wang W, Rowe SM, Aller SG, Turlington M. Evaluation of 1,2,3-Triazoles as Amide Bioisosteres In Cystic Fibrosis Transmembrane Conductance Regulator Modulators VX-770 and VX-809. Chemistry 2019; 25:3662-3674. [PMID: 30650214 PMCID: PMC6469399 DOI: 10.1002/chem.201805919] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/14/2019] [Indexed: 12/25/2022]
Abstract
The 1,2,3-triazole has been successfully utilized as an amide bioisostere in multiple therapeutic contexts. Based on this precedent, triazole analogues derived from VX-809 and VX-770, prominent amide-containing modulators of the cystic fibrosis transmembrane conductance regulator (CFTR), were synthesized and evaluated for CFTR modulation. Triazole 11, derived from VX-809, displayed markedly reduced efficacy in F508del-CFTR correction in cellular TECC assays in comparison to VX-809. Surprisingly, triazole analogues derived from potentiator VX-770 displayed no potentiation of F508del, G551D, or WT-CFTR in cellular Ussing chamber assays. However, patch clamp analysis revealed that triazole 60 potentiates WT-CFTR similarly to VX-770. The efficacy of 60 in the cell-free patch clamp experiment suggests that the loss of activity in the cellular assay could be due to the inability of VX-770 triazole derivatives to reach the CFTR binding site. Moreover, in addition to the negative impact on biological activity, triazoles in both structural classes displayed decreased metabolic stability in human microsomes relative to the analogous amides. In contrast to the many studies that demonstrate the advantages of using the 1,2,3-triazole, these findings highlight the negative impacts that can arise from replacement of the amide with the triazole and suggest that caution is warranted when considering use of the 1,2,3-triazole as an amide bioisostere.
Collapse
Affiliation(s)
- Jake E. Doiron
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia 30165 (USA),
| | - Christina A. Le
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama 35205 (USA),
| | - Britton K. Ody
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia 30165 (USA),
| | - Jonathon B. Brace
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia 30165 (USA),
| | - Savannah J. Post
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia 30165 (USA),
| | - Nathan L. Thacker
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia 30165 (USA),
| | - Harrison M. Hill
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia 30165 (USA),
| | - Gary W. Breton
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia 30165 (USA),
| | - Matthew J. Mulder
- Vanderbilt Center for Neuroscience Drug Discovery, Nashville, Tennessee 37232 (USA)
| | - Sichen Chang
- Vanderbilt Center for Neuroscience Drug Discovery, Nashville, Tennessee 37232 (USA)
| | - Thomas M. Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Nashville, Tennessee 37232 (USA)
| | - Liping Tang
- Departments of Medicine and Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama 35205 (USA)
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35205 (USA)
| | - Wei Wang
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35205 (USA)
- Department of Cell, Development, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35205 (USA)
| | - Steven M. Rowe
- Departments of Medicine and Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama 35205 (USA)
- Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35205 (USA)
- Department of Cell, Development, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama 35205 (USA)
| | - Stephen G. Aller
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, Alabama 35205 (USA),
| | - Mark Turlington
- Department of Chemistry and Biochemistry, Berry College, Mount Berry, Georgia 30165 (USA),
| |
Collapse
|
14
|
da Silva VD, de Faria BM, Colombo E, Ascari L, Freitas GPA, Flores LS, Cordeiro Y, Romão L, Buarque CD. Design, synthesis, structural characterization and in vitro evaluation of new 1,4-disubstituted-1,2,3-triazole derivatives against glioblastoma cells. Bioorg Chem 2018; 83:87-97. [PMID: 30343205 DOI: 10.1016/j.bioorg.2018.10.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/25/2018] [Accepted: 10/04/2018] [Indexed: 12/21/2022]
Abstract
A new series of 1,4-disubstituted-1,2,3-triazole derivatives were synthesized through the copper-catalyzed azide-alkyne 1,3-dipolar cycloaddition (Click chemistry) and their inhibitory activities were evaluated against different human glioblastoma (GBM) cell lines, including highly drug-resistant human cell lines GBM02, GBM95. The most effective compounds were 9d, containing the methylenoxy moiety linked to triazole and the tosyl-hydrazone group, and the symmetrical bis-triazole 10a, also containing methylenoxy moiety linked to triazole. Single crystal X-ray diffraction analysis was employed for structural elucidation of compound 9d. In silico analyses of physicochemical, pharmacokinetic, and toxicological properties suggest that compounds 8a, 8b, 8c, 9d, and 10a are potential candidates for central nervous system-acting drugs.
