1
|
Ratnaparkhi MP, Salvankar SS, Tekade AR, Kulkarni GM. Core-Shell Nanoparticles for Pulmonary Drug Delivery. Pharm Nanotechnol 2025; 13:90-116. [PMID: 38265371 DOI: 10.2174/0122117385277725231120043600] [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: 08/17/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 01/25/2024]
Abstract
Nanoscale drug delivery systems have provoked interest for application in various therapies on account of their ability to elevate the intracellular concentration of drugs inside target cells, which leads to an increase in efficacy, a decrease in dose, and dose-associated adverse effects. There are several types of nanoparticles available; however, core-shell nanoparticles outperform bare nanoparticles in terms of their reduced cytotoxicity, high dispersibility and biocompatibility, and improved conjugation with drugs and biomolecules because of better surface characteristics. These nanoparticulate drug delivery systems are used for targeting a number of organs, such as the colon, brain, lung, etc. Pulmonary administration of medicines is a more appealing method as it is a noninvasive route for systemic and locally acting drugs as the pulmonary region has a wide surface area, delicate blood-alveolar barrier, and significant vascularization. A core-shell nano-particulate drug delivery system is more effective in the treatment of various pulmonary disorders. Thus, this review has discussed the potential of several types of core-shell nanoparticles in treating various diseases and synthesis methods of core-shell nanoparticles. The methods for synthesis of core-shell nanoparticles include solid phase reaction, liquid phase reaction, gas phase reaction, mechanical mixing, microwave- assisted synthesis, sono-synthesis, and non-thermal plasma technology. The basic types of core-shell nanoparticles are metallic, magnetic, polymeric, silica, upconversion, and carbon nanomaterial- based core-shell nanoparticles. With this special platform, it is possible to integrate the benefits of both core and shell materials, such as strong serum stability, effective drug loading, adjustable particle size, and immunocompatibility.
Collapse
Affiliation(s)
- Mukesh P Ratnaparkhi
- Department of Pharmaceutics, Marathwada Mitra Mandal's College of Pharmacy, Thergaon, Pune, Maharashtra, 411033, India
| | - Shailendra S Salvankar
- Department of Pharmaceutics, Marathwada Mitra Mandal's College of Pharmacy, Thergaon, Pune, Maharashtra, 411033, India
| | - Avinash R Tekade
- Department of Pharmaceutics, Marathwada Mitra Mandal's College of Pharmacy, Thergaon, Pune, Maharashtra, 411033, India
| | - Gajanan M Kulkarni
- Department of Pharmaceutics, Marathwada Mitra Mandal's College of Pharmacy, Thergaon, Pune, Maharashtra, 411033, India
| |
Collapse
|
2
|
Ashkarran AA, Lin Z, Rana J, Bumpers H, Sempere L, Mahmoudi M. Impact of Nanomedicine in Women's Metastatic Breast Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2301385. [PMID: 37269217 PMCID: PMC10693652 DOI: 10.1002/smll.202301385] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/16/2023] [Indexed: 06/04/2023]
Abstract
Metastatic breast cancer is responsible for 90% of mortalities among women suffering from various types of breast cancers. Traditional cancer treatments such as chemotherapy and radiation therapy can cause significant side effects and may not be effective in many cases. However, recent advances in nanomedicine have shown great promise in the treatment of metastatic breast cancer. For example, nanomedicine demonstrated robust capacity in detection of metastatic cancers at early stages (i.e., before the metastatic cells leave the initial tumor site), which gives clinicians a timely option to change their treatment process (for example, instead of endocrine therapy they may use chemotherapy). Here recent advances in nanomedicine technology in the identification and treatment of metastatic breast cancers are reviewed.
Collapse
Affiliation(s)
- Ali Akbar Ashkarran
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Zijin Lin
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Jatin Rana
- Division of Hematology and Oncology, Michigan State University, East Lansing, MI, 48824, USA
| | - Harvey Bumpers
- Department of Surgery, Michigan State University, East Lansing, MI, 48824, USA
| | - Lorenzo Sempere
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, 48824, USA
- Connors Center for Women's Health & Gender Biology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
3
|
Hatshan MR, Saquib Q, Siddiqui MA, Faisal M, Ahmad J, Al-Khedhairy AA, Shaik MR, Khan M, Wahab R, Matteis VD, Adil SF. Effectiveness of Nonfunctionalized Graphene Oxide Nanolayers as Nanomedicine against Colon, Cervical, and Breast Cancer Cells. Int J Mol Sci 2023; 24:9141. [PMID: 37298090 PMCID: PMC10252622 DOI: 10.3390/ijms24119141] [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: 03/05/2023] [Revised: 04/26/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Recent studies in nanomedicine have intensively explored the prospective applications of surface-tailored graphene oxide (GO) as anticancer entity. However, the efficacy of nonfunctionalized graphene oxide nanolayers (GRO-NLs) as an anticancer agent is less explored. In this study, we report the synthesis of GRO-NLs and their in vitro anticancer potential in breast (MCF-7), colon (HT-29), and cervical (HeLa) cancer cells. GRO-NLs-treated HT-29, HeLa, and MCF-7 cells showed cytotoxicity in the MTT and NRU assays via defects in mitochondrial functions and lysosomal activity. HT-29, HeLa, and MCF-7 cells treated with GRO-NLs exhibited substantial elevations in ROS, disturbances of the mitochondrial membrane potential, an influx of Ca2+, and apoptosis. The qPCR quantification showed the upregulation of caspase 3, caspase 9, bax, and SOD1 genes in GRO-NLs-treated cells. Western blotting showed the depletion of P21, P53, and CDC25C proteins in the above cancer cell lines after GRO-NLs treatment, indicating its function as a mutagen to induce mutation in the P53 gene, thereby affecting P53 protein and downstream effectors P21 and CDC25C. In addition, there may be a mechanism other than P53 mutation that controls P53 dysfunction. We conclude that nonfunctionalized GRO-NLs exhibit prospective biomedical application as a putative anticancer entity against colon, cervical, and breast cancers.
Collapse
Affiliation(s)
- Mohammad Rafe Hatshan
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| | - Quaiser Saquib
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Maqsood A. Siddiqui
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Mohammad Faisal
- Botany and Microbiology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Javed Ahmad
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Abdulaziz A. Al-Khedhairy
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Mohammed Rafi Shaik
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| | - Mujeeb Khan
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| | - Rizwan Wahab
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Valeria De Matteis
- Department of Mathematics and Physics “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy;
| | - Syed Farooq Adil
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| |
Collapse
|
4
|
Zhao YQ, Li LJ, Zhou EF, Wang JY, Wang Y, Guo LM, Zhang XX. Lipid-Based Nanocarrier Systems for Drug Delivery: Advances and Applications. PHARMACEUTICAL FRONTS 2022. [DOI: 10.1055/s-0042-1751036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Lipid-based nanocarriers have been extensively investigated for drug delivery due to their advantages including biodegradability, biocompatibility, nontoxicity, and nonimmunogenicity. However, the shortcomings of traditional lipid-based nanocarriers such as insufficient targeting, capture by the reticuloendothelial system, and fast elimination limit the efficiency of drug delivery and therapeutic efficacy. Therefore, a series of multifunctional lipid-based nanocarriers have been developed to enhance the accumulation of drugs in the lesion site, aiming for improved diagnosis and treatment of various diseases. In this review, we summarized the advances and applications of lipid-based nanocarriers from traditional to novel functional lipid preparations, including liposomes, stimuli-responsive lipid-based nanocarriers, ionizable lipid nanoparticles, lipid hybrid nanocarriers, as well as biomembrane-camouflaged nanoparticles, and further discussed the challenges and prospects of this system. This exploration may give a complete idea viewing the lipid-based nanocarriers as a promising choice for drug delivery system, and fuel the advancement of pharmaceutical products by materials innovation and nanotechnology.
Collapse
Affiliation(s)
- Yan-Qi Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Li-Jun Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Er-Fen Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, People's Republic of China
| | - Jiang-Yue Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- School of Pharmacy, East China University of Science and Technology, Shanghai, People's Republic of China
| | - Ying Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Lin-Miao Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Xin-Xin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, People's Republic of China
| |
Collapse
|
5
|
Xu D, Tang WJ, Zhu YZ, Liu Z, Yang K, Liang MX, Chen X, Wu Y, Tang JH, Zhang W. Hyperthermia promotes exosome secretion by regulating Rab7b while increasing drug sensitivity in adriamycin-resistant breast cancer. Int J Hyperthermia 2022; 39:246-257. [PMID: 35100921 DOI: 10.1080/02656736.2022.2029585] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE To investigate the mechanism through which hyperthermia promotes exosome secretion and drug sensitivity in adriamycin-resistant breast cancer. MATERIALS AND METHODS We first evaluated the effect of hyperthermia on adriamycin-resistant breast cancer viability and used transmission electron microscopy, nanoparticle tracking analysis, and a bicinchoninic acid kit to validate the effect of hyperthermia on exosome secretion. The effective targeting molecules and pathways changed by hyperthermia were explored by RNA microarray and verified in vitro. The adriamycin-resistant MCF-7/ADR cells co-incubated with the exosomes produced by MCF-7/ADR cells after hyperthermia were assessed. The uptake of exosomes by MCF-7/ADR cells after hyperthermia treatment was evaluated by confocal microscopy. Finally, the mechanism through which hyperthermia promotes exosome secretion by hyperthermia was determined. RESULTS Hyperthermia significantly suppressed the growth of adriamycin-resistant breast cancer cells and increased drug sensitivity by upregulating FOS and CREB5, genes related to longer overall survival in breast cancer patients. Moreover, hyperthermia promoted exosome secretion through Rab7b, a small GTPase that controls endosome transport. The upregulated FOS and CREB5 antioncogenes can be transferred to MCF-7/ADR cells by hyperthermia-treated MCF-7/ADR cell-secreted exosomes. CONCLUSIONS Our results demonstrated a novel function of hyperthermia in promoting exosome secretion in adriamycin-resistant breast cancer cells and revealed the effects of hyperthermia on tumor cell biology. These hyperthermia-triggered exosomes can carry antitumor genes to the residual tumor and tumor microenvironment, which may be more beneficial to the effects of hyperthermia. These results represent an exploration of the relationship between therapeutic strategies and exosome biology.
