1
|
Szymaszek P, Tyszka-Czochara M, Ortyl J. Iridium(III) complexes as novel theranostic small molecules for medical diagnostics, precise imaging at a single cell level and targeted anticancer therapy. Eur J Med Chem 2024; 276:116648. [PMID: 38968786 DOI: 10.1016/j.ejmech.2024.116648] [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: 05/14/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Medical applications of iridium (III) complexes include their use as state-of-the-art theranostic agents - molecules that combine therapeutic and diagnostic functions into a single entity. These complexes offer a promising avenue in medical diagnostics, precision imaging at single-cell resolution and targeted anticancer therapy due to their unique properties. In this review we report a short summary of their application in medical diagnostics, imaging at single-cell level and targeted anticancer therapy. The exceptional photophysical properties of Iridium (III) complexes, including their brightness and photostability, make them excellent candidates for bioimaging. They can be used to image cellular processes and the microenvironment within single cells with unprecedented clarity, aiding in the understanding of disease mechanisms at the molecular level. Moreover the iridium (III) complexes can be designed to selectively target cancer cells,. Upon targeting, these complexes can act as photosensitizers for photodynamic therapy (PDT), generating reactive oxygen species (ROS) upon light activation to induce cell death. The integration of diagnostic and therapeutic capabilities in Iridium (III) complexes offers the potential for a holistic approach to cancer treatment, enabling not only the precise eradication of cancer cells but also the real-time monitoring of treatment efficacy and disease progression. This aligns with the goals of personalized medicine, offering hope for more effective and less invasive cancer treatment strategies.
Collapse
Affiliation(s)
- Patryk Szymaszek
- Department of Biotechnology and Physical Chemistry, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155, Kraków, Poland
| | | | - Joanna Ortyl
- Department of Biotechnology and Physical Chemistry, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155, Kraków, Poland; Photo HiTech Ltd., Bobrzyńskiego 14, 30-348, Kraków, Poland; Photo4Chem ltd., Juliusza Lea 114/416A-B, 31-133, Kraków, Poland.
| |
Collapse
|
2
|
Ruan ML, Ni WX, Chu JCH, Lam TL, Law KC, Zhang Y, Yang G, He Y, Zhang C, Fung YME, Liu T, Huang T, Lok CN, Chan SLF, Che CM. Iridium(III) carbene complexes as potent girdin inhibitors against metastatic cancers. Proc Natl Acad Sci U S A 2024; 121:e2316615121. [PMID: 38861602 PMCID: PMC11194514 DOI: 10.1073/pnas.2316615121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 04/27/2024] [Indexed: 06/13/2024] Open
Abstract
Many cancer-driving protein targets remain undruggable due to a lack of binding molecular scaffolds. In this regard, octahedral metal complexes with unique and versatile three-dimensional structures have rarely been explored as inhibitors of undruggable protein targets. Here, we describe antitumor iridium(III) pyridinium-N-heterocyclic carbene complex 1a, which profoundly reduces the viability of lung and breast cancer cells as well as cancer patient-derived organoids at low micromolar concentrations. Compound 1a effectively inhibits the growth of non-small-cell lung cancer and triple-negative breast cancer xenograft tumors, impedes the metastatic spread of breast cancer cells, and can be modified into an antibody-drug conjugate payload to achieve precise tumor delivery in mice. Identified by thermal proteome profiling, an important molecular target of 1a in cellulo is Girdin, a multifunctional adaptor protein that is overexpressed in cancer cells and unequivocally serves as a signaling hub for multiple pivotal oncogenic pathways. However, specific small-molecule inhibitors of Girdin have not yet been developed. Notably, 1a exhibits high binding affinity to Girdin with a Kd of 1.3 μM and targets the Girdin-linked EGFR/AKT/mTOR/STAT3 cancer-driving pathway, inhibiting cancer cell proliferation and metastatic activity. Our study reveals a potent Girdin-targeting anticancer compound and demonstrates that octahedral metal complexes constitute an untapped library of small-molecule inhibitors that can fit into the ligand-binding pockets of key oncoproteins.
Collapse
Affiliation(s)
- Mei-Ling Ruan
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
- National Key Laboratory of Green Pesticide, College of Chemistry, Central China Normal University, Wuhan430079, China
| | - Wen-Xiu Ni
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou515041, Guangdong, China
- Chemistry and Chemical Engineering of Guangdong Laboratory, Shantou515041, Guangdong, China
| | - Jacky C. H. Chu
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Tsz-Lung Lam
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kwok-Chung Law
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yiwei Zhang
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Guanya Yang
- AI And Life Sciences Institute (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Ying He
- AI And Life Sciences Institute (Hong Kong) Limited, Hong Kong Science Park, Shatin, Hong Kong, China
| | - Chunlei Zhang
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yi Man Eva Fung
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Tao Liu
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou515041, Guangdong, China
- Chemistry and Chemical Engineering of Guangdong Laboratory, Shantou515041, Guangdong, China
| | - Tao Huang
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou515041, Guangdong, China
- Chemistry and Chemical Engineering of Guangdong Laboratory, Shantou515041, Guangdong, China
| | - Chun-Nam Lok
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Sharon Lai-Fung Chan
- Department of Applied Biology and Chemical Biology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Chi-Ming Che
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, Shatin, Hong Kong, China
- State Key Laboratory of Synthetic Chemistry and Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
3
|
Wang L, Wang X, Chen F, Song YQ, Nao SC, Chan DSH, Wong CY, Wang W, Leung CH. A glycyrrhetinic acid-iridium(III) conjugate as a theranostic NIR probe for hepatocellular carcinoma with mitochondrial-targeting ability. Eur J Med Chem 2024; 264:115995. [PMID: 38043488 DOI: 10.1016/j.ejmech.2023.115995] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/05/2023]
Abstract
Hepatocellular carcinoma (HCC) is a major contributor to global mortality rates, but current treatment options have limitations. Advanced theranostics are needed to effectively integrate diagnosis and therapeutic of HCC. Glycyrrhetinic acid (GA) has abundant binding sites with glycyrrhetinic acid receptors (GA-Rs) on the surface of HCC cells and has also been reported to possess ligands with mitochondrial-targeting capability but with limited efficacy. Herein, we report a near-infrared (NIR) luminescent theranostic complex 1 through conjugating an iridium(III) complex to GA, which exhibits the desired photophysical properties and promotes mitochondrial-targeting capability. Complex 1 was selectively taken up by HepG2 liver cancer cells and was imaged within mitochondria with NIR emission. Complex 1 targeted mitochondria and opened mitochondrial permeability transition pores (MPTPs), resulting in ROS accumulation, mitochondrial damage, disruption of Bax/Bcl-2 equilibrium, and tumor cell apoptosis, resulting in significantly improved anticancer activity compared to GA. This work offers a methodology for developing multifunctional theranostic probes with amplified specificity and efficacy.
Collapse
Affiliation(s)
- Ling Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Xueliang Wang
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 South Gaoxin Road, Shenzhen, 518057, China
| | - Feng Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Ying-Qi Song
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Sang-Cuo Nao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | | | - Chun-Yuen Wong
- Department of Chemistry, City University of Hong Kong, Hong Kong, 999077, China
| | - Wanhe Wang
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, Shaanxi, 710072, China; Research & Development Institute of Northwestern Polytechnical University in Shenzhen, 45 South Gaoxin Road, Shenzhen, 518057, China.
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao, 999078, China; Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, 999078, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, 999078, China.
| |
Collapse
|
4
|
Bai Y, Aodeng G, Ga L, Hai W, Ai J. Research Progress of Metal Anticancer Drugs. Pharmaceutics 2023; 15:2750. [PMID: 38140091 PMCID: PMC10747151 DOI: 10.3390/pharmaceutics15122750] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer treatments, including traditional chemotherapy, have failed to cure human malignancies. The main reasons for the failure of these treatments are the inevitable drug resistance and serious side effects. In clinical treatment, only 5 percent of the 50 percent of cancer patients who are able to receive conventional chemotherapy survive. Because of these factors, being able to develop a drug and treatment that can target only cancer cells without affecting normal cells remains a big challenge. Since the special properties of cisplatin in the treatment of malignant tumors were accidentally discovered in the last century, metal anticancer drugs have become a research hotspot. Metal anticancer drugs have unique pharmaceutical properties, such as ruthenium metal drugs with their high selectivity, low toxicity, easy absorption by tumor tissue, excretion, and so on. In recent years, efficient and low-toxicity metal antitumor complexes have been synthesized. In this paper, the scientific literature on platinum (Pt), ruthenium (Ru), iridium (Ir), gold (Au), and other anticancer complexes was reviewed by referring to a large amount of relevant literature at home and abroad.
Collapse
Affiliation(s)
- Yun Bai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| | - Gerile Aodeng
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| | - Lu Ga
- College of Pharmacy, Inner Mongolia Medical University, Jinchuankaifaqu, Hohhot 010110, China;
| | - Wenfeng Hai
- Inner Mongolia Key Laboratory of Carbon Nanomaterials, Nano Innovation Institute (NII), College of Chemistry and Materials Science, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Jun Ai
- Inner Mongolia Key Laboratory of Environmental Chemistry, College of Chemistry and Enviromental Science, Inner Mongolia Normal University, 81 Zhaowudalu, Hohhot 010022, China; (Y.B.); (G.A.)
| |
Collapse
|
5
|
Yang Y, Gao Y, Sun Y, Zhao J, Gou S. Study on the Multimodal Anticancer Mechanism of Ru(II)/Ir(III) Complexes Bearing a Poly(ADP-ribose) Polymerase 1 Inhibitor. J Med Chem 2023; 66:13731-13745. [PMID: 37788351 DOI: 10.1021/acs.jmedchem.3c01156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
A series of novel ruthenium(II) and iridium(III) complexes (Ru1-Ru3 and Ir1-Ir3) with different ancillary ligands and a PARP-1-inhibitory chelating ligand 2-(2,3-dibromo-4,5-dimethoxybenzylidene)hydrazine-1-carbothioamide (L1) were designed and prepared. The target complexes were structurally characterized by NMR and ESI-MS techniques. Among them, the crystal and molecular structures of Ir1 and Ir2 were also determined by X-ray crystallography. These complexes retained the PARP-1 enzyme inhibitory effect of L1 and showed potent antiproliferative activity on the tested cancer cell lines. The ruthenium(II) complexes Ru1-Ru3 were found to be more cytotoxic than the iridium(III) complexes Ir1-Ir3. Further investigations revealed that the most active complex Ru3 induced apoptosis in MCF-7 cells by multiple modes, inclusive of inducing DNA damage, suppressing DNA damage repair, disturbing cell cycle distribution, decreasing the mitochondrial membrane potential, and increasing the intracellular reactive oxygen species levels.
