1
|
Wang Y, Yu X, Gu Y, Li W, Zhu K, Chen L, Tang Y, Liu G. XGraphCDS: An explainable deep learning model for predicting drug sensitivity from gene pathways and chemical structures. Comput Biol Med 2024; 168:107746. [PMID: 38039896 DOI: 10.1016/j.compbiomed.2023.107746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/29/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023]
Abstract
Cancer is a highly complex disease characterized by genetic and phenotypic heterogeneity among individuals. In the era of precision medicine, understanding the genetic basis of these individual differences is crucial for developing new drugs and achieving personalized treatment. Despite the increasing abundance of cancer genomics data, predicting the relationship between cancer samples and drug sensitivity remains challenging. In this study, we developed an explainable graph neural network framework for predicting cancer drug sensitivity (XGraphCDS) based on comparative learning by integrating cancer gene expression information and drug chemical structure knowledge. Specifically, XGraphCDS consists of a unified heterogeneous network and multiple sub-networks, with molecular graphs representing drugs and gene enrichment scores representing cell lines. Experimental results showed that XGraphCDS consistently outperformed most state-of-the-art baselines (R2 = 0.863, AUC = 0.858). We also constructed a separate in vivo prediction model by using transfer learning strategies with in vitro experimental data and achieved good predictive power (AUC = 0.808). Simultaneously, our framework is interpretable, providing insights into resistance mechanisms alongside accurate predictions. The excellent performance of XGraphCDS highlights its immense potential in aiding the development of selective anti-tumor drugs and personalized dosing strategies in the field of precision medicine.
Collapse
Affiliation(s)
- Yimeng Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Xinxin Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yaxin Gu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Weihua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Keyun Zhu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Long Chen
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Guixia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
2
|
Pastuch-Gawołek G, Szreder J, Domińska M, Pielok M, Cichy P, Grymel M. A Small Sugar Molecule with Huge Potential in Targeted Cancer Therapy. Pharmaceutics 2023; 15:913. [PMID: 36986774 PMCID: PMC10056414 DOI: 10.3390/pharmaceutics15030913] [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: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
The number of cancer-related diseases is still growing. Despite the availability of a large number of anticancer drugs, the ideal drug is still being sought that would be effective, selective, and overcome the effect of multidrug resistance. Therefore, researchers are still looking for ways to improve the properties of already-used chemotherapeutics. One of the possibilities is the development of targeted therapies. The use of prodrugs that release the bioactive substance only under the influence of factors characteristic of the tumor microenvironment makes it possible to deliver the drug precisely to the cancer cells. Obtaining such compounds is possible by coupling a therapeutic agent with a ligand targeting receptors, to which the attached ligand shows affinity and is overexpressed in cancer cells. Another way is to encapsulate the drug in a carrier that is stable in physiological conditions and sensitive to conditions of the tumor microenvironment. Such a carrier can be directed by attaching to it a ligand recognized by receptors typical of tumor cells. Sugars seem to be ideal ligands for obtaining prodrugs targeted at receptors overexpressed in cancer cells. They can also be ligands modifying polymers' drug carriers. Furthermore, polysaccharides can act as selective nanocarriers for numerous chemotherapeutics. The proof of this thesis is the huge number of papers devoted to their use for modification or targeted transport of anticancer compounds. In this work, selected examples of broad-defined sugars application for improving the properties of both already-used drugs and substances exhibiting anticancer activity are presented.
Collapse
Affiliation(s)
- Gabriela Pastuch-Gawołek
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| | - Julia Szreder
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Monika Domińska
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Mateusz Pielok
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Piotr Cichy
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
| | - Mirosława Grymel
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Silesian University of Technology, B. Krzywoustego 4, 44-100 Gliwice, Poland
- Biotechnology Centre, Silesian University of Technology, B. Krzywoustego 8, 44-100 Gliwice, Poland
| |
Collapse
|
3
|
A kNGR Peptide-Tethered Lipid–Polymer Hybrid Nanocarrier-Based Synergistic Approach for Effective Tumor Therapy: Development, Characterization, Ex-Vivo, and In-Vivo Assessment. Pharmaceutics 2022; 14:pharmaceutics14071401. [PMID: 35890297 PMCID: PMC9320317 DOI: 10.3390/pharmaceutics14071401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
The present study aims to design, develop and characterize kNGR (Asn-Gly-Arg) peptide-conjugated lipid–polymer-based nanoparticles for the target-specific delivery of anticancer bioactive(s), i.e., Paclitaxel (PTX). The kNGR-PEG-DSPE conjugate was synthesized and characterized by using spectral analysis. The dual-targeted PLGA–lecithin–PEG core-shell nanoparticles (PLNs-kNGR-NPs) were synthesized using a modified nanoprecipitation process, and their physiological properties were determined. The results support that, compared to other NPs, PLNs-kNGR-NPs are highly cytotoxic, owing to higher apoptosis and intracellular uptake. The significance of rational nanoparticle design for synergistic treatment is shown by the higher tumor volume inhibition percentage rate (59.7%), compared to other designed formulations in Balb/c mice in the HT-1080 tumor-induced model. The overall results indicate that the PLNs-kNGR-NPs-based hybrid lipid–polymer nanoparticles present the highest therapeutic efficacy against solid tumor overexpressing the CD13 receptors.
Collapse
|
4
|
Wu J, Wang X, Li H, Qu M, Sun W, Yan X, Zhao Z, Li B. A hollow chitosan-coated PLGA microsphere to enhance drug delivery and anticancer efficiency. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
5
|
Pu DB, Guo SQ, Ni DX, Lin J, Gao JB, Li XN, Zhang RH, Li XL, Luo C, Chen SJ, Xiao WL. Spiroarborin, an ent-Clerodane Homodimer from Callicarpa arborea as an Inhibitor of the Eleven-Nineteen Leukemia (ENL) Protein by Targeting the YEATS Domain. JOURNAL OF NATURAL PRODUCTS 2022; 85:317-326. [PMID: 35029993 DOI: 10.1021/acs.jnatprod.1c00775] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
A spiro ent-clerodane homodimer with a rare 6/6/6/6/6-fused pentacyclic scaffold, spiroarborin (1), together with four new monomeric analogues (2-5), were isolated from Callicarpa arborea. Their structures were elucidated by comprehensive spectroscopic data analysis, quantum-chemical calculations, and X-ray diffraction. A plausible biosynthetic pathway of 1 was proposed, and a biomimetic synthesis of its derivative was accomplished. Compound 1 showed a potent inhibitory effect by directly binding to the YEATS domain of the 11-19 leukemia (ENL) protein with an IC50 value of 7.3 μM. This gave a KD value of 5.0 μM, as recorded by a surface plasmon resonance binding assay.
Collapse
Affiliation(s)
- De-Bing Pu
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China
| | - Si-Qi Guo
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
- School of Pharmacy, Nanchang University, Nanchang 330006, People's Republic of China
| | - Dong-Xuan Ni
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China
| | - Jing Lin
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China
| | - Jun-Bo Gao
- State Key Laboratory of CAD&CG, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xiao-Ning Li
- School of Pharmaceutical Sciences, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Rui-Han Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China
| | - Cheng Luo
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Shi-Jie Chen
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resources, Ministry of Education, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, Yunnan University, Kunming 650091, People's Republic of China
| |
Collapse
|
6
|
Nunes M, Silva PMA, Coelho R, Pinto C, Resende A, Bousbaa H, Almeida GM, Ricardo S. Generation of Two Paclitaxel-Resistant High-Grade Serous Carcinoma Cell Lines With Increased Expression of P-Glycoprotein. Front Oncol 2021; 11:752127. [PMID: 34745981 PMCID: PMC8566917 DOI: 10.3389/fonc.2021.752127] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Debulking surgery followed by chemotherapy are the standard of care for high-grade serous carcinoma. After an initial good response to treatment, the majority of patients relapse with a chemoresistant profile, leading to a poor overall survival. Chemotherapy regimens used in high-grade serous carcinomas are based in a combination of classical chemotherapeutic drugs, namely, Carboplatin and Paclitaxel. The mechanisms underlying drug resistance and new drug discovery are crucial to improve patients’ survival. To uncover the molecular mechanisms of chemoresistance and test drugs capable of overcoming this resistant profile, it is fundamental to use good cellular models capable of mimicking the chemoresistant disease. Herein, we established two high-grade serous carcinoma cell lines with intrinsic resistance to Carboplatin and induced Paclitaxel resistance (OVCAR8 PTX R C and OVCAR8 PTX R P) derived from the OVCAR8 cell line. These two chemoresistant cell line variants acquired an enhanced resistance to Paclitaxel-induced cell death by increasing the drug efflux capacity, and this resistance was stable in long-term culture and following freeze/thaw cycles. The mechanism underlying Paclitaxel resistance resides in a significant increase in P-glycoprotein expression and, when this drug efflux pump was blocked with Verapamil, cells re-acquired Paclitaxel sensitivity. We generated two high-grade serous carcinoma cell lines, with a double-chemoresistant (Carboplatin and Paclitaxel) phenotype that mimics the majority of tumor recurrences in ovarian cancer context. This robust tool is suitable for preliminary drug testing towards the development of therapeutic strategies to overcome chemoresistance.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto/Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Patrícia M A Silva
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Gandra, Portugal.,TOXRUN, Toxicology Research Unit, University Institute of Health Sciences, Advanced Polytechnic and University Cooperative (CESPU), Gandra, Portugal
| | - Ricardo Coelho
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Carla Pinto
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Gandra, Portugal.,Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Porto, Portugal
| | - Albina Resende
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Gandra, Portugal.,Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Porto, Portugal
| | - Hassan Bousbaa
- CESPU, Institute of Research and Advanced Training in Health Sciences and Technologies (IINFACTS), Gandra, Portugal
| | - Gabriela M Almeida
- Expression Regulation in Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto/Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal.,Faculty of Medicine from University of Porto (FMUP), Porto, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto/Institute of Molecular Pathology and Immunology of the University of Porto (Ipatimup), Porto, Portugal.,TOXRUN, Toxicology Research Unit, University Institute of Health Sciences, Advanced Polytechnic and University Cooperative (CESPU), Gandra, Portugal.,Faculty of Medicine from University of Porto (FMUP), Porto, Portugal
| |
Collapse
|
7
|
Abstract
The author describes his 60-year career in studying the chemistry of natural products, which includes structural, synthetic, and biosynthetic studies of natural products ranging from insect pigments, antibiotics, and fecal mutagens to taxol and other anticancer natural products as well as antimalarial natural products. One of the compounds discussed, napabucasin, is now an anticancer drug in phase III clinical trials.
