1
|
Li R, Yu J, Ye D, Liu S, Zhang H, Lin H, Feng J, Deng K. Conotoxins: Classification, Prediction, and Future Directions in Bioinformatics. Toxins (Basel) 2025; 17:78. [PMID: 39998095 PMCID: PMC11860864 DOI: 10.3390/toxins17020078] [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: 12/28/2024] [Revised: 01/25/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Conotoxins, a diverse family of disulfide-rich peptides derived from the venom of Conus species, have gained prominence in biomedical research due to their highly specific interactions with ion channels, receptors, and neurotransmitter systems. Their pharmacological properties make them valuable molecular tools and promising candidates for therapeutic development. However, traditional conotoxin classification and functional characterization remain labor-intensive, necessitating the increasing adoption of computational approaches. In particular, machine learning (ML) techniques have facilitated advancements in sequence-based classification, functional prediction, and de novo peptide design. This review explores recent progress in applying ML and deep learning (DL) to conotoxin research, comparing key databases, feature extraction techniques, and classification models. Additionally, we discuss future research directions, emphasizing the integration of multimodal data and the refinement of predictive frameworks to enhance therapeutic discovery.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kejun Deng
- The Clinical Hospital of Chengdu Brain Science Institute, School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China; (R.L.); (J.Y.); (D.Y.); (S.L.); (H.Z.); (H.L.); (J.F.)
| |
Collapse
|
2
|
Shen X, Ma Y, Luo H, Abdullah R, Pan Y, Zhang Y, Zhong C, Zhang B, Zhang G. Peptide Aptamer-Paclitaxel Conjugates for Tumor Targeted Therapy. Pharmaceutics 2024; 17:40. [PMID: 39861688 PMCID: PMC11768741 DOI: 10.3390/pharmaceutics17010040] [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: 11/13/2024] [Revised: 12/23/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Traditional paclitaxel therapy often results in significant side effects due to its non-specific targeting of cancer cells. Peptide aptamer-paclitaxel conjugates present a promising alternative by covalently attaching paclitaxel to a versatile peptide aptamer via a linker. Compared to antibody-paclitaxel conjugates, peptide aptamer-paclitaxel conjugates offer several advantages, including a smaller size, lower immunogenicity, improved tissue penetration, and easier engineering. Methods: This review provides an in-depth analysis of the multifunctional peptide aptamers in these conjugates, emphasizing their structural features, therapeutic efficacy, and challenges in clinical applications. Results: This analysis highlights the potential of peptide aptamer-paclitaxel conjugates as a novel and effective approach for targeted cancer therapy. By harnessing the unique properties of peptide aptamers, these conjugates demonstrate significant promise in improving drug delivery efficiency while reducing the adverse effects associated with traditional paclitaxel therapy. Conclusions: The incorporation of peptide aptamers into paclitaxel conjugates offers a promising pathway for developing more efficient and targeted cancer therapies. However, further research and clinical studies are essential to fully unlock the therapeutic potential of these innovative conjugates and enhance patient outcomes.
Collapse
Affiliation(s)
- Xinyang Shen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuan Ma
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (Y.M.)
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Hang Luo
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (Y.M.)
| | - Razack Abdullah
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Yufei Pan
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Yihao Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (Y.M.)
| | - Chuanxin Zhong
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Baoting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; (Y.M.)
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone &Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| |
Collapse
|
3
|
Chary PS, Shaikh S, Rajana N, Bhavana V, Mehra NK. Unlocking nature's arsenal: Nanotechnology for targeted delivery of venom toxins in cancer therapy. BIOMATERIALS ADVANCES 2024; 162:213903. [PMID: 38824828 DOI: 10.1016/j.bioadv.2024.213903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/24/2024] [Accepted: 05/19/2024] [Indexed: 06/04/2024]
Abstract
AIM The aim of the present review is to shed light on the nanotechnological approaches adopted to overcome the shortcomings associated with the delivery of venom peptides which possess inherent anti-cancer properties. BACKGROUND Venom peptides although have been reported to demonstrate anti-cancer effects, they suffer from several disadvantages such as in vivo instability, off-target adverse effects, limited drug loading and low bioavailability. This review presents a comprehensive compilation of different classes of nanocarriers while underscoring their advantages, disadvantages and potential to carry such peptide molecules for in vivo delivery. It also discusses various nanotechnological aspects such as methods of fabrication, analytical tools to assess these nanoparticulate formulations, modulation of nanocarrier polymer properties to enhance loading capacity, stability and improve their suitability to carry toxic peptide drugs. CONCLUSION Nanotechnological approaches bear great potential in delivering venom peptide-based molecules as anticancer agents by enhancing their bioavailability, stability, efficacy as well as offering a spatiotemporal delivery approach. However, the challenges associated with toxicity and biocompatibility of nanocarriers must be duly addressed. PERSPECTIVES The everlasting quest for new breakthroughs for safer delivery of venom peptides in human subjects is fuelled by unmet clinical needs in the current landscape of chemotherapy. In addition, exhaustive efforts are required in obtaining and purifying the venom peptides followed by designing and optimizing scale up technologies.
Collapse
Affiliation(s)
- Padakanti Sandeep Chary
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Samia Shaikh
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Naveen Rajana
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Valamla Bhavana
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India.
| |
Collapse
|
4
|
Sun X, Hu J, Ren M, Chang H, Zhangsun D, Zhang B, Dong S. Stapling Cysteine[2,4] Disulfide Bond of α-Conotoxin LsIA and Its Potential in Target Delivery. Mar Drugs 2024; 22:314. [PMID: 39057423 PMCID: PMC11278161 DOI: 10.3390/md22070314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
α-Conotoxins, as selective nAChR antagonists, can be valuable tools for targeted drug delivery and fluorescent labeling, while conotoxin-drug or conotoxin-fluorescent conjugates through the disulfide bond are rarely reported. Herein, we demonstrate the [2,4] disulfide bond of α-conotoxin as a feasible new chemical modification site. In this study, analogs of the α-conotoxin LsIA cysteine[2,4] were synthesized by stapling with five linkers, and their inhibitory activities against human α7 and rat α3β2 nAChRs were maintained. To further apply this method in targeted delivery, the alkynylbenzyl bromide linker was synthesized and conjugated with Coumarin 120 (AMC) and Camptothecin (CPT) by copper-catalyzed click chemistry, and then stapled between cysteine[2,4] of the LsIA to construct a fluorescent probe and two peptide-drug conjugates. The maximum emission wavelength of the LsIA fluorescent probe was 402.2 nm, which was essentially unchanged compared with AMC. The cytotoxic activity of the LsIA peptide-drug conjugates on human A549 was maintained in vitro. The results demonstrate that the stapling of cysteine[2,4] with alkynylbenzyl bromide is a simple and feasible strategy for the exploitation and utilization of the α-conotoxin LsIA.
Collapse
Affiliation(s)
- Xin Sun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.S.); (J.H.); (M.R.); (D.Z.)
| | - Jiangnan Hu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.S.); (J.H.); (M.R.); (D.Z.)
| | - Maomao Ren
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.S.); (J.H.); (M.R.); (D.Z.)
| | - Hong Chang
- Hainan Academy of Inspection and Testing, Haikou 570228, China;
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.S.); (J.H.); (M.R.); (D.Z.)
| | - Baojian Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.S.); (J.H.); (M.R.); (D.Z.)
| | - Shuai Dong
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; (X.S.); (J.H.); (M.R.); (D.Z.)
| |
Collapse
|
5
|
Chen X, Zou Z, Li W, Dong X, Chen Y, Lu Y, Zhu M, Li M, Lin B. α-Conotoxin recombinant protein ImI-AFP3 efficiently inhibits the growth and migration of lung cancer cells. Protein Expr Purif 2024; 215:106405. [PMID: 37979629 DOI: 10.1016/j.pep.2023.106405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/11/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
α-Conotoxin ImI is a selective antagonist of alpha7 nicotinic acetylcholine receptor (α7 nAChR) that is involved in cancer development. Human alpha fetoprotein domain 3 (AFP3) is a prototype of anticancer agents. In an effort to design drugs for anticancer treatments, we fused the ImI peptide to AFP3 as a fusion protein for testing. The fusion protein (ImI-AFP3) was highly expressed in the insect Bac-to-Bac system. The purified fusion protein was found to have improved anticancer activity and synergized with the drug gefitinib to inhibit the growth and migration of A549 and NCI-H1299 lung cancer cells. Our data have demonstrated that the recombinant protein ImI-AFP3 is a promising candidate for drug development to suppress lung cancer cell growth, especially to suppress hepatoid adenocarcinoma of the lung (HAL) cell growth.
Collapse
Affiliation(s)
- Xiaobing Chen
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan Province, PR China
| | - Zijuan Zou
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan Province, PR China
| | - Wei Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan Province, PR China
| | - Xu Dong
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan Province, PR China
| | - Yi Chen
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan Province, PR China
| | - Yan Lu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan Province, PR China
| | - Mingyue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan Province, PR China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan Province, PR China; Institution of Tumor, Hainan Medical University, Haikou, 570102, Hainan Province, PR China.
| | - Bo Lin
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical University, Haikou, 571199, Hainan Province, PR China.
| |
Collapse
|
6
|
Gu J, You J, Liang H, Zhan J, Gu X, Zhu Y. Engineered bone marrow mesenchymal stem cell-derived exosomes loaded with miR302 through the cardiomyocyte specific peptide can reduce myocardial ischemia and reperfusion (I/R) injury. J Transl Med 2024; 22:168. [PMID: 38368334 PMCID: PMC10874538 DOI: 10.1186/s12967-024-04981-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/12/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND MicroRNA (miRNA)-based therapies have shown great potential in myocardial repair following myocardial infarction (MI). MicroRNA-302 (miR302) has been reported to exert a protective effect on MI. However, miRNAs are easily degraded and ineffective in penetrating cells, which limit their clinical applications. Exosomes, which are small bioactive molecules, have been considered as an ideal vehicle for miRNAs delivery due to their cell penetration, low immunogenicity and excellent stability potential. Herein, we explored cardiomyocyte-targeting exosomes as vehicles for delivery of miR302 into cardiomyocyte to potentially treat MI. METHODS To generate an efficient exosomal delivery system that can target cardiomyocytes, we engineered exosomes with cardiomyocyte specific peptide (CMP, WLSEAGPVVTVRALRGTGSW). Afterwards, the engineered exosomes were characterized and identified using transmission electron microscope (TEM) and Nanoparticle Tracking Analysis (NTA). Later on, the miR302 mimics were loaded into the engineered exosomes via electroporation technique. Subsequently, the effect of the engineered exosomes on myocardial ischemia and reperfusion (I/R) injury was evaluated in vitro and in vivo, including MTT, ELISA, real-time quantitative polymerase chain reaction (PCR), western blot, TUNNEL staining, echocardiogram and hematoxylin and eosin (HE) staining. RESULTS Results of in vitro experimentation showed that DSPE-PEG-CMP-EXO could be more efficiently internalized by H9C2 cells than unmodified exosomes (blank-exosomes). Importantly, compared with the DSPE-PEG-CMP-EXO group, DSPE-PEG-CMP-miR302-EXO significantly upregulated the expression of miR302, while exosomes loaded with miR302 could enhance proliferation of H9C2 cells. Western blot results showed that the DSPE-PEG-CMP-miR302-EXO significantly increased the protein level of Ki67 and Yap, which suggests that DSPE-PEG-CMP-miR302-EXO enhanced the activity of Yap, the principal downstream effector of Hippo pathway. In vivo, DSPE-PEG-CMP-miR302-EXO improved cardiac function, attenuated myocardial apoptosis and inflammatory response, as well as reduced infarct size significantly. CONCLUSION In conclusion, our findings suggest that CMP-engineered exosomes loaded with miR302 was internalized by H9C2 cells, an in vitro model for cardiomyocytes coupled with potential enhancement of the therapeutic effects on myocardial I/R injury.
