1
|
Jiang B, Zheng Y, Xue T, Wu J, Song H, Zhou S, Li Y, Gong J, Wei M, Ji X, Wei M, Wang L, Gong J, Liu M, Wang A, Zhang K, Lv K, Zheng Y. Identification of selenium-containing benzamides as potent microtubule-targeting antitumor agents. Bioorg Chem 2025; 159:108355. [PMID: 40090150 DOI: 10.1016/j.bioorg.2025.108355] [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: 01/27/2025] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025]
Abstract
IMB5046, a microtubule inhibitor discovered by our team, served as the lead compound for designing a series of selenium-containing benzoates and benzamides. Among these, compound 2g emerged as a lead candidate, demonstrating potent antiproliferative activity. Mechanistic studies revealed that 2g bound to the colchicine site of tubulin, caused G2/M cell cycle arrest, and generated ROS. Notably, 2g exhibited exceptional efficacy in P-gp overexpressing MCF7/ADR and KBV200 cell lines, with drug-resistance indices (DRI) of 0.83 and 0.58, respectively, significantly outperforming colchicine (DRIs: 25.4 and 8.03) and paclitaxel (DRIs: 41.0 and 4.96). In an MCF-7 xenograft model, 2g (25 mg/kg, IP) achieved a tumor growth inhibition rate of 57.2 %, surpassing IMB5046 (47.6 %). To enhance solubility and pharmacokinetics, prodrug 2g-P was developed, showing 69 % bioavailability but reduced in vivo efficacy. Further investigation is warranted to elucidate the factors underlying the discrepancy, such as the efficiency of prodrug-to-drug conversion and intracellular accumulation of active 2g. In summary, our study not only identified a novel selenium-containing lead compound, but also provided important insights into prodrug design. These findings lay a solid foundation for the development of next-generation microtubule-targeting agents capable of overcoming drug resistance.
Collapse
Affiliation(s)
- Bin Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Department of Pharmaceutical Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Yijia Zheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Tiezheng Xue
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Department of Pharmaceutical Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Jizhou Wu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Huijuan Song
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Sheng Zhou
- Department of Pharmaceutical Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Yujing Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiaqi Gong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Meng Wei
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaorui Ji
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Meijiao Wei
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lujun Wang
- Department of Pharmacy, Medical Supplies Center of PLA General Hospital, China
| | - Jianhua Gong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mingliang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Apeng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Kai Zhang
- Department of Pharmaceutical Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China.
| | - Kai Lv
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yanbo Zheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
2
|
Lim H, Lee JH, Park SH, Lee JH, Jang H, Yang SB, Seo M, Lee S, Park J. Tumor-specific biochemical nanoconversion of self-assembled peptide-conjugated paclitaxel-docetaxel-based nanoparticles. NANO CONVERGENCE 2025; 12:20. [PMID: 40285925 PMCID: PMC12033163 DOI: 10.1186/s40580-025-00487-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Docetaxel (DTX, 1) and paclitaxel (PTX, 2) are famous cytotoxic agents widely used in cancer therapy, however, their low specificity for tumor cells often results in severe systemic toxicity. Beyond conventional prodrug strategies, this study introduces a novel nanoconversion technology that chemically modifies DTX to form self-assembled nanoparticles (NPs), which subsequently convert into a paclitaxel-mimicking molecule (PTXm, 3). Hydrophilic acetylated Phe-Arg-Arg-Phe peptide ((Ac)FRRF, 4) and hydrophobic docetaxel were conjugated to prepare self-assembled (Ac)FRRF-DTX NPs. The selective cleavage of the Arg-Phe bond by cathepsin B, which is abundant in cancer cells, facilitated the nanoconversion of PTXm (3) from (Ac)FRRF-DTX NPs, demonstrating effective cytotoxic effects. Utilizing the cleavage site of peptide and specific sequences (ex. Arg-Arg-Phe), this approach does not simply act as a prodrug but allows the nanomaterial to transform into another cytotoxic biomolecule within tumors. (Ac)FRRF-DTX NPs exhibited remarkable physicochemical properties, superior anti-cancer efficacy, and low toxicity, showcasing an innovative conversion in peptide-conjugated nanomedicine. Unlike traditional prodrug chemistry, this tumor-specific nanoconversion process involves the biochemical transformation of DTX (1) into PTXm (3) via enzymatic action. Overall, this study provides an outstanding example of chemical drug molecular modification through the concept of nanoconversion.
Collapse
Affiliation(s)
- Hansol Lim
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Jae-Hyeon Lee
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - So-Hyeon Park
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Jun-Hyuck Lee
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Hyesu Jang
- College of Pharmacy, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Seong-Bin Yang
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Minho Seo
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea
| | - Seokwoo Lee
- College of Pharmacy, Chungnam National University, Daejeon, 34134, Republic of Korea.
| | - Jooho Park
- Department of Applied Life Science, BK21 Program, Konkuk University, Chungju, 27478, Republic of Korea.
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungju, 27478, Republic of Korea.
