1
|
Owoseni O, Adekiya TA, Akinboye ES, Adesina SK. Development of a Prostate-Specific Antigen Targeted Dual Drug Conjugate for Prostate Cancer Therapy. ACS OMEGA 2025; 10:17611-17625. [PMID: 40352517 PMCID: PMC12059896 DOI: 10.1021/acsomega.4c11483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 05/14/2025]
Abstract
Prostate cancer (PC) is the most frequently diagnosed cancer and the second leading cause of cancer deaths in American men. While localized cases can be cured through surgery or local radiation, metastatic PC lacks curative therapy. The challenges to the success of treating advanced PC include the adverse effects of current treatment strategies, the continuous generation of cancer cells by cancer stem cells, and tumor heterogeneity. To overcome these challenges, researchers have explored the prodrug approach for targeted, combination drug delivery or release of cytotoxic agents specifically to sites of metastatic PC to improve therapeutic efficacy while decreasing systemic toxicity. The objective of this study is to develop a dual drug conjugate with a prostate-specific antigen (PSA) peptide recognition sequence for PC-specific combination SN-38 and cabazitaxel delivery in the treatment of advanced PC. To achieve this, His-Ser-Ser-Lys-Leu-Glu terminated with the dibenzocyclooctyne (DBCO) functional group conjugated to SN-38 via a leucine spacer was synthesized. Similarly, His-Ser-Ser-Lys-Leu-Glu terminated with azido poly(ethylene glycol) conjugated to cabazitaxel via a leucine spacer was synthesized. The conjugates were linked together via click chemistry to synthesize a dual drug conjugate. In vitro exposure to exogenous PSA was found to trigger the release of cytotoxic drugs. The dual drug conjugate exhibited time- and concentration-dependent cytotoxicity in PC-3 and LNCaP PC cells. The observed cytotoxicity was also dependent on PSA expression in the cellular models tested. This study demonstrates the potential of peptide-drug conjugates for the delivery of combination chemotherapeutics for selective cytotoxicity to PC cells.
Collapse
Affiliation(s)
- Oluwanifemi
B. Owoseni
- Department
of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington
D.C. 20059, United States
| | - Tayo Alex Adekiya
- Department
of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington
D.C. 20059, United States
| | - Emmanuel S. Akinboye
- Department
of Chemistry, Howard University, Washington D.C. 20059, United States
| | - Simeon K. Adesina
- Department
of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington
D.C. 20059, United States
| |
Collapse
|
2
|
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
|
3
|
Sadat SMA, Vakili MR, Abd-El Hafeez SI, Paladino M, Hall DG, Weinfeld M, Lavasanifar A. Synergistic Nanomedicine Delivering Topoisomerase I Toxin (SN-38) and Inhibitors of Polynucleotide Kinase 3'-Phosphatase (PNKP) for Enhanced Treatment of Colorectal Cancer. Mol Pharm 2024; 21:3240-3255. [PMID: 38785196 DOI: 10.1021/acs.molpharmaceut.4c00007] [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] [Indexed: 05/25/2024]
Abstract
Inhibitors of a DNA repair enzyme known as polynucleotide kinase 3'-phosphatase (PNKP) are expected to show synergistic cytotoxicity in combination with topoisomerase I (TOP1) inhibitors in cancer. In this study, the synergistic cytotoxicity of a novel inhibitor of PNKP, i.e., A83B4C63, with a potent TOP1 inhibitor, i.e., SN-38, against colorectal cancer cells was investigated. Polymeric micelles (PMs) for preferred tumor delivery of A83B4C63, developed through physical encapsulation of this compound in methoxy poly(ethylene oxide)-poly(α-benzyl carboxylate-ε-caprolactone) (mPEO-b-PBCL) micelles, were combined with SN-38 in free or PM form. The PM form of SN-38 was prepared through chemical conjugation of SN-38 to the functional end group of mPEO-b-PBCL and further assembly of mPEO-b-PBCL-SN-38 in water. Moreover, mixed micelles composed of mPEO-b-PBCL and mPEO-b-PBCL-SN-38 were used to co-load A83B4C63 and SN-38 in the same nanoformulation. The loading content (% w/w) of the SN-38 and A83B4C63 to mPEO-b-PBCL in the co-loaded formulation was 7.91 ± 0.66 and 16.13 ± 0.11% (w/w), respectively, compared to 15.67 ± 0.34 (% w/w) and 23.06 ± 0.63 (% w/w) for