1
|
Xu X, Zhang J, Wang T, Li J, Rong Y, Wang Y, Bai C, Yan Q, Ran X, Wang Y, Zhang T, Sun J, Jiang Q. Emerging non-antibody‒drug conjugates (non-ADCs) therapeutics of toxins for cancer treatment. Acta Pharm Sin B 2024; 14:1542-1559. [PMID: 38572098 PMCID: PMC10985036 DOI: 10.1016/j.apsb.2023.11.029] [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: 09/04/2023] [Revised: 10/31/2023] [Accepted: 11/23/2023] [Indexed: 04/05/2024] Open
Abstract
The non-selective cytotoxicity of toxins limits the clinical relevance of the toxins. In recent years, toxins have been widely used as warheads for antibody‒drug conjugates (ADCs) due to their efficient killing activity against various cancer cells. Although ADCs confer certain targeting properties to the toxins, low drug loading capacity, possible immunogenicity, and other drawbacks also limit the potential application of ADCs. Recently, non-ADC delivery strategies for toxins have been extensively investigated. To further understand the application of toxins in anti-tumor, this paper provided an overview of prodrugs, nanodrug delivery systems, and biomimetic drug delivery systems. In addition, toxins and their combination strategies with other therapies were discussed. Finally, the prospect and challenge of toxins in cancer treatment were also summarized.
Collapse
Affiliation(s)
- Xiaolan Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiaming Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tao Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jing Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yukang Rong
- School of Education, University of Nottingham, Nottingham NG7 2RD, UK
| | - Yanfang Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chenxia Bai
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qing Yan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaohua Ran
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yingli Wang
- Department of Pharmacy, Linyi People's Hospital, Shandong University, Linyi 276000, China
| | - Tianhong Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qikun Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| |
Collapse
|
2
|
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
|
3
|
Dai Y, Zhang Y, Ye T, Chen Y. Synthesis and Antitumor Evaluation of Biotin-SN38-Valproic Acid Conjugates. Molecules 2023; 28:molecules28093936. [PMID: 37175346 PMCID: PMC10179906 DOI: 10.3390/molecules28093936] [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: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Despite the strong anticancer activity of SN38 (7-ethyl-10-hydroxy-camptothecin), the severe side effects and loss of anticancer activity caused by the lack of selectivity to cancer cells and hydrolysis of ring E prevent its clinical application. To address the issue, herein a multifunctional SN38 derivative (compound 9) containing biotin (tumor-targeting group) and valproic acid (histone deacetylase inhibitor, HDACi) was synthesized via click chemistry and evaluated using MTT assay. The in vitro cytotoxicity study showed that compound 9 exhibited superior cytotoxicity than irinotecan against human cervical cancer HeLa cells, albeit it was inferior to SN38. More significantly, compound 9 significantly reduced toxicity in mouse embryonic fibroblast NIH3T3 cells, indicating that compound 9 had the capacity to enhance tumor targeting due to its cell selectivity. Further studies demonstrated that, compared with irinotecan, compound 9 induced similar apoptosis of cancer cells. Consequently, compound 9 can not only improve its tumor-targeting ability mediated by biotin but also exert potent anticancer activity through the effect of SN38 and valproic acid, indicating that the design concept is an effective strategy for the structural modification of SN38.
Collapse
Affiliation(s)
- Yi Dai
- College of Pharmaceutical Science, Anhui Xinhua University, Hefei 230088, China
| | - Yang Zhang
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230031, China
| | - Tianxiang Ye
- College of Pharmaceutical Science, Anhui Xinhua University, Hefei 230088, China
| | - Yue Chen
- College of Pharmaceutical Science, Anhui Xinhua University, Hefei 230088, China
| |
Collapse
|
4
|
Bhosale M, Jeelani I, Nawaz A, Abe H, Padhye S. Site-Specific Binding of Anticancer Drugs to Human Serum Albumin. Anticancer Agents Med Chem 2022; 22:2876-2884. [PMID: 35331098 DOI: 10.2174/1871520622666220324094033] [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: 08/10/2021] [Revised: 11/15/2021] [Accepted: 01/13/2022] [Indexed: 11/22/2022]
Abstract
The interaction of drugs with proteins plays a very important role in the distribution of the drug. Human serum albumin (HSA) is the most abundant protein in the human body and showing great binding characteristics has gained a lot of importance pharmaceutically. It plays an essential role in the pharmacokinetics of a number of drugs and hence several reports are available on the interaction of drugs with HSA. It can bind to cancer drugs and thus it is crucial to look at the binding characteristics of these drugs with HSA. Herein we summarize the binding properties of some anti-cancer drugs by specifically looking into the binding site with HSA. The number of drugs binding at Sudlow's site I situated in subdomain II A is more than the drugs binding at Sudlow's site II.
