1
|
Yin G, Ruan Q, Jiang Y, Feng J, Han P, Wang Q, Li Z, Zhang J. Novel 99mTc-Labeled Mannose Derivative as a Highly Promising Single Photon Emission Computed Tomography Probe for Tumor Imaging. J Med Chem 2024; 67:15796-15806. [PMID: 39058751 DOI: 10.1021/acs.jmedchem.4c01425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
18F-2-fluoro-2-deoxy-d-glucose ([18F]FDG) has been the most used positron emission tomography imaging agent for clinical applications. Single photon emission computed tomography (SPECT) imaging is cheaper and used more widely for diagnostic use, but there is no SPECT tumor imaging agent for clinical applications comparable to [18F]FDG. Mannose is a C2 epimer of glucose and can also be transported into tumor cells via glucose transporters (GLUTs). To develop a novel SPECT tumor imaging agent with satisfactory tumor uptake and tumor/nontarget ratios, here a mannose derivative (CN7DM) was synthesized and radiolabeled with technetium-99m to prepare [99mTc]Tc-CN7DM. The six-coordinated structure of [99mTc]Tc-CN7DM was confirmed by the corresponding rhenium compound (Re-CN7DM). [99mTc]Tc-CN7DM was transported into cancer cells via GLUTs and may be trapped in the cancer cells by electrostatic attraction. The probe exhibited high uptake in tumors and low uptake in nontarget tissues in mice bearing different tumors, indicating that [99mTc]Tc-CN7DM exhibited promising potential for SPECT tumor imaging and warranted further clinical investigation.
Collapse
Affiliation(s)
- Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, School of Physics and Astronomy, Beijing Normal University, Beijing 100875, China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junhong Feng
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
- Department of Nuclear Technology and Application, China Institute of Atomic Energy, Beijing 102413, China
| | - Peiwen Han
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Qianna Wang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Zuojie Li
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of Ministry of Education, NMPA (National Medical Products Administration) Key Laboratory for Research and Evaluation of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| |
Collapse
|
2
|
Yan W, Zhou Y, Yuan X, Bai P, Tang M, Chen L, Wei H, Yang J. The cytotoxic natural compound erianin binds to colchicine site of β-tubulin and overcomes taxane resistance. Bioorg Chem 2024; 150:107569. [PMID: 38905886 DOI: 10.1016/j.bioorg.2024.107569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
Erianin, a natural compound derived from Dendrobium, has shown significant anticancer properties against a wide range of cancer cells. Despite the identification of multiple mechanisms of action for erianin, none of these mechanisms fully account for its broad-spectrum effect. In this study, we aimed to identify the cellular target and underlying mechanism responsible for the broad-spectrum antitumor effects of erianin. We found that erianin effectively inhibited tubulin polymerization in cancer cells and purified tubulin. Through competition binding assays and X-ray crystallography, it was revealed that erianin bound to the colchicine site of β-tubulin. Importantly, the X-ray crystal structure of the tubulin-erianin complex was solved, providing clear insight into the orientation and position of erianin in the colchicine-binding site. Erianin showed activity against paclitaxel-resistant cells, evidenced by G2/M cell cycle arrest, apoptosis-related PARP and Caspase-3 cleavage, and in vivo xenograft studies. The study concluded that erianin bound reversibly to the colchicine site of β-tubulin, inhibited tubulin polymerization, and displayed anticancer activity against paclitaxel-resistant cells, offering valuable insights for further exploration as potential anticancer agents.
Collapse
Affiliation(s)
- Wei Yan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yongzhao Zhou
- Integrated Care Management Center, West China Hospital, Sichuan University, China.
| | - Xue Yuan
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Peng Bai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Lijuan Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haoche Wei
- Department of General Surgery, Gastric Cancer Center, Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| | - Jianhong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Shi Y, Wei L, Jin F, Wang J, Cao H, Yang Y, Gao L. Colchicine disrupts bile acid metabolic homeostasis by affecting the enterohepatic circulation in mice. J Appl Toxicol 2024; 44:863-873. [PMID: 38311468 DOI: 10.1002/jat.4587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/06/2024]
Abstract
Although the medicinal properties of colchicine (COL) have been widely known for centuries, its toxicity has been the subject of controversy. The narrow therapeutic window causes COL to induce gastrointestinal adverse effects even when taken at recommended doses, mainly manifested as nausea, vomiting, and diarrhea. However, the mechanism of COL-induced gastrointestinal toxic reactions remains obscure. In the present study, the mice were dosed with COL (2.5 mg/kg b.w./day) for a week to explore the effect of COL on bile acid metabolism and the mechanism of COL-induced diarrhea. The results showed that COL treatment affected liver biochemistry in mice, resulting in a significant down-regulation of the mRNA expression levels of bile acid biosynthesis regulators Cyp7a1, Cyp8b1, Cyp7b1, and Cyp27a1 in liver tissues. The mRNA expression levels of bile acid transporters Ntcp, Oatp1, Mrp2, Ibabp, Mrp3, Osta, and Ostb in liver and ileum tissues were also significantly down-regulated. In addition, COL treatment significantly inhibited the mRNA expression levels of Fxr and its downstream target genes Shp, Lrh1, and Fgf15 in liver and ileum tissues, affecting the feedback regulation of bile acid biosynthesis. More importantly, the inhibition of COL on bile acid transporters in ileal and hepatic tissues affected bile acid recycling in the ileum as well as their reuptake in the liver, leading to a significantly increased accumulation of bile acids in the colon, which may be an important cause of diarrhea. In conclusion, our study revealed that COL treatment affected bile acid biosynthesis and enterohepatic circulation, thereby disrupting bile acid metabolic homeostasis in mice.
Collapse
Affiliation(s)
- Yongpeng Shi
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Li Wei
- Gansu Provincial Clinical Research Center for Laboratory Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Fang Jin
- Gansu Provincial Clinical Research Center for Laboratory Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Ji Wang
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Hanwen Cao
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Ying Yang
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lan Gao
- School of Life Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
4
|
Ren W, Deng Y, Ward JD, Vairin R, Bai R, Wanniarachchi HI, Hamal KB, Tankoano PE, Tamminga CS, Bueno LMA, Hamel E, Mason RP, Trawick ML, Pinney KG. Synthesis and biological evaluation of structurally diverse 6-aryl-3-aroyl-indole analogues as inhibitors of tubulin polymerization. Eur J Med Chem 2024; 263:115794. [PMID: 37984295 PMCID: PMC11019941 DOI: 10.1016/j.ejmech.2023.115794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 11/22/2023]
Abstract
The synthesis and evaluation of small-molecule inhibitors of tubulin polymerization remains a promising approach for the development of new therapeutic agents for cancer treatment. The natural products colchicine and combretastatin A-4 (CA4) inspired significant drug discovery campaigns targeting the colchicine site located on the beta-subunit of the tubulin heterodimer, but so far these efforts have not yielded an approved drug for cancer treatment in human patients. Interest in the colchicine site was enhanced by the discovery that a subset of colchicine site agents demonstrated dual functionality as both potent antiproliferative agents and effective vascular disrupting agents (VDAs). Our previous studies led to the discovery and development of a 2-aryl-3-aroyl-indole analogue (OXi8006) that inhibited tubulin polymerization and demonstrated low nM IC50 values against a variety of human cancer cell lines. A water-soluble phosphate prodrug salt (OXi8007), synthesized from OXi8006, displayed promising vascular disrupting activity in mouse models of cancer. To further extend structure-activity relationship correlations, a series of 6-aryl-3-aroyl-indole analogues was synthesized and evaluated for their inhibition of tubulin polymerization and cytotoxicity against human cancer cell lines. Several structurally diverse molecules in this small library were strong inhibitors of tubulin polymerization and of MCF-7 and MDA-MB-231 human breast cancer cells. One of the most promising analogues (KGP591) caused significant G2/M arrest of MDA-MB-231 cells, disrupted microtubule structure and cell morphology in MDA-MB-231 cells, and demonstrated significant inhibition of MDA-MB-231 cell migration in a wound healing (scratch) assay. A phosphate prodrug salt, KGP618, synthesized from its parent phenolic precursor, KGP591, demonstrated significant reduction in bioluminescence signal when evaluated in vivo against an orthotopic model of kidney cancer (RENCA-luc) in BALB/c mice, indicative of VDA efficacy. The most active compounds from this series offer promise as anticancer therapeutic agents.
Collapse
Affiliation(s)
- Wen Ren
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Yuling Deng
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Jacob D Ward
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Rebecca Vairin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD, 21702, United States.
| | - Hashini I Wanniarachchi
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Khagendra B Hamal
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Pouguiniseli E Tankoano
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Caleb S Tamminga
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Lorena M A Bueno
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD, 21702, United States.
| | - Ralph P Mason
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| |
Collapse
|
5
|
Ren W, Vairin R, Ward JD, Francis R, VanNatta J, Bai R, Tankoano PE, Deng Y, Hamel E, Trawick ML, Pinney KG. Structure Guided Design, Synthesis, and Biological Evaluation of Oxetane-Containing Indole Analogues. Bioorg Med Chem 2023; 92:117400. [PMID: 37556912 PMCID: PMC10848874 DOI: 10.1016/j.bmc.2023.117400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 08/11/2023]
Abstract
The oxetane functional group offers a variety of potential advantages when incorporated within appropriate therapeutic agents as a ketone surrogate. OXi8006, a 2-aryl-3-aroyl-indole analogue, functions as a small-molecule inhibitor of tubulin polymerization that has a dual mechanism of action as both an antiproliferative agent and a tumor-selective vascular disrupting agent. Replacement of the bridging ketone moiety in OXi8006 with an oxetane functional group has expanded structure activity relationship (SAR) knowledge and provided insights regarding oxetane incorporation within this class of molecules. A new synthetic method using an oxetane-containing tertiary alcohol subjected to Lewis acid catalyzed conditions led to successful Friedel-Crafts alkylation and yielded fourteen new oxetane-containing indole-based molecules. This synthetic approach represents the first method to successfully install an oxetane ring at the 3-position of a 2-aryl-indole system. Several analogues showed potent cytotoxicity (micromolar GI50 values) against human breast cancer cell lines (MCF-7 and MDA-MB-231) and a pancreatic cancer cell line (PANC-1), although they proved to be ineffective as inhibitors of tubulin polymerization. Molecular docking studies comparing colchicine with the OXi8006-oxetane analogue 5m provided a rationale for the differential interaction of these molecules with the colchicine site on the tubulin heterodimer.
