1
|
Amini M, Barazandeh Tehrani M, Moghimirad P, Boumi S, Ostad S. Design, Synthesis, and Biological Evaluation of New Di-arylimidazole-quinazolinone Hybrid. HETEROCYCLES 2023. [DOI: 10.3987/com-22-14801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
2
|
Zaki I, Moustafa AMY, Beshay BY, Masoud RE, Elbastawesy MAI, Abourehab MAS, Zakaria MY. Design and synthesis of new trimethoxylphenyl-linked combretastatin analogues loaded on diamond nanoparticles as a panel for ameliorated solubility and antiproliferative activity. J Enzyme Inhib Med Chem 2022; 37:2679-2701. [PMID: 36154552 PMCID: PMC9518609 DOI: 10.1080/14756366.2022.2116016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
A new series of vinyl amide-, imidazolone-, and triazinone-linked combretastatin A-4 analogues have been designed and synthesised. These compounds have been evaluated for their cytotoxic activity against MDA-MB-231 breast cancer cells. The triazinone-linked combretastatin analogues (6 and 12) exhibited the most potent cytotoxic activity, in sub-micromolar concentration compared with combretastatin A-4 as a reference standard. The results of β-tubulin polymerisation inhibition assay appear to correlate well with the ability to inhibit β-tubulin polymerisation. Additionally, these compounds were subjected to biological assays relating to cell cycle aspects and apoptosis induction. In addition, the most potent compound 6 was loaded on PEG-PCL modified diamond nanoparticles (PEG-PCL-NDs) and F4 was picked as the optimum formula. F4 exhibited enhanced solubility and release over the drug suspension. In the comparative cytotoxic activity, PEG-PCL modified F4 was capable of diminishing the IC50 by around 2.89 times for nude F4, while by 3.48 times relative to non-formulated compound 6.
Collapse
Affiliation(s)
- Islam Zaki
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Amal M Y Moustafa
- Chemistry Department, Faculty of Science, Port Said University, Port Said, Egypt
| | - Botros Y Beshay
- Pharmaceutical Sciences (Pharmaceutical Chemistry) Department, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria, Egypt
| | - Reham E Masoud
- Clinical Pharmacology Department, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Mohammed A I Elbastawesy
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, Minia University, Minia, Egypt
| | - Mohamed Y Zakaria
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| |
Collapse
|
3
|
Kostin RK, Marshavin AS. Pyrazoles, isoxazoles, and 1,2,3-triazoles as analogs of the natural cytostatic combretastatin A-4: efficient routes of synthesis, tubulin inhibition, and cytotoxicity. Chem Heterocycl Compd (N Y) 2022. [DOI: 10.1007/s10593-021-03025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
4
|
Paidakula S, Nerella S, Kankala S, Kankala RK. Recent Trends in Tubulin-Binding Combretastatin A-4 Analogs for Anticancer Drug Development. Curr Med Chem 2021; 29:3748-3773. [PMID: 34856892 DOI: 10.2174/0929867328666211202101641] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/20/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022]
Abstract
Although significant progress over several decades has been evidenced in cancer therapy, there still remains a need for the development of novel and effective therapeutic strategies to treat several relapsed and intractable cancers. In this regard, tubulin protein has become one of the efficient and major targets for anticancer drug discovery. Considering the antimitotic ability, several tubulin inhibitors have been developed to act against various cancers. Among various tubulin inhibitors available, combretastatin-A4 (CA-4), a naturally occurring lead molecule, offers exceptional cytotoxicity (including the drug-resistant cell lines) and antivascular effects. Although CA-4 offers exceptional therapeutic efficacy, several new advancements have been proposed, such as the structural modification via A and B rings, as well as cis-olefinic bridging, which provide highly efficient analogs with improved tubulin-binding efficiency to meet the anticancer drug development requirements. This review systematically emphasizes the recent trends and latest developments in the anticancer drug design & discovery, using CA-4 analogs as the tubulin inhibiting agents, highlighting their structure-activity relationships (SAR) and resultant pharmacological efficacies.