Collapse
Affiliation(s)
- Mrinalini Bhosale
- Department of Chemistry, Abeda Inamdar Senior College of Arts, Science and Commerce, Savitribai Phule Pune University, Pune 411001, India
| | - Ishtiaq Jeelani
- Graduate School of Innovative Life Science, University of Toyama, Toyama, 3190 Gofuku 930-8555, Japan
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, 2630 Sugitani 930-0194, Japan
| | - Allah Nawaz
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, 2630 Sugitani 930-0194, Japan
| | - Hitoshi Abe
- Faculty of Engineering, University of Toyama, Toyama, 3190 Gofuku 930-8555, Japan
| | - Subhash Padhye
- Department of Chemistry, Abeda Inamdar Senior College of Arts, Science and Commerce, Savitribai Phule Pune University, Pune 411001, India
| |
Collapse
|
5
|
Hu Y, Yu D, Zhang X. 9-amino acid cyclic peptide-decorated sorafenib polymeric nanoparticles for the efficient in vitro nursing care analysis of hepatocellular carcinoma. Process Biochem 2021. [DOI: 10.1016/j.procbio.2020.09.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
6
|
Bai YH, Yun XJ, Xue Y, Zhou T, Sun X, Gao YJ. A novel oncolytic adenovirus inhibits hepatocellular carcinoma growth. J Zhejiang Univ Sci B 2020; 20:1003-1013. [PMID: 31749347 DOI: 10.1631/jzus.b1900089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To evaluate the inhibitory role of a novel oncolytic adenovirus (OA), GP73-SphK1sR-Ad5, on the growth of hepatocellular carcinoma (HCC). METHODS GP73-SphK1sR-Ad5 was constructed by integrating Golgi protein 73 (GP73) promoter and sphingosine kinase 1 (SphK1)-short hairpin RNA (shRNA) into adenovirus serotype 5 (Ad5), and transfecting into HCC Huh7 cells and normal human liver HL-7702 cells. The expression of SphK1 and adenovirus early region 1 (E1A) was detected by quantitative real-time PCR (qRT-PCR) and western blot, respectively. Cell viability was detected by methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay, and apoptotic rate was determined by flow cytometry. An Huh7 xenograft model was established in mice injected intratumorally with GP73-SphK1sR-Ad5. Twenty days after injection, the tumor volume and weight, and the survival time of the mice were recorded. The histopathological changes in tumor tissues were observed by hematoxylin-eosin (HE) staining and transmission electron microscopy (TEM). RESULTS Transfection of GP73-SphK1sR-Ad5 significantly upregulated E1A and downregulated SphK1 in Huh7 cells, but not in HL7702 cells. GP73-SphK1sR-Ad5 transfection significantly decreased the viability and increased the apoptotic rate of Huh7 cells, but had no effect on HL7702 cells. Intratumoral injection of GP73-SphK1sR-Ad5 into the Huh7 xenograft mouse model significantly decreased tumor volume and weight, and prolonged survival time. It also significantly decreased the tumor infiltration area and blood vessel density, and increased the percentages of cells with nucleus deformation and cells with condensed chromatin in tumor tissues. CONCLUSIONS GP73-SphK1sR-Ad5 serves as a novel OA and can inhibit HCC progression with high specificity and efficacy.
Collapse
Affiliation(s)
- Yu-Huan Bai
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China.,Department of Gastroenterology, the Second People's Hospital of Liaocheng, Linqing 252600, China
| | - Xiao-Jing Yun
- Department of Gastroenterology, the Second People's Hospital of Liaocheng, Linqing 252600, China
| | - Yan Xue
- Department of Gastroenterology, the Second People's Hospital of Liaocheng, Linqing 252600, China
| | - Ting Zhou
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xin Sun
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Yan-Jing Gao
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
7
|
Fan Q, Cui X, Guo H, Xu Y, Zhang G, Peng B. Application of rare earth-doped nanoparticles in biological imaging and tumor treatment. J Biomater Appl 2020; 35:237-263. [DOI: 10.1177/0885328220924540] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Rare earth-doped nanoparticles have been widely used in disease diagnosis, drug delivery, tumor therapy, and bioimaging. Among various bioimaging methods, the fluorescence imaging technology based on the rare earth-doped nanoparticles can visually display the cell activity and lesion evolution in living animals, which is a powerful tool in biological technology and has being widely applied in medical and biological fields. Especially in the band of near infrared (700–1700 nm), the emissions show the characteristics of deep penetration due to low absorption, low photon scattering, and low autofluorescence interference. Furthermore, the rare earth-doped nanoparticles can be endowed with the water solubility, biocompatibility, drug-loading ability, and the targeting ability for different tumors by surface functionalization. This confirms its potential in the cancer diagnosis and treatment. In this review, we summarized the recent progress in the application of rare earth-doped nanoparticles in the field of bioimaging and tumor treatment. The luminescent mechanism, properties, and structure design were also discussed.