Collapse
Affiliation(s)
- Wen Ren
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Rebecca Vairin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Jacob D Ward
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ricardo Francis
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Jenny VanNatta
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Pouguiniseli E Tankoano
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Yuling Deng
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD 21702, United States
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX 76798-7348, United States.
| |
Collapse
|
6
|
Hao SH, Ye LY, Yang C. The landscape of pathophysiology guided therapeutic strategies for gout treatment. Expert Opin Pharmacother 2023; 24:1993-2003. [PMID: 38037803 DOI: 10.1080/14656566.2023.2291073] [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: 10/12/2023] [Accepted: 11/30/2023] [Indexed: 12/02/2023]
Abstract
INTRODUCTION Gout is a common autoinflammatory disease caused by hyperuricemia with acute and/or chronic inflammation as well as tissue damage. Currently, urate-lowering therapy (ULT) and anti-inflammatory therapy are used as first-line strategies for gout treatment. However, traditional drugs for gout treatment exhibit some unexpected side effects and are not suitable for certain patients due to their comorbidity with other chronic disease. AREAS COVERED In this review, we described the pathophysiology of hyperuricemia and monosodium urate (MSU) crystal induced inflammatory response during gout development in depth and comprehensively summarized the advances in the investigation of promising ULT drugs as well as anti-inflammatory drugs that might be safer and more effective for gout treatment. EXPERT OPINION New drugs that are developed based on these molecular mechanisms exhibited great efficacy on reduction of disease burden both in vitro and in vivo, implying their potential for clinical application. Moreover, hyperthermia also showed regulation effect on MSU crystals formation and the signaling pathways involved in inflammation.
Collapse
Affiliation(s)
- Sai Heng Hao
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lin Yan Ye
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chang Yang
- Department of Hematology of First Affiliated Hospital, and Department of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Montecinos F, Sackett DL. Structural Changes, Biological Consequences, and Repurposing of Colchicine Site Ligands. Biomolecules 2023; 13:biom13050834. [PMID: 37238704 DOI: 10.3390/biom13050834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/06/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Microtubule-targeting agents (MTAs) bind to one of several distinct sites in the tubulin dimer, the subunit of microtubules. The binding affinities of MTAs may vary by several orders of magnitude, even for MTAs that specifically bind to a particular site. The first drug binding site discovered in tubulin was the colchicine binding site (CBS), which has been known since the discovery of the tubulin protein. Although highly conserved throughout eukaryotic evolution, tubulins show diversity in their sequences between tubulin orthologs (inter-species sequence differences) and paralogs (intraspecies differences, such as tubulin isotypes). The CBS is promiscuous and binds to a broad range of structurally distinct molecules that can vary in size, shape, and affinity. This site remains a popular target for the development of new drugs to treat human diseases (including cancer) and parasitic infections in plants and animals. Despite the rich knowledge about the diversity of tubulin sequences and the structurally distinct molecules that bind to the CBS, a pattern has yet to be found to predict the affinity of new molecules that bind to the CBS. In this commentary, we briefly discuss the literature evidencing the coexistence of the varying binding affinities for drugs that bind to the CBS of tubulins from different species and within species. We also comment on the structural data that aim to explain the experimental differences observed in colchicine binding to the CBS of β-tubulin class VI (TUBB1) compared to other isotypes.
Collapse
Affiliation(s)
- Felipe Montecinos
- Protein Expression Laboratory, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dan L Sackett
- Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Murelli RP, Berkowitz AJ, Zuschlag DW. Carbocycloaddition Strategies for Troponoid Synthesis. Tetrahedron 2023; 130:133175. [PMID: 36777111 PMCID: PMC9910567 DOI: 10.1016/j.tet.2022.133175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Tropone is the prototypical aromatic 7-membered ring, and can be found in virtually any undergraduate textbook as a key example of non-benzenoid aromaticity. Aside from this important historical role, tropone is also of high interest as a uniquely reactive synthon in complex chemical synthesis as well as a valuable chemotype in drug design. More recently, there has been growing interest in the utility of tropones for catalysis and material science. Thus, synthetic strategies capable of synthesizing functional tropones are key to fully exploiting the potential of this aromatic ring system. Cycloaddition reactions are particularly powerful methods for constructing carbocycles, and these strategies in turn have proven to be powerful for generating troponoids. The following review article provides an overview of strategies for troponoids wherein the 7-membered carbocycle is generated through a cycloaddition reaction. Representative examples of each strategy are also provided.
Collapse
Affiliation(s)
- Ryan P Murelli
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, 10016, United States
| | - Alex J Berkowitz
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, NY, 11210, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, United States
| | - Daniel W Zuschlag
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, 10016, United States
| |
Collapse
|
9
|
Guo K, Ma X, Li J, Zhang C, Wu L. Recent advances in combretastatin A-4 codrugs for cancer therapy. Eur J Med Chem 2022; 241:114660. [PMID: 35964428 DOI: 10.1016/j.ejmech.2022.114660] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
CA4 is a potent microtubule polymerization inhibitor and vascular disrupting agent. However, the in vivo efficiency of CA4 is limited owing to its poor pharmacokinetics resulting from its high lipophilicity and low water solubility. To improve the water solubility, CA4 phosphate (CA4P) has been developed and shows potent antivascular and antitumor effects. CA4P had been evaluated as a vascular disrupting agent in previousc linical trials. However, it had been discontinued due to the lack of a meaningful improvement in progression-free survival and unfavorable partial response data. Codrug is a drug design approach to chemically bind two or more drugs to improve therapeutic efficiency or decrease adverse effects. This review describes the progress made over the last twenty years in developing CA4-based codrugs to improve the therapeutic profile and achieve targeted delivery to cancer tissues. It also discusses the existing problems and the developmental prospects of CA4 codrugs.
Collapse
Affiliation(s)
- Kerong Guo
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xin Ma
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jian Li
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chong Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liqiang Wu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
10
|
Bai R, Smith AB, Pettit GR, Hamel E. The interaction of spongistatin 1 with tubulin. Arch Biochem Biophys 2022; 727:109296. [PMID: 35594923 PMCID: PMC10062379 DOI: 10.1016/j.abb.2022.109296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/27/2022] [Accepted: 05/15/2022] [Indexed: 11/26/2022]
Abstract
A tritiated derivative of the sponge-derived natural product spongistatin 1 was prepared, and its interactions with tubulin were examined. [3H]Spongistatin 1 was found to bind rapidly to tubulin at a single site (the low specific activity of the [3H]spongistatin 1, 0.75 Ci/mmol, prevented our defining an association rate), and the inability of spongistatin 1 to cause an aberrant assembly reaction was confirmed. Spongistatin 1 bound to tubulin very tightly, and we could detect no significant dissociation reaction from tubulin. The tubulin-[3H]spongistatin 1 complex did dissociate in 8 M urea, so there was no evidence for covalent bond formation. Apparent KD values were obtained by Scatchard analysis of binding data and by Hummel-Dreyer chromatography (3.5 and 1.1 μM, respectively). The effects of a large cohort of vinca domain drugs on the binding of [3H]spongistatin 1 to tubulin were evaluated. Compounds that did not cause aberrant assembly reactions (halichondrin B, eribulin, maytansine, and rhizoxin) caused little inhibition of [3H]spongistatin 1 binding. Little inhibition also occurred with the peptides dolastatin 15, its active pentapeptide derivative, vitilevuamide, or diazonamide A, nor with the vinca alkaloid vinblastine. Strong inhibition was observed with dolastatin 10, hemiasterlin, and cryptophycin 1, all of which cause aberrant assembly reactions that might actually mask the spongistatin 1 binding site. Spongistatin 5 was found to be a competitive inhibitor of [3H]spongistatin 1 binding, with an apparent Ki of 2.2 μM. We propose that the strong picomolar cytotoxicity of spongistatin 1 probably derives from its extremely tight binding to tubulin.
Collapse
Affiliation(s)
- Ruoli Bai
- Molecular Pharmacology Branch (RB, EH), 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, USA
| | - Amos B Smith
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - George R Pettit
- Laboratory for Discovery of Anti-Cancer and Anti-Infective Drugs, School of Molecular Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Ernest Hamel
- Molecular Pharmacology Branch (RB, EH), 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, USA.
| |
Collapse
|
11
|
Ma Z, Chen J, Jin K, Chen X. Colchicine and coronary heart disease risks: A meta-analysis of randomized controlled clinical trials. Front Cardiovasc Med 2022; 9:947959. [PMID: 36176989 PMCID: PMC9512890 DOI: 10.3389/fcvm.2022.947959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Background Several trials have considered the safety and clinical benefits of colchicine as a treatment option for secondary prevention in patients with coronary atherosclerotic heart disease (CAD), but its safety and clinical benefits remain controversial. The purpose of this study was to explore the clinical benefits of colchicine, focusing on certain subgroups of patients. Methods Randomized controlled trials (RCTs) of colchicine in subjects with acute or chronic CAD compared with controls were included to assess all-cause mortality, non-cardiovascular mortality, gastrointestinal adverse effects, diarrhea, MACE, cardiovascular mortality, MI, stroke, and revascularization. We analyzed the association of cardiovascular, mortality, and gastrointestinal risk with colchicine in all subjects. We also focused on the cardiovascular risk of colchicine in subgroups with different drug doses, different treatment durations, age, gender, and associated comorbidities. Results This meta-analysis included 15 clinical RCTs, including 13,539 subjects. Colchicine reduced the risk of MACE (RR: 0.65; 95% CI: 0.38–0.77, p for heterogeneity < 0.01; I2 = 70%; p < 0.01), stroke (RR: 0.48; 95% CI: 0.30–0.76; p heterogeneity = 0.52; I2 = 0%; p < 0.01), MI by 40% (RR: 0.60; 95% CI: 0.43–0.83; p for heterogeneity = 0.01; I2 = 59%; p < 0.01) and risk of revascularization (RR: 0.68; 95% CI: 0.56–0.83; p for heterogeneity = 0.17; I2 = 40%; p < 0.01), but had no significant effect on risk of cardiovascular death and risk of all-cause mortality. In addition, colchicine increased the risk of gastrointestinal side effects and diarrhea. In a subgroup analysis, low-dose colchicine and treatment duration > 1 month reduced the risk of MACE, MI, stroke, and revascularization. Also, the cardiovascular benefits of colchicine were observed in subjects up to 65 years of age. The results showed that hypertension and diabetes did not have a specific effect on colchicine and MACE risk. Conclusion Colchicine has a positive effect in reducing the incidence of MACE, MI, stroke, and revascularization, but can increase the risk of gastrointestinal and diarrhea events. Low-dose colchicine significantly reduces the risk of MACE more than high-dose colchicine, and the benefits of long-term treatment are higher than those of short-term treatment. Long-term low-dose colchicine treatment may significantly reduce the risk of cardiovascular events. Furthermore, colchicine significantly reduced the risk of cardiovascular events in patients up to 65 years of age, but it did not appear to reduce cardiovascular risk in patients over 65 years of age or in preoperative PCI patients. Systematic review registration [https://www.crd.york.ac.uk/prospero/], identifier [CDR42022332170].
Collapse
Affiliation(s)
- Zijun Ma
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Jun Chen
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
- Jun Chen,
| | - Kaiqin Jin
- Department of Cardiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin Chen
- Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
- *Correspondence: Xin Chen,
| |
Collapse
|
12
|
Reddel CJ, Pennings GJ, Chen VM, Gnanenthiran S, Kritharides L. Colchicine as a Modulator of Platelet Function: A Systematic Review. Semin Thromb Hemost 2022; 48:552-567. [PMID: 35882248 DOI: 10.1055/s-0042-1749660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
The microtubule inhibitor and anti-inflammatory agent colchicine is used to treat a range of conditions involving inflammasome activation in monocytes and neutrophils, and is now known to prevent coronary and cerebrovascular events. In vitro studies dating back more than 50 years showed a direct effect of colchicine on platelets, but as little contemporary attention has been paid to this area, we have critically reviewed the effects of colchicine on diverse aspects of platelet biology in vitro and in vivo. In this systematic review we searched Embase, Medline, and PubMed for articles testing platelets after incubation with colchicine and/or reporting a clinical effect of colchicine treatment on platelet function, including only papers available in English and excluding reviews and conference abstracts. We identified 98 relevant articles and grouped their findings based on the type of study and platelet function test. In vitro, colchicine inhibits traditional platelet functions, including aggregation, clotting, degranulation, and platelet-derived extracellular vesicle formation, although many of these effects were reported at apparently supraphysiological concentrations. Physiological concentrations of colchicine inhibit collagen- and calcium ionophore-induced platelet aggregation and internal signaling. There have been limited studies of in vivo effects on platelets. The colchicine-platelet interaction has the potential to contribute to colchicine-mediated reduction in cardiovascular events, but there is a pressing need for high quality clinical research in this area.