Collapse
Affiliation(s)
- Jianjun Gu
- Department of Cardiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Cardiology, Northern Jiangsu People's Hospital, 98 Nantong West Road, Yangzhou, Jiangsu, China
| | - Jia You
- Department of Internal Medicine, Yangzhou Maternal and Child Health Care Hospital, Yangzhou, 225001, Jiangsu, China
| | - Hao Liang
- Department of Cardiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Cardiology, Northern Jiangsu People's Hospital, 98 Nantong West Road, Yangzhou, Jiangsu, China
| | - Jiacai Zhan
- Department of Cardiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Cardiology, Northern Jiangsu People's Hospital, 98 Nantong West Road, Yangzhou, Jiangsu, China
| | - Xiang Gu
- Department of Cardiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Cardiology, Northern Jiangsu People's Hospital, 98 Nantong West Road, Yangzhou, Jiangsu, China
| | - Ye Zhu
- Department of Cardiology, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.
- Department of Cardiology, Northern Jiangsu People's Hospital, 98 Nantong West Road, Yangzhou, Jiangsu, China.
| |
Collapse
|
7
|
Abdelmessih R, Xu J, Hung FR, Auguste DT. Integration of an LPAR1 Antagonist into Liposomes Enhances Their Internalization and Tumor Accumulation in an Animal Model of Human Metastatic Breast Cancer. Mol Pharm 2023; 20:5500-5514. [PMID: 37844135 PMCID: PMC10631474 DOI: 10.1021/acs.molpharmaceut.3c00348] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
Lysophosphatidic acid receptor 1 (LPAR1) is elevated in breast cancer. The deregulation of LPAR1, including the function and level of expression, is linked to cancer initiation, progression, and metastasis. LPAR1 antagonists, AM095 or Ki16425, may be effective therapeutic molecules, yet their limited water solubility hinders in vivo delivery. In this study, we report on the synthesis of two liposomal formulations incorporating AM095 or Ki16425, embedded within the lipid bilayer, as targeted nanocarriers for metastatic breast cancer (MBC). The data show that the Ki16425 liposomal formulation exhibited a 50% increase in internalization by MBC mouse epithelial cells (4T1) and a 100% increase in tumor accumulation in a mouse model of MBC compared with that of a blank liposomal formulation (control). At the same time, normal mouse epithelial cells (EpH-4Ev) internalized the Ki16425 liposomal formulation 25% lesser than the control formulation. Molecular dynamics simulations show that the integration of AM095 or Ki16425 modified the physical and mechanical properties of the lipid bilayer, making it more flexible in these liposomal formulations compared with liposomes without drug. The incorporation of an LPAR1 antagonist within a liposomal drug delivery system represents a viable therapeutic approach for targeting the LPA-LPAR1 axis, which may hinder the progression of MBC.
Collapse
Affiliation(s)
- Rudolf
G. Abdelmessih
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Jiaming Xu
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Francisco R. Hung
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Debra T. Auguste
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
8
|
Tae HS, Adams DJ. Nicotinic acetylcholine receptor subtype expression, function, and pharmacology: Therapeutic potential of α-conotoxins. Pharmacol Res 2023; 191:106747. [PMID: 37001708 DOI: 10.1016/j.phrs.2023.106747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
The pentameric nicotinic acetylcholine receptors (nAChRs) are typically classed as muscle- or neuronal-type, however, the latter has also been reported in non-neuronal cells. Given their broad distribution, nAChRs mediate numerous physiological and pathological processes including synaptic transmission, presynaptic modulation of transmitter release, neuropathic pain, inflammation, and cancer. There are 17 different nAChR subunits and combinations of these subunits produce subtypes with diverse pharmacological properties. The expression and role of some nAChR subtypes have been extensively deciphered with the aid of knock-out models. Many nAChR subtypes expressed in heterologous systems are selectively targeted by the disulfide-rich α-conotoxins. α-Conotoxins are small peptides isolated from the venom of cone snails, and a number of them have potential pharmaceutical value.
Collapse
|
9
|
Zhang B, Ren M, Yang F, Li R, Yu L, Luo A, Zhangsun D, Luo S, Dong S. Oligo-basic amino acids, potential nicotinic acetylcholine receptor inhibitors. Biomed Pharmacother 2022; 152:113215. [PMID: 35667234 DOI: 10.1016/j.biopha.2022.113215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/21/2022] [Accepted: 05/26/2022] [Indexed: 11/26/2022] Open
Abstract
Oligo-basic amino acids have been extensively studied in molecular biology and pharmacology, but the inhibitory activity on nicotinic acetylcholine receptors (nAChRs) was unknown. In this study, the inhibitory activity of 8 oligopeptides, including both basic and acidic amino acids, was evaluated on 9 nAChR subtypes by a two-electrode voltage clamp (TEVC). Among them, the oligo-lysine K9, K12, d-K9, d-K9F, and oligo-arginine R9 showed nanomolar inhibitory activity on various nAChRs, especially for α7 and α9α10 nAChRs. d-K9 containing N-Fmoc protecting group (d-K9F) has an enhanced inhibitory activity on most of the nAChRs, including 47-fold promotion on α1β1δε nAChR. However, H9 and H12 only showed weak inhibitory activity on α9α10 and α1β1δε nAChRs, and the acidic oligopeptide D9 has no inhibitory activity on nAChRs. Flexible docking of K9 in α10(+) α9(-) and α7(+) α7(-) binding pockets showed particularly strong dipole-dipole interactions, which may be responsible for the inhibition of nAChRs. These results demonstrated that oligo-basic amino acids have the potential to be the lead compounds as selective nAChR subtype inhibitors, and oligo-lysines deserved to be modified for further exploitation and utilization. On the other hand, the toxicity and side effects of these nAChR inhibitory peptides should be contemplated in the application.
Collapse
Affiliation(s)
- Baojian Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Maomao Ren
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Fang Yang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Rui Li
- School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Liutong Yu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - An Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Sulan Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Shuai Dong
- Key Laboratory of Tropical Biological Resources of Ministry of Education, Key Laboratory for Marine Drugs of Haikou, School of Pharmaceutical Sciences, Hainan University, Haikou, China.
| |
Collapse
|
10
|
Joglekar AV, Dehari D, Anjum MM, Dulla N, Chaudhuri A, Singh S, Agrawal AK. Therapeutic potential of venom peptides: insights in the nanoparticle-mediated venom formulations. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-022-00415-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Abstract
Background
Venoms are the secretions produced by animals, generally for the purpose of self-defense or catching a prey. Biochemically venoms are mainly composed of proteins, lipids, carbohydrates, ions, etc., and classified into three major classes, viz. neurotoxic, hemotoxic and cytotoxic based upon their mode of action. Venoms are composed of different specific peptides/toxins which are responsible for their unique biological actions. Though venoms are generally seen as a source of death, scientifically venom is a complex biochemical substance having a specific pharmacologic action which can be used as agents to diagnose and cure a variety of diseases in humans.
Main body
Many of these venoms have been used since centuries, and their specified therapies can also be found in ancient texts such as Charka Samhita. The modern-day example of such venom therapeutic is captopril, an antihypertensive drug developed from venom of Bothrops jararaca. Nanotechnology is a modern-day science of building materials on a nanoscale with advantages like target specificity, increased therapeutic response and diminished side effects. In the present review we have introduced the venom, sources and related constituents in brief, by highlighting the therapeutic potential of venom peptides and focusing more on the nanoformulations-based approaches. This review is an effort to compile all such report to have an idea about the future direction about the nanoplatforms which should be focused to have more clinically relevant formulations for difficult to treat diseases.
Conclusion
Venom peptides which are fatal in nature if used cautiously and effectively can save life. Several research findings suggested that many of the fatal diseases can be effectively treated with venom peptides. Nanotechnology has emerged as novel strategy in diagnosis, treatment and mitigation of diseases in more effective ways. A variety of nanoformulation approaches have been explored to enhance the therapeutic efficacy and reduce the toxicity and targeted delivery of the venom peptide conjugated with it. We concluded that venom peptides along with nanoparticles can evolve as the new era for potential treatments of ongoing and untreatable diseases.
Graphical Abstract
Collapse
|
11
|
Chrna5 is overexpressed in psoriasis patients and promotes psoriasis-like inflammation in mouse models. J Invest Dermatol 2022; 142:2978-2987.e6. [PMID: 35513071 DOI: 10.1016/j.jid.2022.04.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 11/20/2022]
Abstract
It is well known that psoriasis is closely related to smoking, and the cholinergic receptor nicotinic subunit alpha-5 (Chrna5) plays an important role in smoking-related diseases. However, studies on the relationship between Chrna5 and psoriasis are limited. This study aimed to examine the role of Chrna5 in psoriasis development and pathogenesis. Analysis in psoriatic tissues and imiquimod (IMQ)-induced mouse models showed that Chrna5 was highly expressed in psoriatic lesional skin. To further verify the function of Chrna5, we constructed Chrna5-knockout mice and induced the psoriasis model. We found that Chrna5 knockout significantly reduced the severity of psoriasis and could regulate inflammation via the mitogen-activated protein kinase kinase kinase 1 (MEKK1)/c-Jun NH(2)-terminal kinase (JNK)-MAPK/NF-κB pathway. The single-cell sequencing results revealed that after Chrna5 knockout, the keratinocyte subpopulation was significantly reduced and the related Janus kinase/signal transduction and activator of transcription (JAK/STAT) signaling pathway was downregulated, further indicating the importance of Chrna5 in psoriasis. Human keratinocytes were analyzed, and silencing Chrna5 inhibited keratinocyte proliferation and migration. In summary, Chrna5 played important roles in the development and pathogenesis of psoriasis, and targeting Chrna5 may be an effective strategy for the treatment of psoriasis.