Collapse
Affiliation(s)
- Veronica D da Silva
- Laboratório de Síntese orgânica, Pontifícia Universidade Católica do Rio de Janeiro, 22451-900 Rio de Janeiro, RJ, Brazil
| | - Bruna M de Faria
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Eduardo Colombo
- Laboratório de Síntese orgânica, Pontifícia Universidade Católica do Rio de Janeiro, 22451-900 Rio de Janeiro, RJ, Brazil
| | - Lucas Ascari
- Faculdade de Farmácia, UFRJ, RJ 21941-902, Brazil
| | - Gabriella P A Freitas
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Leonã S Flores
- Laboratório de Difração de raios X, UFJF, MG 36036-900, Brazil
| | | | - Luciana Romão
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Camilla D Buarque
- Laboratório de Síntese orgânica, Pontifícia Universidade Católica do Rio de Janeiro, 22451-900 Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
15
|
Synergistic inhibition of the Hedgehog pathway by newly designed Smo and Gli antagonists bearing the isoflavone scaffold. Eur J Med Chem 2018; 156:554-562. [DOI: 10.1016/j.ejmech.2018.07.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/25/2018] [Accepted: 07/07/2018] [Indexed: 01/19/2023]
|
16
|
Christodoulou MS, Dapiaggi F, Ghiringhelli F, Pieraccini S, Sironi M, Lucafò M, Curci D, Decorti G, Stocco G, Chirumamilla CS, Vanden Berghe W, Balaguer P, Michel BY, Burger A, Beccalli EM, Passarella D, Martinet N. Imidazo[2,1- b]benzothiazol Derivatives as Potential Allosteric Inhibitors of the Glucocorticoid Receptor. ACS Med Chem Lett 2018; 9:339-344. [PMID: 29670697 PMCID: PMC5900336 DOI: 10.1021/acsmedchemlett.7b00527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoid receptor (GCR) transactivation reporter gene assays were used as an initial high-throughput screening on a diversified library of 1200 compounds for their evaluation as GCR antagonists. A class of imidazo[2,1-b]benzothiazole and imidazo[2,1-b]benzoimidazole derivatives were identified for their ability to modulate GCR transactivation and anti-inflammatory transrepression effects utilizing GCR and NF-κB specific reporter gene assays. Modeling studies on the crystallographic structure of the GCR ligand binding domain provided three new analogues bearing the tetrahydroimidazo[2,1-b]benzothiazole scaffold able to antagonize the GCR in the presence of dexamethasone (DEX) and also defined their putative binding into the GCR structure. Both mRNA level measures of GCR itself and its target gene GILZ, on cells treated with the new analogues, showed a GCR transactivation inhibition, thus suggesting a potential allosteric inhibition of the GCR.