Collapse
Affiliation(s)
- Caroline J Reddel
- ANZAC Research Institute, University of Sydney, Concord Repatriation General Hospital, Sydney, Australia
| | - Gabrielle J Pennings
- ANZAC Research Institute, University of Sydney, Concord Repatriation General Hospital, Sydney, Australia
| | - Vivien M Chen
- ANZAC Research Institute, University of Sydney, Concord Repatriation General Hospital, Sydney, Australia.,Department of Haematology, Concord Repatriation General Hospital, Sydney, Australia
| | - Sonali Gnanenthiran
- ANZAC Research Institute, University of Sydney, Concord Repatriation General Hospital, Sydney, Australia.,Department of Cardiology, Concord Repatriation General Hospital, Sydney, Australia
| | - Leonard Kritharides
- ANZAC Research Institute, University of Sydney, Concord Repatriation General Hospital, Sydney, Australia.,Department of Cardiology, Concord Repatriation General Hospital, Sydney, Australia
| |
Collapse
|
13
|
Boichuk S, Syuzov K, Bikinieva F, Galembikova A, Zykova S, Gankova K, Igidov S, Igidov N. Computational-Based Discovery of the Anti-Cancer Activities of Pyrrole-Based Compounds Targeting the Colchicine-Binding Site of Tubulin. Molecules 2022; 27:2873. [PMID: 35566235 PMCID: PMC9101527 DOI: 10.3390/molecules27092873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 12/10/2022] Open
Abstract
Despite the tubulin-binding agents (TBAs) that are widely used in the clinic for cancer therapy, tumor resistance to TBAs (both inherited and acquired) significantly impairs their effectiveness, thereby decreasing overall survival (OS) and progression-free survival (PFS) rates, especially for the patients with metastatic, recurrent, and unresectable forms of the disease. Therefore, the development of novel effective drugs interfering with the microtubules' dynamic state remains a big challenge in current oncology. We report here about the novel ethyl 2-amino-1-(furan-2-carboxamido)-5-(2-aryl/tert-butyl-2-oxoethylidene)-4-oxo-4,5-dihydro-1H-pyrrole-3-carboxylates (EAPCs) exhibiting potent anti-cancer activities against the breast and lung cancer cell lines in vitro. This was due to their ability to inhibit tubulin polymerization and induce cell cycle arrest in M-phase. As an outcome, the EAPC-treated cancer cells exhibited a significant increase in apoptosis, which was evidenced by the expression of cleaved forms of PARP, caspase-3, and increased numbers of Annexin-V-positive cells. By using the in silico molecular modeling methods (e.g., induced-fit docking, binding metadynamics, and unbiased molecular dynamics), we found that EAPC-67 and -70 preferentially bind to the colchicine-binding site of tubulin. Lastly, we have shown that the EAPCs indicated above and colchicine utilizes a similar molecular mechanism to inhibit tubulin polymerization via targeting the T7 loop in the β-chain of tubulin, thereby preventing the conformational changes in the tubulin dimers required for their polymerization. Collectively, we identified the novel and potent TBAs that bind to the colchicine-binding site and disrupt the microtubule network. As a result of these events, the compounds induced a robust cell cycle arrest in M-phase and exhibited potent pro-apoptotic activities against the epithelial cancer cell lines in vitro.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
- Department of Radiotherapy and Radiology, Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
- Biologically Active Terpenoids Laboratory, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Kirill Syuzov
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
| | - Firuza Bikinieva
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
| | - Svetlana Zykova
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| | - Ksenia Gankova
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| | - Sergei Igidov
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| | - Nazim Igidov
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| |
Collapse
|
14
|
Computational-Based Discovery of the Anti-Cancer Activities of Pyrrole-Based Compounds Targeting the Colchicine-Binding Site of Tubulin. Molecules 2022. [PMID: 35566235 DOI: 10.3390/molecules27092873.(] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Despite the tubulin-binding agents (TBAs) that are widely used in the clinic for cancer therapy, tumor resistance to TBAs (both inherited and acquired) significantly impairs their effectiveness, thereby decreasing overall survival (OS) and progression-free survival (PFS) rates, especially for the patients with metastatic, recurrent, and unresectable forms of the disease. Therefore, the development of novel effective drugs interfering with the microtubules' dynamic state remains a big challenge in current oncology. We report here about the novel ethyl 2-amino-1-(furan-2-carboxamido)-5-(2-aryl/tert-butyl-2-oxoethylidene)-4-oxo-4,5-dihydro-1H-pyrrole-3-carboxylates (EAPCs) exhibiting potent anti-cancer activities against the breast and lung cancer cell lines in vitro. This was due to their ability to inhibit tubulin polymerization and induce cell cycle arrest in M-phase. As an outcome, the EAPC-treated cancer cells exhibited a significant increase in apoptosis, which was evidenced by the expression of cleaved forms of PARP, caspase-3, and increased numbers of Annexin-V-positive cells. By using the in silico molecular modeling methods (e.g., induced-fit docking, binding metadynamics, and unbiased molecular dynamics), we found that EAPC-67 and -70 preferentially bind to the colchicine-binding site of tubulin. Lastly, we have shown that the EAPCs indicated above and colchicine utilizes a similar molecular mechanism to inhibit tubulin polymerization via targeting the T7 loop in the β-chain of tubulin, thereby preventing the conformational changes in the tubulin dimers required for their polymerization. Collectively, we identified the novel and potent TBAs that bind to the colchicine-binding site and disrupt the microtubule network. As a result of these events, the compounds induced a robust cell cycle arrest in M-phase and exhibited potent pro-apoptotic activities against the epithelial cancer cell lines in vitro.
Collapse
|
15
|
Liu Z, Zhang Y, Shen N, Sun J, Tang Z, Chen X. Destruction of tumor vasculature by vascular disrupting agents in overcoming the limitation of EPR effect. Adv Drug Deliv Rev 2022; 183:114138. [PMID: 35143895 DOI: 10.1016/j.addr.2022.114138] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/27/2021] [Accepted: 02/03/2022] [Indexed: 02/08/2023]
Abstract
Nanomedicine greatly improves the efficiency in the delivery of antitumor drugs into the tumor, but insufficient tumoral penetration impairs the therapeutic efficacy of most nanomedicines. Vascular disrupting agent (VDA) nanomedicines are distributed around the tumor vessels due to the low tissue penetration in solid tumors, and the released drugs can selectively destroy immature tumor vessels and block the supply of oxygen and nutrients, leading to the internal necrosis of the tumors. VDAs can also improve the vascular permeability of the tumor, further increasing the extravasation of VDA nanomedicines in the tumor site, markedly reducing the dependence of nanomedicines on the enhanced permeability and retention effect (EPR effect). This review highlights the progress of VDA nanomedicines in recent years and their application in cancer therapy. First, the mechanisms of different VDAs are introduced. Subsequently, different strategies of delivering VDAs are described. Finally, multiple combination strategies with VDA nanomedicines in cancer therapy are described in detail.
Collapse
|
16
|
Nicolaou KC, Yu R, Lu Z, Alvarez FG. Total Synthesis of Gukulenin B via Sequential Tropolone Functionalizations. J Am Chem Soc 2022; 144:5190-5196. [PMID: 35275627 DOI: 10.1021/jacs.2c01305] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The synthesis of functionalized aromatic compounds is a central theme of research for modern organic chemistry. Despite the increasing finesse in the functionalization of five- and six-membered aromatic rings, their seven-membered-ring sibling, tropolone (2-hydroxy-2,4,6-cycloheptatrien-1-one), remains a challenging target for synthetic derivatization. This challenge primarily emanates from the unique structural and chemical properties of tropolonoid compounds, which often lead to unexpected and undesired reaction outcomes under conditions developed for the functionalizations of other aromatic moieties. Herein, we describe the total synthesis of one of the most complex natural tropolonoids, gukulenin B. Our synthetic route features a series of site-selective aromatic C-H bond functionalizations and C-C bond formations, whose reaction conditions are judiciously tuned to allow uncompromised performance on the tropolone nucleus. The flexibility and modularity of our synthesis are expected to facilitate further synthetic and biological studies of the gukulenin family of cytotoxins. In addition, the methods and tactics developed herein for the functionalization of the tropolone moiety could inspire and enable chemists of multiple disciplines to take advantage of this privileged yet underexplored structural motif.
Collapse
Affiliation(s)
- K C Nicolaou
- Department of Chemistry, BioScience Research Collaborative, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Ruocheng Yu
- Department of Chemistry, BioScience Research Collaborative, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Zhaoyong Lu
- Department of Chemistry, BioScience Research Collaborative, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| | - Fernando G Alvarez
- Department of Chemistry, BioScience Research Collaborative, Rice University, 6100 Main Street, Houston, Texas 77005, United States
| |
Collapse
|
17
|
Goulart HA, Bartz RH, Peglow TJ, Barcellos AM, Cervo R, Cargnelutti R, Jacob RG, Lenardão EJ, Perin G. Synthesis of Seleno-Dibenzocycloheptenones/Spiro[5.5]Trienones by Radical Cyclization of Biaryl Ynones. J Org Chem 2022; 87:4273-4283. [PMID: 35245049 DOI: 10.1021/acs.joc.1c03112] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We report herein an alternative method for the synthesis of seleno-dibenzocycloheptenones and seleno-spiro[5.5]trienones through the radical cyclization of biaryl ynones in the presence of diorganyl diselenides, using Oxone as a green oxidizing agent. The reactions were conducted using acetonitrile as the solvent in a sealed tube at 100 °C. The protocol is operationally simple and scalable, exhibits high regioselectivity, and allows the synthesis of 24 dibenzocycloheptenones/spiro[5.5]trienones in yields of up to 99%, 17 of which are unpublished compounds. Additionally, synthetic transformations of the prepared compounds, such as oxidation and reduction reactions, are demonstrated.