| |
Collapse
|
3
|
Byrne AJ, Bright SA, McKeown JP, Bergin A, Twamley B, McElligott AM, Noorani S, Kandwal S, Fayne D, O’Boyle NM, Williams DC, Meegan MJ. Synthesis and Pro-Apoptotic Effects of Nitrovinylanthracenes and Related Compounds in Chronic Lymphocytic Leukaemia (CLL) and Burkitt's Lymphoma (BL). Molecules 2023; 28:8095. [PMID: 38138584 PMCID: PMC10746112 DOI: 10.3390/molecules28248095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/30/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Chronic lymphocytic leukaemia (CLL) is a malignancy of the immune B lymphocyte cells and is the most common leukaemia diagnosed in developed countries. In this paper, we report the synthesis and antiproliferative effects of a series of (E)-9-(2-nitrovinyl)anthracenes and related nitrostyrene compounds in CLL cell lines and also in Burkitt's lymphoma (BL) cell lines, a rare form of non-Hodgkin's immune B-cell lymphoma. The nitrostyrene scaffold was identified as a lead structure for the development of effective compounds targeting BL and CLL. The series of structurally diverse nitrostyrenes was synthesised via Henry-Knoevenagel condensation reactions. Single-crystal X-ray analysis confirmed the structure of (E)-9-chloro-10-(2-nitrobut-1-en-1-yl)anthracene (19f) and the related 4-(anthracen-9-yl)-1H-1,2,3-triazole (30a). The (E)-9-(2-nitrovinyl)anthracenes 19a, 19g and 19i-19m were found to elicit potent antiproliferative effects in both BL cell lines EBV-MUTU-1 (chemosensitive) and EBV+ DG-75 (chemoresistant) with >90% inhibition at 10 μM. Selected (E)-9-(2-nitrovinyl)anthracenes demonstrated potent antiproliferative activity in CLL cell lines, with IC50 values of 0.17 μM (HG-3) and 1.3 μM (PGA-1) for compound 19g. The pro-apoptotic effects of the most potent compounds 19a, 19g, 19i, 19l and 19m were demonstrated in both CLL cell lines HG-3 and PGA-1. The (E)-nitrostyrene and (E)-9-(2-nitrovinyl)anthracene series of compounds offer potential for further development as novel chemotherapeutics for CLL.
Collapse
Affiliation(s)
- Andrew J. Byrne
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (J.P.M.); (M.J.M.)
| | - Sandra A. Bright
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (S.K.); (D.F.); (D.C.W.)
| | - James. P. McKeown
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (J.P.M.); (M.J.M.)
| | - Adam Bergin
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (J.P.M.); (M.J.M.)
| | - Brendan Twamley
- School of Chemistry, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland;
| | - Anthony M. McElligott
- Discipline of Haematology, School of Medicine, Trinity Translational Medicine Institute, St. James’s Hospital and Trinity College, Dublin 8, D08 W9RT Dublin, Ireland;
| | - Sara Noorani
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (J.P.M.); (M.J.M.)
| | - Shubhangi Kandwal
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (S.K.); (D.F.); (D.C.W.)
| | - Darren Fayne
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (S.K.); (D.F.); (D.C.W.)
| | - Niamh M. O’Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (J.P.M.); (M.J.M.)
| | - D. Clive Williams
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (S.K.); (D.F.); (D.C.W.)
| | - Mary J. Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse St, Dublin 2, D02 R590 Dublin, Ireland (J.P.M.); (M.J.M.)
| |
Collapse
|
4
|
Zhao Q, Lv X, Dong Y, Hong H, Zheng Y, Yang L, Gong J. IMB5036 overcomes resistance to multiple chemotherapeutic drugs in human cancer cells through pyroptosis by targeting the KH-type splicing regulatory protein. Life Sci 2023; 328:121941. [PMID: 37451400 DOI: 10.1016/j.lfs.2023.121941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/26/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
AIMS IMB5036 is a pyridazinone compound with antiproliferative and antitumour activity against hepatoma and pancreatic cancer. In this study, we attempted to identify the target protein of IMB5036 and test its potential for overcoming multidrug resistance and inducing pyroptosis. MATERIALS AND METHODS We examined the effects of IMB5036 on cancer cells by in vitro assays, a molecular docking model and in vivo tumour models. We performed pull-down experiments using biotinylated IMB5036 and identified the binding proteins. Gene knockdown were used to investigate the oncogenic role of KH-type splicing regulatory protein (KSRP). Western blot was used to detect for mechanism-associated molecules. KEY FINDINGS IMB5036 could overcome resistance to multiple chemotherapeutic drugs at the cellular level and in vivo. Furthermore, IMB5036 was not a P-glycoprotein (P-gp) substrate and downregulated the expression of P-gp. We identified KSRP as a binding protein of IMB5036. The knockdown of KSRP inhibited the proliferation of MCF7 and MCF7/adriamycin (MCF7/ADR) cells. In addition, IMB5036 induced pyroptosis in both MCF7 and MCF7/ADR cells via KSRP. SIGNIFICANCE We found IMB5036 binds to KSRP and overcomes multidrug resistance via gasdermin E (GSDME)-dependent pyroptosis.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xing Lv
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanqun Dong
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanyu Hong
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Biopharmaceuticals, School of Pharmaceutics Sciences, Wenzhou Medical University, Zhejiang, China
| | - Yanbo Zheng
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Lijun Yang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Jianhua Gong
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Doan NQH, Nguyen NTK, Duong VB, Nguyen HTT, Vong LB, Duong DN, Nguyen NTT, Nguyen TLT, Do TTH, Truong TN. Synthesis, Biological Evaluation, and Molecular Modeling Studies of 1-Aryl-1 H-pyrazole-Fused Curcumin Analogues as Anticancer Agents. ACS OMEGA 2022; 7:33963-33984. [PMID: 36188331 PMCID: PMC9520563 DOI: 10.1021/acsomega.2c02933] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/08/2022] [Indexed: 05/28/2023]
Abstract
Addressing the growing burden of cancer and the shortcomings of chemotherapy in cancer treatment are the current research goals. Research to overcome the limitations of curcumin and to improve its anticancer activity via its heterocycle-fused monocarbonyl analogues (MACs) has immense potential. In this study, 32 asymmetric MACs fused with 1-aryl-1H-pyrazole (7a-10h) were synthesized and characterized to develop new curcumin analogues. Subsequently, via initial screening for cytotoxic activity, nine compounds exhibited potential growth inhibition against MDA-MB-231 (IC50 2.43-7.84 μM) and HepG2 (IC50 4.98-14.65 μM), in which seven compounds showing higher selectivities on two cancer cell lines than the noncancerous LLC-PK1 were selected for cell-free in vitro screening for effects on microtubule assembly activity. Among those, compounds 7d, 7h, and 10c showed effective inhibitions of microtubule assembly at 20.0 μM (40.76-52.03%), indicating that they could act as microtubule-destabilizing agents. From the screening results, three most potential compounds, 7d, 7h, and 10c, were selected for further evaluation of cellular effects on breast cancer MDA-MB-231 cells. The apoptosis-inducing study indicated that these three compounds could cause morphological changes at 1.0 μM and could enhance caspase-3 activity (1.33-1.57 times) at 10.0 μM in MDA-MB-231 cells, confirming their apoptosis-inducing activities. Additionally, in cell cycle analysis, compounds 7d and 7h at 2.5 μM and 10c at 5.0 μM also arrested MDA-MB-231 cells in the G2/M phase. Finally, the results from in silico studies revealed that the predicted absorption, distribution, metabolism, excretion, and the toxicity (ADMET) profile of the most potent MACs might have several advantages in addition to potential disadvantages, and compound 7h could bind into (ΔG -10.08 kcal·mol-1) and access wider space at the colchicine-binding site (CBS) than that of colchicine or nocodazole via molecular docking studies. In conclusion, our study serves as a basis for the design of promising synthetic compounds as anticancer agents in the future.