Collapse
Affiliation(s)
- Di Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Wen-Juan Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Yi-Zhi Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P. R. China
| | - Zhen Liu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Kai Yang
- School of Clinical Medicine, Xuzhou Medical University, Xuzhou, P. R. China
| | - Ming-Xing Liang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Xiu Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Yang Wu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Jin-Hai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Wei Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
6
|
Li C, Liu Y, Wei M, Liu J, Yu X, Hu P, Liu Y. A novel core-shell rifampicin/isoniazid electrospun nanofiber membrane for long time drug dissolution. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
7
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
8
|
Ganesan K, Wang Y, Gao F, Liu Q, Zhang C, Li P, Zhang J, Chen J. Targeting Engineered Nanoparticles for Breast Cancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13111829. [PMID: 34834243 PMCID: PMC8623926 DOI: 10.3390/pharmaceutics13111829] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/11/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022] Open
Abstract
Breast cancer (BC) is the second most common cancer in women globally after lung cancer. Presently, the most important approach for BC treatment consists of surgery, followed by radiotherapy and chemotherapy. The latter therapeutic methods are often unsuccessful in the treatment of BC because of their various side effects and the damage incurred to healthy tissues and organs. Currently, numerous nanoparticles (NPs) have been identified and synthesized to selectively target BC cells without causing any impairments to the adjacent normal tissues or organs. Based on an exploratory study, this comprehensive review aims to provide information on engineered NPs and their payloads as promising tools in the treatment of BC. Therapeutic drugs or natural bioactive compounds generally incorporate engineered NPs of ideal sizes and shapes to enhance their solubility, circulatory half-life, and biodistribution, while reducing their side effects and immunogenicity. Furthermore, ligands such as peptides, antibodies, and nucleic acids on the surface of NPs precisely target BC cells. Studies on the synthesis of engineered NPs and their impact on BC were obtained from PubMed, Science Direct, and Google Scholar. This review provides insights on the importance of engineered NPs and their methodology for validation as a next-generation platform with preventive and therapeutic effects against BC.
Collapse
Affiliation(s)
- Kumar Ganesan
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong, Hong Kong, China; (K.G.); (Y.W.); (Q.L.)
| | - Yan Wang
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong, Hong Kong, China; (K.G.); (Y.W.); (Q.L.)
| | - Fei Gao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (F.G.); (C.Z.)
| | - Qingqing Liu
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong, Hong Kong, China; (K.G.); (Y.W.); (Q.L.)
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen 518063, China
| | - Chen Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (F.G.); (C.Z.)
| | - Peng Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China;
| | - Jinming Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (F.G.); (C.Z.)
- Correspondence: (J.Z.); (J.C.); Tel.: +852-3917-6479 (J.C.)
| | - Jianping Chen
- Li Ka Shing Faculty of Medicine, School of Chinese Medicine, The University of Hong Kong, Hong Kong, China; (K.G.); (Y.W.); (Q.L.)
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen 518063, China
- Correspondence: (J.Z.); (J.C.); Tel.: +852-3917-6479 (J.C.)
| |
Collapse
|
9
|
Chowdhury P, Ghosh U, Samanta K, Jaggi M, Chauhan SC, Yallapu MM. Bioactive nanotherapeutic trends to combat triple negative breast cancer. Bioact Mater 2021; 6:3269-3287. [PMID: 33778204 PMCID: PMC7970221 DOI: 10.1016/j.bioactmat.2021.02.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/27/2021] [Accepted: 02/28/2021] [Indexed: 02/09/2023] Open
Abstract
The management of aggressive breast cancer, particularly, triple negative breast cancer (TNBC) remains a formidable challenge, despite treatment advancement. Although newer therapies such as atezolizumab, olaparib, and sacituzumab can tackle the breast cancer prognosis and/or progression, but achieved limited survival benefit(s). The current research efforts are aimed to develop and implement strategies for improved bioavailability, targetability, reduce systemic toxicity, and enhance therapeutic outcome of FDA-approved treatment regimen. This review presents various nanoparticle technology mediated delivery of chemotherapeutic agent(s) for breast cancer treatment. This article also documents novel strategies to employ cellular and cell membrane cloaked (biomimetic) nanoparticles for effective clinical translation. These technologies offer a safe and active targeting nanomedicine for effective management of breast cancer, especially TNBC.
Collapse
Affiliation(s)
- Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Upasana Ghosh
- Department of Biomedical Engineering, School of Engineering, Rutgers University, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Kamalika Samanta
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C. Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M. Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| |
Collapse
|
10
|
Forouhari S, Beygi Z, Mansoori Z, Hajsharifi S, Heshmatnia F, Gheibihayat SM. Liposomes: Ideal drug delivery systems in breast cancer. Biotechnol Appl Biochem 2021; 69:1867-1884. [PMID: 34505736 DOI: 10.1002/bab.2253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022]
Abstract
Breast cancer (BC) has been recognized as the most common type of cancer in females across the world, accounting for 12% of each cancer case. In this sense, better diagnosis and screening have been thus far proven to contribute to higher survival rates. Moreover, traditional (or standard) chemotherapy is still known as one of the several prominent therapeutic options available, though it suffers from unsuitable cell selectivity, severe consequences, as well as resistance. In this regard, nanobased drug delivery systems (DDSs) are likely to provide promising grounds for BC treatment. Liposomes are accordingly effective nanosystems, having the benefits of multiple formulations verified to treat different diseases. Such systems possess specific features, including smaller size, biodegradability, hydrophobic/hydrophilic characteristics, biocompatibility, lower toxicity, as well as immunogenicity, which can all lead to considerable efficacy in treating various types of cancer. As chemotherapy uses drugs to target tumors, generates higher drug concentrations in tumors, which can provide for their slow release, and enhances drug stability, it can be improved via liposomes in DDSs for BC treatment. Therefore, the present study aims to review the existing issues regarding BC treatment and discuss liposome-based targeting in order to overcome barriers to conventional drug therapy.
Collapse
Affiliation(s)
- Sedighe Forouhari
- Infertility Research Center, Research Center of Quran, Hadith, and Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Beygi
- Department of Nursing and Midwife, Maybod Branch, Islamic Azad University, Maybod, Iran
| | - Zahra Mansoori
- Faculty of Educational Sciences and Psychology, Department of Sports Sciences, Shiraz University, Shiraz, Iran
| | - Sara Hajsharifi
- Student Research Committee, Department of Midwifery, Fatemeh (PBUH) School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Heshmatnia
- Student Research Committee, Department of Midwifery, Fatemeh (PBUH) School of Nursing and Midwifery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
11
|
Zhuang Y, Zhao Y, Wang B, Wang Q, Cai T, Cai Y. Strategies for Preparing Different Types of Lipid Polymer Hybrid Nanoparticles in Targeted Tumor Therapy. Curr Pharm Des 2021; 27:2274-2288. [PMID: 33222665 DOI: 10.2174/1381612826666201120155558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/27/2020] [Indexed: 11/22/2022]
Abstract
At present, cancer is one of the most common diseases in the world, causing a large number of deaths and seriously affecting people's health. The traditional treatment of cancer is mainly surgery, radiotherapy or chemotherapy. Conventional chemotherapy is still an important treatment, but it has some shortcomings, such as poor cell selectivity, serious side effects, drug resistance and so on. Nanoparticle administration can improve drug stability, reduce toxicity, prolong drug release time, prolong system half-life, and bring broad prospects for tumor therapy. Lipid polymer hybrid nanoparticles (LPNs), which combine the advantages of polymer core and phospholipid shell to form a single platform, have become multi-functional drug delivery platforms. This review introduces the basic characteristics, structure and preparation methods of LPNs, and discusses targeting strategies of LPNs in tumor therapy in order to overcome the defects of traditional drug therapy.
Collapse
Affiliation(s)
- Yong Zhuang
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Yiye Zhao
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510315, China
| | - Bingyue Wang
- Guangzhou Jiayuan Medical and Pharmaceutical Technology Co., Ltd., Guangzhou 510663, China
| | - Qi Wang
- Guangzhou Jiayuan Medical and Pharmaceutical Technology Co., Ltd., Guangzhou 510663, China
| | - Tiange Cai
- College of Life Science, Liaoning University, Shenyang 110036, China
| | - Yu Cai
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| |
Collapse
|
12
|
Liu Y, Li C, Chen J, Han Y, Wei M, Liu J, Yu X, Li F, Hu P, Fu L, Liu Y. Electrospun high bioavailable rifampicin–isoniazid-polyvinylpyrrolidone fiber membranes. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-021-01957-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
13
|
Rahman S, Kumar V, Kumar A, Abdullah TS, Rather IA, Jan AT. Molecular Perspective of Nanoparticle Mediated Therapeutic Targeting in Breast Cancer: An Odyssey of Endoplasmic Reticulum Unfolded Protein Response (UPR ER) and Beyond. Biomedicines 2021; 9:biomedicines9060635. [PMID: 34199484 PMCID: PMC8229605 DOI: 10.3390/biomedicines9060635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is the second most frequent cause of death among women. Representing a complex and heterogeneous type of cancer, its occurrence is attributed by both genetic (gene mutations, e.g., BRCA1, BRCA2) and non-genetic (race, ethnicity, etc.) risk factors. The effectiveness of available treatment regimens (small molecules, cytotoxic agents, and inhibitors) decreased due to their poor penetration across biological barriers, limited targeting, and rapid body clearance along with their effect on normal resident cells of bone marrow, gastrointestinal tract, and hair follicles. This significantly reduced their clinical outcomes, which led to an unprecedented increase in the number of cases worldwide. Nanomedicine, a nano-formulation of therapeutics, emerged as a versatile delivering module for employment in achieving the effective and target specific delivery of pharmaceutical payloads. Adoption of nanotechnological approaches in delivering therapeutic molecules to target cells ensures not only reduced immune response and toxicity, but increases the stability of therapeutic entities in the systemic circulation that averts their degradation and as such increased extravasations and accumulation via enhanced permeation and the retention (EPR) effect in target tissues. Additionally, nanoparticle (NP)-induced ER stress, which enhances apoptosis and autophagy, has been utilized as a combative strategy in the treatment of cancerous cells. As nanoparticles-based avenues have been capitalized to achieve better efficacy of the new genera of therapeutics with enhanced specificity and safety, the present study is aimed at providing the fundamentals of BC, nanotechnological modules (organic, inorganic, and hybrid) employed in delivering different therapeutic molecules, and mechanistic insights of nano-ER stress induced apoptosis and autophagy with a perspective of exploring this avenue for use in the nano-toxicological studies. Furthermore, the current scenario of USA FDA approved nano-formulations and the future perspective of nanotechnological based interventions to overcome the existing challenges are also discussed.