Collapse
Affiliation(s)
- David G I Kingston
- Department of Chemistry and the Virginia Tech Center for Drug Discovery, M/C 0212, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
8
|
Small interfering RNA (siRNA) to target genes and molecular pathways in glioblastoma therapy: Current status with an emphasis on delivery systems. Life Sci 2021; 275:119368. [PMID: 33741417 DOI: 10.1016/j.lfs.2021.119368] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 02/08/2023]
Abstract
Glioblastoma multiforme (GBM) is one of the worst brain tumors arising from glial cells, causing many deaths annually. Surgery, chemotherapy, radiotherapy and immunotherapy are used for GBM treatment. However, GBM is still an incurable disease, and new approaches are required for its successful treatment. Because mutations and amplifications occurring in several genes are responsible for the progression and aggressive behavior of GBM cells, genetic approaches are of great importance in its treatment. Small interfering RNA (siRNA) is a new emerging tool to silence the genes responsible for disease progression, particularly cancer. SiRNA can be used for GBM treatment by down-regulating genes such as VEGF, STAT3, ELTD1 or EGFR. Furthermore, the use of siRNA can promote the chemosensitivity of GBM cells. However, the efficiency of siRNA in GBM is limited via its degradation by enzymes, and its off-targeting effects. SiRNA-loaded carriers, especially nanovehicles that are ligand-functionalized by CXCR4 or angiopep-2, can be used for the protection and targeted delivery of siRNA. Nanostructures can provide a platform for co-delivery of siRNA plus anti-tumor drugs as another benefit. The prepared nanovehicles should be stable and biocompatible in order to be tested in human studies.
Collapse
|
9
|
Atanasov AG, Zotchev SB, Dirsch VM, Supuran CT. Natural products in drug discovery: advances and opportunities. Nat Rev Drug Discov 2021; 20:200-216. [PMID: 33510482 PMCID: PMC7841765 DOI: 10.1038/s41573-020-00114-z] [Citation(s) in RCA: 1926] [Impact Index Per Article: 642.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 02/07/2023]
Abstract
Natural products and their structural analogues have historically made a major contribution to pharmacotherapy, especially for cancer and infectious diseases. Nevertheless, natural products also present challenges for drug discovery, such as technical barriers to screening, isolation, characterization and optimization, which contributed to a decline in their pursuit by the pharmaceutical industry from the 1990s onwards. In recent years, several technological and scientific developments - including improved analytical tools, genome mining and engineering strategies, and microbial culturing advances - are addressing such challenges and opening up new opportunities. Consequently, interest in natural products as drug leads is being revitalized, particularly for tackling antimicrobial resistance. Here, we summarize recent technological developments that are enabling natural product-based drug discovery, highlight selected applications and discuss key opportunities.
Collapse
Affiliation(s)
- Atanas G Atanasov
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland.
- Department of Pharmacognosy, University of Vienna, Vienna, Austria.
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Vienna, Austria.
| | - Sergey B Zotchev
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Verena M Dirsch
- Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Claudiu T Supuran
- Università degli Studi di Firenze, NEUROFARBA Dept, Sezione di Scienze Farmaceutiche, Florence, Italy.
| |
Collapse
|
10
|
Nada H, Elkamhawy A, Lee K. Structure Activity Relationship of Key Heterocyclic Anti-Angiogenic Leads of Promising Potential in the Fight against Cancer. Molecules 2021; 26:molecules26030553. [PMID: 33494492 PMCID: PMC7865909 DOI: 10.3390/molecules26030553] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/13/2021] [Accepted: 01/18/2021] [Indexed: 12/11/2022] Open
Abstract
Pathological angiogenesis is a hallmark of cancer; accordingly, a number of anticancer FDA-approved drugs act by inhibiting angiogenesis via different mechanisms. However, the development process of the most potent anti-angiogenics has met various hurdles including redundancy, multiplicity, and development of compensatory mechanisms by which blood vessels are remodeled. Moreover, identification of broad-spectrum anti-angiogenesis targets is proved to be required to enhance the efficacy of the anti-angiogenesis drugs. In this perspective, a proper understanding of the structure activity relationship (SAR) of the recent anti-angiogenics is required. Various anti-angiogenic classes have been developed over the years; among them, the heterocyclic organic compounds come to the fore as the most promising, with several drugs approved by the FDA. In this review, we discuss the structure–activity relationship of some promising potent heterocyclic anti-angiogenic leads. For each lead, a molecular modelling was also carried out in order to correlate its SAR and specificity to the active site. Furthermore, an in silico pharmacokinetics study for some representative leads was presented. Summarizing, new insights for further improvement for each lead have been reviewed.
Collapse
Affiliation(s)
- Hossam Nada
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
| | - Ahmed Elkamhawy
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Korea
| |
Collapse
|
11
|
Tang Z, Tang N, Jiang S, Bai Y, Guan C, Zhang W, Fan S, Huang Y, Lin H, Ying Y. The Chemosensitizing Role of Metformin in Anti-Cancer Therapy. Anticancer Agents Med Chem 2021; 21:949-962. [PMID: 32951587 DOI: 10.2174/1871520620666200918102642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/23/2020] [Accepted: 08/08/2020] [Indexed: 11/22/2022]
Abstract
Chemoresistance, which leads to the failure of chemotherapy and further tumor recurrence, presents the largest hurdle for the success of anti-cancer therapy. In recent years, metformin, a widely used first-line antidiabetic drug, has attracted increasing attention for its anti-cancer effects. A growing body of evidence indicates that metformin can sensitize tumor responses to different chemotherapeutic drugs, such as hormone modulating drugs, anti-metabolite drugs, antibiotics, and DNA-damaging drugs via selective targeting of Cancer Stem Cells (CSCs), improving the hypoxic microenvironment, and by suppressing tumor metastasis and inflammation. In addition, metformin may regulate metabolic programming, induce apoptosis, reverse Epithelial to Mesenchymal Transition (EMT), and Multidrug Resistance (MDR). In this review, we summarize the chemosensitization effects of metformin and focus primarily on its molecular mechanisms in enhancing the sensitivity of multiple chemotherapeutic drugs, through targeting of mTOR, ERK/P70S6K, NF-κB/HIF-1 α, and Mitogen- Activated Protein Kinase (MAPK) signaling pathways, as well as by down-regulating the expression of CSC genes and Pyruvate Kinase isoenzyme M2 (PKM2). Through a comprehensive understanding of the molecular mechanisms of chemosensitization provided in this review, the rationale for the use of metformin in clinical combination medications can be more systematically and thoroughly explored for wider adoption against numerous cancer types.>.
Collapse
Affiliation(s)
- Zhimin Tang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Nan Tang
- Nanchang Joint Program, Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Shanshan Jiang
- Institute of Hematological Research, Shanxi Provincial People's Hospital, Xian 710000, China
| | - Yangjinming Bai
- Nanchang Joint Program, Queen Mary School, Nanchang University, Nanchang 330006, China
| | - Chenxi Guan
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Wansi Zhang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Shipan Fan
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou 510005, China
| | - Yonghong Huang
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Hui Lin
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Ying Ying
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology and Department of Pathophysiology, Schools of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| |
Collapse
|
12
|
Ko G, Kim T, Ko E, Park D, Lee Y. Synergistic Enhancement of Paclitaxel-induced Inhibition of Cell Growth by Metformin in Melanoma Cells. Dev Reprod 2019; 23:119-128. [PMID: 31321352 PMCID: PMC6635613 DOI: 10.12717/dr.2019.23.2.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/27/2019] [Accepted: 06/08/2019] [Indexed: 11/25/2022]
Abstract
Melanoma is one of the most aggressive and treatment-resistant malignancies.
Antidiabetic drug metformin has been reported to inhibit cell proliferation and
metastasis in many cancers, including melanoma. Metformin suppresses the
mammalian target of rapamycin (mTOR) and our previous study showed that it also
inhibits the activity of extracellular signal-regulated kinase (ERK). Paclitaxel
is currently prescribed for treatment of melanoma. However, paclitaxel induced
the activation of ERK/mitogen-activated protein kinase (MAPK) pathway, a cell
signaling pathway implicated in cell survival and proliferation. Therefore, we
reasoned that combined treatment of paclitaxel with metformin could be more
effective in the suppression of cell proliferation than treatment of paclitaxel
alone. Here, we investigated the combinatory effect of paclitaxel and metformin
on the cell survival in SK-MEL-28 melanoma cell line. Our study shows that the
combination of paclitaxel and metformin has synergistic effect on cell survival
and suppresses the expression of proteins involved in cancer metastasis. These
findings suggest that the combination of paclitaxel and metformin can be a
possible therapeutic option for treatment of melanoma.
