1
|
He YH, Xiang H, Long ZJ, Jin ZX, Hu JF, Mao YC, Xiong J. Structurally diverse sesquiterpenes with anti-leukemia activity from the rare medicinal plant Liriodendron chinense. Bioorg Chem 2025; 158:108341. [PMID: 40073594 DOI: 10.1016/j.bioorg.2025.108341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/25/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
A further phytochemical investigation on the branches and leaves of Liriodendron chinense, a rare medicinal and ornamental plant endemic to China, yielded thirty-four sesquiterpenes with diverse skeletons. The isolated compounds comprise 12 new naturally occurring sesquiterpenoids (1-12) and 22 known analogues (13-34). The new structures were elucidated by spectroscopic data analyses, and their absolute configurations (except for 10) were determined by electronic circular dichroism spectra, quantum-chemical calculations, and X-ray crystallography. Among them, liriochinolide A (1) is a rare chlorine-substituted germacrane-type sesquiterpenoid from the terrestrial plants. Bioassay evaluations revealed that most of the isolates exhibited remarkable cytotoxicity against three human leukemia cell lines (MV-4-11, HL-60 and Raji), with IC50 values ranging from 2.27 to 9.69 μM. Further exploration of anti-leukemia mechanism demonstrated that the bioactive compounds 5, 8, and 18 significantly induced the cell apoptosis, which was associated with down-regulation of the anti-apoptotic proteins BCL-2 and MCL-1.
Collapse
Affiliation(s)
- Yu-Hang He
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Hong Xiang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Zi-Jie Long
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Ze-Xin Jin
- Institute of Natural Medicine and Health Products, School of Pharmaceutical Sciences, Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Taizhou University, Zhejiang 318000, PR China
| | - Jin-Feng Hu
- Institute of Natural Medicine and Health Products, School of Pharmaceutical Sciences, Zhejiang Provincial Key Laboratory of Plant Evolutionary Ecology and Conservation, Taizhou University, Zhejiang 318000, PR China.
| | - Yi-Cheng Mao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, PR China.
| | - Juan Xiong
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai 201203, PR China.
| |
Collapse
|
2
|
Mihaylova R, Momekova D, Elincheva V, Momekov G. Immunoconjugates as an Efficient Platform for Drug Delivery: A Resurgence of Natural Products in Targeted Antitumor Therapy. Pharmaceuticals (Basel) 2024; 17:1701. [PMID: 39770542 PMCID: PMC11677665 DOI: 10.3390/ph17121701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
The present review provides a detailed and comprehensive discussion on antibody-drug conjugates (ADCs) as an evolving new modality in the current therapeutic landscape of malignant diseases. The principle concepts of targeted delivery of highly toxic agents forsaken as stand-alone drugs are examined in detail, along with the biochemical and technological tools for their successful implementation. An extensive analysis of ADCs' major components is conducted in parallel with their function and impact on the stability, efficacy, safety, and resistance profiles of the immunoconjugates. The scope of the article covers the major classes of currently validated natural compounds used as payloads, with an emphasis on their structural and mechanistic features, natural origin, and distribution. Future perspectives in ADCs' design are thoroughly explored, addressing their inherent or emerging challenges and limitations. The survey also provides a comprehensive overview of the molecular rationale for active tumor targeting of ADC-based platforms, exploring the cellular biology and clinical relevance of validated tumor markers used as a "homing" mechanism in both hematological and solid tumor malignancies.
Collapse
Affiliation(s)
- Rositsa Mihaylova
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Denitsa Momekova
- Department “Pharmaceutical Technology and Biopharmaceutics”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria;
| | - Viktoria Elincheva
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| | - Georgi Momekov
- Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (V.E.); (G.M.)
| |
Collapse
|
3
|
Li Y, Cheng Z, Zhou W, Wang L, Li X, Xia G, Lu W, Zhu S. Synthesis and evaluation of homocamptothecin antibody-drug conjugates for cancer treatment. Eur J Med Chem 2024; 279:116899. [PMID: 39321689 DOI: 10.1016/j.ejmech.2024.116899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
As an emerging tumor therapeutic strategy, antibody-drug conjugates (ADCs) overcome the high toxicity of traditional small molecule chemotherapy and improve the targeting of treatment. In this study, we successfully constructed a novel ADC, Tras-16b, for the first time using homocamptothecin 16b as the payload. Tras-16b, at a dose of 3 mg/kg, exhibited comparable anti-tumor activity to Enhertu and demonstrated an enhanced safety profile in the NCI-N87 xenograft model. Notably, this is the first ADC developed based on homocamptothecin, marking a significant advancement with promising prospects for the structural modification of camptothecin ADCs.
Collapse
Affiliation(s)
- Yalong Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, PR China
| | - Zhiyang Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, PR China; Central Research Institute, Shanghai Pharmaceuticals Holding Company Limited, Shanghai, 201203, PR China
| | - Wei Zhou
- Central Research Institute, Shanghai Pharmaceuticals Holding Company Limited, Shanghai, 201203, PR China
| | - Lei Wang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, PR China
| | - Xiaomei Li
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, PR China
| | - Guangxin Xia
- Central Research Institute, Shanghai Pharmaceuticals Holding Company Limited, Shanghai, 201203, PR China.
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, PR China.
| | - Shulei Zhu
- Innovation Center for AI and Drug Discovery, School of Pharmacy, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, PR China.
| |
Collapse
|
4
|
Kamle M, Pandhi S, Mishra S, Barua S, Kurian A, Mahato DK, Rasane P, Büsselberg D, Kumar P, Calina D, Sharifi-Rad J. Camptothecin and its derivatives: Advancements, mechanisms and clinical potential in cancer therapy. Med Oncol 2024; 41:263. [PMID: 39382779 DOI: 10.1007/s12032-024-02527-x] [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: 07/30/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024]
Abstract
Camptothecin (CPT), an alkaloid isolated from the Camptotheca tree, has demonstrated significant anticancer properties in a range of malignancies. However, its therapeutic efficacy is limited by its hydrophobicity, poor bioavailability, and systemic toxicity. Derivatives, analogues, and nanoformulations of CPT have been synthesized to overcome these limitations. The aim of this review is to comprehensively analyze existing studies to evaluate the therapeutic efficacy, mechanistic aspects, and clinical potential of CPT and its modified forms, including derivatives, analogues, and nanoformulations, in cancer treatment. A comprehensive literature review was performed using PubMed/Medline, Scopus, and Web of Science databases; articles were selected based on specific inclusion criteria, and data were extracted on the pharmacological profile, clinical studies, and therapeutic efficacy of CPT and its different forms. Current evidence suggests that derivatives and analogues of CPT have improved water solubility, bioavailability, and reduced systemic toxicity compared to CPT. Nanoformulations further enhance targeted delivery and reduce off-target effects. Clinical trials indicate promising outcomes with enhanced survival rates and lower side effects. CPT and its modified forms hold significant promise as potent anticancer agents. Ongoing research and clinical trials are essential for establishing their long-term efficacy and safety; the evidence overwhelmingly supports further development and clinical testing of these compounds.
Collapse
Affiliation(s)
- Madhu Kamle
- Applied Microbiology Laboratory, Department of Forestry, North Eastern Regional Institute of Science and Technology, Nirjuli, India
| | - Shikha Pandhi
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Sadhna Mishra
- Faculty of Agricultural Sciences, GLA University, Mathura, 281406, India
| | - Sreejani Barua
- Indian Institute of Technology Kharagpur, Kharagpur, 721302, India
| | - Anju Kurian
- Department of Post Graduate Studies and Research in Food Science, St. Aloysius College (Autonomous), Mangalore, 575003, India
| | - Dipendra Kumar Mahato
- CASS Food Research Centre, School of Exercise and Nutrition Sciences, Deakin University, Burwood, VIC, 3125, Australia
| | - Prasad Rasane
- Department of Food Technology and Nutrition, School of Agriculture, Lovely Professional University, Phagwara, 144411, India
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, P.O. Box 24144, Doha, Qatar
| | - Pradeep Kumar
- Department of Botany, University of Lucknow, Uttar Pradesh, Lucknow, India.
- College of Life Science & Biotechnology, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, 092301, Ecuador.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
5
|
Don TM, Hong YT, Jheng PR, Chuang EY, Huang YC. Improved camptothecin encapsulation and efficacy by environmentally sensitive block copolymer/chitosan/fucoidan nanoparticles for targeting lung cancer cells. Int J Biol Macromol 2024; 277:133901. [PMID: 39038585 DOI: 10.1016/j.ijbiomac.2024.133901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/24/2024]
Abstract
In this study, thermo-sensitive poly(N-isopropyl acrylamide) (PNP) was polymerized with pH-sensitive poly(acrylic acid) (PAA) to prepare a PAA-b-PNP block copolymer. Above its cloud point, the block copolymer self-assembled into nanoparticles (NPs), encapsulating the anticancer drug camptothecin (CPT) in situ. Chitosan (CS) and fucoidan (Fu) further modified these NPs, forming Fu-CPT-NPs to enhance biocompatibility, drug encapsulation efficiency (EE), and loading content (LC), crucially facilitating P-selectin targeting of lung cancer cells through a drug delivery system. The EE and LC reached 82 % and 3.5 %, respectively. According to transmission electron microscope observation, these Fu-CPT-NPs had uniform spherical shapes with an average diameter of ca. 250 nm. They could maintain their stability in a pH range of 5.0-6.8. In vitro experimental results revealed that the Fu-CPT-NPs exhibited good biocompatibility and had anticancer activity after encapsulating CPT. It could deliver CPT to cancer cells by targeting P-selectin, effectively increasing cell uptake and inducing cell apoptosis. Animal study results showed that the Fu-CPT-NPs inhibited lung tumor growth by increasing tumor cell apoptosis without causing significant tissue damage related to generating reactive oxygen species in lung cancer cells. This system can effectively improve drug-delivery efficiency and treatment effects and has great potential for treating lung cancer.
