1
|
Mitra A, Roy R, Paul S. Modulating the Self-Assembly of a Camptothecin Prodrug with Paclitaxel for Anticancer Combination Therapy: A Molecular Dynamics Approach. J Phys Chem B 2024; 128:10799-10812. [PMID: 39230512 DOI: 10.1021/acs.jpcb.4c04798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Camptothecin (CPT) and paclitaxel (PTX), derived from natural products, are recognized for their significant efficacy in clinical cancer treatments. Despite its therapeutic advantages, CPT is challenged by issues of toxicity and solubility, necessitating its use in conjugation with other compounds for enhanced compatibility. This study delves into the coassembly mechanism of Evans blue-conjugated camptothecin (EB-CPT) with PTX, aiming to elucidate their synergistic potential in combination therapy applications, employing all-atom molecular dynamics simulations. The EB-CPT prodrug is reported to form a self-aggregated cluster. Our findings suggest that increasing the PTX concentration induces a dispersion of EB-CPT clusters, thereby disrupting their inherent self-assembly. This disruption is explained to be facilitated by the coassembly of EB-CPT and PTX. With increasing concentration of PTX, a lengthening of the coassembled structures is observed, supporting the experimental findings of tube-like coassembled structures at higher weight ratios of PTX. Hydrophobic interactions and π-π stacking are the primary forces responsible for the formation of both self- and coassembled structures. Interestingly, the structural analysis reveals that the CPT moiety of EB-CPT is less involved in assemblies due to steric hindrances. Instead, the interaction and coassembly processes are predominantly mediated by the EB derivative component of the prodrug. This research underscores the critical role of the solubilizing agent, EB derivative, in mediating the flexibility and interaction of CPT in combination therapy strategies, particularly with PTX, thus emphasizing the importance of conjugates for therapeutic developments. Furthermore, the molecular insights into the interaction sites and mechanisms facilitating coassembly between EB-CPT and PTX contribute valuable knowledge to the field, highlighting the potential of these nanomedicine combinations in advancing cancer treatment modalities.
Collapse
Affiliation(s)
- Anandita Mitra
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| | - Rituparna Roy
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| | - Sandip Paul
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| |
Collapse
|
2
|
Li Z, Tan W, Li X, Wang Y, Dang Z, Zhang Z, Guan S, Zhu S, Li F, Zhang M. Unlocking lysosomal acidity to activate membranolytic module for accurately cancer theranostics. Bioorg Chem 2024; 153:107764. [PMID: 39232344 DOI: 10.1016/j.bioorg.2024.107764] [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: 02/17/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Chemotherapy drug efflux, toxic side effects, and low efficacy against drug-resistant cells have plagued safe and efficient cancer theranostics. However, the materials or methods that resolve these defects all-in-one are scarce. Here, a new cancer theranostics strategy is proposed by utilizing changes in lysosomal acidity in cancer cells to activate the membranolytic model to overcome these obstacles together. Therefore, a simple fluorescent anthracene derivative Lyso-Mito is developed, which has a perfect pKa (4.62) value that falls between the pH of lysosomes in cancer and normal cells. Lyso-Mito itself can precisely target and convert the pH perturbation of lysosomes in cancer cells to fluorescent response and membranolytic module activity to accomplish the low drug efflux, weak toxic side effects, and low drug-resistant cancer diagnosis and treatment without linking other functional units or any additional assistance. Hereby, a new cancer theranostics strategy of integrating organelle microenvironment and the membranolytic model is realized.
Collapse
Affiliation(s)
- Zhuo Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Wenjia Tan
- China-Japan Union Hospital of Jilin University, Changchun 130041, China
| | - Xinru Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - YaJun Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zetao Dang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zhaoxia Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Shuwen Guan
- College of Life Science, Jilin University, Changchun 130012, China
| | - Shoujun Zhu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Feng Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| | - Ming Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| |
Collapse
|
3
|
Li J, Wang X, Zhang H, Hu X, Peng X, Jiang W, Zhuo L, Peng Y, Zeng G, Wang Z. Fenamates: Forgotten treasure for cancer treatment and prevention: Mechanisms of action, structural modification, and bright future. Med Res Rev 2024. [PMID: 39171404 DOI: 10.1002/med.22079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 08/03/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024]
Abstract
Fenamates as classical nonsteroidal anti-inflammatory agents are widely used for relieving pain. Preclinical studies and epidemiological data highlight their chemo-preventive and chemotherapeutic potential for cancer. However, comprehensive reviews of fenamates in cancer are limited. To accelerate the repurposing of fenamates, this review summarizes the results of fenamates alone or in combination with existing chemotherapeutic agents. This paper also explores targets of fenamates in cancer therapy, including COX, AKR family, AR, gap junction, FTO, TEAD, DHODH, TAS2R14, ion channels, and DNA. Besides, this paper discusses other mechanisms, such as regulating Wnt/β-catenin, TGF-β, p38 MAPK, and NF-κB pathway, and the regulation of the expressions of Sp, EGR-1, NAG-1, ATF-3, ErbB2, AR, as well as the modulation of the tumor immune microenvironment. Furthermore, this paper outlined the structural modifications of fenamates, highlighting their potential as promising leads for anticancer drugs.
Collapse
Affiliation(s)
- Junfang Li
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaodong Wang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Honghua Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoling Hu
- School of Pharmacy, Lanzhou University, Lanzhou, Gansu, China
| | - Xue Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weifan Jiang
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Linsheng Zhuo
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yan Peng
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Guo Zeng
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zhen Wang
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Postdoctoral Station for Basic Medicine, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
4
|
Geng WC, Jiang ZT, Chen SL, Guo DS. Supramolecular interaction in the action of drug delivery systems. Chem Sci 2024; 15:7811-7823. [PMID: 38817563 PMCID: PMC11134347 DOI: 10.1039/d3sc04585d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/27/2024] [Indexed: 06/01/2024] Open
Abstract
Complex diseases and diverse clinical needs necessitate drug delivery systems (DDSs), yet the current performance of DDSs is far from ideal. Supramolecular interactions play a pivotal role in various aspects of drug delivery, encompassing biocompatibility, drug loading, stability, crossing biological barriers, targeting, and controlled release. Nevertheless, despite having some understanding of the role of supramolecular interactions in drug delivery, their incorporation is frequently overlooked in the design and development of DDSs. This perspective provides a brief analysis of the involved supramolecular interactions in the action of drug delivery, with a primary emphasis on the DDSs employed in the clinic, mainly liposomes and polymers, and recognized phenomena in research, such as the protein corona. The supramolecular interactions implicated in various aspects of drug delivery systems, including biocompatibility, drug loading, stability, spatiotemporal distribution, and controlled release, were individually analyzed and discussed. This perspective aims to trigger a comprehensive and systematic consideration of supramolecular interactions in the further development of DDSs. Supramolecular interactions embody the true essence of the interplay between the majority of DDSs and biological systems.
Collapse
Affiliation(s)
- Wen-Chao Geng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Ze-Tao Jiang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Shi-Lin Chen
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| |
Collapse
|
5
|
Yang C, Ding Y, Mao Z, Wang W. Nanoplatform-Mediated Autophagy Regulation and Combined Anti-Tumor Therapy for Resistant Tumors. Int J Nanomedicine 2024; 19:917-944. [PMID: 38293604 PMCID: PMC10826716 DOI: 10.2147/ijn.s445578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024] Open
Abstract
The overall cancer incidence and death toll have been increasing worldwide. However, the conventional therapies have some obvious limitations, such as non-specific targeting, systemic toxic effects, especially the multidrug resistance (MDR) of tumors, in which, autophagy plays a vital role. Therefore, there is an urgent need for new treatments to reduce adverse reactions, improve the treatment efficacy and expand their therapeutic indications more effectively and accurately. Combination therapy based on autophagy regulators is a very feasible and important method to overcome tumor resistance and sensitize anti-tumor drugs. However, the less improved efficacy, more systemic toxicity and other problems limit its clinical application. Nanotechnology provides a good way to overcome this limitation. Co-delivery of autophagy regulators combined with anti-tumor drugs through nanoplatforms provides a good therapeutic strategy for the treatment of tumors, especially drug-resistant tumors. Notably, the nanomaterials with autophagy regulatory properties have broad therapeutic prospects as carrier platforms, especially in adjuvant therapy. However, further research is still necessary to overcome the difficulties such as the safety, biocompatibility, and side effects of nanomedicine. In addition, clinical research is also indispensable to confirm its application in tumor treatment.
Collapse
Affiliation(s)
- Caixia Yang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
6
|
Komedchikova EN, Kolesnikova OA, Syuy AV, Volkov VS, Deyev SM, Nikitin MP, Shipunova VO. Targosomes: Anti-HER2 PLGA nanocarriers for bioimaging, chemotherapy and local photothermal treatment of tumors and remote metastases. J Control Release 2024; 365:317-330. [PMID: 37996056 DOI: 10.1016/j.jconrel.2023.11.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 11/25/2023]
Abstract
Developing combined cancer therapy strategies is of utmost importance as it can enhance treatment efficacy, overcome drug resistance, and ultimately improve patient outcomes by targeting multiple pathways and mechanisms involved in cancer growth and progression. Specifically, the potential of developing a combination chemo&photothermal therapy using targeted polymer nanoparticles as nanocarriers offers a promising approach for synergistic cancer treatment by combining the benefits of both therapies, such as targeted drug delivery and localized hyperthermia. Here, we report the first targeted anti-HER2 PLGA nanocarriers, called targosomes, that simultaneously possess photothermal, chemotherapeutic and diagnostic properties using only molecular payloads. Biocompatible poly(lactic-co-glycolic acid), PLGA, nanoparticles were loaded with photosensitizer phthalocyanine, diagnostic dye Nile Blue, and chemotherapeutic drug irinotecan, which was chosen as a result of screening a panel of theragnostic nanoparticles. The targeted delivery to cell surface oncomarker HER2 was ensured by nanoparticle modification with the anti-HER2 monoclonal antibody, trastuzumab, using the one-pot synthesis method without chemical conjugation. The irradiation tests revealed prominent photothermal properties of nanoparticles, namely heating by 35 °C in 10 min. Nanoparticles exhibited a 7-fold increase in binding and nearly an 18-fold increase in cytotoxicity for HER2-overexpressing cells compared to cells lacking HER2 expression. This enhancement of cytotoxicity was further amplified by >20-fold under NIR light irradiation. In vivo studies proved the efficacy of nanoparticles for bioimaging of primary tumor and metastasis sites and demonstrated 93% tumor growth inhibition, making these nanoparticles excellent candidates for translation into theragnostic applications.