Collapse
|
12
|
Nicotine treatment regulates PD-L1 and PD-L2 expression via inhibition of Akt pathway in HER2-type breast cancer cells. PLoS One 2022; 17:e0260838. [PMID: 35085258 PMCID: PMC8794171 DOI: 10.1371/journal.pone.0260838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023] Open
Abstract
The immune checkpoint molecules such as PD-L1 and PD-L2 have a substantial contribution to cancer immunotherapy including breast cancer. Microarray expression profiling identified several molecular subtypes, namely luminal-type (with a good-prognosis), HER2-type (with an intermediate-prognosis), and triple-negative breast cancer (TNBC)-type (with a poor-prognosis). We found that PD-L1 and PD-L2 mRNA expressions were highly expressed in TNBC-type cell lines (HCC1937, MDA-MB-231), moderately expressed in HER2-type cell line (SK-BR-3), and poorly expressed in luminal-type cell lines (MDA-MB-361, MCF7). The PD-L1 and PD-L2 expression in SK-BR-3 cells, but not those in HCC1937 and MDA-MB-231 cells, decreased by nicotine stimulation in a dose-dependent manner. In addition, nicotine treatment decreased the phosphorylation of Akt in SK-BR-3 cells, but not in other cell lines. These results show that nicotine regulates the expression of immune checkpoint molecules, PD-L1 and PD-L2, via inhibition of Akt phosphorylation. This findings may provide the new therapeutic strategies for the treatment of breast cancer.
Collapse
|
13
|
Rizwanullah M, Ahmad MZ, Ghoneim MM, Alshehri S, Imam SS, Md S, Alhakamy NA, Jain K, Ahmad J. Receptor-Mediated Targeted Delivery of Surface-ModifiedNanomedicine in Breast Cancer: Recent Update and Challenges. Pharmaceutics 2021; 13:2039. [PMID: 34959321 PMCID: PMC8708551 DOI: 10.3390/pharmaceutics13122039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
Breast cancer therapeutic intervention continues to be ambiguous owing to the lack of strategies for targeted transport and receptor-mediated uptake of drugs by cancer cells. In addition to this, sporadic tumor microenvironment, prominent restrictions with conventional chemotherapy, and multidrug-resistant mechanisms of breast cancer cells possess a big challenge to even otherwise optimal and efficacious breast cancer treatment strategies. Surface-modified nanomedicines can expedite the cellular uptake and delivery of drug-loaded nanoparticulate constructs through binding with specific receptors overexpressed aberrantly on the tumor cell. The present review elucidates the interesting yet challenging concept of targeted delivery approaches by exploiting different types of nanoparticulate systems with multiple targeting ligands to target overexpressed receptors of breast cancer cells. The therapeutic efficacy of these novel approaches in preclinical models is also comprehensively discussed in this review. It is concluded from critical analysis of related literature that insight into the translational gap between laboratories and clinical settings would provide the possible future directions to plug the loopholes in the process of development of these receptor-targeted nanomedicines for the treatment of breast cancer.
Collapse
Affiliation(s)
- Md. Rizwanullah
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
| | - Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia;
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia;
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (S.S.I.)
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.M.); (N.A.A.)
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.M.); (N.A.A.)
| | - Keerti Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)—Raebareli, Lucknow 226002, India;
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 11001, Saudi Arabia;
| |
Collapse
|
14
|
Clarke MR, Jones B, Squires CLM, Imhoff FM, Harwood DT, Rhodes L, Selwood AI, McNabb PS, Baird SK. Cyclic Imine Pinnatoxin G is Cytotoxic to Cancer Cell Lines via Nicotinic Acetylcholine Receptor-Driven Classical Apoptosis. JOURNAL OF NATURAL PRODUCTS 2021; 84:2035-2042. [PMID: 34170700 DOI: 10.1021/acs.jnatprod.1c00418] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Pinnatoxin G is a cyclic imine neurotoxin produced by dinoflagellates that has been reported in shellfish. Like other members of the pinnatoxin family, it has been shown to have its effects via antagonism of the nicotinic acetylcholine receptors, with preferential binding to the α7 subunit often upregulated in cancer. Because increased activity of α7 nicotinic acetylcholine receptors contributes to increased growth and resistance to apoptosis, the effect of pinnatoxin G on cancer cell viability was tested. In a panel of six cancer cell lines, all cell types lost viability, but HT29 colon cancer and LN18 and U373 glioma cell lines were more sensitive than MDA-MB-231 breast cancer cells, PC3 prostate cancer cells, and U87 glioma cells, correlating with expression levels of α7, α4, and α9 nicotinic acetylcholine receptors. Some loss of cell viability could be attributed to cell cycle arrest, but significant levels of classical apoptosis were found, characterized by caspase activity, phosphatidylserine exposure, mitochondrial membrane permeability, and fragmented DNA. Intracellular Ca2+ levels also dropped immediately upon pinnatoxin G treatment, which may relate to antagonism of nicotinic acetylcholine receptor-mediated Ca2+ inflow. In conclusion, pinnatoxin G can decrease cancer cell viability, with both cytostatic and cytotoxic effects.
Collapse
Affiliation(s)
- Mitchell R Clarke
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin 9016, New Zealand
| | - Ben Jones
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin 9016, New Zealand
| | - Chloe L M Squires
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin 9016, New Zealand
| | - Floriane M Imhoff
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin 9016, New Zealand
| | - D Tim Harwood
- Cawthron Institute, Private Bag 2, Nelson 7040, New Zealand
| | - Lesley Rhodes
- Cawthron Institute, Private Bag 2, Nelson 7040, New Zealand
| | | | - Paul S McNabb
- Cawthron Institute, Private Bag 2, Nelson 7040, New Zealand
| | - Sarah K Baird
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin 9016, New Zealand
| |
Collapse
|
15
|
Afrashteh Nour M, Hajiasgharzadeh K, Kheradmand F, Asadzadeh Z, Bolandi N, Baradaran B. Nicotinic acetylcholine receptors in chemotherapeutic drugs resistance: An emerging targeting candidate. Life Sci 2021; 278:119557. [PMID: 33930371 DOI: 10.1016/j.lfs.2021.119557] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022]
Abstract
There is no definitive cure for cancer, and most of the current chemotherapy drugs have limited effects due to the development of drug resistance and toxicity at high doses. Therefore, there is an ongoing need for identifying the causes of chemotherapeutic resistance, and it will be possible to develop innovative treatment approaches based on these novel targeting candidates. Cigarette smoking is known to be one of the main causes of resistance to chemotherapeutic agents. Nicotine as a component of cigarette smoke is an exogenous activator of nicotinic acetylcholine receptors (nAChRs). It can inhibit apoptosis, increase cell proliferation and cell survival, reducing the cytotoxic effects of chemotherapy drugs and cause a reduced therapeutic response. Recent studies have demonstrated that nAChRs and their downstream signaling pathways have considerable implications in different cancer's initiation, progression, and chemoresistance. In some previous studies, nAChRs have been targeted to obtain better efficacies for chemotherapeutics. Besides, nAChRs-based therapies have been used in combination with chemotherapy drugs to reduce the side effects. This strategy requires lower doses of chemotherapy drugs compared to the conditions that must be used alone. Here, we discussed the experimental and clinical studies that show the nAChRs involvement in response to chemotherapy agents. Also, controversies relating to the effects of nAChR on chemotherapy-induced apoptosis are in our focus in this review article. Delineating the complex influences of nAChRs would be of great interest in establishing new effective chemotherapy regimens.
Collapse
Affiliation(s)
- Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Khalil Hajiasgharzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Kheradmand
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadia Bolandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
16
|
Alven S, Aderibigbe BA. The Therapeutic Efficacy of Dendrimer and Micelle Formulations for Breast Cancer Treatment. Pharmaceutics 2020; 12:E1212. [PMID: 33333778 PMCID: PMC7765183 DOI: 10.3390/pharmaceutics12121212] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is among the most common types of cancer in women and it is the cause of a high rate of mortality globally. The use of anticancer drugs is the standard treatment approach used for this type of cancer. However, most of these drugs are limited by multi-drug resistance, drug toxicity, poor drug bioavailability, low water solubility, poor pharmacokinetics, etc. To overcome multi-drug resistance, combinations of two or more anticancer drugs are used. However, the combination of two or more anticancer drugs produce toxic side effects. Micelles and dendrimers are promising drug delivery systems that can overcome the limitations associated with the currently used anticancer drugs. They have the capability to overcome drug resistance, reduce drug toxicity, improve the drug solubility and bioavailability. Different classes of anticancer drugs have been loaded into micelles and dendrimers, resulting in targeted drug delivery, sustained drug release mechanism, increased cellular uptake, reduced toxic side effects of the loaded drugs with enhanced anticancer activity in vitro and in vivo. This review article reports the biological outcomes of dendrimers and micelles loaded with different known anticancer agents on breast cancer in vitro and in vivo.
Collapse
Affiliation(s)
| | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa;
| |
Collapse
|
17
|
Bjørn-Yoshimoto WE, Ramiro IBL, Yandell M, McIntosh JM, Olivera BM, Ellgaard L, Safavi-Hemami H. Curses or Cures: A Review of the Numerous Benefits Versus the Biosecurity Concerns of Conotoxin Research. Biomedicines 2020; 8:E235. [PMID: 32708023 PMCID: PMC7460000 DOI: 10.3390/biomedicines8080235] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/17/2020] [Accepted: 07/19/2020] [Indexed: 01/18/2023] Open
Abstract
Conotoxins form a diverse group of peptide toxins found in the venom of predatory marine cone snails. Decades of conotoxin research have provided numerous measurable scientific and societal benefits. These include their use as a drug, diagnostic agent, drug leads, and research tools in neuroscience, pharmacology, biochemistry, structural biology, and molecular evolution. Human envenomations by cone snails are rare but can be fatal. Death by envenomation is likely caused by a small set of toxins that induce muscle paralysis of the diaphragm, resulting in respiratory arrest. The potency of these toxins led to concerns regarding the potential development and use of conotoxins as biological weapons. To address this, various regulatory measures have been introduced that limit the use and access of conotoxins within the research community. Some of these regulations apply to all of the ≈200,000 conotoxins predicted to exist in nature of which less than 0.05% are estimated to have any significant toxicity in humans. In this review we provide an overview of the many benefits of conotoxin research, and contrast these to the perceived biosecurity concerns of conotoxins and research thereof.
Collapse
Affiliation(s)
- Walden E. Bjørn-Yoshimoto
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; (W.E.B.-Y.); (I.B.L.R.)
| | - Iris Bea L. Ramiro
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; (W.E.B.-Y.); (I.B.L.R.)
| | - Mark Yandell
- Eccles Institute of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA;
- Utah Center for Genetic Discovery, University of Utah, Salt Lake City, UT 84112, USA
| | - J. Michael McIntosh
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA; (J.M.M.); (B.M.O.)