Collapse
Affiliation(s)
- Safikur Rahman
- Department of Botany, Munshi Singh College, BR Ambedkar Bihar University, Muzaffarpur 845401, India;
| | - Vijay Kumar
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Korea;
| | - Anuj Kumar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Korea;
| | - Tasduq S. Abdullah
- Council of Scientific and Industrial Research–Indian Institute of Integrative Medicine (CSIR–IIIM), Jammu 180001, India;
| | - Irfan A. Rather
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), P.O. Box 80141, Jeddah 21589, Saudi Arabia
- Correspondence: (I.A.R.); (A.T.J.)
| | - Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, India
- Correspondence: (I.A.R.); (A.T.J.)
| |
Collapse
|
14
|
Uda RM, Yoshida N, Iwasaki T, Hayashi K. pH-triggered solubility and cytotoxicity changes of malachite green derivatives incorporated in liposomes for killing cancer cells. J Mater Chem B 2021; 8:8242-8248. [PMID: 32794526 DOI: 10.1039/d0tb01346c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Three different malachite green leuco derivatives (MG-Xs) are incorporated in liposomes. In all three cases, a substituent (X) is covalently linked to the central carbon atom, abbreviated as MG-OH, MG-OCH3, and MG-CN. The three MG-X compounds are solubilized separately in liposome membranes and become cationic (MG+) and water soluble under acidic conditions. MG+ is consequently released from the liposome to the aqueous exterior. Their release behavior corresponds to their ionization ability: MG-OH > MG-OCH3 > MG-CN. The cellular uptake of the liposomes, the cytotoxic effect, and the location of MG+ in cancer cells are investigated using murine cells derived from colon cancer (Colon 26 cells) and human embryonic kidney cells (HEK 293 cells). The toxic effect on cancer cells is correlated to the ionization ability of MG-Xs. The liposomes effectively deliver MG+via the endocytic pathway, resulting in the cytotoxicity of liposomes containing MG-OH which is higher than that of free MG-OH and MG+. The difference in the phospholipids constituting the liposome membranes barely had an effect on the ionization ratio and the cytotoxicity of MG-OH. Confocal fluorescence microscopic observations revealed that MG+ is ultimately transported into the nuclei after being released in acidic cellular compartments.
Collapse
Affiliation(s)
- Ryoko M Uda
- Department of Chemical Engineering, National Institute of Technology, Nara College, Yata 22, Yamato-koriyama, Nara 639-1080, Japan.
| | - Nao Yoshida
- Department of Chemical Engineering, National Institute of Technology, Nara College, Yata 22, Yamato-koriyama, Nara 639-1080, Japan.
| | - Tomoyuki Iwasaki
- Division of Analytical Bio-medicine, Advanced Research Support Center, Ehime University, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Keita Hayashi
- Department of Chemical Engineering, National Institute of Technology, Nara College, Yata 22, Yamato-koriyama, Nara 639-1080, Japan.
| |
Collapse
|
15
|
Cui L, Liu W, Liu H, Qin Q, Wu S, He S, Zhang Z, Pang X, Zhu C. Cascade-Targeting of Charge-Reversal and Disulfide Bonds Shielding for Efficient DOX Delivery of Multistage Sensitive MSNs-COS-SS-CMC. Int J Nanomedicine 2020; 15:6153-6165. [PMID: 32884269 PMCID: PMC7443036 DOI: 10.2147/ijn.s252769] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Although pH and redox sensitiveness have been extensively investigated to improve therapeutic efficiency, the effect of disulfide bonds location and pH-triggered charge-reversal on cascade-targeting still need to be further evaluated in cancer treatment with multi-responsive nanoparticles. PURPOSE The aim of this study was to design multi-responsive DOX@MSNs-COS-NN-CMC, DOX@MSNs-COS-SS-CMC and DOX@MSNs-COS-CMC-SS and systematically investigate the effects of disulfide bonds location and charge-reversal on the cancer cell specificity, endocytosis mechanisms and antitumor efficiency. RESULTS In vitro drug release rate of DOX@MSNs-COS-SS-CMC in tumor environments was 7-fold higher than that under normal physiological conditions after 200 h. Furthermore, the fluorescence intensity of DOX@MSNs-COS-SS-CMC and DOX@MSNs-COS-CMC-SS was 1.9-fold and 1.3-fold higher than free DOX at pH 6.5 and 10 mM GSH. In addition, vesicular transport might be a factor that affects the uptake efficiency of DOX@MSNs-COS-SS-CMC and DOX@MSNs-COS-CMC-SS. The clathrin-mediated endocytosis and endosomal escape of DOX@MSNs-COS-SS-CMC enhanced cellular internalization and preserved highly controllable drug release into the perinuclear of HeLa cells. DOX@MSNs-COS-SS-CMC exhibited a synergistic chemotherapy in preeminent tumor inhibition and less side effects of cardiotoxicity. CONCLUSION The cascade-targeting of charge-reversal and disulfide bonds shielding would be a highly personalized strategy for cervical cancer treatment.
Collapse
Affiliation(s)
- Lan Cui
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou450001, People’s Republic of China
| | - Wentao Liu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou450001, People’s Republic of China
| | - Hao Liu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou450001, People’s Republic of China
| | - Qian Qin
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou450001, People’s Republic of China
- Bio & Soft Matter, Institute of Condensed Matter and Nanosciences, Universite Catholique de Louvain, Louvain-la-NeuveB-1348, Belgium
| | - Shuangxia Wu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou450001, People’s Republic of China
| | - Suqin He
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou450001, People’s Republic of China
- Henan Key Laboratory of Advanced Nylon Materials and Application, Zhengzhou University, Zhengzhou450001, People’s Republic of China
| | - Zhenya Zhang
- Department of Chemistry, Changwon National University of Korea, Changwon-city, Gyeongnam-do51140, Republic of Korea
| | - Xinchang Pang
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou450001, People’s Republic of China
| | - Chengshen Zhu
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou450001, People’s Republic of China
| |
Collapse
|
16
|
Omer ME, Halwani M, Alenazi RM, Alharbi O, Aljihani S, Massadeh S, Al Ghoribi M, Al Aamery M, Yassin AE. Novel Self-Assembled Polycaprolactone-Lipid Hybrid Nanoparticles Enhance the Antibacterial Activity of Ciprofloxacin. SLAS Technol 2020; 25:598-607. [PMID: 32734812 DOI: 10.1177/2472630320943126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Ciprofloxacin (CIP), a widely used antibiotic, is a poor biopharmaceutical resulting in low bioavailability. We optimized a CIP polymer-lipid hybrid nanoparticle (CIP-PLN) delivery system to enhance its biopharmaceutical attributes and the overall therapeutic performance. CIP-PLN formulations were prepared by a direct emulsification-solvent-evaporation method. Varying the type and ratio of lipid was tried to optimize a CIP-PLN formulation. All the prepared formulations were evaluated for their particle size, polydispersity index, zeta potential, physical stability, and drug entrapment efficiency. The drug in vitro release profile was also studied. Antibacterial activities were tested by the agar diffusion method for all CIP-PLN formulations against an Escherichia coli clinical bacterial isolate (EC04). CIP-PLN formulations showed average sizes in the range of 133.9 ± 1.7 nm to 217.1 ± 0.8 nm, exhibiting high size uniformity as indicated by polydispersity indices lower than 0.25. The entrapment efficiency was close to 80% for all formulations. The differential scanning calorimetry (DSC) thermograms indicated the existence of CIP in the amorphous state in all PLN formulations. Fourier transform infrared spectra indicated deep incorporation of molecular CIP within the polymer matrix. The release profile of CIP from PLN formulas showed a uniform prolonged drug profile, extended for a week from most formulations with a zero-order kinetics. The antibacterial activity of CIP-PLN formulations showed significantly higher antibacterial activity only with F4 containing lecithin as the lipid component. In conclusion, we successfully optimized a CIP-PLN formulation with a low nanoparticle size in a close range, high percentage of entrapment efficiency and drug loading, uniform prolonged release rate, and higher antibacterial activity against the EC04 clinical isolate.
Collapse
Affiliation(s)
- Mustafa E Omer
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, Saudi Arabia
| | - Majed Halwani
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, Saudi Arabia
| | - Rayan M Alenazi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Omar Alharbi
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Shokran Aljihani
- King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, Saudi Arabia
| | - Salam Massadeh
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, Saudi Arabia
| | - Majed Al Ghoribi
- King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, Saudi Arabia
| | - Manal Al Aamery
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, Saudi Arabia
| | - Alaa Eldeen Yassin
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard, Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
17
|
Bio-functionalized CuO nanoparticles induced apoptotic activities in human breast carcinoma cells and toxicity against Aspergillus flavus: An in vitro approach. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.01.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
18
|
Singhal A, Sinha N, Kumari P, Purkayastha M. Synthesis and Applications of Hydrogels in Cancer Therapy. Anticancer Agents Med Chem 2020; 20:1431-1446. [PMID: 31958041 DOI: 10.2174/1871521409666200120094048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 11/10/2019] [Accepted: 12/04/2019] [Indexed: 11/22/2022]
Abstract
Hydrogels are water-insoluble, hydrophilic, cross-linked, three-dimensional networks of polymer chains having the ability to swell and absorb water but do not dissolve in it, that comprise the major difference between gels and hydrogels. The mechanical strength, physical integrity and solubility are offered by the crosslinks. The different applications of hydrogels can be derived based on the methods of their synthesis, response to different stimuli, and their different kinds. Hydrogels are highly biocompatible and have properties similar to human tissues that make it suitable to be used in various biomedical applications, including drug delivery and tissue engineering. The role of hydrogels in cancer therapy is highly emerging in recent years. In the present review, we highlighted different methods of synthesis of hydrogels and their classification based on different parameters. Distinctive applications of hydrogels in the treatment of cancer are also discussed.
Collapse
Affiliation(s)
- Anchal Singhal
- Department of Chemistry, St. Joseph's College (Autonomous), Bangalore-560027, India
| | - Niharika Sinha
- Department of Chemistry, Gautam Buddha University, Noida, India
| | - Pratibha Kumari
- Department of Chemistry, Deshbandhu College, University of Delhi, New Delhi, India
| | | |
Collapse
|
19
|
Ghorbanizamani F, Moulahoum H, Zihnioglu F, Timur S. Nanohybrid carriers: the yin–yang equilibrium between natural and synthetic in biomedicine. Biomater Sci 2020; 8:3237-3247. [DOI: 10.1039/d0bm00401d] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Nanocarriers are key players in biomedicine applications. The development of hybrid nanoparticles stems from the need to enhance their quality by lowering disadvantages and fusing the positive qualities of both natural and synthetic materials.