Collapse
Affiliation(s)
- Suresh Paidakula
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | - Srinivas Nerella
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | - Shravankumar Kankala
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | | |
Collapse
|
5
|
Sameem B, Moghadam ES, Darabi M, Shahsavari Z, Amini M. Triarylpyrazole Derivatives as Potent Cytotoxic Agents; Synthesis and Bioactivity Evaluation "Pyrazole Derivatives as Anticancer Agent". Drug Res (Stuttg) 2021; 71:388-394. [PMID: 34010979 DOI: 10.1055/a-1498-1714] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND During the last recent years, several anti-cancer agents were introduced for the treatment of diverse kinds of cancer. Despite their potential in the treatment of cancer, drug resistance and adverse toxicity such as peripheral neuropathy are some of the negative criteria of anti-cancer agents and for this reason, the design and synthesis of new anti-cancer agents are important. OBJECTIVE Design, synthesis, and anticancer activity evaluation of some pyrazole derivatives. METHODS A series of Target compounds were prepared using multistep synthesis. Their cytotoxic activity against three different human cancer cell lines namely human colon carcinoma cells (HT-29), epithelial carcinoma cells (U-87MG), pancreatic cancerous cells (Panc-1) as well as AGO1522 normal cell line using in vitro 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay was investigated. RESULTS 1,3-Diaryl-5-(3,4,5-trimethoxyphenyl)-4,5-dihydro-1H-pyrazole and 1,3-Diaryl-5-(3,4,5-trimethoxyphenyl)- 1H-pyrazole were synthesized in good yields and their structure and purity were confirmed using 1H-NMR, 13C-NMR, and elemental analysis. Generally, the synthesized scaffolds exhibited good cytotoxicity against cancerous cell lines in comparison to the reference standard, paclitaxel. Compounds 3A: and 3C: , in Annexin V/ PI staining assay, exerted remarkable activity in apoptosis induction in HT-29 cell lines. Both of them also led to cell cycle arrest in the sub-G1 phase which is inconsistent with the results of apoptosis assay. CONCLUSION Concerning obtained results, it is interesting to synthesis more pyrazole derivatives as anticancer agents.
Collapse
Affiliation(s)
- Bilqees Sameem
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Saeedian Moghadam
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.,Department of Chemistry, College of Science, Sultan Qaboos University, Muscat, Sultanate of Oman
| | - Majid Darabi
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Shahsavari
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Rahimzadeh Oskuei S, Mirzaei S, Reza Jafari-Nik M, Hadizadeh F, Eisvand F, Mosaffa F, Ghodsi R. Design, synthesis and biological evaluation of novel imidazole-chalcone derivatives as potential anticancer agents and tubulin polymerization inhibitors. Bioorg Chem 2021; 112:104904. [PMID: 33933802 DOI: 10.1016/j.bioorg.2021.104904] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/03/2021] [Accepted: 04/07/2021] [Indexed: 12/30/2022]
Abstract
Novel imidazole-chalcone derivatives were designed and synthesized as tubulin polymerization inhibitors and anticancer agents. The antiproliferative activity of the imidazole-chalcone was assessed on some human cancer cell lines including A549 (adenocarcinoma human alveolar basal epithelial cells), MCF-7 (human breast cancer cells), MCF-7/MX (mitoxantrone resistant human breast cancer cells), and HEPG2 (human hepatocellular carcinoma cells). Generally, the imidazole-chalcone derivatives exhibited more cytotoxicity on A549 cancer cells in comparison to the other three cell lines, among them compounds 9j' and 9g showed significant cytotoxicity with IC50 values ranging from 7.05 to 63.43 μM against all the four human cancer cells. The flow cytometry analysis of A549 cancer cells treated with 9g and 9j' displayed that these compounds induced cell cycle arrest at the G2/M phase at low concentrations and increased the number of apoptotic cells (cells in subG1 phase) at higher concentrations. They have also inhibited tubulin polymerization similar to combretastatin A-4 (CA-4). Annexin V binding staining assay in A549 cancer cells revealed that compound 9j' induced apoptosis (early and late). Finally, molecular docking studies of 9j' into the colchicine-binding site of tubulin presented the probable interactions of these compounds with tubulin.
Collapse
Affiliation(s)
- Sara Rahimzadeh Oskuei
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Salimeh Mirzaei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Reza Jafari-Nik
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzin Hadizadeh
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farhad Eisvand
- Department of Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Razieh Ghodsi
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
7
|
Boumi S, Moghimirad J, Amanlou M, Ostad SN, Tavajohi S, Amini M. Synthesis, Evaluation of Biological Activity, Docking and Molecular Dynamic Studies of Pyrimidine Derivatives. LETT ORG CHEM 2021. [DOI: 10.2174/1570178617999200706005824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The microtubule is composed of αβ-tubulin heterodimers and is an attractive target for the
design of anticancer drugs. Over the years, various compounds have been developed and their effect on
tubulin polymerization has been studied. Despite great efforts to make an effective drug, no drug has
been introduced which inhibit colchicine binding site. In the current work, a series of pyrimidine derivatives
were designed and synthesized. Furthermore, their cytotoxic activities were evaluated and molecular
docking studies were performed. Twenty compounds of pyrimidine were synthesized in 2 different
groups. In the first group, 4,6-diaryl pyrimidine was connected to the third aryl group via thiomethylene
spacer. In the second group, this linker was substituted by S-CH2-triazole moiety. The cytotoxic
activity of these compounds was evaluated against 4 different cell lines (HT-29, MCF-7, T47D,
NIH3T3). Compounds 6d, 6m, 6p showed potent cytotoxic activity against MCF7 cancerous cell lines.