Collapse
Affiliation(s)
- Michael S. Christodoulou
- DISFARM,
Sezione di Chimica Generale e Organica “A. Marchesini”, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Federico Dapiaggi
- Department
of Chemistry, Università degli Studi
di Milano, Via Golgi
19, 20133 Milano Italy
| | - Francesca Ghiringhelli
- Department
of Chemistry, Università degli Studi
di Milano, Via Golgi
19, 20133 Milano Italy
| | - Stefano Pieraccini
- Department
of Chemistry, Università degli Studi
di Milano, Via Golgi
19, 20133 Milano Italy
- Istituto
di Scienze e Tecnologie Molecolari (INSTM), CNR, and INSTM, UdR Milano, Via Golgi 19, 20133 Milano, Italy
| | - Maurizio Sironi
- Department
of Chemistry, Università degli Studi
di Milano, Via Golgi
19, 20133 Milano Italy
- Istituto
di Scienze e Tecnologie Molecolari (INSTM), CNR, and INSTM, UdR Milano, Via Golgi 19, 20133 Milano, Italy
| | - Marianna Lucafò
- Department
of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy
| | - Debora Curci
- PhD
School in Reproduction and Developmental Sciences, University of Trieste, 34127 Trieste, Italy
| | - Giuliana Decorti
- Department
of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy
- Institute
for Maternal and Child Health - IRCCS “Burlo Garofolo”, 34127 Trieste, Italy
| | - Gabriele Stocco
- Department
of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Chandra Sekhar Chirumamilla
- Laboratory
of Protein Chemistry, Proteomics and Epigenetic Signalling, Department
of Biomedical Sciences, University of Antwerp
(UA), 2000 Antwerpen, Belgium
| | - Wim Vanden Berghe
- Laboratory
of Protein Chemistry, Proteomics and Epigenetic Signalling, Department
of Biomedical Sciences, University of Antwerp
(UA), 2000 Antwerpen, Belgium
| | - Patrick Balaguer
- IRCM,
INSERM U1194, Université Montpellier, ICM, 208 rue des Apothicaires, F-34298 Montpellier, France
| | - Benoît Y. Michel
- Université Côte d’Azur, CNRS, Institut
de Chimie
de Nice, UMR 7272, Parc Valrose, 06108 Nice Cedex 2, France
| | - Alain Burger
- Université Côte d’Azur, CNRS, Institut
de Chimie
de Nice, UMR 7272, Parc Valrose, 06108 Nice Cedex 2, France
| | - Egle M. Beccalli
- DISFARM,
Sezione di Chimica Generale e Organica “A. Marchesini”, Università degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy
| | - Daniele Passarella
- Department
of Chemistry, Università degli Studi
di Milano, Via Golgi
19, 20133 Milano Italy
| | - Nadine Martinet
- Université Côte d’Azur, CNRS, Institut
de Chimie
de Nice, UMR 7272, Parc Valrose, 06108 Nice Cedex 2, France
| |
Collapse
|
17
|
Bonandi E, Christodoulou MS, Fumagalli G, Perdicchia D, Rastelli G, Passarella D. The 1,2,3-triazole ring as a bioisostere in medicinal chemistry. Drug Discov Today 2017; 22:1572-1581. [PMID: 28676407 DOI: 10.1016/j.drudis.2017.05.014] [Citation(s) in RCA: 441] [Impact Index Per Article: 55.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/16/2017] [Accepted: 05/25/2017] [Indexed: 10/25/2022]
Abstract
1,2,3-Triazole is a well-known scaffold that has a widespread occurrence in different compounds characterized by several bioactivities, such as antimicrobial, antiviral, and antitumor effects. Moreover, the structural features of 1,2,3-triazole enable it to mimic different functional groups, justifying its wide use as a bioisostere for the synthesis of new active molecules. Here, we provide an overview of the 1,2,3-triazole ring as a bioisostere for the design of drug analogs, highlighting relevant recent examples.
Collapse
Affiliation(s)
- Elisa Bonandi
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | - Michael S Christodoulou
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy; Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Gaia Fumagalli
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | - Dario Perdicchia
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | - Giulio Rastelli
- Dipartimento di Scienze della Vita, Università degli Studi di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy
| | - Daniele Passarella
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy.