Collapse
Affiliation(s)
| | - Hichem Moulahoum
- Biochemistry Department
- Faculty of Science
- Ege University
- Bornova
- Turkey
| | - Figen Zihnioglu
- Biochemistry Department
- Faculty of Science
- Ege University
- Bornova
- Turkey
| | - Suna Timur
- Biochemistry Department
- Faculty of Science
- Ege University
- Bornova
- Turkey
| |
Collapse
|
20
|
Thakur V, Kutty RV. Recent advances in nanotheranostics for triple negative breast cancer treatment. J Exp Clin Cancer Res 2019; 38:430. [PMID: 31661003 PMCID: PMC6819447 DOI: 10.1186/s13046-019-1443-1] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/10/2019] [Indexed: 12/20/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most complex and aggressive type of breast cancer encountered world widely in women. Absence of hormonal receptors on breast cancer cells necessitates the chemotherapy as the only treatment regime. High propensity to metastasize and relapse in addition to poor prognosis and survival motivated the oncologist, nano-medical scientist to develop novel and efficient nanotherapies to solve such a big TNBC challenge. Recently, the focus for enhanced availability, targeted cellular uptake with minimal toxicity is achieved by nano-carriers. These smart nano-carriers carrying all the necessary arsenals (drugs, tracking probe, and ligand) designed in such a way that specifically targets the TNBC cells at site. Articulating the targeted delivery system with multifunctional molecules for high specificity, tracking, diagnosis, and treatment emerged as theranostic approach. In this review, in addition to classical treatment modalities, recent advances in nanotheranostics for early and effective diagnostic and treatment is discussed. This review highlighted the recently FDA approved immunotherapy and all the ongoing clinical trials for TNBC, in addition to nanoparticle assisted immunotherapy. Futuristic but realistic advancements in artificial intelligence (AI) and machine learning not only improve early diagnosis but also assist clinicians for their workup in TNBC. The novel concept of Nanoparticles induced endothelial leakiness (NanoEL) as a way of tumor invasion is also discussed in addition to classical EPR effect. This review intends to provide basic insight and understanding of the novel nano-therapeutic modalities in TNBC diagnosis and treatment and to sensitize the readers for continue designing the novel nanomedicine. This is the first time that designing nanoparticles with stoichiometric definable number of antibodies per nanoparticle now represents the next level of precision by design in nanomedicine.
Collapse
Affiliation(s)
- Vikram Thakur
- Department of Virology, Postgraduate Institute of Medical Education and Research, PGIMER, Chandigarh, 160012 India
| | - Rajaletchumy Veloo Kutty
- Faculty of Chemical and Process Engineering Technology, College of Engineering Technology,University Malaysia Pahang, Tun Razak Highway, 26300 Kuantan, Pahang Malaysia
- Center of Excellence for Advanced Research in Fluid Flow, University Malaysia Pahang, 26300, Kuantan, Pahang Malaysia
| |
Collapse
|
21
|
Li H, Li H, Yu W, Huang S, Liu Y, Zhang N, Yuan J, Xu X, Duan S, Hu Y. PEGylated hyaluronidase/NIR induced drug controlled release system for synergetic chemo-photothermal therapy of hepatocellular carcinoma. Eur J Pharm Sci 2019; 133:127-136. [PMID: 30779981 DOI: 10.1016/j.ejps.2019.02.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 01/26/2019] [Accepted: 02/15/2019] [Indexed: 01/10/2023]
Abstract
In recent years, cancer treatment has been facing the challenge of increasing antitumor efficiency and avoiding severe adverse effects simultaneously. In this study, we designed a controlled release drug delivery system, doxorubicin (Dox)-loaded and hyaluronic acid (HA)-modified PEGylated gold nanocages (AuNCs), which was designated as PEG-HAn-AuNCs/Dox (n represented 10n HA repeating units were modified on each AuNC). In this system, AuNCs were applied as the photothermal cores, Dox was employed as the model drug, HA was applied as the tumor-microenvironment responsive switch to achieve controlled release, and poly (ethylene glycol) (PEG) was used as the stealth polymer to prolong systemic circulation time. Firstly, we evaluated the physical and chemical properties of the PEG-HAn-AuNCs/Dox with different ratios of HA to AuNC and found that PEG-HA4-AuNCs/Dox was the optimal. Secondly, PEG-HA4-AuNCs/Dox revealed the feature of controlled release, namely, the drug release was triggered by hyaluronidase (HAase) and accelerated by the acidic pH and near-infrared (NIR) irradiation. And then PEG-HA4-AuNCs/Dox could be effectively delivered to a cultured SMMC-7721 cell line in vitro and the tumor tissues of the subcutaneous mouse models of hepatocellular carcinoma (HCC) in vivo. Finally, the results demonstrated the synergetic therapy, namely the combination of chemotherapy and photothermal therapy (PTT) (defined as chemo-photothermal therapy) mediated by PEG-HA4-AuNCs/Dox, could efficiently inhibit the tumor growth both in vitro and in vivo. Therefore, the advantages of PEG-HA4-AuNCs/Dox endowed it as a great potential candidate for HCC treatment.
Collapse
Affiliation(s)
- Huili Li
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Huanjie Li
- General Hospital of Xuzhou Mining Group, Quanshan District Coal Road No. 32, Xuzhou, 221006, PR China
| | - Wei Yu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Shengnan Huang
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Ying Liu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Ningxia Zhang
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Jinxiu Yuan
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Xin Xu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China
| | - Shaofeng Duan
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, PR China; Henan International Joint Laboratory for Nuclear Protein Regulation, School of Medical Sciences, Henan University, N. Jinming Ave., Kaifeng, Henan 475004, PR China.
| | - Yurong Hu
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China; Key Laboratory of Key Technology of Drug Preparation, Ministry of Education, Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou 450001, PR China.
| |
Collapse
|
22
|
Pawar A, Prabhu P. Nanosoldiers: A promising strategy to combat triple negative breast cancer. Biomed Pharmacother 2019; 110:319-341. [DOI: 10.1016/j.biopha.2018.11.122] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/10/2018] [Accepted: 11/25/2018] [Indexed: 12/16/2022] Open
|
23
|
Liang W, Huang Y, Lu D, Ma X, Gong T, Cui X, Yu B, Yang C, Dong C, Shuang S. β-Cyclodextrin⁻Hyaluronic Acid Polymer Functionalized Magnetic Graphene Oxide Nanocomposites for Targeted Photo-Chemotherapy of Tumor Cells. Polymers (Basel) 2019; 11:E133. [PMID: 30960117 PMCID: PMC6401939 DOI: 10.3390/polym11010133] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/29/2018] [Accepted: 01/08/2019] [Indexed: 01/25/2023] Open
Abstract
A multifunctional targeted drug delivery platform (CDHA⁻MGO) has been successfully constructed by grafting β-cyclodextrin⁻hyaluronic acid polymers (CDHA) to Fe₃O₄⁻graphene oxide (MGO). The obtained CDHA⁻MGO nanocomposite has good water-dispersibility, easy magnetic separation, high near-infrared (NIR) photothermal heating, and excellent biocompatibility. The β-cyclodextrin-hyaluronic acid polymers efficaciously enhance the doxorubicin (DOX) loading amount up to 485.43 mg·g-1. Meanwhile, the Fe₃O₄⁻graphene oxide provides a facile photothermal response mechanism to handle the NIR-triggered release of DOX in weak acidic solvent environments. Significantly, the DOX-loaded nanocomposite (DOX@CDHA⁻MGO) has displayed CD44 receptor-mediated active targeting recognition and chemo-photothermal synergistic therapy of hepatoma cells. These findings suggest that the as-prepared drug delivery platform would be of valuable potential for cancer-targeted photo-chemotherapy.
Collapse
Affiliation(s)
- Wenting Liang
- Institute of Environmental Science, Department of Chemistry, Shanxi University, Taiyuan 030006, China.
| | - Yu Huang
- Institute of Environmental Science, Department of Chemistry, Shanxi University, Taiyuan 030006, China.
| | - Dongtao Lu
- Institute of Environmental Science, Department of Chemistry, Shanxi University, Taiyuan 030006, China.
| | - Xuewen Ma
- Institute of Environmental Science, Department of Chemistry, Shanxi University, Taiyuan 030006, China.
| | - Tao Gong
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China.
| | - Xiaodong Cui
- Institute of Environmental Science, Department of Chemistry, Shanxi University, Taiyuan 030006, China.
| | - Baofeng Yu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China.
| | - Cheng Yang
- State Key Laboratory of Biotherapy, College of Chemistry, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Chuan Dong
- Institute of Environmental Science, Department of Chemistry, Shanxi University, Taiyuan 030006, China.
| | - Shaomin Shuang
- Institute of Environmental Science, Department of Chemistry, Shanxi University, Taiyuan 030006, China.