Collapse
Affiliation(s)
- E N Komedchikova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - O A Kolesnikova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - A V Syuy
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - V S Volkov
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - S M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - M P Nikitin
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - V O Shipunova
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; Nanobiomedicine Division, Sirius University of Science and Technology, 354340 Sochi, Russia.
| |
Collapse
|
7
|
Qin M, Xia H, Xu W, Chen B, Wang Y. The spatiotemporal journey of nanomedicines in solid tumors on their therapeutic efficacy. Adv Drug Deliv Rev 2023; 203:115137. [PMID: 37949414 DOI: 10.1016/j.addr.2023.115137] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
The rapid development of nanomedicines is revolutionizing the landscape of cancer treatment, while effectively delivering them into solid tumors remains a formidable challenge. Currently, there is a huge disconnect on therapeutic response between regulatory approved nanomedicines and laboratory reported nanoparticles. The discrepancy is mainly resulted from the failure of using the classic overall pharmacokinetics behaviors of nanomedicines in tumors to predict the antitumor efficacy. Increasing evidence has revealed that the therapeutic efficacy predominantly relies on the intratumoral spatiotemporal distribution of nanomedicines. This review focuses on the spatiotemporal distribution of systemically administered chemotherapeutic nanomedicines in solid tumor. Firstly, the intratumoral biological barriers that regulate the spatiotemporal distribution of nanomedicines are described in detail. Next, the influences on antitumor efficacy caused by the spatial distribution and temporal drug release of nanomedicines are emphatically analyzed. Then, current methodologies for evaluating the spatiotemporal distribution of nanomedicines are summarized. Finally, the advanced strategies to positively modulate the spatiotemporal distribution of nanomedicines for an optimal tumor therapy are comprehensively reviewed.
Collapse
Affiliation(s)
- Mengmeng Qin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wenhao Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; Chemical Biology Center, Peking University, Beijing, China.
| |
Collapse
|
8
|
Wang W, Zhang Y, Wang Z, Liu X, Lu S, Hu X. A Native Drug-Free Macromolecular Therapeutic to Trigger Mutual Reinforcing of Endoplasmic Reticulum Stress and Mitochondrial Dysfunction for Cancer Treatment. ACS NANO 2023. [PMID: 37257082 DOI: 10.1021/acsnano.3c03450] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Drug-free macromolecular therapeutics are promising alternatives to traditional drugs. Nanomedicines with multiple organelles targeting can potentially increase the efficacy. Herein, a drug-free macromolecular therapeutic was designed to formulate endoplasmic reticulum (ER) and mitochondria dual-targeting nanoparticles (EMT-NPs), which can synergistically elicit ER stress and mitochondrial dysfunction. In vitro experiments indicated that EMT-NPs could effectively enter ER and mitochondria at an approximate ratio of 2 to 3. Subsequently, EMT-NPs could upregulate ER stress-related protein expression (IRE1α, CHOP), boosting calcium ion (Ca2+) efflux and activating the caspase-12 signaling cascade in cancer cells. In addition, EMT-NPs induced direct oxidative stress in mitochondria; some mitochondrial-related apoptotic events such as decreased mitochondrial membrane potential (MMP), upregulation of Bax, cytochrome c release, and caspase-3 activation were also observed for tumor cells upon incubation with EMT-NPs. Furthermore, the leaked Ca2+ from ER could induce mitochondrial Ca2+ overloading to further augment cancer cell apoptosis. In brief, mitochondrial and ER signaling networks collaborated well to promote cancer cell death. Extended photoacoustic and fluorescence imaging served well for the treatment of in vivo patient-derived xenografts cancer model. This drug-free macromolecular strategy with multiple subcellular targeting provides a potential paradigm for cancer theranostics in precision nanomedicine.
Collapse
Affiliation(s)
- Wenhui Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science & Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yongteng Zhang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Zeshu Wang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Xueping Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science & Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Siyu Lu
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450000, China
| | - Xianglong Hu
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, Anhui, China
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| |
Collapse
|
9
|
Yu L, Xie L, Chen Z, Guo H, Zhang Y, Wang H, Wang R, Zhou X, Lei Z, Lu D. Target Embolization Combined with Multimodal Thermal Ablation for Solid Tumors by Smart Poly(amino acid)s Nanocomposites. ACS Biomater Sci Eng 2023; 9:2683-2693. [PMID: 37083337 DOI: 10.1021/acsbiomaterials.2c01274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Noninterventional embolization does not require the use of a catheter, and the treatment of solid tumors in combination with thermal ablation can avoid some of the risks of the surgical procedure. Therefore, we developed an efficient tumor microenvironment-gelled nanocomposites with poly [(l-glutamic acid-ran-l-tyrosine)-b-l-serine-b-l-cysteine] (PGTSCs) coated-nanoparticles (Fe3O4&Au@PGTSCs), from which the prepared PGTSCs were given possession of pH response to an acidic tumor microenvironment. Fe3O4&Au@PGTSC in noninterventional embolization treatment not only achieved the smart targeted medicine delivery but also meshed with noninvasive multimodal thermal ablation therapy and multimodal imaging of solid tumors via intravenous injection. It was worth noting that the results of animal experiments in vivo demonstrated that Fe3O4&Au@PGTSCs have specific tumor accumulation and embolization and thermal ablation effects; at 10 days postinjection, only scars were found at the tumor site. After 20 days, the tumors of model mice completely disappeared. This device is easier to treat solid tumors based on the slightly acidic tumor environment.
Collapse
Affiliation(s)
- Lili Yu
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China
| | - Liyuan Xie
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China
| | - Zhengpeng Chen
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China
| | - Hongyun Guo
- Gansu Provincial Academic for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou 730070, P. R. China
| | - Yongdong Zhang
- Gansu Provincial Academic for Medical Research, Gansu Provincial Cancer Hospital, Lanzhou 730070, P. R. China
| | - Haijun Wang
- Gansu Provincial Hospital, Lanzhou 730000, P. R. China
| | - Rong Wang
- Gansu Provincial Hospital, Lanzhou 730000, P. R. China
| | - Xing Zhou
- Gansu Provincial Hospital, Lanzhou 730000, P. R. China
| | - Ziqiang Lei
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China
| | - Dedai Lu
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou 730070, P. R. China
| |
Collapse
|
10
|
Wang X, Xiong H, Zhang P, Liu Y, Gao C, Zhou Z, Sun J, Diao M. Intratympanic microcrystals of dexamethasone and lipoic acid for the treatment of cisplatin-induced inner ear injury. Colloids Surf B Biointerfaces 2023; 223:113191. [PMID: 36739674 DOI: 10.1016/j.colsurfb.2023.113191] [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: 12/04/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Steroids (anti-inflammatory drugs) combined with antioxidants are frequently prescribed to treat cisplatin (CP)-induced hearing loss in the clinic. Compared to systemic administration of free drugs, local drug delivery systems offer better therapeutic qualities and patient compliance since they not only can bypass the blood-labyrinth barrier but also can perform sustained release. In this work, dexamethasone (DEX) and lipoic acid (LA) non-spherical microcrystals (MCs) were prepared without complicated chemical modification. Following a series of physical characterizations, including morphology, stability and injectability, dissolution and round window membrane distribution of MCs, DEX MCs, LA MCs and the simple mixture of DEX MCs + LA MCs (combination group) were administered in guinea pigs by intratympanic injection. We found that LA MCs enabled improvement of DEX absorption in the combination group compared to a single dose. In addition, no significant morphological changes or inflammatory responses were observed in cochlear tissue, indicating good biocompatibility. Finally, the combination group also demonstrated synergistic therapeutic effect for protecting hair cells against CP-induced damage. The local co delivery of DEX MCs and LA MCs offers a new strategy for the treatment of CP-induced inner ear injury since they provide sustained anti-inflammatory and antioxidant effects simultaneously.
Collapse
Affiliation(s)
- Xiangxiang Wang
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Haixia Xiong
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Peili Zhang
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Ya Liu
- Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; National Clinical Medical Research Center for Otolaryngology Diseases, Beijing 100048, China
| | - Chang Gao
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Zhimin Zhou
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Jianjun Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Peking University International Hospital, Beijing 102206, China.
| | - Mingfang Diao
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; National Clinical Medical Research Center for Otolaryngology Diseases, Beijing 100048, China.
| |
Collapse
|
11
|
Emerging potential of 5-Fluorouracil-loaded chitosan nanoparticles in cancer therapy. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
12
|
Kumar N, Oqmhula K, Hongo K, Takagi K, Yusa SI, Rajan R, Matsumura K. Mechanistic insights and importance of hydrophobicity in cationic polymers for cancer therapy. J Mater Chem B 2023; 11:1456-1468. [PMID: 36661268 DOI: 10.1039/d2tb02059a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Development of molecules that can be effectively used for killing cancer cells remains a research topic of interest in drug discovery. However, various limitations of small molecules and nanotechnology-based drug-delivery systems hinder the development of chemotherapeutics. To resolve this issue, this study describes the potential application of polymeric molecules as anticancer drug candidates. We describe the design and synthesis of novel anticancer polymers containing hydrophobic groups. We established the fact that the cationic homopolymer (PAMPTMA) does not show any anticancer activity on its own; however, the insertion of hydrophobic moieties in copolymers (PAMPTMA-r-BuMA, PAMPTMA-r-HexMA, and PAMPTMA-r-OctMA) enhances their anticancer activity with a very low IC50 value (60 μg mL-1 for HepG2 cells). Mechanistic investigations were carried out using LDH leakage assay, cellular uptake, DOSY NMR and molecular dynamics to study the interaction between the polymer and the cell membrane as well as the role of hydrophobicity in enhancing this interaction. The results demonstrated that polymers are attracted by the anionic cancer cell membrane, which then leads to the insertion of hydrophobic groups inside the cell membrane, causing its disruption and ultimate lysis of the cell. This study demonstrates a novel and better approach for the rational design and discovery of new polymeric anticancer agents with improved efficacy.
Collapse
Affiliation(s)
- Nishant Kumar
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| | - Kenji Oqmhula
- School of Information Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Kenta Hongo
- Research Center for Advanced Computing Infrastructure, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Kengo Takagi
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji, Hyogo 671-2280, Japan
| | - Shin-Ichi Yusa
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji, Hyogo 671-2280, Japan
| | - Robin Rajan
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| | - Kazuaki Matsumura
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| |
Collapse
|
13
|
Detappe A, Nguyen HVT, Jiang Y, Agius MP, Wang W, Mathieu C, Su NK, Kristufek SL, Lundberg DJ, Bhagchandani S, Ghobrial IM, Ghoroghchian PP, Johnson JA. Molecular bottlebrush prodrugs as mono- and triplex combination therapies for multiple myeloma. NATURE NANOTECHNOLOGY 2023; 18:184-192. [PMID: 36702954 PMCID: PMC10032145 DOI: 10.1038/s41565-022-01310-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 12/06/2022] [Indexed: 05/19/2023]
Abstract
Cancer therapies often have narrow therapeutic indexes and involve potentially suboptimal combinations due to the dissimilar physical properties of drug molecules. Nanomedicine platforms could address these challenges, but it remains unclear whether synergistic free-drug ratios translate to nanocarriers and whether nanocarriers with multiple drugs outperform mixtures of single-drug nanocarriers at the same dose. Here we report a bottlebrush prodrug (BPD) platform designed to answer these questions in the context of multiple myeloma therapy. We show that proteasome inhibitor (bortezomib)-based BPD monotherapy slows tumour progression in vivo and that mixtures of bortezomib, pomalidomide and dexamethasone BPDs exhibit in vitro synergistic, additive or antagonistic patterns distinct from their corresponding free-drug counterparts. BPDs carrying a statistical mixture of three drugs in a synergistic ratio outperform the free-drug combination at the same ratio as well as a mixture of single-drug BPDs in the same ratio. Our results address unanswered questions in the field of nanomedicine, offering design principles for combination nanomedicines and strategies for improving current front-line monotherapies and combination therapies for multiple myeloma.
