1
|
Naguib BH, Elsebaie HA, Nafie MS, Mohamady S, Albujuq NR, Samir Ayed A, Nada D, Khalil AF, Hefny SM, Tawfik HO, Shaldam MA. Fragment-based design and synthesis of coumarin-based thiazoles as dual c-MET/STAT-3 inhibitors for potential antitumor agents. Bioorg Chem 2024; 151:107682. [PMID: 39137597 DOI: 10.1016/j.bioorg.2024.107682] [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: 06/30/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 08/15/2024]
Abstract
c-MET and STAT-3 are significant targets for cancer treatments. Here, we describe a class of very effective dual STAT-3 and c-MET inhibitors with coumarin-based thiazoles (3a-o) as its scaffold. Spectroscopic evidence (NMR, HRMS, and HPLC) validated the structural discoveries of the new compounds. The cytotoxic activity of these compounds was also tested against a panel of cancer cells in accordance with US-NCI guidelines. Compound 3g proved to be active at 10 µM, thus it was automatically scheduled to be tested at five doses. Towards SNB-75 (CNS cancer cell line), compound 3g showed notable in vitro anti-cancer activity with GI50 = 1.43 μM. For the molecular targets, compound 3g displayed potent activity towards STAT-3 and c-MET having IC50 of 4.7 µM and 12.67, respectively, compared to Cabozantinib (IC50 = 15 nM of c-MET) and STAT-3-IN-3 (IC50 = 2.1 µM of STAT-3). Moreover, compound 3g significantly induced apoptosis in SNB-75 cells, causing a 3.04-fold increase in apoptotic cell death (treated cells exhibited 11.53 % overall apoptosis, against 3.04 % in reference cells) and a 3.58-fold increase in necrosis. Moreover, it arrests cells at the G2 phase. Dual inhibition of c-MET and STAT-3 protein kinase was further validated using RT-PCR. The target compound's binding mechanism was determined by the application of molecular docking.
Collapse
Affiliation(s)
- Bassem H Naguib
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
| | - Heba A Elsebaie
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Samy Mohamady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
| | - Nader R Albujuq
- Department of Chemistry, School of Science, The University of Jordan, Amman 11942, Jordan
| | - Aya Samir Ayed
- Zoology Department, Faculty of Science, Suez Canal University, P.O. 41522, Ismailia, Egypt
| | - Dina Nada
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University, Egypt
| | - Ahmed F Khalil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Salma M Hefny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, P.O. Box 33516, Kafrelsheikh, Egypt
| |
Collapse
|
2
|
Di Stefano M, Masoni S, Bononi G, Poli G, Galati S, Gado F, Manzi S, Vagaggini C, Brai A, Caligiuri I, Asif K, Rizzolio F, Macchia M, Chicca A, Sodi A, Di Bussolo V, Minutolo F, Meier P, Gertsch J, Granchi C, Dreassi E, Tuccinardi T. Design, synthesis, ADME and biological evaluation of benzylpiperidine and benzylpiperazine derivatives as novel reversible monoacylglycerol lipase (MAGL) inhibitors. Eur J Med Chem 2024; 263:115916. [PMID: 37976705 DOI: 10.1016/j.ejmech.2023.115916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/17/2023] [Accepted: 10/25/2023] [Indexed: 11/19/2023]
Abstract
The degradation of the endocannabinoid 2-arachidonoylglycerol is mediated by the enzyme monoacylglycerol lipase (MAGL), thus generating arachidonic acid, the precursor of prostaglandins and other pro-inflammatory mediators. MAGL also contributes to the hydrolysis of monoacylglycerols into glycerol and fatty acids in peripheral body districts, which may act as pro-tumorigenic signals. For this reason, MAGL inhibitors have been considered as interesting therapeutic agents for their anti-nociceptive, anti-inflammatory, antioxidant and anti-cancer properties. So far, only a limited series of reversible MAGL inhibitors, which are devoid of side effects shown by irreversible inhibitors in animal models, have been reported. Here we optimized a class of benzylpiperidine and benzylpiperazine-based compounds for a reversible MAGL inhibition. The best MAGL inhibitors of this class, compounds 28 and 29, showed a very good inhibition potency, both on the isolated enzyme and in U937 cells, as confirmed by molecular modeling studies that predicted their binding mode into the MAGL active site. Both compounds are characterized by a high selectivity for MAGL versus other serine hydrolases including enzymes of the endocannabinoid system, as confirmed by ABPP experiments in mouse brain membranes. Moreover, very good properties concerning ADME parameters and low in vivo toxicity have been observed for both compounds.
Collapse
Affiliation(s)
- Miriana Di Stefano
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy; Department of Life Sciences, University of Siena, Via Aldo Moro, 2, 53100, Siena, Italy
| | - Samuele Masoni
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Giulia Bononi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Salvatore Galati
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Francesca Gado
- Department of Pharmaceutical Sciences, University of Milan, Via Luigi Mangiagalli 25, 20133, Milan, Italy
| | - Simone Manzi
- Department of Pharmaceutical Sciences, University of Milan, Via Luigi Mangiagalli 25, 20133, Milan, Italy
| | - Chiara Vagaggini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 2, 53100, Siena, Italy
| | - Annalaura Brai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 2, 53100, Siena, Italy
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081, Aviano, Italy
| | - Kanwal Asif
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University, 30123, Venezia, Italy
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081, Aviano, Italy; Department of Molecular Sciences and Nanosystems, Ca' Foscari University, 30123, Venezia, Italy
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012, Bern, Switzerland
| | - Andrea Sodi
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139, Florence, Italy
| | - Valeria Di Bussolo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Filippo Minutolo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy; Center for Instrument Sharing of the University of Pisa (CISUP), Lungarno Pacinotti 43, 56126, Pisa, Italy
| | - Philip Meier
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012, Bern, Switzerland
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012, Bern, Switzerland
| | - Carlotta Granchi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy; Center for Instrument Sharing of the University of Pisa (CISUP), Lungarno Pacinotti 43, 56126, Pisa, Italy.
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 2, 53100, Siena, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy; Center for Instrument Sharing of the University of Pisa (CISUP), Lungarno Pacinotti 43, 56126, Pisa, Italy
| |
Collapse
|
3
|
Chen Y, Zhai N, Zhu Y, Yue P, Verma N, Brotherton-Pleiss C, Fu W, Nakamura K, Chen W, Kawakami J, Murali R, Tius MA, Lopez-Tapia F, Turkson J. Azetidine ring, salicylic acid, and salicylic acid bioisosteres as determinants of the binding characteristics of novel potent compounds to Stat3. Bioorg Med Chem Lett 2024; 97:129565. [PMID: 38008341 DOI: 10.1016/j.bmcl.2023.129565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 11/08/2023] [Accepted: 11/22/2023] [Indexed: 11/28/2023]
Affiliation(s)
- Yue Chen
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Ning Zhai
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Yinsong Zhu
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Peibin Yue
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Nagendra Verma
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Christine Brotherton-Pleiss
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo St., Honolulu, HI 96813, USA; Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI 96825, USA
| | - Wenzhen Fu
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo St., Honolulu, HI 96813, USA; Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI 96825, USA
| | - Kayo Nakamura
- Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI 96825, USA
| | - Weiliang Chen
- Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI 96825, USA
| | - Joel Kawakami
- Department of Natural Sciences and Mathematics, Chaminade University, 3140 Waialae Avenue, Honolulu, HI 96816, USA
| | - Ramachandran Murali
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA
| | - Marcus A Tius
- Cancer Biology Program, University of Hawaii Cancer Center, University of Hawaii at Manoa, 701 Ilalo St., Honolulu, HI 96813, USA; Department of Chemistry, University of Hawaii, Manoa, 2545 McCarthy Mall, Honolulu, HI 96825, USA
| | - Francisco Lopez-Tapia
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA.