Collapse
Affiliation(s)
- Helen A Goulart
- Laboratório de Síntese Orgânica Limpa - LASOL, CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Ricardo H Bartz
- Laboratório de Síntese Orgânica Limpa - LASOL, CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Thiago J Peglow
- Laboratório de Síntese Orgânica Limpa - LASOL, CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Angelita M Barcellos
- Laboratório de Síntese Orgânica Limpa - LASOL, CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Rodrigo Cervo
- Departamento de Química, CCNE, Universidade Federal de Santa Maria - UFSM, 97105-900, Santa Maria, RS, Brazil
| | - Roberta Cargnelutti
- Departamento de Química, CCNE, Universidade Federal de Santa Maria - UFSM, 97105-900, Santa Maria, RS, Brazil
| | - Raquel G Jacob
- Laboratório de Síntese Orgânica Limpa - LASOL, CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Eder J Lenardão
- Laboratório de Síntese Orgânica Limpa - LASOL, CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Gelson Perin
- Laboratório de Síntese Orgânica Limpa - LASOL, CCQFA, Universidade Federal de Pelotas - UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| |
Collapse
|
18
|
Bell CJ, Potts KG, Hitt MM, Pink D, Tuszynski JA, Lewis JD. Novel colchicine derivative CR42-24 demonstrates potent anti-tumor activity in urothelial carcinoma. Cancer Lett 2022; 526:168-179. [PMID: 34838691 DOI: 10.1016/j.canlet.2021.11.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/09/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023]
Abstract
Bladder cancers, and specifically urothelial carcinoma, have few effective treatment options, and tumors typically develop resistance against standard of care chemotherapies leading to significant mortality. The development of alternative therapies with increased selectivity and improved tolerability would significantly impact this patient population. Here, we investigate a novel colchicine derivative, CR42-24, with increased selectivity for the βIII tubulin subtype as a treatment for urothelial carcinoma. βIII tubulin is a promising target due to its low expression in healthy tissues and its clinical association with poor prognosis. This study demonstrated that CR42-24 is selectively cytotoxic to several cancer cell lines at low nanomolar IC50, with high activity in bladder cancer cell lines both in vitro and in vivo. CR42-24 monotherapy in an aggressive urothelial carcinoma xenograft model results in effective control when treated early. We observed significant ablation of large tumors and patient-derived xenografts at low doses with excellent tolerability. CR42-24 was highly synergistic in combination with the standard of care chemotherapies gemcitabine and cisplatin, further increasing its therapeutic potential as a novel treatment for urothelial carcinoma.
Collapse
Affiliation(s)
- Clayton J Bell
- Department of Oncology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Kyle G Potts
- Department Microbiology, Immunology & Infectious Diseases, Alberta Children's Hospital Research Institute, T2N-4N1, Canada
| | - Mary M Hitt
- Department of Oncology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Desmond Pink
- Department of Oncology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Jack A Tuszynski
- Department of Oncology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - John D Lewis
- Department of Oncology, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada.
| |
Collapse
|
19
|
Abstract
Fluorescence spectroscopy is routinely used for the determination of the interaction of a ligand with a protein. The quick detection of the interaction between the ligand and the protein is one of the most significant advantages of fluorescence spectroscopic methods. In this chapter, we have described assays to monitor drug -tubulin interactions using several fluorescence spectroscopic techniques. We have provided detailed protocols for different assays for investigating tubulin-drug interactions with key practical considerations for performing the experiments. We have also discussed how to deduce the binding parameters by fitting the fluorescence change data in different binding isotherms. Further, we have described detailed protocols to monitor the binding site of a ligand on tubulin by competitive inhibition. Though the methods are described for tubulin, these methods can also be used to monitor any drug -protein interactions.
Collapse
Affiliation(s)
- Anuradha Kumari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Dulal Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India.
| |
Collapse
|
20
|
Rozario AM, Duwé S, Elliott C, Hargreaves RB, Moseley GW, Dedecker P, Whelan DR, Bell TDM. Nanoscale characterization of drug-induced microtubule filament dysfunction using super-resolution microscopy. BMC Biol 2021; 19:260. [PMID: 34895240 PMCID: PMC8665533 DOI: 10.1186/s12915-021-01164-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 10/11/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The integrity of microtubule filament networks is essential for the roles in diverse cellular functions, and disruption of its structure or dynamics has been explored as a therapeutic approach to tackle diseases such as cancer. Microtubule-interacting drugs, sometimes referred to as antimitotics, are used in cancer therapy to target and disrupt microtubules. However, due to associated side effects on healthy cells, there is a need to develop safer drug regimens that still retain clinical efficacy. Currently, many questions remain open regarding the extent of effects on cellular physiology of microtubule-interacting drugs at clinically relevant and low doses. Here, we use super-resolution microscopies (single-molecule localization and optical fluctuation based) to reveal the initial microtubule dysfunctions caused by nanomolar concentrations of colcemid. RESULTS We identify previously undetected microtubule (MT) damage caused by clinically relevant doses of colcemid. Short exposure to 30-80 nM colcemid results in aberrant microtubule curvature, with a trend of increased curvature associated to increased doses, and curvatures greater than 2 rad/μm, a value associated with MT breakage. Microtubule fragmentation was detected upon treatment with ≥ 100 nM colcemid. Remarkably, lower doses (< 20 nM after 5 h) led to subtle but significant microtubule architecture remodelling characterized by increased curvature and suppression of microtubule dynamics. CONCLUSIONS Our results support the emerging hypothesis that microtubule-interacting drugs induce non-mitotic effects in cells, and establish a multi-modal imaging assay for detecting and measuring nanoscale microtubule dysfunction. The sub-diffraction visualization of these less severe precursor perturbations compared to the established antimitotic effects of microtubule-interacting drugs offers potential for improved understanding and design of anticancer agents.
Collapse
Affiliation(s)
- Ashley M Rozario
- School of Chemistry, Monash University, Clayton, 3800, Australia
| | - Sam Duwé
- Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Cade Elliott
- School of Chemistry, Monash University, Clayton, 3800, Australia
| | | | - Gregory W Moseley
- Department of Microbiology, Monash Biomedicine Discovery Institute, Clayton, 3800, Australia
| | - Peter Dedecker
- Department of Chemistry, KU Leuven, 3001, Leuven, Belgium
| | - Donna R Whelan
- La Trobe Institute for Molecular Science, La Trobe University, Bendigo, 3552, Australia.
| | - Toby D M Bell
- School of Chemistry, Monash University, Clayton, 3800, Australia.
| |
Collapse
|
21
|
Krishna R, Shivankar B. A review on liquid chromatographic analysis of colchicine in the forensic and medical perspective. TOXICOLOGIE ANALYTIQUE ET CLINIQUE 2021. [DOI: 10.1016/j.toxac.2021.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
22
|
Krzywik J, Nasulewicz-Goldeman A, Mozga W, Wietrzyk J, Huczyński A. Novel Double-Modified Colchicine Derivatives Bearing 1,2,3-Triazole: Design, Synthesis, and Biological Activity Evaluation. ACS OMEGA 2021; 6:26583-26600. [PMID: 34661013 PMCID: PMC8515607 DOI: 10.1021/acsomega.1c03948] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/15/2021] [Indexed: 05/08/2023]
Abstract
A series of 1,4-disubstituted 1,2,3-triazoles having 10-demethoxy-10-N-methylaminocolchicine core were designed and synthesized via the Cu(I)-catalyzed "click" reaction and screened for their in vitro cytotoxicity against four cancer cell lines (A549, MCF-7, LoVo, LoVo/DX) and one noncancerous cell line (BALB/3T3). Indexes of resistance (RI) and selectivity (SI) were also determined to assess the potential of the analogues to break drug resistance of the LoVo/DX cells and to verify their selectivity toward killing cancer cells over normal cells. The compounds with an ester or amide moiety in the fourth position of 1,2,3-triazole of 10-N-methylaminocolchicine turned out to have the greatest therapeutic potential (low IC50 values and favorable SI values), much better than that of unmodified colchicine or doxorubicin and cisplatin. Thus, they make a valuable clue for the further search for a drug having a colchicine scaffold.
Collapse
Affiliation(s)
- Julia Krzywik
- Department
of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
- TriMen
Chemicals, Piłsudskiego
141, 92-318 Łódź, Poland
| | - Anna Nasulewicz-Goldeman
- Hirszfeld
Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wrocław, Poland
| | - Witold Mozga
- TriMen
Chemicals, Piłsudskiego
141, 92-318 Łódź, Poland
| | - Joanna Wietrzyk
- Hirszfeld
Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wrocław, Poland
| | - Adam Huczyński
- Department
of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
- . Tel: +48618291673
| |
Collapse
|
23
|
Shi Y, Cai H, Niu Z, Li J, Pan G, Tian H, Wei L, Chen L, Yang P, Wang J, Cao H, Gao L. Acute oral colchicine caused gastric mucosal injury and disturbance of associated microbiota in mice. Toxicology 2021; 461:152908. [PMID: 34453961 DOI: 10.1016/j.tox.2021.152908] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/22/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022]
Abstract
Colchicine (COL), an ancient and well-known drug, has been used in clinical practice for centuries. On the other hand, COL has also attracted extensive concerns for its potent toxic effects, especially gastrointestinal adverse reactions (nausea, vomiting, and diarrhea) before clinical symptoms relief. In this study, we used a rodent model to study the effects of COL on gastric mucosa and associated microbiota. The mice were exposed to various concentrations of COL (0.1, 0.5, and 2.5 mg kg-1 body weight per day) for 7 days, and the results showed that COL treatment caused severe gastric mucosal damage, accompanied by a significant decrease in gastric mucosal proinflammatory cytokines (IL-1β, IL-6, and TNF-α). The 16S rRNA gene sequencing revealed that COL significantly perturbed the gastric microbiota composition and reduced the gastric microbiota diversity in mice. Also, we identified bacterial biomarkers associated with diarrhea, including phylum Firmicutes, class Bacilli, order Lactobacillales, family Lactobacillaceae, genu Lactobacillus, and genu Blautia, suggesting that COL-triggered adverse reactions are closely related to gastric microbial perturbations. Our findings open new paths for understanding the mechanism of COL-related adverse gastrointestinal reactions, broadening the scientific view on the interaction between drugs and host gastrointestinal microbiota.
Collapse
Affiliation(s)
- Yongpeng Shi
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Hui Cai
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, China
| | - Zhanyu Niu
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Jiande Li
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Gaowei Pan
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Huanbing Tian
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Li Wei
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Linchi Chen
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Pengfei Yang
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Ji Wang
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Hanwen Cao
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lan Gao
- School of Life Sciences, Lanzhou University, Lanzhou, China.
| |
Collapse
|
24
|
Komogortsev AN, Lichitsky BV, Melekhina VG. Straightforward one-step approach towards novel derivatives of 9-oxo-5,6,7,9-tetrahydrobenzo[9,10]heptaleno[3,2-b]furan-12-yl)acetic acid based on the multicomponent reaction of colchiceine, arylglyoxals and Meldrum’s acid. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2021.153292] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
25
|
Krzywik J, Maj E, Nasulewicz-Goldeman A, Mozga W, Wietrzyk J, Huczyński A. Synthesis and antiproliferative screening of novel doubly modified colchicines containing urea, thiourea and guanidine moieties. Bioorg Med Chem Lett 2021; 47:128197. [PMID: 34116158 DOI: 10.1016/j.bmcl.2021.128197] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 11/28/2022]
Abstract
A new series of 10-demethoxy-10-methylaminocolchicines bearing urea, thiourea or aguanidine moieties at position C7 has been designed, synthesized and evaluated for in vitro anticancer activity against different cancer cell lines (A549, MCF-7, LoVo, LoVo/DX). The majority of the new derivatives were active in the nanomolar range and were characterized by lower IC50 values than cisplatin or doxorubicin. Two ureas (4 and 8) and thioureas (19 and 25) were found to be good antiproliferative agents (low IC50 values and high SI) and could prove to be promising candidates for further research in the field of anticancer drugs based on the colchicine skeleton.
