1
|
Su Y, Jin G, Zhou H, Yang Z, Wang L, Mei Z, Jin Q, Lv S, Chen X. Development of stimuli responsive polymeric nanomedicines modulating tumor microenvironment for improved cancer therapy. MEDICAL REVIEW (2021) 2023; 3:4-30. [PMID: 37724108 PMCID: PMC10471091 DOI: 10.1515/mr-2022-0048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 02/16/2023] [Indexed: 09/20/2023]
Abstract
The complexity of the tumor microenvironment (TME) severely hinders the therapeutic effects of various cancer treatment modalities. The TME differs from normal tissues owing to the presence of hypoxia, low pH, and immune-suppressive characteristics. Modulation of the TME to reverse tumor growth equilibrium is considered an effective way to treat tumors. Recently, polymeric nanomedicines have been widely used in cancer therapy, because their synthesis can be controlled and they are highly modifiable, and have demonstrated great potential to remodel the TME. In this review, we outline the application of various stimuli responsive polymeric nanomedicines to modulate the TME, aiming to provide insights for the design of the next generation of polymeric nanomedicines and promote the development of polymeric nanomedicines for cancer therapy.
Collapse
Affiliation(s)
- Yuanzhen Su
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Materials Science and Engineering, Peking University, Beijing, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui Province, China
| | - Guanyu Jin
- School of Materials Science and Engineering, Peking University, Beijing, China
- Department of Chemistry, Capital Normal University, Beijing, China
| | - Huicong Zhou
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Zhaofan Yang
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Lanqing Wang
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Zi Mei
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Qionghua Jin
- Department of Chemistry, Capital Normal University, Beijing, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
- School of Materials Science and Engineering, Peking University, Beijing, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui Province, China
| |
Collapse
|
2
|
Application of Gold Nanoparticle-Based Materials in Cancer Therapy and Diagnostics. CHEMENGINEERING 2021. [DOI: 10.3390/chemengineering5040069] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Several metal nanoparticles have been developed for medical application. While all have their benefits, gold nanoparticles (AuNPs) are ideal in cancer therapy and diagnosis as they are chemically inert and minimally toxic. Several studies have shown the potential of AuNPs in the therapeutic field, as photosensitizing agents in sonochemical and photothermal therapy and as drug delivery, as well as in diagnostics and theranostics. Although there is a significant number of reviews on the application of AuNPs in cancer medicine, there is no comprehensive review on their application both in therapy and diagnostics. Therefore, considering the high number of studies on AuNPs’ applications, this review summarizes data on the application of AuNPs in cancer therapy and diagnostics. In addition, we looked at the influence of AuNPs’ shape and size on their biological properties. We also present the potential use of hybrid materials based on AuNPs in sonochemical and photothermal therapy and the possibility of their use in diagnostics. Despite their potential, the use of AuNPs and derivatives in cancer medicine still has some limitations. In this review, we provide an overview of the biological, physicochemical, and legal constraints on using AuNPs in cancer medicine.
Collapse
|
3
|
Shin Y, Husni P, Kang K, Lee D, Lee S, Lee E, Youn Y, Oh K. Recent Advances in pH- or/and Photo-Responsive Nanovehicles. Pharmaceutics 2021; 13:725. [PMID: 34069233 PMCID: PMC8157172 DOI: 10.3390/pharmaceutics13050725] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 01/10/2023] Open
Abstract
The combination of nanotechnology and chemotherapy has resulted in more effective drug design via the development of nanomaterial-based drug delivery systems (DDSs) for tumor targeting. Stimulus-responsive DDSs in response to internal or external signals can offer precisely controlled delivery of preloaded therapeutics. Among the various DDSs, the photo-triggered system improves the efficacy and safety of treatment through spatiotemporal manipulation of light. Additionally, pH-induced delivery is one of the most widely studied strategies for targeting the acidic micro-environment of solid tumors. Accordingly, in this review, we discuss representative strategies for designing DDSs using light as an exogenous signal or pH as an endogenous trigger.