Collapse
Affiliation(s)
- Yuliang Yang
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Ya Gao
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Yanyan Sun
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jian Zhao
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| | - Shaohua Gou
- Pharmaceutical Research Center and School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, Southeast University, Nanjing 211189, China
| |
Collapse
|
6
|
Yu J, Zhong B, Zhao L, Hou Y, Ai N, Lu JJ, Ge W, Chen X. Fighting drug-resistant lung cancer by induction of NAD(P)H:quinone oxidoreductase 1 (NQO1)-mediated ferroptosis. Drug Resist Updat 2023; 70:100977. [PMID: 37321064 DOI: 10.1016/j.drup.2023.100977] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/17/2023]
Abstract
Drug resistance is a major challenge in cancer treatment. The substrates of NAD(P)H:quinone oxidoreductase 1 (NQO1) show a promising anticancer effect in clinical trials. We previously identified a natural NQO1 substrate 2-methoxy-6-acetyl-7-methyljuglone (MAM) with a potent anticancer effect. The present study was designed to explore the efficacy of MAM in fighting against drug-resistant non-small cell lung cancer (NSCLC). The anticancer effect of MAM was evaluated in cisplatin-resistant A549 and AZD9291-resistant H1975 cells. The interaction of MAM with NQO1 was measured by cellular thermal shift assay and drug affinity responsive target stability assay. The NQO1 activity and expression were measured using NQO1 recombinant protein, Western blotting, and immunofluorescence staining assay. The roles of NQO1 were examined by NQO1 inhibitor, small interfering RNA (siRNA), and short hairpin RNA (shRNA). The roles of reactive oxygen species (ROS), labile iron pool (LIP), and lipid peroxidation were determined. MAM induced significant cell death in drug-resistant cells with similar potency to that of parental cells, which were completely abolished by NQO1 inhibitor, NQO1 siRNA, and iron chelators. MAM activates and binds to NQO1, which triggers ROS generation, LIP increase, and lipid peroxidation. MAM significantly suppressed tumor growth in the tumor xenograft zebrafish model. These results showed that MAM induced ferroptosis by targeting NQO1 in drug-resistant NSCLC cells. Our findings provided a novel therapeutic strategy for fighting against drug resistance by induction of NQO1-mediated ferroptosis.
Collapse
Affiliation(s)
- Jie Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao Special Administrative Region of China; School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Bingling Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao Special Administrative Region of China
| | - Lin Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao Special Administrative Region of China
| | - Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao Special Administrative Region of China
| | - Nana Ai
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region of China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao Special Administrative Region of China
| | - Wei Ge
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region of China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao Special Administrative Region of China; Department of Pharmaceutical Sciences, Faculty of Health Scien ces, University of Macau, Taipa, Macao Special Administrative Region of China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao Special Administrative Region of China.
| |
Collapse
|
7
|
Kanbe A, Yokoi K, Yamada Y, Tsurui M, Kitagawa Y, Hasegawa Y, Ogata D, Yuasa J, Aoki S. Optical Resolution of Carboxylic Acid Derivatives of Homoleptic Cyclometalated Iridium(III) Complexes via Diastereomers Formed with Chiral Auxiliaries. Inorg Chem 2023; 62:11325-11341. [PMID: 37432912 PMCID: PMC10369494 DOI: 10.1021/acs.inorgchem.3c00685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Indexed: 07/13/2023]
Abstract
We report on a facile method for the optical resolution of cyclometalated iridium(III) (Ir(III)) complexes via diastereomers formed with chiral auxiliaries. The racemic carboxylic acids of Ir(III) complexes (fac-4 (fac-Ir(ppyCO2H)3 (ppy: 2-phenylpyridine)), fac-6 (fac-Ir(tpyCO2H)3 (tpy: 2-(4'-tolyl)pyridine)), and fac-13 (fac-Ir(mpiqCO2H)3 (mpiq: 1-(4'-methylphenyl)isoquinoline))) were converted into the diastereomers, Δ- and Λ-forms of fac-9 (from fac-6), fac-10 (from fac-4), fac-11 (from fac-6), and fac-14 (from fac-13), respectively, by the condensation with (1R,2R)-1,2-diaminocyclohexane or (1R,2R)-2-aminocyclohexanol. The resulting diastereomers were separated by HPLC (with a nonchiral column) or silica gel column chromatography, and their absolute stereochemistry was determined by X-ray single-crystal structure analysis and CD (circular dichroism) spectra. Spectra of all diastereomers of the Ir(III) complexes are reported. Hydrolysis of the ester moieties of Δ- and Λ-forms of fac-10, fac-11, and fac-14 gave both enantiomers of the corresponding carboxylic acid derivatives in the optically pure forms, Δ-fac and Λ-fac-4, -6, and -13, respectively.
Collapse
Affiliation(s)
- Azusa Kanbe
- Faculty
of Pharmaceutical Science, Tokyo University
of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Kenta Yokoi
- Faculty
of Pharmaceutical Science, Tokyo University
of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Yasuyuki Yamada
- Department
of Chemistry, Graduate School of Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
- Research
Center for Materials Science, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8602, Japan
- JST,
PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Makoto Tsurui
- Graduate
School of Chemical Sciences and Engineering, Hokkaido University, N13W8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| | - Yuichi Kitagawa
- Faculty of
Engineering, Hokkaido University, Kita-13, Nishi-8, Kita-Ku, Sapporo, Hokkaido 060-8628, Japan
- Institute
for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Kita-21, Nishi-10, Kita-Ku, Sapporo, Hokkaido 001-0021, Japan
| | - Yasuchika Hasegawa
- Faculty of
Engineering, Hokkaido University, Kita-13, Nishi-8, Kita-Ku, Sapporo, Hokkaido 060-8628, Japan
- Institute
for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Kita-21, Nishi-10, Kita-Ku, Sapporo, Hokkaido 001-0021, Japan
| | - Daiji Ogata
- Faculty
of Science, Tokyo University of Science, 1-3 Kagurazaka,
Shinjuku-ku, Tokyo 162-8601, Japan
| | - Junpei Yuasa
- Faculty
of Science, Tokyo University of Science, 1-3 Kagurazaka,
Shinjuku-ku, Tokyo 162-8601, Japan
| | - Shin Aoki
- Faculty
of Pharmaceutical Science, Tokyo University
of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Research
Institute for Science and Technology (RIST), Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
- Research
Institute for Biomedical Science (RIBS), Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| |
Collapse
|
8
|
Harwood SJ, Smith CR, Lawson JD, Ketcham JM. Selected Approaches to Disrupting Protein-Protein Interactions within the MAPK/RAS Pathway. Int J Mol Sci 2023; 24:ijms24087373. [PMID: 37108538 PMCID: PMC10139024 DOI: 10.3390/ijms24087373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Within the MAPK/RAS pathway, there exists a plethora of protein-protein interactions (PPIs). For many years, scientists have focused efforts on drugging KRAS and its effectors in hopes to provide much needed therapies for patients with KRAS-mutant driven cancers. In this review, we focus on recent strategies to inhibit RAS-signaling via disrupting PPIs associated with SOS1, RAF, PDEδ, Grb2, and RAS.
Collapse
Affiliation(s)
| | | | - J David Lawson
- Mirati Therapeutics, 3545 Cray Court, San Diego, CA 92121, USA
| | - John M Ketcham
- Mirati Therapeutics, 3545 Cray Court, San Diego, CA 92121, USA
| |
Collapse
|
9
|
Lim JM, Supianto M, Kim TY, Kim BS, Park JW, Jang HH, Lee HJ. Fluorescent Lateral Flow Assay with Carbon Nanodot Conjugates for Carcinoembryonic Antigen. BIOCHIP JOURNAL 2023. [DOI: 10.1007/s13206-022-00093-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
10
|
Shahabadi N, Zendehcheshm S, Mahdavi M. Exploring the In‐Vitro Antibacterial Activity and Protein (Human Serum Albumin, Human Hemoglobin and Lysozyme) Interaction of Hexagonal Silver Nanoparticle Obtained from Wood Extract of Wild Cherry Shrub. ChemistrySelect 2023. [DOI: 10.1002/slct.202204672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Nahid Shahabadi
- Inorganic Chemistry Department, Faculty of Chemistry Razi University Kermanshah Iran
- Medical Biology Research Center (MBRC) Kermanshah University of Medical Sciences Kermanshah Iran
| | - Saba Zendehcheshm
- Inorganic Chemistry Department, Faculty of Chemistry Razi University Kermanshah Iran
- Medical Biology Research Center (MBRC) Kermanshah University of Medical Sciences Kermanshah Iran
| | - Mohammad Mahdavi
- Inorganic Chemistry Department, Faculty of Chemistry Razi University Kermanshah Iran
| |
Collapse
|
11
|
Zhao J, Gao Y, He W, Wang W, Hu W, Sun Y. Synthesis, characterization and biological evaluation of two cyclometalated iridium(III) complexes containing a glutathione S-transferase inhibitor. J Inorg Biochem 2023; 238:112050. [PMID: 36332411 DOI: 10.1016/j.jinorgbio.2022.112050] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/12/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
The cyclometalated iridium(III) compounds have been intensively studied for health-related applications due to their outstanding luminescent properties and multiple anticancer modes of action. Herein, two iridium(III) compounds Ir-1 and Ir-3 containing glutathione S-transferase inhibitor (GSTi) were developed and studied together with two unfunctionalized compounds Ir-2 and Ir-4 as a comparison. Biological study indicated that GSTi-bearing complexes Ir-1 and Ir-3 exert a synergistic effect on the inhibition of cancer cells. The photophysical properties of Ir-1 ∼ Ir-4 were investigated by UV/vis absorption and fluorescence spectroscopy and rationalized with TD-DFT calculations. As expected, GSTi-bearing complexes Ir-1 and Ir-3 exhibited considerable cytotoxicity against both A549 and cisplatin-resistant A549/cis cancer cells, much higher than the unfunctionalized iridium compounds Ir-2 and Ir-4. Further study indicated that Ir-1 and Ir-3 mainly localize in the mitochondria of tumor cells, and exert their cytotoxicity via generating ROS and inhibiting GST activity. The flow cytometry investigations demonstrated that Ir-1 and Ir-3 can arrest the cell cycle in S phase and induce the cell death through apoptosis process. Overall, the complexation of GST inhibitors with cyclometalated iridium(III) agents provides an effective way for potentiating the cytotoxicity of iridium(III) anticancer agents and resensitizing the efficacy against cisplatin resistant cancer cells.