Collapse
Affiliation(s)
- Julia Krzywik
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland; TriMen Chemicals, Piłsudskiego 141, 92-318 Łódź, Poland
| | - Ewa Maj
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wrocław, Poland
| | - Anna Nasulewicz-Goldeman
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wrocław, Poland
| | - Witold Mozga
- TriMen Chemicals, Piłsudskiego 141, 92-318 Łódź, Poland
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wrocław, Poland
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland.
| |
Collapse
|
26
|
Sana S, Reddy VG, Srinivasa Reddy T, Tokala R, Kumar R, Bhargava SK, Shankaraiah N. Cinnamide derived pyrimidine-benzimidazole hybrids as tubulin inhibitors: Synthesis, in silico and cell growth inhibition studies. Bioorg Chem 2021; 110:104765. [PMID: 33677248 DOI: 10.1016/j.bioorg.2021.104765] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/12/2021] [Accepted: 02/20/2021] [Indexed: 12/14/2022]
Abstract
An approach in modern medicinal chemistry to discover novel bioactive compounds is by mimicking diverse complementary pharmacophores. In extension of this strategy, a new class of piperazine-linked cinnamide derivatives of benzimidazole-pyrimidine hybrids have been designed and synthesized. Their in vitro cytotoxicity profiles were explored on selected human cancer cell lines. Specifically, structural comparison of target hybrids with tubulin-DAMA-colchicine and tubulin-nocodazole complexes has exposed a deep position of benzimidazole ring into the αT5 loop. All the synthesized compounds were demonstrated modest to interesting cytotoxicity against different cancer cell lines. The utmost cytotoxicity has shown with an amine linker of benzimidazole-pyrimidine series, with specificity toward A549 (lung cancer) cell line. The most potent compound in this series was 18i, which inhibited cancer cell growth at micromolar concentrations ranging 2.21-7.29 µM. Flow cytometry studies disclosed that 18i inhibited the cells in G2/M phase of cell cycle. The potent antitumor activity of 18i resulted from enhanced microtubule disruption at a similar level as nocodazole on β-tubulin antibody, explored using immunofluorescence staining. The most active compound 18i also inhibited tubulin polymerization with an IC50 of 5.72 ± 0.51 µM. In vitro biological analysis of 18i presented apoptosis induction on A549 cells with triggering of ROS generation and loss of mitochondrial membrane potential, resulting in DNA injury. In addition, 18i displayed impairment in cellular migration and inhibited the colony formation. Notably, the safety profile of most potent compound 18i was revealed by screening against normal human pulmonary epithelial cells (L132: IC50: 69.25 ± 5.95 μM). The detailed binding interactions of 18i with tubulin was investigated by employing molecular docking, superimposition and free energy analyses. Thus remarks made in this study established that pyrimidine-benzimidazole hybrids as a new class of tubulin polymerization inhibitors with significant anticancer activity.
Collapse
Affiliation(s)
- Sravani Sana
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Velma Ganga Reddy
- Centre for Advanced Materials & Industrial Chemistry (CAMIC), School of Science, RMIT University, GPO Box 2476, Melbourne 3001, Australia.
| | - T Srinivasa Reddy
- Centre for Advanced Materials & Industrial Chemistry (CAMIC), School of Science, RMIT University, GPO Box 2476, Melbourne 3001, Australia
| | - Ramya Tokala
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Rahul Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Suresh K Bhargava
- Centre for Advanced Materials & Industrial Chemistry (CAMIC), School of Science, RMIT University, GPO Box 2476, Melbourne 3001, Australia
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| |
Collapse
|
27
|
An insight into the anticancer potential of carbamates and thiocarbamates of 10-demethoxy-10-methylaminocolchicine. Eur J Med Chem 2021; 215:113282. [PMID: 33611191 DOI: 10.1016/j.ejmech.2021.113282] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/20/2022]
Abstract
Colchicine shows very high antimitotic activity, therefore, it is used as a lead compound for generation of new anticancer agents. In the hope of developing novel, useful drugs with more favourable pharmacological profiles, a series of doubly modified colchicine derivatives has been designed, synthesized and characterized. These novel carbamate or thiocarbamate derivatives of 10-demethoxy-10-methylaminocolchicine have been tested for their antiproliferative activity against four human cancer cell lines. Additionally, their mode of action has been evaluated as colchicine binding site inhibitors, using molecular docking studies. Most of the tested compounds showed greater cytotoxicity (IC50 in a low nanomolar range) and were characterized by a higher selectivity index than standard chemotherapeutics such as cisplatin and doxorubicin as well as unmodified colchicine. Their pharmacological use in cancer therapy could possibly be accomplished with lower dosages and result in less acute toxicity problems than in the case of colchicine. In addition, we present a QSAR model for predicting the antiproliferative activity of doubly modified derivatives for two tumour cell lines.
Collapse
|
28
|
Kurup R, Galougahi KK, Figtree G, Misra A, Patel S. The Role of Colchicine in Atherosclerotic Cardiovascular Disease. Heart Lung Circ 2021; 30:795-806. [PMID: 33461916 DOI: 10.1016/j.hlc.2020.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/10/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023]
Abstract
Colchicine, an inexpensive immunomodulatory drug used traditionally to treat gout and familial Mediterranean fever, is rapidly accumulating basic and clinical evidence for a therapeutic role in atherosclerotic cardiovascular disease. Its athero-protective properties are thought to be mainly related to its effect on tubulin polymerisation, enabling a broad range of effect on multiple atherosclerotic plaque cell types and cellular processes, including cell division, cell migration as well as pro-inflammatory cytokine and chemokine secretion. These properties indicate the potential to favourably affect all stages of atherosclerotic plaque development including formation, progression, destabilisation, and plaque rupture. This review focusses on the pharmacology of colchicine, the mechanisms by which it modulates atherosclerosis pathobiology, and summarises the current clinical evidence for its use along with the upcoming clinical trial landscape. Given the current lack of primary immunomodulatory drugs in the treatment of atherosclerosis, colchicine is a promising candidate to fill this therapeutic gap.
Collapse
Affiliation(s)
- Rahul Kurup
- The Heart Research Institute, Sydney, NSW, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia. https://twitter.com/drrahulkurup
| | - Keyvan Karimi Galougahi
- The Heart Research Institute, Sydney, NSW, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Gemma Figtree
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Ashish Misra
- The Heart Research Institute, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Sanjay Patel
- The Heart Research Institute, Sydney, NSW, Australia; Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
29
|
Sharma A, Talimarada D, Yadav UP, Singh N, Reddy AS, Bag D, Biswas K, Baidya A, Borale AN, Shinde D, Singh S, Holla H. Design and Synthesis of New Tubulin Polymerization Inhibitors Inspired from Combretastatin A‐4: An Anticancer Agent. ChemistrySelect 2020. [DOI: 10.1002/slct.202003170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Akanksha Sharma
- Department of Chemistry Central University of Karnataka Kalaburagi 585367 India
| | | | - Umesh Prasad Yadav
- Department of Human Genetics and Molecular Medicine Central University of Punjab Bathinda 151001 India
| | - Nidhi Singh
- Centre for Chemical and Pharmaceutical Sciences Central University of Punjab Bathinda 151001 India
| | - A. Sudharshan Reddy
- Department of Chemistry Central University of Karnataka Kalaburagi 585367 India
| | - Debojyoti Bag
- Department of Chemistry Central University of Karnataka Kalaburagi 585367 India
| | - Krishna Biswas
- Department of Chemistry Central University of Karnataka Kalaburagi 585367 India
| | - Amit Baidya
- Department of Chemistry Central University of Karnataka Kalaburagi 585367 India
| | - Asha N Borale
- Department of Chemistry Central University of Karnataka Kalaburagi 585367 India
| | | | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine Central University of Punjab Bathinda 151001 India
| | - Harish Holla
- Department of Chemistry Central University of Karnataka Kalaburagi 585367 India
| |
Collapse
|
30
|
Borys F, Joachimiak E, Krawczyk H, Fabczak H. Intrinsic and Extrinsic Factors Affecting Microtubule Dynamics in Normal and Cancer Cells. Molecules 2020; 25:E3705. [PMID: 32823874 PMCID: PMC7464520 DOI: 10.3390/molecules25163705] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/18/2022] Open
Abstract
Microtubules (MTs), highly dynamic structures composed of α- and β-tubulin heterodimers, are involved in cell movement and intracellular traffic and are essential for cell division. Within the cell, MTs are not uniform as they can be composed of different tubulin isotypes that are post-translationally modified and interact with different microtubule-associated proteins (MAPs). These diverse intrinsic factors influence the dynamics of MTs. Extrinsic factors such as microtubule-targeting agents (MTAs) can also affect MT dynamics. MTAs can be divided into two main categories: microtubule-stabilizing agents (MSAs) and microtubule-destabilizing agents (MDAs). Thus, the MT skeleton is an important target for anticancer therapy. This review discusses factors that determine the microtubule dynamics in normal and cancer cells and describes microtubule-MTA interactions, highlighting the importance of tubulin isoform diversity and post-translational modifications in MTA responses and the consequences of such a phenomenon, including drug resistance development.
Collapse
Affiliation(s)
- Filip Borys
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, 00-664 Warsaw, Poland;
| | - Ewa Joachimiak
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
| | - Hanna Krawczyk
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 3 Noakowskiego Street, 00-664 Warsaw, Poland;
| | - Hanna Fabczak
- Laboratory of Cytoskeleton and Cilia Biology Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland;
| |
Collapse
|
31
|
Krzywik J, Aminpour M, Maj E, Mozga W, Wietrzyk J, Tuszyński JA, Huczyński A. New Series of Double-Modified Colchicine Derivatives: Synthesis, Cytotoxic Effect and Molecular Docking. Molecules 2020; 25:molecules25153540. [PMID: 32748887 PMCID: PMC7435877 DOI: 10.3390/molecules25153540] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023] Open
Abstract
Colchicine is a well-known anticancer compound showing antimitotic effect on cells. Its high cytotoxic activity against different cancer cell lines has been demonstrated many times. In this paper we report the syntheses and spectroscopic analyses of novel colchicine derivatives obtained by structural modifications at C7 (carbon-nitrogen single bond) and C10 (methylamino group) positions. All the obtained compounds have been tested in vitro to determine their cytotoxicity toward A549, MCF-7, LoVo, LoVo/DX, and BALB/3T3 cell lines. The majority of obtained derivatives exhibited higher cytotoxicity than colchicine, doxorubicin and cisplatin against the tested cancerous cell lines. Additionally, most of the presented derivatives were able to overcome the resistance of LoVo/DX cells. Additionally, their mode of binding to β-tubulin was evaluated in silico. Molecular docking studies showed that apart from the initial amides 1 and 2, compound 14, which had the best antiproliferative activity (IC50 = 0.1–1.6 nM), stood out also in terms of its predicted binding energy and probably binds best into the active site of βI-tubulin isotype.