Collapse
Affiliation(s)
- Nam Q. H. Doan
- Faculty
of Pharmacy, Van Lang University, Ho Chi Minh City 700000, Vietnam
| | - Ngan T. K. Nguyen
- Department
of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Vu B. Duong
- Department
of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Ha T. T. Nguyen
- School
of Biomedical Engineering, International University, Vietnam National University Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Long B. Vong
- School
of Biomedical Engineering, International University, Vietnam National University Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Diem N. Duong
- Immunology
Lab, Vaccines and Biologicals Production Department, Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Nguyet-Thu T. Nguyen
- Immunology
Lab, Vaccines and Biologicals Production Department, Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Tuyen L. T. Nguyen
- Saigon
Pharmaceutical Sciences and Technologies Center, Ho Chi Minh City 700000, Vietnam
| | - Tuoi T. H. Do
- Department
of Pharmacology, Faculty of Pharmacy, University
of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| | - Tuyen N. Truong
- Department
of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam
| |
Collapse
|
6
|
Nitrobenzoate-Derived Compound X8 Impairs Vascular Development in Zebrafish. Int J Mol Sci 2022; 23:ijms23147788. [PMID: 35887139 PMCID: PMC9316178 DOI: 10.3390/ijms23147788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/11/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022] Open
Abstract
Proper growth and patterning of blood vessels are critical for embryogenesis. Chemicals or environmental hormones may interfere with vascular growth and cause developmental defects. Nitrobenzoate-based compounds have been demonstrated to have a wide range of biological and pharmacological functions, leading to the development of numerous 4-nitrobenzoate derivatives for clinical application. In this study, we tested a novel nitrobenzoate-derived compound, X8, and investigated its effects on vascular development using zebrafish as a model organism. We first determined the survival rate of embryos after the addition of exogenous X8 (0.5, 1, 3, 5, and 10 μM) to the fish medium and determined a sublethal dose of 3 μM for use in further assays. We used transgenic fish to examine the effects of X8 treatment on vascular development. At 25–32 h postfertilization (hpf), X8 treatment impaired the growth of intersegmental vessels (ISVs) and caudal vein plexuses (CVPs). Moreover, X8-treated embryos exhibited pericardial edema and circulatory defects at 60–72 hpf, suggesting the effects of X8 in vasculature. Apoptosis tests showed that the vascular defects were likely caused by the inhibition of proliferation and migration. To investigate the molecular impacts underlying the defects in the vasculature of X8-treated fish, the expression levels of vascular markers, including ephrinb2, mrc1, and stabilin, were assessed, and the decreased expression of those genes was detected, indicating that X8 inhibited the expression of vascular genes. Finally, we showed that X8 treatment disrupted exogenous GS4012-induced angiogenesis in Tg(flk:egfp) zebrafish embryos. In addition, vascular defects were enhanced during cotreatment with X8 and the VEGFR2 inhibitor SU5416, suggesting that X8 treatment causes vascular defects mediated by disruption of VEGF/VEGFR2 signaling. Collectively, our findings indicate that X8 could be developed as a novel antiangiogenic agent.
Collapse
|
7
|
Zheng YB, Dong YQ, Si SY, Zhen YS, Gong JH. IMB5476, a novel microtubule inhibitor, induces mitotic catastrophe and overcomes multidrug resistance in tumors. Eur J Pharmacol 2022; 919:174802. [PMID: 35143830 DOI: 10.1016/j.ejphar.2022.174802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/22/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023]
Abstract
IMB5046 is a nitrobenzoate microtubule inhibitor we reported previously. During screening of its structural analogues, we identified a novel compound IMB5476 with increased aqueous solubility. Here, its antitumor activity and the underlying mechanism were investigated. IMB5476 disrupted microtubule networks in cells and arrested cell cycle at G2/M phase. It inhibited purified tubulin polymerization in vitro. Competition assay indicated that it bound to tubulin at the colchicine pocket. Further experiments proved that it induced cell death by mitotic catastrophe and apoptosis. Notably, it was a poor substrate of P-glycoprotein and exhibited potent cytotoxicity against drug-resistant tumor cells. In addition, IMB5476 could inhibit angiogenesis in vitro. IMB5476 also inhibited the growth of drug-resistant KBV200 xenografts in mice. Conclusively, our data reveal a novel nitrobenzoate microtubule inhibitor with improved aqueous solubility and can overcome multidrug resistance.