mPEO-b-PBCL micelles loading individual drugs. Notably, the average diameter of PMs co-encapsulating both SN-38 and A83B4C63 was larger than that of PMs encapsulating either of these compounds alone but still lower than 60 nm. The release of A83B4C63 from PMs co-encapsulating both drugs was 76.36 ± 1.41% within 24 h, which was significantly higher than that of A83B4C63-encapsulated micelles (42.70 ± 0.72%). In contrast, the release of SN-38 from PMs co-encapsulating both drugs was 44.15 ± 2.61% at 24 h, which was significantly lower than that of SN-38-conjugated PMs (74.16 ± 3.65%). Cytotoxicity evaluations by the MTS assay as analyzed by the Combenefit software suggested a clear synergy between PM/A83B4C63 (at a concentration range of 10-40 μM) and free SN-38 (at a concentration range of 0.001-1 μM). The synergistic cytotoxic concentration range for SN-38 was narrowed down to 0.1-1 or 0.01-1 μM when combined with PM/A83B4C63 at 10 or 20-40 μM, respectively. In general, PMs co-encapsulating A83B4C63 and SN-38 at drug concentrations within the synergistic range (10 μM for A83B4C63 and 0.05-1 μM for SN-38) showed slightly less enhancement of SN-38 anticancer activity than a combination of individual micelles, i.e., A83B4C63 PMs + SN-38 PMs at the same molar concentrations. This was attributed to the slower release of SN-38 from the SN-38 and A83B4C63 co-encapsulated PMs compared to PMs only encapsulating SN-38. Cotreatment of cells with TOP1 inhibitors and A83B4C63 formulation enhanced the expression level of γ-HA2X, cleaved PARP, caspase-3, and caspase-7 in most cases. This trend was more consistent and notable for PMs co-encapsulating both A83B4C63 and SN-38. The overall result from the study shows a synergy between PMs of SN-38 and A83B4C63 as a mixture of two PMs for individual drugs or PMs co-encapsulating both drugs.
Collapse
Affiliation(s)
- Sams M A Sadat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Mohammad Reza Vakili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Sara I Abd-El Hafeez
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Marco Paladino
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Dennis G Hall
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Michael Weinfeld
- Department of Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
- Department of Chemical and Material Engineering, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
4
|
Tan B, Lan X, Zhang Y, Liu P, Jin Q, Wang Z, Liang Z, Song W, Xuan Y, Sun Y, Li Y. Effect of 23‑hydroxybetulinic acid on lung adenocarcinoma and its mechanism of action. Exp Ther Med 2024; 27:239. [PMID: 38633355 PMCID: PMC11019653 DOI: 10.3892/etm.2024.12527] [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] [Received: 09/12/2023] [Accepted: 02/12/2024] [Indexed: 04/19/2024] Open
Abstract
The present study aimed to investigate the effect and mechanism of Pulsatilla compounds on lung adenocarcinoma. The representative drug chosen was the compound 23-HBA. GeneCards, Swiss target prediction, DisGeNET and TCMSP were used to screen out related genes, and MTT and flow cytometry assays were used to verify the inhibitory effect of Pulsatilla compounds on the proliferation of lung adenocarcinoma cells. Subsequently, the optimal target, peroxisome proliferator-activated receptor (PPAR)-γ, was selected using bioinformatics analysis, and its properties of low expression in lung adenocarcinoma cells and its role as a tumor suppressor gene were verified by western blot assay. The pathways related to immunity and inflammation, vascular function, cell proliferation, differentiation, development and apoptosis with the highest degree of enrichment and the mechanisms were explored through Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. Finally, the clinical prognosis in terms of the survival rate of patients in whom the drug is acting on the target was analyzed using the GEPIA database. The results indicated that Pulsatilla compounds can inhibit the proliferation of lung adenocarcinoma cells by blocking the cell cycle at the G1 phase. Subsequently, the related PPAR-γ gene was verified as a tumor suppressor gene. Further analysis demonstrated that this finding was related to the PPAR signaling pathway and mitochondrial reactive oxygen