Collapse
Affiliation(s)
- Julia Krzywik
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland;
- TriMen Chemicals, Piłsudskiego 141, 92-318 Łódź, Poland;
| | - Maral Aminpour
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada; (M.A.); (J.A.T.)
| | - Ewa Maj
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wrocław, Poland; (E.M.); (J.W.)
| | - Witold Mozga
- TriMen Chemicals, Piłsudskiego 141, 92-318 Łódź, Poland;
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wrocław, Poland; (E.M.); (J.W.)
| | - Jack A. Tuszyński
- Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada; (M.A.); (J.A.T.)
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi, 24, 10129 Torino, Italy
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland;
- Correspondence: ; Tel.: +48-61-8291673
| |
Collapse
|
32
|
Yue Q, Liu T, Cheng Z. Protective Effect of Colchicine on LPS-Induced Lung Injury in Rats via Inhibition of P-38, ERK1/2, and JNK Activation. Pharmacology 2020; 105:639-644. [PMID: 32688359 DOI: 10.1159/000504759] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/12/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Acute lung injury (ALI), a commonly detected syndrome, is characterized by the accumulation of neutrophils and leucocytes, and inflammation of pulmonary tissues. OBJECTIVE The present study was designed to investigate the effect and underlying mechanism of colchicine on LPS-induced lung injury. METHODS The rats were divided randomly into 6 groups of 10 each: normal control, untreated, and 4 colchicine (5, 10, 15, and 20 mg/kg) treatment groups. ALI was induced in rats by the administration of 20 μg LPS intratracheally. Rats in the normal control and untreated groups were injected normal saline, while those in the treatment groups received 5, 10, 15, and 20 mg/kg doses of colchicine daily for 1 month. ELISA was used for determination of interleukin (IL)-1β, IL-6, tumour necrosis factor (TNF)-α, superoxide dismutase (SOD), and leucocytes in the rat bronchoalveolar lavage fluid (BALF). The expression of P-38, JNK, and Erk-1/2 was analysed by Western blotting. RESULTS In LPS-administered TC-1 cells, the levels of IL-1β, IL-6, and TNF-α were markedly higher. Treatment with colchicine reduced the levels of IL-1β, IL-6, and TNF-α in LPS-administered TC-1 cells. Colchicine treatment caused a marked reduction in LPS-induced accumulation of inflammatory cells in the rat lungs. The LPS-induced aggregation of leucocytes and neutrophils in the rat BALF was also suppressed markedly on treatment with colchicine. Treatment of the lung injury in rats with colchicine caused a marked decrease in the level of IL-1β, IL-6, and TNF-α in BALF. The LPS-mediated suppression of SOD in the rat BALF was prevented by treatment with colchicine. Treatment of the rats with colchicine attenuated the LPS-induced activation of P-38, Erk1/2, and JNK in pulmonary tissues. CONCLUSION In summary, colchicine treatment prevents LPS-induced lung damage in rats through targeting activation of P-38, ERK1/2, and JNK. Therefore, colchicine may be used for the development of treatment for ALI.
Collapse
Affiliation(s)
- Qianyu Yue
- Department of Respiratory Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tian Liu
- Respiratory Medicine, People's Hospital of Qingyuan County, Baoding, China
| | - Zhongfeng Cheng
- Emergency Department, The Second People's Hospital of Yunnan Province, Kunming, China,
| |
Collapse
|
33
|
Gracheva IA, Shchegravina ES, Schmalz HG, Beletskaya IP, Fedorov AY. Colchicine Alkaloids and Synthetic Analogues: Current Progress and Perspectives. J Med Chem 2020; 63:10618-10651. [PMID: 32432867 DOI: 10.1021/acs.jmedchem.0c00222] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Colchicine, the main alkaloid of Colchicum autumnale, is one of the most famous natural molecules. Although colchicine belongs to the oldest drugs (in use since 1500 BC), its pharmacological potential as a lead structure is not yet fully exploited. This review is devoted to the synthesis and structure-activity relationships (SAR) of colchicine alkaloids and their analogues with modified A, B, and C rings, as well as hybrid compounds derived from colchicinoids including prodrugs, conjugates, and delivery systems. The systematization of a vast amount of information presented to date will create a paradigm for future studies of colchicinoids for neoplastic and various other diseases.
Collapse
Affiliation(s)
- Iuliia A Gracheva
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russian Federation
| | - Ekaterina S Shchegravina
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russian Federation
| | | | - Irina P Beletskaya
- Department of Chemistry, M. V. Lomonosov Moscow State University, Moscow 119992, Russian Federation
| | - Alexey Yu Fedorov
- Department of Chemistry, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russian Federation
| |
Collapse
|
34
|
Gasic I, Groendyke BJ, Nowak RP, Yuan JC, Kalabathula J, Fischer ES, Gray NS, Mitchison TJ. Tubulin Resists Degradation by Cereblon-Recruiting PROTACs. Cells 2020; 9:E1083. [PMID: 32349222 PMCID: PMC7290497 DOI: 10.3390/cells9051083] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 12/22/2022] Open
Abstract
Dysregulation of microtubules and tubulin homeostasis has been linked to developmental disorders, neurodegenerative diseases, and cancer. In general, both microtubule-stabilizing and destabilizing agents have been powerful tools for studies of microtubule cytoskeleton and as clinical agents in oncology. However, many cancers develop resistance to these agents, limiting their utility. We sought to address this by developing a different kind of agent: tubulin-targeted small molecule degraders. Degraders (also known as proteolysis-targeting chimeras (PROTACs)) are compounds that recruit endogenous E3 ligases to a target of interest, resulting in the target's degradation. We developed and examined several series of α- and β-tubulin degraders, based on microtubule-destabilizing agents. Our results indicate, that although previously reported covalent tubulin binders led to tubulin degradation, in our hands, cereblon-recruiting PROTACs were not efficient. In summary, while we consider tubulin degraders to be valuable tools for studying the biology of tubulin homeostasis, it remains to be seen whether the PROTAC strategy can be applied to this target of high clinical relevance.
Collapse
Affiliation(s)
- Ivana Gasic
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA;
| | - Brian J. Groendyke
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; (R.P.N.); (J.C.Y.); (J.K.); (E.S.F.); (N.S.G.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Radosław P. Nowak
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; (R.P.N.); (J.C.Y.); (J.K.); (E.S.F.); (N.S.G.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - J. Christine Yuan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; (R.P.N.); (J.C.Y.); (J.K.); (E.S.F.); (N.S.G.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Joann Kalabathula
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; (R.P.N.); (J.C.Y.); (J.K.); (E.S.F.); (N.S.G.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; (R.P.N.); (J.C.Y.); (J.K.); (E.S.F.); (N.S.G.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Nathanael S. Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; (R.P.N.); (J.C.Y.); (J.K.); (E.S.F.); (N.S.G.)
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy J. Mitchison
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
35
|
Barreca M, Stathis A, Barraja P, Bertoni F. An overview on anti-tubulin agents for the treatment of lymphoma patients. Pharmacol Ther 2020; 211:107552. [PMID: 32305312 DOI: 10.1016/j.pharmthera.2020.107552] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 04/14/2020] [Indexed: 01/19/2023]
Abstract
Anti-tubulin agents constitute a large class of compounds with broad activity both in solid tumors and hematologic malignancies, due to the interference with microtubule dynamics. Since microtubules play crucial roles in the regulation of the mitotic spindles, the interference with their function usually leads to a block in cell division with arrest at the metaphase/anaphase junction of mitosis, followed to apoptosis. This explains the reason why tubulin-binding agents (TBAs) proved to be extremely active in patients with cancer. Several anti-tubulin agents are indicated in the treatment of patients with lymphomas both alone and in combination chemotherapy regimens. The article reviews the literature on classic and more recent anti-tubulin agents, providing an insight into their mechanisms of action and their use in the treatment of lymphoma.
Collapse
Affiliation(s)
- Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy; Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland,; Faculty of Biomedical Sciences, USI, Lugano, Switzerland
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland,; Oncology Institute of Southern Switzerland, Bellinzona, Switzerland,.
| |
Collapse
|
36
|
Synthesis, Antiproliferative Activity and Molecular Docking Studies of Novel Doubly Modified Colchicine Amides and Sulfonamides as Anticancer Agents. Molecules 2020; 25:molecules25081789. [PMID: 32295119 PMCID: PMC7221574 DOI: 10.3390/molecules25081789] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/05/2020] [Accepted: 04/11/2020] [Indexed: 12/18/2022] Open
Abstract
Colchicine is a well-known compound with strong antiproliferative activity that has had limited use in chemotherapy because of its toxicity. In order to create more potent anticancer agents, a series of novel colchicine derivatives have been obtained by simultaneous modification at C7 (amides and sulfonamides) and at C10 (methylamino group) positions and characterized by spectroscopic methods. All the synthesized compounds have been tested in vitro to evaluate their cytotoxicity toward A549, MCF-7, LoVo, LoVo/DX and BALB/3T3 cell lines. Additionally, the activity of the studied compounds was investigated using computational methods involving molecular docking of the colchicine derivatives to β-tubulin. The majority of the obtained derivatives exhibited higher cytotoxicity than colchicine, doxorubicin or cisplatin against tested cancer cell lines. Furthermore, molecular modeling studies of the obtained compounds revealed their possible binding modes into the colchicine binding site of tubulin.
Collapse
|
37
|
Sun FY, Liu L, Yu Y, Ruan XM, Wang CY, Hu QW, Wu DX, Sun G. MicroRNA-mediated responses to colchicine treatment in barley. PLANTA 2020; 251:44. [PMID: 31907626 DOI: 10.1007/s00425-019-03326-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 12/06/2019] [Indexed: 06/10/2023]
Abstract
In Hordeum vulgare, nine differentially expressed novel miRNAs were induced by colchicine. Five novel miRNA in colchicine solution showed the opposite expression patterns as those in water. Colchicine is a commonly used agent for plant chromosome set doubling. MicroRNA-mediated responses to colchicine treatment in plants have not been characterized. Here, we characterized new microRNAs induced by colchicine treatment in Hordeum vulgare using high-throughput sequencing. Our results showed that 39 differentially expressed miRNAs were affected by water treatment, including 34 novel miRNAs and 5 known miRNAs; 42 miRNAs, including 37 novel miRNAs and 5 known miRNAs, were synergistically affected by colchicine and water, and 9 differentially expressed novel miRNAs were induced by colchicine. The novel_mir69, novel_mir57, novel_mir75, novel_mir38, and novel_mir56 in colchicine treatment showed the opposite expression patterns as those in water. By analyzing these 9 differentially expressed novel miRNAs and their targets, we found that novel_mir69, novel_mir56 and novel_mir25 co-target the genes involving the DNA repair pathway. Based on our results, microRNA-target regulation network under colchicine treatment was proposed, which involves actin, cell cycle regulation, cell wall synthesis, and the regulation of oxidative stress. Overall, the results demonstrated the critical role of microRNAs mediated responses to colchicine treatment in plants.