Collapse
Affiliation(s)
- Alexandre Detappe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Hung V-T Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Window Therapeutics, Boston, MA, USA
| | - Yivan Jiang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Window Therapeutics, Boston, MA, USA
| | - Michael P Agius
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Wencong Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Clelia Mathieu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nang K Su
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Samantha L Kristufek
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David J Lundberg
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sachin Bhagchandani
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - P Peter Ghoroghchian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
14
|
Wu YY, Liao WH, Niu ZL, Zhou SH, Wu TT, Li Z, Zhao QH, Xu JY, Xie MJ. Gallium Metal-Organic Nanoparticles with Albumin-Stabilized and Loaded Graphene for Enhanced Delivery to HCT116 Cells. Int J Nanomedicine 2023; 18:225-241. [PMID: 36660337 PMCID: PMC9844232 DOI: 10.2147/ijn.s386253] [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: 08/25/2022] [Accepted: 12/12/2022] [Indexed: 01/15/2023] Open
Abstract
Background Gallium (III) metal-organic complexes have been shown to have the ability to inhibit tumor growth, but the poor water solubility of many of the complexes precludes further application. The use of materials with high biocompatibility as drug delivery carriers for metal-organic complexes to enhance the bioavailability of the drug is a feasible approach. Methods Here, we modified the ligands of gallium 8-hydroxyquinolinate complex with good clinical anticancer activity by replacing the 8-hydroxyquinoline ligands with 5-bromo-8-hydroxyquinoline (HBrQ), and the resulting Ga(III) + HBrQ complex had poor water solubility. Two biocompatible materials, bovine serum albumin (BSA) and graphene oxide (GO), were used to synthesize the corresponding Ga(III) + HBrQ complex nanoparticles (NPs) BSA/Ga/HBrQ NPs and GO/Ga/HBrQ NPs in different ways to enhance the drug delivery of the metal complex. Results Both of BSA/Ga/HBrQ NPs and GO/Ga/HBrQ NPs can maintain stable existence in different solution states. In vitro cytotoxicity test showed that two nanomedicines had excellent anti-proliferation effect on HCT116 cells, which shown higher level of intracellular ROS and apoptosis ratio than that of cisplatin and oxaliplatin. In addition, the superior emissive properties of BSA/Ga/HBrQ NPs and GO/Ga/HBrQ NPs allow their use for in vivo imaging showing highly effective therapy in HCT116 tumor-bearing mouse models. Conclusion The use of biocompatible materials for the preparation of NPs against poorly biocompatible metal-organic complexes to construct drug delivery systems is a promising strategy that can further improve drug delivery and therapeutic efficacy.
Collapse
Affiliation(s)
- Yuan-yuan Wu
- School of Chemical Science and Technology, Yunnan University, Kunming, People’s Republic of China
| | - Wen-Hui Liao
- School of Chemical Science and Technology, Yunnan University, Kunming, People’s Republic of China
| | - Zong-ling Niu
- School of Chemical Science and Technology, Yunnan University, Kunming, People’s Republic of China
| | - Si-Han Zhou
- School of Chemical Science and Technology, Yunnan University, Kunming, People’s Republic of China
| | - Tian-Tian Wu
- School of Chemical Science and Technology, Yunnan University, Kunming, People’s Republic of China
| | - Zhe Li
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Qi-Hua Zhao
- School of Chemical Science and Technology, Yunnan University, Kunming, People’s Republic of China
| | - Jing-Yuan Xu
- Department of Chemical Biology and Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Ming-jin Xie
- School of Chemical Science and Technology, Yunnan University, Kunming, People’s Republic of China,Correspondence: Ming-jin Xie, Email
| |
Collapse
|
15
|
Yin M, Xu X, Han H, Dai J, Sun R, Yang L, Xie J, Wang Y. Preparation of triangular silver nanoparticles and their biological effects in the treatment of ovarian cancer. J Ovarian Res 2022; 15:121. [PMID: 36411490 PMCID: PMC9680130 DOI: 10.1186/s13048-022-01056-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 10/26/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND In recent years, silver nanoparticles (AgNPs) have gradually been widely used, especially in the field of anticancer medicine. Ovarian cancer (OC) is the gynaecological malignancy with the highest mortality rate, and the current treatment is still based on surgery, chemotherapy and postoperative targeted therapy. Therefore, the development of safe and effective nanoparticles for targeted therapy of OC is very important. This study aimed to prepare a new type of triangular silver nanoparticles (tAgNPs) and evaluate the anticancer properties for OC in vitro and in vivo. METHODS The tAgNPs were chemically synthesized and characterized using scanning electron microscopy (SEM), ultraviolet (UV) spectrophotometry and other techniques. By performing cell-based tests, such as cell counting kit-8 (CCK-8), plate colony formation, cell apoptosis, reactive oxygen species (ROS), and western blot (WB) assays, the inhibitory effects and related mechanisms of tAgNPs on OC cells were analysed.The anticancer effect of tAgNPs in vivo was verified by a SKOV3 tumor-bearing mouse model. RESULTS Five types of tAgNPs with different colours were successfully synthesized, with a particle size of 25-50 nm and a good dispersion. The results of in vitro experiments showed that tAgNPs treatment reduced the viability and proliferation of SKOV3 cells, arrested the cell cycle in G0/G1 phase, inhibited the expression levels of proliferation-related factors and cyclins, and promoted cell apoptosis by producing ROS and increasing caspase-3 activity. Consistent with the results of in vitro experiments, in vivo animal experiments also showed that tAgNPs significantly inhibited the proliferation of ovarian cancer. More importantly, no obvious toxic and side effects were observed. CONCLUSIONS In this study, a novel triangular AgNPs was successfully prepared. tAgNPs are very stable, significantly inhibit the proliferation of OC cells and tumour growth in tumour-bearing mice, providing a promising nanotargeted therapy for OC.
Collapse
Affiliation(s)
- Man Yin
- grid.449428.70000 0004 1797 7280Department of Clinical Medicine, Jining Medical University, Jining, 272000 Shandong China
| | - Xiangyu Xu
- grid.449428.70000 0004 1797 7280Laboratory of New Antitumor Drug Molecular Design & Synthesis, College of Basic Medical, Jining Medical University, Jining, 272067 Shandong Province China
| | - Hui Han
- grid.452252.60000 0004 8342 692XDepartment of Gynecology, Affiliated Hospital of Jining Medical University, Gu Huai Road, No.89 Jining, 272029 Shandong China
| | - Jiahui Dai
- grid.449428.70000 0004 1797 7280Department of Clinical Medicine, Jining Medical University, Jining, 272000 Shandong China
| | - Ronghe Sun
- grid.449428.70000 0004 1797 7280Department of Clinical Medicine, Jining Medical University, Jining, 272000 Shandong China
| | - Linqing Yang
- grid.452252.60000 0004 8342 692XDepartment of Gynecology, Affiliated Hospital of Jining Medical University, Gu Huai Road, No.89 Jining, 272029 Shandong China
| | - Junyu Xie
- grid.449428.70000 0004 1797 7280Department of Clinical Medicine, Jining Medical University, Jining, 272000 Shandong China
| | - Yunfei Wang
- grid.452252.60000 0004 8342 692XDepartment of Gynecology, Affiliated Hospital of Jining Medical University, Gu Huai Road, No.89 Jining, 272029 Shandong China
| |
Collapse
|
16
|
Sun S, Yang Y, Gao Z, Jiang H, Ye L, Lai Y, Shen Z, Wu ZS. Endogenous Stimuli-Responsive Autonomous Separation of Dual-Targeting DNA Guided Missile from Nanospacecraft for Intelligent Targeted Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:45201-45216. [PMID: 36184788 DOI: 10.1021/acsami.2c13624] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Most conventional chemotherapeutics indiscriminately kill both cancerous and healthy cells and cause toxic side effects, limiting the maximum tolerated dose and thereby compromising therapeutic efficacy. To address this challenge, here dual-targeting intelligent DNA guided missile (GM)-integrated nanospacecraft (NSC) (abbreviated as GM-NSC) is demonstrated for staged chemotherapeutic drug delivery exclusively into cancer cells and then mitochondria (not into healthy cells). GM-NSC is essentially a core/shell nanocomposite composed of gold nanoparticles (AuNPs) surrounded by a high-density multilayer DNA crown that is self-assembled from DNA tetrahedral units (DNA Tetra) in a highly ordered manner. Each tetrahedral structural unit is equipped with three functional components: a cancer cell-targeting aptamer pointing toward the outside environment, a hidden mitochondria-targeting triphenylphosphonium (TPP), and an explosive bolt (E-bolt). GM-NSC can remain intact in fetal bovine serum solution over 12 h and has 53-fold improved systemic stability. Each GM-NSC accommodates 1250 anticancer doxorubicin (Dox), achieving a 48-63-fold improved drug payload capacity. When systemically administrated into a tumor-bearing xenograft murine model, Dox-loaded GM-NSC enters into tumor sites with 18-fold improved specificity followed by autonomous separation of GMs from the NSC core and specific mitochondrial accumulation due to the explosion of E-bolt upon stimuli of endogenous miRNAs. About 80% of Dox uptaken is transferred into mitochondria and induces mitochondria-mediated apoptosis. As a result, the growth of malignant tumor is almost 100% inhibited without detectable toxicity to healthy tissues. Due to the desirable systemic stability, good biocompatibility, high cargo loading capability, satisfactory in vivo biodistribution, and therapeutic efficacy without adverse effects, intelligible GM-NSC is expected to become an alternative drug delivery system for precision cancer therapy.