Collapse
Affiliation(s)
- Yan-Bo Zheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yan-Qun Dong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Yi Si
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yong-Su Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Hua Gong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
8
|
Zhuang Y, Yang G, Wu S, Chen J, Guo J, Quan D, Zhang T, Yang Z, Tan S, Ji Y, Chen Z, Lv L. 5-arylalkynyl-2-benzoyl thiophene: a novel microtubule inhibitor exhibits antitumor activity without neurological toxicity. Am J Cancer Res 2022; 12:229-246. [PMID: 35141015 PMCID: PMC8822276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 06/15/2021] [Indexed: 06/14/2023] Open
Abstract
The composition of microtubules involving several steps, including the polymerization and depolymerization of α-tubulin and β-tubulin heterodimers. Microtubule-targeting agents can increase or inhibit microtubule polymerization, thereby disrupting the dynamic process and stalling cells in G2/M phase. Microtubule-targeting agents are generally cytotoxic, which neurological toxicity being one of the significant adverse events associated. We recently reported a novel 5-arylalkynyl-2-benzoyl thiophene (PST-3) that exhibited broad-spectrum cellular cytotoxicity and in vivo potency with high safety. PST-3 was a substrate of p-gp, which could not cross the blood-brain barrier and lead to less neurotoxicity. The antitumor activities in vitro demonstrated that PST-3 combined with the colchicine-binding site on microtubule, induces morphological changes, disrupts microtubule networks, inhibits polymerization of tubulin, arrests breast cancer cells in the G2/M phase of the cell cycle and induces apoptosis. Evaluation of the antitumor effect in vivo demonstrated that PST-3 elicited MDA-MB-468 tumor %T/C of 11.75%, whereas elicited MCF7 tumor %T/C of 44.38% in breast cancer xenograft models. Besides, in vivo experiments of a higher dose (60 mg/kg) of PST-3 treatment for 21 days did not produce any significant neurotoxicity. These results provide evidence that PST-3 might possess the potential to be developed into a new microtubule inhibitor without neurological toxicity.
Collapse
Affiliation(s)
- Yuxin Zhuang
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
- State Key Laboratory of Quality Research in Chinese Medicines, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and TechnologyMacau, People’s Republic of China
| | - Guang Yang
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
- Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University)Zhuhai 519000, People’s Republic of China
| | - Shaoyu Wu
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| | - Jianjun Chen
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| | - Jiayin Guo
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| | - Dongling Quan
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| | - Tingting Zhang
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| | - Zichao Yang
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| | - Shaobin Tan
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| | - Yuheng Ji
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| | - Zhipeng Chen
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| | - Lin Lv
- School of Pharmaceutical Science, Southern Medical UniversityGuangzhou 510515, People’s Republic of China
| |
Collapse
|
9
|
Ayanlaja AA, Hong X, Cheng B, Zhou H, Kanwore K, Alphayo-Kambey P, Zhang L, Tang C, Adeyanju MM, Gao D. Susceptibility of cytoskeletal-associated proteins for tumor progression. Cell Mol Life Sci 2021; 79:13. [PMID: 34964908 PMCID: PMC11072373 DOI: 10.1007/s00018-021-04101-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/11/2021] [Accepted: 12/16/2021] [Indexed: 10/19/2022]
Abstract
The traditional functions of cytoskeletal-associated proteins (CAPs) in line with polymerization and stabilization of the cytoskeleton have evolved and are currently underrated in oncology. Although therapeutic drugs have been developed to target the cytoskeletal components directly in cancer treatment, several recently established therapeutic agents designed for new targets block the proliferation of cancer cells and suppress resistance to existing target agents. It would seem like these targets only work toward inhibiting the polymerization of cytoskeletal components or hindering mitotic spindle formation in cancer cells, but a large body of literature points to CAPs and their culpability in cell signaling, molecular conformation, organelle trafficking, cellular metabolism, and genomic modifications. Here, we review those underappreciated functions of CAPs, and we delineate the implications of cellular signaling instigated by evasive properties induced by aberrant expression of CAPs in response to stress or failure to exert normal functions. We present an analogy establishing CAPs as vulnerable targets for cancer systems and credible oncotargets. This review establishes a paradigm in which the cancer machinery may commandeer the conventional functions of CAPs for survival, drug resistance, and energy generation; an interesting feature overdue for attention.
Collapse
Affiliation(s)
- Abiola Abdulrahman Ayanlaja
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Department of Neurology, Johns Hopkins University School of Medicine, 201 N Broadway, Baltimore, MD, 21287, USA
| | - Xiaoliang Hong
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Bo Cheng
- The Affiliated Oriental Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Han Zhou
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Kouminin Kanwore
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Piniel Alphayo-Kambey
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Lin Zhang
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Chuanxi Tang
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | | | - Dianshuai Gao
- Public Experimental Laboratory, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
10
|
Huang YT, Hsu YT, Chen YF, Shen MR. Super-Resolution Microscopy Reveals That Stromal Interaction Molecule 1 Trafficking Depends on Microtubule Dynamics. Front Physiol 2021; 12:762387. [PMID: 34803742 PMCID: PMC8602801 DOI: 10.3389/fphys.2021.762387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/18/2021] [Indexed: 12/23/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) is an essential pathway for Ca2+ signaling, and regulates various vital cellular functions. It is triggered by the endoplasmic reticulum Ca2+ sensor stromal interaction molecule 1 (STIM1). Illustration of STIM1 spatiotemporal structure at the nanometer scale during SOCE activation provides structural and functional insights into the fundamental Ca2+ homeostasis. In this study, we used direct stochastic optical reconstruction microscopy (dSTORM) to revisit the dynamic process of the interaction between STIM1, end-binding protein (EB), and microtubules to the ER-plasma membrane. Using dSTORM, we found that“powder-like”STIM1 aggregates into “trabecular-like” architectures toward the cell periphery during SOCE, and that an intact microtubule network and EB1 are essential for STIM1 trafficking. After thapsigargin treatment, STIM1 can interact with EB1 regardless of undergoing aggregation. We generated STIM1 variants adapted from a real-world database and introduced them into SiHa cells to clarify the impact of STIM1 mutations on cancer cell behavior. The p.D76G and p.D84Y variants locating on the Ca2+ binding domain of STIM1 result in inhibition of focal adhesion turnover, Ca2+ influx during SOCE and subsequent cell migration. Inversely, the p.R643C variant on the microtubule interacting domain of STIM1 leads to dissimilar consequence and aggravates cell migration. These findings imply that STIM1 mutational patterns have an impact on cancer metastasis, and therefore could be either a prognostic marker or a novel therapeutic target to inhibit the malignant behavior of STIM1-mediated cancer cells. Altogether, we generated novel insight into the role of STIM1 during SOCE activation, and uncovered the impact of real-world STIM1 variants on cancer cells.