- George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA
- Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA
| | - Baldomero M. Olivera
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA; (J.M.M.); (B.M.O.)
| | - Lars Ellgaard
- Department of Biology, Linderstrøm-Lang Centre for Protein Science, University of Copenhagen, 2200 Copenhagen N, Denmark;
| | - Helena Safavi-Hemami
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark; (W.E.B.-Y.); (I.B.L.R.)
- School of Biological Sciences, University of Utah, Salt Lake City, UT 84112, USA; (J.M.M.); (B.M.O.)
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
18
|
Li Z, Zhang Y, Zhu C, Guo T, Xia Q, Hou X, Liu W, Feng N. Folic acid modified lipid-bilayer coated mesoporous silica nanoparticles co-loading paclitaxel and tanshinone IIA for the treatment of acute promyelocytic leukemia. Int J Pharm 2020; 586:119576. [PMID: 32603839 DOI: 10.1016/j.ijpharm.2020.119576] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/14/2020] [Accepted: 06/21/2020] [Indexed: 12/16/2022]
Abstract
In this work, paclitaxel (Ptx) combined with tanshinone IIA (TanIIA) was found to show synergistic effect on inducing apoptosis of human acute promyelocytic leukemia (APL) cell line NB4, and the anti-tumor effect was strongest when its molar ratio was 1:1. To enhance the efficacy and reduce side effects, an active targeting drug delivery system with mesoporous silica nanoparticles (MSNs) coated with folic acid (FA) modified PEGylated lipid-bilayer (LB) membrane (FA-LB-MSNs) was established for co-loading drugs. The drug loadings of Ptx and TanIIA in FA-LB-MSNs were 5.5% and 1.8%, respectively. Compared with the uncoated MSNs, the FA-LB-MSNs showed a sustained drug release, and Ptx and TanIIA released synchronously from the carriers. By means of biological adhesion between FA and its receptors, the uptake of FA-LB-MSNs by NB4 cells was significantly higher than that of uncoated preparations, and Ptx combined with TanIIA had strong synergistic effect to enhance the apoptosis and differentiation of NB4 cells. The results of pharmacodynamics in vivo showed that the FA-LB-MSNs targeted tumor in nude mice more effectively than the compared formulations without FA modification. The Ptx and TanIIA-loaded FA-LB-MSNs group showed significantly better effects on inducing apoptosis and inhibiting tumor growth than the reference groups, which agreed with the results of anti-tumor experiments in vitro. Furthermore, no toxicity was observed to the heart, liver, spleen, lung and kidney of the tumor-bearing animals, indicating good biocompatibility of the prepared novel nanocarriers. This study confirmed the synergistic therapeutic effect of Ptx and TanIIA on APL, and the superior of FA-LB-MSNs as co-loaded nanocarriers for active targeted therapy of tumors.
Collapse
Affiliation(s)
- Zhe Li
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Yongtai Zhang
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Chunyun Zhu
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Teng Guo
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Qing Xia
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Xuefeng Hou
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China
| | - Wei Liu
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Nianping Feng
- Department of Pharmaceutical Sciences School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park Pudong New District, Shanghai 201203, China.
| |
Collapse
|
19
|
He J, Xiao H, Li B, Peng Y, Li X, Wang Y, Adamus G, Kowalczuk M, Shuai X. The programmed site-specific delivery of the angiostatin sunitinib and chemotherapeutic paclitaxel for highly efficient tumor treatment. J Mater Chem B 2020; 7:4953-4962. [PMID: 31411627 DOI: 10.1039/c9tb01159e] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Malignant solid tumors are composed of tumor cells, stromal cells and the complex networks of the tumor microenvironment (TME), which is the underlying cause of the unsatisfactory outcome of conventional chemotherapy approaches only aimed at cancer cell killing. In this study, a novel TME-responsive polymeric micelle has been developed for the programmed site-specific delivery of the angiostatin sunitinib and chemotherapeutic paclitaxel (PTX). The pH-sensitive micelle core encapsulates PTX, while β-cyclodextrin molecules being conjugated to the micelle shell via matrix metalloproteinase 2 (MMP-2) sensitive peptides include sunitinib. Following the pH and MMP-2 dual sensitive structure design, the micelle may sequentially release sunitinib inside the tumor extracellular matrix and PTX into cancer cells through responding to enriched MMP-2 levels and decreased pH, respectively. Consequently, the anti-angiogenesis effect of sunitinib and tumor cell-killing effect of PTX synergize, resulting in highly efficient tumor treatment.
Collapse
Affiliation(s)
- Jin He
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| | - Hong Xiao
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| | - Bo Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| | - Yuan Peng
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| | - Xiaoxia Li
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| | - Yong Wang
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| | - Grazyna Adamus
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland.
| | - Marek Kowalczuk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Sklodowskiej 34, 41-819 Zabrze, Poland. and Wolverhampton School of Sciences, Faculty of Science and Engineering, University of Wolverhampton, Wulfruna Street, Wolverhampton, WV1 1LY, UK
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| |
Collapse
|
20
|
Xu J, Cui Z, Ge X, Luo Y, Xu F. Polymers prepared through an “ATRP polymerization–esterification” strategy for dual temperature- and reduction-induced paclitaxel delivery. RSC Adv 2020; 10:28891-28901. [PMID: 35520090 PMCID: PMC9055954 DOI: 10.1039/d0ra05422d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 07/21/2020] [Indexed: 11/21/2022] Open
Abstract
A dual temperature- and reduction-responsive nanovehicle with 29.36% paclitaxel loading was fabricated using an “ATRP polymerization–esterification” method for tumor suppression.
Collapse
Affiliation(s)
- JingWen Xu
- School of Food and Biological Engineering
- Shaanxi University of Science and Technology
- Xi'an 710021
- China
| | - ZhuoMiao Cui
- Key Laboratory of Macromolecular Science of Shaanxi Province
- School of Chemistry and Chemical Engineering
- Shaanxi Normal University
- Xi'an 710062
- China
| | - Xin Ge
- The First Affiliated Hospital of USTC
- Division of Life Science and Medicine
- University of Science and Technology of China
- Hefei
- China
| | - YanLing Luo
- Key Laboratory of Macromolecular Science of Shaanxi Province
- School of Chemistry and Chemical Engineering
- Shaanxi Normal University
- Xi'an 710062
- China
| | - Feng Xu
- Key Laboratory of Macromolecular Science of Shaanxi Province
- School of Chemistry and Chemical Engineering
- Shaanxi Normal University
- Xi'an 710062
- China
| |
Collapse
|
21
|
Surface engineering of nanomaterials with phospholipid-polyethylene glycol-derived functional conjugates for molecular imaging and targeted therapy. Biomaterials 2019; 230:119646. [PMID: 31787335 DOI: 10.1016/j.biomaterials.2019.119646] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 11/16/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
Abstract
In recent years, phospholipid-polyethylene glycol-derived functional conjugates have been widely employed to decorate different nanomaterials, due to their excellent biocompatibility, long blood circulation characteristics, and specific targeting capability. Numerous in vivo studies have demonstrated that nanomedicines peripherally engineered with phospholipid-polyethylene glycol-derived functional conjugates show significantly increased selective and efficient internalization by target cells/tissues. Targeting moieties including small-molecule ligands, peptides, proteins, and antibodies are generally conjugated onto PEGylated phospholipids to decorate liposomes, micelles, hybrid nanoparticles, nanocomplexes, and nanoemulsions for targeted delivery of diagnostic and therapeutic agents to diseased sites. In this review, the synthesis methods of phospholipid-polyethylene glycol-derived functional conjugates, biophysicochemical properties of nanomedicines decorated with these conjugates, factors dominating their targeting efficiency, as well as their applications for in vivo molecular imaging and targeted therapy were summarized and discussed.
Collapse
|
22
|
Identification of nicotinic acetylcholine receptor subunits in different lung cancer cell lines and the inhibitory effect of alpha-conotoxin TxID on lung cancer cell growth. Eur J Pharmacol 2019; 865:172674. [PMID: 31634461 DOI: 10.1016/j.ejphar.2019.172674] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 09/08/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022]
Abstract
Lung cancer is an aggressive tumor with high incidence and mortality rate. There was growing evidence supporting that nicotinic acetylcholine receptors (nAChRs) play vital role inlung cancer development. In this study, the expression of α3, α4, α5, α6, α7, α9, α10, β2, β3, β4 nAChR subunits on protein and mRNA level were studied in A549, NCI-H1299, NCI-H1688, DMS114 and normal human embryonic lung fibroblast (HEL) cell lines by real-time quantitative PCR (qPCR) and Western blot assay respectively. The results indicated that most of these nAChR subunits were expressed in these five cell lines. Compared with normal cells, the expression of α3 and β4 nAChR subunits were upregulated in A549 and NCI-H1299. Thus, we treated A549 and NCI-H1299 with an antagonist α-conotoxin TxID which potently and selectively blocks α3β4 nAChRs. TxID treatment could inhibit A549 and NCI-H1299 cell growth and enhance the inhibitory effect of adriamycin when treated simultaneously. To sum up, our study identified the expression of nAChR subunits in different lung cells and the anti-tumor effect of α-conotoxin TxID, which may provide novel strategies for lung cancer therapy.
Collapse
|
23
|
Li CL, Lin YK, Chen HA, Huang CY, Huang MT, Chang YJ. Smoking as an Independent Risk Factor for Hepatocellular Carcinoma Due to the α7-Nachr Modulating the JAK2/STAT3 Signaling Axis. J Clin Med 2019; 8:jcm8091391. [PMID: 31492006 PMCID: PMC6780871 DOI: 10.3390/jcm8091391] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a worldwide health problem. Currently, there is no effective clinical therapeutic strategy for HCC. Smoking is associated with several malignant diseases including cancers. EXPERIMENTAL APPROACH However, the impact of smoking on HCC is still unresolved. Retrospectively reviewed HCC patients diagnosed between 1 January 2010 and 31 December 2015 at Taipei Medical University-Shuang Ho Hospital (Ministry of Health and Welfare). We found that smoking was associated with a poor prognosis, especially recurrence and patient survival after curative surgery using a clinicopathological analysis. RESULTS Our univariate and multivariate analyses showed that the α7-nicotinic acetylcholine receptor (α7-nAChR) was an oncogene and risk factor for post-resection recurrence. The α7-nAChR was overexpressed in HCC tissues compared to their non-tumor counterparts. Silencing the α7-nAChR reduced the viability of HCC cells, suppressed cellular proliferation, attenuated migration and invasion, and diminished the tumor's sphere-formation ability, with concurrent downregulation of expression levels of the TGR5, p-JAK2, p-STAT3 (Tyr705/Ser727), RhoA, ROCK1, MMP2, and MMP9 proteins. Furthermore, a positive correlation was found between α7-nAChR and JAK2 expressions (p = 0.01) in HCC specimens, as well as their membranous co-localization. CONCLUSION Together, we demonstrated that the α7-nAChR may be an independent prognosticator of the progression and prognosis of HCC patients. These findings suggest that the α7-nAChR drives the progression and recurrence of HCC through JAK2/STAT3 signaling and is a novel target for anti-HCC therapy.