| | - James Turkson
- Department of Medicine, Division of Medical Oncology, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA; Cancer Biology Program, Cedars-Sinai Cancer, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA.
| |
Collapse
|
4
|
Rafiq H, Hu J, Hakami MA, Hazazi A, Alamri MA, Alkhatabi HA, Mahmood A, Alotaibi BS, Wadood A, Huang X. Identification of novel STAT3 inhibitors for liver fibrosis, using pharmacophore-based virtual screening, molecular docking, and biomolecular dynamics simulations. Sci Rep 2023; 13:20147. [PMID: 37978263 PMCID: PMC10656421 DOI: 10.1038/s41598-023-46193-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/29/2023] [Indexed: 11/19/2023] Open
Abstract
The signal transducer and activator of transcription 3 (STAT3) plays a fundamental role in the growth and regulation of cellular life. Activation and over-expression of STAT3 have been implicated in many cancers including solid blood tumors and other diseases such as liver fibrosis and rheumatoid arthritis. Therefore, STAT3 inhibitors are be coming a growing and interesting area of pharmacological research. Consequently, the aim of this study is to design novel inhibitors of STAT3-SH3 computationally for the reduction of liver fibrosis. Herein, we performed Pharmacophore-based virtual screening of databases including more than 19,481 commercially available compounds and in-house compounds. The hits obtained from virtual screening were further docked with the STAT3 receptor. The hits were further ranked on the basis of docking score and binding interaction with the active site of STAT3. ADMET properties of the screened compounds were calculated and filtered based on drug-likeness criteria. Finally, the top five drug-like hit compounds were selected and subjected to molecular dynamic simulation. The stability of each drug-like hit in complex with STAT3 was determined by computing their RMSD, RMSF, Rg, and DCCM analyses. Among all the compounds Sa32 revealed a good docking score, interactions, and stability during the entire simulation procedure. As compared to the Reference compound, the drug-like hit compound Sa32 showed good docking scores, interaction, stability, and binding energy. Therefore, we identified Sa32 as the best small molecule potent inhibitor for STAT3 that will be helpful in the future for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Huma Rafiq
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, Abdul Wali Khan University, Mardan, Pakistan
| | - Junjian Hu
- Department of Central Laboratory, Dongguan Songshan Lake Central Hospital, Dongguan, China
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia
| | - Mubarak A Alamri
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Hind A Alkhatabi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Arif Mahmood
- Center for Medical Genetics and Human Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Bader S Alotaibi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | - Abdul Wadood
- Department of Biochemistry, Computational Medicinal Chemistry Laboratory, Abdul Wali Khan University, Mardan, Pakistan.
| | - Xiaoyun Huang
- Department of Neurology, Dongguan Songshan Lake Central Hospital, Dongguan, China.
| |
Collapse
|
5
|
In silico drug design and molecular docking of novel amidophosphonates and sulfamidophosphonates as inhibitors of urokinase-type plasminogen activator. J INDIAN CHEM SOC 2022. [DOI: 10.1016/j.jics.2022.100650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
6
|
Bononi G, Di Stefano M, Poli G, Ortore G, Meier P, Masetto F, Caligiuri I, Rizzolio F, Macchia M, Chicca A, Avan A, Giovannetti E, Vagaggini C, Brai A, Dreassi E, Valoti M, Minutolo F, Granchi C, Gertsch J, Tuccinardi T. Reversible Monoacylglycerol Lipase Inhibitors: Discovery of a New Class of Benzylpiperidine Derivatives. J Med Chem 2022; 65:7118-7140. [PMID: 35522977 PMCID: PMC9150076 DOI: 10.1021/acs.jmedchem.1c01806] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Monoacylglycerol
lipase (MAGL) is the enzyme responsible for the
metabolism of 2-arachidonoylglycerol in the brain and the hydrolysis
of peripheral monoacylglycerols. Many studies demonstrated beneficial
effects deriving from MAGL inhibition for neurodegenerative diseases,
inflammatory pathologies, and cancer. MAGL expression is increased
in invasive tumors, furnishing free fatty acids as pro-tumorigenic
signals and for tumor cell growth. Here, a new class of benzylpiperidine-based
MAGL inhibitors was synthesized, leading to the identification of 13, which showed potent reversible and selective MAGL inhibition.
Associated with MAGL overexpression and the prognostic role in pancreatic
cancer, derivative 13 showed antiproliferative activity
and apoptosis induction, as well as the ability to reduce cell migration
in primary pancreatic cancer cultures, and displayed a synergistic
interaction with the chemotherapeutic drug gemcitabine. These results
suggest that the class of benzylpiperidine-based MAGL inhibitors have
potential as a new class of therapeutic agents and MAGL could play
a role in pancreatic cancer.
Collapse
Affiliation(s)
- Giulia Bononi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Miriana Di Stefano
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy.,Department of Life Sciences, University of Siena, Via Aldo Moro, 2, 53100 Siena, Italy
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Gabriella Ortore
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Philip Meier
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland
| | - Francesca Masetto
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, DeBoelelaan 1117, 1081HV Amsterdam, The Netherlands
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy.,Department of Molecular Sciences and Nanosystems, Ca' Foscari University, 30123 Venezia, Italy
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Science, Mashhad 91886-17871, Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, DeBoelelaan 1117, 1081HV Amsterdam, The Netherlands.,Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, via Giovannini 13, 56017 San Giuliano Terme, Pisa, Italy
| | - Chiara Vagaggini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 2, 53100 Siena, Italy
| | - Annalaura Brai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 2, 53100 Siena, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro, 2, 53100 Siena, Italy
| | - Massimo Valoti
- Department of Life Sciences, University of Siena, Via Aldo Moro, 2, 53100 Siena, Italy
| | - Filippo Minutolo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy.,Center for Instrument Sharing of the University of Pisa (CISUP), Lungarno Pacinotti 43, 56126 Pisa, Italy
| | - Carlotta Granchi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy.,Center for Instrument Sharing of the University of Pisa (CISUP), Lungarno Pacinotti 43, 56126 Pisa, Italy
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy.,Center for Instrument Sharing of the University of Pisa (CISUP), Lungarno Pacinotti 43, 56126 Pisa, Italy
| |
Collapse
|
7
|
Abdalla AN, Di Stefano M, Poli G, Tuccinardi T, Bader A, Vassallo A, Abdallah ME, El-Readi MZ, Refaat B, Algarni AS, Ahmad R, Alkahtani HM, Abdel-Aziz AAM, El-Azab AS, Alqathama A. Co-Inhibition of P-gp and Hsp90 by an Isatin-Derived Compound Contributes to the Increase of the Chemosensitivity of MCF7/ADR-Resistant Cells to Doxorubicin. Molecules 2021; 27:molecules27010090. [PMID: 35011321 PMCID: PMC8746493 DOI: 10.3390/molecules27010090] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is a complex and multi-drug resistant (MDR) disease, which could result in the failure of many chemotherapeutic clinical agents. Discovering effective molecules from natural products or by derivatization from known compounds is the interest of many research studies. The first objective of the present study is to investigate the cytotoxic combinatorial, chemosensitizing, and apoptotic effects of an isatin derived compound (5,5-diphenylimidazolidine-2,4-dione conjugated with 5-substituted isatin, named HAA2021 in the present study) against breast cancer cells (MCF7) and breast cancer cells resistant to doxorubicin (MCF7/ADR) when combined with doxorubicin. The second objective is to investigate the binding mode of HAA2021 withP-glycoprotein (P-gp) and heat shock protein 90 (Hsp90), and to determine whether their co-inhibition by HAA2021 contribute to the increase of the chemosensitization of MCF7/ADR cells to doxorubicin. The combination of HAA2021, at non-toxic doses, with doxorubicin synergistically inhibited the proliferation while inducing significant apoptosis in MCF7 cells. Moreover, HAA2021 increased the chemosensitization of MCF7/ADR cells to doxorubicin, resulting in increased cytotoxicity/selectivity and apoptosis-inducing efficiency compared with the effect of doxorubicin or HAA2021 alone against MCF7/ADR cells. Molecular modeling showed that two molecules of HAA2021 bind to P-gp at the same time, causing P-gp inhibitory effect of the MDR efflux pump, and accumulation of Rhodamine-123 (Rho123) in MCF7/ADR cells. Furthermore, HAA2021 stably interacted with Hsp90α more efficiently compared with 17-N-allylamino-17-demethoxygeldanamycin (17-AAG), which was confirmed with the surface plasmon resonance (SPR) and molecular modeling studies. Additionally, HAA2021 showed multi-target effects via the inhibition of Hsp90 and nuclear factor kappa B (NF-𝜅B) proteins in MCF7 and MCF7/ADR cells. Results of real time-PCR also confirmed the synergistic co-inhibition of P-gp/Hsp90α genes in MCF7/ADR cells. Further pharmacokinetic and in vivo studies are warranted for HAA2021 to confirm its anticancer capabilities.