Collapse
Affiliation(s)
- Shujuan Sun
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Ya Yang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Zhihua Gao
- Key Laboratory of Laboratory Medicine (Ministry of Education, China), Zhejiang Provincial Key Laboratory of Medicine Genetics, School of Laboratory Medicine and Life Sciences, Institute of Institute of Functional Nucleic Acids and Personalized Cancer Theranostics, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hao Jiang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Liyun Ye
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Yuqi Lai
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| | - Zhifa Shen
- Key Laboratory of Laboratory Medicine (Ministry of Education, China), Zhejiang Provincial Key Laboratory of Medicine Genetics, School of Laboratory Medicine and Life Sciences, Institute of Institute of Functional Nucleic Acids and Personalized Cancer Theranostics, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 305108, China
| |
Collapse
|
17
|
Effect of amphiphilic phosphorous dendrons on the conformation, secondary structure, and zeta potential of albumin and thrombin. Polym Bull (Berl) 2022. [DOI: 10.1007/s00289-022-04512-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
18
|
Zhou H, Wang Y, Hou Y, Zhang Z, Wang Q, Tian X, Lu H. Co‐delivery of Cisplatin and Chlorin e6 by Poly(phosphotyrosine) for Synergistic Chemotherapy and Photodynamic Therapy. CHINESE J CHEM 2022. [DOI: 10.1002/cjoc.202200334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Haisen Zhou
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering Peking University Beijing 100871 People's Republic of China
| | - Yaoyi Wang
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering Peking University Beijing 100871 People's Republic of China
| | - Yingqin Hou
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering Peking University Beijing 100871 People's Republic of China
| | - Zhengkui Zhang
- Department of General Surgery Peking University First Hospital Beijing 100034 People's Republic of China
| | - Qi Wang
- Department of General Surgery Peking University First Hospital Beijing 100034 People's Republic of China
| | - Xiaodong Tian
- Department of General Surgery Peking University First Hospital Beijing 100034 People's Republic of China
| | - Hua Lu
- Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering Peking University Beijing 100871 People's Republic of China
| |
Collapse
|
19
|
Evaluation and Efficacy Modified Carvacrol and Anti-cancer Peptide Against Cell Line Gastric AGS. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10426-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
20
|
Bai Y, Li X, Li M, Shang Q, Yang J, Fan L, Tian W. Host-guest interaction-based supramolecular prodrug self-assemblies for GSH-consumption augmented chemotherapy. J Mater Chem B 2022; 10:4952-4958. [PMID: 35723649 DOI: 10.1039/d2tb00989g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The over-expressed cellular glutathione (GSH) severely restricts the chemotherapeutic efficacy due to the GSH-induced detoxification of chemical drugs. Herein, how to construct effective drug delivery systems with GSH-consumption property is still a general concern and a major challenge. In this study, the host-guest interactions between water-soluble pillar[6]arene (WP[6]) and chlorambucil-arylboronic acid (Cb-BA) were utilized to construct supramolecular prodrug self-assemblies (SPSAs) with specific stimuli-responsive property. Notably, the BA moiety could not only consume GSH but also rapidly bind curcumin (Cur), which could inhibit the thioredoxin reductase (TrxR) to further reduce the GSH biosynthesis pathway. Benefiting from the functionality of BA-Cur conjugates, the GSH levels could be significantly downregulated, paving a novel way to enhance chemotherapeutic efficacy. In vitro and in vivo investigations demonstrated that this two-pronged GSH-depletion strategy could amplify the cellular oxidative stress and achieve excellent anti-tumor efficacy.
Collapse
Affiliation(s)
- Yang Bai
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Xihua Li
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Muqiong Li
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China.
| | - Qingqing Shang
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Jing Yang
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Li Fan
- School of Pharmacy, Air Force Medical University, Xi'an 710032, China.
| | - Wei Tian
- MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
21
|
Bai Y, Liu C, Yang J, Liu C, Shang Q, Tian W. Supramolecular self-assemblies based on water-soluble pillar[6]arene and drug-drug conjugates for the combination of chemotherapy. Colloids Surf B Biointerfaces 2022; 217:112606. [PMID: 35660745 DOI: 10.1016/j.colsurfb.2022.112606] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
Abstract
Although some co-drug delivery systems have been reported to treat cancer, how to optimal design these nano-systems with enhanced therapeutic efficacy is still a major challenge. As for the nitrogen mustard drugs chlorambucil (Cb), the overexpressed glutathione (GSH) in cancer tissue is responsible for their detoxification and reduced bioavailability. In this paper, chlorambucil-oxoplatin (Cb-Pt) was prepared to fabricate water-soluble pillar[6]arene (WP[6]) based supramolecular drug-drug self-assemblies (SDSAs). Remarkably, after the transcytosis by cancer cells, SDSAs was reduced by GSH to re lease Cb and higher toxic cisplatin, accompanying with the declining GSH level and ascending ROS level. Moreover, in vitro and in vivo experiments demonstrated that SDSAs with oxidative stress amplification strategy exhibited excellent therapeutic effect. This strategy might be useful for the synergistic co-drug based chemotherapy field.
Collapse
Affiliation(s)
- Yang Bai
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Caiping Liu
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Jing Yang
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Chengfei Liu
- Shaanxi Key Laboratory of Polymer Science & Technology, OME Key Laboratory of Supernormal Material Physics & Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Qingqing Shang
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Wei Tian
- Shaanxi Key Laboratory of Polymer Science & Technology, OME Key Laboratory of Supernormal Material Physics & Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, China.
| |
Collapse
|
22
|
Silverman L, Bhatti G, Wulff JE, Moffitt MG. Improvements in Drug-Delivery Properties by Co-Encapsulating Curcumin in SN-38-Loaded Anticancer Polymeric Nanoparticles. Mol Pharm 2022; 19:1866-1881. [PMID: 35579267 DOI: 10.1021/acs.molpharmaceut.2c00005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
SN-38 is an immensely potent anticancer agent although its use necessitates encapsulation to overcome issues of poor solubility and stability. Since SN-38 is a notoriously challenging drug to encapsulate, new avenues to increase encapsulation efficiency in polymer nanoparticles (PNPs) are needed. In this paper, we show that nanoprecipitation with curcumin (CUR) increases SN-38 encapsulation efficiencies in coloaded SN-38/CUR-PNPs based on poly(ε-caprolactone)-block-poly(ethylene glycol) (PCL-b-PEG) by up to a factor of 10. In addition, we find a dramatic decrease in PNP polydispersities, from 0.34 to 0.07, as the initial CUR-to-polymer ratio increases from 0 to 10, with only a modest increase in PNP size (from 40 to 55 nm). Compared to coloaded PNP formation using nanoprecipitation in the bulk or in a gas-liquid, a two-phase microfluidic reactor shows similar trends with respect to CUR content, although improvements in SN-38 encapsulation efficiencies both with and without CUR are found using the microfluidic method. Additional precipitation studies without copolymer suggest that CUR increases the dispersion of SN-38 in the solvent medium of micelle formation, which may contribute to the observed encapsulation enhancement. Cytotoxicity studies of unencapsulated SN-38/CUR mixtures show that addition of CUR does not significantly affect SN-38 potency against either U87 (glioblastoma) or A204 (rhabdomyosarcoma) cell lines. However, we find significant differences in the potencies of SN-38/CUR-PNP formulations depending on initial CUR amounts, with an optimized formulation showing subnanomolar cytotoxicity against A204 cells, significantly more potent than either free SN-38 or PNPs containing only SN-38.
Collapse
Affiliation(s)
- Lisa Silverman
- Department of Chemistry, University of Victoria, P.O. Box 1700, Stn CSC, Victoria, British Coloumbia V8W 2Y2, Canada
| | - Gitika Bhatti
- Department of Chemistry, University of Victoria, P.O. Box 1700, Stn CSC, Victoria, British Coloumbia V8W 2Y2, Canada
| | - Jeremy E Wulff
- Department of Chemistry, University of Victoria, P.O. Box 1700, Stn CSC, Victoria, British Coloumbia V8W 2Y2, Canada
| | - Matthew G Moffitt
- Department of Chemistry, University of Victoria, P.O. Box 1700, Stn CSC, Victoria, British Coloumbia V8W 2Y2, Canada
| |
Collapse
|
23
|
Hu C, Wei G, Zhu F, Wu A, Luo L, Shen S, Zhang J. Platinum-Based Nanocomposite Pt@BSA as an Efficient Electrochemical Biosensing Interface for Rapid and Ultrasensitive Determination of Folate Receptor-Positive Tumor Cells. ACS APPLIED BIO MATERIALS 2022; 5:3038-3048. [PMID: 35544589 DOI: 10.1021/acsabm.2c00332] [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] [Indexed: 12/24/2022]
Abstract
Developing a cytosensing strategy based on electrochemical approaches has attracted wide interest due to the low cost, rapid response, and simple instrumentation. In this work, an electrochemical cytosensor employing the Pt@BSA nanocomposite as the biosensing substrate along with the covalent cross-linking of targeting molecules folic acid (FA) was constructed for highly sensitive determination of folate receptor (FR)-positive tumor cells. The prepared Pt@BSA nanocomposite revealed excellent biocompatibility for cell adhesion and proliferation, which was confirmed by cell viability evaluation using thiazolyl blue tetrazolium bromide (MTT) colorimetric methods. Due to the satisfactory electrical conductivity originating from Pt@BSA and the high binding affinity of FA to FR on the cell surface, an ultrasensitive and specific cytosensing device was designed for rapid and quantitative determination of HeLa cells (a model system) by differential pulse voltammetry (DPV) tests. This proposed cytosensor resulted in a wide HeLa cell determination range of 2.8 × 101-2.8 × 106 cells mL-1 with a low DPV detection limit of 9 cells mL-1. The developed cytosensing approach exhibited highly specific recognition of FR-positive tumor cells, excellent inter-assay reproducibility with a relative standard deviation (RSD) of 4.7%, acceptable intra-assay precision, and favorable storage stability, expanding the application of electrochemical measurement technology in the biomedical field of early detection and diagnosis of cancers.
Collapse
Affiliation(s)
- Chenyi Hu
- Institute of Fuel Cells, Key Laboratory of Power Machinery and Engineering of MOE, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guanghua Wei
- SJTU-Paris Tech Elite Institute of Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fengjuan Zhu
- Institute of Fuel Cells, Key Laboratory of Power Machinery and Engineering of MOE, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Aiming Wu
- Institute of Fuel Cells, Key Laboratory of Power Machinery and Engineering of MOE, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Liuxuan Luo
- Institute of Fuel Cells, Key Laboratory of Power Machinery and Engineering of MOE, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuiyun Shen
- Institute of Fuel Cells, Key Laboratory of Power Machinery and Engineering of MOE, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Junliang Zhang
- Institute of Fuel Cells, Key Laboratory of Power Machinery and Engineering of MOE, School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
24
|
Mignani S, Shi X, Rodrigues J, Tomás H, Majoral JP. Dendrimer nanoplatforms for veterinary medicine applications: A concise overview. Drug Discov Today 2022; 27:1251-1260. [PMID: 34999213 DOI: 10.1016/j.drudis.2022.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/07/2021] [Accepted: 01/04/2022] [Indexed: 02/08/2023]
Abstract
Within the nanoparticle (NP) space, dendrimers are becoming increasingly important in the field of nanomedicine, not only to treat human diseases, but also in veterinary medicine, which represents a new therapeutic approach. Major applications include using dendrimers to tackle highly contagious foot-and-mouth disease virus (FMDV) and swine fever virus (SFV) in pigs, FMDV in cattle, hypothermic circulatory arrest (HCA) in dogs, rabies, and H9N2 avian influenza virus in chickens. As we review here, intramuscular (im) subcutaneous (sc), intravenous (iv), and intraperitoneal (ip) routes of administration can be used for the successful application of dendrimers in animals.