Collapse
Affiliation(s)
- Trong-Ming Don
- Department of Chemical and Materials Engineering, Tamkang University, New Taipei City, Taiwan
| | - Yu-Ting Hong
- College of Life Sciences, Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan
| | - Pei-Ru Jheng
- Graduate Institute of Biomedical Materials and Tissue Engineering, International PhD Program in Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, International PhD Program in Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan.
| | - Yi-Cheng Huang
- College of Life Sciences, Department of Food Science, National Taiwan Ocean University, Keelung, Taiwan.
| |
Collapse
|
6
|
Lu Y, Liang Z, Liu L, Zhou Y, Liu C, Zhao Z, Zheng T, Du Q, Liu W. Discovery of novel quinoline scaffold selective estrogen receptor degraders (SERDs) for treatment of ER positive breast cancer with enhanced antiproliferative bioactivity through immunogenic cell death (ICD) effects. Eur J Med Chem 2024; 275:116534. [PMID: 38870830 DOI: 10.1016/j.ejmech.2024.116534] [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: 02/15/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/15/2024]
Abstract
Combination therapy proven to be an effective therapeutic approach for estrogen receptor (ER)-positive breast cancer. Currently, cyclin-dependent kinase 4/6 (CDK4/6) inhibitors are combined with aromatase inhibitors (AIs) or selective estrogen receptor degraders (SERDs) as first-line therapy for advanced ER-positive breast cancer. Herein, a new family of quinoline scaffold SERDs was synthesized and evaluated in MCF-7 cells. Among them, compounds 18j and 24d exhibited remarkable MCF-7 inhibition, both alone and in combination with ribociclib (CDK4/6 inhibitor), in vitro and in vivo. Meanwhile, compounds 18j and 24d effectively degraded ER and inhibited ER downstream signaling pathways. Interestingly, compounds 18j and 24d induced endoplasmic reticulum stress (ERS) and triggered immunogenic cell death (ICD) via damage-associated molecular patterns (DAMPs) in MCF-7 cells. These findings highlight the immune-related and enhanced antiproliferative effects of oral SERDs in ER positive breast cancer treatment.
Collapse
Affiliation(s)
- Yunlong Lu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, 210023, PR China
| | - Zhenlin Liang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Lijuan Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Yanyu Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Chao Liu
- Department of Pharmacy, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029, PR China; School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Zhihao Zhao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Tianpeng Zheng
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Qianming Du
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, PR China; School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China.
| | - Wukun Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|
7
|
Banadka A, Narasimha SW, Dandin VS, Naik PM, Vennapusa AR, Melmaiee K, Vemanna RS, Al-Khayri JM, Thiruvengadam M, Nagella P. Biotechnological approaches for the production of camptothecin. Appl Microbiol Biotechnol 2024; 108:382. [PMID: 38896329 PMCID: PMC11186875 DOI: 10.1007/s00253-024-13187-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024]
Abstract
Camptothecin (CPT), an indole alkaloid popular for its anticancer property, is considered the third most promising drug after taxol and famous alkaloids from Vinca for the treatment of cancer in humans. Camptothecin was first identified in Camptotheca acuminata followed by several other plant species and endophytic fungi. Increased harvesting driven by rising global demand is depleting the availability of elite plant genotypes, such as Camptotheca acuminata and Nothapodytes nimmoniana, crucial for producing alkaloids used in treating diseases like cancer. Conservation of these genotypes for the future is imperative. Therefore, research on different plant tissue culture techniques such as cell suspension culture, hairy roots, adventitious root culture, elicitation strategies, and endophytic fungi has been adopted for the production of CPT to meet the increasing demand without affecting the source plant's existence. Currently, another strategy to increase camptothecin yield by genetic manipulation is underway. The present review discusses the plants and endophytes that are employed for camptothecin production and throws light on the plant tissue culture techniques for the regeneration of plants, callus culture, and selection of cell lines for the highest camptothecin production. The review further explains the simple, accurate, and cost-effective extraction and quantification methods. There is enormous potential for the sustainable production of CPT which could be met by culturing of suitable endophytes or plant cell or organ culture in a bioreactor scale production. Also, different gene editing tools provide opportunities for engineering the biosynthetic pathway of CPT, and the overall CPT production can be improved . KEY POINTS: • Camptothecin is a naturally occurring alkaloid with potent anticancer properties, primarily known for its ability to inhibit DNA topoisomerase I. • Plants and endophytes offer a potential approach for camptothecin production. • Biotechnology approaches like plant tissue culture techniques enhanced camptothecin production.
Collapse
Affiliation(s)
- Akshatha Banadka
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, 560 029, Karnataka, India
| | - Sudheer Wudali Narasimha
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, 560 029, Karnataka, India
| | | | - Poornanand M Naik
- Department of Botany, Karnatak University, Dharwad, 580003, Karnataka, India
| | | | - Kalpalatha Melmaiee
- Department of Agriculture and Natural Resources, Delaware State University, Dover, DE, 19901, USA
| | - Ramu S Vemanna
- Laboratory of Plant Functional Genomics, Regional Center for Biotechnology, Faridabad, 121001, Haryana, India
| | - Jameel M Al-Khayri
- Department of Agricultural Biotechnology, College of Agriculture and Food Sciences, King Faisal University, Al- Ahsa, 31982, Saudi Arabia.
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, South Korea
| | - Praveen Nagella
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, 560 029, Karnataka, India.
| |
Collapse
|
8
|
Zhou X. Recent advances of tryptanthrin and its derivatives as potential anticancer agents. RSC Med Chem 2024; 15:1127-1147. [PMID: 38665827 PMCID: PMC11042161 DOI: 10.1039/d3md00698k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/03/2024] [Indexed: 04/28/2024] Open
Abstract
Tryptanthrin is one of the well-known natural alkaloids with a broad spectrum of biological activities and can act as anti-inflammatory, anticancer, antibacterial, antifungal, antiviral, antitubercular, and other agents. Owing to its potent anticancer activity, tryptanthrin has been widely explored for the therapy of various cancers besides being effective against other diseases. Tryptanthrin with a pharmacological indoloquinazoline moiety can not only be modified by different functional groups to achieve various tryptanthrin derivatives, which may realize the improvement of anticancer activity, but also bind with different metal ions to obtain varied tryptanthrin metal complexes as potential anticancer agents, due to their higher anticancer activities in comparison with tryptanthrin (or its derivatives) and cisplatin. This review outlines the recent advances in the syntheses, structures, and anticancer activities of tryptanthrin derivatives and their metal complexes, trying to reveal their structure-activity relationships and to provide a helpful way for medicinal chemists in the development of new and effective tryptanthrin-based anticancer agents.
Collapse
Affiliation(s)
- Xiaofeng Zhou
- Second Clinical Medicine College of Lanzhou University Lanzhou China
| |
Collapse
|
9
|
Sirinyildiz F, Unay S. N-methyl-D-aspartate receptors and thymoquinone induce apoptosis and alteration in mitochondria in colorectal cancer cells. Med Oncol 2024; 41:123. [PMID: 38652404 DOI: 10.1007/s12032-024-02348-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 02/27/2024] [Indexed: 04/25/2024]
Abstract
Colon cancer is on the rise in both men and women. In addition to traditional treatment methods, herbal treatments from complementary and alternative medicine are actively followed. Naturally derived from plants, thymoquinone (TQ) has drawn a lot of attention in the field of cancer treatment. MK-801, an N-methyl-D-aspartate agonist, is used to improve memory and plasticity, but it has also lately been explored as a potential cancer treatment. This study aimed to determine the roles of N-Methyl-D-Aspartate agonists and Thymoquinone on mitochondria and apoptosis. HT-29 cells were treated with different TQ and MK-801 concentrations. We analyzed cell viability, apoptosis, and alteration of mitochondria. Cell viability significantly decreased depending on doses of TQ and MK-801. Apoptosis and mitochondrial dysfunctions induced by low and high doses of TQ and MK-801. Our study emphasizes the need for further safety evaluation of MK-801 due to the potential toxicity risk of TQ and MK-801. Optimal and toxic doses of TQ and MK-801 were determined for the treatment of colon cancer. It should be considered as a possibility that colon cancer can be treated with TQ and MK-801.
Collapse
Affiliation(s)
- Ferhat Sirinyildiz
- Faculty of Medicine, Department of Physiology, Aydin Adnan Menderes University, Aydin, Turkey
| | - Simge Unay
- Faculty of Medicine, Department of Biophysics, Lokman Hekim University, Ankara, Turkey.
| |
Collapse
|
10
|
Yang S, Lin HS, Zhang L, Chi-Lui Ho P. Formulating 10-hydroxycamptothecin into nanoemulsion with functional excipient tributyrin: An innovative strategy for targeted hepatic cancer chemotherapy. Int J Pharm 2024; 654:123945. [PMID: 38403088 DOI: 10.1016/j.ijpharm.2024.123945] [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: 11/03/2023] [Revised: 02/10/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
This study aimed to develop an innovative dosage form for 10-hydroxycamptothecin (HCPT), a chemotherapeutic agent with limited aqueous solubility and stability, to enhance its parenteral delivery and targeting to hepatic cancer. We formulated HCPT into a nanoemulsion using tributyrin, a dietary component with histone deacetylase inhibitor activity. The resulting HCPT-loaded tributyrin nanoemulsion (Tri-HCPT-E) underwent extensive evaluations. Tri-HCPT-E significantly improved the aqueous solubility, stability, and anti-cancer activities in HepG2 cells. Pharmacokinetic studies confirmed the increased stability and hepatic targeting, with Tri-HCPT-E leading to a 120-fold increase in plasma exposure of intact HCPT and a 10-fold increase in hepatic exposure compared to the commercial free solution. Co-administration of 17α-ethynylestradiol, an up-regulator of low-density lipoprotein (LDL) receptor, further enhanced the distribution and metabolism of HCPT, demonstrating an association between the LDL receptor pathway and hepatic targeting. Most importantly, Tri-HCPT-E exhibited superior in vivo anti-cancer efficacy in a mouse xenograft model compared to the commercial formulation, without causing escalated hepatic or renal toxicity. In conclusion, formulating HCPT into a nanoemulsion with tributyrin has proven to be an innovative and effective strategy for targeted hepatic cancer chemotherapy while tributyrin, a pharmacologically active dietary component, has emerged as a promising functional excipient for drug delivery.