Collapse
Affiliation(s)
- Ashraf N. Abdalla
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.B.); (A.S.A.); (A.A.)
- Medicinal and Aromatic Plants Research Institute, National Center for Research, Khartoum 2404, Sudan
- Correspondence: or
| | - Miriana Di Stefano
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.S.); (G.P.); (T.T.)
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.S.); (G.P.); (T.T.)
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (M.D.S.); (G.P.); (T.T.)
| | - Ammar Bader
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.B.); (A.S.A.); (A.A.)
| | - Antonio Vassallo
- Dipartimento di Scienze, Università Degli Studi della Basilicata, 85100 Potenza, Italy;
| | - Mohamed E. Abdallah
- Department of Clinical Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.Z.E.-R.)
| | - Mahmoud Zaki El-Readi
- Department of Clinical Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.Z.E.-R.)
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Bassem Refaat
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Alanood S. Algarni
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.B.); (A.S.A.); (A.A.)
| | - Rizwan Ahmad
- Natural Products and Alternative Medicines, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia;
| | - Hamad M. Alkahtani
- College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (H.M.A.); (A.A.-M.A.-A.); (A.S.E.-A.)
| | - Alaa A.-M. Abdel-Aziz
- College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (H.M.A.); (A.A.-M.A.-A.); (A.S.E.-A.)
| | - Adel S. El-Azab
- College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (H.M.A.); (A.A.-M.A.-A.); (A.S.E.-A.)
| | - Aljawharah Alqathama
- College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (A.B.); (A.S.A.); (A.A.)
| |
Collapse
|
8
|
Beshay BY, Abdellatef AA, Loksha YM, Fahmy SM, Habib NS, Bekhit AEDA, Georghiou PE, Hayakawa Y, Bekhit AA. Design and synthesis of 2-Substituted-4-benzyl-5-methylimidazoles as new potential Anti-breast cancer agents to inhibit oncogenic STAT3 functions. Bioorg Chem 2021; 113:105033. [PMID: 34089945 DOI: 10.1016/j.bioorg.2021.105033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/17/2021] [Accepted: 05/24/2021] [Indexed: 12/31/2022]
Abstract
STAT3 signaling is known to be associated with tumorigenesis and further cancer cell-intrinsic activation of STAT3 leads to altered regulation of several oncogenic processes. Given the importance of STAT3 in cancer development and progression particularly breast cancer, it is crucial to discover new chemical entities of STAT3 inhibitor to develop anti-breast cancer drug candidates. Herein, 4-benzyl-2-benzylthio-5-methyl-1H-imidazole (2a) and 4-benzyl-5-methyl-2-[(2,6-difluorobenzyl)thio]-1H-imidazole (2d) from a group of thirty imidazole-bearing compounds showed greater STAT3 inhibition than their lead compounds VS1 and the oxadiazole derivative MD77. Within all tested compounds, ten derivatives effectively inhibited the growth of the two tested breast cancer cells with IC50 values ranging from 6.66 to 26.02 µM. In addition, the most potent derivatives 2a and 2d inhibited the oncogenic function of STAT3 as seen in the inhibition of colony formation and IL-6 production of breast cancer cell lines. Modeling studies provided evidence for the possible interactions of the synthesized compounds with the key residues of the STAT3-SH2 domain. Collectively, our present study suggests 2-substituted-4-benzyl-5-methylimidazoles are a new class of anti-cancer drug candidates to inhibit oncogenic STAT3 function.
Collapse
Affiliation(s)
- Botros Y Beshay
- Department of Pharmaceutical Chemistry, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria, Egypt
| | - Amira A Abdellatef
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Yasser M Loksha
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University, Al-Arish, North Sinai, Egypt
| | - Salwa M Fahmy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, 2152 Alexandria, Egypt
| | - Nargues S Habib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, 2152 Alexandria, Egypt
| | | | - Paris E Georghiou
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Yoshihiro Hayakawa
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan.
| | - Adnan A Bekhit
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, 2152 Alexandria, Egypt; Pharmacy Program, Allied Health Department, College of Health and Sport Sciences, University of Bahrain, Bahrain; Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| |
Collapse
|
9
|
Mohammadian J, Mahmoudi S, Pourmohammad P, Pirouzpanah M, Salehnia F, Maroufi NF, Samadi N, Sabzichi M. Formulation of Stattic as STAT3 inhibitor in nanostructured lipid carriers (NLCs) enhances efficacy of doxorubicin in melanoma cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2020; 393:2315-2323. [PMID: 32653978 DOI: 10.1007/s00210-020-01942-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/03/2020] [Indexed: 01/29/2023]
Abstract
Nowadays, nanoparticle-based combination therapy has been emerging as huge innovation in cancer treatment. Here, we studied the effect of Stattic (STAT3 inhibitor) loaded in nanostructured lipid carriers (NLCs) on enhancing the efficacy, cytotoxicity, and induction of apoptosis of doxorubicin in B16F10 mouse melanoma cancer cell. The evaluation of Stattic-loaded NLCs has been done in terms of zeta potential, particle size, scanning electron microscope (SEM), and cellular uptake. MTT assay was applied to evaluate the cell proliferation. Apoptotic cell death and identification of early and late apoptosis were assessed by DAPI staining and Annexin V/PI staining, respectively. Real-time RT-PCR was applied to measure the effects of doxorubicin and/or Stattic on key apoptotic genes such as Bad, Survivin, HIF1, and STAT3. The Stattic formulated into NLCs shown mean particle size of 56 ± 7 nm which was confirmed by SEM. The IC50 values for Stattic and doxorubicin were 2.95 ± 0.52 μM and 1.21 ± 0.36 μM, respectively. Stattic-loaded NLCs diminished percent of cell proliferation from 68 ± 6.8 to 54 ± 3.7% (p < 0.05). Combinational treatment of the cells with Stattic-loaded nanoparticles and doxorubicin give rise to a significant increase in the percentage of apoptosis (p < 0.05). The study of gene expression profile has shown a remarkable decrease in anti-apoptotic gene, Survivin, along with smooth decline in HIF1 as angiogenesis intermediator and increase in Bad mRNA levels. Our results recommend that NLCs as novel technology have potent strategy to augment efficacy of current chemotherapeutic agent in melanoma cancer cells.