Collapse
Affiliation(s)
- Ching-Li Li
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Surgery, Sijhih Cathay General Hospital, New Taipei City 221, Taiwan.
| | - Yen-Kuang Lin
- Biostatistics Center, Taipei Medical University, Taipei 110, Taiwan.
| | - Hsin-An Chen
- Department of Surgery, Taipei Medical University, Shuang Ho Hospital, New Taipei City 235, Taiwan.
- Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Chien-Yu Huang
- Department of Surgery, Taipei Medical University, Shuang Ho Hospital, New Taipei City 235, Taiwan.
| | - Ming-Te Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Surgery, Taipei Medical University, Shuang Ho Hospital, New Taipei City 235, Taiwan.
- Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Yu-Jia Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- International PhD Program in Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan.
- Cancer Research Center and Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
24
|
Abstract
In order to improve stability of a peptide marine drug lead, α-conotoxin TxID, we synthesized and modified TxID at the N-terminal with DSPE-PEG-NHS by a nucleophilic substitution reaction to prepare the DSPE-PEG-TxID for the first time. The reaction conditions, including solvent, ratio, pH, and reaction time, were optimized systematically and the optimal one was reacted in dimethyl formamide at pH 8.2 with triethylamine at room temperature for 120 h. The in vitro stabilities in serum, simulated gastric juice, and intestinal fluid were tested, and improved dramatically compared with TxID. The PEG-modified peptide was functionally tested on α3β4 nicotinic acetylcholine receptor (nAChR) heterologously expressed in Xenopus laevis oocytes. The DSPE-PEG-TxID showed an obvious inhibition effect on α3β4 nAChR. All in all, the PEG modification of TxID was improved in stability, resistance to enzymatic degradation, and may prolong the half-life in vivo, which may pave the way for the future application in smoking cessation and drug rehabilitation, as well as small cell lung cancer.
Collapse
|
25
|
Yang Y, Wang Z, Peng Y, Ding J, Zhou W. A Smart pH-Sensitive Delivery System for Enhanced Anticancer Efficacy via Paclitaxel Endosomal Escape. Front Pharmacol 2019; 10:10. [PMID: 30733675 PMCID: PMC6353802 DOI: 10.3389/fphar.2019.00010] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/07/2019] [Indexed: 12/17/2022] Open
Abstract
Micelles are highly attractive nano-drug delivery systems for targeted cancer therapy. While they have been demonstrated to significantly alleviate the side-effects of their cargo drugs, the therapy outcomes are usually suboptimal partially due to ineffective drug release and endosome entrapment. Stimulus-responsive nanoparticles have allowed controlled drug release in a smart fashion, and we want to use this concept to design novel micelles. Herein, we reported pH-sensitive paclitaxel (PTX)-loaded poly (ethylene glycol)-phenylhydrazone-dilaurate (PEG-BHyd-dC12) micelles (PEG-BHyd-dC12/PTX). The micelles were spherical, with an average particle size of ∼135 nm and a uniform size distribution. The pH-responsive properties of the micelles were certified by both colloidal stability and drug release profile, where the particle size was strikingly increased accompanied by faster drug release as pH decreased from 7.4 to 5.5. As a result, the micelles exhibited much stronger cytotoxicity than the pH-insensitive counterpart micelles against various types of cancer cells due to the hydrolysis of the building block polymers and subsequent rapid PTX release. Overall, these results demonstrate that the PEG-BHyd-dC12 micelle is a promising drug delivery system for cancer therapy.
Collapse
Affiliation(s)
- Yihua Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmaceutical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Zhe Wang
- Xiangya International Academy of Translational Medicine, Central South University, Changsha, China
| | - Ying Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| |
Collapse
|
26
|
Raza F, Zafar H, You X, Khan A, Wu J, Ge L. Cancer nanomedicine: focus on recent developments and self-assembled peptide nanocarriers. J Mater Chem B 2019; 7:7639-7655. [DOI: 10.1039/c9tb01842e] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The applications of nanoparticulate drug delivery have received abundant interest in the field of cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics
| | - Hajra Zafar
- School of Pharmacy
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Xinru You
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong, Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Asifullah Khan
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing
- China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong, Province
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- P. R. China
| | - Liang Ge
- State Key Laboratory of Natural Medicines and Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing
- China
| |
Collapse
|
27
|
Kang SJ, Jeong HY, Kim MW, Jeong IH, Choi MJ, You YM, Im CS, Song IH, Lee TS, Park YS. Anti-EGFR lipid micellar nanoparticles co-encapsulating quantum dots and paclitaxel for tumor-targeted theranosis. NANOSCALE 2018; 10:19338-19350. [PMID: 30307008 DOI: 10.1039/c8nr05099f] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Cancer theranosis is an emerging field of personalized medicine which enables individual anti-cancer treatment by monitoring the therapeutic responses of cancer patients. Based on a consideration of the nano-bio interactions related to the blood circulation of systemically administered nanoparticles in humans, as well as extravasation and active targeting, lipid micellar nanoparticles were co-loaded with paclitaxel (PTX) and quantum dots (QDs) to generate a theranostic delivery vehicle. To provide with a tumor-targeting capability, either an antibody or an aptamer against the epidermal growth factor receptor (EGFR) was conjugated to the micelle surface. The QD-containing micelles (QDMs), antibody-coupled QDMs (immuno-QDMs), and aptamer-coupled QDMs (aptamo-QDMs) were able to effectively circulate in blood for at least 8 h when administered intravenously into mice bearing EGFR-positive LS174T tumor xenografts. In vivo fluorescence imaging and a bio-distribution study showed that both the immuno-QDMs and aptamo-QDMs were largely localized in the tumor tissue. The tumor targeting capability enhanced the therapeutic efficacy of PTX for the target cancer cells. Both the immuno-PTX-QDMs and the aptamo-PTX-QDMs caused a stronger inhibition of LS174T tumor growth in mice, compared to the non-targeted PTX-QDMs. These results suggest that the anti-EGFR immuno-PTX-QDMs and anti-EGFR aptamo-PTX-QDMs could be utilized as a tumor-targeted theranostic delivery system for cancer treatment in the clinic.
Collapse
Affiliation(s)
- Seong Jae Kang
- Department of Biomedical Laboratory Science, Yonsei University, Wonju, Republic of Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sun T, Zhan B, Zhang W, Qin D, Xia G, Zhang H, Peng M, Li SA, Zhang Y, Gao Y, Lee WH. Carboxymethyl chitosan nanoparticles loaded with bioactive peptide OH-CATH30 benefit nonscar wound healing. Int J Nanomedicine 2018; 13:5771-5786. [PMID: 30310280 PMCID: PMC6165789 DOI: 10.2147/ijn.s156206] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background Nonscar wound healing is a desirable treatment for cutaneous wounds worldwide. Peptide OH-CATH30 (OH30) from king cobra can selectively regulate the innate immunity and create an anti-inflammatory micro-environment which might benefit nonscar wound healing. Purpose To overcome the enzymatic digestion and control release of OH30, OH30 encapsulated in carboxymethyl chitosan nanoparticles (CMCS-OH30 NP) were prepared and their effects on wound healing were evaluated. Methods CMCS-OH30 NP were prepared by mild ionic gelation method and properties of the prepared CMCS-OH30 NP were determined by dynamic light scattering. Encapsulation efficiency, stability and release profile of OH30 from prepared CMCS-OH30 NP were determined by HPLC. Cytotoxicity, cell migration and cellular uptake of CMCS-OH30 NP were determined by conventional methods. The effects of prepared CMCS-OH30 NP on the wound healing was investigated by full-thickness excision animal models. Results The release of encapsulated OH30 from prepared CMCS-OH30 NP was maintained for at least 24 h in a controlled manner. CMCSOH30 NP enhanced the cell migration but had no effects on the metabolism and proliferation of keratinocytes. In the full-thickness excision animal models, the CMCS-OH30 NP treatment significantly accelerated the wound healing compared with CMCS or OH30 administration alone. Histopathological examination suggested that CMCS-OH30 NP promoted wound healing by enhancing the granulation tissue formation through the re-epithelialized and neovascularized composition. CMCS-OH30 NP induced a steady anti-inflammatory cytokine IL10 expression but downregulated the expressions of several pro-inflammatory cytokines. Conclusion The prepared biodegradable drug delivery system accelerates the healing and shows better prognosis because of the combined effects of OH30 released from the nanoparticles.
Collapse
Affiliation(s)
- Tongyi Sun
- Department of Bioengineering, School of Bioscience and Technology, Weifang Medical University, Weifang 261053, Shandong, China,
| | - Bo Zhan
- Department of Pharmaceutics, School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, China, .,Key Laboratory of Bioactive Peptide of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China,
| | - Weifen Zhang
- Department of Pharmaceutics, School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, China,
| | - Di Qin
- Department of Bioengineering, School of Bioscience and Technology, Weifang Medical University, Weifang 261053, Shandong, China,
| | - Guixue Xia
- Department of Pharmaceutics, School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, China,
| | - Huijie Zhang
- Department of Bioengineering, School of Bioscience and Technology, Weifang Medical University, Weifang 261053, Shandong, China, .,Key Laboratory of Bioactive Peptide of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China,
| | - Meiyu Peng
- Department of Immunology, School of Clinical Medicine, Weifang Medical University, Weifang 261053, Shandong, China
| | - Sheng-An Li
- Key Laboratory of Bioactive Peptide of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China,
| | - Yun Zhang
- Key Laboratory of Bioactive Peptide of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China,
| | - Yuanyuan Gao
- Department of Pharmaceutics, School of Pharmacy, Weifang Medical University, Weifang 261053, Shandong, China,
| | - Wen-Hui Lee
- Department of Bioengineering, School of Bioscience and Technology, Weifang Medical University, Weifang 261053, Shandong, China, .,Key Laboratory of Bioactive Peptide of Yunnan Province/Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, Yunnan, China,
| |
Collapse
|
29
|
Mei D, Zhao L, Chen B, Zhang X, Wang X, Yu Z, Ni X, Zhang Q. α-Conotoxin ImI-modified polymeric micelles as potential nanocarriers for targeted docetaxel delivery to α7-nAChR overexpressed non-small cell lung cancer. Drug Deliv 2018; 25:493-503. [PMID: 29426250 PMCID: PMC6058686 DOI: 10.1080/10717544.2018.1436097] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
A micelle system modified with α-Conotoxin ImI (ImI), a potently antagonist for alpha7 nicotinic acetylcholine receptor (α7-nAChR) previously utilized for targeting breast cancer, was constructed. Its targeting efficiency and cytotoxicity against non-small cell lung cancer (NSCLC) highly expressing α7-nAChR was investigated. A549, a non-small cell lung cancer cell line, was selected as the cell model. The cellular uptake study showed that the optimal modification ratio of ImI on micelle surface was 5% and ImI-modification increased intracellular delivery efficiency to A549 cells via receptor-mediated endocytosis. Intracellular Ca2+ transient assay demonstrated that ImI modification led to enhanced molecular interaction between nanocarriers and A549 cells. The in vivo near-infrared fluorescence imaging further revealed that ImI-modified micelles could facilitate the drug accumulation in tumor sites compared with non-modified micelles via α7-nAChR mediation. Moreover, docetaxel (DTX) was loaded in ImI-modified nanomedicines to evaluate its in vitro cytotoxicity. As a result, DTX-loaded ImI-PMs exhibited greater anti-proliferation effect on A549 cells compared with non-modified micelles. Generally, our study proved that ImI-modified micelles had targeting ability to NSCLC in addition to breast cancer and it may provide a promising strategy to deliver drugs to NSCLC overexpressing α7-nAChR.