Collapse
Affiliation(s)
- Gihyun Ko
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Taehyung Kim
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Eunjeong Ko
- Dept. of Medicine, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Deokbae Park
- Histology, Jeju National University School of Medicine, Jeju 63243, Korea
| | - Youngki Lee
- Histology, Jeju National University School of Medicine, Jeju 63243, Korea
| |
Collapse
|
13
|
Choules MP, Klein LL, Lankin DC, McAlpine JB, Cho SH, Cheng J, Lee H, Suh JW, Jaki BU, Franzblau SG, Pauli GF. Residual Complexity Does Impact Organic Chemistry and Drug Discovery: The Case of Rufomyazine and Rufomycin. J Org Chem 2018; 83:6664-6672. [PMID: 29792329 PMCID: PMC6006449 DOI: 10.1021/acs.joc.8b00988] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
![]()
Residual complexity (RC) involves
the impact of subtle but critical structural and biological features on drug lead validation, including unexplained effects related to unidentified impurities. RC commonly plagues drug discovery efforts due to the inherent imperfections
of chromatographic separation methods. The new diketopiperazine, rufomyazine
(6), and the previously known antibiotic, rufomycin (7), represent a prototypical case of RC that (almost) resulted
in the misassignment of biological activity. The case exemplifies
that impurities well below the natural abundance of 13C
(1.1%) can be highly relevant and calls for advanced analytical characterization
of drug leads with extended molar dynamic ranges of >1:1,000 using
qNMR and LC-MS. Isolated from an actinomycete strain, 6 was originally found to be active against Mycobacterium
tuberculosis with a minimum inhibitory concentration (MIC)
of 2 μg/mL and high selectivity. As a part of lead validation,
the dipeptide was synthesized and surprisingly found to be inactive.
The initially observed activity was eventually attributed to
a very minor contamination (0.24% [m/m]) with a highly active cyclic
peptide (MIC ∼ 0.02 μM), subsequently identified as an
analogue of 7. This study illustrates the serious implications
RC can exert on organic chemistry and drug discovery, and what efforts
are vital to improve lead validation and efficiency, especially in
NP-related drug discovery programs.
Collapse
|
14
|
Ojima I, Wang X, Jing Y, Wang C. Quest for Efficacious Next-Generation Taxoid Anticancer Agents and Their Tumor-Targeted Delivery. JOURNAL OF NATURAL PRODUCTS 2018; 81:703-721. [PMID: 29468872 PMCID: PMC5869464 DOI: 10.1021/acs.jnatprod.7b01012] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Indexed: 05/28/2023]
Abstract
Paclitaxel and docetaxel are among the most widely used chemotherapeutic drugs against various types of cancer. However, these drugs cause undesirable side effects as well as drug resistance. Therefore, it is essential to develop next-generation taxoid anticancer agents with better pharmacological properties and improved activity especially against drug-resistant and metastatic cancers. The SAR studies by the authors have led to the development of numerous highly potent novel second- and third-generation taxoids with systematic modifications at the C-2, C-10, and C-3' positions. The third-generation taxoids showed virtually no difference in potency against drug-resistant and drug-sensitive cell lines. Some of the next-generation taxoids also exhibited excellent potency against cancer stem cells. This account summarizes concisely investigations into taxoids over 25 years based on a strong quest for the discovery and development of efficacious next-generation taxoids. Discussed herein are SAR studies on different types of taxoids, a common pharmacophore proposal for microtubule-stabilizing anticancer agents and its interesting history, the identification of the paclitaxel binding site and its bioactive conformation, characteristics of the next-generation taxoids in cancer cell biology, including new aspects of their mechanism of action, and the highly efficacious tumor-targeted drug delivery of potent next-generation taxoids.
Collapse
Affiliation(s)
- Iwao Ojima
- Department of Chemistry and Institute
of Chemical Biology & Drug Discovery, Stony Brook University−State University of New York, Stony Brook, New York 11794-3400, United States
| | - Xin Wang
- Department of Chemistry and Institute
of Chemical Biology & Drug Discovery, Stony Brook University−State University of New York, Stony Brook, New York 11794-3400, United States
| | - Yunrong Jing
- Department of Chemistry and Institute
of Chemical Biology & Drug Discovery, Stony Brook University−State University of New York, Stony Brook, New York 11794-3400, United States
| | - Changwei Wang
- Department of Chemistry and Institute
of Chemical Biology & Drug Discovery, Stony Brook University−State University of New York, Stony Brook, New York 11794-3400, United States
| |
Collapse
|
15
|
Meng Z, Lv Q, Lu J, Yao H, Lv X, Jiang F, Lu A, Zhang G. Prodrug Strategies for Paclitaxel. Int J Mol Sci 2016; 17:E796. [PMID: 27223283 PMCID: PMC4881612 DOI: 10.3390/ijms17050796] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/04/2016] [Accepted: 05/11/2016] [Indexed: 01/08/2023] Open
Abstract
Paclitaxel is an anti-tumor agent with remarkable anti-tumor activity and wide clinical uses. However, it is also faced with various challenges especially for its poor water solubility and low selectivity for the target. To overcome these disadvantages of paclitaxel, approaches using small molecule modifications and macromolecule modifications have been developed by many research groups from all over the world. In this review, we discuss the different strategies especially prodrug strategies that are currently used to make paclitaxel more effective.
Collapse
Affiliation(s)
- Ziyuan Meng
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| | - Quanxia Lv
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| | - Jun Lu
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Houzong Yao
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Xiaoqing Lv
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| | - Feng Jiang
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
- The State Key Laboratory Base of Novel Functional Materials and Preparation Science, Faculty of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| | - Aiping Lu
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| | - Ge Zhang
- Institution for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Research Group of Precision Medicine and Innovative Drug, HKBU (Hong Kong Baptist University) (Haimen) Institute of Science and Technology, Haimen 226100, China.
| |
Collapse
|
16
|
Pérez-Estrada S, Sayar N, Granja JR. Towards taxane analogues synthesis by dienyne ring closing metathesis. Org Chem Front 2016. [DOI: 10.1039/c6qo00321d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The synthesis of highly functionalized 16,17,18-trinortaxane analogues based on a dienyne cyclization is described.
Collapse
Affiliation(s)
- S. Pérez-Estrada
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica
- Universidade de Santiago de Compostela
- 15782 Santiago de Compostela
- Spain
| | - N. Sayar
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica
- Universidade de Santiago de Compostela
- 15782 Santiago de Compostela
- Spain
| | - J. R. Granja
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CIQUS) and Departamento de Química Orgánica
- Universidade de Santiago de Compostela
- 15782 Santiago de Compostela
- Spain
| |
Collapse
|
17
|
Ojima I, Kumar K, Awasthi D, Vineberg JG. Drug discovery targeting cell division proteins, microtubules and FtsZ. Bioorg Med Chem 2014; 22:5060-77. [PMID: 24680057 PMCID: PMC4156572 DOI: 10.1016/j.bmc.2014.02.036] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 01/25/2014] [Accepted: 02/18/2014] [Indexed: 12/16/2022]
Abstract
Eukaryotic cell division or cytokinesis has been a major target for anticancer drug discovery. After the huge success of paclitaxel and docetaxel, microtubule-stabilizing agents (MSAs) appear to have gained a premier status in the discovery of next-generation anticancer agents. However, the drug resistance caused by MDR, point mutations, and overexpression of tubulin subtypes, etc., is a serious issue associated with these agents. Accordingly, the discovery and development of new-generation MSAs that can obviate various drug resistances has a significant meaning. In sharp contrast, prokaryotic cell division has been largely unexploited for the discovery and development of antibacterial drugs. However, recent studies on the mechanism of bacterial cytokinesis revealed that the most abundant and highly conserved cell division protein, FtsZ, would be an excellent new target for the drug discovery of next-generation antibacterial agents that can circumvent drug-resistances to the commonly used drugs for tuberculosis, MRSA and other infections. This review describes an account of our research on these two fronts in drug discovery, targeting eukaryotic as well as prokaryotic cell division.