Collapse
Affiliation(s)
- Jamal Mohammadian
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shiva Mahmoudi
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Science, Ardabil, Iran
| | - Pirouz Pourmohammad
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Science, Ardabil, Iran
| | - Mohammad Pirouzpanah
- Department of Genetic and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, 34755, Istanbul, Turkey
| | - Fatemeh Salehnia
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Fathi Maroufi
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Drug Applied Research Center, and Department of Medical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Sabzichi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
10
|
Granchi C, Bononi G, Ferrisi R, Gori E, Mantini G, Glasmacher S, Poli G, Palazzolo S, Caligiuri I, Rizzolio F, Canzonieri V, Perin T, Gertsch J, Sodi A, Giovannetti E, Macchia M, Minutolo F, Tuccinardi T, Chicca A. Design, synthesis and biological evaluation of second-generation benzoylpiperidine derivatives as reversible monoacylglycerol lipase (MAGL) inhibitors. Eur J Med Chem 2020; 209:112857. [PMID: 33045662 DOI: 10.1016/j.ejmech.2020.112857] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
An interesting enzyme of the endocannabinoid system is monoacylglycerol lipase (MAGL). This enzyme, which metabolizes the endocannabinoid 2-arachidonoylglycerol (2-AG), has attracted great interest due to its involvement in several physiological and pathological processes, such as cancer progression. Experimental evidences highlighted some drawbacks associated with the use of irreversible MAGL inhibitors in vivo, therefore the research field concerning reversible inhibitors is rapidly growing. In the present manuscript, the class of benzoylpiperidine-based MAGL inhibitors was further expanded and optimized. Enzymatic assays identified some compounds in the low nanomolar range and steered molecular dynamics simulations predicted the dissociation itinerary of one of the best compounds from the enzyme, confirming the observed structure-activity relationship. Biological evaluation, including assays in intact U937 cells and competitive activity-based protein profiling experiments in mouse brain membranes, confirmed the selectivity of the selected compounds for MAGL versus other components of the endocannabinoid system. An antiproliferative ability in a panel of cancer cell lines highlighted their potential as potential anticancer agents. Future studies on the potential use of these compounds in the clinical setting are also supported by the inhibition of cell growth observed both in cancer organoids derived from high grade serous ovarian cancer patients and in pancreatic ductal adenocarcinoma primary cells, which showed genetic and histological features very similar to the primary tumors.
Collapse
Affiliation(s)
- Carlotta Granchi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Giulia Bononi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Rebecca Ferrisi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Eleonora Gori
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Giulia Mantini
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, DeBoelelaan 1117, 1081HV, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana per La Scienza, Via Giovannini 13, 56017, San Giuliano Terme, Pisa, Italy
| | - Sandra Glasmacher
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012, Bern, Switzerland
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081, Aviano, Italy
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081, Aviano, Italy
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081, Aviano, Italy; Department of Molecular Sciences and Nanosystems, Ca' Foscari University, 30123, Venezia, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081, Aviano, Italy; Department of Medical, Surgical and Health Sciences, Università Degli Studi di Trieste, Strada di Fiume 447, Trieste, Italy
| | - Tiziana Perin
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081, Aviano, Italy
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012, Bern, Switzerland
| | - Andrea Sodi
- Department of Neurosciences, Psychology, Drug Research and Child Health Eye Clinic, University of Florence, AOU Careggi, 50139, Florence, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, DeBoelelaan 1117, 1081HV, Amsterdam, the Netherlands; Cancer Pharmacology Lab, Fondazione Pisana per La Scienza, Via Giovannini 13, 56017, San Giuliano Terme, Pisa, Italy
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Filippo Minutolo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126, Pisa, Italy.
| | - Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012, Bern, Switzerland
| |
Collapse
|
11
|
Novel Approaches to Target Mutant FLT3 Leukaemia. Cancers (Basel) 2020; 12:cancers12102806. [PMID: 33003568 PMCID: PMC7600363 DOI: 10.3390/cancers12102806] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/24/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is a haematologic disease in which oncogenic mutations in the receptor tyrosine kinase FLT3 frequently lead to leukaemic development. Potent treatment of AML patients is still hampered by inefficient targeting of leukemic stem cells expressing constitutive active FLT3 mutants. This review summarizes the current knowledge about the regulation of FLT3 activity at cellular level and discusses therapeutical options to affect the tumor cells and the microenvironment to impair the haematological aberrations. Abstract Fms-like tyrosine kinase 3 (FLT3) is a member of the class III receptor tyrosine kinases (RTK) and is involved in cell survival, proliferation, and differentiation of haematopoietic progenitors of lymphoid and myeloid lineages. Oncogenic mutations in the FLT3 gene resulting in constitutively active FLT3 variants are frequently found in acute myeloid leukaemia (AML) patients and correlate with patient’s poor survival. Targeting FLT3 mutant leukaemic stem cells (LSC) is a key to efficient treatment of patients with relapsed/refractory AML. It is therefore essential to understand how LSC escape current therapies in order to develop novel therapeutic strategies. Here, we summarize the current knowledge on mechanisms of FLT3 activity regulation and its cellular consequences. Furthermore, we discuss how aberrant FLT3 signalling cooperates with other oncogenic lesions and the microenvironment to drive haematopoietic malignancies and how this can be harnessed for therapeutical purposes.
Collapse
|
12
|
Abstract
Background:
Molecular docking is probably the most popular and profitable approach in
computer-aided drug design, being the staple technique for predicting the binding mode of bioactive
compounds and for performing receptor-based virtual screening studies. The growing attention received
by docking, as well as the need for improving its reliability in pose prediction and virtual screening
performance, has led to the development of a wide plethora of new docking algorithms and scoring
functions. Nevertheless, it is unlikely to identify a single procedure outperforming the other ones in
terms of reliability and accuracy or demonstrating to be generally suitable for all kinds of protein targets.
Methods:
In this context, consensus docking approaches are taking hold in computer-aided drug design.
These computational protocols consist in docking ligands using multiple docking methods and then
comparing the binding poses predicted for the same ligand by the different methods. This analysis is
usually carried out calculating the root-mean-square deviation among the different docking results obtained
for each ligand, in order to identify the number of docking methods producing the same binding
pose.
Results:
The consensus docking approaches demonstrated to improve the quality of docking and virtual
screening results compared to the single docking methods. From a qualitative point of view, the improvement
in pose prediction accuracy was obtained by prioritizing ligand binding poses produced by a
high number of docking methods, whereas with regards to virtual screening studies, high hit rates were
obtained by prioritizing the compounds showing a high level of pose consensus.
Conclusion:
In this review, we provide an overview of the results obtained from the performance assessment
of various consensus docking protocols and we illustrate successful case studies where consensus
docking has been applied in virtual screening studies.
Collapse
Affiliation(s)
- Giulio Poli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | |
Collapse
|
13
|
Bechlem K, Aissaoui M, Belhani B, Rachedi KO, Bouacida S, Bahadi R, Djouad SE, Ben Mansour R, Bouaziz M, Almalki F, Ben Hadda T, Berredjem M. Synthesis, X-ray crystallographic study and molecular docking of new α-sulfamidophosphonates: POM analyses of their cytotoxic activity. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.127990] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
14
|
Bisio A, Schito AM, Pedrelli F, Danton O, Reinhardt JK, Poli G, Tuccinardi T, Bürgi T, De Riccardis F, Giacomini M, Calzia D, Panfoli I, Schito GC, Hamburger M, De Tommasi N. Antibacterial and ATP Synthesis Modulating Compounds from Salvia tingitana. JOURNAL OF NATURAL PRODUCTS 2020; 83:1027-1042. [PMID: 32182064 PMCID: PMC7997632 DOI: 10.1021/acs.jnatprod.9b01024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Indexed: 05/05/2023]
Abstract
A surface extract of the aerial parts of Salvia tingitana afforded a nor-sesterterpenoid (1) and eight new sesterterpenoids (2-̵9), along with five known sesterterpenoids, five labdane and one abietane diterpenoid, one sesquiterpenoid, and four flavonoids. The structures of the new compounds were established by 1D and 2D NMR spectroscopy, HRESIMS, and VCD data and Mosher's esters analysis. The antimicrobial activity of compounds was evaluated against 30 human pathogens including 27 clinical strains and three isolates of marine origin for their possible implications on human health. The methyl ester of salvileucolide (10), salvileucolide-6,23-lactone (11), sclareol (15), and manool (17) were the most active against Gram-positive bacteria. The compounds were also tested for the inhibition of ATP production in purified mammalian rod outer segments. Terpenoids 10, 11, 15, and 17 inhibited ATP production, while only 17 inhibited also ATP hydrolysis. Molecular modeling studies confirmed the capacity of 17 to interact with mammalian ATP synthase. A significant reduction of ATP production in the presence of 17 was observed in Enterococcus faecalis and E. faecium isolates.