Collapse
Affiliation(s)
- Yi-Ting Huang
- Department of Pharmacology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Ting Hsu
- Institute of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Division of Hematology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Meng-Ru Shen
- Department of Pharmacology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
11
|
Zhu H, Li W, Shuai W, Liu Y, Yang L, Tan Y, Zheng T, Yao H, Xu J, Zhu Z, Yang DH, Chen ZS, Xu S. Discovery of novel N-benzylbenzamide derivatives as tubulin polymerization inhibitors with potent antitumor activities. Eur J Med Chem 2021; 216:113316. [PMID: 33676300 DOI: 10.1016/j.ejmech.2021.113316] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 01/17/2023]
Abstract
A series of novel N-benzylbenzamide derivatives were designed and synthesized as tubulin polymerization inhibitors. Among fifty-one target compounds, compound 20b exhibited significant antiproliferative activities with IC50 values ranging from 12 to 27 nM against several cancer cell lines, and possessed good plasma stability and satisfactory physicochemical properties. Mechanism studies demonstrated that 20b bound to the colchicine binding site and displayed potent anti-vascular activity. Notably, the corresponding disodium phosphate 20b-P exhibited an excellent safety profile with the LD50 value of 599.7 mg/kg (i.v. injection), meanwhile, it significantly inhibited tumor growth and decreased microvessel density in liver cancer cell H22 allograft mouse model without obvious toxicity. Collectively, 20b and 20b-P are novel promising anti-tubulin agents with more druggable properties and deserve to be further investigated for cancer therapy.
Collapse
Affiliation(s)
- Huajian Zhu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Wenlong Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Wen Shuai
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Yang Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Limei Yang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Yuchen Tan
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Tiandong Zheng
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Hong Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, UK
| | - Dong-Hua Yang
- College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York, 11439, United States
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York, 11439, United States
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China.
| |
Collapse
|
12
|
Xia LY, Zhang YL, Yang R, Wang ZC, Lu YD, Wang BZ, Zhu HL. Tubulin Inhibitors Binding to Colchicine-Site: A Review from 2015 to 2019. Curr Med Chem 2021; 27:6787-6814. [PMID: 31580244 DOI: 10.2174/0929867326666191003154051] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/25/2019] [Accepted: 08/22/2019] [Indexed: 11/22/2022]
Abstract
Due to the three domains of the colchicine-site which is conducive to the combination with small molecule compounds, colchicine-site on the tubulin has become a common target for antitumor drug development, and accordingly, a large number of tubulin inhibitors binding to the colchicine-site have been reported and evaluated over the past years. In this study, tubulin inhibitors targeting the colchicine-site and their application as antitumor agents were reviewed based on the literature from 2015 to 2019. Tubulin inhibitors were classified into ten categories according to the structural features, including colchicine derivatives, CA-4 analogs, chalcone analogs, coumarin analogs, indole hybrids, quinoline and quinazoline analogs, lignan and podophyllotoxin derivatives, phenothiazine analogs, N-heterocycle hybrids and others. Most of them displayed potent antitumor activity, including antiproliferative effects against Multi-Drug-Resistant (MDR) cell lines and antivascular properties, both in vitro and in vivo. In this review, the design, synthesis and the analysis of the structure-activity relationship of tubulin inhibitors targeting the colchicine-site were described in detail. In addition, multi-target inhibitors, anti-MDR compounds, and inhibitors bearing antitumor activity in vivo are further listed in tables to present a clear picture of potent tubulin inhibitors, which could be beneficial for medicinal chemistry researchers.
Collapse
Affiliation(s)
- Lin-Ying Xia
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Ya-Liang Zhang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Rong Yang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Zhong-Chang Wang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Ya-Dong Lu
- Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, Nanjing 210008, P.R. China
| | - Bao-Zhong Wang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Hai-Liang Zhu
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China
| |
Collapse
|
13
|
Cui H, Arnst K, Miller DD, Li W. Recent Advances in Elucidating Paclitaxel Resistance Mechanisms in Non-small Cell Lung Cancer and Strategies to Overcome Drug Resistance. Curr Med Chem 2020; 27:6573-6595. [DOI: 10.2174/0929867326666191016113631] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/06/2019] [Accepted: 09/12/2019] [Indexed: 12/12/2022]
Abstract
Paclitaxel (PTX) is a first-line drug for late-stage non-small cell lung cancer (NSCLC) patients
who do not benefit from targeted therapy or immunotherapy. However, patients invariably develop
resistance to PTX upon prolonged treatments. Although diverse mechanisms leading to PTX
resistance have been well-documented in the literature, strategies to overcome PTX resistance in
NSCLC based on these mechanisms are still challenging. In this article, we reviewed recent advancements
elucidating major mechanisms of PTX resistance in NSCLC, including the overexpression of
ABC transporters, alternations to tubulin structures, and the involvement of cytokines, miRNAs, kinase
signaling pathways, and epithelial-mesenchymal transition. Potential markers of PTX resistance or
PTX response that could help to direct treatment decisions and restore cellular sensitivity to PTX were
also discussed. Finally, we summarized the corresponding strategies to overcome PTX resistance in
NSCLC cells, which might provide new insights into clinical trials and benefit lung cancer patients in
the future.