| |
Collapse
|
24
|
Sun J, Wang Z, Cao A, Sheng R. Synthesis of crosslinkable diblock terpolymers PDPA-b-P(NMS-co-OEG) and preparation of shell-crosslinked pH/redox-dual responsive micelles as smart nanomaterials. RSC Adv 2019; 9:34535-34546. [PMID: 35529956 PMCID: PMC9073896 DOI: 10.1039/c9ra05082e] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/25/2019] [Indexed: 12/16/2022] Open
Abstract
Crosslinked polymer nanomaterials have attracted great attention due to their stability and highly controllable drug delivery; herein, a series of well-defined amphiphilic PDPA-b-P(NMS-co-OEG) diblock terpolymers (P1–P3) were designed and prepared via RAFT polymerization and were self-assembled into non-cross-linked (NCL) nanomicelles, which were further prepared into shell-cross-linked (SCL) micelles via cystamine-based in situ shell cross-linking. Using P3 as an optimized polymer, SCL-P3 micelles were prepared, which demonstrated remarkable pH/redox-dual responsive behaviour. For drug delivery, camptothecin (CPT)-loaded SCL-P3 micelles were prepared and showed much higher CPT-loading capability than their NCL-P3 counterparts. Notably, the SCL-P3 micelles showed good synergistic pH/redox-dual responsive CPT release properties, making them potential “smart” nanocarriers for drug delivery. A series of well-defined amphiphilic PDPA-b-P(NMS-co-OEG) diblock terpolymers were prepared via RAFT polymerization and self-assembled into non-cross-linked nanomicelles, and then shell-cross-linked micelles via cystamine-based in situ shell cross-linking.![]()
Collapse
Affiliation(s)
- Jingjing Sun
- Department of Radiology
- Shanghai Tenth People's Hospital
- School of Medicine
- Tongji University
- Shanghai 200072
| | - Zhao Wang
- Key Laboratory of Synthetic and Self-assembly Chemistry for Organic Functional Molecules
- Shanghai Institute of Organic Chemistry
- CAS
- Shanghai
- China
| | - Amin Cao
- Key Laboratory of Synthetic and Self-assembly Chemistry for Organic Functional Molecules
- Shanghai Institute of Organic Chemistry
- CAS
- Shanghai
- China
| | - Ruilong Sheng
- Department of Radiology
- Shanghai Tenth People's Hospital
- School of Medicine
- Tongji University
- Shanghai 200072
| |
Collapse
|
25
|
Alsuraifi A, Curtis A, Lamprou DA, Hoskins C. Stimuli Responsive Polymeric Systems for Cancer Therapy. Pharmaceutics 2018; 10:E136. [PMID: 30131473 PMCID: PMC6161138 DOI: 10.3390/pharmaceutics10030136] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 12/14/2022] Open
Abstract
Nanoscale polymers systems have dominated the revolution of drug delivery advancement. Their potential in the fight against cancer is unrivalled with other technologies. Their functionality increase, targeting ability and stimuli responsive nature have led to a major boom in research focus. This review article concentrates on the use of these smart polymers in cancer therapy. Nanotechnologies have shown potential as drug carriers leading to increased drug efficacy and penetration. Multifunctional smart carriers which can release their payload upon an external or internal trigger such as pH or temperature are proving to be major frontrunners in the development of effective strategies to overcome this disease with minimal patient side effects.
Collapse
Affiliation(s)
- Ali Alsuraifi
- Institute of Science and Technology in Medicine, Keele University, Keele ST5 5BG, UK.
- College of Dentistry, University of Basrah, Basrah 61004, Iraq.
| | - Anthony Curtis
- Institute of Science and Technology in Medicine, Keele University, Keele ST5 5BG, UK.
| | | | - Clare Hoskins
- Institute of Science and Technology in Medicine, Keele University, Keele ST5 5BG, UK.
| |
Collapse
|
26
|
Barattin M, Mattarei A, Balasso A, Paradisi C, Cantù L, Del Favero E, Viitala T, Mastrotto F, Caliceti P, Salmaso S. pH-Controlled Liposomes for Enhanced Cell Penetration in Tumor Environment. ACS APPLIED MATERIALS & INTERFACES 2018; 10:17646-17661. [PMID: 29737834 DOI: 10.1021/acsami.8b03469] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
An innovative pH-switchable colloidal system that can be exploited for site-selective anticancer drug delivery has been generated by liposome decoration with a new novel synthetic non-peptidic oligo-arginine cell-penetration enhancer (CPE) and a quenching PEGylated counterpart that detaches from the vesicle surface under the acidic conditions of tumors. The CPE module ( Arg4- DAG) is formed by four arginine units conjugated to a first-generation (G1) 2,2-bis(hydroxymethyl)propionic acid (bis-MPA)/2,2-bis(aminomethyl)propionic acid (bis-AMPA) polyester dendron terminating with 1,2-distearoyl-3-azidopropane for liposome bilayer insertion. The zeta potential of the Arg4- DAG-decorated liposomes increased up to +32 mV as the Arg4- DAG/lipids molar ratio increased. The Arg4- DAG liposome shielding at pH 7.4 was provided by methoxy-PEG5 kDa-polymethacryloyl sulfadimethoxine (mPEG5 kDa-SDM8) with 7.1 apparent p Ka. Zeta potential, surface plasmon resonance and synchrotron small-angle X-ray scattering analyses showed that at pH 7.4 mPEG5 kDa-SDM8 associates with polycationic Arg4- DAG-decorated liposomes yielding liposomes with neutral zeta potential. At pH 6.5, which mimics the tumor environment, mPEG5 kDa-SDM8 detaches from the liposome surface yielding Arg4- DAG exposure. Flow cytometry and confocal microscopy showed a 30-fold higher HeLa cancer cell association of the Arg4- DAG-decorated liposomes compared to non-decorated liposomes. At pH 7.4, the mPEG5 kDa-SDM8-coated liposomes undergo low cell association while remarkable cell association occurred at pH 6.5, which allowed for the controlled intracellular delivery of model macromolecules and small molecules loaded in the liposome under tumor conditions.
Collapse
Affiliation(s)
- Michela Barattin
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| | - Andrea Mattarei
- Department of Chemical Sciences , University of Padova , Via F. Marzolo 1 , Padova 35131 , Italy
| | - Anna Balasso
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| | - Cristina Paradisi
- Department of Chemical Sciences , University of Padova , Via F. Marzolo 1 , Padova 35131 , Italy
| | - Laura Cantù
- Department of Medical Biotechnologies and Traslational Medicine , University of Milano , LITA, Via F.lli Cervi, 93 , Segrate 20090 , Italy
| | - Elena Del Favero
- Department of Medical Biotechnologies and Traslational Medicine , University of Milano , LITA, Via F.lli Cervi, 93 , Segrate 20090 , Italy
| | - Tapani Viitala
- Centre for Drug Research and Division of Pharmaceutical Biosciences, Faculty of Pharmacy , University of Helsinki , Viikinkaari 5 , Helsinki FI-00014 , Finland
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Via F. Marzolo 5 , Padova 35131 , Italy
| |
Collapse
|
27
|
Zhang B, Yang X, Li P, Guo C, Ren X, Li J. Preparation of chitosan sulfate and vesicle formation with a conventional cationic surfactant. Carbohydr Polym 2018; 183:240-245. [DOI: 10.1016/j.carbpol.2017.12.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/06/2017] [Accepted: 12/13/2017] [Indexed: 11/25/2022]
|
28
|
Zhu P, Liu Z, Nie J, He Y. Reversible CO2-Responsive and Photopolymerizable Prepolymers for Stepwise Regulation on Demand. Ind Eng Chem Res 2018. [DOI: 10.1021/acs.iecr.7b04383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Pengcheng Zhu
- State
Key Laboratory of Chemical Resource Engineering, Ministry of Education, Beijing University of Chemical Technology, Beijing 100029, PR China
- Key
Laboratory of Carbon Fiber and Functional Polymers (Beijing University
of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing 100029, PR China
- College
of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Zhixin Liu
- College
of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Jun Nie
- State
Key Laboratory of Chemical Resource Engineering, Ministry of Education, Beijing University of Chemical Technology, Beijing 100029, PR China
- College
of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| | - Yong He
- Key
Laboratory of Carbon Fiber and Functional Polymers (Beijing University
of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing 100029, PR China
- College
of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, PR China
| |
Collapse
|
29
|
Zaki AM, Carbone P. How the Incorporation of Pluronic Block Copolymers Modulates the Response of Lipid Membranes to Mechanical Stress. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:13284-13294. [PMID: 29084428 DOI: 10.1021/acs.langmuir.7b02244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
We employ atomistic molecular dynamics simulations to investigate the effect that the incorporation of the nonionic amphiphilic copolymer known as Pluronic L64 has on the mechanical stability of a DPPC membrane. The simulations reveal that the incorporation of the polymer chains leads to membranes that can sustain increasing mechanical stresses. Analysis of mechanical, structural, and dynamic properties of the membrane shows that the polymer chains interact strongly with the lipids in the vicinity, restraining their mobility and imparting better mechanical stability to the membrane. The hybrid membranes under tension remain thicker, more ordered, and stiffer in comparison to their lipid analogues. Trans-bilayer lipid movements (flip-flop) are observed and appear to be triggered by the presence of the polymer chains. A careful analysis of the pore formation under high tensions reveals two distinctive mechanisms that depend on the distribution of the hydrophilic polymer blocks in the bilayer. Finally, the rate of growth of the formed membrane defects is slowed down in the presence of polymers. These findings show that Pluronic block copolymers could be exploited for the formation of optimized hybrid nanodevices with controlled elastic and dynamic properties.