Collapse
Affiliation(s)
- Angela Bisio
- Department
of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Anna M. Schito
- Department
of Integrated Surgical and Diagnostical Sciences, University of Genova, Largo Rosanna Benzi 8, 16145 Genova, Italy
| | - Francesca Pedrelli
- Department
of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Ombeline Danton
- Department
of Pharmaceutical Sciences, University of
Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Jakob K. Reinhardt
- Department
of Pharmaceutical Sciences, University of
Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Giulio Poli
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Tiziano Tuccinardi
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Thomas Bürgi
- Department
of Chemical Physics, University of Geneva, 30 Quai Ernest-Ansermet, 1211 Genève 4, Switzerland
| | - Francesco De Riccardis
- Department
of Chemistry and Biology, University of
Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy
| | - Mauro Giacomini
- Department
of Informatics Bioengineering Robotics and System Engineering, University of Genova, Via all’Opera Pia, 13, 16145 Genova, Italy
| | - Daniela Calzia
- Department
of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Isabella Panfoli
- Department
of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Gian Carlo Schito
- Department
of Pharmacy, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Matthias Hamburger
- Department
of Pharmaceutical Sciences, University of
Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Nunziatina De Tommasi
- Department
of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy
| |
Collapse
|
15
|
Discovery of Novel µ-Opioid Receptor Inverse Agonist from a Combinatorial Library of Tetrapeptides through Structure-Based Virtual Screening. Molecules 2019; 24:molecules24213872. [PMID: 31717871 PMCID: PMC6865014 DOI: 10.3390/molecules24213872] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/24/2019] [Accepted: 10/26/2019] [Indexed: 11/18/2022] Open
Abstract
Morphine, oxycodone, fentanyl, and other µ-opioid receptors (MOR) agonists have been used for decades in antinociceptive therapies. However, these drugs are associated with numerous side effects, such as euphoria, addiction, respiratory depression, and adverse gastrointestinal reactions, thus, circumventing these drawbacks is of extensive importance. With the aim of identifying novel peptide ligands endowed with MOR inhibitory activity, we developed a virtual screening protocol, including receptor-based pharmacophore screening, docking studies, and molecular dynamics simulations, which was used to filter an in-house built virtual library of tetrapeptide ligands. The three top-scored compounds were synthesized and subjected to biological evaluation, revealing the identity of a hit compound (peptide 1) endowed with appreciable MOR inverse agonist effect and selectivity over δ-opioid receptors. These results confirmed the reliability of our computational approach and provided a promising starting point for the development of new potent MOR modulators.
Collapse
|
16
|
D'Andrea F, Vagelli G, Granchi C, Guazzelli L, Tuccinardi T, Poli G, Iacopini D, Minutolo F, Di Bussolo V. Synthesis and Biological Evaluation of New Glycoconjugated LDH Inhibitors as Anticancer Agents. Molecules 2019; 24:E3520. [PMID: 31569409 PMCID: PMC6804087 DOI: 10.3390/molecules24193520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/17/2022] Open
Abstract
Conjugation of known biologically active molecules to carbohydrate frameworks represents a valuable option for the preparation of hybrid, structurally-related families of compounds with the aim of modulating their biological response. Therefore, we present here a study on the preparation of d-galacto, d-manno, d-gluco, and d-lactose glycoconjugates of an established N-hydroxyindole-based (NHI) inhibitor of lactated dehydrogenase (LDH). Structural variations involved the sugar stereochemistry and size as well as the anchoring point of the NHI on the carbohydrate frame (either C-1 or C-6). In the case of the galactose anomeric glycoconjugate (C-1), intriguing solvent-dependent effects were observed in the glycosylation stereochemical outcome. The biological activity of the deprotected glycoconjugates in contrasting lactate formation and cancer cell proliferation are described.
Collapse
Affiliation(s)
- Felicia D'Andrea
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy.
| | - Giulia Vagelli
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi 3, 56124 Pisa, Italy.
| | - Carlotta Granchi
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy.
| | - Lorenzo Guazzelli
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy.
| | - Tiziano Tuccinardi
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy.
| | - Giulio Poli
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy.
| | - Dalila Iacopini
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi 3, 56124 Pisa, Italy.
| | - Filippo Minutolo
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy.
| | - Valeria Di Bussolo
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via G. Moruzzi 3, 56124 Pisa, Italy.
| |
Collapse
|
17
|
Russo Spena C, De Stefano L, Poli G, Granchi C, El Boustani M, Ecca F, Grassi G, Grassi M, Canzonieri V, Giordano A, Tuccinardi T, Caligiuri I, Rizzolio F. Virtual screening identifies a PIN1 inhibitor with possible antiovarian cancer effects. J Cell Physiol 2019; 234:15708-15716. [PMID: 30697729 DOI: 10.1002/jcp.28224] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/10/2019] [Indexed: 02/06/2023]
Abstract
Peptidyl-prolyl cis-trans isomerase, NIMA-interacting 1 (PIN1) is a peptidyl-prolyl isomerase that binds phospho-Ser/Thr-Pro motifs in proteins and catalyzes the cis-trans isomerization of proline peptide bonds. PIN1 is overexpressed in several cancers including high-grade serous ovarian cancer. Since few therapies are effective against this cancer, PIN1 could be a therapeutic target but effective PIN1 inhibitors are lacking. To identify molecules with in vivo inhibitory effects on PIN1, we used consensus docking to model existing PIN1-ligand X-ray structures and to screen a chemical database for candidate inhibitors. Ten molecules were selected and tested in cellular assays, leading to the identification of VS10 that bound and inhibited PIN1. VS10 treatment reduced the viability of ovarian cancer cell lines by inducing proteasomal PIN1 degradation, without effects on PIN1 transcription, and also reduced the levels of downstream targets β-catenin, cyclin D1, and pSer473-Akt. VS10 is a selective PIN1 inhibitor that may offer new opportunities for treating PIN1-overexpressing tumors.