Collapse
Affiliation(s)
- Hongmei Cui
- Department of Pharmaceutical Science, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Kinsie Arnst
- Department of Pharmaceutical Science, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D. Miller
- Department of Pharmaceutical Science, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Wei Li
- Department of Pharmaceutical Science, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
14
|
Zheng Y, Gong J, Zhen Y. Focal adhesion kinase is activated by microtubule-depolymerizing agents and regulates membrane blebbing in human endothelial cells. J Cell Mol Med 2020; 24:7228-7238. [PMID: 32452639 PMCID: PMC7339229 DOI: 10.1111/jcmm.15273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/01/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Microtubule-depolymerizing agents can selectively disrupt tumor vessels via inducing endothelial membrane blebbing. However, the mechanism regulating blebbing is largely unknown. IMB5046 is a newly discovered microtubule-depolymerizing agent. Here, the functions of focal adhesion kinase (FAK) during IMB5046-induced blebbing and the relevant mechanism are studied. We found that IMB5046 induced membrane blebbing and reassembly of focal adhesions in human vascular endothelial cells. Both FAK inhibitor and knock-down expression of FAK inhibited IMB5046-induced blebbing. Mechanism study revealed that IMB5046 induced the activation of FAK via GEF-H1/ Rho/ ROCK/ MLC2 pathway. cRGD peptide, a ligand of integrin, also blocked IMB5046-induced blebbing. After activation, FAK further promoted the phosphorylation of MLC2. This positive feedback loop caused more intensive actomyosin contraction and continuous membrane blebbing. FAK inhibitor blocked membrane blebbing via inhibiting actomyosin contraction, and stimulated stress fibre formation via promoting the phosphorylation of HSP27. Conclusively, these results demonstrate that FAK is a molecular switch controlling endothelial blebbing and stress fibre formation. Our study provides a new molecular mechanism for microtubule-depolymerizing agents to be used as vascular disrupting agents.
Collapse
Affiliation(s)
- Yan‐Bo Zheng
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jian‐Hua Gong
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yong‐Su Zhen
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
15
|
Ong MS, Deng S, Halim CE, Cai W, Tan TZ, Huang RYJ, Sethi G, Hooi SC, Kumar AP, Yap CT. Cytoskeletal Proteins in Cancer and Intracellular Stress: A Therapeutic Perspective. Cancers (Basel) 2020; 12:cancers12010238. [PMID: 31963677 PMCID: PMC7017214 DOI: 10.3390/cancers12010238] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/20/2022] Open
Abstract
Cytoskeletal proteins, which consist of different sub-families of proteins including microtubules, actin and intermediate filaments, are essential for survival and cellular processes in both normal as well as cancer cells. However, in cancer cells, these mechanisms can be altered to promote tumour development and progression, whereby the functions of cytoskeletal proteins are co-opted to facilitate increased migrative and invasive capabilities, proliferation, as well as resistance to cellular and environmental stresses. Herein, we discuss the cytoskeletal responses to important intracellular stresses (such as mitochondrial, endoplasmic reticulum and oxidative stresses), and delineate the consequences of these responses, including effects on oncogenic signalling. In addition, we elaborate how the cytoskeleton and its associated molecules present themselves as therapeutic targets. The potential and limitations of targeting new classes of cytoskeletal proteins are also explored, in the context of developing novel strategies that impact cancer progression.
Collapse
Affiliation(s)
- Mei Shan Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (M.S.O.); (S.D.); (C.E.H.)
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (M.S.O.); (S.D.); (C.E.H.)
| | - Clarissa Esmeralda Halim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (M.S.O.); (S.D.); (C.E.H.)
| | - Wanpei Cai
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore (T.Z.T.); (R.Y.-J.H.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore (T.Z.T.); (R.Y.-J.H.)
| | - Ruby Yun-Ju Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore (T.Z.T.); (R.Y.-J.H.)
- School of Medicine, College of Medicine, National Taiwan University, No. 1 Ren Ai Road Sec. 1, Taipei City 10617, Taiwan
- Department of Obstetrics and Gynaecology, National University Hospital, National University Health System, Singapore 119074, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Medical Science Cluster, Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Shing Chuan Hooi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (M.S.O.); (S.D.); (C.E.H.)
- Medical Science Cluster, Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Correspondence: (S.C.H.); (A.P.K.); (C.T.Y.); Tel.: +65-6516-3294 (S.C.H. & C.T.Y.); +65-6873-5456 (A.P.K.); Fax: +65-6778-8161 (S.C.H. & C.T.Y.); +65-6873-9664 (A.P.K.)
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore (T.Z.T.); (R.Y.-J.H.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Medical Science Cluster, Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
- Correspondence: (S.C.H.); (A.P.K.); (C.T.Y.); Tel.: +65-6516-3294 (S.C.H. & C.T.Y.); +65-6873-5456 (A.P.K.); Fax: +65-6778-8161 (S.C.H. & C.T.Y.); +65-6873-9664 (A.P.K.)
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (M.S.O.); (S.D.); (C.E.H.)
- Medical Science Cluster, Cancer Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
- Correspondence: (S.C.H.); (A.P.K.); (C.T.Y.); Tel.: +65-6516-3294 (S.C.H. & C.T.Y.); +65-6873-5456 (A.P.K.); Fax: +65-6778-8161 (S.C.H. & C.T.Y.); +65-6873-9664 (A.P.K.)
| |
Collapse
|
16
|
Colchicine-Binding Site Inhibitors from Chemistry to Clinic: A Review. Pharmaceuticals (Basel) 2020; 13:ph13010008. [PMID: 31947889 PMCID: PMC7168938 DOI: 10.3390/ph13010008] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
It is over 50 years since the discovery of microtubules, and they have become one of the most important drug targets for anti-cancer therapies. Microtubules are predominantly composed of the protein tubulin, which contains a number of different binding sites for small-molecule drugs. There is continued interest in drug development for compounds targeting the colchicine-binding site of tubulin, termed colchicine-binding site inhibitors (CBSIs). This review highlights CBSIs discovered through diverse sources: from natural compounds, rational design, serendipitously and via high-throughput screening. We provide an update on CBSIs reported in the past three years and discuss the clinical status of CBSIs. It is likely that efforts will continue to develop CBSIs for a diverse set of cancers, and this review provides a timely update on recent developments.