Collapse
Affiliation(s)
- Jian Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China; Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Ya Gao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Weiyu He
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Wei Wang
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Weiwei Hu
- Jiangsu Key Laboratory of Regional Resource Exploitation and Medicinal Research, Huaiyin Institute of Technology, Huaian 223003, China.
| | - Yanyan Sun
- School of Chemistry and Life Sciences, Suzhou University of Science and Technology, Suzhou 215009, China.
| |
Collapse
|
12
|
Hu X, Guo L, Liu M, Zhang Q, Gong Y, Sun M, Feng S, Xu Y, Liu Y, Liu Z. Increasing Anticancer Activity with Phosphine Ligation in Zwitterionic Half-Sandwich Iridium(III), Rhodium(III), and Ruthenium(II) Complexes. Inorg Chem 2022; 61:20008-20025. [PMID: 36426422 DOI: 10.1021/acs.inorgchem.2c03279] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The synthesis and biological assessment of neutral or cationic platinum group metal-based anticancer complexes have been extremely studied, whereas there are few reports on the corresponding zwitterionic complexes. Herein, the synthesis, characterization, and bioactivity of zwitterionic half-sandwich phosphine-imine iridium(III), rhodium(III), and ruthenium(II) complexes were presented. The sulfonated phosphine-imine ligand and a group of zwitterionic half-sandwich P,N-chelating organometallic complexes were fully characterized by nuclear magnetic resonance (NMR), mass spectrum (electrospray ionization, ESI), elemental analysis, and X-ray crystallography. The solution stability of these complexes and their spectral properties were also determined. Notably, almost all of these complexes showed enhanced anticancer activity against model HeLa and A549 cancer cells than the corresponding zwitterionic pyridyl-imine N,N-chelating iridium(III) and ruthenium(II) complexes, which have exhibited inactive or low active in our previous work. The increase in the lipophilic property and intracellular uptake levels of these zwitterionic P,N-chelating complexes appeared to be associated with their superior cytotoxicity. In addition, these complexes showed biomolecular interactions with bovine serum albumin (BSA). The flow cytometry studies indicated that the representative complex Ir1 could induce early-stage apoptosis in A549 cells. Further, confocal microscopy imaging analysis displayed that Ir1 entered A549 cells through the energy-dependent pathway, targeted lysosome, and could cause lysosomal damage. In particular, these complexes could impede cell migration in A549 cells.
Collapse
Affiliation(s)
- Xueyan Hu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Lihua Guo
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Mengqi Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Qiuya Zhang
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yuwen Gong
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Mengru Sun
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Shenghan Feng
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Youzhi Xu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yiming Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Zhe Liu
- School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| |
Collapse
|
13
|
Graf M, Böttcher H, Mayer P, Metzler‐Nolte N, Thavalingam S, Czerwieniec R. Cytotoxic Activities of Bis‐cyclometalated Iridium(III) Complexes Containing Chloro‐substituted κ
2
N‐terpyridines. Z Anorg Allg Chem 2022. [DOI: 10.1002/zaac.202200047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Marion Graf
- Department Chemie Ludwig-Maximilians-Universität Butenandtstrasse 5–13 (D) 1377 München Germany
| | - Hans‐Christian Böttcher
- Department Chemie Ludwig-Maximilians-Universität Butenandtstrasse 5–13 (D) 1377 München Germany
| | - Peter Mayer
- Department Chemie Ludwig-Maximilians-Universität Butenandtstrasse 5–13 (D) 1377 München Germany
| | - Nils Metzler‐Nolte
- Faculty for Chemistry and Biochemistry Chair of Inorganic Chemistry I – Bioinorganic Chemistry Ruhr University Bochum Universitätsstrasse 150 44801 Bochum Germany
| | - Sugina Thavalingam
- Faculty for Chemistry and Biochemistry Chair of Inorganic Chemistry I – Bioinorganic Chemistry Ruhr University Bochum Universitätsstrasse 150 44801 Bochum Germany
| | - Rafał Czerwieniec
- Institute of Physical and Theoretical Chemistry University Regensburg Universitätsstrasse 31 3053 Regensburg Germany
| |
Collapse
|
14
|
Tuning the emission maxima of iridium systems using benzimidazole-based cyclometallating framework. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2021.118446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
15
|
Chen C, Wang Y, Zhang D, Wang J. Construction of bifunctionalized Co-V mixed metal oxide nanosheets with Co3+-Rich surfaces and oxygen defect derived from LDHs as nanozyme for antibacterial application. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.09.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
16
|
|
17
|
Li G, Liu H, Feng R, Kang TS, Wang W, Ko CN, Wong CY, Ye M, Ma DL, Wan JB, Leung CH. A bioactive ligand-conjugated iridium(III) metal-based complex as a Keap1-Nrf2 protein-protein interaction inhibitor against acetaminophen-induced acute liver injury. Redox Biol 2021; 48:102129. [PMID: 34526248 PMCID: PMC8710994 DOI: 10.1016/j.redox.2021.102129] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/30/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023] Open
Abstract
Hepatotoxicity caused by an overdose of acetaminophen (APAP) is the leading reason for acute drug-related liver failure. Nuclear factor erythroid-2-related factor 2 (Nrf2) is a protein that helps to regulate redox homeostasis and coordinate stress responses via binding to the Kelch-like ECH-associated protein 1 (Keap1). Targeting the Keap1-Nrf2 interaction has recently emerged as a potential strategy to alleviate liver injury caused by APAP. Here, we designed and synthesized a number of iridium (III) and rhodium (III) complexes bearing ligands with reported activity against oxidative stress, which is associated with Nrf2 transcriptional activation. The iridium (III) complex 1 bearing a bioactive ligand 2,9-dimethyl-1,10-phenanthroline and 4-chloro-2-phenylquinoline, a derivative of the bioactive ligand 2-phenylquinoline, was identified as a direct small-molecule inhibitor of the Keap1–Nrf2 protein-protein interaction. 1 could stabilize Keap1 protein, upregulate HO-1 and NQO1, and promote Nrf2 nuclear translocation in normal liver cells. Moreover, 1 reversed APAP-induced liver damage by disrupting Keap1–Nrf2 interaction and without inducing organ damage and immunotoxicity in mice. Our study demonstrates the identification of a selective and efficacious antagonist of Keap1–Nrf2 interaction possessed good cellular permeability in cellulo and ideal pharmacokinetic parameters in vivo, and, more importantly, validates the feasibility of conjugating metal complexes with bioactive ligands to generate metal-based drug leads as non-toxic Keap1–Nrf2 interaction inhibitors for treating APAP-induced acute liver injury. 1 reversed APAP-induced liver damage by disrupting Keap1–Nrf2 interaction without inducing organ damage or immunotoxicity. Complex 1 possessed good cellular permeability in cellulo and ideal pharmacokinetic parameters in vivo. Conjugating metal complexes with bioactive ligands opens a novel avenue for the treatment of APAP-induced liver damage.
Collapse
Affiliation(s)
- Guodong Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Hao Liu
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Ruibing Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Tian-Shu Kang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Wanhe Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China; Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Chung-Nga Ko
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Chun-Yuen Wong
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Hong Kong SAR, China
| | - Min Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China.
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China.
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Macao SAR, China.
| |
Collapse
|
18
|
Ye RR, Peng W, Chen BC, Jiang N, Chen XQ, Mao ZW, Li RT. Mitochondria-targeted artesunate conjugated cyclometalated iridium(iii) complexes as potent anti-HepG2 hepatocellular carcinoma agents. Metallomics 2021; 12:1131-1141. [PMID: 32453319 DOI: 10.1039/d0mt00060d] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) poses a serious threat to people's health worldwide. Artesunate (ART), one of the classical antimalarial drugs, has recently been shown to exert significant cytotoxicity in various cancers, but its bioavailability is low. Cyclometalated iridium(iii) complexes have emerged as a promising class of anticancer therapeutic agents. Herein, through conjugation of two of them, three novel Ir(iii)-ART conjugates, [Ir(C-N)2(bpy-ART)](PF6) (bpy = 2,2'-bipyridine, C-N = 2-phenylpyridine (ppy, Ir-ART-1), 2-(2-thienyl)pyridine (thpy, Ir-ART-2), and 2-(2,4-difluorophenyl)pyridine (dfppy, Ir-ART-3)) have been synthesized, and their potential as anti-HCC agents was evaluated. We demonstrate that Ir-ART-1-3 display higher cytotoxicity against HCC cell lines than normal liver cells, and they can especially locate to mitochondria of HepG2 cells and induce a series of mitochondria-mediated apoptosis events. Moreover, Ir-ART-1-3 can regulate the cell cycle and inhibit metastasis of HepG2 cells. Finally, in vivo antitumor evaluation also demonstrates the inhibitory activity of Ir-ART-1 on tumor growth. Taken together, these Ir(iii)-ART conjugates have the potential to become drug candidates for future anti-HCC treatments.
Collapse
Affiliation(s)
- Rui-Rong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Wan Peng
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Bi-Chun Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Ning Jiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Xuan-Qin Chen
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, P. R. China.
| | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, P. R. China.
| |
Collapse
|
19
|
Nayeem N, Contel M. Exploring the Potential of Metallodrugs as Chemotherapeutics for Triple Negative Breast Cancer. Chemistry 2021; 27:8891-8917. [PMID: 33857345 DOI: 10.1002/chem.202100438] [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: 02/03/2021] [Indexed: 12/11/2022]
Abstract
This review focuses on studies of coordination and organometallic compounds as potential chemotherapeutics against triple negative breast cancer (TNBC) which has one of the poorest prognoses and worst survival rates from all breast cancer types. At present, chemotherapy is still the standard of care for TNBC since only one type of targeted therapy has been recently developed. References for metal-based compounds studied in TNBC cell lines will be listed, and those of metal-specific reviews, but a detailed overview will also be provided on compounds studied in vivo (mostly in mice models) and those compounds for which some preliminary mechanistic data was obtained (in TNBC cell lines and tumors) and/or for which bioactive ligands have been used. The main goal of this review is to highlight the most promising metal-based compounds with potential as chemotherapeutic agents in TNBC.
Collapse
Affiliation(s)
- Nazia Nayeem
- Brooklyn College Cancer Center BCCC-CURE, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Biology PhD Program, The Graduate Center, The City University of New York, 365 5th Avenue, New York, New York, 11006, USA
| | - Maria Contel
- Brooklyn College Cancer Center BCCC-CURE, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Department of Chemistry, Brooklyn College, The City University of New York, 2900 Bedford Avenue, Brooklyn, New York, 11210, USA.,Biology PhD Program, The Graduate Center, The City University of New York, 365 5th Avenue, New York, New York, 11006, USA.,Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, 365 5th Avenue, New York, New York, 11006, USA.,University of Hawaii Cancer Center, 701 Ilalo St, Honolulu, Hawaii, 96813, USA
| |
Collapse
|
20
|
Huang Y, Yang Z, Li F, Zhao H, Li C, Yu N, Hamilton DJ, Li Z. 64Cu/ 177Lu-DOTA-diZD, a Small-Molecule-Based Theranostic Pair for Triple-Negative Breast Cancer. J Med Chem 2021; 64:2705-2713. [PMID: 33646782 DOI: 10.1021/acs.jmedchem.0c01957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite advances in targeted therapies, the prognosis for patients with triple-negative breast cancer (TNBC) is poor because there are few actionable molecular targets. The dependence of solid tumor growth on angiogenesis prompted our development of angiogenic-receptor-targeted radionuclide therapy (TRT) to treat TNBC by targeted delivery of therapeutic doses of ionizing radiation to tumors. A high-affinity vascular endothelial growth factor receptor (VEGFR)-targeted agent, diZD, was synthesized and labeled with 177Lu and 64Cu by 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) chelator giving the TRT agent, 177Lu-DOTA-diZD, and PET imaging agent, 64Cu-DOTA-diZD. We showed that "64Cu/177Lu"-DOTA-diZD radiotracers are a promising theranostic pair for TNBC. 4T1-bearing mice treated with 177Lu-DOTA-diZD-based TRT survived with a median of 28 days, which was significantly longer than that of control mice as 18 days. Anti-PD1 immunotherapy resulted in a shorter median survival of 16 days. This work presents for the first time that small-molecule VEGFR-oriented TRT is a promising therapeutic option to treat "immunogenic cold" TNBC.