Collapse
Affiliation(s)
- Afroditi Maria Zaki
- School of Chemical Engineering and Analytical Science, The University of Manchester , Oxford Road, Manchester M13 9PL, United Kingdom
| | - Paola Carbone
- School of Chemical Engineering and Analytical Science, The University of Manchester , Oxford Road, Manchester M13 9PL, United Kingdom
| |
Collapse
|
30
|
de Vries WC, Grill D, Tesch M, Ricker A, Nüsse H, Klingauf J, Studer A, Gerke V, Ravoo BJ. Reversible Stabilisierung von Vesikeln: redox-responsive Polymer-Nanocontainer für den Transport in das Zellinnere. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201702620] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Wilke C. de Vries
- Organisch-Chemisches Institut und Center for Soft Nanoscience; Westfälische Wilhelms-Universität Münster; Correnstraße 40 48149 Münster Deutschland
| | - David Grill
- Institut für Medizinische Biochemie, Zentrum für Molekularbiologie der Entzündung; Westfälische Wilhelms-Universität Münster; Von-Esmarch-Straße 56 48149 Münster Deutschland
| | - Matthias Tesch
- Organisch-Chemisches Institut und Center for Soft Nanoscience; Westfälische Wilhelms-Universität Münster; Correnstraße 40 48149 Münster Deutschland
| | - Andrea Ricker
- Institut für Medizinische Physik und Biophysik; Westfälische Wilhelms-Universität Münster; Robert-Koch-Straße 31 48149 Münster Deutschland
| | - Harald Nüsse
- Institut für Medizinische Physik und Biophysik; Westfälische Wilhelms-Universität Münster; Robert-Koch-Straße 31 48149 Münster Deutschland
| | - Jürgen Klingauf
- Institut für Medizinische Physik und Biophysik; Westfälische Wilhelms-Universität Münster; Robert-Koch-Straße 31 48149 Münster Deutschland
| | - Armido Studer
- Organisch-Chemisches Institut und Center for Soft Nanoscience; Westfälische Wilhelms-Universität Münster; Correnstraße 40 48149 Münster Deutschland
| | - Volker Gerke
- Institut für Medizinische Biochemie, Zentrum für Molekularbiologie der Entzündung; Westfälische Wilhelms-Universität Münster; Von-Esmarch-Straße 56 48149 Münster Deutschland
| | - Bart Jan Ravoo
- Organisch-Chemisches Institut und Center for Soft Nanoscience; Westfälische Wilhelms-Universität Münster; Correnstraße 40 48149 Münster Deutschland
| |
Collapse
|
31
|
de Vries WC, Grill D, Tesch M, Ricker A, Nüsse H, Klingauf J, Studer A, Gerke V, Ravoo BJ. Reversible Stabilization of Vesicles: Redox-Responsive Polymer Nanocontainers for Intracellular Delivery. Angew Chem Int Ed Engl 2017; 56:9603-9607. [DOI: 10.1002/anie.201702620] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/18/2017] [Indexed: 12/28/2022]
Affiliation(s)
- Wilke C. de Vries
- Organic Chemistry Institute and Center for Soft Nanoscience; Westfälische Wilhelms-Universität Münster; Correnstrasse 40 48149 Münster Germany
| | - David Grill
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation; Westfälische Wilhelms-Universität Münster; Von-Esmarch-Strasse 56 48149 Münster Germany
| | - Matthias Tesch
- Organic Chemistry Institute and Center for Soft Nanoscience; Westfälische Wilhelms-Universität Münster; Correnstrasse 40 48149 Münster Germany
| | - Andrea Ricker
- Institute of Medical Physics and Biophysics; Westfälische Wilhelms-Universität Münster; Robert-Koch-Strasse 31 48149 Münster Germany
| | - Harald Nüsse
- Institute of Medical Physics and Biophysics; Westfälische Wilhelms-Universität Münster; Robert-Koch-Strasse 31 48149 Münster Germany
| | - Jürgen Klingauf
- Institute of Medical Physics and Biophysics; Westfälische Wilhelms-Universität Münster; Robert-Koch-Strasse 31 48149 Münster Germany
| | - Armido Studer
- Organic Chemistry Institute and Center for Soft Nanoscience; Westfälische Wilhelms-Universität Münster; Correnstrasse 40 48149 Münster Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation; Westfälische Wilhelms-Universität Münster; Von-Esmarch-Strasse 56 48149 Münster Germany
| | - Bart Jan Ravoo
- Organic Chemistry Institute and Center for Soft Nanoscience; Westfälische Wilhelms-Universität Münster; Correnstrasse 40 48149 Münster Germany
| |
Collapse
|
32
|
Cabeza L, Ortiz R, Prados J, Delgado ÁV, Martín-Villena MJ, Clares B, Perazzoli G, Entrena JM, Melguizo C, Arias JL. Improved antitumor activity and reduced toxicity of doxorubicin encapsulated in poly(ε-caprolactone) nanoparticles in lung and breast cancer treatment: An in vitro and in vivo study. Eur J Pharm Sci 2017; 102:24-34. [DOI: 10.1016/j.ejps.2017.02.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 01/31/2017] [Accepted: 02/15/2017] [Indexed: 11/29/2022]
|
33
|
Ling L, Yao C, Du Y, Ismail M, He R, Hou Y, Zhang Y, Li X. Assembled liposomes of dual podophyllotoxin phospholipid: preparation, characterization and in vivo anticancer activity. Nanomedicine (Lond) 2017; 12:657-672. [DOI: 10.2217/nnm-2016-0396] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Aim: A novel amphiphilic prodrug dual podophyllotoxin (PPT) succinate glycerophosphorylcholine (Di-PPT-GPC) assembled liposomes was developed to improve efficiency of PPT. Materials & methods: Di-PPT-GPC liposomes were prepared by thin film technique and characterized by dynamic light scattering and cryo-electron microscopy. Results: In vitro release studies showed that Di-PPT-GPC liposomes could significantly release PPT in weakly acidic environment but had good stability under biological conditions. Methyl tetrazolium assay data revealed that the liposomes have comparable cytotoxicities to free PPT against MCF-7, HeLa and U87 cells. More importantly, in vivo antitumor evaluation indicated that Di-PPT-GPC liposomes exhibited favorable tumor growth inhibition without side effects. Conclusion: Di-PPT-GPC liposomes might have potential to promote the therapeutic effect of PPT for cancer therapy.
Collapse
Affiliation(s)
- Longbing Ling
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Chen Yao
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yawei Du
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Muhammad Ismail
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ruiyu He
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yongpeng Hou
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ying Zhang
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xinsong Li
- School of Chemistry & Chemical Engineering, Southeast University, Nanjing 211189, PR China
| |
Collapse
|
34
|
Wang M. Emerging Multifunctional NIR Photothermal Therapy Systems Based on Polypyrrole Nanoparticles. Polymers (Basel) 2016; 8:E373. [PMID: 30974650 PMCID: PMC6432477 DOI: 10.3390/polym8100373] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/05/2016] [Accepted: 10/10/2016] [Indexed: 01/09/2023] Open
Abstract
Near-infrared (NIR)-light-triggered therapy platforms are now considered as a new and exciting possibility for clinical nanomedicine applications. As a promising photothermal agent, polypyrrole (PPy) nanoparticles have been extensively studied for the hyperthermia in cancer therapy due to their strong NIR light photothermal effect and excellent biocompatibility. However, the photothermal application of PPy based nanomaterials is still in its preliminary stage. Developing PPy based multifunctional nanomaterials for cancer treatment in vivo should be the future trend and object for cancer therapy. In this review, the synthesis of PPy nanoparticles and their NIR photothermal conversion performance were first discussed, followed by a summary of the recent progress in the design and implementation on the mulitifunctionalization of PPy or PPy based therapeutic platforms, as well as the introduction of their exciting biomedical applications based on the synergy between the photothermal conversion effect and other stimulative responsibilities.
Collapse
Affiliation(s)
- Mozhen Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
35
|
Wang J, Zhang X, Cen Y, Lin X, Wu Q. Antitumor gemcitabine conjugated micelles from amphiphilic comb-like random copolymers. Colloids Surf B Biointerfaces 2016; 146:707-15. [DOI: 10.1016/j.colsurfb.2016.07.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 01/08/2023]
|
36
|
Gu L, Wang N, Nusblat LM, Soskind R, Roth CM, Uhrich KE. pH-responsive amphiphilic macromolecular carrier for doxorubicin delivery. J BIOACT COMPAT POL 2016. [DOI: 10.1177/0883911516643219] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In this work, pH-sensitive amphiphilic macromolecules are designed to possess good biocompatibility and drug loading while employing an acid-sensitive linkage to trigger drug release at tumor tissues. Specifically, two pH-sensitive amphiphilic macromolecules were synthesized with a hydrazone linkage between the hydrophobic and hydrophilic segments. The chemical structure, molecular weight, critical micelle concentration, micelle size, and pH-triggered cleavage of the amphiphilic macromolecules were characterized via matrix-assisted laser desorption/ionization time-of-flight, nuclear magnetic resonance, and dynamic light scattering techniques. Drug loading and release as well as cytotoxicity studies were performed using doxorubicin. Hydrodynamic diameters of the micelles formed with pH-sensitive amphiphilic macromolecules were within an optimal range for cellular uptake. The critical micelle concentration values were 10–8–10–6 M, indicating micellar stability upon dilution. The degradation products of the amphiphilic macromolecules after acidic incubation were identified using mass spectrometry, nuclear magnetic resonance, and dynamic light scattering methods. A pH-dependent release profile of the doxorubicin-encapsulated amphiphilic macromolecules was observed. Cytotoxicity studies against two cancer cell lines, MDA-MB-231 human breast cancer cells and A549 lung cancer cells, showed that doxorubicin encapsulated in pH-sensitive amphiphilic macromolecules decreased cell viability more efficiently than free doxorubicin, possibly due to the toxicity of the amphiphilic macromolecule degradation products. Resulting from enhanced release at acidic pH due to hydrolysis of the hydrazone linkage, pH-sensitive amphiphilic macromolecules also had improved efficacy toward cancer cells compared to other carriers (e.g. Pluronics®). These findings indicate that pH-sensitive amphiphilic macromolecules can potentially be applied as anticancer drug delivery vehicles to achieve controlled release and improved therapeutic effects.
Collapse
Affiliation(s)
- Li Gu
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ning Wang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Leora M Nusblat
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Rose Soskind
- Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Charles M Roth
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Kathryn E Uhrich
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| |
Collapse
|
37
|
Sonawane SJ, Kalhapure RS, Rambharose S, Mocktar C, Vepuri SB, Soliman M, Govender T. Ultra-small lipid-dendrimer hybrid nanoparticles as a promising strategy for antibiotic delivery: In vitro and in silico studies. Int J Pharm 2016; 504:1-10. [PMID: 26992817 DOI: 10.1016/j.ijpharm.2016.03.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/13/2016] [Indexed: 11/19/2022]
Abstract
The purpose of this study was to explore the preparation of a new lipid-dendrimer hybrid nanoparticle (LDHN) system to effectively deliver vancomycin against methicillin-resistant Staphylococcus aureus (MRSA) infections. Spherical LDHNs with particle size, polydispersity index and zeta potential of 52.21±0.22 nm, 0.105±0.01, and -14.2±1.49 mV respectively were prepared by hot stirring and ultrasonication using Compritol 888 ATO, G4 PAMAM- succinamic acid dendrimer, and Kolliphor RH-40. Vancomycin encapsulation efficiency (%) in LDHNs was almost 4.5-fold greater than in lipid-polymer hybrid nanoparticles formulated using Eudragit RS 100. Differential scanning calorimetry and Fourier transform-infrared studies confirmed the formation of LDHNs. The interactions between the drug-dendrimer complex and lipid molecules using in silico modeling revealed the molecular mechanism behind the enhanced encapsulation and stability. Vancomycin was released from LDHNs over the period of 72 h with zero order kinetics and super case II transport mechanism. The minimum inhibitory concentration (MIC) against S. aureus and MRSA were 15.62 μg/ml and 7.81 μg/ml respectively. Formulation showed sustained activity with MIC of 62.5 μg/ml against S. aureus and 500 μg/ml against MRSA at the end of 72 and 54 h period respectively. The results suggest that the LDHN system can be an effective strategy to combat resistant infections.