species (ROS) production. Finally, the clinical prognosis was found to be improved, as the survival rate of patients was increased. In conclusion, Pulsatilla compounds were indicated to inhibit the viability and proliferation of lung adenocarcinoma H1299 cells, and the mechanism of action was related to PPAR-γ, the PPAR signaling pathway and mitochondrial ROS. The present study provides novel insight to further explore the treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Boyu Tan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xiaoxu Lan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yifan Zhang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Pai Liu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Qiyao Jin
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Zhiqiang Wang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Zhidong Liang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Wei Song
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Ye Xuan
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yunxiao Sun
- Department of Pediatrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| |
Collapse
|
5
|
Dai Y, Qian M, Li Y. Structural Modification Endows Small-Molecular SN38 Derivatives with Multifaceted Functions. Molecules 2023; 28:4931. [PMID: 37446591 DOI: 10.3390/molecules28134931] [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: 05/29/2023] [Revised: 06/21/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
As a camptothecin derivative, 7-ethyl-10-hydroxycamptothecin (SN38) combats cancer by inhibiting topoisomerase I. SN38 is one of the most active compounds among camptothecin derivatives. In addition, SN38 is also a theranostic reagent due to its intrinsic fluorescence. However, the poor water solubility, high systemic toxicity and limited action against drug resistance and metastasis of tumor cells of SN38 indicates that there is great space for the structural modification of SN38. From the perspective of chemical modification, this paper summarizes the progress of SN38 in improving solubility, increasing activity, reducing toxicity and possessing multifunction and analyzes the strategies of structure modification to provide a reference for drug development based on SN38.
Collapse
Affiliation(s)
- Yi Dai
- College of Pharmaceutical Science, Anhui Xinhua University, Hefei 230088, China
- Department of Chemistry, University of Science and Technology of China, Hefei 230026, China
| | - Meng Qian
- College of Pharmaceutical Science, Anhui Xinhua University, Hefei 230088, China
| | - Yan Li
- College of Pharmaceutical Science, Anhui Xinhua University, Hefei 230088, China
| |
Collapse
|
6
|
Wu D, Zhang W, Li Y, Zhao Z, Ji W, Liu H, Yang G. Gold nanorods-loaded chitosan-based nanomedicine platform enabling an effective tumor regression in vivo. Int J Pharm 2023; 632:122561. [PMID: 36586640 DOI: 10.1016/j.ijpharm.2022.122561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/09/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
The clinical utility of 7-ethyl-10-hydroxycamptothecin (SN-38) is hampered by its low water solubility and reduced bioactivity at neutral or alkaline conditions. The rational design of an effective drug delivery system that can significantly enhance the therapeutic index of SN-38 and achieve complete tumor regression still remains a challenge. Herein, chitosan-based hybrid nanoparticles system co-loading with chemotherapeutic drug SN-38 and gold nanorods (AuNRs) was engineered for effective combinational photothermal-chemotherapy. To increase the solubility of SN-38, soluble polymeric prodrug poly (l-glutamic acid)-SN38 (l-PGA-SN38) was firstly synthesized and then complexed with chitosan to form stable nanomedicine via a mild and facile way without using any organic solvent or surfactant. Upon introducing AuNRs into chitosan-based nanomedicine by coordination interaction between the amine group of chitosan and AuNRs, the hybrid nanoparticles exhibited distinct synergistic therapeutic effect compared with single chemotherapy or photothermal treatment in vitro and in vivo. Almost complete tumor regression was achieved after 21-day treatment of the developed hybrid nanoparticles and showed no recurrence for at least 60 days.