Collapse
Affiliation(s)
- Serge Mignani
- Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, 45, Rue des Saints Peres, 75006 Paris, France; CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal.
| | - Xiangyang Shi
- CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal; College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, PR China.
| | - João Rodrigues
- CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal; School of Materials Science and Engineering, Center for Nano Energy Materials, Northwestern Polytechnical University, Xi'an 710072, China.
| | - Helena Tomás
- CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, 31077 Toulouse Cedex 4, France; Université Toulouse, 118 Route de Narbonne, 31077 Toulouse Cedex 4, France.
| |
Collapse
|
25
|
Evaluation of Liposome-Loaded Microbubbles as a Theranostic Tool in a Murine Collagen-Induced Arthritis Model. Sci Pharm 2022. [DOI: 10.3390/scipharm90010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by severe inflammation of the synovial tissue. Here, we assess the feasibility of liposome-loaded microbubbles as theranostic agents in a murine arthritis model. First, contrast-enhanced ultrasound (CEUS) was used to quantify neovascularization in this model since CEUS is well-established for RA diagnosis in humans. Next, the potential of liposome-loaded microbubbles and ultrasound (US) to selectively enhance liposome delivery to the synovium was evaluated with in vivo fluorescence imaging. This procedure is made very challenging by the presence of hard joints and by the limited lifetime of the microbubbles. The inflamed knee joints were exposed to therapeutic US after intravenous injection of liposome-loaded microbubbles. Loaded microbubbles were found to be quickly captured by the liver. This resulted in fast clearance of attached liposomes while free and long-circulating liposomes were able to accumulate over time in the inflamed joints. Our observations show that murine arthritis models are not well-suited for evaluating the potential of microbubble-mediated drug delivery in joints given: (i) restricted microbubble passage in murine synovial vasculature and (ii) limited control over the exact ultrasound conditions in situ given the much shorter length scale of the murine joints as compared to the therapeutic wavelength.
Collapse
|
26
|
Karpus A, Mignani S, Zablocka M, Majoral JP. Crown Macromolecular Derivatives: Stepwise Design of New Types of Polyfunctionalized Phosphorus Dendrimers. J Org Chem 2022; 87:3433-3441. [PMID: 35142502 DOI: 10.1021/acs.joc.1c03009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Phosphorus dendrimers are used for many applications in different domains including nanomedicine as cargo of drugs or as species active per se but also in a variety of other fields ranging from nanoscience to catalysis. Their properties depend on the nature of their internal structure and mainly of the diversity and versatility of the functional groups located on their outer shell. Therefore, there is a need to diversify their structure in order to use them for new applications and to propose alternative synthetic pathways to be built easily, at each step and in high yield a family of original stable phosphorus dendrimers of different generations. Such a goal is illustrated in this report with the original synthesis of 14 new phosphorus dendrimers of generation 0 to 2 and the possibility to modify at will their internal structure and the nature of their functional end groups.
Collapse
Affiliation(s)
- Andrii Karpus
- Laboratoire de Chimie de Coordination (LCC), CNRS, 205 route de Narbonne, Toulouse Cedex 4 31077, France.,LCC-CNRS Université de Toulouse, 118 route de Narbonne, Toulouse Cedex 4 31077, France
| | - Serge Mignani
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, 45, rue des Saints Peres, Paris 75006, France.,CQM─Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, Funchal 9020-105, Portugal
| | - Maria Zablocka
- Polish Academy of Sciences, Centre of Molecular and Macromolecular Studies, Sienkiewicza 112, Lodz 90001, Poland
| | - Jean Pierre Majoral
- Laboratoire de Chimie de Coordination (LCC), CNRS, 205 route de Narbonne, Toulouse Cedex 4 31077, France.,LCC-CNRS Université de Toulouse, 118 route de Narbonne, Toulouse Cedex 4 31077, France
| |
Collapse
|
27
|
Mignani S, Shi X, Rodrigues J, Tomás H, Majoral JP. Dendrimer nanoplatforms for veterinary medicine applications: A concise overview. Drug Discov Today 2022. [DOI: https://doi.org/10.1016/j.drudis.2022.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
28
|
Lu D, Chen M, Yu L, Chen Z, Guo H, Zhang Y, Han Z, Xu T, Wang H, Zhou X, Zhou Z, Teng G. Smart-Polypeptide-Coated Mesoporous Fe 3O 4 Nanoparticles: Non-Interventional Target-Embolization/Thermal Ablation and Multimodal Imaging Combination Theranostics for Solid Tumors. NANO LETTERS 2021; 21:10267-10278. [PMID: 34878286 DOI: 10.1021/acs.nanolett.1c03340] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Tumor theranostics hold great potential for personalized medicine in the future, and transcatheter arterial embolization (TAE) is an important clinical treatment for unresectable or hypervascular tumors. In order to break the limitation, simplify the procedure of TAE, and achieve ideal combinatorial theranostic capability, here, a kind of triblock-polypeptide-coated perfluoropentane-loaded mesoporous Fe3O4 nanocomposites (PFP-m-Fe3O4@PGTTCs) were prepared for non-interventional target-embolization, magnetic hyperthermia, and multimodal imaging combination theranostics of solid tumors. The results of systematic animal experiments by H22-tumor-bearing mice and VX2-tumor-bearing rabbits in vivo indicated that PFP-m-Fe3O4@PGTTC-6.3 has specific tumor accumulation and embolization effects. The tumors' growth has been inhibited and the tumors disappeared 4 weeks and ≤15 days post-injection with embolization and magnetic hyperthermia combination therapy, respectively. The results also showed an excellent effect of magnetic resonance/ultrasound/SPECT multimodal imaging. This pH-responsive non-interventional embolization combinatorial theranostics system provides a novel embolization and multifunctional theranostic candidate for solid tumors.
Collapse
Affiliation(s)
- Dedai Lu
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Mingshu Chen
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Lili Yu
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Zhengpeng Chen
- Key Laboratory of Eco-functional Polymer Materials of the Ministry of Education, Key Laboratory of Eco-environmental Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, 730070, China
| | - Hongyun Guo
- Institute of Gansu Medical Science Research, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Yongdong Zhang
- Institute of Gansu Medical Science Research, Gansu Provincial Cancer Hospital, Lanzhou, 730050, China
| | - Zhiming Han
- Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Tingting Xu
- Zhongda Hospital Southeast University, Jiangsu Key Laboratory of Molecular Imaging and Function Imaging, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Haijun Wang
- Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Xing Zhou
- Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Zubang Zhou
- Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Gaojun Teng
- Zhongda Hospital Southeast University, Jiangsu Key Laboratory of Molecular Imaging and Function Imaging, School of Medicine, Southeast University, Nanjing, 210009, China
| |
Collapse
|
29
|
Zhang Y, An J, Shao Y, Yu N, Yue S, Sun H, Zhang J, Gu W, Xia Y, Zhang J, Xu Y, Zhong Z. CD38-Directed Vincristine Nanotherapy for Acute Lymphoblastic Leukemia. Biomacromolecules 2021; 23:377-387. [PMID: 34913676 DOI: 10.1021/acs.biomac.1c01342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy in children. Although intensive chemotherapy greatly improved the survival rate, it is often accompanied by severe and lifelong side effects as a result of weak ALL selectivity. The intensive and poorly selective chemotherapy is also detrimental to patients' immune system. There is an urgent need to develop more selective and less toxic chemotherapy for ALL. Here, we report daratumumab-polymersome-vincristine (DP-VCR) as a CD38-directed nanotherapy for ALL. DP-VCR showed selective uptake in CD38-positive 697 and Nalm-6-Luc ALL cells and potent anti-ALL activity with an IC50 as low as 0.06 nM VCR, which was 13.7-fold more potent than free VCR. In contrast, no toxicity to human peripheral blood mononuclear cells was detected for DP-VCR even at 108.3 nM VCR. The apoptotic assays confirmed a high selectivity of DP-VCR to CD38-positive ALL cells. DP-VCR exhibited superior treatment of both 697 and Nalm-6-Luc orthotopic ALL models to all controls, as revealed by significant survival benefit and marked reduction of leukemia burden in bone marrow, blood, spleen, and liver. Importantly, DP-VCR induced few side effects. DP-VCR emerges as a safe and potent nanotherapy for CD38-positive ALL.
Collapse
Affiliation(s)
- Yifan Zhang
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Jingnan An
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215123, P. R. China
| | - Yu Shao
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Na Yu
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Shujing Yue
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Huanli Sun
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Jubin Zhang
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215123, P. R. China
| | - Wenxing Gu
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Yifeng Xia
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, P. R. China
| | - Yang Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
30
|
Ma Y, Wang C, Zhu L, Yu C, Lu B, Wang Y, Ding Y, Dong CM, Yao Y. Polydopamine-drug conjugate nanocomposites based on ZIF-8 for targeted cancer photothermal-chemotherapy. J Biomed Mater Res A 2021; 110:954-963. [PMID: 34913253 DOI: 10.1002/jbm.a.37344] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/19/2021] [Accepted: 12/04/2021] [Indexed: 12/26/2022]
Abstract
Stimuli-responsive prodrug-based nanoplatform with synergistic antitumor activity is of central importance to the development of promising nanomedicines for cancer therapy. Here, we describe a polydopamine-drug conjugate nanocomposite (ZP-PDA-DOX) with targeted cancer photothermal-chemotherapy (PTT-CT), which constructed by a gradual copolymerization of dopamine (DA) and pH-sensitive dopamine-derived prodrug (DA-DOX) into the porous channels of zeolite imidazolate frameworks-8 (ZIF-8), followed by PEGylation with amino-terminated folic acid-polyethylene glycol (NH2 -PEG-FA) to acquire the high biocompatibility, specificity, and excellent tumor-targeting property. The incorporation of polydopamine strengthened the stability and dispersion of ZIF-8, and also conferred photothermal conversion effect. In the tumor acidic microenvironment, the acid-labile hydrazone linker of DA-DOX and ZIF-8 promptly degraded to release activated DOX. Moreover, the generated hyperthermia due to the high photothermal conversion efficiency of PDA component could accelerate drug release, and simultaneously thermally ablate tumor tissue to maximize the DOX-induced CT, which could also assist PTT to eradicate tumor cells. This study provides a promising strategy for targeted cancer PTT-CT with synergistic anti-tumor effect.