Between these compounds, compound 6p did not show cytotoxic activity against NIH- 3T3 (normal
cell) cell line. Docking studies show that these compounds occupy colchicine binding site in tubulin
protein and probably their anticancer mechanism is inhibition of tubulin polymerization. Altogether,
with respect to obtained results, it is attractive and beneficial to further investigation on pyrimidine
scaffold as antimitotic agents. Attention to the selectivity index of 6p on MCF7 cell line could be valuable
in design new chemical agents for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shahin Boumi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran,Iran
| | - Jafar Moghimirad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran,Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran,Iran
| | - Seyed Nasser Ostad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Poisoning Research Center, Tehran University of Medical Sciences, Tehran,Iran
| | - Shohreh Tavajohi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Poisoning Research Center, Tehran University of Medical Sciences, Tehran,Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran,Iran
| |
Collapse
|
8
|
Boumi S, Moghimirad J, Ostad SN, Amanlou M, Tavajohi S, Amini M. Synthesis, Biological Evaluation and Docking Study of New Pyrimidine Compounds as Anticancer Agents. Drug Res (Stuttg) 2020; 71:284-290. [PMID: 33285580 DOI: 10.1055/a-1306-0202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVES The microtubule is composed of αβ tubulin heterodimers and is an attractive target for the design of anticancer drugs. Over the years, various compounds have been developed and their effect on tubulin polymerization has been studied. Despite a great efforts to make an effective drug, no drug has been introduced which inhibit Colchicine binding site. METHODS In the current work a series of pyrimidine derivatives were designed and synthesized. Furthermore their cytotoxic activities were evaluated and molecular docking studies were performed. Twelve compounds of pyrimidine were synthesized in 3 different groups. In the first group, 4,6-diaryl pyrimidine was connected to the third aryl group via thio-methylene spacer. In the second group, this linker was substituted by sulfoxide-methylene moiety and in the third group sulfone-methylene group was used as spacer. RESULTS The cytotoxic activity of these compounds were evaluated against 3 different cancerous cell lines (HT-29, MCF-7, T47D) as well as normal cell line (NIH3T3). Compounds in group 2 showed the best cytotoxicity and compound 7D: showed the most potent cytotoxic activity against all cell lines. Molecular modelling studies revealed that compound 7D: could strongly bind to the colchicine binding site of tubulin. CONCLUSION Altogether, with respect to obtained results, it is attractive and beneficial to further investigation on pyrimidine scaffold as antimitotic agents.
Collapse
Affiliation(s)
- Shahin Boumi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Moghimirad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Nasser Ostad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Shohreh Tavajohi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Poisoning Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Moghadam ES, Saravani F, Hamel E, Shahsavari Z, Alipour M, Hosseinkhani S, Ostad S, Amini M. Design, Synthesis and In Vitro Anti-Cancer Evaluation of Novel Derivatives of 2-(2-Methyl-1,5-diaryl-1H-pyrrol-3-yl)-2-oxo-N-(pyridin-3- yl)acetamide. Med Chem 2020; 16:340-349. [PMID: 31032753 DOI: 10.2174/1573406415666190425153717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Several anti-tubulin agents were introduced for the cancer treatment so far. Despite successes in the treatment of cancer, these agents cause toxic side effects, including peripheral neuropathy. Comparing anti-tubulin agents, indibulin seemed to cause minimal peripheral neuropathy, but its poor aqueous solubility and other potential clinical problems have led to its remaining in a preclinical stage. METHODS Herein, indibulin analogues were synthesized and evaluated for their in vitro anti-cancer activity using MTT assay (on the MCF-7, T47-D, MDA-MB231 and NIH-3T3 cell lines), annexin V/PI staining assay, cell cycle analysis, anti-tubulin assay and caspase 3/7 activation assay. RESULTS One of the compounds, 4a, showed good anti-proliferative activity against MCF-7 cells (IC50: 7.5 μM) and low toxicity on a normal cell line (IC50 > 100 μM). All of the tested compounds showed lower cytotoxicity on normal cell line in comparison to reference compound, indibulin. In the annexin V/PI staining assay, induction of apoptosis in the MCF-7 cell line was observed. Cell cycle analysis illustrated an increasing proportion of cells in the sub-G-1 phase, consistent with an increasing proportion of apoptotic cells. No increase in G2/M cells was observed, consistent with the absence of anti-tubulin activity. A caspase 3/7 assay protocol showed that apoptosis induction by more potent compounds was due to activation of caspase 3. CONCLUSION Newly synthesized compounds exerted acceptable anticancer activity and further investigation of current scaffold would be beneficial.