Collapse
Affiliation(s)
- Danjun Wu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Wangyang Zhang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yi Li
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zejing Zhao
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Weili Ji
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hong Liu
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gensheng Yang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
7
|
Zhang R, Luo Y, Du C, Wu L, Wang Y, Chen Y, Li S, Jiang X, Xie Y. Synthesis and biological evaluation of novel SN38-glucose conjugate for colorectal cancer treatment. Bioorg Med Chem Lett 2023; 81:129128. [PMID: 36639036 DOI: 10.1016/j.bmcl.2023.129128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
7-Ethyl-10-hydroxycamptothecin (SN38), the bioactive metabolite of irinotecan (CPT-11), has been shown to be 100-1000 times more effective than CPT-11. However, the poor water solubility and bioavailability of SN38 constrained its clinical application. In this study, we synthesized a novel SN38-glucose conjugate (FSY04) to address this issue. Our in vitro studies indicated that FSY04 had a potent inhibitory ability against colorectal cancer (CRC) cell lines of SW-480 and HCT-116 compared to the inhibitory capacity of CPT-11. Interestingly, FSY04 possessed lower cytotoxicity against normal cell lines of LO2 and 293T in contrast with CPT-11. Moreover, FSY04 is more active than CPT-11 in inducing apoptosis, inhibiting migration, and invasion. In vivo experiments suggested that half of the equivalent of FSY04 inhibited the growth of SW480 in the xenograft tumor model better than one equivalent of CPT-11. Our data demonstrated FSY04 to be a promising agent in CRC therapy.
Collapse
Affiliation(s)
- Ruiming Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, PR China
| | - Yi Luo
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Chenghao Du
- Department of Biological Sciences, USC Dana and David Dornsife College of Letters, Arts and Sciences, Los Angeles 90089, USA
| | - Ling Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, PR China
| | - Yankang Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, PR China
| | - Yuanduan Chen
- Guizhou Jinqianguo Biotechnology Co. Ltd., Bijie 551714, PR China
| | - Shouqian Li
- Guizhou Jinqianguo Biotechnology Co. Ltd., Bijie 551714, PR China
| | - Xin Jiang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Yongmei Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, PR China.
| |
Collapse
|
8
|
Anti-tumor effects and mechanism of a novel camptothecin derivative YCJ100. Life Sci 2022; 311:121105. [DOI: 10.1016/j.lfs.2022.121105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/08/2022] [Accepted: 10/16/2022] [Indexed: 11/18/2022]
|
9
|
Li J, Miao P, Guan X, Gao F, Khan AJ, Wang T, Zhang F. Interaction Between 7-Ethyl-10-Hydroxycamptothecin and β-Lactoglobulin Based on Molecular Docking and Molecular Dynamics Simulations. J MACROMOL SCI B 2021. [DOI: 10.1080/00222348.2021.1945080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Jiawei Li
- Pharmacy Laboratory, Inner Mongolia International Mongolian Hospital, Hohhot, China
- Biomedical Nanocenter, School of Life Science, Inner Mongolia Agricultural University, Hohhot, P. R. China
| | - Pandeng Miao
- Pharmacy Laboratory, Inner Mongolia International Mongolian Hospital, Hohhot, China
- Biomedical Nanocenter, School of Life Science, Inner Mongolia Agricultural University, Hohhot, P. R. China
| | - Xiaoying Guan
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital, Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Feng Gao
- Biomedical Nanocenter, School of Life Science, Inner Mongolia Agricultural University, Hohhot, P. R. China
| | - Abdul Jamil Khan
- Biomedical Nanocenter, School of Life Science, Inner Mongolia Agricultural University, Hohhot, P. R. China
| | - Tegexibaiyin Wang
- Pharmacy Laboratory, Inner Mongolia International Mongolian Hospital, Hohhot, China
| | - Feng Zhang
- Pharmacy Laboratory, Inner Mongolia International Mongolian Hospital, Hohhot, China
- Biomedical Nanocenter, School of Life Science, Inner Mongolia Agricultural University, Hohhot, P. R. China
- Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital, Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
He W, Du Y, Wang T, Wang J, Cheng L, Li X. Redox responsive 7-ethyl-10-hydroxycamptothecin (SN38) lysophospholipid conjugate: synthesis, assembly and anticancer evaluation. Int J Pharm 2021; 606:120856. [PMID: 34229071 DOI: 10.1016/j.ijpharm.2021.120856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/01/2021] [Accepted: 07/01/2021] [Indexed: 12/25/2022]
Abstract
7-Ethyl-10-hydroxycamptothecin (SN38), a potent camptothecin derivative specifically targeting DNA topoisomerase I cleavage complexes, has shown great potential in the treatment of solid tumors. Because of its poor solubility and chemical and metabolic stability, the clinical application of SN38 is highly limited. To address these problems, a novel redox-responsive SN38 conjugate based liposomal formulation is developed in this report. First, SN38 was conjugated with lysophospholipid by using a cleavable disulfide bond linker. After that, the conjugate (SN38-SS-PC) was assembled into liposomes by thin film method. Dynamic lightscattering(DLS) characterization indicated that SN38-SS-PC liposomes possessed a narrow size distribution (172.8 ± 10.5 nm) and negative charged zeta potential (-8.9 ± 0.3 mV). The results of storage and physiological stabilities showed that SN38-SS-PC liposomes was stable under different conditions. More importantly, a reduction responsive release of parent drug SN38 was observed in the medium containing glutathione (GSH). In addition, SN38-SS-PC liposomes had a much more rapid cellular uptake behavior against cancer cells. The enhanced anti-cancer efficacy of SN38-SS-PC liposomes was further demonstrated by in vitro cytotoxicity assay against MCF-7 and A549 cells. Under in vivo evaluation in 4 T1 xenograft tumor model, SN38-SS-PC liposomes were observed to have lower systemic toxicity and higher tumor inhibition rate of 53.3% compared with the commercialized SN38 prodrug Irinotecan (Ir). In summary, SN38-SS-PC liposomes could be a promising redox responsive delivery system of SN38 for cancer therapy.
Collapse
Affiliation(s)
- Wei He
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yawei Du
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Tao Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ji Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Lei Cheng
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xinsong Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| |
Collapse
|
11
|
Yang XY, Zhao HY, Lei H, Yuan B, Mao S, Xin M, Zhang SQ. Synthesis and Biological Evaluation of 10-Substituted Camptothecin Derivatives with Improved Water Solubility and Activity. ChemMedChem 2020; 16:1000-1010. [PMID: 33241878 DOI: 10.1002/cmdc.202000753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/27/2020] [Indexed: 11/09/2022]
Abstract
Despite remarkable clinical achievements, camptothecin (CPT) still suffers from poor solubility and severe toxicity. Therefore, it is necessary to redevelop CPT derivatives as supplementary antitumor agents with good water solubility and small side effects. In this work, 27 camptothecin derivatives were synthesized and screened for their cytotoxicity against A549 (lung) and HCT-116 (colon) cancer cell lines. Among them, compound B7, 7-ethyl-10-(2-oxo-2-(4-methylpiperidin-1-yl)ethoxy)camptothecin,was demonstrated in vitro to be a more potent antitumor agent than SN-38 by comparison of their inhibitory activities against cell proliferation and colony formation and interference effect on process of cell cycle and cell apoptosis. Additionally, a molecular docking model revealed that B7 can interact with the topoisomerase I-DNA complex, and that the solubility of B7 reached 5.73 μg/mL in water. Moreover, B7 significantly inhibited tumor growth in an A549 xenograft model at dosages of 0.4 and 2.0 mg/kg, and exhibited minimum lethal doses comparable to those of irinotecan. These results indicated that B7, with improved solubility, enhanced activity and acceptable acute toxicity, can be used as a lead compound for the development of novel anticancer agents.