Collapse
Affiliation(s)
- Yuxuan Ma
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| | - Chenwei Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| | - Lvming Zhu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| | - Chunmei Yu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| | - Bing Lu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| | - Yang Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| | - Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| | - Chang-Ming Dong
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai, China
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, China
| |
Collapse
|
31
|
Peng K, Liang BB, Liu W, Mao ZW. What blocks more anticancer platinum complexes from experiment to clinic: Major problems and potential strategies from drug design perspectives. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214210] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
Gao M, Deng H, Zhang W. Hyaluronan-based Multifunctional Nano-carriers for Combination Cancer Therapy. Curr Top Med Chem 2021; 21:126-139. [PMID: 32962617 DOI: 10.2174/1568026620666200922113846] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/30/2022]
Abstract
Hyaluronan (HA) is a natural linear polysaccharide that has excellent hydrophilicity, biocompatibility, biodegradability, and low immunogenicity, making it one of the most attractive biopolymers used for biomedical researches and applications. Due to the multiple functional sites on HA and its intrinsic affinity for CD44, a receptor highly expressed on various cancer cells, HA has been widely engineered to construct different drug-loading nanoparticles (NPs) for CD44-targeted anti-tumor therapy. When a cocktail of drugs is co-loaded in HA NP, a multifunctional nano-carriers could be obtained, which features as a highly effective and self-targeting strategy to combat cancers with CD44 overexpression. The HA-based multidrug nano-carriers can be a combination of different drugs, various therapeutic modalities, or the integration of therapy and diagnostics (theranostics). Up to now, there are many types of HA-based multidrug nano-carriers constructed by different formulation strategies, including drug co-conjugates, micelles, nano-gels and hybrid NP of HA and so on. This multidrug nano-carrier takes the full advantages of HA as an NP matrix, drug carriers and targeting ligand, representing a simplified and biocompatible platform to realize the targeted and synergistic combination therapy against the cancers. In this review, recent progress of HA-based multidrug nano-carriers for combination cancer therapy is summarized and the potential challenges for translational applications have been discussed.
Collapse
Affiliation(s)
- Menghan Gao
- State Key Laboratory of Medical Molecular Biology & Department of Biomedical Engineering, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Hong Deng
- State Key Laboratory of Medical Molecular Biology & Department of Biomedical Engineering, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Weiqi Zhang
- State Key Laboratory of Medical Molecular Biology & Department of Biomedical Engineering, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| |
Collapse
|
33
|
Wang Q, Wu C, Li X, Yang D, Shi L. Cisplatin and paclitaxel co-delivery nanosystem for ovarian cancer chemotherapy. Regen Biomater 2021; 8:rbab015. [PMID: 35707698 DOI: 10.1093/rb/rbab015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/04/2021] [Accepted: 03/22/2021] [Indexed: 12/24/2022] Open
Abstract
We have designed and developed an effective drug delivery system using biocompatible polymer of poly (ethylene glycol)-polyaspartic acid (mPEG-PAsp) for co-loading the chemotherapy drugs paclitaxel (PTX) and cisplatin (CP) in one nano-vehicle. This study aimed to improve the anti-cancer efficacy of combinations of chemotherapy drugs and reduce their side effects. mPEG-PAsp-(PTX/Pt) nano-micelles disperse well in aqueous solution and have a narrow size distribution (37.8 ± 3.2 nm) in dynamic light scattering (DLS). Drug release profiles found that CP released at pH 5.5 was significantly faster than that at pH 7.4. MPEG-PAsp-(PTX/Pt) nano-micelles displayed a significantly higher tumor inhibitory effect than mPEG-PAsp-PTX nano-micelles when the polymer concentrations reached 50 μg/mL. Our data indicated that polymer micelles of mPEG-PAsp loaded with the combined drug exert synergistic anti-tumor efficacy on SKOV3 ovarian cells via different action mechanisms. Results from our studies suggested that mPEG-PAsp-(PTX/Pt) nano-micelles are promising alternatives for carrying and improving the delivery of therapeutic drugs with different water solubilities.
Collapse
Affiliation(s)
- Qiaoying Wang
- Department of Medicine, Leshan Vocational and Technical College, No. 1336, Middle Section of Qingyijiang Avenue, Shizhong District, Leshan City, Sichuan Province, China
| | - Changqiang Wu
- Medical Imaging Key Laboratory of Sichuan Province and School of Medical Imaging, North Sichuan Medical College, 55 Dongshun Road, Gaoping District, Nanchong, Sichuan Province, China
| | - Xiaoting Li
- Department of Medicine, Leshan Vocational and Technical College, No. 1336, Middle Section of Qingyijiang Avenue, Shizhong District, Leshan City, Sichuan Province, China
| | - Dixiao Yang
- Department of Medicine, Leshan Vocational and Technical College, No. 1336, Middle Section of Qingyijiang Avenue, Shizhong District, Leshan City, Sichuan Province, China
| | - Liangjun Shi
- Department of Medicine, Leshan Vocational and Technical College, No. 1336, Middle Section of Qingyijiang Avenue, Shizhong District, Leshan City, Sichuan Province, China
| |
Collapse
|
34
|
Zhou L, Du C, Zhang R, Dong C. Stimuli-responsive dual drugs-conjugated polydopamine nanoparticles for the combination photothermal-cocktail chemotherapy. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2020.02.043] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
35
|
Majoral JP, Zablocka M, Ciepluch K, Milowska K, Bryszewska M, Shcharbin D, Katir N, El Kadib A, Caminade AM, Mignani S. Hybrid phosphorus–viologen dendrimers as new soft nanoparticles: design and properties. Org Chem Front 2021. [DOI: 10.1039/d1qo00511a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Design of new families of dendritic soft nanoparticles constituted of phosphorus, viologen and carbosilane fragments and their properties as nanomaterials and applications in biology.
Collapse
Affiliation(s)
- Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination
- CNRS
- UPR 8241
- 31077 Toulouse CEDEX 4
- France
| | - Maria Zablocka
- Center of Molecular and Macromolecular Studies
- Polish Academy of Science
- 90001 Lodz
- Poland
| | - Karol Ciepluch
- Division of Medical Biology
- Jan Kochanowski University
- Kielce
- Poland
| | - Katarzyna Milowska
- Department of General Biophysics
- Faculty of Biology and Environmental Protection
- University of Lodz
- Lodz
- Poland
| | - Maria Bryszewska
- Department of General Biophysics
- Faculty of Biology and Environmental Protection
- University of Lodz
- Lodz
- Poland
| | | | - Nadia Katir
- Euromed Research Center
- Engineering Division
- Euro-Med University of Fes (UEMF)
- Fès
- Morocco
| | - Abdelkrim El Kadib
- Euromed Research Center
- Engineering Division
- Euro-Med University of Fes (UEMF)
- Fès
- Morocco
| | - Anne-Marie Caminade
- Laboratoire de Chimie de Coordination
- CNRS
- UPR 8241
- 31077 Toulouse CEDEX 4
- France
| | - Serge Mignani
- Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique
- Université Paris Descartes
- PRES Sorbonne Paris Cité
- CNRS UMR 860
- 75006 Paris
| |
Collapse
|
36
|
Mignani S, Shi X, Karpus A, Majoral JP. Non-invasive intranasal administration route directly to the brain using dendrimer nanoplatforms: An opportunity to develop new CNS drugs. Eur J Med Chem 2021; 209:112905. [PMID: 33069435 PMCID: PMC7548078 DOI: 10.1016/j.ejmech.2020.112905] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
There are several routes of administration to the brain, including intraparenchymal, intraventricular, and subarachnoid injections. The blood-brain barrier (BBB) impedes the permeation and access of most drugs to the central nervous system (CNS), and consequently, many neurological diseases remain undertreated. For past decades, to circumvent this effect, several nanocarriers have been developed to deliver drugs to the brain. Importantly, intranasal (IN) administration can allow direct delivery of drugs into the brain through the anatomical connection between the nasal cavity and brain without crossing the BBB. In this regard, dendrimers may possess great potential to deliver drugs to the brain by IN administration, bypassing the BBB and reducing systemic exposure and side effects, to treat diseases of the CNS. In this original concise review, we highlighted the few examples advocated regarding the use of dendrimers to deliver CNS drugs directly via IN. This review highlighed the few examples of the association of dendrimer encapsulating drugs (e.g., small compounds: haloperidol and paeonol; macromolecular compounds: dextran, insulin and calcitonin; and siRNA) using IN administration. Good efficiencies were observed. In addition, we will present the in vivo effects of PAMAM dendrimers after IN administration, globally, showing no general toxicity.
Collapse
Affiliation(s)
- Serge Mignani
- Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, 45, Rue des Saints Peres, 75006, Paris, France; CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105, Funchal, Portugal.
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, PR China.
| | - Andrii Karpus
- Laboratoire de Chimie de Coordination Du CNRS, 205 Route de Narbonne, 31077, Toulouse, Cedex 4, France; Université Toulouse 118 Route de Narbonne, 31077, Toulouse, Cedex 4, France
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination Du CNRS, 205 Route de Narbonne, 31077, Toulouse, Cedex 4, France; Université Toulouse 118 Route de Narbonne, 31077, Toulouse, Cedex 4, France.
| |
Collapse
|
37
|
Astani S, Salehi R, Massoumi B, Massoudi A. Co-delivery of cisplatin and doxorubicin by carboxylic acid functionalized poly (hydroxyethyl methacrylate)/reduced graphene nanocomposite for combination chemotherapy of breast cancer cells. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2020; 32:657-677. [PMID: 33347395 DOI: 10.1080/09205063.2020.1855393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
In this study a novel pH-responsive magnetic nanocomposite based on reduced graphene oxide was developed for combination of doxorubicin (Dox)-cisplatin (Cis) delivery to destroy the MCF-7 cell line. For this purpose, polyhydroxyethyl methacrylate (PHEMA) was bonded to the reduced graphene oxide through ATRP polymerization using grafting from method. Then the PHEMA hydroxy groups were converted to succinyloxy groups by polyesterification with succinic anhydride. The physicochemical properties of the nanocomposite were investigated via FTIR, SEM, XRD, DLS and TGA analysis. Unique structure of nanocomposite led to simultaneous encapsulation of Dox (75%) and Cis (82%) through ionic interaction, π-π stacking and hydrogen bonding. The obtained nanocomposite was uptake by MCF-7 cells at early first hour because of nanocomposite small size (below 70 nm). Cell viability assay results revealed that the Dox&Cis-loaded nanocomposite showed the highest rate of MCF-7 cells at lowest concentration (IC50 = 0.798 µg/mL) compared to treatment groups received single drug-loaded nanocomposite and free drugs. Dox&Cis-loaded nanocomposite exhibited a synergistic influence with the combination index (CI) value <1. The cell cycle analysis results revealed that the highest amount of apoptosis (cells population in sub G1 was 75%) was observed in the Dox&Cis-loaded nanocomposite treatment group compared with the single drug-loaded nanocomposite and free drugs. Our findings confirmed that combinational therapy by Dox and Cis graphene oxide-based nanocomposite has increased the cytotoxicity in MCF-7 cells by stimulating the apoptotic response.