Collapse
Affiliation(s)
- Shili Yang
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Hai-Shu Lin
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore; College of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China.
| | - Li Zhang
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore
| | - Paul Chi-Lui Ho
- Department of Pharmacy, National University of Singapore, Singapore 117543, Singapore; School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Malaysia.
| |
Collapse
|
11
|
Chen Z, Liu M, Wang N, Xiao W, Shi J. Unleashing the Potential of Camptothecin: Exploring Innovative Strategies for Structural Modification and Therapeutic Advancements. J Med Chem 2024; 67:3244-3273. [PMID: 38421819 DOI: 10.1021/acs.jmedchem.3c02115] [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: 03/02/2024]
Abstract
Camptothecin (CPT) is a potent anti-cancer agent targeting topoisomerase I (TOP1). However, CPT has poor pharmacokinetic properties, causes toxicities, and leads to drug resistance, which limit its clinical use. In this paper, to review the current state of CPT research. We first briefly explain CPT's TOP1 inhibition mechanism and the key hurdles in CPT drug development. Then we examine strategies to overcome CPT's limitations through structural modifications and advanced delivery systems. Though modifications alone seem insufficient to fully enhance CPT's therapeutic potential, structure-activity relationship analysis provides insights to guide optimization of CPT analogs. In comparison, advanced delivery systems integrating controlled release, imaging capabilities, and combination therapies via stimulus-responsive linkers and targeting moieties show great promise for improving CPT's pharmacological profile. Looking forward, multifaceted approaches combining selective CPT derivatives with advanced delivery systems, informed by emerging biological insights, hold promise for fully unleashing CPT's anti-cancer potential.
Collapse
Affiliation(s)
- Zheng Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Maoyu Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenjing Xiao
- Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu 610083, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
12
|
Schmitt S, Machui P, Mai I, Herterich S, Wunder S, Cyprys P, Gerlach M, Ochtrop P, Hackenberger CP, Schumacher D, Helma J, Vogl AM, Kasper MA. Design and Evaluation of Phosphonamidate-Linked Exatecan Constructs for Highly Loaded, Stable, and Efficacious Antibody-Drug Conjugates. Mol Cancer Ther 2024; 23:199-211. [PMID: 37828728 PMCID: PMC10831470 DOI: 10.1158/1535-7163.mct-23-0359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/30/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023]
Abstract
Topoisomerase I (TOP1) Inhibitors constitute an emerging payload class to engineer antibody-drug conjugates (ADC) as next-generation biopharmaceutical for cancer treatment. Existing ADCs are using camptothecin payloads with lower potency and suffer from limited stability in circulation. With this study, we introduce a novel camptothecin-based linker-payload platform based on the highly potent camptothecin derivative exatecan. First, we describe general challenges that arise from the hydrophobic combination of exatecan and established dipeptidyl p-aminobenzyl-carbamate (PAB) cleavage sites such as reduced antibody conjugation yields and ADC aggregation. After evaluating several linker-payload structures, we identified ethynyl-phosphonamidates in combination with a discrete PEG24 chain to compensate for the hydrophobic PAB-exatecan moiety. Furthermore, we demonstrate that the identified linker-payload structure enables the construction of highly loaded DAR8 ADCs with excellent solubility properties. Head-to-head comparison with Enhertu, an approved camptothecin-based ADC, revealed improved target-mediated killing of tumor cells, excellent bystander killing, drastically improved linker stability in vitro and in vivo and superior in vivo efficacy over four tested dose levels in a xenograft model. Moreover, we show that ADCs based on the novel exatecan linker-payload platform exhibit antibody-like pharmacokinetic properties, even when the ADCs are highly loaded with eight drug molecules per antibody. This ADC platform constitutes a new and general solution to deliver TOP1 inhibitors with highest efficiency to the site of the tumor, independent of the antibody and its target, and is thereby broadly applicable to various cancer indications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Christian P.R. Hackenberger
- Chemical Biology Department, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|
13
|
Park J, Kang SK, Kwon WS, Jeong I, Kim TS, Yu SY, Cho SW, Chung HC, Rha SY. Novel HER2-targeted therapy to overcome trastuzumab resistance in HER2-amplified gastric cancer. Sci Rep 2023; 13:22648. [PMID: 38114573 PMCID: PMC10730520 DOI: 10.1038/s41598-023-49646-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023] Open
Abstract
Trastuzumab is used to treat HER2-amplified metastatic gastric cancer; however, most patients become trastuzumab-resistant within a year. Knowledge of the mechanisms underlying trastuzumab resistance is required to overcome this limitation. Here, we aimed to elucidate this resistance mechanism using four trastuzumab-resistant (TR) cell lines and investigate the efficacy of HER2-targeted therapies to overcome treatment resistance. Each TR cell line had different phenotypic characteristics. Interestingly, HER2 expression remained as high as the parental cell lines in TR cell lines, suggesting that HER2-targeted agents were still useful. As expected, three tyrosine kinase inhibitors (lapatinib, neratinib, and tucatinib) and one antibody-drug conjugate (trastuzumab deruxtecan: T-DXd) exhibited good antitumor effects against TR cell lines. We further investigated the potential biological mechanism of T-DXd. When treated with trastuzumab or T-DXd, HER2 or its downstream signals were disrupted in parental cell lines, but not in TR cell lines. Moreover, T-DXd induced the expression of pH2A.X and cPARP and caused cell cycle arrest in the S or G2-M phase in TR cell lines. T-DXd showed promising antitumor activity in both parental and TR cell lines, suggesting that it is a potential candidate for overcoming trastuzumab resistance.
Collapse
Affiliation(s)
- Juin Park
- Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sun Kyoung Kang
- Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Woo Sun Kwon
- Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Inhye Jeong
- Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Tae Soo Kim
- Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Seo Young Yu
- Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sang Woo Cho
- Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hyun Cheol Chung
- Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Yonsei Cancer Center, Yonsei University Health System, Seoul, 03722, Republic of Korea
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sun Young Rha
- Song-Dang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Department of Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Yonsei Cancer Center, Yonsei University Health System, Seoul, 03722, Republic of Korea.
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
14
|
Sofi FA, Tabassum N. Natural product inspired leads in the discovery of anticancer agents: an update. J Biomol Struct Dyn 2023; 41:8605-8628. [PMID: 36255181 DOI: 10.1080/07391102.2022.2134212] [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: 05/16/2022] [Accepted: 10/03/2022] [Indexed: 10/24/2022]
Abstract
Natural products have emerged as major leads for the discovery and development of new anti-cancer drugs. The plant-derived anti-cancer drugs account for approximately 60% and the quest for new anti-cancer agents is in progress. Anti-cancer leads have been isolated from plants, animals, marine organisms, and microorganisms from time immemorial. The process of semisynthetic modifications of the parent lead has led to the generation of new anti-cancer agents with improved therapeutic efficacy and minimal side effects. The various chemo-informatics tools, bioinformatics, high-throughput screening, and combinatorial synthesis are able to deliver the new natural product lead molecules. Plant-derived anticancer agents in either late preclinical development or early clinical trials include taxol, vincristine, vinblastine, topotecan, irinotecan, etoposide, paclitaxel, and docetaxel. Similarly, anti-cancer agents from microbial sources include dactinomycin, bleomycin, mitomycin C, and doxorubicin. In this review, we highlighted the importance of natural products leads in the discovery and development of novel anti-cancer agents. The semisynthetic modifications of the parent lead to the new anti-cancer agent are also presented. Further, the leads in the preclinical settings with the potential to become effective anticancer agents are also reviewed.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Firdoos Ahmad Sofi
- Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Srinagar, Jammu & Kashmir, India
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, School of Applied Sciences and Technology, University of Kashmir, Srinagar, Jammu & Kashmir, India
| |
Collapse
|
15
|
Ravi A, Pathigoolla A, Balan H, Gupta R, Raj G, Varghese R, Sureshan KM. Adamantoid Scaffolds for Multiple Cargo Loading and Cellular Delivery as β-Cyclodextrin Inclusion Complexes. Angew Chem Int Ed Engl 2023; 62:e202307324. [PMID: 37384430 DOI: 10.1002/anie.202307324] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/24/2023] [Accepted: 06/29/2023] [Indexed: 07/01/2023]
Abstract
There is huge demand for developing guests that bind β-CD and can conjugate multiple cargos for cellular delivery. We synthesized trioxaadamantane derivatives, which can conjugate up to three cargos per guest. 1 H NMR titration and isothermal titration calorimetry revealed these guests form 1 : 1 inclusion complexes with β-CD with association constants in the order of 103 M-1 . Co-crystallization of β-CD with guests yielded crystals of their 1 : 1 inclusion complexes as determined by single-crystal X-ray diffraction. In all cases, trioxaadamantane core is buried within the hydrophobic cavity of β-CD and three hydroxyl groups are exposed outside. We established biocompatibility using representative candidate G4 and its inclusion complex with β-CD (β-CD⊂G4), by MTT assay using HeLa cells. We incubated HeLa cells with rhodamine-conjugated G4 and established cellular cargo delivery using confocal laser scanning microscopy (CLSM) and fluorescence-activated cell sorting (FACS) analysis. For functional assay, we incubated HeLa cells with β-CD-inclusion complexes of G4-derived prodrugs G6 and G7, containing one and three units of the antitumor drug (S)-(+)-camptothecin, respectively. Cells incubated with β-CD⊂G7 displayed the highest internalization and uniform distribution of camptothecin. β-CD⊂G7 showed higher cytotoxicity than G7, camptothecin, G6 and β-CD⊂G6, affirming the efficiency of adamantoid derivatives in high-density loading and cargo delivery.