Collapse
Affiliation(s)
- Yuqian Huang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhen Yang
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Feng Li
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Hong Zhao
- Cancer Center, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Chun Li
- Departments of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Nam Yu
- Houston Radiology Associates and Department of Radiology, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Dale J Hamilton
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas 77030, United States
| | - Zheng Li
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas 77030, United States
| |
Collapse
|
21
|
Sun Q, Wang Y, Fu Q, Ouyang A, Liu S, Wang Z, Su Z, Song J, Zhang Q, Zhang P, Lu D. Sulfur‐Coordinated Organoiridium(III) Complexes Exert Breast Anticancer Activity via Inhibition of Wnt/β‐Catenin Signaling. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202015009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Qi Sun
- Guangdong Key Laboratory for Genome Stability & Disease Prevention International Cancer Center Department of Pharmacology Shenzhen University Health Science Center Shenzhen 518060 China
| | - Yi Wang
- College of Chemistry and Environmental Engineering Shenzhen University Shenzhen 518060 P. R. China
- Key Laboratory for Advanced Materials of MOE School of Chemistry & Molecular Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Qiuxia Fu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention International Cancer Center Department of Pharmacology Shenzhen University Health Science Center Shenzhen 518060 China
| | - Ai Ouyang
- College of Chemistry and Environmental Engineering Shenzhen University Shenzhen 518060 P. R. China
| | - Shanshan Liu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention International Cancer Center Department of Pharmacology Shenzhen University Health Science Center Shenzhen 518060 China
| | - Zhongyuan Wang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention International Cancer Center Department of Pharmacology Shenzhen University Health Science Center Shenzhen 518060 China
| | - Zijie Su
- Guangdong Key Laboratory for Genome Stability & Disease Prevention International Cancer Center Department of Pharmacology Shenzhen University Health Science Center Shenzhen 518060 China
| | - Jiaxing Song
- Guangdong Key Laboratory for Genome Stability & Disease Prevention International Cancer Center Department of Pharmacology Shenzhen University Health Science Center Shenzhen 518060 China
| | - Qianling Zhang
- College of Chemistry and Environmental Engineering Shenzhen University Shenzhen 518060 P. R. China
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering Shenzhen University Shenzhen 518060 P. R. China
| | - Desheng Lu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention International Cancer Center Department of Pharmacology Shenzhen University Health Science Center Shenzhen 518060 China
| |
Collapse
|
22
|
Tisi R, Spinelli M, Palmioli A, Airoldi C, Cazzaniga P, Besozzi D, Nobile MS, Mazzoleni E, Arnhold S, De Gioia L, Grandori R, Peri F, Vanoni M, Sacco E. The Multi-Level Mechanism of Action of a Pan-Ras Inhibitor Explains its Antiproliferative Activity on Cetuximab-Resistant Cancer Cells. Front Mol Biosci 2021; 8:625979. [PMID: 33681292 PMCID: PMC7925909 DOI: 10.3389/fmolb.2021.625979] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/11/2021] [Indexed: 12/04/2022] Open
Abstract
Ras oncoproteins play a crucial role in the onset, maintenance, and progression of the most common and deadly human cancers. Despite extensive research efforts, only a few mutant-specific Ras inhibitors have been reported. We show that cmp4–previously identified as a water-soluble Ras inhibitor– targets multiple steps in the activation and downstream signaling of different Ras mutants and isoforms. Binding of this pan-Ras inhibitor to an extended Switch II pocket on HRas and KRas proteins induces a conformational change that down-regulates intrinsic and GEF-mediated nucleotide dissociation and exchange and effector binding. A mathematical model of the Ras activation cycle predicts that the inhibitor severely reduces the proliferation of different Ras-driven cancer cells, effectively cooperating with Cetuximab to reduce proliferation even of Cetuximab-resistant cancer cell lines. Experimental data confirm the model prediction, indicating that the pan-Ras inhibitor is an appropriate candidate for medicinal chemistry efforts tailored at improving its currently unsatisfactory affinity.
Collapse
Affiliation(s)
- Renata Tisi
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Michela Spinelli
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy.,SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, Milan, Italy
| | - Alessandro Palmioli
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Cristina Airoldi
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy.,SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, Milan, Italy
| | - Paolo Cazzaniga
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, Milan, Italy.,Bicocca Bioinformatics, Biostatistics and Bioimaging Centre - B4, Milano, Italy
| | - Daniela Besozzi
- SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, Milan, Italy.,Bicocca Bioinformatics, Biostatistics and Bioimaging Centre - B4, Milano, Italy
| | - Marco S Nobile
- Bicocca Bioinformatics, Biostatistics and Bioimaging Centre - B4, Milano, Italy.,Department of Industrial Engineering and Innovation Sciences, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Elisa Mazzoleni
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Simone Arnhold
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Luca De Gioia
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy.,SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, Milan, Italy
| | - Rita Grandori
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Francesco Peri
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy.,SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, Milan, Italy
| | - Elena Sacco
- Department of Biotechnology and Biosciences, University of Milan-Bicocca, Milan, Italy.,SYSBIO-ISBE-IT-Candidate National Node of Italy for ISBE, Research Infrastructure for Systems Biology Europe, Milan, Italy
| |
Collapse
|
23
|
Wu K, Ho S, Wu C, Wang HD, Ma D, Leung C. Simultaneous blocking of the pan-RAF and S100B pathways as a synergistic therapeutic strategy against malignant melanoma. J Cell Mol Med 2021; 25:1972-1981. [PMID: 33377602 PMCID: PMC7882986 DOI: 10.1111/jcmm.15994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/14/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
Melanoma is a very aggressive form of skin cancer. Although BRAF inhibitors have been utilized for melanoma therapy, advanced melanoma patients still face a low five-year survival rate. Recent studies have shown that CRAF can compensate for BRAF depletion via regulating DNA synthesis to remain melanoma proliferation. Hence, targeting CRAF either alone or in combination with other protein pathways is a potential avenue for melanoma therapy. Based on our previously reported CRAF-selective inhibitor for renal cancer therapy, we have herein discovered an analogue (complex 1) from the reported CRAF library suppresses melanoma cell proliferation and melanoma tumour growth in murine models of melanoma via blocking the S100B and RAF pathways. Intriguingly, we discovered that inhibiting BRAF together with S100B exerts a novel synergistic effect to significantly restore p53 transcription activity and inhibit melanoma cell proliferation, whereas blocking BRAF together with CRAF only had an additive effect. We envision that blocking the pan-RAF and S100B/p53 pathways might be a novel synergistic strategy for melanoma therapy and that complex 1 is a potential inhibitor against melanoma via blocking the pan-RAF and S100B pathways.
Collapse
Affiliation(s)
- Ke‐Jia Wu
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacao SARChina
| | - Shih‐Hsin Ho
- State Key Laboratory of Urban Water Resource and EnvironmentSchool of EnvironmentHarbin Institute of TechnologyHarbinChina
| | - Chun Wu
- Department of ChemistryHong Kong Baptist UniversityKowloon TongHong Kong
| | - Hui‐Min D. Wang
- Graduate Institute of Biomedical Engineering National Chung Hsing UniversityTaichungTaiwan
- Graduate Institute of MedicineCollege of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
- Department of Medical Laboratory Science and BiotechnologyChina Medical UniversityTaichung CityTaiwan
| | - Dik‐Lung Ma
- Department of ChemistryHong Kong Baptist UniversityKowloon TongHong Kong
| | - Chung‐Hang Leung
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauMacao SARChina
| |
Collapse
|
24
|
Sun Q, Wang Y, Fu Q, Ouyang A, Liu S, Wang Z, Su Z, Song J, Zhang Q, Zhang P, Lu D. Sulfur-Coordinated Organoiridium(III) Complexes Exert Breast Anticancer Activity via Inhibition of Wnt/β-Catenin Signaling. Angew Chem Int Ed Engl 2021; 60:4841-4848. [PMID: 33244858 DOI: 10.1002/anie.202015009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Indexed: 12/12/2022]
Abstract
The sulfur-coordinated organoiridium(III) complexes pbtIrSS and ppyIrSS, which contain C,N and S,S (dithione) chelating ligands, were found to inhibit breast cancer tumorigenesis and metastasis by targeting Wnt/β-catenin signaling for the first time. Treatment with pbtIrSS and ppyIrSS induces the degradation of LRP6, thereby decreasing the protein levels of DVL2, β-catenin and activated β-catenin, resulting in downregulation of Wnt target genes CD44 and survivin. Additionally, pbtIrSS and ppyIrSS can suppress cell migration and invasion of breast cancer cells. Furthermore, both complexes show the ability to inhibit sphere formation and mediate the stemness properties of breast cancer cells. Importantly, pbtIrSS exerts potent anti-tumor and anti-metastasis effects in mouse xenograft models through the blockage of Wnt/β-catenin signaling. Taken together, our results indicate that pbtIrSS has great potential to be developed as a breast cancer therapeutic agent with a novel mechanism.