Collapse
Affiliation(s)
- Iwao Ojima
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA; Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY 11794-3400, USA.
| | - Kunal Kumar
- Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Divya Awasthi
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| | - Jacob G Vineberg
- Department of Chemistry, Stony Brook University, Stony Brook, NY 11794-3400, USA
| |
Collapse
|
18
|
Kingston DGI, Snyder JP. The quest for a simple bioactive analog of paclitaxel as a potential anticancer agent. Acc Chem Res 2014; 47:2682-91. [PMID: 25052294 PMCID: PMC4139185 DOI: 10.1021/ar500203h] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Paclitaxel (PTX), introduced into the clinic in 1991, has revealed itself as an effective antimicrotubule drug for treatment of a range of otherwise intractable cancers. Along with docetaxel (DTX) and in combination with other agents such as cisplatin, it has proven to be a first-line therapy. Unfortunately, PTX and DTX carry severe liabilities such as debilitating side effects, rapid onset of resistance, and rather complex molecular structures offering substantial challenges to ease of synthetic manipulation. Consequently, the past 15 years has witnessed many efforts to synthesize and test highly modified analogs based on intuitive structural similarity relationships with the PTX molecular skeleton, as well as efforts to mimic the conformational profile of the ligand observed in the macromolecular tubulin-PTX complex. Highly successful improvements in potency, up to 50-fold increases in IC50, have been achieved by constructing bridges between distal centers in PTX that imitate the conformer of the electron crystallographic binding pose. Much less successful have been numerous attempts to truncate PTX by replacing the baccatin core with simpler moieties to achieve PTX-like potencies and applying a wide range of flexible synthesis-based chemistries. Reported efforts, characterized by a fascinating array of baccatin substitutes, have failed to surpass the bioactivities of PTX in both microtubule disassembly assays and cytotoxicity measurements against a range of cell types. Most of the structures retain the main elements of the PTX C13 side chain, while seeking a smaller rigid bicycle as a baccatin replacement adorned with substituents to mimic the C2 benzoyl moiety and the oxetane ring. We surmise that past studies have been handicapped by solubility and membrane permeability issues, but primarily by the existence of an expansive taxane binding pocket and the discrepancy in molecular size between PTX and the pruned analogs. A number of these molecules offer molecular volumes 50-60% that of PTX, fewer contacts with the tubulin protein, severe mismatches with the PTX pharmacophore, lessened capacity to dispel binding site waters contributing to ΔGbind, and unanticipated binding poses. The latter is a critical drawback if molecular designs of simpler PTX structures are based on a perceived or known PTX binding conformation. We conclude that design and synthesis of a highly cytotoxic tubulin-assembly agent based on the paclitaxel pharmacophore remains an unsolved challenge, but one that can be overcome by focus on the architecture of the taxane binding site independent of the effective, but not unique, hand-in-glove match represented by the PTX-tubulin complex.
Collapse
Affiliation(s)
- David G. I. Kingston
- Department
of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia, 24061 United States
| | - James P. Snyder
- Department
of Chemistry, Emory University, Atlanta, Georgia, 30322 United States
| |
Collapse
|
19
|
Grimmer C, Moore TW, Padwa A, Prussia A, Wells G, Wu S, Sun A, Snyder JP. Antiviral atropisomers: conformational energy surfaces by NMR for host-directed myxovirus blockers. J Chem Inf Model 2014; 54:2214-23. [PMID: 25058809 DOI: 10.1021/ci500204j] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Biologically active organic molecules characterized by a high single bond torsional barrier generate isolable isomers (atropisomers) and offer a unique stereochemical component to the design of selective therapeutic agents. The present work presents a nanomolar active inhibitor of myxoviruses, which most likely acts by blocking one or more cellular host proteins but also, serendipitously, exhibits axial chirality with an energy barrier of ΔG((++)) ≥30 kcal/mol. The latter has been probed by variable temperature NMR and microwave irradiation and by high level DFT transition state analysis and force field calculations. Full conformational profiles of the corresponding (aR,S) and (aS,S) atropisomers at ambient temperature were derived by conformer deconvolution with NAMFIS (NMR Analysis by Molecular Flexibility In Solution) methodology to generate seven and eight individual conformations, each assigned a % population. An accurate evaluation of a key torsion angle at the center of the molecules associated with a (3)JC-S-C-H coupling constant was obtained by mapping the S-C bond rotation with the MPW1PW91/6-31G-d,p DFT method followed by fitting the resulting dihedral angles and J-values to a Karplus expression. Accordingly, we have developed a complete conformational profile of diastereomeric atropisomers consistent with both high and low rotational barriers. We expect this assessment to assist the rationalization of the selectivity of the two (aR,S) and (aS,S) forms against host proteins, while offering insights into their divergent toxicity behavior.
Collapse
Affiliation(s)
- Craig Grimmer
- School of Chemistry and Physics, University of KwaZulu-Natal , Pietermaritzburg, South Africa
| | | | | | | | | | | | | | | |
Collapse
|
20
|
SONG RONGFENG, LI XIAOJUN, CHENG XIAOLIANG, FU AIRONG, WANG YANHUA, FENG YANJUN, XIONG YAN. Paclitaxel-loaded trimethyl chitosan-based polymeric nanoparticle for the effective treatment of gastroenteric tumors. Oncol Rep 2014; 32:1481-8. [DOI: 10.3892/or.2014.3344] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 06/25/2014] [Indexed: 11/05/2022] Open
|
21
|
Ojima I, Kamath A, Seitz JD. Taxol, Taxoids, and Related Taxanes. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2014. [DOI: 10.1002/9783527676545.ch04] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
22
|
Coderch C, Tang Y, Klett J, Zhang SE, Ma YT, Shaorong W, Matesanz R, Pera B, Canales A, Jiménez-Barbero J, Morreale A, Díaz JF, Fang WS, Gago F. A structure-based design of new C2- and C13-substituted taxanes: tubulin binding affinities and extended quantitative structure-activity relationships using comparative binding energy (COMBINE) analysis. Org Biomol Chem 2013; 11:3046-56. [PMID: 23532250 DOI: 10.1039/c3ob40407b] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ten novel taxanes bearing modifications at the C2 and C13 positions of the baccatin core have been synthesized and their binding affinities for mammalian tubulin have been experimentally measured. The design strategy was guided by (i) calculation of interaction energy maps with carbon, nitrogen and oxygen probes within the taxane-binding site of β-tubulin, and (ii) the prospective use of a structure-based QSAR (COMBINE) model derived from an earlier series comprising 47 congeneric taxanes. The tubulin-binding affinity displayed by one of the new compounds (CTX63) proved to be higher than that of docetaxel, and an updated COMBINE model provided a good correlation between the experimental binding free energies and a set of weighted residue-based ligand-receptor interaction energies for 54 out of the 57 compounds studied. The remaining three outliers from the original training series have in common a large unfavourable entropic contribution to the binding free energy that we attribute to taxane preorganization in aqueous solution in a conformation different from that compatible with tubulin binding. Support for this proposal was obtained from solution NMR experiments and molecular dynamics simulations in explicit water. Our results shed additional light on the determinants of tubulin-binding affinity for this important class of antitumour agents and pave the way for further rational structural modifications.
Collapse
Affiliation(s)
- Claire Coderch
- Área de Farmacología, Departamento de Ciencias Biomédicas, Universidad de Alcalá, E-28871 Alcalá de Henares, Unidad Asociada al Instituto de Química Médica del CSIC, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Díaz JF, Andreu JM, Jiménez-Barbero J. The interaction of microtubules with stabilizers characterized at biochemical and structural levels. Top Curr Chem (Cham) 2013; 286:121-49. [PMID: 23563612 DOI: 10.1007/128_2008_12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Since the discovery of paclitaxel and its peculiar mechanism of cytotoxicity, which has made it and its analogues widely used antitumour drugs, great effort has been made to understand the way they produce their effect in microtubules and to find other products that share this effect without the undesired side effects of low solubility and development of multidrug resistance by tumour cells. This chapter reviews the actual knowledge about the biochemical and structural mechanisms of microtubule stabilization by microtubule stabilizing agents, and illustrates the way paclitaxel and its biomimetics induce microtubule assembly, the thermodynamics of their binding, the way they reach their binding site and the conformation they have when bound.
Collapse
Affiliation(s)
- J F Díaz
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040, Madrid, Spain,
| | | | | |
Collapse
|
24
|
Hao XD, Xie C, Hao YP, Chang J, Sun X. (3 R,4 S)-1-(4-Methoxyphenyl)-2-oxo-4-(3-vinylphenyl)azetidin-3-yl acetate. Acta Crystallogr Sect E Struct Rep Online 2013; 69:o601. [PMID: 23634128 PMCID: PMC3629641 DOI: 10.1107/s1600536813007897] [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: 01/24/2013] [Accepted: 03/21/2013] [Indexed: 11/30/2022]
Abstract
In the title compound, C20H19NO4, the absolute configuration (3R,4S) for the two chiral centres of the molecule has been determined.
Collapse
|
25
|
Oh MH, Kim JS, Lee JY, Park TG, Nam YS. Radio-opaque theranostic nanoemulsions with synergistic anti-cancer activity of paclitaxel and Bcl-2 siRNA. RSC Adv 2013. [DOI: 10.1039/c3ra40883c] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
26
|
Kingston DGI, Tamarkin L, Paciotti GF. Conformationally Constrained and Nanoparticle Targeted Paclitaxels. PURE APPL CHEM 2012; 84:1455-1467. [PMID: 25598555 PMCID: PMC4295212 DOI: 10.1351/pac-con-11-09-02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Paclitaxel (Taxol®) is one of the most important anticancer agents developed over the last 30 years. Its primary mechanism of action is by interaction with the cellular protein tubulin, causing irreversible polymerization to microtubules. A detailed knowledge of this crucial interaction is thus of paramount importance in the design and development of highly potent analogs and also for the potential development of "non-taxane" tubulin polymerization agents. This review briefly describes the discovery and development of taxol, and then describes our work on delineating the tubulin-binding conformation of paclitaxel by a combination of REDOR NMR and molecular modeling. The resulting "T-taxol" conformation was validated by the synthesis of conformationally constrained paclitaxel analogs, which had bioactivities up to twenty-fold higher than those of paclitaxel. The review concludes with recent work on the development of a gold nanoparticle derivative of paclitaxel. This delivery method has the potential to lower the dosage of paclitaxel needed while maintaining or increasing its effectiveness, thus significantly improving the benefits of this important chemotherapeutic agent.