Collapse
Affiliation(s)
- Shahram Astani
- Department of Chemistry, Payame Noor University, Tehran, Iran
| | - Roya Salehi
- Drug Applied Research Centre and Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | |
Collapse
|
38
|
Mignani S, Shi X, Steinmetz A, Majoral JP. Multivalent Copper(II)-Conjugated Phosphorus Dendrimers with Noteworthy In Vitro and In Vivo Antitumor Activities: A Concise Overview. Mol Pharm 2020; 18:65-73. [PMID: 33236637 DOI: 10.1021/acs.molpharmaceut.0c00892] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dendrimers are macromolecules with well-defined, homogeneous, and monodispersed structures that form a branch-like structure. In general, they have a symmetric core, inner shells, and an outer shell. Over the past decade, metallodendritic architectures have developed into a new area in nanomedicine. Due to their versatility and facile customization, phosphorus dendrimers represent interesting platforms for biomedical applications. Metallo-conjugated phosphorus dendrimers have been developed within the dendrimer space, an important part of the chemical space. The first investigation was made using phosphorus dendrimers bearing copper(II) groups on their surface as the original anticancer drug candidates. The aim of this minireview is to present our powerful strategy to find and develop original multivalent copper(II)-conjugated phosphorus dendrimers. The most potent of them is G3 dendrimers with N-(pyridine-2-ylmethylene)ethanamine as the chelating motif complexed with Cu(II) (1G3-Cu), showing very good in vitro and in vivo antiproliferative efficacy. On the basis of these results, 1G3-Cu is a potential clinical candidate having progressed from hit to preclinical candidate status.
Collapse
Affiliation(s)
- Serge Mignani
- Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologique, 45 rue des Saints Peres, 75006 Paris, France.,CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal
| | - Xiangyang Shi
- CQM - Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal.,College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, PR China
| | - Anke Steinmetz
- Sanofi R&D, Integrated Drug Discovery, Centre de Recherche Vitry-Alfortville, 94403 Cedex Vitry-sur-Seine, France
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 route de Narbonne, 31077 Cedex 4 Toulouse, France.,Université Toulouse, 118 route de Narbonne, 31077 Cedex 4 Toulouse, France
| |
Collapse
|
39
|
Cen M, Ding Y, Wang J, Yuan X, Lu B, Wang Y, Yao Y. Cationic Water-Soluble Pillar[5]arene-Modified Cu 2-xSe Nanoparticles: Supramolecular Trap for ATP and Application in Targeted Photothermal Therapy in the NIR-II Window. ACS Macro Lett 2020; 9:1558-1562. [PMID: 35617083 DOI: 10.1021/acsmacrolett.0c00714] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
With the rapid progress of nanotechnology, near-infrared (NIR), light-assisted phototherapy as a minimally invasive local cancer therapy, especially photothermal therapy (PTT), has captured broad research attention in recent years. However, combined target molecules with a PTT system through reversible supramolecular interactions has been reported rarely. In this work, we constructed a supramolecular nanosystem combining ATP capture and target PTT based on cationic pillar[5]arene (CWP5)-functionalized Cu2-xSe nanoparticles (Cu2-xSe@CWP5 NPs). Cu2-xSe@CWP5 NPs, with an average diameter of approximately 100 nm and strong absorption in the near-infrared-II window, were prepared in water through a facile one-step in situ synthesis method, then (4-carboxybutyl)triphenylphosphonium bromide (TPP), a mitochondria-targeted molecule, was modified on the surface of the particles through the host-guest recognition. Upon irradiation with a 1064 nm laser, the obtained Cu2-xSe@CWP5/TPP NPs showed remarkably photothermal ablation capability to HeLa cells. Importantly, our Cu2-xSe@CWP5/TPP NPs exhibited excellent therapeutic effect due to the combination of inhibited hydrolysis of ATP and targeted photothermal therapy upon in vitro and in vivo studies. Significantly, through host-guest interactions, we can modify different types of target molecules within this PTT system at will.
Collapse
Affiliation(s)
- Moupan Cen
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, People’s Republic of China
| | - Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, People’s Republic of China
| | - Jin Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, People’s Republic of China
| | - Xiaolei Yuan
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, People’s Republic of China
| | - Bing Lu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, People’s Republic of China
| | - Yang Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, People’s Republic of China
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, People’s Republic of China
| |
Collapse
|
40
|
Combination of Cytochalasin H and zinc oxide nanoparticles in human breast cancer: an insight into apoptosis study. Biologia (Bratisl) 2020. [DOI: 10.2478/s11756-020-00611-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
41
|
Targeted co-delivery of curcumin and doxorubicin by citric acid functionalized Poly (ε-caprolactone) based micelle in MDA-MB-231 cell. Colloids Surf B Biointerfaces 2020; 194:111225. [DOI: 10.1016/j.colsurfb.2020.111225] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/08/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
|
42
|
Liew HS, Mai CW, Zulkefeli M, Madheswaran T, Kiew LV, Delsuc N, Low ML. Recent Emergence of Rhenium(I) Tricarbonyl Complexes as Photosensitisers for Cancer Therapy. Molecules 2020; 25:E4176. [PMID: 32932573 PMCID: PMC7571230 DOI: 10.3390/molecules25184176] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 02/05/2023] Open
Abstract
Photodynamic therapy (PDT) is emerging as a significant complementary or alternative approach for cancer treatment. PDT drugs act as photosensitisers, which upon using appropriate wavelength light and in the presence of molecular oxygen, can lead to cell death. Herein, we reviewed the general characteristics of the different generation of photosensitisers. We also outlined the emergence of rhenium (Re) and more specifically, Re(I) tricarbonyl complexes as a new generation of metal-based photosensitisers for photodynamic therapy that are of great interest in multidisciplinary research. The photophysical properties and structures of Re(I) complexes discussed in this review are summarised to determine basic features and similarities among the structures that are important for their phototoxic activity and future investigations. We further examined the in vitro and in vivo efficacies of the Re(I) complexes that have been synthesised for anticancer purposes. We also discussed Re(I) complexes in conjunction with the advancement of two-photon PDT, drug combination study, nanomedicine, and photothermal therapy to overcome the limitation of such complexes, which generally absorb short wavelengths.
Collapse
Affiliation(s)
- Hui Shan Liew
- School of Postgraduate Studies, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
| | - Chun-Wai Mai
- Centre for Cancer and Stem Cell Research, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia;
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| | - Mohd Zulkefeli
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| | - Thiagarajan Madheswaran
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| | - Lik Voon Kiew
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Nicolas Delsuc
- Laboratoire des Biomolécules, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, 75005 Paris, France;
| | - May Lee Low
- School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia; (M.Z.); (T.M.)
| |
Collapse
|
43
|
Du C, Ding Y, Qian J, Zhang R, Dong CM. Dual drug-paired polyprodrug nanotheranostics reverse multidrug resistant cancers via mild photothermal-cocktail chemotherapy. J Mater Chem B 2020; 7:5306-5319. [PMID: 31411235 DOI: 10.1039/c9tb01368g] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Combating multidrug resistance (MDR) of tumors is still challenging for clinical chemotherapy, cocktail chemotherapy (CCT), and currently widely-studied nanodrug-based treatments. Inspired by different MDR-overcoming and antitumor mechanisms of CCT and photothermal therapy (PT), a dual drug-paired polyprodrug nanoparticle (PDCN25-CDDP) was constructed to achieve the combination therapy PT-CCT for reversing MDR and combating multidrug resistant cancers. The PT-CCT treatment can greatly downregulate the P-gp expression level and achieve utmost MDR-reversal and antitumor efficacy by both a cocktail effect of CCT and a synergistic effect of CCT with PT; meanwhile, PT can inhibit the expression of heat shock protein 90 and enhance the thermosensitivity of cancer cells. Upon NIR irradiation, PDCN25-CDDPin vivo produced a selective tumor accumulation effect and relatively deep tumor penetration, as evidenced by fluorescent and photoacoustic imaging and CLSM. The mild PT-CCT treatment completely eradicated MCF-7/ADR and OVCAR-3/DDP tumors without skin damage or tumor recurrence for 30 days, exhibiting synergistic MDR-reversal and superior antitumor efficacy in vivo. Importantly, this work provides an innovative strategy for reversing MDR and combating DOX-resistant breast and CDDP-resistant ovarian cancers.
Collapse
Affiliation(s)
- Chang Du
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| | - Yue Ding
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| | - Jiwen Qian
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| | - Rong Zhang
- Department of Obstetrics and Gynecology, Shanghai Fengxian Central Hospital, Southern Medical University, Shanghai 201499, P. R. China.
| | - Chang-Ming Dong
- School of Chemistry and Chemical Engineering, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China.
| |
Collapse
|
44
|
Hou S, Zhou S, Chen S, Lu Q. Polyphosphazene-Based Drug Self-Framed Delivery System as a Universal Intelligent Platform for Combination Therapy against Multidrug-Resistant Tumors. ACS APPLIED BIO MATERIALS 2020; 3:2284-2294. [DOI: 10.1021/acsabm.0c00072] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Shenglei Hou
- School of Chemistry and Chemical Engineering, The State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shiliu Zhou
- School of Chemistry and Chemical Engineering, The State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuangshuang Chen
- School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China
| | - Qinghua Lu
- School of Chemistry and Chemical Engineering, The State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
45
|
Lara-Chacón B, Guerrero-Rodríguez SL, Ramírez-Hernández KJ, Robledo-Rivera AY, Velazquez MAV, Sánchez-Olea R, Calera MR. Gpn3 Is Essential for Cell Proliferation of Breast Cancer Cells Independent of Their Malignancy Degree. Technol Cancer Res Treat 2020; 18:1533033819870823. [PMID: 31431135 PMCID: PMC6704425 DOI: 10.1177/1533033819870823] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Successful therapies for patients with breast cancer often lose their initial effectiveness. Thus, identifying new molecular targets is a constant goal in the fight against breast cancer. Gpn3 is a protein required for RNA polymerase II nuclear targeting in both yeast and human cells. We investigated here the effect of suppressing Gpn3 expression on cell proliferation in a progression series of isogenic cell lines derived from the nontumorigenic MCF-10A breast cells that recapitulate different stages of breast carcinogenesis. Gpn3 protein levels were comparable in all malignant derivatives of the nontumorigenic MCF-10A cells. shRNA-mediated inhibition of Gpn3 expression markedly decreased cell proliferation in all MCF-10A sublines. A fraction of the largest RNA polymerase II subunit Rpb1 was retained in the cytoplasm, but most Rpb1 remained nuclear after suppressing Gpn3 in all cell lines studied. Long-term proliferation experiments in cells with suppressed Gpn3 expression resulted in the eventual loss of all isogenic cell lines but MCF-10CA1d.cl1. In MCF-10CA1d.cl1 cells, Gpn3 knockdown reduced the proliferation of breast cancer stem cells as evaluated by mammosphere assays. After the identification that Gpn3 plays a key role in cell proliferation in mammary epithelial cells independent of the degree of transformation, we also analyzed the importance of Gpn3 in other human breast cancer cell lines from different subtypes. Gpn3 was also required for cell proliferation and nuclear translocation of RNA polymerase II in such cellular models. Altogether, our results show that Gpn3 is essential for breast cancer cell proliferation regardless of the transformation level, indicating that Gpn3 could be considered a molecular target for the development of new antiproliferative therapies. Importantly, our analysis of public data revealed that Gpn3 overexpression was associated with a significant decrease in overall survival in patients with estrogen receptor-positive and Human epidermal growth factor receptor 2 (HER2+) breast cancer, supporting our proposal that targeting Gpn3 could potentially benefit patients with breast cancer.