Collapse
Affiliation(s)
- Xue-Yan Yang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Hong-Yi Zhao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Hao Lei
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Bo Yuan
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Shuai Mao
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - Minghang Xin
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, PR China
| |
Collapse
|
12
|
Fan S, Cao YX, Li GY, Lei H, Attiogbe MKI, Yao JC, Yang XY, Liu YJ, Hei YY, Zhang H, Cao L, Zhang XY, Du SS, Zhang GM, Zhang SQ. F10, a new camptothecin derivative, was identified as a new orally-bioavailable, potent antitumor agent. Eur J Med Chem 2020; 202:112528. [PMID: 32650182 DOI: 10.1016/j.ejmech.2020.112528] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 01/12/2023]
Abstract
Topoisomerases are interesting targets for drug discovery. In the present study, we attached saturated carbon atoms to the 10-position of camptothecin and synthesized 10 new camptothecin derivatives from 10-HCPT or SN-38. The activities of new compounds were evaluated both in vitro and in vivo. The most promising compound F10, 7-ethyl-10-(2-oxo-2-(piperidin-1-yl)ethoxy)camptothecin, inhibited cancer cells growth with the IC50 of 0.002, 0.003, 0.011 and 0.081 μM on Raji, HCT116, A549 and Lovo cells, respectively. Meanwhile, oral administration of F10 remarkably suppressed the HCT116-xenograft tumor growth in the nude-mice model at the dosage of 0.5, 2.0 and 8.0 mg/kg in vivo. Intraperitoneal administration of F10 can completely inhibit Raji-xenograft tumor growth in established NPG mouse model at 2.0 and 4.0 mg/kg. In addition, the minimum lethal doses of F10 and SN-38 in mice by intravenous administration were 80 and 40 mg/kg (or 0.155, 0.102 mmol/kg), respectively. The solubility of F10 reached 9.86 μg/mL in a buffer solution of pH 4.5. The oral bioavailability of F10 achieved 22.4% in mice. The molecular docking model revealed that F10 can interact with topoisomerase I-DNA complex. Our findings indicate that F10 is a new orally-oavailable antitumor agent with potent anticancer effect. Furthermore, attaching a ring hydrophobic moiety to the 10-position of camptothecin provides a favorable approach in the optimization of camptothecin.
Collapse
Affiliation(s)
- Shu Fan
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Yong-Xiao Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Guang-Yan Li
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., LTD, Linyi, Shandong, 276000, PR China
| | - Hao Lei
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Mawusse K I Attiogbe
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Jing-Chun Yao
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., LTD, Linyi, Shandong, 276000, PR China
| | - Xue-Yan Yang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Yan-Jie Liu
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Yuan-Yuan Hei
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Hao Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., LTD, Linyi, Shandong, 276000, PR China
| | - Lei Cao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China.
| | - Xiao-Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Shuai-Shuai Du
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China
| | - Gui-Min Zhang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., LTD, Linyi, Shandong, 276000, PR China
| | - San-Qi Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, PR China.
| |
Collapse
|
13
|
Naumczuk B, Bocian W, Sitkowski J, Kawęcki R, Kozerski L. Solvent-dependent regioselectivity of 2′-deoxyadenosine alkylation by 9-aminomethyl derivatives of SN38. NEW J CHEM 2019. [DOI: 10.1039/c9nj04828f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The investigated compounds in DMSO solution unexpectedly alkylate the N9 nitrogen of 2′-deoxyadenosine, whereas the N6 nitrogen atom is alkylated in water solution.
Collapse
Affiliation(s)
- B. Naumczuk
- National Medicines Institute
- Chełmska 30/34
- Poland
- Institute of Organic Chemistry
- Polish Academy of Sciences
| | - W. Bocian
- National Medicines Institute
- Chełmska 30/34
- Poland
| | - J. Sitkowski
- National Medicines Institute
- Chełmska 30/34
- Poland
- Institute of Organic Chemistry
- Polish Academy of Sciences
| | - R. Kawęcki
- University of Natural Sciences and Humanities
- Faculty of Science
- 3 Maja 54
- Poland
| | - L. Kozerski
- National Medicines Institute
- Chełmska 30/34
- Poland
- Institute of Organic Chemistry
- Polish Academy of Sciences
| |
Collapse
|