Collapse
Affiliation(s)
| | - Lawrence Tamarkin
- CytImmune Sciences, Inc. 105010 Broschart Rd, Rockville, MD 20850, USA
| | - Giulio F Paciotti
- CytImmune Sciences, Inc. 105010 Broschart Rd, Rockville, MD 20850, USA
| |
Collapse
|
27
|
Contini A, Cappelletti G, Cartelli D, Fontana G, Gelmi ML. Molecular dynamics and tubulin polymerization kinetics study on 1,14-heterofused taxanes: evidence of stabilization of the tubulin head-to-tail dimer–dimer interaction. MOLECULAR BIOSYSTEMS 2012; 8:3254-61. [DOI: 10.1039/c2mb25326g] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
28
|
Zhao J, Bane S, Snyder JP, Hu H, Mukherjee K, Slebodnick C, Kingston DGI. Design and synthesis of simplified taxol analogs based on the T-Taxol bioactive conformation. Bioorg Med Chem 2011; 19:7664-78. [PMID: 22071526 PMCID: PMC3225578 DOI: 10.1016/j.bmc.2011.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 09/30/2011] [Accepted: 10/05/2011] [Indexed: 10/16/2022]
Abstract
A series of compounds designed to adopt a conformation similar to the tubulin-binding T-Taxol conformation of the anticancer drug paclitaxel has been synthesized. Both the internally bridged analogs 37-39, 41 and the open-chain analogs 27-29 and 43 were prepared. The bridged analogs 37-39 and 41 were synthesized by Grubbs' metatheses of compounds 30-32 and 33, which, in turn, were prepared by coupling β-lactams 24-26 with alcohols 22 and 23. Both the bridged and the open-chain analogs showed moderate to good cytotoxicity.
Collapse
Affiliation(s)
- Jielu Zhao
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Susan Bane
- Department of Chemistry, State University of New York at Binghamton, Binghamton, NY 13902
| | - James P. Snyder
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, GA 30322
| | - Haipeng Hu
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, GA 30322
| | - Kamalika Mukherjee
- Department of Chemistry, State University of New York at Binghamton, Binghamton, NY 13902
| | - Carla Slebodnick
- Department of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, USA
| | | |
Collapse
|
29
|
Ji X, Gao Y, Chen L, Zhang Z, Deng Y, Li Y. Nanohybrid systems of non-ionic surfactant inserting liposomes loading paclitaxel for reversal of multidrug resistance. Int J Pharm 2011; 422:390-7. [PMID: 22001531 DOI: 10.1016/j.ijpharm.2011.10.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Revised: 08/31/2011] [Accepted: 10/02/2011] [Indexed: 11/19/2022]
Abstract
Three new nanohybrid systems of non-ionic surfactant inserting liposomes loading paclitaxel (PTX) (NLPs) were prepared to overcome multidrug resistance (MDR) in PTX-resistance human lung cancer cell line. Three non-ionic surfactants, Solutol HS 15 (HS-15), pluronic F68 (PF-68) and cremophor EL (CrEL) were inserted into liposomes by film hydration method to form NLPs with an average size of around 110, 180 and 110 nm, respectively. There was an obvious increase of rhodamin 123 (Rh123) accumulation in A549/T cells after treated with nanohybrid systems loading Rh123 (NLRs) when compared with free Rh123 or liposomes loading Rh123 without surfactants (LRs), which indicated the significant inhibition effects of NLRs on drug efflux. The P-gp detection and ATP determination demonstrated that BNLs could not only interfere P-gp expression on the membrane of drug resistant cells, but also decrease ATP level in the cells. The cytotoxicity of NLPs against A549/T cells was higher than PTX loaded liposomes without surfactants (LPs), and the best result was achieved after treated with NLPs2. The apoptotic assay and the cell cycle analysis showed that NLPs could induce more apoptotic cells in drug resistant cells when compared with LPs. These results suggested that NLPs could overcome MDR by combination of drug delivery, P-gp inhibition and ATP depletion, and showed potential for treatment of MDR.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Adenosine Triphosphate/metabolism
- Antineoplastic Agents, Phytogenic/chemistry
- Antineoplastic Agents, Phytogenic/metabolism
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis/drug effects
- Cell Cycle/drug effects
- Cell Line, Tumor
- Chemistry, Pharmaceutical
- Dose-Response Relationship, Drug
- Drug Compounding
- Drug Resistance, Multiple
- Drug Resistance, Neoplasm
- Glycerol/analogs & derivatives
- Glycerol/chemistry
- Humans
- Lipids/chemistry
- Liposomes
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Nanotechnology
- Paclitaxel/chemistry
- Paclitaxel/metabolism
- Paclitaxel/pharmacology
- Particle Size
- Poloxamer/chemistry
- Polyethylene Glycols/chemistry
- Rhodamine 123/metabolism
- Solubility
- Stearic Acids/chemistry
- Surface-Active Agents/chemistry
- Technology, Pharmaceutical/methods
- Time Factors
Collapse
Affiliation(s)
- Xiufeng Ji
- Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai 201203, China
| | | | | | | | | | | |
Collapse
|
30
|
Wang Y, Bian F, Deng S, Shi Q, Ge M, Wang S, Zhang X, Xu S. The key residues of active sites on the catalytic fragment for paclitaxel interacting with poly (ADP-ribose) polymerase. J Biomol Struct Dyn 2011; 28:881-93. [PMID: 21469749 DOI: 10.1080/07391102.2011.10508615] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP) is regarded as a target protein for paclitaxel (PTX) to bind. An important issue is to identify the key residues as active sites for PTX interacting with PARP, which will help to understand the potential drug activity of PTX against cancer cells. Using docking method and MD simulation, we have constructed a refined structure of PTX docked on the catalytic function domain of PARP (PDB code: 1A26). The residues Glu327(988), Tyr246(907), Lys242(903), His165(826), Asp105(766), Gln102(763) and Gln98(759) in PARP are identified as potential sites involved in interaction with PTX according to binding energy (E(b)) between PTX and single residue calculated with B3LYP/6-31G(d,p). These residues form an active binding pocket located on the surface of the catalytic fragment, possibly interacting with the required groups of PTX leading to its activity against cancer cells. It is noted that most of the active sites make conatct with the "southern hemisphere" of PTX except for one residue, Tyr246(907), which interacts with the "northern hemisphere" of PTX. The conformation of PTX in complex with the catalytic fragment is observed as being T-shaped, similar to that complexed with β-tubulin. The total Eb of -269.9 kJ/mol represents the potent interaction between PTX and the catalytic fragment, implying that PTX can readily bind to the active pocket. The tight association of PTX with the catalytic fragment would inhibit PARP activation, suggesting a potential application of PTX as an effective antineoplastic agent.
Collapse
Affiliation(s)
- Yue Wang
- Key Laboratory of Education Ministry for Medicinal Chemistry of Natural Resource, College of Chemical Science and Technology, Yunnan University, Kunming 650091, PR China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Xu S, Chi S, Jin Y, Shi Q, Ge M, Wang S, Zhang X. Molecular dynamics simulation and density functional theory studies on the active pocket for the binding of paclitaxel to tubulin. J Mol Model 2011; 18:377-91. [PMID: 21537957 DOI: 10.1007/s00894-011-1083-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 04/05/2011] [Indexed: 12/29/2022]
Abstract
Paclitaxel (PTX) is used to treat various cancers, but it also causes serious side effects and resistance. To better design similar compounds with less toxicity and more activity against drug-resistant tumors, it is important to clearly understand the PTX-binding pocket formed by the key residues of active sites on β-tubulin. Using a docking method, molecular dynamics (MD) simulation and density functional theory (DFT), we identified some residues (such as Arg278, Asp26, Asp226, Glu22, Glu27, His229, Arg369, Lys218, Ser277 and Thr276) on β-tubulin that are the active sites responsible for interaction with PTX. Another two residues, Leu371 and Gly279, also likely serve as active sites. Most of these sites contact with the "southern hemisphere" of PTX; only one key residue interacts with the "northern hemisphere" of PTX. These key residues can be divided into four groups, which serve as active compositions in the formation of an active pocket for PTX binding to β-tubulin. This active binding pocket enables a very strong interaction (the strength is predicted to be in the range of -327.8 to -365.7 kJ mol(-1)) between β-tubulin and PTX, with various orientated conformations. This strong interaction means that PTX possesses a high level of activity against cancer cells, a result that is in good agreement with the clinical mechanism of PTX. The described PTX pocket and key active residues will be applied to probe the mechanism of tumor cells resistant to PTX, and to design novel analogs with superior properties.