Collapse
|
17
|
Arnst KE, Banerjee S, Chen H, Deng S, Hwang DJ, Li W, Miller DD. Current advances of tubulin inhibitors as dual acting small molecules for cancer therapy. Med Res Rev 2019; 39:1398-1426. [PMID: 30746734 DOI: 10.1002/med.21568] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 12/25/2022]
Abstract
Microtubule (MT)-targeting agents are highly successful drugs as chemotherapeutic agents, and this is attributed to their ability to target MT dynamics and interfere with critical cellular functions, including, mitosis, cell signaling, intracellular trafficking, and angiogenesis. Because MT dynamics vary in the different stages of the cell cycle, these drugs tend to be the most effective against mitotic cells. While this class of drug has proven to be effective against many cancer types, significant hurdles still exist and include overcoming aspects such as dose limited toxicities and the development of resistance. Newer generations of developed drugs attack these problems and alternative approaches such as the development of dual tubulin and kinase inhibitors are being investigated. This approach offers the potential to show increased efficacy and lower toxicities. This review covers different categories of MT-targeting agents, recent advances in dual inhibitors, and current challenges for this drug target.
Collapse
Affiliation(s)
- Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Souvik Banerjee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
18
|
Abstract
The nitro group is considered to be a versatile and unique functional group in medicinal chemistry. Despite a long history of use in therapeutics, the nitro group has toxicity issues and is often categorized as a structural alert or a toxicophore, and evidence related to drugs containing nitro groups is rather contradictory. In general, drugs containing nitro groups have been extensively associated with mutagenicity and genotoxicity. In this context, efforts toward the structure-mutagenicity or structure-genotoxicity relationships have been undertaken. The current Perspective covers various aspects of agents that contain nitro groups, their bioreductive activation mechanisms, their toxicities, and approaches to combat their toxicity issues. In addition, recent advances in the field of anticancer, antitubercular and antiparasitic agents containing nitro groups, along with a patent survey on hypoxia-activated prodrugs containing nitro groups, are also covered.
Collapse
Affiliation(s)
- Kunal Nepali
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Hsueh-Yun Lee
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| | - Jing-Ping Liou
- School of Pharmacy, College of Pharmacy , Taipei Medical University , 250 Wuxing Street , Taipei 11031 , Taiwan
| |
Collapse
|
19
|
Identification and characterization of SSE15206, a microtubule depolymerizing agent that overcomes multidrug resistance. Sci Rep 2018; 8:3305. [PMID: 29459693 PMCID: PMC5818492 DOI: 10.1038/s41598-018-21642-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/06/2018] [Indexed: 12/31/2022] Open
Abstract
Microtubules are highly dynamic structures that form spindle fibres during mitosis and are one of the most validated cancer targets. The success of drugs targeting microtubules, however, is often limited by the development of multidrug resistance. Here we describe the discovery and characterization of SSE15206, a pyrazolinethioamide derivative [3-phenyl-5-(3,4,5-trimethoxyphenyl)-4,5-dihydro-1H-pyrazole-1-carbothioamide] that has potent antiproliferative activities in cancer cell lines of different origins and overcomes resistance to microtubule-targeting agents. Treatment of cells with SSE15206 causes aberrant mitosis resulting in G2/M arrest due to incomplete spindle formation, a phenotype often associated with drugs that interfere with microtubule dynamics. SSE15206 inhibits microtubule polymerization both in biochemical and cellular assays by binding to colchicine site in tubulin as shown by docking and competition studies. Prolonged treatment of cells with the compound results in apoptotic cell death [increased Poly (ADP-ribose) polymerase cleavage and Annexin V/PI staining] accompanied by p53 induction. More importantly, we demonstrate that SSE15206 is able to overcome resistance to chemotherapeutic drugs in different cancer cell lines including multidrug-resistant KB-V1 and A2780-Pac-Res cell lines overexpressing MDR-1, making it a promising hit for the lead optimization studies to target multidrug resistance.
Collapse
|
20
|
New insights into Vinca alkaloids resistance mechanism and circumvention in lung cancer. Biomed Pharmacother 2017; 96:659-666. [DOI: 10.1016/j.biopha.2017.10.041] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 12/22/2022] Open
|
21
|
Bai Z, Gao M, Zhang H, Guan Q, Xu J, Li Y, Qi H, Li Z, Zuo D, Zhang W, Wu Y. BZML, a novel colchicine binding site inhibitor, overcomes multidrug resistance in A549/Taxol cells by inhibiting P-gp function and inducing mitotic catastrophe. Cancer Lett 2017; 402:81-92. [PMID: 28576750 DOI: 10.1016/j.canlet.2017.05.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/18/2017] [Accepted: 05/18/2017] [Indexed: 02/07/2023]
Abstract
Multidrug resistance (MDR) interferes with the efficiency of chemotherapy. Therefore, developing novel anti-cancer agents that can overcome MDR is necessary. Here, we screened a series of colchicine binding site inhibitors (CBSIs) and found that 5-(3, 4, 5-trimethoxybenzoyl)-4-methyl-2-(p-tolyl) imidazol (BZML) displayed potent cytotoxic activity against both A549 and A549/Taxol cells. We further explored the underlying mechanisms and found that BZML caused mitosis phase arrest by inhibiting tubulin polymerization in A549 and A549/Taxol cells. Importantly, BZML was a poor substrate for P-glycoprotein (P-gp) and inhibited P-gp function by decreasing P-gp expression at the protein and mRNA levels. Cell morphology changes and the expression of cycle- or apoptosis-related proteins indicated that BZML mainly drove A549/Taxol cells to die by mitotic catastrophe (MC), a p53-independent apoptotic-like cell death, whereas induced A549 cells to die by apoptosis. Taken together, our data suggest that BZML is a novel colchicine binding site inhibitor and overcomes MDR in A549/Taxol cells by inhibiting P-gp function and inducing MC. Our study also offers a new strategy to solve the problem of apoptosis-resistance.