Collapse
Affiliation(s)
- Arthi Ravi
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Vithura, 695551, India
| | - Atchutarao Pathigoolla
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Vithura, 695551, India
| | - Haripriya Balan
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Vithura, 695551, India
| | - Ria Gupta
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Vithura, 695551, India
| | - Gowtham Raj
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Vithura, 695551, India
| | - Reji Varghese
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Vithura, 695551, India
| | - Kana M Sureshan
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, Thiruvananthapuram, Vithura, 695551, India
| |
Collapse
|
16
|
Huang YC, Zeng YJ, Lin YW, Tai HC, Don TM. In Situ Encapsulation of Camptothecin by Self-Assembly of Poly(acrylic acid)- b-Poly( N-Isopropylacrylamide) and Chitosan for Controlled Drug Delivery. Polymers (Basel) 2023; 15:polym15112463. [PMID: 37299263 DOI: 10.3390/polym15112463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Camptothecin (CPT) has been shown to exhibit anticancer activity against several cancers. Nevertheless, CPT is very hydrophobic with poor stability, and thus its medical application is limited. Therefore, various drug carriers have been exploited for effectively delivering CPT to the targeted cancer site. In this study, a dual pH/thermo-responsive block copolymer of poly(acrylic acid-b-N-isopropylacrylamide) (PAA-b-PNP) was synthesized and applied to encapsulate CPT. At temperatures above its cloud point, the block copolymer self-assembled to form nanoparticles (NPs) and in situ encapsulate CPT, owing to their hydrophobic interaction as evidenced by fluorescence spectrometry. Chitosan (CS) was further applied on the surface through the formation of a polyelectrolyte complex with PAA for improving biocompatibility. The average particle size and zeta potential of the developed PAA-b-PNP/CPT/CS NPs in a buffer solution were 168 nm and -30.6 mV, respectively. These NPs were still stable at least for 1 month. The PAA-b-PNP/CS NPs exhibited good biocompatibility toward NIH 3T3 cells. Moreover, they could protect the CPT at pH 2.0 with a very slow-release rate. At pH 6.0, these NPs could be internalized by Caco-2 cells, followed by intracellular release of the CPT. They became highly swollen at pH 7.4, and the released CPT was able to diffuse into the cells at higher intensity. Among several cancer cell lines, the highest cytotoxicity was observed for H460 cells. As a result, these environmentally-responsive NPs have the potential to be applied in oral administration.
Collapse
Affiliation(s)
- Yi-Cheng Huang
- Department of Food Science, National Taiwan Ocean University, No. 2, Beining Rd., Zhongzheng Dist., Keelung City 202301, Taiwan
| | - Yang-Jie Zeng
- Department of Food Science, National Taiwan Ocean University, No. 2, Beining Rd., Zhongzheng Dist., Keelung City 202301, Taiwan
| | - Yu-Wei Lin
- Department of Chemical and Materials Engineering, Tamkang University, No. 151 Yingzhuan Rd., Tamsui Dist., New Taipei City 251301, Taiwan
| | - Hung-Chih Tai
- Department of Food Science, National Taiwan Ocean University, No. 2, Beining Rd., Zhongzheng Dist., Keelung City 202301, Taiwan
| | - Trong-Ming Don
- Department of Chemical and Materials Engineering, Tamkang University, No. 151 Yingzhuan Rd., Tamsui Dist., New Taipei City 251301, Taiwan
| |
Collapse
|
17
|
Nipate DS, Rangan K, Kumar A. Copper(II)-Catalyzed Synthesis of Pyrrolo[3,4- b]quinolinediones from o-Amino Carbonyl Compounds and Maleimides. Org Lett 2023. [PMID: 36809005 DOI: 10.1021/acs.orglett.3c00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
A copper(II)-catalyzed cascade synthesis of 1H-pyrrolo[3,4-b]quinoline-1,3(2H)-diones has been achieved from readily available o-amino carbonyl compounds and maleimides. This one-pot cascade strategy involves a copper-catalyzed aza-Michael addition followed by condensation and oxidation to deliver the target molecules. The protocol features a broad substrate scope and excellent functional group tolerance and affords products in moderate to good (44-88%) yields.
Collapse
Affiliation(s)
- Dhananjay S Nipate
- Department of Chemistry, Birla Institute of Technology and Science Pilani, Pilani, Rajasthan 333031, India
| | - Krishnan Rangan
- Department of Chemistry, Birla Institute of Technology and Science Pilani, Hyderabad, Telangana 500078, India
| | - Anil Kumar
- Department of Chemistry, Birla Institute of Technology and Science Pilani, Pilani, Rajasthan 333031, India
| |
Collapse
|
18
|
Su M, Chen Y, Jia L, Zhang Z. Camptothecin-Loaded and Manganese Dioxide-Coated Polydopamine Nanomedicine Used for Magnetic Resonance Imaging Diagnosis and Chemo-Photothermal Therapy for Lung Cancer. Int J Nanomedicine 2022; 17:6687-6705. [PMID: 36597434 PMCID: PMC9805739 DOI: 10.2147/ijn.s359300] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 11/08/2022] [Indexed: 12/29/2022] Open
Abstract
Introduction Camptothecin (CPT) is a cytotoxic quinolone alkaloid (isolated from a traditional Chinese medicine Camptotheca acuminata), used for the treatment of various malignancies, which inhibits DNA topoisomerase I (Topo I). However, its drawbacks, such as poor water solubility, stability, and highly toxic side effects, limit its clinical application. Therefore, CPT needs to be prepared as a nanomedicine to improve solubility, reduce side effects, and synergize with other therapies to improve efficacy. Methods In this work, we constructed CPT NPs (nanoparticles), which were CPT-loaded and manganese dioxide (MnO2)-coated polydopamine (PDA) nanomedicine. In vitro, we explored the antitumor effect including CPT NPs-induced cell proliferation inhibition, apoptosis and ferroptosis for tumor cell lines. In vivo, we established LLC tumor-bearing mice model to evaluate their tumor imaging and anticancer effects. Results CPT NPs improve the water solubility and stability of CPT and reduce its toxic effects. It has good biocompatibility, excellent photothermal conversion ability for photothermal therapy (PTT) and pH release in the tumor microenvironment. It can inhibit tumor cell proliferation, induce apoptosis and result in ferroptosis of tumor cells. More significantly, this nanomedicine can provide information for the location and diagnosis of tumors via magnetic resonance imaging. In general, the nanomedicine integrated with diagnosis and treatment has excellent anticancer effect. Discussion Altogether, this nanomedicine possesses the ability to diagnose and therapy through magnetic resonance imaging and chemo-photothermal therapy, respectively. In addition, the integrated diagnosis and treatment nanomedicine has potential clinical application prospects through treating lung cancer with high efficiency and low side effect. It can support the construction of related nano-delivery systems.
Collapse
Affiliation(s)
- Mingliang Su
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| | - Yukun Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China,Correspondence: Lijun Jia; Zhanxia Zhang, Email ;
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
19
|
Zhang L, Chen D, Zhou C, Yin Y, Wang G, Zhu Q, Li S, Kong X. Synthesis of evodiamine and its derivatives through a visible-light-driven intramolecular C-N-cross-coupling reaction. Tetrahedron Lett 2022. [DOI: 10.1016/j.tetlet.2022.154305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
20
|
Yan BB, Zhao Y, Li M, Li K, Dong L, Yang SY, Luo Z, Yu SH. Engineering Multishelled Nanostructures Enables Stepwise Self-Degradability for Drug-Release Optimization. NANO LETTERS 2022; 22:9181-9189. [PMID: 36374229 DOI: 10.1021/acs.nanolett.2c04229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The balance between degradability and drug release kinetics is a major challenge for the development of drug delivery systems. Here we develop hierarchically structured nanoparticles comprising multiple noncontact silica shells using an amorphous calcium carbonate template. The system could be degraded in a sequential fashion on account of the molecularly engineered multishelled structures. The hydrolysis rate of drug-containing cores is inversely correlated with the nanoparticle concentration due to the shielding effect of the hierarchical nanostructure and could be exploited to regulate the release kinetics. Specifically, multishelled nanospheres show a low drug release rate with high doses that increases steadily as the concentration decreases due to continuous degradation, thus stabilizing the local drug concentration for effective tumor therapy. Moreover, the nanoparticles could be eventually degraded completely, which may reduce their health risks. This kind of hierarchically structured silica-based nanoparticle could serve as a sustainable drug depot and provides a new avenue for tumor treatment.
Collapse
Affiliation(s)
- Bei-Bei Yan
- Department of Chemistry, Institute of Biomimetic Materials & Chemistry, Anhui Engineering Laboratory of Biomimetic Materials, Division of Nanomaterials & Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Yang Zhao
- Department of Chemistry, Institute of Biomimetic Materials & Chemistry, Anhui Engineering Laboratory of Biomimetic Materials, Division of Nanomaterials & Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, People's Republic of China
| | - Ke Li
- School of Life Science, Chongqing University, Chongqing 400044, People's Republic of China
| | - Liang Dong
- Department of Chemistry, Institute of Biomimetic Materials & Chemistry, Anhui Engineering Laboratory of Biomimetic Materials, Division of Nanomaterials & Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Si-Yao Yang
- Department of Chemistry, Institute of Biomimetic Materials & Chemistry, Anhui Engineering Laboratory of Biomimetic Materials, Division of Nanomaterials & Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, People's Republic of China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, People's Republic of China
| | - Shu-Hong Yu
- Department of Chemistry, Institute of Biomimetic Materials & Chemistry, Anhui Engineering Laboratory of Biomimetic Materials, Division of Nanomaterials & Chemistry, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, People's Republic of China
| |
Collapse
|
21
|
Li Z, Chen K, Rose P, Zhu YZ. Natural products in drug discovery and development: Synthesis and medicinal perspective of leonurine. Front Chem 2022; 10:1036329. [PMID: 36324522 PMCID: PMC9618625 DOI: 10.3389/fchem.2022.1036329] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/05/2022] [Indexed: 12/03/2022] Open
Abstract
Natural products, those molecules derived from nature, have been used by humans for thousands of years to treat ailments and diseases. More recently, these compounds have inspired chemists to use natural products as structural templates in the development of new drug molecules. One such compound is leonurine, a molecule isolated and characterized in the tissues of Herb leonuri. This molecule has received attention from scientists in recent years due to its potent anti-oxidant, anti-apoptotic, and anti-inflammatory properties. More recently researchers have shown leonurine to be useful in the treatment of cardiovascular and nervous system diseases. Like other natural products such as paclitaxel and artemisinin, the historical development of leonurine as a therapeutic is very interesting. Therefore, this review provided an overview of natural product discovery, through to the development of a potential new drug. Content will summarize known plant sources, the pathway used in the synthesis of leonurine, and descriptions of leonurine’s pharmacological properties in mammalian systems.