Collapse
Affiliation(s)
- Ebrahim S Moghadam
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Farhad Saravani
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, United States
| | - Zahra Shahsavari
- Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Alipour
- Department of Nano Biotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115, Iran.,Department of Advanced Medical Sciences and Technologies, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran.,Research Center for Noncommunicable Diseases, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Saman Hosseinkhani
- Department of Nano Biotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115, Iran.,Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115, Iran
| | - Seyednasser Ostad
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| |
Collapse
|
10
|
Exploring Diverse-Ring Analogues on Combretastatin A4 (CA-4) Olefin as Microtubule-Targeting Agents. Int J Mol Sci 2020; 21:ijms21051817. [PMID: 32155790 PMCID: PMC7084768 DOI: 10.3390/ijms21051817] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/17/2022] Open
Abstract
Combretastatin-4 (CA-4) as a tubulin polymerization inhibitor draws extensive attentions. However, due to its weak stability of cis-olefin and poor metabolic stability, structure modifications on cis-configuration are being performed. In this work, we constructed a series of novel CA-4 analogues with linkers on olefin containing diphenylethanone, cis-locked dihydrofuran, α-substituted diphenylethanone, cyclobutane and cyclohexane on its cis-olefin. Cytotoxic activity of all analogues was measured by an SRB assay. Among them, compound 6b, a by-product in the preparation of diphenylethanone analogues, was found to be the most potent cytotoxic agents against HepG2 cells with IC50 values of less than 0.5 μM. The two isomers of 6b induced cellular apoptosis tested by Annexin V-FITC and propidium iodide (PI) double staining, arrested cells in the G2/M phase by PI staining analysis, and disrupted microtubule network by immunohistochemistry study in HepG2 cells. Moreover, 6b-(E) displayed a dose-dependent inhibition effect for tubulin assembly in in vitro tubulin polymerization assay. In addition, molecular docking studies showed that two isomers of 6b could bind efficiently at colchicine binding site of tubulin similar to CA-4.
Collapse
|
11
|
Nourmahammadi J, Moghadam ES, Shahsavari Z, Amini M. Design, Synthesis and Biological Evaluation of Novel Diaryl Pyrazole Derivatives as Anticancer Agents. LETT ORG CHEM 2020. [DOI: 10.2174/1570178616666190514090158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cancer is one of the major causes of mortality all around the world. Globally, nearly 1 in 6
deaths is due to cancer. Researchers are trying to synthesize new anticancer agents. Previous studies
demonstrated that some pyrazole derivatives could be considered as potential anticancer agents. Herein,
ten novel derivatives of 1,5-diarylpyrazole were synthesized in four step reactions and cytotoxic activity
was investigated by MTT cell viability assay. All of the compounds were characterized by 1H
NMR and 13C NMR and their purity was confirmed by elemental analysis. The cytotoxicity was determined
against three cancerous cell lines (HT-29, U87MG and MDA-MB 468) and AGO1522 as a
normal cell line. Compound 5a showed the best cytotoxic activity on cancerous cell lines in comparison
to paclitaxel. Annexin V/ PI staining assay also showed that compounds 5a and 5i would lead to
significant apoptosis induction in MDA-MB 486 cell line.
Collapse
Affiliation(s)
- Jalal Nourmahammadi
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Ebrahim Saeedian Moghadam
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Zahra Shahsavari
- Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| |
Collapse
|
12
|
Saravani F, Moghadam ES, Salehabadi H, Ostad S, Hamedani MP, Amanlou M, Faramarzi MA, Amini M. Synthesis, Anti-proliferative Evaluation, and Molecular Docking Studies of 3-(alkylthio)-5,6-diaryl-1,2,4-triazines as Tubulin Polymerization Inhibitors. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666180727114216] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background:
The role of microtubules in cell division and signaling, intercellular transport,
and mitosis has been well known. Hence, they have been targeted for several anti-cancer drugs.
Methods:
A series of 3-(alkylthio)-5,6-diphenyl-1,2,4-triazines were prepared and evaluated for
their cytotoxic activities in vitro against three human cancer cell lines; human colon carcinoma cells
HT-29, human breast adenocarcinoma cell line MCF-7, human Caucasian gastric adenocarcinoma
cell line AGS as well as fibroblast cell line NIH-3T3 by MTT assay. Docking simulation was performed
to insert these compounds into the crystal structure of tubulin at the colchicine binding site
to determine a probable binding model. Compound 5d as the most active compound was selected
for studying of microtubule disruption.
Results:
Compound 5d showed potent cytotoxic activity against all cell lines. The molecular modeling
study revealed that some derivatives of triazine strongly bind to colchicine binding site. The
tubulin polymerization assay kit showed that the cytotoxic activity of 5d may be related to inhibition
of tubulin polymerization.
Conclusion:
The cytotoxicity and molecular modeling study of the synthesized compounds with
their inhibition activity in tubulin polymerization demonstrate the potential of triazine derivatives
for development of new anti-cancer agents.