Collapse
Affiliation(s)
- Concetta Russo Spena
- Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, Aviano, Italy.,Doctoral School in Chemistry, University of Trieste, Trieste, Italy
| | - Lucia De Stefano
- Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, Aviano, Italy.,Doctoral School in Chemistry, University of Trieste, Trieste, Italy
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Maguie El Boustani
- Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, Aviano, Italy.,Doctoral School in Molecular Biomedicine, University of Trieste, Trieste, Italy
| | - Fabrizio Ecca
- Experimental and Clinical Pharmacology Unit, IRCCS CRO Aviano-National Cancer Institute, Aviano, Italy
| | - Gabriele Grassi
- Department of Life Sciences, Cattinara University Hospital, University of Trieste, Trieste, Italy
| | - Mario Grassi
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Vincenzo Canzonieri
- Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, Aviano, Italy.,Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania.,Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Antonio Giordano
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania
| | - Isabella Caligiuri
- Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, Aviano, Italy
| | - Flavio Rizzolio
- Pathology Unit, IRCCS CRO Aviano-National Cancer Institute, Aviano, Italy.,Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania.,Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, Venezia-Mestre, Italy
| |
Collapse
|
18
|
Identification of novel inhibitors of signal transducer and activator of transcription 3 over signal transducer and activator of transcription 1 for the treatment of breast cancer by in-silico and in-vitro approach. Process Biochem 2019. [DOI: 10.1016/j.procbio.2019.04.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
19
|
Krasavin M, Shetnev A, Baykov S, Kalinin S, Nocentini A, Sharoyko V, Poli G, Tuccinardi T, Korsakov M, Tennikova TB, Supuran CT. Pyridazinone-substituted benzenesulfonamides display potent inhibition of membrane-bound human carbonic anhydrase IX and promising antiproliferative activity against cancer cell lines. Eur J Med Chem 2019; 168:301-314. [PMID: 30826507 DOI: 10.1016/j.ejmech.2019.02.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/10/2019] [Accepted: 02/12/2019] [Indexed: 01/09/2023]
Abstract
An expanded set of pyridazine-containing benzene sulfonamides was investigated for inhibition of four human carbonic anhydrase isoforms, which revealed a pronounced inhibition trend toward hCA IX, a cancer-related, membrane-bound isoform of the enzyme. Comparison of antiproliferative effects of these compounds against cancer (PANC-1) and normal (ARPE-19) cells at 50 μM concentration narrowed the selection of compounds to the eight which displayed selective growth inhibition toward the cancer cells. More detailed investigation in concentration-dependent mode against normal (ARPE-19) and two cancer cell lines (PANC-1 and SK-MEL-2) identified two lead compounds one of which displayed a notable cytotoxicity toward pancreatic cancer cells while the other targeted the melanoma cells. These findings significantly expand the knowledge base concerning the hCA IX inhibitors whose inhibitory potency against a recombinant enzyme translates into selective anticancer activity under hypoxic conditions which are aimed to model the environment of a growing tumor.
Collapse
Affiliation(s)
- Mikhail Krasavin
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation.
| | - Anton Shetnev
- The Ushinsky Yaroslavl State Pedagogical University, Yaroslavl, 150000, Russian Federation
| | - Sergey Baykov
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Stanislav Kalinin
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Alessio Nocentini
- Neurofarba Department, Universita degli Studi di Firenze, Florence, Italy
| | - Vladimir Sharoyko
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | | | - Mikhail Korsakov
- The Ushinsky Yaroslavl State Pedagogical University, Yaroslavl, 150000, Russian Federation
| | - Tatiana B Tennikova
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Claudiu T Supuran
- Neurofarba Department, Universita degli Studi di Firenze, Florence, Italy.
| |
Collapse
|
20
|
Bizzarri B, Botta L, Aversa D, Mercuri NB, Poli G, Barbieri A, Berretta N, Saladino R. L-DOPA-quinone Mediated Recovery from GIRK Channel Firing Inhibition in Dopaminergic Neurons. ACS Med Chem Lett 2019; 10:431-436. [PMID: 30996775 PMCID: PMC6466524 DOI: 10.1021/acsmedchemlett.8b00477] [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: 10/11/2018] [Accepted: 01/09/2019] [Indexed: 01/07/2023] Open
Abstract
The oxidative degeneration of dopamine-releasing (DAergic) neurons in the substantia nigra pars compacta (SNc) has attracted much interest in preclinical research, due to its involvement in Parkinson's disease manifestations. Evidence exists on the participation of quinone derivatives in mitochondrial dysfunction, alpha synuclein protein aggregation, and protein degradation. With the aim to investigate the role of L-DOPA-quinone in DAergic neuron functions, we synthesized L-DOPA-quinone by use of 2-iodoxybenzoic acid and measured its activity in recovery from dopamine-mediated firing inhibition of SNc neurons. Noteworthy, L-DOPA-quinone counteracts firing inhibition in SNc DAergic neurons caused by GIRK opening. A possible mechanism to explain the effect of L-DOPA-quinone on GIRK channel has been proposed by computational models. Overall, the study showed the possibility that L-DOPA-quinone stabilizes GIRK in a preopen conformation through formation of a covalent adduct with cysteine-65 on the GIRK2 subunit of the protein.
Collapse
Affiliation(s)
- Bruno
M. Bizzarri
- Dipartimento
di Scienze Ecologiche e Biologiche, Università
della Tuscia, Via S. C. De Lellis 44, 01100 Viterbo, Italy
| | - Lorenzo Botta
- Dipartimento
di Scienze Ecologiche e Biologiche, Università
della Tuscia, Via S. C. De Lellis 44, 01100 Viterbo, Italy
| | - Daniela Aversa
- IRCCS
Fondazione Santa Lucia, Via Ardeatina, 306/354, 00142 Rome, Italy
| | - Nicola B. Mercuri
- IRCCS
Fondazione Santa Lucia, Via Ardeatina, 306/354, 00142 Rome, Italy
- Dipartimento
di Medicina dei Sistemi, Università
di Roma Tor Vergata, Via Montpellier, 1, 00133 Rome, Italy
| | - Giulio Poli
- Dipartimento
di Biotecnologie, Chimica e Farmacia, Università
degli studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Alessandro Barbieri
- Dipartimento
di Biotecnologie, Chimica e Farmacia, Università
degli studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Nicola Berretta
- IRCCS
Fondazione Santa Lucia, Via Ardeatina, 306/354, 00142 Rome, Italy
| | - Raffaele Saladino
- Dipartimento
di Scienze Ecologiche e Biologiche, Università
della Tuscia, Via S. C. De Lellis 44, 01100 Viterbo, Italy
| |
Collapse
|
21
|
Lapillo M, Salis B, Palazzolo S, Poli G, Granchi C, Minutolo F, Rotondo R, Caligiuri I, Canzonieri V, Tuccinardi T, Rizzolio F. First-of-its-kind STARD 3 Inhibitor: In Silico Identification and Biological Evaluation as Anticancer Agent. ACS Med Chem Lett 2019; 10:475-480. [PMID: 30996782 DOI: 10.1021/acsmedchemlett.8b00509] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 02/20/2019] [Indexed: 01/17/2023] Open
Abstract
STARD3 is a cellular protein that represents an attractive target for cancer therapy, being overexpressed in breast cancer and implied in the development of colorectal, gastric, and prostate cancers. Unfortunately, no STARD3 inhibitor has been identified yet. In this work, an in silico strategy was applied to predict a reliable binding mode of cholesterol into STARD3 and to develop a pharmacophore-based virtual screening protocol that allowed the identification of the first STARD3 inhibitor ever reported. The identified compound VS1 binds STARD3 with micromolar affinity (IC50 = 35 μM) and shows antiproliferative activity in breast (MCF7 and MDA- MB-231) and colon (HCT-116) cancer cell lines in the same concentration range (IC50 = 49.7-105.5 μM). Although VS1 has a moderate potency, we demonstrated that it specifically targets STARD3 in the cells and induces its degradation. Overall, the results confirm the reliability of the computational strategies herein applied and the identification of the first hit compound for the development of novel potent STARD3 inhibitors.