Collapse
Affiliation(s)
- Zhaoyi Li
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Taipa, Macau, China
| | - Keyuan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Taipa, Macau, China
| | - Peter Rose
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom
| | - Yi Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Taipa, Macau, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- *Correspondence: Yi Zhun Zhu,
| |
Collapse
|
22
|
Pulmonary delivery of liposomes co-loaded with SN38 prodrug and curcumin for the treatment of lung cancer. Eur J Pharm Biopharm 2022; 179:156-165. [PMID: 36064084 DOI: 10.1016/j.ejpb.2022.08.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/22/2022] [Accepted: 08/29/2022] [Indexed: 11/21/2022]
Abstract
A co-delivery system of SN38 (7-ethyl-10-hydroxyl camptothecin) prodrug and CUR (curcumin) was designed for the treatment of lung cancer by pulmonary delivery. SN38 was linked to cell-penetrating peptide (CPP) TAT via a polyethylene glycol (PEG) linker to form the SN38 prodrug (TAT-PEG-SN38). Liposomes co-loaded with amphiphilic TAT-PEG-SN38 and curcumin (Lip-TAT-PEG-SN38/CUR) were successfully prepared by a microfluidic method for the treatment of lung cancer via pulmonary delivery. Lip-TAT-PEG-SN38/CUR showed nanometer-sized sphericity and a particle size of 171.21 nm. Besides, Lip-TAT-PEG-SN38/CUR exhibited enhanced antiproliferative effect, increased cell apoptosis induction and improved cell cycle arrest compared to the single agents in vitro. The combination induced significant tumor inhibition in a BALB/c mouse lung cancer model. These results indicated that our SN38 prodrug and curcumin co-delivery system was a promising candidate for lung cancer treatment.
Collapse
|
23
|
Buchelnikova VA, Rogozhin EA, Barashkova AS, Buchelnikov AS, Evstigneev MP. C 60 Fullerene Clusters Stabilize the Biologically Inactive Form of Topotecan. Chem Res Toxicol 2022; 35:1482-1492. [PMID: 35980010 DOI: 10.1021/acs.chemrestox.2c00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
There is a range of experimental proofs that biologically relevant compounds change their activity in the presence of C60 fullerene clusters in aqueous solution, which most frequently act as a nanoplatform for drug delivery. Inspired by this evidence, we made an effort to investigate the interaction of fullerene clusters with the antibiotic topotecan (TPT). This study proceeded in three steps, namely, UV/vis titration to confirm complexation and in vitro assays on proliferating and nonproliferating cells to elucidate the role of C60 fullerene in the putative change in TPT activity. Surprisingly, although the nonproliferating cell assay is consistent with the titration data and confirms complex formation, it contradicted the results of the proliferating cell assay. The latter showed that the mixture of TPT and fullerene affects the cells in the same way as pure TPT, as if there were no fullerenes in solution at all, whereas the action of TPT was expected to be enhanced. We explained this contradiction by the specific stabilization of the biologically inactive carboxylate form of the antibiotic adsorbed in the alkaline shell of large fullerene clusters, which leads to neutralization of the drug delivery function and almost zero net biological effect of the antibiotic in vitro. The practical outcome of the work is that fullerene clusters can be used for the selective delivery of pH-sensitive drug forms.
Collapse
Affiliation(s)
| | - Eugene A Rogozhin
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow 117997, Russia.,Gause Institute of New Antibiotics, Moscow 119021, Russia
| | - Anna S Barashkova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences, Moscow 117997, Russia
| | | | | |
Collapse
|
24
|
Huang M, Liu C, Shao Y, Zhou S, Hu G, Yin S, Pu W, Yu H. Anti-tumor pharmacology of natural products targeting mitosis. Cancer Biol Med 2022; 19:j.issn.2095-3941.2022.0006. [PMID: 35699421 PMCID: PMC9257311 DOI: 10.20892/j.issn.2095-3941.2022.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cancer has been an insurmountable problem in the history of medical science. The uncontrollable proliferation of cancer cells is one of cancer’s main characteristics, which is closely associated with abnormal mitosis. Targeting mitosis is an effective method for cancer treatment. This review summarizes several natural products with anti-tumor effects related to mitosis, focusing on targeting microtubulin, inducing DNA damage, and modulating mitosis-associated kinases. Furthermore, the main disadvantages of several typical compounds, including drug resistance, toxicity to non-tumor tissues, and poor aqueous solubility and pharmacokinetic properties, are also discussed, together with strategies to address them. Improved understanding of cancer cell mitosis and natural products may pave the way to drug development for the treatment of cancer.
Collapse
Affiliation(s)
- Manru Huang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.,State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Caiyan Liu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.,State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yingying Shao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.,State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shiyue Zhou
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.,State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Gaoyong Hu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.,State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuangshuang Yin
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.,State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Weiling Pu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.,State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Haiyang Yu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.,State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
25
|
Yang Y, Hu D, Lu Y, Chu B, He X, Chen Y, Xiao Y, Yang C, Zhou K, Yuan L, Qian Z. Tumor-targeted/reduction-triggered composite multifunctional nanoparticles for breast cancer chemo-photothermal combinational therapy. Acta Pharm Sin B 2022; 12:2710-2730. [PMID: 35755283 PMCID: PMC9214336 DOI: 10.1016/j.apsb.2021.08.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 08/02/2021] [Indexed: 12/20/2022] Open
Abstract
Breast cancer has become the most commonly diagnosed cancer type in the world. A combination of chemotherapy and photothermal therapy (PTT) has emerged as a promising strategy for breast cancer therapy. However, the intricacy of precise delivery and the ability to initiate drug release in specific tumor sites remains a challenging puzzle. Therefore, to ensure that the therapeutic agents are synchronously delivered to the tumor site for their synergistic effect, a multifunctional nanoparticle system (PCRHNs) is developed, which is grafted onto the prussian blue nanoparticles (PB NPs) by reduction-responsive camptothecin (CPT) prodrug copolymer, and then modified with tumor-targeting peptide cyclo(Asp-d-Phe-Lys-Arg-Gly) (cRGD) and hyaluronic acid (HA). PCRHNs exhibited nano-sized structure with good monodispersity, high load efficiency of CPT, triggered CPT release in response to reduction environment, and excellent photothermal conversion under laser irradiation. Furthermore, PCRHNs can act as a photoacoustic imaging contrast agent-guided PTT. In vivo studies indicate that PCRHNs exhibited excellent biocompatibility, prolonged blood circulation, enhanced tumor accumulation, allow tumor-specific chemo-photothermal therapy to achieve synergistic antitumor effects with reduced systemic toxicity. Moreover, hyperthermia-induced upregulation of heat shock protein 70 in the tumor cells could be inhibited by CPT. Collectively, PCRHNs may be a promising therapeutic way for breast cancer therapy.
Collapse
|
26
|
Dashyan SS, Paronikyan EG, Ayvazyan AS, Mamyan SS, Hunanyan LS. Synthesis and Prediction of Antitumor Activity of New Fused Pyrano[3,4-c]pyridines and Pyrano[4′,3′:4,5]pyrido[2,3-d]pyrimidines. RUSS J GEN CHEM+ 2022. [DOI: 10.1134/s1070363222030069] [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
Abstract
One-pot methods were developed for the preparation of 6-amino-3-isopropyl-7-aryl substituted-8-thioxo(oxo)pyrano[3,4-c]pyridines derivatives. The latter served as starting materials for the synthesis of a new pyrano[4′,3′:4,5]pyrido[2,3-d]pyrimidine heterosystems. The proposed mechanisms of one-pot reactions were presented. Antitumor activity of new synthesized compounds was predicted by in silico methods.
Collapse
|
27
|
Lagunas-Rangel FA, Liu W, Schiöth HB. Can Exposure to Environmental Pollutants Be Associated with Less Effective Chemotherapy in Cancer Patients? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19042064. [PMID: 35206262 PMCID: PMC8871977 DOI: 10.3390/ijerph19042064] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023]
Abstract
Since environmental pollutants are ubiquitous and many of them are resistant to degradation, we are exposed to many of them on a daily basis. Notably, these pollutants can have harmful effects on our health and be linked to the development of disease. Epidemiological evidence together with a better understanding of the mechanisms that link toxic substances with the development of diseases, suggest that exposure to some environmental pollutants can lead to an increased risk of developing cancer. Furthermore, several studies have raised the role of low-dose exposure to environmental pollutants in cancer progression. However, little is known about how these compounds influence the treatments given to cancer patients. In this work, we present a series of evidences suggesting that environmental pollutants such as bisphenol A (BPA), benzo[a]pyrene (BaP), persistent organic pollutants (POPs), aluminum chloride (AlCl3), and airborne particulate matter may reduce the efficacy of some common chemotherapeutic drugs used in different types of cancer. We discuss the potential underlying molecular mechanisms that lead to the generation of this chemoresistance, such as apoptosis evasion, DNA damage repair, activation of pro-cancer signaling pathways, drug efflux and action of antioxidant enzymes, among others.
Collapse
Affiliation(s)
- Francisco Alejandro Lagunas-Rangel
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, BMC Box 593, Husargatan 3, 75124 Uppsala, Sweden;
- Correspondence: (F.A.L.-R.); (H.B.S.)
| | - Wen Liu
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, BMC Box 593, Husargatan 3, 75124 Uppsala, Sweden;
| | - Helgi B. Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, BMC Box 593, Husargatan 3, 75124 Uppsala, Sweden;
- Institute of Translational Medicine and Biotechnology, I. M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya Str. Moscow, 119991 Moscow, Russia
- Correspondence: (F.A.L.-R.); (H.B.S.)
| |
Collapse
|
28
|
Malhotra S, Dumoga S, Mehta S, Rao EP, Mohanty S, Singh N. Engineering Cellular Membrane for Dual Mode Therapy Using NIR Responsive Photosensitizer and Reversible Topoisomerase Inhibition Activity. ACS APPLIED BIO MATERIALS 2022; 5:570-582. [PMID: 35040623 DOI: 10.1021/acsabm.1c01070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Extensive research over past few decades has highlighted the challenges of chemotherapy and prompted the need for multimodality therapy because chemotherapy alone cannot fully eradicate the tumor due to physiological barriers in its effective delivery and systemic side effects. It can be mitigated by adopting nanoparticles as more effective delivery method, but none of them completely prevents drug toxicities. Utilizing multiple therapeutic modes such as phototherapy that can act synergistically with chemotherapy in controlling tumor growth, while reducing the overall dosage, could become a preferred route for cancer management. Careful selection of nanoparticle system, which can simultaneously deliver both drug and photosensitizer, can significantly enhance the therapeutic outcome. Therefore, in this paper, we report development and potential of immune-compatible and long circulating nanoerythrosomes for enhancing the therapeutic potential of camptothecin and indocyanine green against murine cancer model. The RBCs membrane simultaneously loaded the nonpolar drug and amphiphilic photosensitizer in its lipid bilayer, which self-assembled to form stable nanoparticles. These nano constructs absorbed light in the near-infrared region and hence are suitable for targeting deep seated tissues. The dual chemo-phototherapy had great effect on cell viability and had therapeutic value.