Collapse
Affiliation(s)
- Fang-Yao Sun
- College of Agronomy, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Lin Liu
- College of Agronomy, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Yi Yu
- College of Agronomy, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Xin-Ming Ruan
- College of Agronomy, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - Cheng-Yu Wang
- College of Agronomy, Anhui Agricultural University, Hefei, 230036, Anhui, China.
| | - Qun-Wen Hu
- College of Agronomy, Anhui Agricultural University, Hefei, 230036, Anhui, China
| | - De-Xiang Wu
- College of Agronomy, Anhui Agricultural University, Hefei, 230036, Anhui, China.
| | - Genlou Sun
- Biology Department, Saint Mary's University, Halifax, NS, B3H 3C3, Canada.
| |
Collapse
|
38
|
Inhibition of respiratory syncytial virus replication and suppression of RSV-induced airway inflammation in neonatal rats by colchicine. 3 Biotech 2019; 9:392. [PMID: 31656730 DOI: 10.1007/s13205-019-1917-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/24/2019] [Indexed: 12/31/2022] Open
Abstract
The present study investigated the role of colchicine in the treatment of RSV infection. Treatment of BEAS-2B cells following RSV infection with colchicine caused a significant decrease in the number of viral plaques. In RSV-infected BEAS-2B cells' treatment with colchicine leads to a significant up-regulation of both IFN-β1 and RIG-I genes. The levels of interleukin, NO, and MDA were suppressed in BEAS-2B cells infected with RSV by colchicine. The phosphorylation of Stat3, COX-2, and p38 was also suppressed significantly by colchicine. The phosphorylation of IkBα was promoted in RSV-infected BEAS-2B cells' oncolchicine treatment. In neonatal rats, replication of RSV was inhibited significantly by colchicine treatment which was evident by suppression of RSV-L gene expression. A significant decrease in the level of IL-6 and TNF-α was caused in neonatal rat BALF by colchicine treatment. The production of MDA, NO and MPO in the neonatal rat BALF was suppressed markedly by colchicine treatment. Treatment of the neonatal rats infected by RSV with colchicine suppressed the release of IκBα and COX-2 in the pulmonary epithelial cells. Colchicine treatment of the neonatal rats promoted the expression of IFN-α and IFN-β1. In summary, the current study showed that colchicine inhibited RSV infection in neonatal rats through regulation of anti-oxidative factor production. The expression of IFN-β1 and RIG-I genes was also up-regulated in the RSV-infected alveolar epithelial cells by treatment with colchicine. Therefore, colchicine may be developed as the therapeutic agent for the treatment of RSV infection.
Collapse
|
39
|
Liu F, Lin S, Zhang C, Ma J, Han Z, Jia F, Xie W, Li X. The Novel Nature Microtubule Inhibitor Ivalin Induces G2/M Arrest and Apoptosis in Human Hepatocellular Carcinoma SMMC-7721 Cells In Vitro. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E470. [PMID: 31409007 PMCID: PMC6723664 DOI: 10.3390/medicina55080470] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/02/2019] [Accepted: 08/08/2019] [Indexed: 12/11/2022]
Abstract
Background and Objectives: Microtubules are an attractive target for cancer chemotherapy. Previously, we reported that Ivalin exhibited excellent anti-migration and anti-invasion activities in human breast cancer cells. Here, we examined the microtubule inhibition effect of Ivalin in human hepatocellular carcinoma SMMC-7721 cells. Materials and Methods: We used the 3-(4,5-dimethylthiazol)-2,5-diphenyltetrazolium bromide (MTT) assay to evaluate the cell proliferation effect of Ivalin and flow cytometry analysis to detect the apoptotic and cell cycle arrest effects of Ivalin. Immunofluorescence staining was used to measure the effect of Ivalin on the cytoskeleton network, and Western blotting was used to detect the expression levels of Bax, Bcl-2, Cdc2, phosphor-Cdc2, Cdc25A, Cyclin B1, and tubulin. Results: Ivalin induced cell cycle G2/M arrest and subsequent triggered apoptosis in human hepatocellular carcinoma SMMC-7721 cells. Furthermore, microtubules were shown to be involved in Ivalin-meditated apoptosis. In this connection, Ivalin treatment suppressed cellular microtubule network formation by regulating microtubule depolymerization. Moreover, Western blotting revealed Cdc25A and Cyclin B1 were upregulated in Ivalin-meditated cell cycle arrest. Subsequently, the induction of Bax (a proapoptotic protein) and reduction of Bcl-2 (an anti-apoptotic protein) expression were observed in Ivalin-treated SMMC-7721 cells. Conclusion: Ivalin induced microtubule depolymerization, then blocked cells in mitotic phase, and eventually resulted in apoptosis in SMMC-7721 cells. Collectively, these data indicate that Ivalin, acting as a novel inhibitor of microtubules, could be considered as a promising lead in anticancer drug development.
Collapse
Affiliation(s)
- Fangyuan Liu
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Shiqi Lin
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Caiyun Zhang
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Jiahui Ma
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Zhuo Han
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Fujuan Jia
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Weidong Xie
- College of Marine Science, Shandong University, Weihai 264209, China
| | - Xia Li
- College of Marine Science, Shandong University, Weihai 264209, China.
- School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
- The Key Laboratory of Chemistry for Natural Product of Guizhou Province and Chinese Academy of Science, Guiyang 550002, China.
| |
Collapse
|
40
|
Wen J, Liu F, Tao B, Sun S. GSH-responsive anti-mitotic cell penetrating peptide-linked podophyllotoxin conjugate for improving water solubility and targeted synergistic drug delivery. Bioorg Med Chem Lett 2019; 29:1019-1022. [DOI: 10.1016/j.bmcl.2019.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/21/2019] [Accepted: 02/05/2019] [Indexed: 01/23/2023]
|
41
|
Kaul R, Risinger AL, Mooberry SL. Microtubule-Targeting Drugs: More than Antimitotics. JOURNAL OF NATURAL PRODUCTS 2019; 82:680-685. [PMID: 30835122 DOI: 10.1021/acs.jnatprod.9b00105] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Nature has yielded numerous compounds that bind to tubulin/microtubules and disrupt microtubule function. Even with the advent of targeted therapies for cancer, natural products and their derivatives that target microtubules are some of the most effective drugs used in the treatment of solid tumors and hematological malignancies. For decades, these drugs were thought to work solely through their ability to inhibit mitosis. Accumulating evidence demonstrates that their actions are much more complex, in that they also have significant effects on microtubules in nondividing cells that inhibit a diverse range of signaling events important for carcinogenesis. The abilities of these drugs to inhibit oncogenic signaling likely underlies their efficacy, especially in solid tumors. In this review, we describe the role of microtubules in cells, the proliferation paradox of cells in culture as compared to cancers in patients, and evidence that microtubule-targeting drugs inhibit cellular signaling pathways important for tumorigenesis. The potential mechanisms behind differences in the clinical indications and efficacy of these natural-product-derived drugs are also discussed. Microtubules are an important target for structurally diverse natural products, and a fuller understanding of the mechanisms of action of these drugs will promote their optimal use.
Collapse
|
42
|
The effectiveness of colchicine combined with mitomycin C to prolong bleb function in trabeculectomy in rabbits. PLoS One 2019; 14:e0213811. [PMID: 30889194 PMCID: PMC6424470 DOI: 10.1371/journal.pone.0213811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 02/28/2019] [Indexed: 11/30/2022] Open
Abstract
Purpose To investigate the potential of colchicine to improve bleb function after trabeculectomy. Methods To find the maximum usable colchicine concentration, an ocular irritation study was performed with the Draize test at concentrations of 0.001%, 0.01% and 0.1%. Additionally, the synergistic effect of topical colchicine instillation and MMC application to surgical site was evaluated in a rabbit model by measuring changes after trabeculectomy in intraocular pressure (IOP) and bleb morphology score at 3, 7, 14, 21, 28, 35, 42, and 49 days. Results Experiments with a rabbit model of trabeculectomy showed that 0.04% MMC plus 0.01% colchicine was more effective than saline and 0.04% MMC alone in maintaining IOP reduction at days 7–49 (P < 0.01 at all time points) and day 49 (P < 0.05), respectively, while 0.04% MMC alone was more effective than saline only at days 7–35 (P < 0.05 at all time points). 0.04% MMC plus 0.01% colchicine and 0.04% MMC alone were more effective than saline at preserving bleb score at days 7–21 and 35–49 (P < 0.05 at all time points) and at days 7–35 (P < 0.05 at all time points), respectively. Conclusion Colchicine may be a promising adjuvant for strengthening the effect of MMC and improving the survival of the filtering bleb in trabeculectomy.
Collapse
|
43
|
Lynch AM, Eastmond D, Elhajouji A, Froetschl R, Kirsch-Volders M, Marchetti F, Masumura K, Pacchierotti F, Schuler M, Tweats D. Targets and mechanisms of chemically induced aneuploidy. Part 1 of the report of the 2017 IWGT workgroup on assessing the risk of aneugens for carcinogenesis and hereditary diseases. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 847:403025. [PMID: 31699346 DOI: 10.1016/j.mrgentox.2019.02.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/22/2019] [Accepted: 02/20/2019] [Indexed: 02/06/2023]
Abstract
An aneuploidy workgroup was established as part of the 7th International Workshops on Genotoxicity Testing. The workgroup conducted a review of the scientific literature on the biological mechanisms of aneuploidy in mammalian cells and methods used to detect chemical aneugens. In addition, the current regulatory framework was discussed, with the objective to arrive at consensus statements on the ramifications of exposure to chemical aneugens for human health risk assessment. As part of these efforts, the workgroup explored the use of adverse outcome pathways (AOPs) to document mechanisms of chemically induced aneuploidy in mammalian somatic cells. The group worked on two molecular initiating events (MIEs), tubulin binding and binding to the catalytic domain of aurora kinase B, which result in several adverse outcomes, including aneuploidy. The workgroup agreed that the AOP framework provides a useful approach to link evidence for MIEs with aneuploidy on a cellular level. The evidence linking chemically induced aneuploidy with carcinogenicity and hereditary disease was also reviewed and is presented in two companion papers. In addition, the group came to the consensus that the current regulatory test batteries, while not ideal, are sufficient for the identification of aneugens and human risk assessment. While it is obvious that there are many different MIEs that could lead to the induction of aneuploidy, the most commonly observed mechanisms involving chemical aneugens are related to tubulin binding and, to a lesser extent, inhibition of mitotic kinases. The comprehensive review presented here should help with the identification and risk management of aneugenic agents.