Collapse
Affiliation(s)
- Farhad Saravani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design & Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ebrahim Saeedian Moghadam
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design & Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hafezeh Salehabadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design & Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyednasser Ostad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Pirali Hamedani
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design & Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design & Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy and Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Drug Design & Development Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Rezaei Z, Mahdi Didehvar M, Mahdavi M, Azizian H, Hamedifar H, Mohammed EHM, Ostad S, Amini M. Anticancer properties of N-alkyl-2, 4-diphenylimidazo [1, 2-a] quinoxalin-1-amine derivatives; kinase inhibitors. Bioorg Chem 2019; 90:103055. [PMID: 31220669 DOI: 10.1016/j.bioorg.2019.103055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 03/27/2019] [Accepted: 06/06/2019] [Indexed: 12/24/2022]
Abstract
Structure activity correlation revealed that the quinoxaline ring is a satisfactory backbone for anticancer activity and a specific functional group at position 1 and 2 can improve the activity. In this basis, besides quinoxaline, imidazoles as potential anticancer agents were used as a supplementary agents for cancer treatment. In this paper, a new series of N-alkyl-2, 4-diphenylimidazo [1, 2-a] quinoxalin-1-amine derivatives were synthesized in a simple and efficient step. The products are fully characterized by 1H NMR, 13C NMR, FT-IR, HRMS, and CHN elemental analysis. Several starting materials with different functionalities have been used for the synthesis of the final products with high isolated yields. The biological activities of the synthesized compounds were evaluated in kinase inhibition and cytotoxic activity in several cancerous cell lines. All compounds (6) were evaluated for inhibition of the cell proliferation using 4 cancerous cell lines. Five of the more active compounds were studied for determination of IC50%. Compounds 6(32-34) showed good activity on some of cancerous cell lines. The results showed that compound 6-32 has the highest biological activity (IC50% 9.77 for K562 cell line). An IC50% value of 15.84 µM was observed for 6-34. Furthermore 6-34 exhibited inhibition of ABL1 and c-Src kinases with an IC50% value of 5.25 µM and 3.94 µM respectively. Docking simulation was performed to position active synthesized compounds 6-32, 6-33, and 6-34 over the ABL1 active site in two different wild-type (DFG-in and DFG-out motif conformer) and T315I mutant to determine the probable binding orientation, conformation and mode of interaction. According to docking study, the docked location in wild type forms is similar and can be found near the P-loop region while in the case of T315I mutant form, the compounds have a distinct docked location which is close to the αC helix and activation loop. Also, it concluded the role of R1 substituent on phenyl ring produced higher interaction energy. Additionally, the detailed inter-molecular energy and types of non-bonding interaction of these compounds over the wild-type and mutant form of ABL1.
Collapse
Affiliation(s)
- Zahra Rezaei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, 14176 Tehran, Iran
| | - Mir Mahdi Didehvar
- School of Chemistry, University College of Science, University of Tehran, PO Box 14155-6455, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Azizian
- Department of Medicinal Chemistry, School of Pharmacy-International Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Eman H M Mohammed
- Department of Chemistry, Faculty of Sciences, Menoufia University, Shebin EI-Koam, Egypt
| | - Sayednaser Ostad
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran, Iran.
| |
Collapse
|
14
|
Moghadam ES, Saravani F, Ostad S, Tavajohi S, Hamedani MP, Amini M. Design, Synthesis and Anticancer Evaluation of Novel Series of Indibulin Analogues. Med Chem 2019; 15:231-239. [DOI: 10.2174/1573406414666181015145945] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 08/13/2018] [Accepted: 09/30/2018] [Indexed: 11/22/2022]
Abstract
Background:
Cancer is an important cause of human death worldwide. During the last
decades, many anticancer agents with anti-tubulin mechanism have been synthesized or extracted
from nature and some of them also entered clinical use. Indibulin is one of the most potent tubulin
polymerization inhibitors with minimal peripheral neuropathy, which is a big problem by some of
the antimitotic agents such as taxanes and vinka alkaloids. With respect to this giant benefit, herein
we decided to design and synthesize novel indibulin related compounds and investigate their
anticancer activity against HT-29, Caco-2 and T47-D cancerous cell lines as well as NIH-T3T as
normal cell line.
Objective:
The aim of this study was to synthesize new anti-cancer agents and evaluates their cytotoxic
activity on diverse cancerous and normal cell lines.
Method:
Target compounds were synthesized in multistep reaction and cytotoxic activity was
investigated by MTT cell viability assay.
Results:
Herein, nine novel target compounds were synthesized in moderate to good yield. Some
of the compounds exerted good cytotoxic activity against cancerous cell lines. Annexin V/PI staining
showed that compound 4g could induce apoptosis and necrosis in HT-29 cell line.
Conclusion:
It is valuable to do further investigation on compound 4g which showed the highest
activity against HT-29 and Caco-2 (IC50 values are 6.9 and 7 &µM respectively). Also, synthesis of
new derivatives of current synthesized compounds is suggested.