Collapse
Affiliation(s)
- Qi Sun
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yi Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory for Advanced Materials of MOE, School of Chemistry & Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Qiuxia Fu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Ai Ouyang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Shanshan Liu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Zhongyuan Wang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Zijie Su
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jiaxing Song
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Qianling Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Pingyu Zhang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Desheng Lu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| |
Collapse
|
25
|
Natural Products Attenuating Biosynthesis, Processing, and Activity of Ras Oncoproteins: State of the Art and Future Perspectives. Biomolecules 2020; 10:biom10111535. [PMID: 33182807 PMCID: PMC7698260 DOI: 10.3390/biom10111535] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023] Open
Abstract
RAS genes encode signaling proteins, which, in mammalian cells, act as molecular switches regulating critical cellular processes as proliferation, growth, differentiation, survival, motility, and metabolism in response to specific stimuli. Deregulation of Ras functions has a high impact on human health: gain-of-function point mutations in RAS genes are found in some developmental disorders and thirty percent of all human cancers, including the deadliest. For this reason, the pathogenic Ras variants represent important clinical targets against which to develop novel, effective, and possibly selective pharmacological inhibitors. Natural products represent a virtually unlimited resource of structurally different compounds from which one could draw on for this purpose, given the improvements in isolation and screening of active molecules from complex sources. After a summary of Ras proteins molecular and regulatory features and Ras-dependent pathways relevant for drug development, we point out the most promising inhibitory approaches, the known druggable sites of wild-type and oncogenic Ras mutants, and describe the known natural compounds capable of attenuating Ras signaling. Finally, we highlight critical issues and perspectives for the future selection of potential Ras inhibitors from natural sources.
Collapse
|
26
|
Isokotomolide A from Cinnamomum kotoense Induce Melanoma Autophagy and Apoptosis In Vivo and In Vitro. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3425147. [PMID: 33062137 PMCID: PMC7537700 DOI: 10.1155/2020/3425147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/24/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022]
Abstract
Melanoma is an aggressive cancer with high lethality. In order to find new anticancer agents, isokotomolide A (Iso A) and secokotomolide A (Sec A) isolated from Cinnamomum kotoense were identified to be potential bioactive agents against human melanoma but without strong antioxidative properties. Cell proliferation assay displayed Iso A and Sec A treated in the normal human skin cells showed high viabilities. It also verified that two of them possess strong antimelanoma effect in concentration-dependent manners, especially on B16F10, A2058, MeWo, and A375 cells. Wound healing assay presented their excellent antimigratory effects. Through 3-N,3-N,6-N,6-N-Tetramethylacridine-3,6-diamine (acridine orange, AO) staining and Western blot, the autophagy induced by treatment was confirmed, including autophagy-related proteins (Atgs). By using annexin V–FITC/PI double-stain, the apoptosis was confirmed, and both components also triggered the cell cycle arrest and DNA damage. We demonstrated the correlations between the mitogen-activated protein kinase (MAPK) pathway and antimelanoma, such as caspase cascade activations. To further evaluate in vivo experiments, the inhibition of tumor cell growth was verified through the histopathological staining in a xenograft model. In this study, it was confirmed that Iso A and Sec A can encourage melanoma cell death via early autophagy and late apoptosis processes.
Collapse
|
27
|
A Novel Oral Astaxanthin Nanoemulsion from Haematococcus pluvialis Induces Apoptosis in Lung Metastatic Melanoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2647670. [PMID: 32908627 PMCID: PMC7471791 DOI: 10.1155/2020/2647670] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/03/2020] [Indexed: 11/17/2022]
Abstract
Astaxanthin (AST) is a naturally occurring xanthophyll carotenoid having the potential to be used as an anticancer agent; however, the human body has a low bioavailability of AST due to its poor solubility in the water phase. Therefore, we applied D-α-tocopheryl polyethylene glycol succinate (TPGS) as an emulsifier and natural edible peanut oil to form a steady oil-in-water (O/W) nanoemulsion loaded with AST (denoted as TAP-nanoemulsion). TAP-nanoemulsions were stable without the droplet coalescence against thermal treatments (30-90°C), pH value changes (over a range of 2.0-8.0), and ionic strength adjustments (at NaCl concentrations of 100-500 mM) measured by dynamic light scattering (DLS). AST within TAP-nanoemulsion was released up to 80% in a simulated intestinal enzymatic fluid in vitro, and the overall recovery rate was fairly consistent in the Caco-2 cellular model. In order to further evaluate in vivo melanoma inhibitory experiments, we injected the fluorescent-stained B16F10 cells into female C57BL/6 mouse tail veins and treated TAP-nanoemulsion in an oral gavage. qRT-PCR and Western blot demonstrated that TAP-nanoemulsion triggered effectively the apoptosis pathway, including enhancements of cleaved caspase-3 and caspase-9, ataxia-telangiectasia mutated kinase (ATM), and p21WAF1/CIP1 (p21) and decreases of B-cell lymphoma 2 (Bcl-2); cyclins D, D1, and E; mitogen-activated protein kinase (MEK); extracellular signal-regulated kinases (ERK); nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB); and matrix metallopeptidase-1 and metallopeptidase-9 (MMP-1 and MMP-9) in both gene and protein expressions. In conclusion, this study suggests that TAP-nanoemulsion with the oral treatment has a positive chemotherapy effect in melanoma with lung metastases in vivo. As far as we know, this is the first time to demonstrate that an antioxidant in nanoparticle administration cures lung metastatic melanoma.
Collapse
|
28
|
Xu Z, Huang J, Kong D, Yang Y, Guo L, Jia X, Zhong G, Liu Z. Potent half-sandwich Ru(Ⅱ) N^N (aryl-BIAN) complexes: Lysosome-mediated apoptosis, in vitro and in vivo anticancer activities. Eur J Med Chem 2020; 207:112763. [PMID: 32882612 DOI: 10.1016/j.ejmech.2020.112763] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/09/2020] [Accepted: 08/15/2020] [Indexed: 12/22/2022]
Abstract
Herein a new series of organometallic half-sandwich Ru(Ⅱ) complexes bearing aryl-BIAN chelating ligands with various electron-withdrawing and electron-donating substituents have been developed as theranostic agents. All the complexes display much higher anti-proliferative potency than the clinical chemotherapeutic drug cisplatin towards seven cancer cell lines. The anti-proliferative efficacy of these complexes is correlated to their electron-withdrawing ability. Interestingly, complex Ru1 also potently suppresses cancer cell migration in vitro and effectively inhibit tumor growth in vivo in a CT26 colon cancer mouse xenograft model. Mechanisms of action studies display that Ru1 can favorably accumulate in lysosome and exerts anti-cancer potency by inducing a series of events related to lysosomal dysfunction in CT26 cells. Interestingly, inhibition of lysosomal enzymes leads to suppression of cytotoxicity and apoptosis induced by Ru1. Our results elucidate that complex Ru1 can elicit cytotoxicity through lysosome-mediated apoptosis in vitro and suppress tumor growth in vivo.
Collapse
Affiliation(s)
- Zhishan Xu
- College of Chemistry, Chemistry Engineering and Materials Science, Shandong Normal University, Jinan, 250014, China; Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
| | - Jie Huang
- Qingdao University of Science and Technology, Qingdao, 266061, China.
| | - Deliang Kong
- Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
| | - Yuliang Yang
- Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
| | - Lihua Guo
- Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China
| | - Xianglei Jia
- Henan Key Laboratory of Neural Regeneration, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Genshen Zhong
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhe Liu
- Institute of Anticancer Agents Development and Theranostic Application, The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine, Department of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, 273165, China.
| |
Collapse
|
29
|
Covalent and noncovalent constraints yield a figure eight-like conformation of a peptide inhibiting the menin-MLL interaction. Eur J Med Chem 2020; 207:112748. [PMID: 32882610 DOI: 10.1016/j.ejmech.2020.112748] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 08/04/2020] [Accepted: 08/08/2020] [Indexed: 11/21/2022]
Abstract
The interaction between menin and mixed lineage leukemia (MLL) was identified as an interesting target for treating some cancers including acute leukemia. On the basis of the known crystal structure of the MBM1-menin complex (MBM - menin binding motif), several cyclic peptides were designed. Elaboration of the effective cyclization strategy using a metathesis reaction allowed for a successfully large number of derivatives to be obtained. Subsequent optimization of the loop size, as well as N-terminal, central and C-terminal parts of the studied peptides resulted in structures exhibiting low nanomolar activities. A crystal structure of an inhibitor-menin complex revealed a compact conformation of the ligand molecule, which is stabilized not only by the introduction of a covalent linker but also three intramolecular hydrogen bonds. The inhibitor adopts a figure eight-like conformation, which perfectly fits the cleft of menin. We demonstrated that the development of compact, miniprotein-like structures is a highly effective approach for inhibition of protein-protein interactions.
Collapse
|
30
|
Elie BT, Hubbard K, Layek B, Yang WS, Prabha S, Ramos JW, Contel M. Auranofin-Based Analogues Are Effective Against Clear Cell Renal Carcinoma In Vivo and Display No Significant Systemic Toxicity. ACS Pharmacol Transl Sci 2020. [PMID: 32832867 DOI: 10.1021/acsptsci.9b00107/asset/images/large/pt9b00107_0002.jpeg] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Effective pharmacological treatments for patients with advanced clear cell renal carcinoma (ccRCC) are limited. Bimetallic titanium-gold containing compounds exhibit significant cytotoxicity against ccRCC in vitro and in vivo and inhibit invasion and angiogenisis in vitro and markers driving these phenomena. However, in vivo preclinical evaluations of such compounds have not examined their pharmacokinetics, pathology, and hematology. Here we use NOD.CB17-Prkdc SCID/J mice bearing xenograft ccRCC Caki-1 tumors to evaluate the in vivo efficacies of two titanium-gold compounds Titanocref and Titanofin (based on auranofin analogue scaffolds) accompanied by pharmacokinetic and pathology studies. A therapeutic trial was performed over 21 days at 5 mg/kg/72h of Titanocref and 10 mg/kg/72h of Titanofin tracking changes in tumor size. We observed a significant reduction of 51% and 60%, respectively (p < 0.01) in tumor size in the Titanocref- and Titanofin-treated mice compared to the starting size, while the vehicle-treated mice exhibited a tumor size increase of 138% (p < 0.01). Importantly, no signs of pathological complication as a result of treatment were found. In addition, Titanocref and Titanofin treatment reduced angiogenesis by 38% and 54%, respectively. Microarray and qRT-PCR analysis of ccRCC Caki-1 cells treated with Titanocref revealed that the compound alters apoptosis, JNK MAP kinase, and ROS pathways within 3 h of treatment. We further show activation of apoptosis by Titanocref and Titanofin in vivo by caspase 3 assay. Titanocref is active against additional kidney cancer cells. Titanocref and Titanofin are therefore promising candidates for further evaluation toward clinical application in the treatment of ccRCC.