Collapse
Affiliation(s)
- Qi Fan
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- University of Chinese Academy of Sciences (UCAS), Beijing, PR China
| | - Xiaoxia Cui
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Haitao Guo
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Yantao Xu
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Guangwei Zhang
- Zhejiang Fountain Aptitude Technology Inc., Hangzhou, Zhejiang, PR China
| | - Bo Peng
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
8
|
Zhao CK, Xu L, Wang XH, Bao YJ, Wang Y. Synthesis of Dual Target CPT-Ala-Nor Conjugates and Their Biological Activity Evaluation. Anticancer Agents Med Chem 2020; 19:502-508. [PMID: 30663574 DOI: 10.2174/1871520619666190121121933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/18/2018] [Accepted: 01/09/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Target therapy has been one of the important strategies in new drug discovery and the resulting drug resistance has also been a serious problem for concern. At the same time, there are several cancer genes or pathways operating within a given cancer. Given these two things, the combination therapy will be needed for optimal therapeutic effect. OBJECTIVE Camptothecin and norcantharidin were thus chosen to construct a dual anticancer drugs assemblies mainly because CPT was the DNA-topoisomerase I inhibitor and norcantharidin could also suppress the cancer cell growth by inhibiting protein phosphatase. The designed conjugate of camptothecin and norcantharidin linked by alanine was expected to have dual target drug properties. METHODS EDCI/DMAP was chosen as a coupling agent for the coupling of CPT with substituted norcantharidin derivatives and CCK-8 method was used to test the cytotoxicity and intensity on human hepatoma cell line HepG2. Two kinds of enzymes, Top I and CDC 25B were selected to screen the binding affinity in molecular level. RESULTS Nine of dual targets camptothecin derivatives were smoothly synthesized by twice coupling in the condition of EDCI/DMAP in moderate yield. All of the synthesized compounds were characterized by 1HNMR and 13CNMR spectrum and exhibited strong potent inhibition against Hep G2, SW480, BGC803, and PANC-1 cell line in vitro. The newly synthesized camptothecin compounds, such as 3j and 3i have strengthened inhibition activity compared to camptothecin and norcantharidin. CONCLUSION We have successfully synthesized a series of novel camptothecin derivatives constructed from three components of camptothecin, alanine and norcantharidin. These compounds not only preserved strong activity against several cancer cell lines in vitro, but also exhibited potential binding affinity to target Top I and CDC 25B. Therefore, these conjugates linked by alanine could suppress cancer cell growth by inhibiting Top I and protein phosphatase simultaneously, which makes it much valuable as a novel bi-functional target drug candidate to develop in vivo.
Collapse
Affiliation(s)
- Chang K Zhao
- School of Pharmacy, Zunyi Medical University, No.6 Xue Fu West Road, Zunyi City, 563003, China
| | - Lang Xu
- School of Pharmacy, Zunyi Medical University, No.6 Xue Fu West Road, Zunyi City, 563003, China
| | - Xian H Wang
- School of Pharmacy, Zunyi Medical University, No.6 Xue Fu West Road, Zunyi City, 563003, China
| | - Yu J Bao
- School of Pharmacy, Zunyi Medical University, No.6 Xue Fu West Road, Zunyi City, 563003, China
| | - Yuhe Wang
- Department of Pharmacy, Zunyi Medical University Affiliated Hospital, 149 Dalian Road, Zunyi, China
| |
Collapse
|
9
|
Zou J, Chen S, Li Y, Zeng L, Lian G, Li J, Chen S, Huang K, Chen Y. Nanoparticles modified by triple single chain antibodies for MRI examination and targeted therapy in pancreatic cancer. NANOSCALE 2020; 12:4473-4490. [PMID: 32031201 DOI: 10.1039/c9nr04976b] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
UNLABELLED Precise diagnosis and effective treatment are crucial to the prognosis of pancreatic ductal adenocarcinoma (PDAC). Magnetic iron oxide nanoparticles (IONPs) are superior magnetic resonance imaging (MRI) contrast agents, while antibodies are significant immunotherapy reagents. Herein, we firstly generated a novel nanocomposite combining triple single chain antibodies (scAbs) and IONPs for the detection and treatment of PDAC. METHODS Triple scAbs (scAbMUC4, scAbCEACAM6, scFvCD44v6, MCC triple scAbs) were conjugated to the surface of polyethylene glycol modified IONPs (IONPs-PEG), forming the IONPs-PEG-MCC triple scAbs nanocomposite. Characterization of the nanocomposite was performed, and its cytotoxicity, specificity, and apoptosis induction were evaluated. In vivo MRI study and anti-pancreatic cancer effect assessment were performed in tumor-bearing nude mice. RESULTS The size of the IONPs-PEG-MCC triple scAbs nanocomposite was about 23.6 nm. The nanocomposite was non-toxic to normal pancreatic ductal epithelial cells, and could specifically bind to and be internalized by MUC4/CEACAM6/CD44v6-expressing PDAC cells. With an r2 relaxivity of 104.2 mM-1 s-1, the IONPs-PEG-MCC triple scAbs nanocomposite could significantly shorten the MRI T2-weighted signal intensity both in vitro and in vivo. The IONPs-PEG-MCC triple scAbs nanocomposite also showed a favorable anti-pancreatic cancer effect. CONCLUSION In the present study, the IONPs-PEG-MCC triple scAbs nanocomposite was firstly confirmed as a bi-functional nanocomposite in both MRI and treatment, providing its critical clinical transformation potential in PDAC detection and treatment.
Collapse
Affiliation(s)
- Jinmao Zou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120 China.
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Cheng G, Zhang X, Chen Y, Lee RJ, Wang J, Yao J, Zhang Y, Zhang C, Wang K, Yu B. Anticancer activity of polymeric nanoparticles containing linoleic acid-SN38 (LA-SN38) conjugate in a murine model of colorectal cancer. Colloids Surf B Biointerfaces 2019; 181:822-829. [DOI: 10.1016/j.colsurfb.2019.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/09/2019] [Accepted: 06/10/2019] [Indexed: 01/26/2023]
|
11
|
Hydrophobic drug self-delivery systems as a versatile nanoplatform for cancer therapy: A review. Colloids Surf B Biointerfaces 2019; 180:202-211. [DOI: 10.1016/j.colsurfb.2019.04.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/24/2022]
|
12
|
Abdelmoneem MA, Mahmoud M, Zaky A, Helmy MW, Sallam M, Fang JY, Elkhodairy KA, Elzoghby AO. Decorating protein nanospheres with lactoferrin enhances oral COX-2 inhibitor/herbal therapy of hepatocellular carcinoma. Nanomedicine (Lond) 2018; 13:2377-2395. [DOI: 10.2217/nnm-2018-0134] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Lactoferrin (LF)-targeted gliadin nanoparticles (GL-NPs) were developed for targeted oral therapy of hepatocellular carcinoma. Materials & methods: Celecoxib and diosmin were incorporated in the hydrophobic matrix of GL-NPs whose surface was decorated with LF by electrostatic interaction for binding to asialoglycoprotein receptors overexpressed by liver cancer cells. Results: Targeted GL-NPs showed enhanced cytotoxic activity and increased cellular uptake in liver tumor cells compared with nontargeted NPs. Moreover, they demonstrated superior in vivo antitumor effects including reduction in the expression levels of tumor biomarkers and induction of caspase-mediated apoptosis. Ex vivo imaging of isolated organs exhibited extensive accumulation of NPs in livers more than other organs. Conclusion: LF-targeted GL-NPs could be considered as an efficient nanoplatform for targeted oral drug delivery for liver cancer therapy.