Collapse
Affiliation(s)
- Sandeep J Sonawane
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001 Durban, 4000, South Africa
| | - Rahul S Kalhapure
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001 Durban, 4000, South Africa.
| | - Sanjeev Rambharose
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001 Durban, 4000, South Africa
| | - Chunderika Mocktar
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001 Durban, 4000, South Africa
| | - Suresh B Vepuri
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001 Durban, 4000, South Africa
| | - Mahmoud Soliman
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001 Durban, 4000, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Private Bag X54001 Durban, 4000, South Africa.
| |
Collapse
|
38
|
Rao S, Prestidge CA. Polymer-lipid hybrid systems: merging the benefits of polymeric and lipid-based nanocarriers to improve oral drug delivery. Expert Opin Drug Deliv 2016; 13:691-707. [PMID: 26866382 DOI: 10.1517/17425247.2016.1151872] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION A number of biobarriers limit efficient oral drug absorption; both polymer-based and lipid-based nanocarriers have demonstrated properties and delivery mechanisms to overcome these biobarriers in preclinical settings. Moreover, in order to address the multifaceted oral drug delivery challenges, polymer-lipid hybrid systems are now being designed to merge the beneficial features of both polymeric and lipid-based nanocarriers. AREAS COVERED Recent advances in the development of polymer-lipid hybrids with a specific focus on their viability in oral delivery are reviewed. Three classes of polymer-lipid hybrids have been identified, i.e. lipid-core polymer-shell systems, polymer-core lipid-shell systems, and matrix-type polymer-lipid hybrids. We focus on their application to overcome the various biological barriers to oral drug absorption, as exemplified by selected preclinical studies. EXPERT OPINION Numerous studies have demonstrated the superiority of polymer-lipid hybrid systems to their non-hybrid counterparts in providing improved drug encapsulation, modulated drug release, and improved cellular uptake. These features have encouraged their applications in the delivery of chemotherapeutics, proteins, peptides, and vaccines. With further research expected to optimize the manufacturing and scaling up processes and in-depth pre-clinical pharmacological and toxicological assessments, these multifaceted drug delivery systems will have significant clinical impact on the oral delivery of pharmaceuticals and biopharmaceuticals.
Collapse
Affiliation(s)
- Shasha Rao
- a School of Pharmacy and Medical Sciences , University of South Australia , Adelaide , SA , Australia
| | - Clive A Prestidge
- a School of Pharmacy and Medical Sciences , University of South Australia , Adelaide , SA , Australia
| |
Collapse
|
39
|
Huo ZJ, Wang SJ, Wang ZQ, Zuo WS, Liu P, Pang B, Liu K. Novel nanosystem to enhance the antitumor activity of lapatinib in breast cancer treatment: Therapeutic efficacy evaluation. Cancer Sci 2015; 106:1429-37. [PMID: 26177628 PMCID: PMC4637996 DOI: 10.1111/cas.12737] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 07/02/2015] [Accepted: 07/06/2015] [Indexed: 12/20/2022] Open
Abstract
The present study was performed to investigate the therapeutic performance of polymer-lipid hybrid nanoparticles towards the delivery of lapatinib (LPT) in breast cancers. We have successfully developed the lapatinib-loaded polymer-lipid hybrid nanosystem and showed its therapeutic potential in in vitro and in vivo models of breast cancer. The nanoformulations consisted of a polymeric core (poly[lactide-co-glycolide]-D-a-tocopheryl polyethylene glycol 1000 succinate [PLGA-TPGS]), which was then enveloped by a PEGylated lipid layer (DSPE-PEG) (PLPT) to maintain the structural integrity. The PLPT formulation controlled the drug release in pH 7.4 conditions and accelerated the release at pH 5.5 conditions. The PLPT showed a remarkable cellular internalization and efficiently killed the MCF-7 cancer cells in a time- and concentration-dependent manner. Moreover, LPT-loaded nanoparticles effectively induced apoptosis of cancer cells than compared to free LPT. Pharmacokinetic data suggested that nanoparticles could significantly enhance the blood circulation time of LPT by reducing the uptake by a reticuloendothelial system (RES). The prolonged blood circulation of PLPT could allow the preferential accumulation of drug in the tumor tissues. Importantly, PLPT significantly reduced the tumor burden of cancerous mice and effectively controlled the tumor cell proliferation. TUNEL assay further showed a greater apoptosis of tumor tissues in the PLPT treated mice group. Our results suggest that the use of a hybrid system may allow a decrease in the dosage regimen without the loss of therapeutic effect. Overall, lapatinib-loaded hybrid nanoparticles hold great potential for achieving an optimal therapeutic effect in breast cancer treatment. The present anticancer drug delivery system could be potentially applied for the treatment of other cancers.
Collapse
Affiliation(s)
- Zhi-Jun Huo
- Department of Breast Disease Center, Shandong Cancer Hospital & Institute, Jinan, China
| | - Shi-Jiang Wang
- Department of Radiotherapy, Shandong Cancer Hospital & Institute, Jinan, China
| | - Zhi-Qi Wang
- Department of Head and Neck Surgery, Shandong Cancer Hospital & Institute, Jinan, China
| | - Wen-Shu Zuo
- Department of Breast Disease Center, Shandong Cancer Hospital & Institute, Jinan, China
| | - Ping Liu
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, China
| | - Bo Pang
- Department of Neurosurgery, Qilu Hospital, Shandong University, Jinan, China
| | - Kai Liu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital & Institute, Jinan, China
| |
Collapse
|
40
|
Zhang L, Wang Y, Yang Y, Liu Y, Ruan S, Zhang Q, Tai X, Chen J, Xia T, Qiu Y, Gao H, He Q. High Tumor Penetration of Paclitaxel Loaded pH Sensitive Cleavable Liposomes by Depletion of Tumor Collagen I in Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2015; 7:9691-9701. [PMID: 25845545 DOI: 10.1021/acsami.5b01473] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The network of collagen I in tumors could prevent the penetration of drugs loaded in nanoparticles, and this would lead to impaired antitumor efficacy. In this study, free losartan (an angiotensin inhibitor) was injected before treatment to reduce the level of collagen I, which could facilitate the penetration of nanoparticles. Then the pH-sensitive cleavable liposomes (Cl-Lip) were injected subsequently to exert the antitumor effect. The Cl-Lip was constituted by PEG(5K)-Hydrazone-PE and DSPE-PEG(2K)-R8. When the Cl-Lip reached to the tumor site by the enhanced permeability and retention (EPR) effect, PEG(5K)-Hydrazone-PE was hydrolyzed from the Cl-Lip under the low extra-cellular pH conditions of tumors, then the R8 peptide was exposed, and finally liposomes could be internalized into tumor cells by the mediation of R8 peptide. In vitro experiments showed both the cellular uptake of Cl-Lip by 4T1 cells and cytotoxicity of paclitaxel loaded Cl-Lip (PTX-Cl-Lip) were pH sensitive. In vivo experiments showed the Cl-Lip had a good tumor targeting ability. After depletion of collagen I, Cl-Lip could penetrate into the deep place of tumors, the tumor accumulation of Cl-Lip was further increased by 22.0%, and the oxygen distributed in tumor tissues was also enhanced. The antitumor study indicated free losartan in combination with PTX-Cl-Lip (59.8%) was more effective than injection with PTX-Cl-Lip only (37.8%) in 4T1 tumor bearing mice. All results suggested that depletion of collagen I by losartan dramatically increased the penetration of PTX-Cl-Lip and combination of free losartan and PTX-CL-Lip could lead to better antitumor efficacy of chemical drugs. Thus, the combination strategy might be a promising tactic for better treatment of solid tumors with a high level of collagen I.
Collapse
Affiliation(s)
- Li Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Yang Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Yuting Yang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Yayuan Liu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Shaobo Ruan
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Qianyu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Xiaowei Tai
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Jiantao Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Tai Xia
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Yue Qiu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| | - Qin He
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, China
| |
Collapse
|
41
|
Miller-Kleinhenz JM, Bozeman EN, Yang L. Targeted nanoparticles for image-guided treatment of triple-negative breast cancer: clinical significance and technological advances. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 7:797-816. [PMID: 25966677 DOI: 10.1002/wnan.1343] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/23/2015] [Accepted: 03/07/2015] [Indexed: 12/21/2022]
Abstract
Effective treatment of triple-negative breast cancer (TNBC) with its aggressive tumor biology, highly heterogeneous tumor cells, and poor prognosis requires an integrated therapeutic approach that addresses critical issues in cancer therapy. Multifunctional nanoparticles with the abilities of targeted drug delivery and noninvasive imaging for monitoring drug delivery and responses to therapy, such as theranostic nanoparticles, hold great promise toward the development of novel therapeutic approaches for the treatment of TNBC using a single therapeutic platform. The biological and pathological characteristics of TNBC provide insight into several potential molecular targets for current and future nanoparticle-based therapeutics. Extensive tumor stroma, highly proliferative cells, and a high rate of drug resistance are all barriers that must be appropriately addressed in order for these nanotherapeutic platforms to be effective. Utilization of the enhanced permeability and retention effect coupled with active targeting of cell surface receptors expressed by TNBC cells, and tumor-associated endothelial cells, stromal fibroblasts, and macrophages is likely to overcome such barriers to facilitate more effective drug delivery. An in-depth summary of current studies investigating targeted nanoparticles in preclinical TNBC mouse and human xenograft models is presented. This review aims to outline the current status of nanotherapeutic options for TNBC patients, identification of promising molecular targets, challenges associated with the development of targeted nanotherapeutics, the research done by our group as well as by others, and future perspectives on the nanomedicine field and ways to translate current preclinical studies into the clinic.
Collapse
Affiliation(s)
| | - Erica N Bozeman
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Lily Yang
- Winship Cancer Institute, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
42
|
Fang S, Niu Y, Zhu W, Zhang Y, Yu L, Li X. Liposomes assembled from a dual drug-tailed phospholipid for cancer therapy. Chem Asian J 2015; 10:1232-8. [PMID: 25690917 DOI: 10.1002/asia.201500067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Indexed: 12/29/2022]
Abstract
We report a novel dual drug-tailed phospholipid which can form liposomes as a combination of prodrug and drug carrier. An amphiphilic dual chlorambucil-tailed phospholipid (DCTP) was synthesized by a straightforward esterification. With two chlorambucil molecules as hydrophobic tails and one glycerophosphatidylcholine molecule as a hydrophilic head, the DCTP, a phospholipid prodrug, undergoes assembly to form a liposome without any additives by the thin lipid film technique. The DCTP liposomes, as an effective carrier of chlorambucil, exhibited a very high loading capacity and excellent stability. The liposomes had higher cytotoxic effects to cancer cell lines than free DCTP and chlorambucil. The in vivo antitumor activity assessment indicated that the DCTP liposomes could inhibit the tumor growth effectively. This novel strategy of dual drug-tailed phospholipid liposomes may be also applicable to other hydrophobic anticancer drugs which have great potential in cancer therapy.