Collapse
Affiliation(s)
- Ebrahim S. Moghadam
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development, Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Farhad Saravani
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development, Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Seyednasser Ostad
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Shohreh Tavajohi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Morteza P. Hamedani
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development, Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development, Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| |
Collapse
|
15
|
Synthesis and anti-breast cancer activity of novel indibulin related diarylpyrrole derivatives. ACTA ACUST UNITED AC 2019; 27:179-189. [PMID: 30891679 DOI: 10.1007/s40199-019-00260-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/06/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND During recent years, a number of anti-tubulin agents were introduced for treatment of diverse types of cancer. Despite their potential in the treatment of cancer, drug resistance and adverse toxicity, such as peripheral neuropathy, are some of the negative effects of anti-tubulin agents. Among anti-tubulin agents, indibulin was found to cause minimal peripheral neuropathy. Thus far, however, indibulin has not entered clinical usage, caused in part by its poor aqueous solubility and other developmental problems in preclinical evaluation. OBJECTIVES With respect to need for finding potent and safe anticancer agents, in our current research work, we synthesized several indibulin-related diarylpyrrole derivatives and investigated their anti-cancer activity. METHODS Cell cultur studies were perfomred using the MTT cell viability assay on the breast cancer cell lines MCF-7, T47-D, and MDA-MB231 and also NIH-3 T3 cells as representative of a normal cell line. The activity of some of the synthesized compounds for tubulin interaction was studied using colchicine binding and tubulin polymerization assays. The annexin V-FITC/PI method and flow cytometric analysis were used for studying apoptosis induction and cell cycle distribution. RESULTS AND CONCLUSION Two of the synthesized compounds, 4f and 4 g, showed high activity on the MDA-MB231 cell line (IC50 = 11.82 and 13.33 μM, (respectively) and low toxicity on the normal fibroblast cells (IC50 > 100 μM). All of the tested compounds were more potent on T47-D cancer cells and less toxic on NIH-3 T3 normal cells in comparison to reference compound, indibulin. The tubulin polymerization inhibition assay and [3H]colchicine binding assay showed that the main mechanism of cell death by the potent synthesized compounds was not related to an interaction with tubulin. In the annexin V/PI staining assay, the induction of apoptosis in the MCF-7 and MDA-MB231 cell lines was observed. Cell cycle analysis illustrated an increased percentage of sub-G-1 cells in the MDA-MB231 cell line as a further indication of cell death through induction of apoptosis. Graphical abstract Novel Indibulin analogous as anti-breast cancer agents.
Collapse
|
16
|
Saeedian Moghadam E, Saravani F, Ostad S, Tavajohi S, Pirali Hamedani M, Amini M. Design, synthesis and cytotoxicity evaluation of indibulin analogs. HETEROCYCL COMMUN 2018. [DOI: 10.1515/hc-2018-0016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractIndibulin is one of the most potent tubulin polymerization inhibitors with minimal peripheral neuropathy. The design and synthesis of new indibulin analogs were carried out in order to investigate their anti-cancer activity. The target compounds 4a–i were synthesized in multistep reactions starting with the related indole derivatives. Compound 4f shows the highest cytotoxic activity on HT-29 and Caco-2 cell lines with the respective half maximal inhibitory concentration (IC50) values of 5.1 μm and 7.3 μm. In the case of the T47-D cell line, compound 4c exerts the best cytotoxic activity with an IC50 value of 11.5 μm. In the cell cycle analysis on HT-29 cells, compound 4f at 5.1 μm showed an increase in the percentage of cells in the sub-G1 phase. Altogether, nine target compounds were synthesized and characterized by infrared spectroscopy (IR), proton nuclear magnetic resonance (1H NMR), carbon-13 nuclear magnetic resonance (13C NMR), mass spectrometry (MS) and elemental analysis. Some of the compounds show good cytotoxic activity against cancerous cell lines.
Collapse
Affiliation(s)
- Ebrahim Saeedian Moghadam
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Farhad Saravani
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Seyednasser Ostad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Shohreh Tavajohi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Morteza Pirali Hamedani
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy and Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran 1417614411, Iran
| |
Collapse
|
17
|
2-[2-Methyl-5-phenyl-1-(3,4,5-trimethoxyphenyl)-1H-pyrrol-3-yl]-2-oxo-N-(pyridin-4-yl) acetamide. MOLBANK 2018. [DOI: 10.3390/m1002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
18
|
Ameri A, Khodarahmi G, Forootanfar H, Hassanzadeh F, Hakimelahi GH. Hybrid Pharmacophore Design, Molecular Docking, Synthesis, and Biological Evaluation of Novel Aldimine-Type Schiff
Base Derivatives as Tubulin Polymerization Inhibitor. Chem Biodivers 2018; 15:e1700518. [DOI: 10.1002/cbdv.201700518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 12/29/2017] [Indexed: 01/11/2023]
Affiliation(s)
- Alieh Ameri
- Department of Medicinal Chemistry; Faculty of Pharmacy; Kerman University of Medical Sciences; Kerman Iran
| | - Ghadamali Khodarahmi
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
| | - Hamid Forootanfar
- Department of Pharmaceutical Biotechnology; Faculty of Pharmacy; Kerman University of Medical Sciences; Kerman Iran
| | - Farshid Hassanzadeh
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