Collapse
Affiliation(s)
- Mona A Abdelmoneem
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mazen Mahmoud
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Amira Zaky
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21511, Egypt
| | - Maged W Helmy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Damanhur University, Damanhur 22511, Egypt
| | - Marwa Sallam
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan 333, Taiwan
- Research Center for Industry of Human Ecology & Research Center for Chinese Herbal Medicine, Chang Gung University of Science & Technology, Kweishan, Taoyuan 333, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan 333, Taiwan
| | - Kadria A Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed O Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Division of Engineering in Medicine, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technologies, Cambridge, MA 02139, USA
| |
Collapse
|
13
|
Wang XH, Huang M, Zhao CK, Li C, Xu L. Design, synthesis, and biological activity evaluation of campthothecin-HAA-Norcantharidin conjugates as antitumor agents in vitro. Chem Biol Drug Des 2018; 93:986-992. [PMID: 30218487 DOI: 10.1111/cbdd.13397] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/21/2018] [Accepted: 08/08/2018] [Indexed: 11/30/2022]
Abstract
Three components of Camptothecin, hydroxyacetic acid, and functionalized norcantharidins were constructed together to form a novel series of camptothecin derivatives in a good yield. The synthesized campthothecin-HAA-norcantharidin conjugate pro-drugs could suppress cancer cell growth in vitro. These conjugated pro-drug molecules possess therapeutic potential as novel bi-functional conjugates platforms for cancer treatment.
Collapse
Affiliation(s)
- Xian H Wang
- School of Pharmacy, Zunyi Medical University, Zunyi City, Guizhou Province, China
| | - Mei Huang
- Department of Pharmacology and Key Laboratory of Basic Pharmacology of Ministry of Education, Zunyi Medical University, Zunyi City, Guizhou Province, China
| | - Chang K Zhao
- School of Pharmacy, Zunyi Medical University, Zunyi City, Guizhou Province, China
| | - Chan Li
- School of Pharmacy, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Lang Xu
- School of Pharmacy, Zunyi Medical University, Zunyi City, Guizhou Province, China
| |
Collapse
|
14
|
Song Z, Chen X, You X, Huang K, Dhinakar A, Gu Z, Wu J. Self-assembly of peptide amphiphiles for drug delivery: the role of peptide primary and secondary structures. Biomater Sci 2018; 5:2369-2380. [PMID: 29051950 DOI: 10.1039/c7bm00730b] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Peptide amphiphiles (PAs), functionalized with alkyl chains, are capable of self-assembling into various nanostructures. Recently, PAs have been considered as ideal drug carriers due to their good biocompatibility, specific biological functions, and hypotoxicity to normal cells and tissues. Meanwhile, the nanocarriers formed by PAs are able to achieve controlled drug release and enhanced cell uptake in response to the stimulus of the physiological environment or specific biological factors in the location of the lesion. However, the underlying detailed drug delivery mechanism, especially from the aspect of primary and secondary structures of PAs, has not been systematically summarized or discussed. Focusing on the relationship between the primary and secondary structures of PAs and stimuli-responsive drug delivery applications, this review highlights the recent advances, challenges, and opportunities of PA-based functional drug nanocarriers, and their potential pharmaceutical applications are discussed.
Collapse
Affiliation(s)
- Zhenhua Song
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, School of Engineering, Sun Yat-sen University, Guangzhou, 510006, PR China.
| | | | | | | | | | | | | |
Collapse
|
15
|
Zhao CK, Li C, Wang XH, Bao YJ, Yang FH, Huang M. The regio-selective synthesis of 10-hydroxy camptothecin norcantharidin conjugates and their biological activity evaluation in vitro. ROYAL SOCIETY OPEN SCIENCE 2018; 5:172317. [PMID: 30110459 PMCID: PMC6030274 DOI: 10.1098/rsos.172317] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/02/2018] [Indexed: 06/08/2023]
Abstract
A series of conjugates of 10-hydroxy camptothecin (HCPT) with functionalized norcantharidin derivatives were regio-selectively synthesized in the condition of (3-dimethylaminopropyl) ethyl-carbodiimide monohydrochloride in a moderate yield. The synthesized conjugate HCPT pro-drugs can also suppress cancer cell growth in vitro. These conjugated pro-drug constructs possess therapeutic potential as novel bi-functional conjugate platforms for cancer treatment.