Collapse
Affiliation(s)
- Dong Mei
- a Beijing Children's Hospital, Capital Medical University, National Center for Children's Health , Beijing , PR China
| | - Libo Zhao
- a Beijing Children's Hospital, Capital Medical University, National Center for Children's Health , Beijing , PR China
| | - Binlong Chen
- b State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Beijing , PR China
| | - Xiaoyan Zhang
- a Beijing Children's Hospital, Capital Medical University, National Center for Children's Health , Beijing , PR China
| | - Xiaoling Wang
- a Beijing Children's Hospital, Capital Medical University, National Center for Children's Health , Beijing , PR China
| | - Zhiying Yu
- c Department of Pharmacy , Peking University People's Hospital , Beijing , PR China
| | - Xin Ni
- a Beijing Children's Hospital, Capital Medical University, National Center for Children's Health , Beijing , PR China
| | - Qiang Zhang
- b State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences , Peking University , Beijing , PR China
| |
Collapse
|
30
|
Abraham N, Lewis RJ. Neuronal Nicotinic Acetylcholine Receptor Modulators from Cone Snails. Mar Drugs 2018; 16:E208. [PMID: 29899286 PMCID: PMC6024932 DOI: 10.3390/md16060208] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/25/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022] Open
Abstract
Marine cone snails are a large family of gastropods that have evolved highly potent venoms for predation and defense. The cone snail venom has exceptional molecular diversity in neuropharmacologically active compounds, targeting a range of receptors, ion channels, and transporters. These conotoxins have helped to dissect the structure and function of many of these therapeutically significant targets in the central and peripheral nervous systems, as well as unravelling the complex cellular mechanisms modulated by these receptors and ion channels. This review provides an overview of α-conotoxins targeting neuronal nicotinic acetylcholine receptors. The structure and activity of both classical and non-classical α-conotoxins are discussed, along with their contributions towards understanding nicotinic acetylcholine receptor (nAChR) structure and function.
Collapse
Affiliation(s)
- Nikita Abraham
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | - Richard J Lewis
- IMB Centre for Pain Research, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia.
| |
Collapse
|
31
|
Kathle PK, Gautam N, Kesavan K. Tamoxifen citrate loaded chitosan-gellan nanocapsules for breast cancer therapy: development, characterisation and in-vitro cell viability study. J Microencapsul 2018; 35:292-300. [DOI: 10.1080/02652048.2018.1477844] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Pankaj Kumar Kathle
- Department of Pharmaceutics, SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Nivedita Gautam
- Department of Pharmaceutics, SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Karthikeyan Kesavan
- Department of Pharmaceutics, SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| |
Collapse
|
32
|
Hua H, Zhang X, Mu H, Meng Q, Jiang Y, Wang Y, Lu X, Wang A, Liu S, Zhang Y, Wan Z, Sun K. RVG29-modified docetaxel-loaded nanoparticles for brain-targeted glioma therapy. Int J Pharm 2018; 543:179-189. [DOI: 10.1016/j.ijpharm.2018.03.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 03/08/2018] [Accepted: 03/15/2018] [Indexed: 11/28/2022]
|
33
|
Yu S, Wu Y, Xu P, Wang S, Zhangsun D, Luo S. Effects of serum, enzyme, thiol, and forced degradation on the stabilities of αO-Conotoxin GeXIVA[1,2] and GeXIVA [1,4]. Chem Biol Drug Des 2018; 91:1030-1041. [DOI: 10.1111/cbdd.13167] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/22/2017] [Accepted: 12/17/2017] [Indexed: 01/07/2023]
Affiliation(s)
- Shurun Yu
- Key Laboratory of Tropical Biological Resources, Ministry of Education; Hainan University; Haikou China
- Key Laboratory for Marine Drugs of Haikou; Hainan University; Haikou China
- Institute of Tropical Agriculture and Forestry; Hainan University; Haikou China
| | - Yong Wu
- Key Laboratory of Tropical Biological Resources, Ministry of Education; Hainan University; Haikou China
- Key Laboratory for Marine Drugs of Haikou; Hainan University; Haikou China
| | - Pan Xu
- Key Laboratory of Tropical Biological Resources, Ministry of Education; Hainan University; Haikou China
- Key Laboratory for Marine Drugs of Haikou; Hainan University; Haikou China
- Institute of Tropical Agriculture and Forestry; Hainan University; Haikou China
| | - Shuai Wang
- Key Laboratory of Tropical Biological Resources, Ministry of Education; Hainan University; Haikou China
- Key Laboratory for Marine Drugs of Haikou; Hainan University; Haikou China
- Institute of Tropical Agriculture and Forestry; Hainan University; Haikou China
| | - Dongting Zhangsun
- Key Laboratory of Tropical Biological Resources, Ministry of Education; Hainan University; Haikou China
- Key Laboratory for Marine Drugs of Haikou; Hainan University; Haikou China
| | - Sulan Luo
- Key Laboratory of Tropical Biological Resources, Ministry of Education; Hainan University; Haikou China
- Key Laboratory for Marine Drugs of Haikou; Hainan University; Haikou China
| |
Collapse
|
34
|
Tao J, Xu J, Chen F, Xu B, Gao J, Hu Y. Folate acid-Cyclodextrin/Docetaxel induces apoptosis in KB cells via the intrinsic mitochondrial pathway and displays antitumor activity in vivo. Eur J Pharm Sci 2018; 111:540-548. [DOI: 10.1016/j.ejps.2017.10.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/28/2017] [Accepted: 10/30/2017] [Indexed: 10/18/2022]
|
35
|
Zhao J, Chen H, Tang Y, Chen H, Chen G, Yin Y, Li G. Research progresses on the functional polypeptides in the detection and imaging of breast cancer. J Mater Chem B 2018; 6:2510-2523. [DOI: 10.1039/c7tb02541f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Polypeptides as functional groups continue to garner significant interest in the detection and imaging of breast cancer, working as recognition elements, signal sources, building blocks and therapeutic reagents, etc.
Collapse
Affiliation(s)
- Jing Zhao
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Huinan Chen
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Yingying Tang
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Hong Chen
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Guifang Chen
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| | - Yongmei Yin
- Department of Oncology
- The First Affiliated Hospital of Nanjing Medical University
- Nanjing 210029
- China
| | - Genxi Li
- Center for Molecular Recognition and Biosensing
- School of Life Sciences
- Shanghai University
- Shanghai 200444
- China
| |
Collapse
|
36
|
Dutertre S, Nicke A, Tsetlin VI. Nicotinic acetylcholine receptor inhibitors derived from snake and snail venoms. Neuropharmacology 2017. [PMID: 28623170 DOI: 10.1016/j.neuropharm.2017.06.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The nicotinic acetylcholine receptor (nAChR) represents the prototype of ligand-gated ion channels. It is vital for neuromuscular transmission and an important regulator of neurotransmission. A variety of toxic compounds derived from diverse species target this receptor and have been of elemental importance in basic and applied research. They enabled milestone discoveries in pharmacology and biochemistry ranging from the original formulation of the receptor concept, the first isolation and structural analysis of a receptor protein (the nAChR) to the identification, localization, and differentiation of its diverse subtypes and their validation as a target for therapeutic intervention. Among the venom-derived compounds, α-neurotoxins and α-conotoxins provide the largest families and still represent indispensable pharmacological tools. Application of modified α-neurotoxins provided substantial structural and functional details of the nAChR long before high resolution structures were available. α-bungarotoxin represents not only a standard pharmacological tool and label in nAChR research but also for unrelated proteins tagged with a minimal α-bungarotoxin binding motif. A major advantage of α-conotoxins is their smaller size, as well as superior selectivity for diverse nAChR subtypes that allows their development into ligands with optimized pharmacological and chemical properties and potentially novel drugs. In the following, these two groups of nAChR antagonists will be described focusing on their respective roles in the structural and functional characterization of nAChRs and their development into research tools. In addition, we provide a comparative overview of the diverse α-conotoxin selectivities that can serve as a practical guide for both structure activity studies and subtype classification. This article is part of the Special Issue entitled 'Venom-derived Peptides as Pharmacological Tools.'
Collapse
Affiliation(s)
- Sébastien Dutertre
- Institut des Biomolécules Max Mousseron, UMR 5247, Université Montpellier - CNRS, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
| | - Annette Nicke
- Walther Straub Institute for Pharmacology and Toxicology, Ludwig-Maximilians-Universität, Nußbaumstr. 26, 80336 Munich, Germany.
| | - Victor I Tsetlin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya str.16/10, Moscow 117999, Russian Federation
| |
Collapse
|
37
|
Qin B, Liu L, Pan Y, Zhu Y, Wu X, Song S, Han G. PEGylated Solanesol for Oral Delivery of Coenzyme Q 10. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:3360-3367. [PMID: 28418660 DOI: 10.1021/acs.jafc.7b00165] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Coenzyme Q10 (CoQ10) is widely used in preventive or curative treatment of cardiovascular diseases. However, CoQ10 exhibits an extremely low solubility in aqueous medium as well as a poor oral bioavailability. Therefore, solanesyl poly(ethylene glycol) succinate (SPGS) and CoQ10 were formulated as CoQ10-SPGS micelles with a high content of CoQ10 to improve the bioavailability of CoQ10 in rat. Findings indicate that, in the CoQ10-SPGS micelles, SPGS is self-assembled into stable nanosized micelles with a CoQ10 loading capacity of more than 39%. The CoQ10-SPGS micelles exhibit an enhanced photostability upon exposure to simulated sunlight. In vivo experiments demonstrate that, as compared to that of the coarse suspensions of CoQ10, there was three-fold enhancement of oral bioavailability for CoQ10-loaded SPGS micelles depending on varying molecular weight of SPGS. In the encapsulation of CoQ10 by SPGS micelles, the self-assembled nanocarriers with strong muco-adhesive properties lead to increases in the solubility and oral absorption of lipophilic CoQ10 nanoparticles.