| | - Gholam-Hosein Hakimelahi
- Department of Medicinal Chemistry; Faculty of Pharmacy; Isfahan University of Medical Sciences; Isfahan Iran
- Institute of Chemistry; Academia Sinica; Nankang Taipei Taiwan
| |
Collapse
|
19
|
Rossi R, Lessi M, Manzini C, Bellina F. Synthesis and Biological Profiles of 4,5-, 1,5-, and 1,2-Diaryl-1 H -imidazoles. VICINAL DIARYL SUBSTITUTED HETEROCYCLES 2018:83-160. [DOI: 10.1016/b978-0-08-102237-5.00004-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
20
|
Song MY, Cao CY, He QR, Dong QM, Li D, Tang JJ, Gao JM. Constructing novel dihydrofuran and dihydroisoxazole analogues of isocombretastatin-4 as tubulin polymerization inhibitors through [3+2] reactions. Bioorg Med Chem 2017; 25:5290-5302. [PMID: 28803799 DOI: 10.1016/j.bmc.2017.07.048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/19/2022]
Abstract
[3+2] reactions play a key role in constructing various pharmaceutical moleculars. In this study, using Mn(OAc)3 mediated and 1,3-dipolar [3+2] cyclization reactions, 38 novel dihydrofuran and dihydroisoxazole analogues of isoCA-4 were synthesized as inhibitors of tubulin polymerization. Among them, compound 6g was found to be the most potent cytotoxic agents against PC-3 cells with IC50 value of 0.47μM, and compound 5p exhibted highest activity on HeLa cells with IC50 vaule of 2.32µM. Tubulin polymerization assay revealed that 6g was a dose-dependent and effective inhibitor of tubulin assembly. Immunohistochemistry studies and cell cycle distribution analysis indicated that 6g severely disrupted microtubule network and significantly arrested most cells in the G2/M phase of the cell cycle in PC-3 cells. In addition, molecular docking studies showed that two chiral isomers of 6g can bind efficiently and similarly at colchicine binding site of tubulin.
Collapse
Affiliation(s)
- Ming-Yu Song
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, PR China
| | - Chen-Yu Cao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, PR China
| | - Qiu-Rui He
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, PR China
| | - Qing-Miao Dong
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, PR China
| | - Ding Li
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, PR China
| | - Jiang-Jiang Tang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, PR China.
| | - Jin-Ming Gao
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, PR China.
| |
Collapse
|
21
|
Tarade D, Ma D, Pignanelli C, Mansour F, Simard D, van den Berg S, Gauld J, McNulty J, Pandey S. Structurally simplified biphenyl combretastatin A4 derivatives retain in vitro anti-cancer activity dependent on mitotic arrest. PLoS One 2017; 12:e0171806. [PMID: 28253265 PMCID: PMC5333808 DOI: 10.1371/journal.pone.0171806] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 01/26/2017] [Indexed: 12/31/2022] Open
Abstract
The cis-stilbene, combretastatin A4 (CA4), is a potent microtubule targeting and vascular damaging agent. Despite promising results at the pre-clinical level and extensive clinical evaluation, CA4 has yet to be approved for therapeutic use. One impediment to the development of CA4 is an inherent conformational instability about the ethylene linker, which joins two aromatic rings. We have previously published preliminary data regarding structurally simplified biphenyl derivatives of CA4, lacking an ethylene linker, which retain anti-proliferative and pro-apoptotic activity, albeit at higher doses. Our current study provides a more comprehensive evaluation regarding the anti-proliferative and pro-apoptotic properties of biphenyl CA4 derivatives in both 2D and 3D cancerous and non-cancerous cell models. Computational analysis has revealed that cytotoxicity of CA4 and biphenyl analogues correlates with predicted tubulin affinity. Additional mechanistic evaluation of the biphenyl derivatives found that their anti-cancer activity is dependent on prolonged mitotic arrest, in a similar manner to CA4. Lastly, we have shown that cancer cells deficient in the extrinsic pathway of apoptosis experience delayed cell death following treatment with CA4 or analogues. Biphenyl derivatives of CA4 represent structurally simplified analogues of CA4, which retain a similar mechanism of action. The biphenyl analogues warrant in vivo examination to evaluate their potential as vascular damaging agents.
Collapse
Affiliation(s)
- Daniel Tarade
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Dennis Ma
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Christopher Pignanelli
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Fadi Mansour
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Daniel Simard
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - Sean van den Berg
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - James Gauld
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
| | - James McNulty
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Siyaram Pandey
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario, Canada
- * E-mail:
| |
Collapse
|
22
|
Novel Natural Product- and Privileged Scaffold-Based Tubulin Inhibitors Targeting the Colchicine Binding Site. Molecules 2016; 21:molecules21101375. [PMID: 27754459 PMCID: PMC6273505 DOI: 10.3390/molecules21101375] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022] Open
Abstract
Tubulin inhibitors are effective anticancer agents, however, there are many limitations to the use of available tubulin inhibitors in the clinic, such as multidrug resistance, severe side-effects, and generally poor bioavailability. Thus, there is a constant need to search for novel tubulin inhibitors that can overcome these limitations. Natural product and privileged structures targeting tubulin have promoted the discovery and optimization of tubulin inhibitors. This review will focus on novel tubulin inhibitors derived from natural products and privileged structures targeting the colchicine binding site on tubulin.