Collapse
Affiliation(s)
- Chang K. Zhao
- School of Pharmacy, Zunyi Medical University, No. 6 Xue Fu West Road, Xin Pu New District, Zunyi City, 563003, Guizhou Province, People's Republic of China
| | - Chan Li
- School of Pharmacy, Sun Yat-Sen University, 135 Xin Gangxi Road, Haizhu District, Guangzhou 563003, Guangdong Province, People's Republic of China
| | - Xian H. Wang
- School of Pharmacy, Zunyi Medical University, No. 6 Xue Fu West Road, Xin Pu New District, Zunyi City, 563003, Guizhou Province, People's Republic of China
| | - Yu J. Bao
- School of Pharmacy, Zunyi Medical University, No. 6 Xue Fu West Road, Xin Pu New District, Zunyi City, 563003, Guizhou Province, People's Republic of China
| | - Fu H. Yang
- School of Pharmacy, Zunyi Medical University, No. 6 Xue Fu West Road, Xin Pu New District, Zunyi City, 563003, Guizhou Province, People's Republic of China
| | - Mei Huang
- School of Pharmacy, Zunyi Medical University, No. 6 Xue Fu West Road, Xin Pu New District, Zunyi City, 563003, Guizhou Province, People's Republic of China
| |
Collapse
|
16
|
Wang H, Wu J, Xu L, Xie K, Chen C, Dong Y. Albumin nanoparticle encapsulation of potent cytotoxic therapeutics shows sustained drug release and alleviates cancer drug toxicity. Chem Commun (Camb) 2018; 53:2618-2621. [PMID: 28195282 DOI: 10.1039/c6cc08978j] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We here provide the first report on the construction of nanoparticles formulating highly potent cytotoxic therapeutics using albumin. Maytansinoid DM1 can be efficiently integrated into albumin nanoparticles, resulting in remarkable alleviation of in vivo drug toxicity and expanding the repertoire of albumin technology available for cancer therapy.
Collapse
Affiliation(s)
- Hangxiang Wang
- The First Affiliated Hospital, School of Medicine; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310003, P. R. China.
| | - Jiaping Wu
- The First Affiliated Hospital, School of Medicine; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310003, P. R. China.
| | - Li Xu
- The First Affiliated Hospital, School of Medicine; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310003, P. R. China.
| | - Ke Xie
- The First Affiliated Hospital, School of Medicine; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University, Hangzhou, 310003, P. R. China.
| | - Chao Chen
- College of Life Sciences, Huzhou University, Huzhou, 313000, P. R. China
| | - Yuehan Dong
- Department of Applied Engineering, Zhejiang Economic and Trade Polytechnic, Hangzhou, 310018, P. R. China.
| |
Collapse
|
17
|
Xu L, Xu S, Wang H, Zhang J, Chen Z, Pan L, Wang J, Wei X, Xie H, Zhou L, Zheng S, Xu X. Enhancing the Efficacy and Safety of Doxorubicin against Hepatocellular Carcinoma through a Modular Assembly Approach: The Combination of Polymeric Prodrug Design, Nanoparticle Encapsulation, and Cancer Cell-Specific Drug Targeting. ACS APPLIED MATERIALS & INTERFACES 2018; 10:3229-3240. [PMID: 29313660 DOI: 10.1021/acsami.7b14496] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Intervention is urgently required to improve the therapeutic outcome for patients with unresectable hepatocellular carcinomas (HCCs). However, current chemotherapeutics, such as sorafenib and doxorubicin (DOX), provide only limited therapeutic benefits for this devastating disease. In this context, we present a modular assembly approach to the construction of a systemically injectable nanotherapeutic that can efficiently and safely deliver DOX in vivo. To achieve this goal, we covalently attached DOX to a polylactide (PLA) building block (Mw = 2800, n = 36), yielding DOX-PLA conjugate 1. Due to the lipophilicity imparted by PLA, the conjugate 1 coassembled with an amphiphilic lipid, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethylene glycol) 2000] (DSPE-PEG2000), to form nanoparticles (NPs). To achieve preferential tumor accumulation, we additionally decorated the particle surface with an HCC-specific peptide moiety (i.e., SP94). The resulting HCC-targetable DOX-encapsulating NPs (termed tNP-PLA-DOX) exhibited several unique characteristics, including the feasible fabrication of sub-100 nm NPs, substantially delayed drug release profiles of several weeks, HCC cell-specific uptake and tumor accumulation in an in vivo mouse model, as well as alleviated drug toxicity in animals. Collectively, these results show that the integration of multiple components within a single nanocarrier via modular assembly is cost-effective for the creation of safe anticancer nanotherapeutics. The presented DOX-based nanomedicines have potential for enhancing the therapeutic index in patients.
Collapse
Affiliation(s)
- Li Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Shengjun Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Hangxiang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Jun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Zun Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Linhui Pan
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Haiyang Xie
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Lin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Shusen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University , Hangzhou 310003, P. R. China
| |
Collapse
|
18
|
Wang H, Lu Z, Wang L, Guo T, Wu J, Wan J, Zhou L, Li H, Li Z, Jiang D, Song P, Xie H, Zhou L, Xu X, Zheng S. New Generation Nanomedicines Constructed from Self-Assembling Small-Molecule Prodrugs Alleviate Cancer Drug Toxicity. Cancer Res 2017; 77:6963-6974. [PMID: 29055017 DOI: 10.1158/0008-5472.can-17-0984] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/21/2017] [Accepted: 10/13/2017] [Indexed: 11/16/2022]
Abstract
The therapeutic index for chemotherapeutic drugs is determined in part by systemic toxicity, so strategies for dose intensification to improve efficacy must also address tolerability. In addressing this issue, we have investigated a novel combinatorial strategy of reconstructing a drug molecule and using sequential drug-induced nanoassembly to fabricate supramolecular nanomedicines (SNM). Using cabazitaxel as a target agent, we established that individual synthetic prodrugs tethered with polyunsaturated fatty acids were capable of recapitulating self-assembly behavior independent of exogenous excipients. The resulting SNM could be further refined by PEGylation with amphiphilic copolymers suitable for preclinical studies. Among these cabazitaxel derivatives, docosahexaenoic acid-derived compound 1 retained high antiproliferative activity. SNM assembled with compound 1 displayed an unexpected enhancement of tolerability in animals along with effective therapeutic efficacy in a mouse xenograft model of human cancer, compared with free drug administered in its clinical formulation. Overall, our studies showed how attaching flexible lipid chains to a hydrophobic and highly toxic anticancer drug can convert it to a systemic self-deliverable nanotherapy, preserving its pharmacologic efficacy while improving its safety profile. Cancer Res; 77(24); 6963-74. ©2017 AACR.