Collapse
Affiliation(s)
- Benelita T Elie
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States
| | - Karen Hubbard
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Department of Biology, City College of New York, The City University of New York, New York, New York 10031, United States
| | - Buddhadev Layek
- University of Minnesota, College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Won Seok Yang
- University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| | - Swayam Prabha
- University of Minnesota, College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Joe W Ramos
- University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| | - Maria Contel
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| |
Collapse
|
31
|
Ji S, Yang X, Chen X, Li A, Yan D, Xu H, Fei H. Structure-tuned membrane active Ir-complexed oligoarginine overcomes cancer cell drug resistance and triggers immune responses in mice. Chem Sci 2020; 11:9126-9133. [PMID: 34094193 PMCID: PMC8161536 DOI: 10.1039/d0sc03975f] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 11/21/2022] Open
Abstract
The development of chemotherapy, an important cancer treatment modality, is hindered by the frequently found drug-resistance phenomenon. Meanwhile, researchers have been enthused lately by the synergistic use of chemotherapy with emerging immunotherapeutic treatments. In an effort to address both of the two unmet needs, reported herein is a study on a series of membrane active iridium(iii) complexed oligoarginine peptides with a new cell death mechanism capable of overcoming drug resistance as well as stimulating immunological responses. A systematic structure-activity relationship study elucidated the interdependent effects of three structural factors, i.e., hydrophobicity, topology and cationicity, on the regulation of the cytotoxicity of the Ir(iii)-oligoarginine peptides. With the most prominent toxicities, Ir-complexed octaarginines (R8) were found to display a progressive oncotic cell death featuring cell membrane-penetration and eruptive cytoplasmic content release. Consequently, this membrane-centric death mechanism showed promising potential in overcoming multiple chemical drug-resistance of cancer cells. More interestingly, the eruptive mode of cell death proved to be immunogenic by stimulating the dendritic cell maturation and inflammatory factor accumulation in mice tumours. Taking these mechanisms together, this work demonstrates that membrane active compounds may become the next generation chemotherapeutics because of their combined advantages.
Collapse
Affiliation(s)
- Shuangshuang Ji
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China Hefei 230026 PR China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| | - Xiuzhu Yang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| | - Xiaolong Chen
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China Hefei 230026 PR China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| | - Ang Li
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China Hefei 230026 PR China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| | - Doudou Yan
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing 100005 PR China
| | - Haiyan Xu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing 100005 PR China
| | - Hao Fei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China Hefei 230026 PR China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences Suzhou 215123 PR China
| |
Collapse
|
32
|
Purified Astaxanthin from Haematococcus pluvialis Promotes Tissue Regeneration by Reducing Oxidative Stress and the Secretion of Collagen In Vitro and In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4946902. [PMID: 32832000 PMCID: PMC7424503 DOI: 10.1155/2020/4946902] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 06/12/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Intracellular reactive apoptosis and reactive oxygen species (ROS) play a crucial role in ultraviolet- (UV-) induced inflammation and aging reaction in human dermal tissues. This study determines the mechanism by which Haematococcus pluvialis extracts (HPE) and purified astaxanthin (HPA) to promote skin regeneration in the injured tissue in vitro and in vivo. The results show that HPE and HPA decrease the DNA damage and promote the secretion of collagen from the human normal fibroblast cell line (Hs68) in a dose-dependent manner. UV irradiation and HPA reduce oxidative stress damage due to phorbol-12-myristate-13-acetate (PMA). When skin cells are injured by free radicals, cells undergo a programmed cellular death. Cellular apoptotic death is determined using annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) double staining to verify that there is no cell membrane asymmetry and that the nuclear membrane is broken. Inflammatory symptoms and apoptotic injuries to experimental rats in a group that is treated with HPA treated are decreased in a dose-dependent manner after UVB exposure (300 mJ/cm2) for 15 min in vivo, compared to the vehicle control group. These positive results show that HPA repairs UVB-triggered skin tissue injury and aging by conducting electrons out of cells to maintain a low level of oxidative stress so that collagen is synthesized in vitro and in vivo.
Collapse
|
33
|
Graf M, Siegmund D, Gothe Y, Metzler‐Nolte N, Sünkel K. Metal and Substituent Influence on the Cytostatic Activity of Cationic Bis‐cyclometallated Iridium and Rhodium Complexes with Substituted 1,10‐Phenanthrolines as Ancillary Ligands. Z Anorg Allg Chem 2020. [DOI: 10.1002/zaac.201900317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Marion Graf
- Department of Chemistry Ludwig‐Maximilians University Munich Butenandtstr. 9 81377 Munich Germany
| | - Daniel Siegmund
- Inorganic Chemistry I Bioinorganic Chemistry Faculty of Chemistry and Biochemistry Ruhr University Bochum 44801 Bochum Germany
| | - Yvonne Gothe
- Inorganic Chemistry I Bioinorganic Chemistry Faculty of Chemistry and Biochemistry Ruhr University Bochum 44801 Bochum Germany
| | - Nils Metzler‐Nolte
- Inorganic Chemistry I Bioinorganic Chemistry Faculty of Chemistry and Biochemistry Ruhr University Bochum 44801 Bochum Germany
| | - Karlheinz Sünkel
- Department of Chemistry Ludwig‐Maximilians University Munich Butenandtstr. 9 81377 Munich Germany
| |
Collapse
|
34
|
Wang J, Wang Y, Zhang D, Chen C. Intrinsic Oxidase-like Nanoenzyme Co 4S 3/Co(OH) 2 Hybrid Nanotubes with Broad-Spectrum Antibacterial Activity. ACS APPLIED MATERIALS & INTERFACES 2020; 12:29614-29624. [PMID: 32501670 DOI: 10.1021/acsami.0c05141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Improving the antibacterial activity of nanomaterials and avoiding the use of H2O2 are vital for biosecurity and public health. In this work, novel Co4S3/Co(OH)2 hybrid nanotubes (HNTs) for the first time were successfully synthesized through the control of Na2S treatment of Co(CO3)0.35Cl0.20(OH)1.10 precursor. On the basis of Kirkendall effect, acicular precursor was vulcanized to form Co4S3/Co(OH)2 HNTs that possess great properties including favorable storage ability and ideal stability. By tailoring the composition and structure, Co4S3/Co(OH)2 HNTs were found to have profound oxidase-like catalytic activities. When pH = 3 precursor was treated with 900 mg of Na2S, Co4S3/Co(OH)2 HNTs exhibit superior performance. Owing to the outstanding oxidase-like activity, Co4S3/Co(OH)2 HNTs can eliminate Escherichia coli, Pseudomonas aeruginosa, Staphylococcus sciuri, and Bacillus without the help of H2O2. It turned out that the sterilization ability came from the superoxide anion radical generated by Co4S3/Co(OH)2 HNTs. With Co4S3/Co(OH)2 HNTs, the intracellular reactive oxygen species level can be enhanced and the toxicity of H2O2 can be absolutely avoided. Overall, the synthesis of antibacterial nanomaterials is unparalleled and the results of this work would facilitate the utilization in medical science, new energy, and environmental catalysis.
Collapse
Affiliation(s)
- Jin Wang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China
- University of Chinese Academy of Sciences, No. 19 (Jia) Yuquan Road, Beijing 100039, China
- Open Studio for Marine Corrosion and Protection, Pilot National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China
| | - Yi Wang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China
- Open Studio for Marine Corrosion and Protection, Pilot National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China
| | - Dun Zhang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China
- Open Studio for Marine Corrosion and Protection, Pilot National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China
| | - Chao Chen
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China
- University of Chinese Academy of Sciences, No. 19 (Jia) Yuquan Road, Beijing 100039, China
- Open Studio for Marine Corrosion and Protection, Pilot National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China
| |
Collapse
|
35
|
Xue W, Fu T, Zheng G, Tu G, Zhang Y, Yang F, Tao L, Yao L, Zhu F. Recent Advances and Challenges of the Drugs Acting on Monoamine Transporters. Curr Med Chem 2020; 27:3830-3876. [DOI: 10.2174/0929867325666181009123218] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/30/2018] [Accepted: 10/03/2018] [Indexed: 01/06/2023]
Abstract
Background:
The human Monoamine Transporters (hMATs), primarily including hSERT,
hNET and hDAT, are important targets for the treatment of depression and other behavioral disorders
with more than the availability of 30 approved drugs.
Objective:
This paper is to review the recent progress in the binding mode and inhibitory mechanism of
hMATs inhibitors with the central or allosteric binding sites, for the benefit of future hMATs inhibitor
design and discovery. The Structure-Activity Relationship (SAR) and the selectivity for hit/lead compounds
to hMATs that are evaluated by in vitro and in vivo experiments will be highlighted.
Methods:
PubMed and Web of Science databases were searched for protein-ligand interaction, novel
inhibitors design and synthesis studies related to hMATs.
Results:
Literature data indicate that since the first crystal structure determinations of the homologous
bacterial Leucine Transporter (LeuT) complexed with clomipramine, a sizable database of over 100 experimental
structures or computational models has been accumulated that now defines a substantial degree
of structural variability hMATs-ligands recognition. In the meanwhile, a number of novel hMATs
inhibitors have been discovered by medicinal chemistry with significant help from computational models.
Conclusion:
The reported new compounds act on hMATs as well as the structures of the transporters
complexed with diverse ligands by either experiment or computational modeling have shed light on the
poly-pharmacology, multimodal and allosteric regulation of the drugs to transporters. All of the studies
will greatly promote the Structure-Based Drug Design (SBDD) of structurally novel scaffolds with high
activity and selectivity for hMATs.
Collapse
Affiliation(s)
- Weiwei Xue
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Tingting Fu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Guoxun Zheng
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Gao Tu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Yang Zhang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Fengyuan Yang
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-cancer Chinese Medicine of Zhejiang Province, School of Medicine, Hangzhou Normal University, Hangzhou 310036, China
| | - Lixia Yao
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, United States
| | - Feng Zhu
- Innovative Drug Research and Bioinformatics Group, School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, China
| |
Collapse
|
36
|
Wang J, Wang Y, Zhang D, Xu C, Xing R. Dual response mimetic enzyme of novel Co 4S 3/Co 3O 4 composite nanotube for antibacterial application. JOURNAL OF HAZARDOUS MATERIALS 2020; 392:122278. [PMID: 32105950 DOI: 10.1016/j.jhazmat.2020.122278] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/24/2020] [Accepted: 02/10/2020] [Indexed: 06/10/2023]
Abstract
Novel Co4S3/Co3O4 composite nanotubes were first time successfully synthesized through the control of Na2S treatment of Co(CO3)0.35Cl0.20(OH)1.10 precursor and testified to having both oxidase-like and peroxidase-like catalytic activities. Through nanoscale Kirkendall effect, acicular precursor was vulcanized to form Co4S3/Co3O4 nanotubes (NTs). Co4S3/Co3O4 NTs exhibit great properties such as smaller Km and higher Vmax than natural horseradish peroxidase, favorable selectivity and ideal stability. Co4S3/Co3O4 NTs with promising biocompatibility can realize spectral sterilization towards Escherichia coli and Staphylococcus sciuri in the presence of low level H2O2. It turned to be that superoxide anion radical catalyzed by Co4S3/Co3O4 NTs not only oxidizes 3, 3', 5, 5'-tetramethylbenzidine in the catalytic process but also eliminates gram-bacteria during sterilization. The successful synthesis of biofunctional Co4S3/Co3O4 NTs can serve as significant applications of new energy and environmental catalysis.