Collapse
Affiliation(s)
- Benkai Qin
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming, Kaifeng, Henan 475001, China
| | - Lei Liu
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming, Kaifeng, Henan 475001, China
| | - Yangyang Pan
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming, Kaifeng, Henan 475001, China
| | - Yingchun Zhu
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming, Kaifeng, Henan 475001, China
| | - Xiaohe Wu
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming, Kaifeng, Henan 475001, China
| | - Shiyong Song
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming, Kaifeng, Henan 475001, China
| | - Guang Han
- Institute of Pharmacy, Pharmacy College of Henan University , Jinming, Kaifeng, Henan 475001, China
| |
Collapse
|
38
|
Pelaz B, Alexiou C, Alvarez-Puebla RA, Alves F, Andrews AM, Ashraf S, Balogh LP, Ballerini L, Bestetti A, Brendel C, Bosi S, Carril M, Chan WCW, Chen C, Chen X, Chen X, Cheng Z, Cui D, Du J, Dullin C, Escudero A, Feliu N, Gao M, George M, Gogotsi Y, Grünweller A, Gu Z, Halas NJ, Hampp N, Hartmann RK, Hersam MC, Hunziker P, Jian J, Jiang X, Jungebluth P, Kadhiresan P, Kataoka K, Khademhosseini A, Kopeček J, Kotov NA, Krug HF, Lee DS, Lehr CM, Leong KW, Liang XJ, Ling Lim M, Liz-Marzán LM, Ma X, Macchiarini P, Meng H, Möhwald H, Mulvaney P, Nel AE, Nie S, Nordlander P, Okano T, Oliveira J, Park TH, Penner RM, Prato M, Puntes V, Rotello VM, Samarakoon A, Schaak RE, Shen Y, Sjöqvist S, Skirtach AG, Soliman MG, Stevens MM, Sung HW, Tang BZ, Tietze R, Udugama BN, VanEpps JS, Weil T, Weiss PS, Willner I, Wu Y, Yang L, Yue Z, Zhang Q, Zhang Q, Zhang XE, Zhao Y, Zhou X, Parak WJ. Diverse Applications of Nanomedicine. ACS NANO 2017; 11:2313-2381. [PMID: 28290206 PMCID: PMC5371978 DOI: 10.1021/acsnano.6b06040] [Citation(s) in RCA: 823] [Impact Index Per Article: 102.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Indexed: 04/14/2023]
Abstract
The design and use of materials in the nanoscale size range for addressing medical and health-related issues continues to receive increasing interest. Research in nanomedicine spans a multitude of areas, including drug delivery, vaccine development, antibacterial, diagnosis and imaging tools, wearable devices, implants, high-throughput screening platforms, etc. using biological, nonbiological, biomimetic, or hybrid materials. Many of these developments are starting to be translated into viable clinical products. Here, we provide an overview of recent developments in nanomedicine and highlight the current challenges and upcoming opportunities for the field and translation to the clinic.
Collapse
Affiliation(s)
- Beatriz Pelaz
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Christoph Alexiou
- ENT-Department, Section of Experimental Oncology & Nanomedicine
(SEON), Else Kröner-Fresenius-Stiftung-Professorship for Nanomedicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Ramon A. Alvarez-Puebla
- Department of Physical Chemistry, Universitat Rovira I Virgili, 43007 Tarragona, Spain
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| | - Frauke Alves
- Department of Haematology and Medical Oncology, Department of Diagnostic
and Interventional Radiology, University
Medical Center Göttingen, 37075 Göttingen Germany
- Department of Molecular Biology of Neuronal Signals, Max-Planck-Institute for Experimental Medicine, 37075 Göttingen, Germany
| | - Anne M. Andrews
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Sumaira Ashraf
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Lajos P. Balogh
- AA Nanomedicine & Nanotechnology Consultants, North Andover, Massachusetts 01845, United States
| | - Laura Ballerini
- International School for Advanced Studies (SISSA/ISAS), 34136 Trieste, Italy
| | - Alessandra Bestetti
- School of Chemistry & Bio21 Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Cornelia Brendel
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Susanna Bosi
- Department of Chemical
and Pharmaceutical Sciences, University
of Trieste, 34127 Trieste, Italy
| | - Monica Carril
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
- Ikerbasque, Basque Foundation
for Science, 48013 Bilbao, Spain
| | - Warren C. W. Chan
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Chunying Chen
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
| | - Xiaodong Chen
- School of Materials
Science and Engineering, Nanyang Technological
University, Singapore 639798
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine,
National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhen Cheng
- Molecular
Imaging Program at Stanford and Bio-X Program, Canary Center at Stanford
for Cancer Early Detection, Stanford University, Stanford, California 94305, United States
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Department of Instrument
Science and Engineering, School of Electronic Information and Electronical
Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, 200240 Shanghai, China
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials
Science and Engineering, Tongji University, Shanghai, China
| | - Christian Dullin
- Department of Haematology and Medical Oncology, Department of Diagnostic
and Interventional Radiology, University
Medical Center Göttingen, 37075 Göttingen Germany
| | - Alberto Escudero
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
- Instituto
de Ciencia de Materiales de Sevilla. CSIC, Universidad de Sevilla, 41092 Seville, Spain
| | - Neus Feliu
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Mingyuan Gao
- Institute of Chemistry, Chinese
Academy of Sciences, 100190 Beijing, China
| | | | - Yury Gogotsi
- Department of Materials Science and Engineering and A.J. Drexel Nanomaterials
Institute, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Arnold Grünweller
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Zhongwei Gu
- College of Polymer Science and Engineering, Sichuan University, 610000 Chengdu, China
| | - Naomi J. Halas
- Departments of Physics and Astronomy, Rice
University, Houston, Texas 77005, United
States
| | - Norbert Hampp
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Roland K. Hartmann
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Mark C. Hersam
- Departments of Materials Science and Engineering, Chemistry,
and Medicine, Northwestern University, Evanston, Illinois 60208, United States
| | - Patrick Hunziker
- University Hospital, 4056 Basel, Switzerland
- CLINAM,
European Foundation for Clinical Nanomedicine, 4058 Basel, Switzerland
| | - Ji Jian
- Department of Polymer Science and Engineering and Center for
Bionanoengineering and Department of Chemical and Biological Engineering, Zhejiang University, 310027 Hangzhou, China
| | - Xingyu Jiang
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
| | - Philipp Jungebluth
- Thoraxklinik Heidelberg, Universitätsklinikum
Heidelberg, 69120 Heidelberg, Germany
| | - Pranav Kadhiresan
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | | | | | - Jindřich Kopeček
- Biomedical Polymers Laboratory, University of Utah, Salt Lake City, Utah 84112, United States
| | - Nicholas A. Kotov
- Emergency Medicine, University of Michigan, Ann Arbor, Michigan 48019, United States
| | - Harald F. Krug
- EMPA, Federal Institute for Materials
Science and Technology, CH-9014 St. Gallen, Switzerland
| | - Dong Soo Lee
- Department of Molecular Medicine and Biopharmaceutical
Sciences and School of Chemical and Biological Engineering, Seoul National University, Seoul, South Korea
| | - Claus-Michael Lehr
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
- HIPS - Helmhotz Institute for Pharmaceutical Research Saarland, Helmholtz-Center for Infection Research, 66123 Saarbrücken, Germany
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS), 100190 Beijing, China
| | - Mei Ling Lim
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Luis M. Liz-Marzán
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
- Ikerbasque, Basque Foundation
for Science, 48013 Bilbao, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine, Ciber-BBN, 20014 Donostia - San Sebastián, Spain
| | - Xiaowei Ma
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS), 100190 Beijing, China
| | - Paolo Macchiarini
- Laboratory of Bioengineering Regenerative Medicine (BioReM), Kazan Federal University, 420008 Kazan, Russia
| | - Huan Meng
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Helmuth Möhwald
- Department of Interfaces, Max-Planck
Institute of Colloids and Interfaces, 14476 Potsdam, Germany
| | - Paul Mulvaney
- School of Chemistry & Bio21 Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Andre E. Nel
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Shuming Nie
- Emory University, Atlanta, Georgia 30322, United States
| | - Peter Nordlander
- Departments of Physics and Astronomy, Rice
University, Houston, Texas 77005, United
States
| | - Teruo Okano
- Tokyo Women’s Medical University, Tokyo 162-8666, Japan
| | | | - Tai Hyun Park
- Department of Molecular Medicine and Biopharmaceutical
Sciences and School of Chemical and Biological Engineering, Seoul National University, Seoul, South Korea
- Advanced Institutes of Convergence Technology, Suwon, South Korea
| | - Reginald M. Penner
- Department of Chemistry, University of
California, Irvine, California 92697, United States
| | - Maurizio Prato
- Department of Chemical
and Pharmaceutical Sciences, University
of Trieste, 34127 Trieste, Italy
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
- Ikerbasque, Basque Foundation
for Science, 48013 Bilbao, Spain
| | - Victor Puntes
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
- Institut Català de Nanotecnologia, UAB, 08193 Barcelona, Spain
- Vall d’Hebron University Hospital
Institute of Research, 08035 Barcelona, Spain
| | - Vincent M. Rotello
- Department
of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Amila Samarakoon
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Raymond E. Schaak
- Department of Chemistry, The
Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Youqing Shen
- Department of Polymer Science and Engineering and Center for
Bionanoengineering and Department of Chemical and Biological Engineering, Zhejiang University, 310027 Hangzhou, China
| | - Sebastian Sjöqvist
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 141 86 Stockholm, Sweden
| | - Andre G. Skirtach
- Department of Interfaces, Max-Planck
Institute of Colloids and Interfaces, 14476 Potsdam, Germany
- Department of Molecular Biotechnology, University of Ghent, B-9000 Ghent, Belgium
| | - Mahmoud G. Soliman
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Molly M. Stevens
- Department of Materials,
Department of Bioengineering, Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Institute of Biomedical
Engineering, National Tsing Hua University, Hsinchu City, Taiwan,
ROC 300
| | - Ben Zhong Tang
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Hong Kong, China
| | - Rainer Tietze
- ENT-Department, Section of Experimental Oncology & Nanomedicine
(SEON), Else Kröner-Fresenius-Stiftung-Professorship for Nanomedicine, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Buddhisha N. Udugama
- Institute of Biomaterials
and Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| | - J. Scott VanEpps
- Emergency Medicine, University of Michigan, Ann Arbor, Michigan 48019, United States
| | - Tanja Weil
- Institut für
Organische Chemie, Universität Ulm, 89081 Ulm, Germany
- Max-Planck-Institute for Polymer Research, 55128 Mainz, Germany
| | - Paul S. Weiss
- California NanoSystems Institute, Department of Chemistry
and Biochemistry and Department of Psychiatry and Semel Institute
for Neuroscience and Human Behavior, Division of NanoMedicine and Center
for the Environmental Impact of Nanotechnology, and Department of Materials Science
and Engineering, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Itamar Willner
- Institute of Chemistry, The Center for
Nanoscience and Nanotechnology, The Hebrew
University of Jerusalem, Jerusalem 91904, Israel
| | - Yuzhou Wu
- Max-Planck-Institute for Polymer Research, 55128 Mainz, Germany
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 430074 Wuhan, China
| | | | - Zhao Yue
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Qian Zhang
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
| | - Qiang Zhang
- School of Pharmaceutical Science, Peking University, 100191 Beijing, China
| | - Xian-En Zhang
- National Laboratory of Biomacromolecules,
CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience and CAS Key
Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of
China, Beijing 100190, China
| | - Xin Zhou
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Wolfgang J. Parak
- Fachbereich Physik, Fachbereich Medizin, Fachbereich Pharmazie, and Department of Chemistry, Philipps Universität Marburg, 35037 Marburg, Germany
- CIC biomaGUNE, Paseo de Miramón 182, 20014, Donostia - San Sebastián, Spain
| |
Collapse
|
39
|
Utkin YN. Modern trends in animal venom research - omics and nanomaterials. World J Biol Chem 2017; 8:4-12. [PMID: 28289514 PMCID: PMC5329713 DOI: 10.4331/wjbc.v8.i1.4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/20/2016] [Accepted: 12/27/2016] [Indexed: 02/05/2023] Open
Abstract
Animal venom research is a specialized investigation field, in which a number of different methods are used and this array is constantly expanding. Thus, recently emerged omics and nanotechnologies have already been successfully applied to venom research. Animal venoms have been studied for quite a long time. The traditional reductionist approach has been to isolate individual toxins and then study their structure and function. Unfortunately, the characterization of the venom as a whole system and its multiple effects on an entire organism were not possible until recent times. The development of new methods in mass spectrometry and sequencing have allowed such characterizations of venom, encompassing the identification of new toxins present in venoms at extremely low concentrations to changes in metabolism of prey organisms after envenomation. In particular, this type of comprehensive research has become possible due to the development of the various omics technologies: Proteomics, peptidomics, transcriptomics, genomics and metabolomics. As in other research fields, these omics technologies ushered in a revolution for venom studies, which is now entering the era of big data. Nanotechnology is a very new branch of technology and developing at an extremely rapid pace. It has found application in many spheres and has not bypassed the venom studies. Nanomaterials are quite promising in medicine, and most studies combining venoms and nanomaterials are dedicated to medical applications. Conjugates of nanoparticles with venom components have been proposed for use as drugs or diagnostics. For example, nanoparticles conjugated with chlorotoxin - a toxin in scorpion venom, which has been shown to bind specifically to glioma cells - are considered as potential glioma-targeted drugs, and conjugates of neurotoxins with fluorescent semiconductor nanoparticles or quantum dots may be used to detect endogenous targets expressed in live cells. The data on application of omics and nanotechnologies in venom research are systematized concisely in this paper.