Collapse
Affiliation(s)
- Zhaoshi Bai
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Meiqi Gao
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Huijuan Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Jingwen Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yao Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Huan Qi
- Scientific Research Center for Translational Medicine, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Zhengqiang Li
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| |
Collapse
|
22
|
Roy S, Mohanty M, Pasayat S, Majumder S, Senthilguru K, Banerjee I, Reichelt M, Reuter H, Sinn E, Dinda R. Synthesis, structure and cytotoxicity of a series of Dioxidomolybdenum(VI) complexes featuring Salan ligands. J Inorg Biochem 2017; 172:110-121. [PMID: 28448877 DOI: 10.1016/j.jinorgbio.2017.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 04/09/2017] [Accepted: 04/12/2017] [Indexed: 01/02/2023]
Abstract
Seven hexacoordinated cis-dioxidomolybdenum(VI) complexes [MoO2L1-7] (1-7) derived from various tetradentate diamino bis(phenolato) "salan" ligands, N,N'-dimethyl-N,N'-bis-(2-hydroxy-3-X-5-Y-6-Z-benzyl)-1,2-diaminoethane {(X=Br, Y=Me, Z=H (H2L1); X=Me, YCl, Z=H (H2L2); X=iPr, Y=Cl, Z=Me (H2L3)} and N,N'-bis-(2-hydroxy-3-X-5-Y-6-Z-benzyl)-1,2-diaminopropane {(X=Y=tBu, Z=H (H2L4); X=Y=Me, Z=H (H2L5); X=iPr, YCl, Z=Me (H2L6); X=Y=Br, Z=H (H2L7)} containing O-N donor atoms, have been isolated and structurally characterized. The formation of cis-dioxidomolybdenum(VI) complexes was confirmed by elemental analysis, IR, UV-vis and NMR spectroscopy, ESI-MS and cyclic voltammetry. X-ray crystallography showed the O2N2 donor set to define an octahedral geometry in each case. The complexes (1-7) were tested for their in vitro antiproliferative activity against HT-29 and HeLa cancer cell line. IC50 values of the complexes in HT-29 follow the order 6<7<<1<2<5<<3<4 while the order was 6<7<5<1<<3<4<2 in HeLa cells. Some of the complexes proved to be as active as the clinical referred drugs, and the greater potency of 6 and 7 (IC50 values of 6 are 2.62 and 10.74μM and that of 7 is 11.79 and 30.48μM in HT-29 and HeLa cells, respectively) may be dependent on the substituents in the salan ligand environment coordinated to the metal.
Collapse
Affiliation(s)
- Satabdi Roy
- Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Monalisa Mohanty
- Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Sagarika Pasayat
- Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Sudarshana Majumder
- Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Kulanthaivel Senthilguru
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Indranil Banerjee
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Martin Reichelt
- Institute of Chemistry of New Materials, University of Osnabrück, Barbarastraße 6, 49069 Osnabrück, Germany
| | - Hans Reuter
- Institute of Chemistry of New Materials, University of Osnabrück, Barbarastraße 6, 49069 Osnabrück, Germany
| | - Ekkehard Sinn
- Department of Chemistry, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Rupam Dinda
- Department of Chemistry, National Institute of Technology, Rourkela 769008, Odisha, India.
| |
Collapse
|
23
|
Döbber A, Phoa AF, Abbassi RH, Stringer BW, Day BW, Johns TG, Abadleh M, Peifer C, Munoz L. Development and Biological Evaluation of a Photoactivatable Small Molecule Microtubule-Targeting Agent. ACS Med Chem Lett 2017; 8:395-400. [PMID: 28435525 DOI: 10.1021/acsmedchemlett.6b00483] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/15/2017] [Indexed: 01/21/2023] Open
Abstract
Photoremovable protecting groups added to bioactive molecules provide spatial and temporal control of the biological effects. We present synthesis and characterization of the first photoactivatable small-molecule tubulin inhibitor. By blocking the pharmacophoric OH group on compound 1 with photoremovable 4,5-dimethoxy-2-nitrobenzyl moiety we developed the photocaged prodrug 2 that had no effect in biological assays. Short UV light exposure of the derivative 2 or UV-irradiation of cells treated with 2 resulted in fast and potent inhibition of tubulin polymerization, attenuation of cell viability, and apoptotic cell death, implicating release of the parent active compound. This study validates for the first time the photoactivatable prodrug concept in the field of small molecule tubulin inhibitors. The caged derivative 2 represents a novel tool in antitubulin approaches.
Collapse
Affiliation(s)
- Alexander Döbber
- School of Medical
Sciences and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße
76, 24118 Kiel, Germany
| | - Athena F. Phoa
- School of Medical
Sciences and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Ramzi H. Abbassi
- School of Medical
Sciences and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Brett W. Stringer
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
| | - Bryan W. Day
- QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD 4006, Australia
| | - Terrance G. Johns
- Oncogenic Signalling Laboratory and Brain
Cancer Discovery Collaborative, Centre for Cancer Research, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168, Australia
- Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | - Mohammed Abadleh
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße
76, 24118 Kiel, Germany
| | - Christian Peifer
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße
76, 24118 Kiel, Germany
| | - Lenka Munoz
- School of Medical
Sciences and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|