Collapse
Affiliation(s)
- Sichuan Xu
- Key Laboratory of Education Ministry for Medicinal Chemistry of Natural Resource, College of Chemical Science and Technology, Yunnan University, Kunming, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
32
|
George J, Banik NL, Ray SK. Combination of taxol and Bcl-2 siRNA induces apoptosis in human glioblastoma cells and inhibits invasion, angiogenesis and tumour growth. J Cell Mol Med 2010; 13:4205-18. [PMID: 19473291 PMCID: PMC4496127 DOI: 10.1111/j.1582-4934.2008.00539.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Taxol is a powerful chemotherapeutic agent that binds to microtubules to prevent tumour cell division. However, a traditional high dose of taxol may also induce apoptosis in normal cells. The anti-apoptotic molecule Bcl-2 is up-regulated in tumour cells to prevent apoptosis. We designed this study to determine whether use of a low dose of taxol and anti-apoptotic Bcl-2 gene silencing would effectively induce apoptosis in human glioblastoma U251MG cells and also inhibit invasion, angiogenesis and intracranial as well as subcutaneous tumour growth. We treated the cells with either 100 nM taxol or transfected with a plasmid vector expressing Bcl-2 siRNA or both agents together for 72 h. Knockdown of Bcl-2 potentiated efficacy of taxol for cell death. Fluorescence-activated cell sorting analysis, double immunofluorescent staining and TUNEL assay demonstrated apoptosis in about 70% of the cells after treatment with the combination of taxol and Bcl-2 siRNA. In vitro Matrigel invasion assay demonstrated dramatic decrease in glioblastoma cell invasion and in vivo angiogenesis assay showed complete inhibition of neovascularization in athymic nude mice after treatment with the combination. Further, treatment with the combination of taxol and Bcl-2 siRNA caused suppression of intracranial tumour growth and subcutaneous solid tumour development. In conclusion, our results indicate that the combination of taxol and Bcl-2 siRNA effectively induces apoptosis and inhibits glioblastoma cell invasion, angiogenesis and intracranial as well as subcutaneous tumour growth. Therefore, the combination of a low dose of taxol and Bcl-2 siRNA is a promising therapeutic strategy for controlling the aggressive growth of human glioblastoma.
Collapse
Affiliation(s)
- Joseph George
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | | | | |
Collapse
|
33
|
Gassa F, Contini A, Fontana G, Pellegrino S, Gelmi ML. A Highly Diastereoselective Synthesis of α-Hydroxy-β-amino Acid Derivatives via a Lewis Acid Catalyzed Three-Component Condensation Reaction. J Org Chem 2010; 75:7099-106. [DOI: 10.1021/jo1011762] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Federico Gassa
- DISMAB, Sezione di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università di Milano, via Venezian 21, 20133 Milano, Italy
| | - Alessandro Contini
- DISMAB, Sezione di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università di Milano, via Venezian 21, 20133 Milano, Italy
| | | | - Sara Pellegrino
- DISMAB, Sezione di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università di Milano, via Venezian 21, 20133 Milano, Italy
| | - Maria Luisa Gelmi
- DISMAB, Sezione di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università di Milano, via Venezian 21, 20133 Milano, Italy
| |
Collapse
|
34
|
Prussia AJ, Yang Y, Geballe MT, Snyder JP. Cyclostreptin and microtubules: is a low-affinity binding site required? Chembiochem 2010; 11:101-9. [PMID: 19946930 DOI: 10.1002/cbic.200900538] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cyclostreptin (CS) is a recently discovered natural product with cytotoxic activity caused by microtubule stabilization. It is the only known microtubule-stabilizing agent (MSA) that covalently binds to tubulin. It also exhibits the fast-binding kinetics seen for other MSAs. Through careful peptide digestion and mass spectrometry analysis, Buey et al. found that two amino acids are labeled by CS: Asn228, near the known taxane-binding site, and Thr220, in the type I microtubule pore. This led Buey et al. to propose Thr220 resides at the site previously predicted to be a way station or low-affinity site. By using molecular dynamics simulations and structural considerations of the microtubule pore and tubulin dimer, we conclude that postulation of a low-affinity site is unnecessary to explain the available experimental data. An alternative explanation views the microtubule pore as a structural entity that presents a substantial kinetic barrier to ligand passage to the known taxane-binding site-an entry point to the microtubule lumen that becomes completely blocked if cyclostreptin is bound at Thr220. Simulations of the free dimer also suggest a common mechanism of microtubule stabilization for taxane site MSAs through their conformational effect on the M-loop. Such an effect explains the low tubulin polymerization caused by cyclostreptin in vitro despite its covalent attachment.
Collapse
Affiliation(s)
- Andrew J Prussia
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
35
|
Jogalekar AS, Kriel FH, Shi Q, Cornett B, Cicero D, Snyder JP. The discodermolide hairpin structure flows from conformationally stable modular motifs. J Med Chem 2010; 53:155-65. [PMID: 19894728 DOI: 10.1021/jm9015284] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
(+)-Discodermolide (DDM), a polyketide macrolide from marine sponge, is a potent microtubule assembly promoter. Reported solid-state, solution, and protein-bound DDM conformations reveal the unusual result that a common hairpin conformational motif exists in all three microenvironments. No other flexible microtubule binding agent exhibits such constancy of conformation. In the present study, we combine force-field conformational searches with NMR deconvolution in different solvents to compare DDM conformers with those observed in other environments. While several conformational families are perceived, the hairpin form dominates. The stability of this motif is dictated primarily by steric factors arising from repeated modular segments in DDM composed of the C(Me)-CHX-C(Me) fragment. Furthermore, docking protocols were utilized to probe the DDM binding mode in beta-tubulin. A previously suggested pose is substantiated (Pose-1), while an alternative (Pose-2) has been identified. SAR analysis for DDM analogues differentiates the two poses and suggests that Pose-2 is better able to accommodate the biodata.
Collapse
Affiliation(s)
- Ashutosh S Jogalekar
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, USA
| | | | | | | | | | | |
Collapse
|
36
|
Sun L, Geng X, Geney R, Li Y, Simmerling C, Li Z, Lauher JW, Xia S, Horwitz SB, Veith JM, Pera P, Bernacki RJ, Ojima I. Design, synthesis, and biological evaluation of novel C14-C3'BzN-linked macrocyclic taxoids. J Org Chem 2009; 73:9584-93. [PMID: 18975909 DOI: 10.1021/jo801713q] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Novel macrocyclic paclitaxel congeners were designed to mimic the bioactive conformation of paclitaxel. Computational analysis of the "REDOR-Taxol" structure revealed that this structure could be rigidified by connecting the C14 position of the baccatin moiety and the ortho position of C3'N-benzoyl group (C3'BzN), which are ca. 7.5 A apart, with a short linker (4-6 atoms). 7-TES-14beta-allyloxybaccatin III and (3R,4S)-1-(2-alkenylbenzoyl)-beta-lactams were selected as key components, and the Ojima-Holton coupling afforded the corresponding paclitaxel-dienes. The Ru-catalyzed ring-closing metathesis (RCM) of paclitaxel-dienes gave the designed 15- and 16-membered macrocyclic taxoids. However, the RCM reaction to form the designed 14-membered macrocyclic taxoid did not proceed as planned. Instead, the attempted RCM reaction led to the occurrence of an unprecedented novel Ru-catalyzed diene-coupling process, giving the corresponding 15-membered macrocyclic taxoid (SB-T-2054). The biological activities of the novel macrocyclic taxoids were evaluated by tumor cell growth inhibition (i.e., cytotoxicity) and tubulin-polymerization assays. Those assays revealed high sensitivity of cytotoxicity to subtle conformational changes. Among the novel macrocyclic taxoids evaluated, SB-T-2054 is the most active compound, which possesses virtually the same potency as that of paclitaxel. The result may also indicate that SB-T-2054 structure is an excellent mimic of the bioactive conformation of paclitaxel. Computational analysis for the observed structure-activity relationships is also performed and discussed.
Collapse
Affiliation(s)
- Liang Sun
- Department of Chemistry, State University of New York at Stony Brook, Stony Brook, New York 11794-3400, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Sun L, Simmerling C, Ojima I. Recent advances in the study of the bioactive conformation of taxol. ChemMedChem 2009; 4:719-31. [PMID: 19360801 DOI: 10.1002/cmdc.200900044] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Paclitaxel is one of the most important chemotherapeutic drugs in the fight against cancer. This minireview covers the recent advances in the study of the bioactive conformation of paclitaxel in tubulin/microtubules. The tubulin-bound structure of paclitaxel has been studied by means of photoaffinity labeling, cryo-electron microscopy, solid-state NMR, molecular modeling, MD simulations and the synthesis of conformationally restrained analogues and paclitaxel mimics. The bioactive conformation of paclitaxel is important since it could provide critical information that would allow the design of novel analogues with simpler structures and/or increased potency against cancer.