Collapse
Affiliation(s)
- Hangxiang Wang
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China.
| | - Zhongjie Lu
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Lijiang Wang
- Zhejiang-California International Nanosystems Institute, Zhejiang University, Hangzhou, P.R. China
| | - Tingting Guo
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Jiaping Wu
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Jianqin Wan
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Liqian Zhou
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Hui Li
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Zhen Li
- School of Pharmacy, Dalian Medical University, Dalian, P.R. China
| | - Donghai Jiang
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Penghong Song
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Haiyang Xie
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Lin Zhou
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Xiao Xu
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China
| | - Shusen Zheng
- The First Affiliated Hospital; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, P.R. China.
| |
Collapse
|
19
|
Yang Y, Wang X, Liao G, Liu X, Chen Q, Li H, Lu L, Zhao P, Yu Z. iRGD-decorated red shift emissive carbon nanodots for tumor targeting fluorescence imaging. J Colloid Interface Sci 2017; 509:515-521. [PMID: 28923749 DOI: 10.1016/j.jcis.2017.09.007] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/05/2017] [Accepted: 09/05/2017] [Indexed: 10/18/2022]
Abstract
Carbon nanodots (CDs) have been exhibiting increasing applications owing to their luminescence properties and biocompatibility as imaging probes in diagnosis. However, poor tumor targeting and penetration of CDs is still the biggest challenge limiting their tumor imaging efficacy. To improve the tumor targeting and penetration efficiency of CDs, we developed an active tumor targeting imaging system by simply fabricating a tumor-homing penetration peptide iRGD (CRGDKGPDC) to red shift emissive CDs (iRGD-CDs) with a physical method. Particularly, iRGD-CDs showed a small size and red shift fluorescence signals as CDs, which made iRGD-CDs suitable for in vivo fluorescence imaging. iRGD-CDs showed higher cellular uptake in vitro, while presented higher penetration and accumulation in tumor tissue in vivo, leading to better tumor imaging efficacy. In conclusion, decoration with iRGD could significantly increase the permeability of CDs in tumor vessels and tumor tissue, generating more CDs leaking out from tumor vasculature, consequently improving the sensitivity of tumor imaging.
Collapse
Affiliation(s)
- Yuanyuan Yang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Xuefeng Wang
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China
| | - Guochao Liao
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China
| | - Xiqiang Liu
- Guangdong Provincial Key Laboratory of Stomatology, Department of Oral and Maxillofacial Surgery, Guanghua School and Hospital of Stomatology, Sun Yat-Sen University, Guangzhou 510055, PR China
| | - Qiling Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Hongmei Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Ling Lu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Peng Zhao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China.
| | - Zhiqiang Yu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
20
|
Wang H, Chen J, Xu C, Shi L, Tayier M, Zhou J, Zhang J, Wu J, Ye Z, Fang T, Han W. Cancer Nanomedicines Stabilized by π-π Stacking between Heterodimeric Prodrugs Enable Exceptionally High Drug Loading Capacity and Safer Delivery of Drug Combinations. Am J Cancer Res 2017; 7:3638-3652. [PMID: 29109766 PMCID: PMC5667338 DOI: 10.7150/thno.20028] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 07/18/2017] [Indexed: 02/05/2023] Open
Abstract
Combination therapy using distinct mode-of-action drugs has sparked a rapidly growing interest because this paradigm holds promise for improving the therapeutic efficacy of anticancer therapy. However, the current drug combination therapy refers to administering individual drugs together, which is far from a perfect regimen for cancer patients. The aim of this work was to demonstrate that synergistic delivery of two chemotherapeutic drugs in a single nanoparticle reservoir could be achieved through the rational chemical ligation of the drugs followed by supramolecular nano-assembly via blending of the drugs with a minimal amount of matrix. Choosing 7-ethyl-10-hydroxycamptothecin and taxanes, which are rich in aromatic structures, as model compounds, we show that the heterodimeric conjugates of the two agents are miscible with lipids to form systemically injectable nanomedicines. The compatibility between the prodrug conjugates and lipid carriers is substantially augmented by the intermolecular π-π stacking and alleviated polarity, thus enabling an exceptionally high drug loading (DL) capacity (~92%) and a gratifyingly long drug retention time within the micellar core. We further observed superior therapeutic outcomes in a mouse tumor model without detecting accompanying systemic toxicity. This structure-based, self-assembled cancer nanomedicine increased the potency and drug tolerability in animals and thus offers a robust strategy for simultaneously formulating two or more drugs in single nanovehicles.