Collapse
Affiliation(s)
| | | | - Azeddine Elhajouji
- Novartis Institutes for Biomedical Research, Preclinical Safety, Basel, Switzerland
| | | | | | - Francesco Marchetti
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | - Kenichi Masumura
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Kanagawa, Japan
| | - Francesca Pacchierotti
- Health Protection Technology Division, Laboratory of Biosafety and Risk Assessment, ENEA, CR Casaccia, Rome, Italy
| | | | | |
Collapse
|
44
|
Narayanan S, Gupta P, Nazim U, Ali M, Karadkhelkar N, Ahmad M, Chen ZS. Anti-cancer effect of Indanone-based thiazolyl hydrazone derivative on colon cancer cell lines. Int J Biochem Cell Biol 2019; 110:21-28. [PMID: 30794858 DOI: 10.1016/j.biocel.2019.02.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 01/26/2019] [Accepted: 02/19/2019] [Indexed: 11/17/2022]
Abstract
Colorectal cancer is the third leading cause of cancer related deaths in the United States. Currently, Irinotecan, a topoisomerase I inhibitor, is an approved anti-cancer drug for the treatment of patients with advanced or recurrent colorectal cancer. Considering low response rate and events of high toxicity caused by irinotecan, we evaluated a series of thirteen thiazolyl hydrazone derivatives of 1-indanone for their potential antineoplastic activity and four compounds showed promising anti-cancer activity against most of the tested colon cancer cell lines with IC50 values ranging from 0.41 ± 0.19 to 6.85 ± 1.44 μM. It is noteworthy that the compound, N-Indan-1-ylidene-N'-(4-Biphenyl-4-yl-thiazol-2-yl)-hydrazine (ITH-6) is found to be more effective than irinotecan against colon cancer cells, HT-29, COLO 205, and KM 12. Mechanistic studies reveal that ITH-6 arrests these cancer cell lines in G2/M phase of the cell cycle, induces apoptosis and causes an increase in ROS level with a significant reduction in the GSH level. The mechanism of inhibition relates to the inhibition of tubulin polymerization in the mitotic phase. These findings suggest that ITH-6 is a novel drug candidate for the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Silpa Narayanan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Pranav Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Urooj Nazim
- Department of Pharmaceutical Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Mohsin Ali
- Department of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Nishant Karadkhelkar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Mansoor Ahmad
- Department of Pharmaceutical Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| |
Collapse
|
45
|
Abstract
Colchicine, the main alkaloid of the poisonous plant meadow saffron (Colchicum autumnale L.), is a classical drug used for the treatment of gout and familial Mediterranean fever. Although colchicine is not clinically used to treat cancer because of toxicity, it exerts antiproliferative effects through the inhibition of microtubule formation by blocking the cell cycle at the G2/M phase and triggering apoptosis. Colchicine can still be used as a lead compound for the generation of potential anticancer drugs. Thus, numerous analogues of colchicine have been synthesized in the hope of developing novel, useful drugs with more favourable pharmacological profiles. Several colchicine semisynthetics are less toxic than colchicine and research is being carried out on effective, less toxic colchicine semisynthetic formulations with potential drug-delivery strategies directly targeting multiple solid cancers. This review focuses on the anticancer role of some of colchicine-based derivatives and their therapeutic importance.
Collapse
|
46
|
Zawahir S, Gawarammana I, Dargan PI, Abdulghni M, Dawson AH. Activated charcoal significantly reduces the amount of colchicine released from Gloriosa superba in simulated gastric and intestinal media. Clin Toxicol (Phila) 2017; 55:914-918. [PMID: 28535126 PMCID: PMC5942148 DOI: 10.1080/15563650.2017.1325897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 04/11/2017] [Accepted: 04/12/2017] [Indexed: 11/10/2022]
Abstract
BACKGROUND Poisoning with Gloriosa superba, a plant containing colchicine, is common in Sri Lanka. OBJECTIVES This study was to estimate release of colchicine from 5 g of different parts of Gloriosa superba in simulated gastric and intestinal media, and examine the binding efficacy of activated charcoal (AC) to colchicine within this model. METHODS A USP dissolution apparatus-II was used to prepare samples for analysis of colchicine using HPLC. RESULTS Cumulative colchicine release from tuber in gastric media at 120 minutes was significantly higher (2883 μg/g) than in intestinal media (1015 μg/g) (p < .001). Mean ± SD cumulative colchicine concentration over 2 hours from tuber, leaves and trunk in gastric medium was 2883.15 ± 1295.63, 578.25 ± 366.26 and 345.60 ± 200.08 μg/g respectively and the release in intestinal media was 1014.75 ± 268.16, 347.40 ± 262.61 and 251.55 ± 285.72 μg/g respectively. Introduction of 50 g of AC into both media made colchicine undetectable (<0.1 μg/ml). CONCLUSIONS The tuber released the highest quantity of colchicine. The colchicine release and elapse time to achieve saturated, equilibrium dissolution mainly depends on physicochemical properties of plant part. Significant in vitro binding of colchicine to AC suggests that AC has a role in decontamination of patients presenting to hospital after ingestion of Gloriosa superba.
Collapse
Affiliation(s)
- Shukry Zawahir
- Faculty of Medicine, University of Ruhuna, Galle, Sri Lanka
- Faculty of Pharmacy, University of Sydney, Sydney, Australia
| | - Indika Gawarammana
- South Asian Clinical Toxicology Research Collaboration, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
- Department of Medicine, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Paul I. Dargan
- Department of Clinical Toxicology, Guy’s and St. Thomas’s NHS Foundation Trust, London, UK
- Faculty of Life Sciences and Medicine, King’s College London, London, UK
| | - Mahfoudh Abdulghni
- Unaizah College of Pharmacy, Qassim University, Unaizah, Kingdom of Saudi Arabia
| | - Andrew H. Dawson
- South Asian Clinical Toxicology Research Collaboration, Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
- Central Clinical School, The University of Sydney, Sydney, Australia
| |
Collapse
|
47
|
Zhou K, Fleet P, Nevo E, Zhang X, Sun G. Transcriptome analysis reveals plant response to colchicine treatment during on chromosome doubling. Sci Rep 2017; 7:8503. [PMID: 28819246 PMCID: PMC5561261 DOI: 10.1038/s41598-017-08391-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 07/10/2017] [Indexed: 11/09/2022] Open
Abstract
Colchicine was commonly used to artificially double chromosomes while the transcriptome changes in colchicine treated plants has rarely been characterized. To understand the molecular mechanism of colchicine on chromosome doubling, we characterized transcriptome data of diploid orchardgrass root after colchicine treatment. Our results showed that 3381 of differentially expressed genes (DEGs) were mainly affected by water stress, 1258 DEGs that were expressed significantly in sample DacR5tr but not in DacR5ck were considered to be mainly affected by colchicine and combination of water and colchicine. These DEGs mainly regulated by colchicine were enriched to gene ontology (GO) accessions of cation binding, catalytic activity, membrane and transporter activity, and enriched to Kyoto Encyclopedia of Genes and Genome (KEGG) pathways of phenylpropanoid biosynthesis, phenylalanine metabolism, plant hormone signal transduction and starch and sucrose metabolism. Genes related to microtubule, spindle, chromosomal kinetochore, vesicle, cellulose and processes of cytoplasm movement, chromatid segregation, membrane and cell wall development were inhibited by colchicine. Our results revealed that colchicine restrained the microtubules and inhibited gene expression of cytokinesis, which might slow down the cell activity, delay the cell into anaerobic respiration, resulting in apoptosis at late stage, and relieving of waterlogging.
Collapse
Affiliation(s)
- Kai Zhou
- Department of Grassland Science, Animal Science and Technology College, Sichuan Agricultural University, Chengdu, 611130, China
- Department of Biology, Saint Mary's University, Halifax, NS, B3H3C3, Canada
| | - Paige Fleet
- Department of Biology, Saint Mary's University, Halifax, NS, B3H3C3, Canada
| | - Eviatar Nevo
- Institute of Evolution, University of Haifa, Mount Carmel, Haifa, 3498838, Israel
| | - Xinquan Zhang
- Department of Grassland Science, Animal Science and Technology College, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Genlou Sun
- Department of Biology, Saint Mary's University, Halifax, NS, B3H3C3, Canada.
| |
Collapse
|
48
|
Modeling the Colchicum autumnale Tubulin and a Comparison of Its Interaction with Colchicine to Human Tubulin. Int J Mol Sci 2017; 18:ijms18081676. [PMID: 28767055 PMCID: PMC5578066 DOI: 10.3390/ijms18081676] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/27/2017] [Accepted: 07/28/2017] [Indexed: 11/17/2022] Open
Abstract
Tubulin is the target for many small-molecule natural compounds, which alter microtubules dynamics, and lead to cell cycle arrest and apoptosis. One of these compounds is colchicine, a plant alkaloid produced by Colchicum autumnale. While C. autumnale produces a potent cytotoxin, colchicine, and expresses its target protein, it is immune to colchicine’s cytotoxic action and the mechanism of this resistance is hitherto unknown. In the present paper, the molecular mechanisms responsible for colchicine resistance in C. autumnale are investigated and compared to human tubulin. To this end, homology models for C. autumnale α-β tubulin heterodimer are created and molecular dynamics (MD) simulations together with molecular mechanics Poisson–Boltzmann calculations (MM/PBSA) are performed to determine colchicine’s binding affinity for tubulin. Using our molecular approach, it is shown that the colchicine-binding site in C. autumnale tubulin contains a small number of amino acid substitutions compared to human tubulin. However, these substitutions induce significant reduction in the binding affinity for tubulin, and subsequently fewer conformational changes in its structure result. It is suggested that such small conformational changes are insufficient to profoundly disrupt microtubule dynamics, which explains the high resistance to colchicine by C. autumnale.
Collapse
|
49
|
Dalghi MG, Ferreira-Gomes M, Montalbetti N, Simonin A, Strehler EE, Hediger MA, Rossi JP. Cortical cytoskeleton dynamics regulates plasma membrane calcium ATPase isoform-2 (PMCA2) activity. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1413-1424. [DOI: 10.1016/j.bbamcr.2017.05.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/11/2017] [Accepted: 05/15/2017] [Indexed: 01/17/2023]
|
50
|
Rica CI, Naessens T, Pieters L, Apers S. An HPLC Method for the Quantification of Colchicine and Colchicine Derivatives in Gloriosa superba seeds. Nat Prod Commun 2017. [DOI: 10.1177/1934578x1701200817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
An 80% ethanolic extract of Gloriosa superba L. seeds (glory lily, Colchicaceae), as well as a colchicine-poor/colchicoside-rich extract, were shown to exhibit antitumor activity in a murine model for pancreatic cancer. Phytochemical investigations of the 80% ethanolic extract led to the identification of colchicine, 3- O-demethylcolchicine, and colchicoside. The objective of this work was to develop and validate a high performance liquid chromatographic analytical method according to the ICH guidelines for the quantification of these constituents. The calibration model appeared to be linear, ranging from 2.1 μg/mL to 41.9 μg/mL. The method was shown to be precise with respect to time (RSD% of 3.1% for colchicine, 2.9% for 3- O-demethylcolchicine, and 4.7% for colchicoside, 3 days, n = 6) and with respect to the concentration (RSD% of 2.9% for colchicine, 3.0% for 3- O-demethylcolchicine and 4.1% for colchicoside, 3 levels, n = 6). The recovery of colchicine resulted in a mean recovery of 100.02% with a RSD% of 2.1%. The correction factors for colchicoside and 3- O-demethylcolchicine were determined as 1.94 and 1.20, respectively. The total amount of colchicine and colchicine derivatives found in the crude extract of G. superba was 4.6% (m/m) expressed as colchicine and the overall mean of colchicine found in the crude extract was 2.8% (m/m). By using the correction factors, the other constituents of the crude extract could also be quantified, and it was found to contain 1.5% (m/m) colchicoside and 1.3% (m/m) 3- O-demethylcolchicine.
Collapse
Affiliation(s)
- Capistrano I. Rica
- Natural Products & Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp-Wilrijk, Belgium
| | - Tania Naessens
- Natural Products & Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp-Wilrijk, Belgium
| | - Luc Pieters
- Natural Products & Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp-Wilrijk, Belgium
| | - Sandra Apers
- Natural Products & Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp-Wilrijk, Belgium
| |
Collapse
|