Collapse
Affiliation(s)
- Liang Sun
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, State University of New York at Stony Brook, Stony Brook, New York 11794-3400, USA
| | | | | |
Collapse
|
38
|
Nevarez DM, Mengistu YA, Nawarathne IN, Walker KD. An N-aroyltransferase of the BAHD superfamily has broad aroyl CoA specificity in vitro with analogues of N-dearoylpaclitaxel. J Am Chem Soc 2009; 131:5994-6002. [PMID: 19382815 DOI: 10.1021/ja900545m] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The native N-debenzoyl-2'-deoxypaclitaxel:N-benzoyltransferase (NDTBT), from Taxus plants, transfers a benzoyl group from the corresponding CoA thioester to the amino group of the beta-phenylalanine side chain of N-debenzoyl-2'-deoxypaclitaxel, which is purportedly on the paclitaxel (Taxol) biosynthetic pathway. To elucidate the substrate specificity of NDTBT overexpressed in Escherichia coli, the purified enzyme was incubated with semisynthetically derived N-debenzoyltaxoid substrates and aroyl CoA donors (benzoyl; ortho-, meta-, and para-substituted benzoyls; various heterole carbonyls; alkanoyls; and butenoyl), which were obtained from commercial sources or synthesized via a mixed anhydride method. Several unnatural N-aroyl-N-debenzoyl-2'-deoxypaclitaxel analogues were biocatalytically assembled with catalytic efficiencies (V(max)/K(M)) ranging between 0.15 and 1.74 nmol.min(-1).mM(-1). In addition, several N-acyl-N-debenzoylpaclitaxel variants were biosynthesized when N-debenzoylpaclitaxel and N-de(tert-butoxycarbonyl)docetaxel (i.e., 10-deacetyl-N-debenzoylpaclitaxel) were used as substrates. The relative velocity (v(rel)) for NDTBT with the latter two N-debenzoyl taxane substrates ranged between approximately 1% and 200% for the array of aroyl CoAs compared to benzoyl CoA. Interestingly, NDTBT transferred hexanoyl, acetyl, and butyryl more rapidly than butenoyl or benzoyl from the CoA donor to taxanes with isoserinoyl side chains, whereas N-debenzoyl-2'-deoxypaclitaxel was more rapidly converted to its N-benzoyl derivative than to its N-alkanoyl or N-butenoyl congeners. Biocatalytic N-acyl transfer of novel acyl groups to the amino functional group of N-debenzoylpaclitaxel and its 2'-deoxy precursor reveal the surprisingly indiscriminate specificity of this transferase. This feature of NDTBT potentially provides a tool for alternative biocatalytic N-aroylation/alkanoylation to construct next generation taxanes or other novel bioactive diterpene compounds.
Collapse
Affiliation(s)
- Danielle M Nevarez
- Cell and Molecular Biology Program, Michigan State University, East Lansing, Michigan 48824, USA
| | | | | | | |
Collapse
|
39
|
Hodge M, Chen QH, Bane S, Sharma S, Loew M, Banerjee A, Alcaraz AA, Snyder JP, Kingston DGI. Synthesis and bioactivity of a side chain bridged paclitaxel: A test of the T-Taxol conformation. Bioorg Med Chem Lett 2009; 19:2884-7. [PMID: 19359169 PMCID: PMC2684938 DOI: 10.1016/j.bmcl.2009.03.063] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 03/10/2009] [Accepted: 03/10/2009] [Indexed: 10/21/2022]
Abstract
A knowledge of the bioactive tubulin-binding conformation of paclitaxel (Taxol()) is crucial to a full understanding of the bioactivity of this important anticancer drug, and potentially also to the design of simplified analogs. The bioactive conformation has been shown to be best approximated by the T-Taxol conformation. As a further test of this conclusion, the paclitaxel analog 4 was designed as a compound which has all the chemical functionality necessary for activity, but which cannot adopt the T-Taxol conformation. The synthesis and bioassay of 4 confirmed its lack of activity, and thus provided further support for the T-Taxol conformation as the bioactive tubulin-binding conformation.
Collapse
Affiliation(s)
- Mathis Hodge
- Department of Chemistry, Virginia Polytechnic Institute and State University, Blacksburg, 24061, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Herndon JW. The chemistry of the carbon-transition metal double and triple bond: Annual survey covering the Year 2007. Coord Chem Rev 2009. [DOI: 10.1016/j.ccr.2008.12.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
41
|
Abstract
Microtubule-binding drugs (MBD) are widely used in cancer chemotherapy and also have clinically relevant antiangiogenic and vascular-disrupting properties. These antivascular actions are due in part to direct effects on endothelial cells, and all MBDs (both microtubule-stabilizing and microtubule-destabilizing) inhibit endothelial cell proliferation, migration, and tube formation in vitro, actions that are thought to correspond to therapeutic antiangiogenic actions. In addition, the microtubule-destabilizing agents cause prominent changes in endothelial cell morphology, an action associated with rapid vascular collapse in vivo. The effects on endothelial cells occur in vitro at low drug concentrations, which do not affect microtubule gross morphology, do not cause microtubule bundling or microtubule loss and do not induce cell cycle arrest, apoptosis, or cell death. Rather, it has been hypothesized that, at low concentrations, MBDs produce more subtle effects on microtubule dynamics, block critical cell signaling pathways, and prevent the microtubules from properly interacting with transient subcellular assemblies (focal adhesions and adherens junctions) whose subsequent stabilization and/or maturation are required for cell motility and cell-cell interactions. This review will focus on recent studies to define the molecular mechanisms for the antivascular actions of the MBDs, information that could be useful in the identification or design of agents whose actions more selectively target the tumor vasculature.
Collapse
Affiliation(s)
- Edward L Schwartz
- Department of Oncology, Albert Einstein College of Medicine, Bronx, New York 10467, USA.
| |
Collapse
|
42
|
Ojima I, Das M. Recent advances in the chemistry and biology of new generation taxoids. JOURNAL OF NATURAL PRODUCTS 2009; 72:554-565. [PMID: 19239240 PMCID: PMC2727999 DOI: 10.1021/np8006556] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Among the numerous chemotherapeutic drugs, paclitaxel and docetaxel are among the most widely used against various types of cancer. However, these drugs cause undesirable side effects as well as drug resistance. Therefore, it is essential to develop "taxane" anticancer agents with better pharmacological properties and improved activity especially against drug-resistant cancers. Several laboratories have performed extensive SAR studies on paclitaxel. Our SAR studies have led to the development of numerous highly potent novel second- and third-generation taxoids with systematic modifications at the C-2, C-10, and C-3' positions. The third-generation taxoids showed virtually no difference in potency against drug-resistant and drug-sensitive cell lines. Some of the new generation taxoids also exhibited excellent cytotoxicity against pancreatic cell lines expressing multidrug-resistant genes. We have also designed taxoids with strategic fluorine incorporation to investigate their effects on the cytotoxicity and the blockage of known metabolic pathways. Furthermore, we have successfully employed computational biology analysis to design novel macrocyclic taxoids to mimic the bioactive conformation of paclitaxel. This account describes our work on the design, synthesis, and biological evaluation of these novel taxoids, which has led to the discovery of very promising candidates for further preclinical studies.
Collapse
Affiliation(s)
- Iwao Ojima
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, State University of New York at Stony Brook, Stony Brook, New York 11794-3400, USA.
| | | |
Collapse
|
43
|
Calcul L, Chow R, Oliver AG, Tenney K, White KN, Wood AW, Fiorilla C, Crews P. NMR strategy for unraveling structures of bioactive sponge-derived oxy-polyhalogenated diphenyl ethers. JOURNAL OF NATURAL PRODUCTS 2009; 72:443-9. [PMID: 19323567 PMCID: PMC2772101 DOI: 10.1021/np800737z] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The overexpression of the Mcl-1 protein in cancerous cells results in the sequestering of Bak, a key component in the regulation of normal cell apoptosis. Our investigation of the ability of marine-derived small-molecule natural products to inhibit this protein-protein interaction led to the isolation of several bioactive oxy-polyhalogenated diphenyl ethers. A semipure extract, previously obtained from Dysidea (Lamellodysidea) herbacea and preserved in our repository, along with an untouched Dysidea granulosa marine sponge afforded 13 distinct oxy-polyhalogenated diphenyl ethers. Among these isolates were four new compounds, 5, 6, 10, and 12. The structure elucidation of these molecules was complicated by the plethora of structural variants that exist in the literature. During dereplication, we established a systematic method for analyzing this class of compounds. The strategy is governed by trends in the (1)H and (13)C NMR shifts of the aromatic rings, and the success of the strategy was checked by X-ray crystal structure analysis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Phillip Crews
- To whom correspondence should be addressed., Tel.: 831-459-2603. Fax: 831-459-2935.
| |
Collapse
|
44
|
Yang Y, Alcaraz AA, Snyder JP. The tubulin-bound conformation of paclitaxel: T-taxol vs "PTX-NY". JOURNAL OF NATURAL PRODUCTS 2009; 72:422-429. [PMID: 19267457 DOI: 10.1021/np800662j] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Nearly 35 years after its discovery and 11 years after FDA approval of paclitaxel (PTX) as a breakthrough anticancer drug, the 3-D structure of the agent bound to its beta-tubulin target was proposed to be T-Taxol. The latter bioactive form has recently been challenged by the Ojima group with a structure, "PTX-NY" ("REDOR Taxol"), in which the C-13 side chain is proposed to adopt a different conformation and an alternative hydrogen-bonding pattern in the tubulin binding site. Previously, the two conformers were compared to show that only T-Taxol fits the PTX-derived electron crystallographic density. That work has been extended by molecular mechanics and quantum chemical methods to reveal that the PTX-NY conformation is relatively less stable, on average, by 10-11 kcal/mol. In agreement with NMR studies, an 11 ns molecular dynamics treatment for PTX in an explicit water pool locates T-Taxol along the trajectory, but not PTX-NY. Docking of various PTX conformers into the electron crystallographic binding site of tubulin demonstrates that PTX-NY cannot be accommodated unless the pocket is reorganized in violation of the experimental constraints. Finally, analysis of the structures of T-Taxol and PTX-NY for their capacity to predict the existence of superpotent PTX analogues discloses that only the former forecasts such analogues, as now established by the T-Taxol-inspired synthesis of bridged taxanes. In sum, all empirical criteria support T-Taxol as the bound conformation of PTX on beta-tubulin in microtubules.