Collapse
Affiliation(s)
- Sahil Malhotra
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Shweta Dumoga
- Meerut Institute of Engineering and Technology, Meerut 250005, India
| | - Supriya Mehta
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - E Pranshu Rao
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Sujata Mohanty
- Stem Cell Facility, DBT-Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.,Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
29
|
Wang D, Zhang L, Xiao F, Mao GJ, Deng GJ. Electrochemical Selective C3-Thiolation of Quinolines. Org Chem Front 2022. [DOI: 10.1039/d2qo00148a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An electrochemical method has been developed to achieve C3-thiolation of quinoline compounds. This new strategy highlights the maximum atom economy, direct conversion and also the use of simple and readily...
Collapse
|
30
|
Zhou J, Zou Y, Cai Y, Chi F, Huang W, Shi W, Qian H. A designed cyclic peptide based on Trastuzumab used to construct peptide-drug conjugates for its HER2-targeting ability. Bioorg Chem 2021; 117:105453. [PMID: 34736138 DOI: 10.1016/j.bioorg.2021.105453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) has been recognized as an important therapeutic target for its overexpression in many cancers. Trastuzumab is a monoclonal antibody targeting HER2, which has been approved by FDA to treat HER2-positive cancer. In this research, cyclic peptide Cyclo-GCGPep1 was designed based on the binding mode between antibody and HER2 protein in silico, which has been confirmed possessing good affinity with HER2. Cyclo-GCGPep1 was also used to construct peptide-drug conjugates with Camptothecin. Biological evaluations demonstrated that Conjugate 1 has a good antiproliferative activity on SK-BR-3 and NCI-N87 cells. Conjugate 1 retained the pro-apoptotic and Topo I inhibitory ability of Camptothecin. Meanwhile, it has good targeting ability towards HER2-positive cells with the help of Cyclo-GCGPep1. It also has better permeability in the tumor spheroid model than Camptothecin. In summary, the design of cyclic peptide derived from antibody is of significance for the discovery of targeting peptides and Conjugate 1 is expected as a good therapeutic agent for HER2-positive cancers.
Collapse
Affiliation(s)
- Jiaqi Zhou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Yuxing Zou
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Yan Cai
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Fanglian Chi
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Wenlong Huang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China
| | - Wei Shi
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China.
| | - Hai Qian
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China; Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, PR China.
| |
Collapse
|
31
|
Du X, Gao Y, Kang Q, Xing J. Design and Applications of Tumor Microenvironment-Responsive Nanogels as Drug Carriers. Front Bioeng Biotechnol 2021; 9:771851. [PMID: 34746113 PMCID: PMC8569621 DOI: 10.3389/fbioe.2021.771851] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/08/2021] [Indexed: 12/03/2022] Open
Abstract
In recent years, the exploration of tumor microenvironment has provided a new approach for tumor treatment. More and more researches are devoted to designing tumor microenvironment-responsive nanogels loaded with therapeutic drugs. Compared with other drug carriers, nanogel has shown great potential in improving the effect of chemotherapy, which is attributed to its stable size, superior hydrophilicity, excellent biocompatibility, and responsiveness to specific environment. This review primarily summarizes the common preparation techniques of nanogels (such as free radical polymerization, covalent cross-linking, and physical self-assembly) and loading ways of drug in nanogels (including physical encapsulation and chemical coupling) as well as the controlled drug release behaviors. Furthermore, the difficulties and prospects of nanogels as drug carriers are also briefly described.
Collapse
Affiliation(s)
- Xinjing Du
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Yuting Gao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Qi Kang
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinfeng Xing
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
32
|
Wawruszak A, Halasa M, Okon E, Kukula-Koch W, Stepulak A. Valproic Acid and Breast Cancer: State of the Art in 2021. Cancers (Basel) 2021; 13:3409. [PMID: 34298623 PMCID: PMC8306563 DOI: 10.3390/cancers13143409] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Valproic acid (2-propylpentanoic acid, VPA) is a short-chain fatty acid, a member of the group of histone deacetylase inhibitors (HDIs). VPA has been successfully used in the treatment of epilepsy, bipolar disorders, and schizophrenia for over 50 years. Numerous in vitro and in vivo pre-clinical studies suggest that this well-known anticonvulsant drug significantly inhibits cancer cell proliferation by modulating multiple signaling pathways. Breast cancer (BC) is the most common malignancy affecting women worldwide. Despite significant progress in the treatment of BC, serious adverse effects, high toxicity to normal cells, and the occurrence of multi-drug resistance (MDR) still limit the effective therapy of BC patients. Thus, new agents which improve the effectiveness of currently used methods, decrease the emergence of MDR, and increase disease-free survival are highly needed. This review focuses on in vitro and in vivo experimental data on VPA, applied individually or in combination with other anti-cancer agents, in the treatment of different histological subtypes of BC.
Collapse
Affiliation(s)
- Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (M.H.); (E.O.); (A.S.)
| | - Marta Halasa
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (M.H.); (E.O.); (A.S.)
| | - Estera Okon
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (M.H.); (E.O.); (A.S.)
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-093 Lublin, Poland; (M.H.); (E.O.); (A.S.)
| |
Collapse
|
33
|
Yang M, Wang Q, Liu Y, Hao X, Wang C, Liang Y, Chen J, Xiao Y, Kai G. Divergent camptothecin biosynthetic pathway in Ophiorrhiza pumila. BMC Biol 2021; 19:122. [PMID: 34134716 PMCID: PMC8207662 DOI: 10.1186/s12915-021-01051-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/13/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The anticancer drug camptothecin (CPT), first isolated from Camptotheca acuminata, was subsequently discovered in unrelated plants, including Ophiorrhiza pumila. Unlike known monoterpene indole alkaloids, CPT in C. acuminata is biosynthesized via the key intermediate strictosidinic acid, but how O. pumila synthesizes CPT has not been determined. RESULTS In this study, we used nontargeted metabolite profiling to show that 3α-(S)-strictosidine and 3-(S), 21-(S)-strictosidinic acid coexist in O. pumila. After identifying the enzymes OpLAMT, OpSLS, and OpSTR as participants in CPT biosynthesis, we compared these enzymes to their homologues from two other representative CPT-producing plants, C. acuminata and Nothapodytes nimmoniana, to elucidate their phylogenetic relationship. Finally, using labelled intermediates to resolve the CPT biosynthesis pathway in O. pumila, we showed that 3α-(S)-strictosidine, not 3-(S), 21-(S)-strictosidinic acid, is the exclusive intermediate in CPT biosynthesis. CONCLUSIONS In our study, we found that O. pumila, another representative CPT-producing plant, exhibits metabolite diversity in its central intermediates consisting of both 3-(S), 21-(S)-strictosidinic acid and 3α-(S)-strictosidine and utilizes 3α-(S)-strictosidine as the exclusive intermediate in the CPT biosynthetic pathway, which differs from C. acuminata. Our results show that enzymes likely to be involved in CPT biosynthesis in O. pumila, C. acuminata, and N. nimmoniana have evolved divergently. Overall, our new data regarding CPT biosynthesis in O. pumila suggest evolutionary divergence in CPT-producing plants. These results shed new light on CPT biosynthesis and pave the way towards its industrial production through enzymatic or metabolic engineering approaches.
Collapse
Affiliation(s)
- Mengquan Yang
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Core Facility Centre, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Qiang Wang
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang China
- Institute of Plant Biotechnology, School of Life Sciences, Shanghai Normal University, Shanghai, 200234 China
| | - Yining Liu
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Core Facility Centre, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Xiaolong Hao
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang China
| | - Can Wang
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang China
| | - Yuchen Liang
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Core Facility Centre, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Jianbo Chen
- Institute of Plant Biotechnology, School of Life Sciences, Shanghai Normal University, Shanghai, 200234 China
| | - Youli Xiao
- CAS Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Core Facility Centre, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032 China
| | - Guoyin Kai
- Laboratory of Medicinal Plant Biotechnology, College of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053 Zhejiang China
| |
Collapse
|
34
|
Lauria A, La Monica G, Bono A, Martorana A. Quinoline anticancer agents active on DNA and DNA-interacting proteins: From classical to emerging therapeutic targets. Eur J Med Chem 2021; 220:113555. [PMID: 34052677 DOI: 10.1016/j.ejmech.2021.113555] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 12/25/2022]
Abstract
Quinoline is one of the most important and versatile nitrogen heterocycles embodied in several biologically active molecules. Within the numerous quinolines developed as antiproliferative agents, this review is focused on compounds interfering with DNA structure or with proteins/enzymes involved in the regulation of double helix functional processes. In this light, a special focus is given to the quinoline compounds, acting with classical/well-known mechanisms of action (DNA intercalators or Topoisomerase inhibitors). In particular, the quinoline drugs amsacrine and camptothecin (CPT) have been studied as key lead compounds for the development of new agents with improved PK and tolerability properties. Moreover, notable attention has been paid to the quinoline molecules, which are able to interfere with emerging targets involved in cancer progression, as G-quadruplexes or the epigenetic ones (e.g.: histone deacetylase, DNA and histones methyltransferase). The antiproliferative and the enzymatic inhibition data of the reviewed compounds have been analyzed. Furthermore, concerning the SAR (structure-activity relationship) aspects, the most recurrent ligand-protein interactions are summarized, underling the structural requirements for each kind of mechanism of action.