Collapse
|
23
|
Kumar GB, Nayak VL, Sayeed IB, Reddy VS, Shaik AB, Mahesh R, Baig MF, Shareef MA, Ravikumar A, Kamal A. Design, synthesis of phenstatin/isocombretastatin-oxindole conjugates as antimitotic agents. Bioorg Med Chem 2016; 24:1729-40. [DOI: 10.1016/j.bmc.2016.02.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/29/2016] [Accepted: 02/29/2016] [Indexed: 01/11/2023]
|
24
|
Mahdavi M, Hariri R, Saeedi M, Foroumadi A, Shafiee A, Akbarzadeh T. Synthesis of new benzo[f]imidazo[1,2-d][1,4]oxazepines: AgNO3-mediated intramolecular hydroamination. Tetrahedron Lett 2015. [DOI: 10.1016/j.tetlet.2015.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
25
|
(±)Methanodibenzodiazocine tethered [C-H]δ+ functional site: Study towards benzoin condensation and Baylis-Hillman reactions. J CHEM SCI 2015. [DOI: 10.1007/s12039-015-0837-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
Heravi MM, Daraie M, Zadsirjan V. Current advances in the synthesis and biological potencies of tri- and tetra-substituted 1H-imidazoles. Mol Divers 2015; 19:577-623. [PMID: 25863807 DOI: 10.1007/s11030-015-9590-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 03/25/2015] [Indexed: 02/05/2023]
Abstract
In this report, we review the current chemistry progress and in particular the synthesis approaches of tri- and tetra-substituted imidazoles.
Collapse
Affiliation(s)
- Majid M Heravi
- Department of Chemistry, School of Science, Alzahra University, Vanak, Tehran, Iran,
| | | | | |
Collapse
|
27
|
Vosooghi M, Amini M. The discovery and development of cyclooxygenase-2 inhibitors as potential anticancer therapies. Expert Opin Drug Discov 2014; 9:255-67. [PMID: 24483845 DOI: 10.1517/17460441.2014.883377] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION In the past, clinical studies had demonstrated that aspirin and NSAIDs reduce the risk of colorectal cancer. After the discovery of selective prostaglandin-endoperoxide synthase 2 (PTGS2) inhibitors, the further beneficial effects of celecoxib and some other related structures (coxibs) have been demonstrated in both in vivo and in vitro studies. AREAS COVERED The authors illustrate the role of prostaglandins following the overexpression of PTGS2 (COX-2) in signaling pathways. The authors elucidate the role of coxibs in cell proliferation, apoptosis, angiogenesis and multi-drug resistance and discuss the molecular mechanisms involved. The authors also present the strong evidence related to the usefulness of coxibs in several cancer cell lines. EXPERT OPINION There have been a number of PTGS2 (COX-2) selective inhibitors suggested as potential anticancer therapies. In recent years, the development of nanotechnology has also had an impact on chemotherapy. Indeed, nanoparticles of cytotoxic drug carriers have demonstrated potential through their accumulation in cancer cells, and targeting these nanoparticles has been under evaluation. This area could be opened up for coxib development as they are potentially important targets in cancer cells. Further research using celecoxib as a co-drug with PTGS2-overexpressed and PTGS2-independent cancer is still needed.
Collapse
Affiliation(s)
- Mohsen Vosooghi
- Tehran University of Medical Sciences, Faculty of Pharmacy, Drug Design & Development Research Center, Department of Medicinal Chemistry , Tehran , Iran
| | | |
Collapse
|
28
|
Synthesis, cytotoxic evaluation and molecular docking study of 2-alkylthio-4-(2,3,4-trimethoxyphenyl)-5-aryl-thiazoles as tubulin polymerization inhibitors. Bioorg Med Chem 2013; 21:7648-54. [DOI: 10.1016/j.bmc.2013.10.030] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 10/21/2013] [Accepted: 10/22/2013] [Indexed: 11/20/2022]
|
29
|
Salehi M, Ostad SN, Riazi GH, Assadieskandar A, Cheraghi-Shavi T, Shafiee A, Amini M. Synthesis, cytotoxic evaluation, and molecular docking study of 4,5-diaryl-thiazole-2-thione analogs of combretastatin A-4 as microtubule-binding agents. Med Chem Res 2013. [DOI: 10.1007/s00044-013-0754-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
Kleyi P, Frost CL, Tshentu ZR, Torto N. Electrospun nylon 6 nanofibers incorporated with 2-substitutedN-alkylimidazoles and their silver(I) complexes for antibacterial applications. J Appl Polym Sci 2013. [DOI: 10.1002/app.39783] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Phumelele Kleyi
- Department of Chemistry; Rhodes University; Grahamstown South Africa
| | - Carminita L. Frost
- Department of Biochemistry and Microbiology; Nelson Mandela Metropolitan University; Port Elizabeth South Africa
| | | | - Nelson Torto
- Department of Chemistry; Rhodes University; Grahamstown South Africa
| |
Collapse
|