Collapse
Affiliation(s)
| | - Barbara Salis
- Department of Translational Research, Pathology Unit, National Cancer Institute−CRO-IRCSS, 33081 Aviano, Italy
- Doctoral School in Biomolecolar Medicine, University of Trieste, 34127 Trieste, Italy
| | - Stefano Palazzolo
- Department of Translational Research, Pathology Unit, National Cancer Institute−CRO-IRCSS, 33081 Aviano, Italy
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | | | - Rossella Rotondo
- Department of Translational Research, Pathology Unit, National Cancer Institute−CRO-IRCSS, 33081 Aviano, Italy
| | - Isabella Caligiuri
- Department of Translational Research, Pathology Unit, National Cancer Institute−CRO-IRCSS, 33081 Aviano, Italy
| | - Vincenzo Canzonieri
- Department of Translational Research, Pathology Unit, National Cancer Institute−CRO-IRCSS, 33081 Aviano, Italy
| | | | - Flavio Rizzolio
- Department of Translational Research, Pathology Unit, National Cancer Institute−CRO-IRCSS, 33081 Aviano, Italy
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venezia, 30172 Mestre, Italy
| |
Collapse
|
22
|
Granchi C, Lapillo M, Glasmacher S, Bononi G, Licari C, Poli G, el Boustani M, Caligiuri I, Rizzolio F, Gertsch J, Macchia M, Minutolo F, Tuccinardi T, Chicca A. Optimization of a Benzoylpiperidine Class Identifies a Highly Potent and Selective Reversible Monoacylglycerol Lipase (MAGL) Inhibitor. J Med Chem 2019; 62:1932-1958. [DOI: 10.1021/acs.jmedchem.8b01483] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Carlotta Granchi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Margherita Lapillo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sandra Glasmacher
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland
| | - Giulia Bononi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Cristina Licari
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Maguie el Boustani
- Pathology Unit, Department of Molecular Biology and Translational Research, National Cancer Institute and Center for Molecular Biomedicine, 33081 Aviano, Pordenone, Italy
- Doctoral School in Molecular Biomedicine, University of Trieste, 34100 Trieste, Italy
| | - Isabella Caligiuri
- Pathology Unit, Department of Molecular Biology and Translational Research, National Cancer Institute and Center for Molecular Biomedicine, 33081 Aviano, Pordenone, Italy
| | - Flavio Rizzolio
- Pathology Unit, Department of Molecular Biology and Translational Research, National Cancer Institute and Center for Molecular Biomedicine, 33081 Aviano, Pordenone, Italy
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University, 30123 Venezia, Italy
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland
| | - Marco Macchia
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Filippo Minutolo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Andrea Chicca
- Institute of Biochemistry and Molecular Medicine, NCCR TransCure, University of Bern, CH-3012 Bern, Switzerland
| |
Collapse
|
23
|
Guglielmi P, Carradori S, Poli G, Secci D, Cirilli R, Rotondi G, Chimenti P, Petzer A, Petzer JP. Design, Synthesis, Docking Studies and Monoamine Oxidase Inhibition of a Small Library of 1-acetyl- and 1-thiocarbamoyl-3,5-diphenyl-4,5-dihydro-(1H)-pyrazoles. Molecules 2019; 24:molecules24030484. [PMID: 30700029 PMCID: PMC6384781 DOI: 10.3390/molecules24030484] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/24/2019] [Accepted: 01/27/2019] [Indexed: 01/30/2023] Open
Abstract
New N-acetyl/N-thiocarbamoylpyrazoline derivatives were designed and synthesized in high yields to assess their inhibitory activity and selectivity against human monoamine oxidase A and B. The most important chiral compounds were separated into their single enantiomers and tested. The impact of the substituents at N1, C3 and C5 positions as well the influence of the configuration of the C5 on the biological activity were analyzed. Bulky aromatic groups at C5 were not tolerated. p-Prenyloxyaryl moiety at C3 oriented the selectivity toward the B isoform. The results were also corroborated by molecular modelling studies providing new suggestions for the synthesis of privileged structures to serve as lead compounds for the treatment of mood disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Paolo Guglielmi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Simone Carradori
- Department of Pharmacy, "G. D'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy.
| | - Giulio Poli
- Department of Pharmacy, Università di Pisa, via Bonanno 6, 56126 Pisa, Italy.
| | - Daniela Secci
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Roberto Cirilli
- Centro nazionale per il controllo e la valutazione dei farmaci, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Giulia Rotondi
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Paola Chimenti
- Dipartimento di Chimica e Tecnologie del Farmaco, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy.
| | - Anél Petzer
- Pharmaceutical Chemistry, School of Pharmacy and Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2520, South Africa.
| | - Jacobus P Petzer
- Pharmaceutical Chemistry, School of Pharmacy and Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2520, South Africa.
| |
Collapse
|
24
|
Krasavin M, Shetnev A, Sharonova T, Baykov S, Kalinin S, Nocentini A, Sharoyko V, Poli G, Tuccinardi T, Presnukhina S, Tennikova TB, Supuran CT. Continued exploration of 1,2,4-oxadiazole periphery for carbonic anhydrase-targeting primary arene sulfonamides: Discovery of subnanomolar inhibitors of membrane-bound hCA IX isoform that selectively kill cancer cells in hypoxic environment. Eur J Med Chem 2018; 164:92-105. [PMID: 30594030 DOI: 10.1016/j.ejmech.2018.12.049] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/20/2018] [Accepted: 12/20/2018] [Indexed: 01/01/2023]
Abstract
An expanded set of diversely substituted 1,2,4-oxadiazole-containing primary aromatic sulfonamides was synthesized and tested for inhibition of human carbonic anhydrase I, II, IX and XII isoforms. The initial biochemical profiling revealed a significantly more potent inhibition of cancer-related, membrane-bound isoform hCA IX (reaching into submicromolar range), on top of potent inhibition of hCA XII that is another cancer target. The observed structure-activity relationships have been rationalized by molecular modeling. Comparative single-concentration profiling of the carbonic anhydrase inhibitors synthesized for antiproliferative effects against normal (ARPE-19) and cancer (PANC-1) cell lines under chemically induced hypoxia conditions revealed several candidate compounds selectively targeting cancer cells. More in-depth characterization of these leads revealed two structurally related compounds that showed promising selective cytotoxicity against pancreatic cancer (PANC-1) and melanoma (SK-MEL-2) cell lines.
Collapse
Affiliation(s)
- Mikhail Krasavin
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation.
| | - Anton Shetnev
- The Ushinsky Yaroslavl State Pedagogical University, Yaroslavl, 150000, Russian Federation
| | - Tatyana Sharonova
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Sergey Baykov
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Stanislav Kalinin
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Alessio Nocentini
- Neurofarba Department, Universita degli Studi di Firenze, Florence, Italy
| | - Vladimir Sharoyko
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | | | - Sofia Presnukhina
- The Ushinsky Yaroslavl State Pedagogical University, Yaroslavl, 150000, Russian Federation
| | - Tatiana B Tennikova
- Saint Petersburg State University, Saint Petersburg, 199034, Russian Federation
| | - Claudiu T Supuran
- Neurofarba Department, Universita degli Studi di Firenze, Florence, Italy.
| |
Collapse
|
25
|
De Leo M, Huallpa CG, Alvarado B, Granchi C, Poli G, De Tommasi N, Braca A. New diterpenes from Salvia pseudorosmarinus and their activity as inhibitors of monoacylglycerol lipase (MAGL). Fitoterapia 2018; 130:251-258. [DOI: 10.1016/j.fitote.2018.09.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/11/2018] [Accepted: 09/17/2018] [Indexed: 01/10/2023]
|
26
|
Poli G, Jha V, Martinelli A, Supuran CT, Tuccinardi T. Development of a Fingerprint-Based Scoring Function for the Prediction of the Binding Mode of Carbonic Anhydrase II Inhibitors. Int J Mol Sci 2018; 19:ijms19071851. [PMID: 29937490 PMCID: PMC6073570 DOI: 10.3390/ijms19071851] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 06/16/2018] [Accepted: 06/20/2018] [Indexed: 11/16/2022] Open
Abstract
Carbonic anhydrase II (CAII) is a zinc-containing metalloenzyme whose aberrant activity is associated with various diseases such as glaucoma, osteoporosis, and different types of tumors; therefore, the development of CAII inhibitors, which can represent promising therapeutic agents for the treatment of these pathologies, is a current topic in medicinal chemistry. Molecular docking is a commonly used tool in structure-based drug design of enzyme inhibitors. However, there is still a need for improving docking reliability, especially in terms of scoring functions, since the complex pattern of energetic contributions driving ligand–protein binding cannot be properly described by mathematical functions only including approximated energetic terms. Here we report a novel CAII-specific fingerprint-based (IFP) scoring function developed according to the ligand–protein interactions detected in the CAII-inhibitor co-crystal structures of the most potent CAII ligands. Our IFP scoring function outperformed the ability of Autodock4 scoring function to identify native-like docking poses of CAII inhibitors and thus allowed a considerable improvement of docking reliability. Moreover, the ligand–protein interaction fingerprints showed a useful application in the binding mode analysis of structurally diverse CAII ligands.