Collapse
|
21
|
Wang H, Wu J, Xie K, Fang T, Chen C, Xie H, Zhou L, Zheng S. Precise Engineering of Prodrug Cocktails into Single Polymeric Nanoparticles for Combination Cancer Therapy: Extended and Sequentially Controllable Drug Release. ACS APPLIED MATERIALS & INTERFACES 2017; 9:10567-10576. [PMID: 28271714 DOI: 10.1021/acsami.7b01938] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The synergistic combination of two or more chemotherapeutics frequently requires packaging in single delivery vehicles for the sequential release of each substance in a predictable manner. Here, we demonstrate for the first time that the rational engineering of a prodrug cocktail into single polymeric nanoparticles (NPs) can enable the sequential release of chemotherapeutics in a controllable manner. Exploiting combretastatin-A4 (CA4, 1) as a model antiangiogenesis agent, two ester derivatives, 2 and 3, tethered with saturated fatty acids (butanoic and heptanoic acid for 2 and 3, respectively) were synthesized. 7-Ethyl-10-hydroxycamptothecin (SN38) derivative 4, esterified with α-linolenic acid, was used as a cytotoxic drug. Because of their augmented lipophilicity and miscibility, all constructed prodrugs readily assembled with clinically approved polymeric matrices. Results showed that altering the aliphatic chains of modifiers for CA4 chemical derivatization enabled the drug retention capacity within particle systems to be adjusted, leading to the identification of the prodrug cocktail of 2 and 4 as an optimal combination for subsequent preclinical studies. Furthermore, in vivo assessements indicated that the resulting NPs simultaneously formulating 2 and 4 exhibited synergistic activities and outperformed NPs loaded with individual prodrugs 2 or 4 in terms of therapeutic efficacy. These findings highlight a novel and versatile strategy for tailoring chemically disparate prodrug cocktails for adaptation within a single nanoplatform as a potential modality for synergistic cancer therapy.
Collapse
Affiliation(s)
- Hangxiang Wang
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Jiaping Wu
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Ke Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Tao Fang
- Jinhua People's Hospital , Jinhua, Zhejiang Province 321000, P.R. China
| | - Chao Chen
- College of Life Sciences, Huzhou University , Huzhou, Zhejiang Province 313000, P.R. China
| | - Haiyang Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Lin Zhou
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Shusen Zheng
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| |
Collapse
|
22
|
Yuan Y, Xu L, Dai S, Wang M, Wang H. A facile supramolecular approach to fabricate multifunctional upconversion nanoparticles as a versatile platform for drug loading, in vivo delivery and tumor imaging. J Mater Chem B 2017; 5:2425-2435. [PMID: 32264550 DOI: 10.1039/c6tb03381d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multifunctional upconversion nanoparticles (UCNPs) that can be implemented in theranostic applications are particularly attractive scaffolds for precise drug delivery. However, most of the current methods for drug formulation are technically complicated, thereby impeding their use in the clinic. Here, we report on the preparation of a lipophilic cytotoxic prodrug-integrated and polyethylene glycol (PEG)-cloaked UCNPs scaffold through a facile one-pot supramolecular approach. By choosing 7-ethyl-10-hydroxycamptothecin (SN38)-derived prodrug 1 as a model chemotherapeutic, we show that this lipophilic prodrug can be feasibly self-assembled onto the surface of UCNPs, which are cooperatively solubilized by PEGylated phospholipids. The resulting SN38 prodrug 1-encapsulated UCNPs (designated 1@pUCNPs) produce a stable colloidal system in aqueous solution, making it suitable for intravenous injection. The SN38 drug loading capacity in pUCNPs is as high as ∼12.3 wt%, and a sustained drug release profile is observed, indicating that the drug payloads can be transported to targeted tumor sites via the enhanced permeability and retention (EPR) effect. Upconversion luminescence (UCL) imaging, including in vivo and ex vivo imaging, suggests that the drug-loaded pUCNPs remain stable in tumors over a long time and preferentially accumulate in tumors presumably via the EPR effect. Furthermore, the 1@pUCNPs show superior therapeutic outcomes compared with the clinically approved SN38 prodrug CPT-11 in the Bcap-37 mouse model of breast cancer. Collectively, our results demonstrate that pUCNPs facilely constructed in a one-pot self-assembly manner may be used as a versatile platform, enabling synchronous in vivo delivery of poorly water-soluble drugs and tumor imaging.
Collapse
Affiliation(s)
- Yingying Yuan
- The First Affiliated Hospital, School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University, Hangzhou 310003, P. R. China.
| | | | | | | | | |
Collapse
|
23
|
Chen C, Ni S, Zheng Q, Yu M, Wang H. Synthesis, Structure, Biological Evaluation, and Catalysis of Two Pyrazole-Functionalized NHC-RuIIComplexes. Eur J Inorg Chem 2017. [DOI: 10.1002/ejic.201601116] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Chao Chen
- College of Life Sciences; Huzhou University; East 2nd Road 313000 Huzhou China
| | - Shengliang Ni
- College of Life Sciences; Huzhou University; East 2nd Road 313000 Huzhou China
| | - Qing Zheng
- College of Life Sciences; Huzhou University; East 2nd Road 313000 Huzhou China
| | - Meifang Yu
- College of Life Sciences; Huzhou University; East 2nd Road 313000 Huzhou China
| | - Hangxiang Wang
- The First Affiliated Hospital; School of Medicine; Zhejiang University; 310003 Hangzhou China
| |
Collapse
|
24
|
Abet V, Filace F, Recio J, Alvarez-Builla J, Burgos C. Prodrug approach: An overview of recent cases. Eur J Med Chem 2016; 127:810-827. [PMID: 27823878 DOI: 10.1016/j.ejmech.2016.10.061] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 10/26/2016] [Accepted: 10/27/2016] [Indexed: 01/01/2023]
Abstract
In this review we highlight the most modern trends in the prodrug strategy. In drug research and development, the prodrug concept has found a number of useful applications. Selected examples of this approach are provided in this paper and they are classified according to the aim of their design.