Collapse
|
40
|
Zhang X, Yang J, Chen M, Li L, Huan F, Li A, Liu Y, Xia Y, Duan JA, Ma S. Metabolomics profiles delineate uridine deficiency contributes to mitochondria-mediated apoptosis induced by celastrol in human acute promyelocytic leukemia cells. Oncotarget 2016; 7:46557-46572. [PMID: 27374097 PMCID: PMC5216817 DOI: 10.18632/oncotarget.10286] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/20/2016] [Indexed: 11/25/2022] Open
Abstract
Celastrol, extracted from "Thunder of God Vine", is a promising anti-cancer natural product. However, its effect on acute promyelocytic leukemia (APL) and underlying molecular mechanism are poorly understood. The purpose of this study was to explore its effect on APL and underlying mechanism based on metabolomics. Firstly, multiple assays indicated that celastrol could induce apoptosis of APL cells via p53-activated mitochondrial pathway. Secondly, unbiased metabolomics revealed that uridine was the most notable changed metabolite. Further study verified that uridine could reverse the apoptosis induced by celastrol. The decreased uridine was caused by suppressing the expression of gene encoding Dihydroorotate dehydrogenase, whose inhibitor could also induce apoptosis of APL cells. At last, mouse model confirmed that celastrol inhibited tumor growth through enhanced apoptosis. Celastrol could also decrease uridine and DHODH protein level in tumor tissues. Our in vivo study also indicated that celastrol had no systemic toxicity at pharmacological dose (2 mg/kg, i.p., 21 days). Altogether, our metabolomics study firstly reveals that uridine deficiency contributes to mitochondrial apoptosis induced by celastrol in APL cells. Celastrol shows great potential for the treatment of APL.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, China
- Department of Hygienic Analysis and Detection, Nanjing Medical University, Nanjing 211166, China
| | - Jing Yang
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, China
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing 211166, China
| | - Lei Li
- Department of Hygienic Analysis and Detection, Nanjing Medical University, Nanjing 211166, China
| | - Fei Huan
- Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing 211166, China
| | - Aiping Li
- Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing 211166, China
| | - Yanqing Liu
- Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing 211166, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Modern Toxicology of Ministry of Education, Nanjing Medical University, Nanjing 211166, China
| | - Jin-ao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shiping Ma
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
41
|
Yao C, Liu J, Wu X, Tai Z, Gao Y, Zhu Q, Li J, Zhang L, Hu C, Gu F, Gao J, Gao S. Reducible self-assembling cationic polypeptide-based micelles mediate co-delivery of doxorubicin and microRNA-34a for androgen-independent prostate cancer therapy. J Control Release 2016; 232:203-14. [DOI: 10.1016/j.jconrel.2016.04.034] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/30/2016] [Accepted: 04/23/2016] [Indexed: 01/04/2023]
|
42
|
Apoptosis Activation in Human Lung Cancer Cell Lines by a Novel Synthetic Peptide Derived from Conus californicus Venom. Toxins (Basel) 2016; 8:38. [PMID: 26861394 PMCID: PMC4773791 DOI: 10.3390/toxins8020038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 01/26/2016] [Accepted: 01/28/2016] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is one of the most common types of cancer in men and women and a leading cause of death worldwide resulting in more than one million deaths per year. The venom of marine snails Conus contains up to 200 pharmacologically active compounds that target several receptors in the cell membrane. Due to their diversity and specific binding properties, Conus toxins hold great potential as source of new drugs against cancer. We analyzed the cytotoxic effect of a 17-amino acid synthetic peptide (s-cal14.1a) that is based on a native toxin (cal14.1a) isolated from the sea snail Conus californicus. Cytotoxicity studies in four lung cancer cell lines were complemented with measurement of gene expression of apoptosis-related proteins Bcl-2, BAX and the pro-survival proteins NFκB-1 and COX-2, as well as quantification of caspase activity. Our results showed that H1299 and H1437 cell lines treated with s-call4.1a had decreased cell viability, activated caspases, and reduced expression of the pro-survival protein NFκB-1. To our knowledge, this is the first report describing activation of apoptosis in human lung cancer cell lines by s-cal14.1a and we offer insight into the possible mechanism of action.
Collapse
|
43
|
Zhang H, Patel N, Ding S, Xiong J, Wu P. Theranostics for hepatocellular carcinoma with Fe3O4@ZnO nanocomposites. Biomater Sci 2016; 4:288-298. [PMID: 26523294 DOI: 10.1039/c5bm00361j] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The purpose of the current study is to investigate Fe3O4@ZnO nanocomposites as theranostic agents for hepatocellular carcinoma (HCC). Initially, the transferrin receptor antibody (TfR Ab) functionalized Fe3O4@ZnO nanocomposites, followed by loading with doxorubicin (Dox) and denoted as Fe3O4@ZnO/Dox/TfR Ab, were prepared as an all-in-one system allowing for a targeted drug delivery with simultaneous concurrent chemoradiotherapy and magnetic resonance imaging (MRI) monitoring. The diagnostic and therapeutic functionalities for HCC were evaluated in vitro and in a murine orthotopic models using cell viability assays, cell cycle tests, histopathological examinations, and serum biochemistry tests. The results demonstrated that Fe3O4@ZnO/Dox/TfR Ab could deliver Dox into the targeted HCC SMMC-7721 cells to enhance its chemotherapeutic efficiency. Besides, with the addition of short term and low dose X-ray illumination, the Fe3O4@ZnO nanocomposites showed excellent radiosensitizer properties, further attacking the cancer cells. Tumor cells were also mostly arrested at G2/M, resulting in a distinct inhibition of cell proliferation. In vivo, after the treatment, a noninvasive visualization monitoring through MRI showed that tumor growth was significantly suppressed by the targeted chemoradiotherapy mediated by Fe3O4@ZnO/Dox/TfR Ab. Therefore, Fe3O4@ZnO nanocomposites could mediate the theranostic strategy for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Haijun Zhang
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China.
| | - Nishant Patel
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China.
| | - Shuang Ding
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China.
| | - Jian Xiong
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, People's Republic of China.
| | - Pingping Wu
- Jiangsu Cancer Hospital, Nanjing, People's Republic of China
| |
Collapse
|
44
|
Structural modifications in polymeric micelles to impart multifunctionality for improved drug delivery. Ther Deliv 2016; 7:73-87. [PMID: 26769002 DOI: 10.4155/tde.15.90] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Polymeric micelles are macromolecular nanoconstructs which are formed by self-assembly of synthetic amphiphilic block copolymers. These copolymers could be chemically modified to expand their functionality and hence obtain a multifunctional micelle which could serve several functions simultaneously, for example, long circulation time along with active targeting, smart polymeric micelles providing on-demand drug release for example, pH responsive micelles, redox- and light-sensitive micelles, charge-conversion micelles and core/shell cross-linked micelles. Additionally, micelles could be tailored to carry a contrast agent or siRNA/miRNA along with the drug for greater clinical benefit. The focus of the current commentary would be to highlight such chemical modifications which impart multifunctionality to a single carrier and discuss challenges involved in clinical translation of these multifunctional micelles.
Collapse
|
45
|
Yin T, Wang J, Yin L, Shen L, Zhou J, Huo M. Redox-sensitive hyaluronic acid–paclitaxel conjugate micelles with high physical drug loading for efficient tumor therapy. Polym Chem 2015. [DOI: 10.1039/c5py01355k] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Characterization of targeted redox-sensitive micelles self-assembled from polymer–drug conjugates exhibiting conspicuous drug loading capabilities, selective cellular uptake, rapid intracellular disassembly and drug release is presented.
Collapse
Affiliation(s)
- Tingjie Yin
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Jing Wang
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Lifang Yin
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Linjia Shen
- National Engineering and Research Center for Target Drugs
- Lianyungang 222047
- China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Meirong Huo
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| |
Collapse
|