Collapse
Affiliation(s)
- Yutao Yang
- Department of Chemistry, Emory University, Atlanta, Georgia 30322, USA
| | | | | |
Collapse
|
45
|
Ondari ME, Walker KD. Synthesis of 4-deacetyl-1-dimethylsilyl-7-triethylsilylbaccatin III. J Org Chem 2009; 74:2186-8. [PMID: 19199654 DOI: 10.1021/jo802598m] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A one-pot trisilylation step to protect three hydroxyl groups of baccatin III (1), followed by hydride ester cleavage and base hydrolysis of a triethylsilyl ether at C13, provides efficient access to a key intermediate 9 (top path). This route removes two steps from a previously established reaction sequence to 9 (bottom path). In principle, inclusion of the truncated reaction sequence into widely utilized semisynthetic routes to next generation Taxol (paclitaxel) compounds could conceivably shorten the overall process.
Collapse
Affiliation(s)
- Mark E Ondari
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, USA
| | | |
Collapse
|
46
|
The Tubulin Binding Mode of Microtubule Stabilizing Agents Studied by Electron Crystallography. Top Curr Chem (Cham) 2008; 286:209-57. [DOI: 10.1007/128_2008_24] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
|
47
|
Gelmi ML, Nava D, Leone S, Pellegrino S, Baldelli E, Zunino F, Cappelletti G, Cartelli D, Fontana G. Semisynthesis of New D-seco-C-nor-Taxane Derivatives Containing a Polyfunctionalized Furanosyl or Cyclopentenyl or Cyclopentyl C-Ring. J Org Chem 2008; 73:8893-900. [DOI: 10.1021/jo801458j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Maria Luisa Gelmi
- Istituto di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università degli Studi di Milano, via Venezian 21, 20133, Milano, Italy, Indena SPA, via Ripamonti 99, 20141 Milano, Italy, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133 Milano, Italy, and Dipartimento di Biologia, Università degli Studi di Milano, Via Celoria, 26, 20133 Milano, Italy
| | - Donatella Nava
- Istituto di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università degli Studi di Milano, via Venezian 21, 20133, Milano, Italy, Indena SPA, via Ripamonti 99, 20141 Milano, Italy, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133 Milano, Italy, and Dipartimento di Biologia, Università degli Studi di Milano, Via Celoria, 26, 20133 Milano, Italy
| | - Samantha Leone
- Istituto di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università degli Studi di Milano, via Venezian 21, 20133, Milano, Italy, Indena SPA, via Ripamonti 99, 20141 Milano, Italy, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133 Milano, Italy, and Dipartimento di Biologia, Università degli Studi di Milano, Via Celoria, 26, 20133 Milano, Italy
| | - Sara Pellegrino
- Istituto di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università degli Studi di Milano, via Venezian 21, 20133, Milano, Italy, Indena SPA, via Ripamonti 99, 20141 Milano, Italy, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133 Milano, Italy, and Dipartimento di Biologia, Università degli Studi di Milano, Via Celoria, 26, 20133 Milano, Italy
| | - Eleonora Baldelli
- Istituto di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università degli Studi di Milano, via Venezian 21, 20133, Milano, Italy, Indena SPA, via Ripamonti 99, 20141 Milano, Italy, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133 Milano, Italy, and Dipartimento di Biologia, Università degli Studi di Milano, Via Celoria, 26, 20133 Milano, Italy
| | - Franco Zunino
- Istituto di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università degli Studi di Milano, via Venezian 21, 20133, Milano, Italy, Indena SPA, via Ripamonti 99, 20141 Milano, Italy, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133 Milano, Italy, and Dipartimento di Biologia, Università degli Studi di Milano, Via Celoria, 26, 20133 Milano, Italy
| | - Graziella Cappelletti
- Istituto di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università degli Studi di Milano, via Venezian 21, 20133, Milano, Italy, Indena SPA, via Ripamonti 99, 20141 Milano, Italy, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133 Milano, Italy, and Dipartimento di Biologia, Università degli Studi di Milano, Via Celoria, 26, 20133 Milano, Italy
| | - Daniele Cartelli
- Istituto di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università degli Studi di Milano, via Venezian 21, 20133, Milano, Italy, Indena SPA, via Ripamonti 99, 20141 Milano, Italy, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133 Milano, Italy, and Dipartimento di Biologia, Università degli Studi di Milano, Via Celoria, 26, 20133 Milano, Italy
| | - Gabriele Fontana
- Istituto di Chimica Organica “A. Marchesini” Facoltà di Farmacia, Università degli Studi di Milano, via Venezian 21, 20133, Milano, Italy, Indena SPA, via Ripamonti 99, 20141 Milano, Italy, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, 20133 Milano, Italy, and Dipartimento di Biologia, Università degli Studi di Milano, Via Celoria, 26, 20133 Milano, Italy
| |
Collapse
|
48
|
Zimmer SM, Liu J, Clayton JL, Stephens DS, Snyder JP. Paclitaxel binding to human and murine MD-2. J Biol Chem 2008; 283:27916-27926. [PMID: 18650420 PMCID: PMC2562052 DOI: 10.1074/jbc.m802826200] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 06/23/2008] [Indexed: 11/06/2022] Open
Abstract
Paclitaxel (PTX) is an important cancer chemotherapeutic agent that binds to beta-tubulin and prevents mitosis through microtubule overstabilization. Recent evidence also implicates PTX in the induction of apoptosis of cancer cells via the TLR4 innate immune pathway. The TLR4 accessory protein, MD-2, is an essential component for the species-specific proinflammatory activity of PTX on murine cells. However, whether PTX binds to human MD-2 and how MD-2 and TLR4 interact with PTX are not well defined. Recombinant human MD-2 (rhMD-2) was produced in a Pichia pastoris expression system, and the interaction between rhMD-2 and PTX was assessed by an enzyme-linked immunosorbent assay to show that PTX binds rhMD-2. Formation of the latter complex was found to be dose-dependent and inhibited by anti-MD-2 antibody but not by an isotype control antibody. As measured by human tumor necrosis factor alpha production, human THP-1 monocytes expressing TLR4 and MD-2 were poorly responsive to the addition of PTX, but murine macrophages expressing TLR4 and MD-2 responded in a dose-dependent manner. Human embryonic kidney (HEK293) cells transfected with both human TLR4 and human MD-2 or human MD-2 and murine TLR4 were also poorly responsive to PTX (10 microm). However, HEK293 cells transfected with murine MD-2 and human TLR4 or murine MD-2 and murine TLR4 were highly responsive to PTX (10 microm), indicating that the murine MD-2/PTX interaction is required for TLR4 activation. To further define the structural differences for MD-2/TLR4 activation, crystal structures of both murine and human MD-2 were subjected to PTX docking by computational methods. These models indicate that PTX binds in the pocket of both human and mouse MD-2 structures. The species-specific difference between human and murine MD-2 activation of TLR4 by PTX can be explained by alterations of surface charge distribution (i.e. electrostatic potential), binding pocket size, and the locus of PTX binding within the MD-2 pocket, which results in reorganization of the 123-130 amino acid loop. In particular, Phe(126) appears to operate as a bridge for TLR4.MD-2 dimerization in the mouse but not the human protein.
Collapse
Affiliation(s)
- Shanta M Zimmer
- Department of Chemistry, Emory University, Atlanta, Georgia 30322
| | - Jin Liu
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Jaime L Clayton
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322
| | - David S Stephens
- Department of Chemistry, Emory University, Atlanta, Georgia 30322
| | - James P Snyder
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
49
|
Gaspar JR, Qu J, Straubinger NL, Straubinger RM. Highly selective and sensitive assay for paclitaxel accumulation by tumor cells based on selective solid phase extraction and micro-flow liquid chromatography coupled to mass spectrometry. Analyst 2008; 133:1742-8. [PMID: 19082078 DOI: 10.1039/b806856a] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The taxanes are among the most important cancer chemotherapy drugs approved for clinical use in the last two decades. Paclitaxel is used as first-line therapy for a variety of cancers, and numerous drug delivery approaches are under investigation to enhance its selectivity and effectiveness against tumors. One strategy is to produce sustained, low drug levels within the tumor to enhance apoptosis and inhibit angiogenesis. The interest in altering drug concentration/time exposure profiles to improve therapeutic outcomes creates the necessity to quantify low concentrations of paclitaxel in cells or tissues. Here, a selective solid phase extraction (SPE) method, coupled with a capillary liquid chromatography-tandem mass spectrometry (microLC-MS/MS) method, was developed to quantify low, therapeutically relevant concentrations of paclitaxel that could not be analyzed using conventional LC-MS/MS. Under optimized SPE wash and elution conditions, paclitaxel was selectively extracted from biological samples, and most matrix components were removed. A 150 x 0.5 mm ID ODS capillary column was used for microLC separation and the flow rate was 12 microL min(-1). Sample extracts were focused at the front of the microLC column and then eluted with a gradient. The lower limits of detection and quantification were 5 and 20 pg mL(-1), respectively, permitting quantification of paclitaxel in small tissue samples or in cultured cells exposed to low drug concentrations. The quantitative linear range was 20-20 000 pg mL(-1). The ability to quantify these low concentrations of paclitaxel provides an important tool to study the concentration-dependent pharmacological effects of this important drug.
Collapse
Affiliation(s)
- Julio R Gaspar
- The Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Amherst, NY 14260-1200, USA
| | | | | | | |
Collapse
|
50
|
Fang WS, Wang SR. Structural studies of taxol analogues for drug discovery. Expert Opin Drug Discov 2008; 3:1109-22. [DOI: 10.1517/17460441.3.9.1109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|