Collapse
Affiliation(s)
- Antonino Lauria
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy
| | - Gabriele La Monica
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy
| | - Alessia Bono
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy
| | - Annamaria Martorana
- Dipartimento di Scienze e Technologie Biologiche Chimiche e Farmaceutiche "STEBICEF" - University of Palermo, Via Archirafi - 32, 90123, Palermo, Italy.
| |
Collapse
|
35
|
Ahmed EM, Khalil NA, Zaher AF, Alhamaky SM, El-Zoghbi MS. Synthesis, molecular modeling and biological evaluation of new benzo[4,5]thieno[3,2-b]pyran derivatives as topoisomerase I-DNA binary complex poisons. Bioorg Chem 2021; 112:104915. [PMID: 33905973 DOI: 10.1016/j.bioorg.2021.104915] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/05/2021] [Accepted: 04/13/2021] [Indexed: 12/20/2022]
Abstract
A series of new benzo[b]thiophenes 2a-f and benzo[4,5]thieno[3,2-b]pyran derivatives 3a-f and 4a-f were synthesized and their structures were confirmed by elemental analyses and spectral data. All synthesized compounds were evaluated by the National Cancer Institute (NCI, USA) against 60 human tumor cell lines. Compounds 3a-f and 4a-f showed potent cytotoxic effects in one dose assay with mean growth inhibition ranging from 62% to 80%. Six compounds 3a, 3d, 3e, 3f, 4d and 4e were selected by NCI, USA for five dose evaluation against 60 human tumor cell lines. Compounds 3a, 3d, 3e and 3f exhibited very potent and broad spectrum cytotoxicity against almost all cancer cell lines with mean concentration that yield 50% growth inhibition (MG-MID GI50) of 0.1-0.58 µM and mean concentration that produce 100% growth inhibition (MG-MID TGI) of 6.03-10.00 µM. Compounds 4d and 4e exhibited very potent and selective cytotoxic activity against MDA-MB-435 subpanel (melanoma cancer) with GI50 of 0.45 µM and 0.59 µM, respectively. The mechanism of antiproliferative activity was determined for the most active compounds 3a, 3d, 3e, 3f, 4d, and 4evia measuring their half maximal inhibitory concentration (IC50) against topoisomerase I enzyme at different concentrations. Compounds 3a and 3e exhibited excellent activity compared with reference drugs with IC50 of 0.295 µM and 0.219 µM, respectively. Plasmid DNA nicking assay verified that these compounds are topoisomerase I poisons not suppressors. The active compound 3e induced a significant disruption in the cell cycle profile parallel to its effect on apoptosis induction.
Collapse
Affiliation(s)
- Eman M Ahmed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Kasr El-Aini Street, 11562, Egypt
| | - Nadia A Khalil
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Kasr El-Aini Street, 11562, Egypt.
| | - Ashraf F Zaher
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Kasr El-Aini Street, 11562, Egypt
| | - Shimaa M Alhamaky
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Menoufia University, Shibin El kom, Gamal Abd El-Nasir Street, Menoufia, Egypt
| | - Mona S El-Zoghbi
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Menoufia University, Shibin El kom, Gamal Abd El-Nasir Street, Menoufia, Egypt.
| |
Collapse
|
36
|
Sudula SR, Jala R, Siddoju K, Ega JK. One pot synthesis of some new N-allyl and N-benzyl quinazolinones and their anti-inflammatory activity. J INDIAN CHEM SOC 2021. [DOI: 10.1016/j.jics.2021.100033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
37
|
Polymeric micelles coated with hybrid nanovesicles enhance the therapeutic potential of the reversible topoisomerase inhibitor camptothecin in a mouse model. Acta Biomater 2021; 121:579-591. [PMID: 33285325 DOI: 10.1016/j.actbio.2020.11.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/17/2020] [Accepted: 11/27/2020] [Indexed: 12/24/2022]
Abstract
Nanoparticles with longer blood circulation, high loading capacity, controlled release at the targeted site, and preservation of camptothecin (CPT) in its lactone form are the key characteristics for the effective delivery of CPT. In this regard, natural membrane-derived nanovesicles, particularly those derived from RBC membrane, are important. RBC membrane can be engineered to form vesicles or can be coated over synthetic nanoparticles, without losing their basic structural features and can have prolonged circulation time. Here, we developed a hybrid system to encapsulate CPT inside the amphiphilic micelle and coat it with RBC membrane. Thus, it uses the dual ability of polymeric micelles to preserve CPT in its active form, while maintaining its "stealth" effect due to conserved RBC membrane coating. The hybrid system stabilized 60% of the drug in its active form even after 30 h of incubation in serum, in contrast to 15% active form present in free drug formulation after 1 h of incubation. It showed strong retention inside the Ehrlich Ascites Carcinoma (EAC) mice models for at least 72 h, suggesting camouflaging ability conferred by RBC membrane. Additionally, the nano formulation retarded the tumor growth rate more efficiently than free drug, with no evident signs of necrotic skin lesions. Histopathological analysis showed a significant reduction in cardiac atrophy, hepato-renal degeneration, and lung metastasis, which resulted in the increased overall survival of mice treated with the nano formulation. Hence, CPT-loaded polymeric micelles when coated with RBC membrane can prove to be a better system for the delivery of poorly soluble drug camptothecin.
Collapse
|
38
|
Liu Y, Yang Y, Zhang Q, Lu D, Li S, Li J, Yang G, Shan Y. Dynamics of delivering aptamer targeted nano-drugs into cells. J Mater Chem B 2021; 9:952-957. [PMID: 33437980 DOI: 10.1039/d0tb02527e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A targeted nano-drug delivery system has provided great potential and benefits to the diagnosis and therapy of cancers. Cell entry is a critical step for taking effect of the targeted nano-drug. In this report, the dynamics of delivering a single aptamer targeted polyamindoamine-camptothecin-AS1411 (PAMAM-CPT-AS1411) nano-drug into cells was investigated using a force tracing technique based on atomic force microscopy. The results show that the specific interaction of AS1411 and nucleolin, which is overexpressed on cancer cells, enhances the efficiency of the PAMAM-CPT-AS1411 cell entry. Moreover, the specific interaction induced receptor-mediated endocytosis prolongs the duration and decreases the speed of a single PAMAM-CPT-AS1411 cell entry, which is helpful to understand the targeted nano-drugs prolonging the therapeutic drug level. However, the required force for PAMAM-CPT-AS1411 cell entry is not changed. This report will provide a novel and potential method for achieving the precise dynamics of targeted nano-drug delivery.
Collapse
Affiliation(s)
- Yulin Liu
- School of Chemistry and Life Science, Advanced Institute of Materials Science, Changchun University of Technology, Changchun 130012, China.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Packiam K, Dhakshinamoorthy M. Camptothecin: An anticancer drug from Pestalotiopsis microspora Mh458929 – An endophytic fungus isolated from an ethnopharmacologically important medicinal plant Cordia dichotoma G. forst. Pharmacogn Mag 2021. [DOI: 10.4103/pm.pm_417_20] [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] Open
|
40
|
Yahia HB, Sabri S, Essehli R, Kasak P, Drogosz-Stachowicz J, Janecka A, El Bali B. Crystal Growth, Single Crystal Structure, and Biological Activity of Thiazolo-Pyridine Dicarboxylic Acid Derivatives. ACS OMEGA 2020; 5:27756-27765. [PMID: 33163758 PMCID: PMC7643073 DOI: 10.1021/acsomega.0c01769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/06/2020] [Indexed: 06/11/2023]
Abstract
Four novel TPDCA derivatives were prepared via a supersaturation method combining TPDCA with water, N-methyl-2-pyrrolidone (NMP), Na(PO2H2), and ammonia solution: 2(C9H7NO5S)H2O (1), (C9H7NO5S)C5H9NO (2), (C9H7NO5S)Na(PO2H2) (3), and (C9H5NO5S)(NH4)2(H2O) (4). Their crystal structures were determined by single-crystal X-ray diffraction. Compounds (1) and (2) crystallize in the monoclinic space groups P21 and P21/c, respectively, whereas compounds (3) and (4) crystallize in the triclinic space group P1̅. Weak and moderate hydrogen bonds were detected in the four compounds. In the biological tests, (1) and (3) exhibited significant antibacterial activity against Escherichia coli and Staphylococcus aureus; in addition, (1) was cytotoxic against leukemia HL-60 cells with the IC50 value of 158.5 ± 12.5 μM.