| |
Collapse
|
18
|
Alfonsi R, Botta B, Cacchi S, Di Marcotullio L, Fabrizi G, Faedda R, Goggiamani A, Iazzetti A, Mori M. Design, Palladium-Catalyzed Synthesis, and Biological Investigation of 2-Substituted 3-Aroylquinolin-4(1H)-ones as Inhibitors of the Hedgehog Signaling Pathway. J Med Chem 2017; 60:1469-1477. [DOI: 10.1021/acs.jmedchem.6b01135] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Romina Alfonsi
- Department of Molecular
Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
| | - Bruno Botta
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Sandro Cacchi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular
Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
- Istituto Pasteur, Fondazione Cenci-Bolognetti, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
| | - Giancarlo Fabrizi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Roberta Faedda
- Department of Molecular
Medicine, Sapienza University, Viale Regina Elena 291, 00161 Rome, Italy
| | - Antonella Goggiamani
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Antonia Iazzetti
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University, Piazzale A. Moro 5, 00185 Rome, Italy
| | - Mattia Mori
- Center for Life Nano
Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy
| |
Collapse
|
19
|
Gabba A, Robakiewicz S, Taciak B, Ulewicz K, Broggini G, Rastelli G, Krol M, Murphy PV, Passarella D. Synthesis and Biological Evaluation of Migrastatin Macrotriazoles. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600988] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Adele Gabba
- School of Chemistry; National University of Ireland; Galway, Ireland
| | - Stefania Robakiewicz
- Department of Physiological Sciences; Warsaw University of Life Sciences; Warsaw Poland
| | - Bartłomiej Taciak
- Department of Physiological Sciences; Warsaw University of Life Sciences; Warsaw Poland
| | - Katarzyna Ulewicz
- Department of Physiological Sciences; Warsaw University of Life Sciences; Warsaw Poland
| | - Gianluigi Broggini
- Dipartimento di Scienza e Alta Tecnologia; Università degli Studi dell'Insubria; Como Italy
| | - Giulio Rastelli
- Department of Life Sciences; University of Modena and Reggio Emilia; Modena Italy
| | - Magadalena Krol
- Department of Physiological Sciences; Warsaw University of Life Sciences; Warsaw Poland
| | - Paul V. Murphy
- School of Chemistry; National University of Ireland; Galway, Ireland
| | | |
Collapse
|
20
|
Infante P, Alfonsi R, Ingallina C, Quaglio D, Ghirga F, D'Acquarica I, Bernardi F, Di Magno L, Canettieri G, Screpanti I, Gulino A, Botta B, Mori M, Di Marcotullio L. Inhibition of Hedgehog-dependent tumors and cancer stem cells by a newly identified naturally occurring chemotype. Cell Death Dis 2016; 7:e2376. [PMID: 27899820 PMCID: PMC5059851 DOI: 10.1038/cddis.2016.195] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/24/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
Hedgehog (Hh) inhibitors have emerged as valid tools in the treatment of a wide range of cancers. Indeed, aberrant activation of the Hh pathway occurring either by ligand-dependent or -independent mechanisms is a key driver in tumorigenesis. The smoothened (Smo) receptor is one of the main upstream transducers of the Hh signaling and is a validated target for the development of anticancer compounds, as underlined by the FDA-approved Smo antagonist Vismodegib (GDC-0449/Erivedge) for the treatment of basal cell carcinoma. However, Smo mutations that confer constitutive activity and drug resistance have emerged during treatment with Vismodegib. For this reason, the development of new effective Hh inhibitors represents a major challenge for cancer therapy. Natural products have always represented a unique source of lead structures in drug discovery, and in recent years have been used to modulate the Hh pathway at multiple levels. Here, starting from an in house library of natural compounds and their derivatives, we discovered novel chemotypes of Hh inhibitors by mean of virtual screening against the crystallographic structure of Smo. Hh functional based assay identified the chalcone derivative 12 as the most effective Hh inhibitor within the test set. The chalcone 12 binds the Smo receptor and promotes the displacement of Bodipy-Cyclopamine in both Smo WT and drug-resistant Smo mutant. Our molecule stands as a promising Smo antagonist able to specifically impair the growth of Hh-dependent tumor cells in vitro and in vivo and medulloblastoma stem-like cells and potentially overcome the associated drug resistance.
Collapse
Affiliation(s)
- Paola Infante
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Romina Alfonsi
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Cinzia Ingallina
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Deborah Quaglio
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, Italy
| | - Francesca Ghirga
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Ilaria D'Acquarica
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, Italy
| | - Flavia Bernardi
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Laura Di Magno
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Gianluca Canettieri
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Isabella Screpanti
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Alberto Gulino
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| | - Bruno Botta
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, Piazzale Aldo Moro 5, Rome, Italy
| | - Mattia Mori
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Viale Regina Elena 291 Rome, Italy
| | - Lucia Di Marcotullio
- Department of Molecular Medicine, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Sapienza Università di Roma, Viale Regina Elena 291, Rome, Italy
| |
Collapse
|