Collapse
Affiliation(s)
- Shuo Fang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing 210096 (P.R. China)
| | | | | | | | | | | |
Collapse
|
43
|
Park JH, Cho HJ, Termsarasab U, Lee JY, Ko SH, Shim JS, Yoon IS, Kim DD. Interconnected hyaluronic acid derivative-based nanoparticles for anticancer drug delivery. Colloids Surf B Biointerfaces 2014; 121:380-7. [DOI: 10.1016/j.colsurfb.2014.06.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 06/08/2014] [Accepted: 06/09/2014] [Indexed: 01/17/2023]
|
44
|
Yan Q, Zhao Y. Block copolymer self-assembly controlled by the “green” gas stimulus of carbon dioxide. Chem Commun (Camb) 2014; 50:11631-41. [DOI: 10.1039/c4cc03412k] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
CO2 can act as a “green” stimulus to precisely tune the self-assembly of block copolymers.
Collapse
Affiliation(s)
- Qiang Yan
- Département de chimie
- Université de Sherbrooke
- Sherbrooke, Canada J1K 2R1
| | - Yue Zhao
- Département de chimie
- Université de Sherbrooke
- Sherbrooke, Canada J1K 2R1
| |
Collapse
|
45
|
Lee SM, Nguyen ST. Smart Nanoscale Drug Delivery Platforms from Stimuli-Responsive Polymers and Liposomes. Macromolecules 2013; 46:9169-9180. [PMID: 28804160 PMCID: PMC5552073 DOI: 10.1021/ma401529w] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Since the 1960's, stimuli-responsive polymers have been utilized as functional soft materials for biological applications such as the triggered-release delivery of biologically active cargos. Over the same period, liposomes have been explored as an alternative drug delivery system with potentials to decrease the toxic side effects often associated with conventional small-molecule drugs. However, the lack of drug-release triggers and the instability of bare liposomes often limit their practical applications, causing short circulation time and low therapeutic efficacy. This perspective article highlights recent work in integrating these two materials together to achieve a targetable, triggerable nanoscale platform that fulfills all the characteristics of a near-ideal drug delivery system. Through a drop-in, post-synthesis modification strategy, a network of stimuli-responsive polymers can be integrated onto the surface of liposomes to form polymer-caged nanobins, a multifunctional nanoscale delivery platform that allows for multi-drug loading, targeted delivery, triggered drug-release, and theranostic capabilities.
Collapse
Affiliation(s)
- Sang-Min Lee
- Department of Chemistry and Center of Cancer Nanotechnology Excellence, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113
- Department of Chemistry, The Catholic University of Korea, Bucheon, Gyeonggi-do 420-743 Korea
| | - SonBinh T. Nguyen
- Department of Chemistry and Center of Cancer Nanotechnology Excellence, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113
| |
Collapse
|
46
|
Johnson R, Sabnis N, McConathy WJ, Lacko AG. The potential role of nanotechnology in therapeutic approaches for triple negative breast cancer. Pharmaceutics 2013; 5:353-70. [PMID: 24244833 PMCID: PMC3826456 DOI: 10.3390/pharmaceutics5020353] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Triple Negative Breast Cancer, TNBC, a highly aggressive and metastatic type of breast cancer, is characterized by loss of expression of the estrogen receptor (ER), progesterone receptor (PR), and a lack of overexpression of the human epidermal growth factor receptor 2 (HER2). It is a heterogeneous group of tumors with diverse histology, molecular uniqueness and response to treatment. Unfortunately, TNBC patients do not benefit from current anti-HER2 or hormone positive targeted breast cancer treatments; consequently, these patients rely primarily on chemotherapy. However, the 5-year survival rate for woman with metastatic TNBC is less than 30%. As a result of ineffective treatments, TNBC tumors often progress to metastatic lesions in the brain and lung. Brain metastases of invasive breast cancer are associated with 1 and 2 year survival rate of 20% and <2% respectively. Because the only current systemic treatment for TNBC is chemotherapy, alternative targeted therapies are urgently needed to improve the prognosis for TNBC patients. This review is focused on opportunities for developing new approaches for filling the current void in an effective treatment for TNBC patients.
Collapse
Affiliation(s)
- Rebecca Johnson
- Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; E-Mails: (R.J.); (N.S.)
| | - Nirupama Sabnis
- Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; E-Mails: (R.J.); (N.S.)
- LipoMedics LLC., Fort Worth, TX 76107, USA; E-Mail: (W.J.M.)
| | | | - Andras G. Lacko
- Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; E-Mails: (R.J.); (N.S.)
- LipoMedics LLC., Fort Worth, TX 76107, USA; E-Mail: (W.J.M.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-817-735-2132
| |
Collapse
|
47
|
Hong BJ, Swindell EP, MacRenaris KW, Hankins PL, Chipre AJ, Mastarone DJ, Ahn RW, Meade TJ, O’Halloran TV, Nguyen ST. pH-Responsive Theranostic Polymer-Caged Nanobins (PCNs): Enhanced Cytotoxicity and T1 MRI Contrast by Her2-Targeting. PARTICLE & PARTICLE SYSTEMS CHARACTERIZATION : MEASUREMENT AND DESCRIPTION OF PARTICLE PROPERTIES AND BEHAVIOR IN POWDERS AND OTHER DISPERSE SYSTEMS 2013; 30:770-774. [PMID: 24516291 PMCID: PMC3916701 DOI: 10.1002/ppsc.201300158] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
A PCN theranostic platform comprises a doxorubicin (DXR)-loaded liposomal core and an acid-sensitive polymer shell that is functionalized with Herceptin and GdIII-based MRI contrast agents. In vitro testing reveals a 14-fold increase in DXR-based cytotoxicity versus a non-targeted analogue and an 120-fold improvement in cellular GdIII-uptake in comparison with clinically approved DOTA-GdIII, leading to significant T1 MRI contrast enhancement.
Collapse
Affiliation(s)
- Bong Jin Hong
- Department of Chemistry, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| | - Elden P. Swindell
- Department of Chemical & Biological Engineering, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| | - Keith W. MacRenaris
- Department of Chemistry, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| | - Patrick L. Hankins
- Department of Chemistry, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| | - Anthony J. Chipre
- Department of Chemistry, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| | - Daniel J. Mastarone
- Department of Chemistry, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| | - Richard W. Ahn
- Department of Chemistry, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| | - Thomas J. Meade
- Department of Chemistry, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| | - Thomas V. O’Halloran
- Department of Chemistry, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| | - SonBinh T. Nguyen
- Department of Chemistry, Northwestern University, 2145 Sheridan Rd. Evanston, IL 60208-3113 (USA)
| |
Collapse
|
48
|
Srinivas G, Mohan RV, Kelkar AD. Polymer micelle assisted transport and delivery of model hydrophilic components inside a biological lipid vesicle: a coarse-grain simulation study. J Phys Chem B 2013; 117:12095-104. [PMID: 23952604 DOI: 10.1021/jp405381k] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Understanding drug transportation and delivery mechanism from a molecular viewpoint is essential to find better treatment pathways. Despite the fact that many significant drugs such as anticancer doxorubicin and mitoxantrone are predominantly hydrophilic, an efficient methodology to deliver hydrophilic drug components is not well established. Here we explore this problem by studying "patchy" polymeric micelle assisted hydrophilic component transportation across a lipid membrane and delivery inside a biological lipid vesicle. Using the MARTINI force field as the basis, we study the interaction of polymeric micelle with DPPC lipid vesicles in detail. In order to facilitate hydrophilic drug transportation study, a primitive CG model for hydrophilic drug component is used. Extensive simulations carried out over hundreds of nanoseconds demonstrate successful encapsulation, transportation of hydrophilic components by patchy polymeric micelles. Results show the polymeric micelle releases a significant portion of hydrophilic contents inside the lipid vesicle. The present simulation study also reveals a possible mechanism for efficient hydrophilic component transportation and delivery. Insights from this study could potentially help the experimental community to design better delivery vehicles, especially for hydrophilic drug molecules.
Collapse
Affiliation(s)
- Goundla Srinivas
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering (JSNN), North Carolina State A&T University , 2907 East Lee Street, Greensboro, North Carolina 27401, United States
| | | | | |
Collapse
|
49
|
Zhai Y, Guo S, Liu C, Yang C, Dou J, Li L, Zhai G. Preparation and in vitro evaluation of apigenin-loaded polymeric micelles. Colloids Surf A Physicochem Eng Asp 2013. [DOI: 10.1016/j.colsurfa.2013.03.051] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
50
|
O'Halloran TV, Ahn R, Hankins P, Swindell E, Mazar AP. The many spaces of uPAR: delivery of theranostic agents and nanobins to multiple tumor compartments through a single target. Am J Cancer Res 2013; 3:496-506. [PMID: 23843897 PMCID: PMC3706693 DOI: 10.7150/thno.4953] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 02/18/2013] [Indexed: 12/22/2022] Open
Abstract
The urokinase plasminogen activator (uPA) system is a proteolytic system comprised of uPA, a cell surface receptor for uPA (uPAR), and an inhibitor of uPA (PAI-1) and is implicated in many aspects of tumor growth and metastasis. The uPA system has been identified in nearly all solid tumors examined to date as well as several hematological malignancies. In adults, transient expression of the uPA system is observed during wound healing and inflammatory processes while only limited expression is identified in healthy, quiescent tissue. Members of the uPA system are expressed not only on cancer cells but also on tumor-associated stromal cells. These factors make the uPA system an ideal therapeutic target for cancer therapies. To date most therapeutics targeted at the uPA system have been inhibitors of either the uPA-uPAR interaction or uPA proteolysis but have not shown robust anti-tumor activity. There is now mounting evidence that uPAR participates in a complex signaling network central to its role in cancer progression, which provides a basis for the hypothesis that uPAR may be a marker for cancer stem cells. Several new uPAR-directed therapies have recently been developed based on this new information. A monoclonal antibody has been developed that disrupts the interactions of uPAR with signaling partners and is poised to enter the clinic. In addition, nanoscale drug delivery vehicles targeted to the uPA system using monoclonal antibodies, without disrupting the normal functioning of the system, are also in development. This review will highlight some of these new discoveries and the new uPA system-based therapeutic approaches that have arisen from them.
Collapse
|