Collapse
Affiliation(s)
- Hamdi Ben Yahia
- Qatar
Environment and Energy Research Institute (QEERI 2.0), Hamad Bin Khalifa University, Qatar Foundation, P.O. Box 34110 Doha, Qatar
| | - Souhir Sabri
- Qatar
Environment and Energy Research Institute (QEERI 2.0), Hamad Bin Khalifa University, Qatar Foundation, P.O. Box 34110 Doha, Qatar
| | - Rachid Essehli
- Energy
and Transportation Science Division, Oak
Ridge National Laboratory, Oak Ridge, 37831-2008 Tennessee, United States
| | - Peter Kasak
- Centre
for Advanced Materials (CAM), Qatar University, 2713 Doha, Qatar
| | - Joanna Drogosz-Stachowicz
- Department
of Biomolecular Chemistry, Medical University
of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Anna Janecka
- Department
of Biomolecular Chemistry, Medical University
of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Brahim El Bali
- Independent
scientist, LMSAC, Po. Box 717, Oujda 60000, Morocco
| |
Collapse
|
41
|
Moise IM, Bîcu E, Farce A, Dubois J, Ghinet A. Indolizine-phenothiazine hybrids as the first dual inhibitors of tubulin polymerization and farnesyltransferase with synergistic antitumor activity. Bioorg Chem 2020; 103:104184. [DOI: 10.1016/j.bioorg.2020.104184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/03/2020] [Accepted: 08/07/2020] [Indexed: 01/21/2023]
|
42
|
Beghennou A, Passador K, Passador A, Corcé V, Thorimbert S, Botuha C. Synthetic Strategy Studies for a Concise Access to Functionalized Pyrano[4,3‐
b
]pyridin‐7‐ones: An Entry to Semi‐Rigid Analogs of Antihistamines. European J Org Chem 2020. [DOI: 10.1002/ejoc.202001016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Anissa Beghennou
- Institut Parisien de Chimie Moléculaire Sorbonne Université CNRS 4 place Jussieu 75005 Paris France
| | - Kevin Passador
- Institut Parisien de Chimie Moléculaire Sorbonne Université CNRS 4 place Jussieu 75005 Paris France
| | - Anthony Passador
- Institut Parisien de Chimie Moléculaire Sorbonne Université CNRS 4 place Jussieu 75005 Paris France
| | - Vincent Corcé
- Institut Parisien de Chimie Moléculaire Sorbonne Université CNRS 4 place Jussieu 75005 Paris France
| | - Serge Thorimbert
- Institut Parisien de Chimie Moléculaire Sorbonne Université CNRS 4 place Jussieu 75005 Paris France
| | - Candice Botuha
- Institut Parisien de Chimie Moléculaire Sorbonne Université CNRS 4 place Jussieu 75005 Paris France
| |
Collapse
|
43
|
Chu B, Qu Y, He X, Hao Y, Yang C, Yang Y, Hu D, Wang F, Qian Z. ROS‐Responsive Camptothecin Prodrug Nanoparticles for On‐Demand Drug Release and Combination of Chemotherapy and Photodynamic Therapy. ADVANCED FUNCTIONAL MATERIALS 2020. [DOI: 10.1002/adfm.202005918] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Bingyang Chu
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center for Biotherapy Chengdu Sichuan 610041 P. R. China
| | - Ying Qu
- Department of Hematology and Research Laboratory of Hematology State Key Laboratory of Biotherapy West China Hospital Sichuan University Collaborative Innovation Center for Biotherapy Chengdu Sichuan 610041 P. R. China
| | - Xinlong He
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center for Biotherapy Chengdu Sichuan 610041 P. R. China
| | - Ying Hao
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center for Biotherapy Chengdu Sichuan 610041 P. R. China
| | - Chengli Yang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center for Biotherapy Chengdu Sichuan 610041 P. R. China
| | - Yun Yang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center for Biotherapy Chengdu Sichuan 610041 P. R. China
| | - Danrong Hu
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center for Biotherapy Chengdu Sichuan 610041 P. R. China
| | - Fangfang Wang
- Department of Hematology and Research Laboratory of Hematology State Key Laboratory of Biotherapy West China Hospital Sichuan University Collaborative Innovation Center for Biotherapy Chengdu Sichuan 610041 P. R. China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Collaborative Innovation Center for Biotherapy Chengdu Sichuan 610041 P. R. China
| |
Collapse
|
44
|
Penteado F, Bettanin L, Machado K, Perin G, Alves D, Lenardão EJ. Sonochemistry and Copper Catalysis: An Efficient Duo in the Synthesis of Chalcogenylindolizines. ASIAN J ORG CHEM 2020. [DOI: 10.1002/ajoc.202000318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Filipe Penteado
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Luana Bettanin
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Kethelyn Machado
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Gelson Perin
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Diego Alves
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Eder J. Lenardão
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| |
Collapse
|
45
|
Zheng T, Zhou H, Li X, Peng D, Yang Y, Zeng Y, Liu H, Ren J, Zhao Y. RBMX is required for activation of ATR on repetitive DNAs to maintain genome stability. Cell Death Differ 2020; 27:3162-3176. [PMID: 32494026 DOI: 10.1038/s41418-020-0570-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 01/11/2023] Open
Abstract
ATR is a master regulator of cell response to replication stress. Adequate activation of ATR is essential for preventing genome aberrance induced by replication defect. However, the mechanism underlying ATR activation is not fully understood. Here, we identify that RBMX is an ssDNA binding protein that orchestrates a novel pathway to activate ATR. Using super-resolution STORM, we observe that RBMX and RPA bind to adjacent but nonoverlapping sites on ssDNA in response to replication stress. RBMX then binds to and facilitates positioning of TopBP1, which activates nearby ATR associated with RPA. In addition, ATR activation by ssDNA-RBMX-TopBP1 is independent of ssDNA-dsDNA junction and 9-1-1 complex. ChIP-seq analysis reveals that RBMX/RPA are highly enriched on repetitive DNAs, which are considered as fragile sites with high replication stress. RBMX depletion leads to defective localization of TopBP1 to replication stressed sites and inadequate activation of ATR. Furthermore, cells with deficient RBMX demonstrate replication defect, leading to formation of micronuclei and a high rate of sister-chromatin exchange, indicative of genome instability. Together, the results identify a new ssDNA-RBMX-TopBP1 pathway that is specifically required for activation of ATR on repetitive DNAs. Therefore, RBMX is a key factor to ensure genome stability during replication.
Collapse
Affiliation(s)
- Tian Zheng
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Haoxian Zhou
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xiaocui Li
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Di Peng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510006, China
| | - Yiding Yang
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yanru Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510006, China
| | - Haiying Liu
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jian Ren
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510006, China
| | - Yong Zhao
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China. .,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510006, China.
| |
Collapse
|
46
|
Penteado F, Gomes CS, Monzon LI, Perin G, Silveira CC, Lenardão EJ. Photocatalytic Synthesis of 3-Sulfanyl- and 1,3-Bis(sulfanyl)indolizines Mediated by Visible Light. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000162] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Filipe Penteado
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| | - Caroline S. Gomes
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| | - Loana I. Monzon
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| | - Gelson Perin
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| | - Claudio C. Silveira
- Departamento de Química; Universidade Federal de Santa Maria - UFSM; CEP: 97105-900 Santa Maria RS Brazil
| | - Eder J. Lenardão
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| |
Collapse
|
47
|
Oliveira CG, Romero-Canelón I, Silva MM, Coverdale JPC, Maia PIS, Batista AA, Castelli S, Desideri A, Sadler PJ, Deflon VM. Palladium(ii) complexes with thiosemicarbazones derived from pyrene as topoisomerase IB inhibitors. Dalton Trans 2020; 48:16509-16517. [PMID: 31670343 DOI: 10.1039/c9dt02570g] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
New palladium complexes with thiosemicarbazonate ligands derived from pyrene exhibit potent antiproliferative activity against A2780 and cisplatin-resistant A2780Cis human ovarian cancer cells, which is dependent on substituent groups of the thiosemicarbazone ligands. Cellular accumulation and distribution studies confirmed that palladium enters the cell nucleus. DNA and topoisomerase IB studies show that one complex is a potent TopIB inhibitor, with selectivity for cancer versus normal cells.
Collapse
Affiliation(s)
- Carolina G Oliveira
- São Carlos Institute of Chemistry, University of São Paulo, 13560-970, São Carlos, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sun Y, Liang Y, Hao N, Fu X, He B, Han S, Cao J, Ma Q, Xu W, Sun Y. Novel polymeric micelles as enzyme-sensitive nuclear-targeted dual-functional drug delivery vehicles for enhanced 9-nitro-20(S)-camptothecin delivery and antitumor efficacy. NANOSCALE 2020; 12:5380-5396. [PMID: 32022069 DOI: 10.1039/c9nr10574c] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
9-Nitro-20(S)-camptothecin (9-NC) is a broad-spectrum antitumor drug used in tumor treatments, but its clinical applications and antitumor efficacy are limited by its structural instability, poor solubility, and extremely low drug utilization in tumor tissues. In this study, enzyme-sensitive nuclear-targeted dual-functional polymeric micelles were developed for 9-NC delivery with a high drug loading content (12.93 ± 0.88%), steady-state circulation, and a rapid attack at the "heart" of tumor cells. Briefly, chrysin (CHR) as a π-conjugated moiety was immobilized on the PCL terminal in the TAT-PCL amphiphiles and combined with the ALAL peptide as a linker on HA chains to yield the ultimate CHR-PCL-TAT-ALAL-HA (HATPC) amphiphiles. Spherical 9-NC-loaded micelles were obtained from the self-assembly of the dual-functional amphiphiles comprising HATPC and 9-NC with uniform nanosize (121.6 ± 5.79 nm), well-distributed morphology (PDI: 0.256), and negative surface charge (-23.2 ± 0.5 mV), yielding high stability during blood circulation. In this drug delivery system, HA acts as an active tumor-targeting instrument via CD44-receptor-mediated endocytosis; further, the ALAL peptide could be cutoff in the lysosomes of the tumor cells due to the high expression of cathepsin B, leading to lysosomal escape, while the secondary polymeric micelles targeted the tumor cell nucleus via the exposed TAT peptide. The enzyme sensitivity and nuclei targetability of the 9-NC/HATPC micelles were confirmed by dynamic light scattering and confocal laser scanning microscopy analyses. As compared to free 9-NC and traditional mPEG2k-PCL2k polymeric micelles, 9-NC/HATPC micelles were the most concentrated in the tumor cell nucleus; therefore, they exhibited the highest cytotoxicity against SKOV3 tumor cells both in vitro (IC50 = 0.03 μg mL-1) and in vivo. This enzyme-sensitive nuclear-targeted dual-functional drug delivery system involving HATPC provided a new and promising strategy for enhanced 9-NC delivery and antitumor efficacy.
Collapse
Affiliation(s)
- Yalin Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Na Hao
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xiaoheng Fu
- Department of Clinical laboratory, No.971 Hospital of the People's Liberation Army Navy, Qingdao 266021, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Shangcong Han
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Jie Cao
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Qingming Ma
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| | - Wen Xu
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
49
|
Sarker SD, Nahar L, Miron A, Guo M. Anticancer natural products. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2020. [DOI: 10.1016/bs.armc.2020.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
50
|
Chen TC, Yu DS, Chen SJ, Chen CL, Lee CC, Hsieh YY, Chang LC, Guh JH, Lin JJ, Huang HS. Design, synthesis and biological evaluation of tetracyclic azafluorenone derivatives with topoisomerase I inhibitory properties as potential anticancer agents. ARAB J CHEM 2019. [DOI: 10.1016/j.arabjc.2016.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|