Collapse
Affiliation(s)
- Valentina Abet
- Departamento de Química Orgánica, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain
| | - Fabiana Filace
- Departamento de Química Orgánica, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain
| | - Javier Recio
- Departamento de Química Orgánica, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain
| | - Julio Alvarez-Builla
- Departamento de Química Orgánica, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain.
| | - Carolina Burgos
- Departamento de Química Orgánica, Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
25
|
Integrating a novel SN38 prodrug into the PEGylated liposomal system as a robust platform for efficient cancer therapy in solid tumors. Int J Pharm 2016; 512:39-48. [PMID: 27544846 DOI: 10.1016/j.ijpharm.2016.08.036] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/26/2016] [Accepted: 08/17/2016] [Indexed: 12/20/2022]
Abstract
Liposomal nanoassemblies have been used extensively as carriers for the delivery of both lipophilic and hydrophilic drugs. They represent a mature, versatile technology with considerable potential for improving the pharmacokinetics of drugs. However, the formulation of many chemotherapeutics into liposome systems has posed a significant challenge due to their incompatible physicochemical properties, as was the case with camptothecin-based chemotherapeutics. Here, we present a rational paradigm of potent chemotherapeutics that were reconstructed and subsequently integrated into liposomal nanoassemblies. Using SN38 (7-ethyl-10-hydroxy camptothecin) as a model drug, a lipophilic prodrug 1 (designated as LA-SN38) was constructed by tethering the linoleic acid (LA) moiety via esterification, which was further facilitated to form liposomal nanoparticles (LipoNP) through supramolecular nanoassembly. The resulting 1-loaded LipoNP exhibited sustained drug release kinetics and decreased cellular uptake by macrophage cells. Uptake by tumor cells was enhanced relative to our previous supramolecular nanoparticles (SNP 1), which were derived from the self-assembling prodrug 1. Notably, LipoNP outperformed SNP 1 in terms of pharmacokinetics and in vivo therapeutic efficacy in both human BEL-7402 hepatocellular carcinoma (HCC) and HCT-116 colorectal cancer-derived xenograft mouse models. These results were likely due to the improved systemic circulation and preferential accumulation of nanodrugs in tumors. Hence, our results suggest that the combination of liposomal delivery platforms with rational prodrug engineering may emerge as a promising approach for the effective and safe delivery of anticancer chemotherapeutics.
Collapse
|
26
|
Wang J, Wang H, Li J, Liu Z, Xie H, Wei X, Lu D, Zhuang R, Xu X, Zheng S. iRGD-Decorated Polymeric Nanoparticles for the Efficient Delivery of Vandetanib to Hepatocellular Carcinoma: Preparation and in Vitro and in Vivo Evaluation. ACS APPLIED MATERIALS & INTERFACES 2016; 8:19228-37. [PMID: 27381493 DOI: 10.1021/acsami.6b03166] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Molecularly targeted agents that are designed to target specific lesions have been proven effective as clinical cancer therapies; however, most currently available therapeutic agents are poorly water-soluble and require oral administration, thereby resulting in low bioavailability and a high risk of side effects due to dose intensification. The rational engineering of systemically injectable medicines that encapsulate such therapeutic payloads may revolutionize anticancer therapies and remains an under-explored area of drug development. Here, the injectable delivery of a nanomedicine complexed with an oral multitargeted kinase inhibitor, vandetanib (vanib), was explored using polymeric nanoparticles (NPs) to achieve the selective accumulation of drug payloads within tumor lesions. To demonstrate this concept, we used biodegradable amphiphilic block copolymer poly(ethylene glycol)-block-poly(D, L-lactic acid) (PEG-PLA) to nanoprecipitate this potent agent to form water-soluble NPs that are suitable for intravenous administration. NP-vanib induced cytotoxic activity by inhibiting the angiogenetic events mediated by VEGFR and EGFR kinases in tested cancer cells and inhibited the growth, tube formation and metastasis of HUVECs. The intravenously injection of NP-vanib into mice bearing HCC BEL-7402 xenografts more effectively inhibited the tumor than the oral administration of vanib. In addition, due to the modular design of these NPs, the drug-loaded particles can easily be decorated with iRGD, a tumor-homing and -penetrating peptide motif, which further improved the in vivo performance of these vanib-loaded NPs. Our results demonstrate that reformulating targeted therapeutic agents in NPs permits their systemic administration and thus significantly improves the potency of currently available, orally delivered agents.
Collapse
Affiliation(s)
- Jianguo Wang
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| | - Hangxiang Wang
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| | - Jie Li
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| | - Zhikun Liu
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| | - Haiyang Xie
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| | - Xuyong Wei
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| | - Di Lu
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| | - Runzhou Zhuang
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| | - Xiao Xu
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| | - Shusen Zheng
- The First Affiliated Hospital, School of Medicine, Zhejiang University , Hangzhou, 310003, PR China
| |
Collapse
|