Collapse
Affiliation(s)
- Giulio Poli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy.
| | - Vibhu Jha
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy.
| | | | - Claudiu T Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Sesto Fiorentino, 50019 Florence, Italy.
| | | |
Collapse
|
27
|
Anticancer Activity of Euplotin C, Isolated from the Marine Ciliate Euplotes crassus, Against Human Melanoma Cells. Mar Drugs 2018; 16:md16050166. [PMID: 29772645 PMCID: PMC5983297 DOI: 10.3390/md16050166] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 05/08/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Cutaneous melanoma is the most serious type of skin cancer, so new cytotoxic weapons against novel targets in melanoma are of great interest. Euplotin C (EC), a cytotoxic secondary metabolite of the marine ciliate Euplotes crassus, was evaluated in the present study on human cutaneous melanoma cells to explore its anti-melanoma activity and to gain more insight into its mechanism of action. EC exerted a marked cytotoxic effect against three different human melanoma cell lines (A375, 501Mel and MeWo) with a potency about 30-fold higher than that observed in non-cancer cells (HDFa cells). A pro-apoptotic activity and a decrease in melanoma cell migration by EC were also observed. At the molecular level, the inhibition of the Erk and Akt pathways, which control many aspects of melanoma aggressiveness, was shown. EC cytotoxicity was antagonized by dantrolene, a ryanodine receptor (RyR) antagonist, in a concentration-dependent manner. A role of RyR as a direct target of EC was also suggested by molecular modelling studies. In conclusion, our data provide the first evidence of the anti-melanoma activity of EC, suggesting it may be a promising new scaffold for the development of selective activators of RyR to be used for the treatment of melanoma and other cancer types.
Collapse
|
28
|
Dal Piaz F, Vera Saltos MB, Franceschelli S, Forte G, Marzocco S, Tuccinardi T, Poli G, Nejad Ebrahimi S, Hamburger M, De Tommasi N, Braca A. Drug Affinity Responsive Target Stability (DARTS) Identifies Laurifolioside as a New Clathrin Heavy Chain Modulator. JOURNAL OF NATURAL PRODUCTS 2016; 79:2681-2692. [PMID: 27704815 DOI: 10.1021/acs.jnatprod.6b00627] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Five new diterpenes (1-5) and a megastigmane derivative (6) were isolated from the aerial parts of Euphorbia laurifolia, along with several known compounds. Their structures were elucidated by NMR, MS, and ECD and by chemical methods. A chemical proteomics drug affinity responsive target stability (DARTS) approach to investigate the lathyrane diterpene 1, laurifolioside, on its putative cellular target(s) was performed. Clathrin heavy chain 1, a protein mainly involved in selective uptake of proteins, viruses, and other macromolecules at the plasma membrane of cells, was identified as the major interaction partner of compound 1. The modulation of clathrin activity by 1 was studied through microscopy, molecular docking, and molecular dynamics studies, suggesting a new activity of lathyrane diterpenes in the modulation of trafficking pathways.
Collapse
Affiliation(s)
- Fabrizio Dal Piaz
- Dipartimento di Farmacia, Università di Salerno , Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy
| | - Mariela Beatriz Vera Saltos
- Departamento de Ciencias de la Vida, Universidad de las Fuerzas Armadas , ESPE, Avenida General Rumiñahui s/n, Sangolqui, Ecuador
| | - Silvia Franceschelli
- Dipartimento di Farmacia, Università di Salerno , Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy
| | - Giovanni Forte
- Dipartimento di Farmacia, Università di Salerno , Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy
| | - Stefania Marzocco
- Dipartimento di Farmacia, Università di Salerno , Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy
| | - Tiziano Tuccinardi
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 33, 56126 Pisa, Italy
| | - Giulio Poli
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 33, 56126 Pisa, Italy
| | - Samad Nejad Ebrahimi
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University , G. C., Evin, Tehran, Iran
| | - Matthias Hamburger
- Division of Pharmaceutical Biology, Department of Pharmaceutical Sciences, University of Basel , Basel, Switzerland
| | - Nunziatina De Tommasi
- Dipartimento di Farmacia, Università di Salerno , Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy
| | - Alessandra Braca
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 33, 56126 Pisa, Italy
| |
Collapse
|
29
|
Milella L, Milazzo S, De Leo M, Vera Saltos MB, Faraone I, Tuccinardi T, Lapillo M, De Tommasi N, Braca A. α-Glucosidase and α-Amylase Inhibitors from Arcytophyllum thymifolium. JOURNAL OF NATURAL PRODUCTS 2016; 79:2104-2112. [PMID: 27509358 DOI: 10.1021/acs.jnatprod.6b00484] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Three new coumarins (1-3), a prenylated flavanone (4), and two iridoids (5 and 6), together with 17 known secondary metabolites, were isolated from the aerial parts of Arcytophyllum thymifolium. The structures of the new compounds were elucidated on the basis of their spectroscopic data. The potential hypoglycemic properties of the new and known compounds were evaluated by measuring their α-amylase and α-glucosidase inhibitory effects. The iridoid asperulosidic acid (15) and the flavonoid rhamnetin (13) showed the highest activities versus α-amylase (IC50 = 69.4 ± 3.1 and 73.9 ± 5.9 μM, respectively). In turn, the new eriodictyol derivative 4 exhibited the most potent effect as an α-glucosidase inhibitor, with an IC50 value of 28.1 ± 2.6 μM, and was more active than acarbose, used as a positive control. Modeling studies were also performed to suggest the interaction mode of compound 4 in the α-glucosidase enzyme active site.
Collapse
Affiliation(s)
- Luigi Milella
- Dipartimento di Scienze, Università degli Studi della Basilicata , Viale dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Stella Milazzo
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 6 and 33, 56126 Pisa, Italy
| | - Marinella De Leo
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 6 and 33, 56126 Pisa, Italy
| | - Mariela Beatriz Vera Saltos
- Departamento de Ciencias de la Vida, Universidad de las Fuerzas Armadas, ESPE , Avenida General Rumiñahui s/n, Sangolqui, Ecuador
| | - Immacolata Faraone
- Dipartimento di Scienze, Università degli Studi della Basilicata , Viale dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Tiziano Tuccinardi
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 6 and 33, 56126 Pisa, Italy
- Centro Interdipartimentale di Ricerca "Nutraceutica e Alimentazione per la Salute", Università di Pisa , Via del Borghetto 80, 56124 Pisa, Italy
| | - Margherita Lapillo
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 6 and 33, 56126 Pisa, Italy
| | - Nunziatina De Tommasi
- Dipartimento di Farmacia, Università degli Studi di Salerno , Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy
| | - Alessandra Braca
- Dipartimento di Farmacia, Università di Pisa , Via Bonanno 6 and 33, 56126 Pisa, Italy
- Centro Interdipartimentale di Ricerca "Nutraceutica e Alimentazione per la Salute", Università di Pisa , Via del Borghetto 80, 56124 Pisa, Italy
| |
Collapse
|