Collapse
Affiliation(s)
- Jin Wang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; University of Chinese Academy of Sciences, No. 19 (Jia) Yuquan Road, Beijing 100039, China; Open Studio for Marine Corrosion and Protection, Pilot National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China
| | - Yi Wang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Open Studio for Marine Corrosion and Protection, Pilot National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China.
| | - Dun Zhang
- CAS Key Laboratory of Marine Environmental Corrosion and Bio-fouling, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; Open Studio for Marine Corrosion and Protection, Pilot National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China; Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China.
| | - Chaojie Xu
- University of Chinese Academy of Sciences, No. 19 (Jia) Yuquan Road, Beijing 100039, China; CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road,Qingdao 266071, China
| | - Ronge Xing
- Center for Ocean Mega-Science, Chinese Academy of Sciences, No. 7 Nanhai Road, Qingdao 266071, China; CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, No. 7 Nanhai Road,Qingdao 266071, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, No. 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
37
|
Ho PY, Ho CL, Wong WY. Recent advances of iridium(III) metallophosphors for health-related applications. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213267] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
38
|
Elie BT, Hubbard K, Layek B, Yang WS, Prabha S, Ramos JW, Contel M. Auranofin-Based Analogues Are Effective Against Clear Cell Renal Carcinoma In Vivo and Display No Significant Systemic Toxicity. ACS Pharmacol Transl Sci 2020; 3:644-654. [PMID: 32832867 DOI: 10.1021/acsptsci.9b00107] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Indexed: 01/03/2023]
Abstract
Effective pharmacological treatments for patients with advanced clear cell renal carcinoma (ccRCC) are limited. Bimetallic titanium-gold containing compounds exhibit significant cytotoxicity against ccRCC in vitro and in vivo and inhibit invasion and angiogenisis in vitro and markers driving these phenomena. However, in vivo preclinical evaluations of such compounds have not examined their pharmacokinetics, pathology, and hematology. Here we use NOD.CB17-Prkdc SCID/J mice bearing xenograft ccRCC Caki-1 tumors to evaluate the in vivo efficacies of two titanium-gold compounds Titanocref and Titanofin (based on auranofin analogue scaffolds) accompanied by pharmacokinetic and pathology studies. A therapeutic trial was performed over 21 days at 5 mg/kg/72h of Titanocref and 10 mg/kg/72h of Titanofin tracking changes in tumor size. We observed a significant reduction of 51% and 60%, respectively (p < 0.01) in tumor size in the Titanocref- and Titanofin-treated mice compared to the starting size, while the vehicle-treated mice exhibited a tumor size increase of 138% (p < 0.01). Importantly, no signs of pathological complication as a result of treatment were found. In addition, Titanocref and Titanofin treatment reduced angiogenesis by 38% and 54%, respectively. Microarray and qRT-PCR analysis of ccRCC Caki-1 cells treated with Titanocref revealed that the compound alters apoptosis, JNK MAP kinase, and ROS pathways within 3 h of treatment. We further show activation of apoptosis by Titanocref and Titanofin in vivo by caspase 3 assay. Titanocref is active against additional kidney cancer cells. Titanocref and Titanofin are therefore promising candidates for further evaluation toward clinical application in the treatment of ccRCC.
Collapse
Affiliation(s)
- Benelita T Elie
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States
| | - Karen Hubbard
- Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Department of Biology, City College of New York, The City University of New York, New York, New York 10031, United States
| | - Buddhadev Layek
- University of Minnesota, College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Won Seok Yang
- University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| | - Swayam Prabha
- University of Minnesota, College of Pharmacy, Minneapolis, Minnesota 55455, United States
| | - Joe W Ramos
- University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| | - Maria Contel
- Department of Chemistry, Brooklyn College, The City University of New York, Brooklyn, New York 11210, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,Biology, Chemistry and Biochemistry PhD Programs, The Graduate Center, The City University of New York, New York, New York 10016, United States.,University of Hawaii Cancer Center, Honolulu, Hawaii 96813, United States
| |
Collapse
|
39
|
Mitochondria-targeted phosphorescent cyclometalated iridium(III) complexes: synthesis, characterization, and anticancer properties. J Biol Inorg Chem 2020; 25:597-607. [PMID: 32232583 DOI: 10.1007/s00775-020-01783-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/25/2020] [Indexed: 01/09/2023]
Abstract
Cyclometalated iridium(III) complexes represent a promising approach to developing new anticancer metallodrugs. In this work, three phosphorescent cyclometalated iridium(III) complexes Ir1-Ir3 have been explored as mitochondria-targeted anticancer agents. All three complexes display higher antiproliferative activity than cisplatin against the cancer cells screened, and with the IC50 values ranging from 0.23 to 5.6 μM. Colocalization studies showed that these complexes are mainly localized in the mitochondria. Mechanism studies show that these complexes exert their anticancer efficacy through initiating a series of events related to mitochondrial dysfunction, including depolarization of mitochondrial membrane potential (MMP), elevation of intracellular reactive oxygen species (ROS) levels, and induction of apoptosis. Mitochondria-targted cyclometalated iridium complexes induce apoptosis through depolarized mitochondria, elevation of intracellular ROS and activated caspase.
Collapse
|
40
|
Cheng SS, Yang GJ, Wang W, Leung CH, Ma DL. The design and development of covalent protein-protein interaction inhibitors for cancer treatment. J Hematol Oncol 2020; 13:26. [PMID: 32228680 PMCID: PMC7106679 DOI: 10.1186/s13045-020-00850-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
Protein-protein interactions (PPIs) are central to a variety of biological processes, and their dysfunction is implicated in the pathogenesis of a range of human diseases, including cancer. Hence, the inhibition of PPIs has attracted significant attention in drug discovery. Covalent inhibitors have been reported to achieve high efficiency through forming covalent bonds with cysteine or other nucleophilic residues in the target protein. Evidence suggests that there is a reduced risk for the development of drug resistance against covalent drugs, which is a major challenge in areas such as oncology and infectious diseases. Recent improvements in structural biology and chemical reactivity have enabled the design and development of potent and selective covalent PPI inhibitors. In this review, we will highlight the design and development of therapeutic agents targeting PPIs for cancer therapy.
Collapse
Affiliation(s)
- Sha-Sha Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, SAR, China
| | - Guan-Jun Yang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, SAR, China
| | - Wanhe Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon, 999077, Hong Kong, China.,Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Chung-Hang Leung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, SAR, China.
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon, 999077, Hong Kong, China.
| |
Collapse
|
41
|
Pan F, Wang Q, Li S, Huang R, Wang X, Liao X, Mo H, Zhang L, Zhou X. Prognostic value of key genes of the JAK-STAT signaling pathway in patients with cutaneous melanoma. Oncol Lett 2020; 19:1928-1946. [PMID: 32194688 PMCID: PMC7039088 DOI: 10.3892/ol.2020.11287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 11/22/2019] [Indexed: 01/05/2023] Open
Abstract
The Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway is involved in cell immunity, division and death, as well as in tumor formation. The expression of key genes in the JAK-STAT signaling pathway in different types of cancer serves different roles. However, few reports are available on the prognostic value of the genes of the JAK-STAT signaling pathway in skin cutaneous melanoma (SKCM). The potential prognostic value of gene expression in the JAK-STAT signaling pathway in patients with SKCM was analyzed in the present study using data obtained from The Cancer Genome Atlas. To predict the potential functions and mechanisms of these genes in SKCM, gene set enrichment analysis (GSEA) and bioinformatics analysis were performed. A nomogram model including gene expression level and high risk factors was used to predict the risk level of prognostic. High expression levels of STAT1, STAT3, STAT4 and STAT5B, and low expression levels of STAT6 were associated with favorable prognosis [adjusted P<0.001; hazard ratio (HR), 0.595; 95% confidence interval (CI), 0.455–0.778; adjusted P=0.018; HR, 0.725; 95% CI, 0.555–0.947; adjusted P<0.001; HR, 0.590; 95% CI, 0.450–0.773; adjusted P=0.007; HR, 0.690; 95% CI, 0.526–0.940; and adjusted P=0.026; HR, 0.737, 95% CI, 0.563–0.964, respectively]. GSEA results demonstrated that these genes were involved in cell differentiation, invasion, adhesion, migration, cycle, colony formation and mitogen-activated protein kinase signaling. The combination of genes with favorable prognosis had a better effect on the overall survival (univariate survival analysis, P<0.05). The results of the present study suggest that STAT1, STAT3, STAT4, STAT5B and STAT6 gene expression may be used as a potential prognostic biomarker of SKCM, and the combined outcomes may exhibit a stronger interaction and higher survival time for SKCM.