Collapse
Affiliation(s)
- Bárbara Lara-Chacón
- 1 Instituto de Fisica Manuel Sandoval Vallarta, Universidad Autonoma de San Luis Potos, San Luis Potosi, Mexico
| | | | - Karla J Ramírez-Hernández
- 1 Instituto de Fisica Manuel Sandoval Vallarta, Universidad Autonoma de San Luis Potos, San Luis Potosi, Mexico
| | | | - Marco Antonio Velasco Velazquez
- 2 Departamento de Farmacología y Unidad Periférica de Investigación en Biomedicina Traslacional, México city, México.,3 Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Roberto Sánchez-Olea
- 1 Instituto de Fisica Manuel Sandoval Vallarta, Universidad Autonoma de San Luis Potos, San Luis Potosi, Mexico
| | - Mónica Raquel Calera
- 1 Instituto de Fisica Manuel Sandoval Vallarta, Universidad Autonoma de San Luis Potos, San Luis Potosi, Mexico
| |
Collapse
|
46
|
Wu T, Zhang Q, Hu H, Yang F, Li K, Zhang Y, Shi C. Enhancing cellular morphological changes and ablation of cancer cells via the interaction of drug co-loaded magnetic nanosystems in weak rotating magnetic fields. RSC Adv 2020; 10:14471-14481. [PMID: 35497149 PMCID: PMC9051889 DOI: 10.1039/d0ra01458c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 03/25/2020] [Indexed: 11/21/2022] Open
Abstract
Tetrandrine and Fe3O4 nanoparticle co-loaded PLGA nanosystems produce rotational movement and promote tetrandrine release, causing a dual apoptotic effect to tumors.
Collapse
Affiliation(s)
- Tingting Wu
- Department of Pharmacy
- Union Hospital
- Tongji Medical College
- Huazhong University of Science & Technology (HUST)
- Wuhan
| | - Qian Zhang
- Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education
- Department of Instrument Science and Engineering
- School of Electronic Information and Electrical Engineering
- Institute of Nano Biomedicine and Engineering
- Shanghai Jiao Tong University
| | - Huiping Hu
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation
- Tongji Medical College of Huazhong
- University of Science and Technology
- Wuhan
- China
| | - Fang Yang
- Cixi Institute of Biomedical Engineering
- CAS Key Laboratory of Magnetic Materials and Devices
- Key Laboratory of Additive Manufacturing Materials of Zhejiang Province
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
| | - Ke Li
- Department of Pharmacy
- Union Hospital
- Tongji Medical College
- Huazhong University of Science & Technology (HUST)
- Wuhan
| | - Yu Zhang
- Department of Pharmacy
- Union Hospital
- Tongji Medical College
- Huazhong University of Science & Technology (HUST)
- Wuhan
| | - Chen Shi
- Department of Pharmacy
- Union Hospital
- Tongji Medical College
- Huazhong University of Science & Technology (HUST)
- Wuhan
| |
Collapse
|
47
|
Santos A, Veiga F, Figueiras A. Dendrimers as Pharmaceutical Excipients: Synthesis, Properties, Toxicity and Biomedical Applications. MATERIALS (BASEL, SWITZERLAND) 2019; 13:E65. [PMID: 31877717 PMCID: PMC6981751 DOI: 10.3390/ma13010065] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/14/2019] [Accepted: 12/18/2019] [Indexed: 12/31/2022]
Abstract
The European Medicines Agency (EMA) and the Current Good Manufacturing Practices (cGMP) in the United States of America, define excipient as the constituents of the pharmaceutical form other than the active ingredient, i.e., any component that is intended to furnish pharmacological activity. Although dendrimers do not have a pharmacopoeia monograph and, therefore, cannot be recognized as a pharmaceutical excipient, these nanostructures have received enormous attention from researchers. Due to their unique properties, like the nanoscale uniform size, a high degree of branching, polyvalency, aqueous solubility, internal cavities, and biocompatibility, dendrimers are ideal as active excipients, enhancing the solubility of poorly water-soluble drugs. The fact that the dendrimer's properties are controllable during their synthesis render them promising agents for drug-delivery applications in several pharmaceutical formulations. Additionally, dendrimers can be used for reducing the drug toxicity and for the enhancement of the drug efficacy. This review aims to discuss the properties that turn dendrimers into pharmaceutical excipients and their potential applications in the pharmaceutical and biomedical fields.
Collapse
Affiliation(s)
- Ana Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3004-531 Coimbra, Portugal; (A.S.); (F.V.)
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3004-531 Coimbra, Portugal; (A.S.); (F.V.)
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3004-531 Coimbra, Portugal
| | - Ana Figueiras
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3004-531 Coimbra, Portugal; (A.S.); (F.V.)
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3004-531 Coimbra, Portugal
| |
Collapse
|
48
|
Wang T, Zhang J, Hou T, Yin X, Zhang N. Selective targeting of tumor cells and tumor associated macrophages separately by twin-like core-shell nanoparticles for enhanced tumor-localized chemoimmunotherapy. NANOSCALE 2019; 11:13934-13946. [PMID: 31305839 DOI: 10.1039/c9nr03374b] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Tumor associated macrophage (TAM)-based immunotherapy has been presented as a promising strategy in cancer therapy. The combination of TAM-based immunotherapy with sorafenib (SF) could be conceivably quite more effective in hepatocellular carcinoma (HCC) treatment. A co-delivery system was superior in improving the co-accumulation of two drugs in tumor tissues for chemoimmunotherapy, while in the case of selective targeting of separated cells such as tumor cells and immune cells, a novel targeted co-delivery strategy was badly required. In this study, twin-like core-shell nanoparticles (TCN) were developed for synchronous biodistribution and separated cell targeting delivery of SF and TAM re-polarization agents IMD-0354 to cancer cells and TAM to enhance tumor-localized chemoimmunotherapy, respectively. First of all, SF loaded cationic lipid-based nanoparticles (SF-CLN) and mannose-modified IMD-0354 loaded cationic lipid-based nanoparticles (M-IMD-CLN) were prepared, respectively. SF on the surface of SF-CLN and mannose on the M-IMD-CLN were regarded as targeting ligands for selective targeting delivery of SF-CLN and M-IMD-CLN to cancer cells and TAM separately. Then, pH-responsive charge reversal polymer O-carboxymethyl-chitosan (CMCS) was coated on the SF-CLN and M-IMD-CLN to obtain twin-like CMCS/SF-CLN and CMCS/M-IMD-CLN, respectively. The results of cellular uptake assay on Hepa1-6 cells and RAW 264.7 cells in vitro, respectively, as well as the results of tumor tissue distribution of SF and IMD-0354 in vivo suggested that CMCS/SF-CLN and CMCS/M-IMD-CLN exhibited similar properties in vitro and synchronous biodistribution in vivo, and were efficient at separated cell targeting delivery. What's more, the results of antitumor efficiency in vivo and phenotype analysis of TAM in tumor tissues proved that CMCS/SF-CLN and CMCS/M-IMD-CLN exhibited superior synergistic antitumor efficacy and M2-type TAM polarization ability compared with SF treatment in Hepa1-6 tumor bearing mice. Consequently, TCN which was the combination of co-administration and nano-drug delivery systems has great potential to be used in tumor-localized chemoimmunotherapy in clinics.
Collapse
Affiliation(s)
- Tianqi Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| | - Jing Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| | - Teng Hou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| | - Xiaolan Yin
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| |
Collapse
|
49
|
Farahavar G, Abolmaali SS, Gholijani N, Nejatollahi F. Antibody-guided nanomedicines as novel breakthrough therapeutic, diagnostic and theranostic tools. Biomater Sci 2019; 7:4000-4016. [PMID: 31355391 DOI: 10.1039/c9bm00931k] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Recent advances in nanotechnology, such as the development of various types of nanoparticles and hybrid nanomaterials, have revolutionized nanomedicine. The small size, customizable surface, enhanced solubility, and multi-functionality endow the nanoparticles with an ability to interact with complex cellular and biological functions in new ways. Furthermore, these systems can deliver drugs to specific tissues and provide a targeted therapy. For this purpose, different categories of molecules, particularly antibodies, have been used as ligands. Antibody-conjugated nanomaterials can significantly enhance the efficiency of nanomedicines, especially in the field of cancer. This review is focused on three major medical applications of antibody-conjugated nanomaterials, namely, therapeutic, diagnostic and theranostic applications. To provide comprehensive information on the topic and an overview of these hybrid nanomaterials for biomedical applications, a brief summary of nanomaterials and antibodies is given. Moreover, the review has depicted the potential applications of antibody-conjugated nanomaterials in different fields and their capabilities to empower nanomedicine, particularly in relation to the treatment and detection of malignancies.
Collapse
Affiliation(s)
- Ghazal Farahavar
- Pharmaceutical Nanotechnology Department, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Samira Sadat Abolmaali
- Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| | - Nasser Gholijani
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Foroogh Nejatollahi
- Shiraz HIV/AIDS research center, Institute of health, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
50
|
Tangeretin-Assisted Platinum Nanoparticles Enhance the Apoptotic Properties of Doxorubicin: Combination Therapy for Osteosarcoma Treatment. NANOMATERIALS 2019; 9:nano9081089. [PMID: 31362420 PMCID: PMC6723885 DOI: 10.3390/nano9081089] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/25/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022]
Abstract
Osteosarcoma (OS) is the most common type of cancer and the most frequent malignant bone tumor in childhood and adolescence. Nanomedicine has become an indispensable field in biomedical and clinical research, with nanoparticles (NPs) promising to increase the therapeutic efficacy of anticancer drugs. Doxorubicin (DOX) is a commonly used chemotherapeutic drug against OS; however, it causes severe side effects that restrict its clinical applications. Here, we investigated whether combining platinum NPs (PtNPs) and DOX could increase their anticancer activity in human bone OS epithelial cells (U2OS). PtNPs with nontoxic, effective, thermally stable, and thermoplasmonic properties were synthesized and characterized using tangeretin. We examined the combined effects of PtNPs and DOX on cell viability, proliferation, and morphology, reactive oxygen species (ROS) generation, lipid peroxidation, nitric oxide, protein carbonyl content, antioxidants, mitochondrial membrane potential (MMP), adenosine tri phosphate (ATP) level, apoptotic and antiapoptotic gene expression, oxidative stress-induced DNA damage, and DNA repair genes. PtNPs and DOX significantly inhibited U2OS viability and proliferation in a dose-dependent manner, increasing lactate dehydrogenase leakage, ROS generation, and malondialdehyde, nitric oxide, and carbonylated protein levels. Mitochondrial dysfunction was confirmed by reduced MMP, decreased ATP levels, and upregulated apoptotic/downregulated antiapoptotic gene expression. Oxidative stress was a major cause of cytotoxicity and genotoxicity, confirmed by decreased levels of various antioxidants. Furthermore, PtNPs and DOX increased 8-oxo-dG and 8-oxo-G levels and induced DNA damage and repair gene expression. Combination of cisplatin and DOX potentially induce apoptosis comparable to PtNPs and DOX. To the best of our knowledge, this is the first report to describe the combined effects of PtNPs and DOX in OS.
Collapse
|