Collapse
Affiliation(s)
- Fuqiang Pan
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Qiaoqi Wang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Sizhu Li
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Rui Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Haiyan Mo
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Liming Zhang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| | - Xiang Zhou
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530000, P.R. China
| |
Collapse
|
42
|
Wu KJ, Ho SH, Dong JY, Fu L, Wang SP, Liu H, Wu C, Leung CH, Wang HMD, Ma DL. Aliphatic Group-Tethered Iridium Complex as a Theranostic Agent against Malignant Melanoma Metastasis. ACS APPLIED BIO MATERIALS 2020; 3:2017-2027. [DOI: 10.1021/acsabm.9b01156] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ke-Jia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macao SAR, China
| | - Shih-Hsin Ho
- State Key Laboratory of Urban Water Resource and Environment, School of Environment, Harbin Institute of Technology, Harbin 150090, China
| | - Jia-Yi Dong
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa 999078, Macao SAR, China
| | - Ling Fu
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Shuang-Peng Wang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Taipa 999078, Macao SAR, China
| | - Hao Liu
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong, China
| | - Chun Wu
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong, China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa 999078, Macao SAR, China
| | - Hui-Min David Wang
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung 402, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong 999077, Hong Kong, China
| |
Collapse
|
43
|
Kacar S, Unver H, Sahinturk V. A mononuclear copper(II) complex containing benzimidazole and pyridyl ligands: Synthesis, characterization, and antiproliferative activity against human cancer cells. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2019.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
44
|
Li J, Lu YR, Lin IF, Kang W, Chen HB, Lu HF, Wang HMD. Reversing UVB-induced photoaging with Hibiscus sabdariffa calyx aqueous extract. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:672-681. [PMID: 31583701 DOI: 10.1002/jsfa.10063] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/14/2019] [Accepted: 09/22/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Hibiscus sabdariffa is commonly used in daily life and its extract is applied widely in food and cosmetics. However, it has not been evaluated for its anti-aging effects. RESULTS Hibiscus sabdariffa calyx aqueous extract (HSCAE) has shown potential collagenase activity suppression effects, together with tyrosinase activity inhibition, and anti-oxidation as a free radical scavenger. The current investigation demonstrated that HSCAE was not cytotoxic in skin fibroblasts, and it significantly decreased ultraviolet B (UVB)-induced reactive oxygen species (ROS) on a flow cytometry assay. Moreover, HSCAE reduced matrix metalloproteinase (MMP) expression, increased tissue inhibition of metalloproteinase (TIMP)-1 level, and enhanced collagen content by inhibiting collagenase activity. It also blocked mRNA and protein expressions of melanin production pathway key factors, including the microphthalmia-associated transcription factor (MITF), tyrosinase, tyrosinase-related protein-1 (TRP-1), and dopachrome tautomerase-2 (TRP-2). CONCLUSION These results demonstrated, for the first time, the potential of HSCAE as a natural antioxidant with the ability to maintain collagen production and to decrease melanin syntheses under UVB radiation, for anti-aging effects. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jian Li
- College of Food and Biological Engineering, Jimei University, Xiamen, China
| | - Yi-Ru Lu
- Department of Bachelor Program of Biotechnology, National Chung Hsing University, Taichung City, Taiwan
| | - I-Fan Lin
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Wenyi Kang
- Joint International Research Laboratory of Food & Medicine Resource Function, Henan Province, Henan University, Kaifeng, China
| | - Hong-Bin Chen
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, China
| | - Hsu-Feng Lu
- Department of Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Hui-Min David Wang
- College of Food and Biological Engineering, Jimei University, Xiamen, China
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, China
- Graduate Institute of Biomedical Engineering, National Chung Hsing University, Taichung City, Taiwan
- College of Medicine, Kaohsiung Medical University, Kaohsiung City, Taiwan
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City, Taiwan
| |
Collapse
|
45
|
Orabi A, Abou El-Nour K, Youssef M, Salem H. Novel and highly effective composites of silver and zinc oxide nanoparticles with some transition metal complexes against different microorganisms. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2018.06.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
46
|
Xu Z, Yang Y, Jia X, Guo L, Ge X, Zhong G, Chen S, Liu Z. Novel cyclometalated iridium(iii) phosphine-imine (P^N) complexes: highly efficient anticancer and anti-lung metastasis agents in vivo. Inorg Chem Front 2020. [DOI: 10.1039/c9qi01492f] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Iridium(iii)-based complexes with phosphine-imine (P^N) ligands are synthesized and authenticated. The combined treatment with Ir(iii) and BIX01294 potently inhibited tumour growth and lung metastasis in vitro and in vivo.
Collapse
Affiliation(s)
- Zhishan Xu
- College of Chemistry
- Chemistry Engineering and Materials Science
- Shandong Normal University
- Jinan
- China
| | - Yuliang Yang
- Institute of Anticancer Agents Development and Theranostic Application
- The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine
- Department of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu
| | - Xianglei Jia
- Henan Key Laboratory of Neural Regeneration
- The First Affiliated Hospital of Xinxiang Medical University
- Weihui 453100
- China
| | - Lihua Guo
- Institute of Anticancer Agents Development and Theranostic Application
- The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine
- Department of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu
| | - Xingxing Ge
- Institute of Anticancer Agents Development and Theranostic Application
- The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine
- Department of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu
| | - Genshen Zhong
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine
- School of Laboratory Medicine
- Xinxiang Medical University
- Xinxiang
- China
| | - Shujiao Chen
- Institute of Anticancer Agents Development and Theranostic Application
- The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine
- Department of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu
| | - Zhe Liu
- Institute of Anticancer Agents Development and Theranostic Application
- The Key Laboratory of Life-Organic Analysis and Key Laboratory of Pharmaceutical Intermediates and Analysis of Natural Medicine
- Department of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu
| |
Collapse
|
47
|
Synthesis, characterization, apoptosis, ROS, autophagy and western blotting studies of cyclometalated iridium(III) complexes. INORG CHEM COMMUN 2020. [DOI: 10.1016/j.inoche.2019.107594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
48
|
Li Y, Wang KN, He L, Ji LN, Mao ZW. Synthesis, photophysical and anticancer properties of mitochondria-targeted phosphorescent cyclometalated iridium(III) N-heterocyclic carbene complexes. J Inorg Biochem 2019; 205:110976. [PMID: 31926377 DOI: 10.1016/j.jinorgbio.2019.110976] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/12/2019] [Accepted: 12/24/2019] [Indexed: 11/27/2022]
Abstract
Metal N-Heterocyclic carbene (NHC) complexes are expected to be new opportunities for the development of anticancer metallodrugs. In this work, two near-infrared (NIR) emitting iridium(III)-NHC complexes Ir1 and Ir2 have been explored as mitochondria-targeted anticancer and photodynamic agents. These complexes are more cytotoxic than cisplatin against the cancer cells screened, and display higher cytotoxicity in the presence of 450 nm and 630 nm LED light. Colocalization and quantitative studies indicated that these complexes could specially localize to mitochondria. Mechanism studies show that these complexes increase intracellular reactive oxygen species (ROS) level, reduce mitochondrial membrane potential (MMP) and induce some degree of early apoptosis. Further studies found that Ir1could induce mitophagy at dark and necrocytosis under the irradiation of 630 nm LED light. The in vitro and in vivo photoxicity studies revealed that Ir1 is a promising photodynamic therapy (PDT) agent and could significantly inhibit tumor growth.
Collapse
Affiliation(s)
- Yi Li
- Key Laboratory of Theoretical Organic Chemistry and Function Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China; MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Kang-Nan Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Liang He
- College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Liang-Nian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-sen University, Guangzhou, 510275, PR China.
| |
Collapse
|
49
|
Ahmad N, Ahmad R, Alam MA, Ahmad FJ, Rub RA. Quantification and Evaluation of Glycyrrhizic Acid-loaded Surface Decorated Nanoparticles by UHPLC-MS/MS and used in the Treatment of Cerebral Ischemia. CURR PHARM ANAL 2019. [DOI: 10.2174/1573412914666180530073613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Glycyrrhizic Acid (GRA), a potent antioxidant triterpene saponin glycoside
and neuroprotective properties exhibits an important role in the treatment of neurological disorders i.e.
cerebral ischemia. GRA is water soluble, therefore it’s have low bioavailability in the brain.
Objective:
To enhance brain bioavailability for intranasally administered Glycyrrhizic Acidencapsulated-
chitosan-coated-PCL-Nanoparticles (CS-GRA-PCL-NPs).
Methods:
Chitosan-coated-PCL-Nanoparticles (CS-PCL-NPs) were developed through double emulsification-
solvent evaporation technique and further characterized for particle size, zeta potential, size
distribution, encapsulation efficiency as well as in vitro drug release. UPLC triple quadrupole Qtrap
MS/MS method was developed to evaluate brain-drug uptake for optimized CS-GRA-PCL-NPs and to
determine its pharmacokinetic in rat’s brain as well as plasma.
Results:
Mean particles size (231.47±7.82), polydispersity index (PDI) i.e. (0.216±0.030) and entrapment
efficiency (65.69±5.68) was determined for developed NPs. UPLC triple quadrupole Qtrap MS/MS
method study showed a significantly high mucoadhesive potential of CS-GRA-PCL-NPs and least for
conventional and homogenized nanoformulation; elution time for GRA and internal standard (IS) Hydrocortisone
as 0.37 and 1.94 min at m/z 821.49/113.41 and 363.45/121.40 were observed, respectively. Furthermore,
intra and inter-assay (%CV) of 0.49-5.48, %accuracy (90.00-99.09%) as well as a linear dynamic
range (10.00 ng/mL -2000.0 ng/mL), was observed. Pharmacokinetic studies in Wistar rat brain
exhibited a high AUC0-24 alongwith an amplified Cmax (p** < 0.01) as compared to i.v. treated group.
Conclusion:
Intranasal administration of developed CS-coated-GRA-loaded-PCL-NPs enhanced the
drug bioavailability in rat brain along with successfully UPLC-MS/MS method and thus preparation of
GRA-NPs may help treat cerebral ischemia effectively. The toxicity studies performed at the end
revealed safe nature of optimized nanoformulation.
Collapse
Affiliation(s)
- Niyaz Ahmad
- Department of Pharmaceutics, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Rizwan Ahmad
- Department of Natural Products and Alternative Medicine, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Md Aftab Alam
- Department of Pharmaceutics, School of Medical and Allied Sciences, Galgotias University, Gautam Budh Nagar, Greater Noida-201310, India
| | - Farhan Jalees Ahmad
- Nanomedicine Lab, Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| | - Rehan Abdur Rub
- Nanomedicine Lab, Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| |
Collapse
|
50
|
Varghese NM, Senthil V, Jose S, Thomas C, Harindran J. Development and Validation of a Specific RP-HPLC Method for Simultaneous Estimation of Anti-retroviral Drugs: Application to Nanoparticulate Formulation System. CURR PHARM ANAL 2019. [DOI: 10.2174/1573412914666180621110432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Objective:
A simple, accurate and economical reverse phase- high performance liquid chromatographic
method has been developed for the simultaneous quantitative estimation of two anti-retro
viral drugs, Etravirine and Elvitegravir in nanoparticulate formulations for the first time. This method is
a novel analytical technique for the detection of the both the drugs concurrently, as there is no method
available for their simultaneous quantification, to the best of our knowledge.
Methods:
Optimization and validation of the chromatographic conditions were completed according to
the standard ICH guidelines. The separation was done on a C18 column (250 mm x 4.6mm, 5μm) using
methanol and phosphate buffer of pH (5.6) as the mobile phase in the ratio 78:22 v/v at a flow rate of
1ml/min for a short run time of 13 min. The detection wavelength was 285nm and the column temperature
was maintained at 32oC.
Results:
The developed method was linear over 10 to 160 μg/ml with a regression coefficient of 0.999
for each. The LOD values were 4.83 and 9.25 µg/ml while LOQ values were 14.63 and 28.01 µg/ml for
etravirine and elvitegravir respectively. The recovery values obtained by etravirine and elvitegravir
were between 97.6% and 100.8%.
Conclusion:
The method was specific, precise, fast and accurate with good inter and intra day precision.
The method was also effectively employed for the characterization and simultaneous quantification
of both drugs in nanoparticulate formulation.
Collapse
Affiliation(s)
- Nila Mary Varghese
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ootacamund, Mysuru, India
| | - Venkatachalam Senthil
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ootacamund, Mysuru, India
| | - Sajan Jose
- Department of Pharmaceutical Sciences, Centre for Professional and Advanced Studies Cheruvandoor, Ettumanoor, Kottayam, Kerala, India
| | - Cinu Thomas
- Department of Pharmaceutical Sciences, Centre for professional and advanced studies Cheruvandoor, Ettumanoor, Kottayam, Kerala, India
| | - Jyoti Harindran
- Department of Pharmaceutical Sciences, Centre for professional and advanced studies Cheruvandoor, Ettumanoor, Kottayam, Kerala, India
| |
Collapse
|