Collapse
Affiliation(s)
- Yuseon Shin
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University and College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Seoul 06974, Korea; (Y.S.); (P.H.); (K.K.); (D.L.); (S.L.)
| | - Patihul Husni
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University and College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Seoul 06974, Korea; (Y.S.); (P.H.); (K.K.); (D.L.); (S.L.)
| | - Kioh Kang
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University and College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Seoul 06974, Korea; (Y.S.); (P.H.); (K.K.); (D.L.); (S.L.)
| | - Dayoon Lee
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University and College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Seoul 06974, Korea; (Y.S.); (P.H.); (K.K.); (D.L.); (S.L.)
| | - Sehwa Lee
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University and College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Seoul 06974, Korea; (Y.S.); (P.H.); (K.K.); (D.L.); (S.L.)
| | - Eunseong Lee
- Division of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea;
| | - Yuseok Youn
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea;
| | - Kyungtaek Oh
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University and College of Pharmacy, Chung-Ang University, 221 Heukseok-dong, Seoul 06974, Korea; (Y.S.); (P.H.); (K.K.); (D.L.); (S.L.)
| |
Collapse
|
4
|
Xue Y, Bai H, Peng B, Fang B, Baell J, Li L, Huang W, Voelcker NH. Stimulus-cleavable chemistry in the field of controlled drug delivery. Chem Soc Rev 2021; 50:4872-4931. [PMID: 33734247 DOI: 10.1039/d0cs01061h] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Stimulus-cleavable nanoscale drug delivery systems are receiving significant attention owing to their capability of achieving exquisite control over drug release via the exposure to specific stimuli. Central to the construction of such systems is the integration of cleavable linkers showing susceptibility to one stimulus or several stimuli with drugs, prodrugs or fluorogenic probes on the one hand, and nanocarriers on the other hand. This review summarises recent advances in stimulus-cleavable linkers from various research areas and the corresponding mechanisms of linker cleavage and biological applications. The feasibility of extending their applications to the majority of nanoscale drug carriers including nanomaterials, polymers and antibodies are further highlighted and discussed. This review also provides general design guidelines to incorporate stimulus-cleavable linkers into nanocarrier-based drug delivery systems, which will hopefully spark new ideas and applications.
Collapse
Affiliation(s)
- Yufei Xue
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia
| | - Bin Fang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Jonathan Baell
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia
| | - Lin Li
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China.
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (Nanjing Tech), 30 South Puzhu Road, Nanjing, 211816, P. R. China
| | - Nicolas Hans Voelcker
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an 710072, China. and Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria 3168, Australia and Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia. and Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia and Department of Materials Science & Engineering, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
5
|
Spanedda MV, Bourel-Bonnet L. Cyclic Anhydrides as Powerful Tools for Bioconjugation and Smart Delivery. Bioconjug Chem 2021; 32:482-496. [PMID: 33662203 DOI: 10.1021/acs.bioconjchem.1c00023] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cyclic anhydrides are potent tools for bioconjugation; therefore, they are broadly used in the functionalization of biomolecules and carriers. The pH-dependent stability and reactivity, as well as the physical properties, can be tuned by the structure of the cyclic anhydride used; thus, their application in smart delivery systems has become very important. This review intends to cover the last updates in the use of cyclic anhydrides as pH-sensitive linkers, their differences in reactivity, and the latest applications found in bioconjugation chemistry or chemical biology, and when possible, in drug delivery.
Collapse
Affiliation(s)
- Maria Vittoria Spanedda
- Laboratoire de Conception et Application de Molécules Bioactives, 3Bio team, ITI InnoVec, UMR 7199 - CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, BP 60024, 67401 Illkirch Cedex, France
| | - Line Bourel-Bonnet
- Laboratoire de Conception et Application de Molécules Bioactives, 3Bio team, ITI InnoVec, UMR 7199 - CNRS/Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, BP 60024, 67401 Illkirch Cedex, France
| |
Collapse
|
6
|
Das SS, Bharadwaj P, Bilal M, Barani M, Rahdar A, Taboada P, Bungau S, Kyzas GZ. Stimuli-Responsive Polymeric Nanocarriers for Drug Delivery, Imaging, and Theragnosis. Polymers (Basel) 2020; 12:E1397. [PMID: 32580366 PMCID: PMC7362228 DOI: 10.3390/polym12061397] [Citation(s) in RCA: 242] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
In the past few decades, polymeric nanocarriers have been recognized as promising tools and have gained attention from researchers for their potential to efficiently deliver bioactive compounds, including drugs, proteins, genes, nucleic acids, etc., in pharmaceutical and biomedical applications. Remarkably, these polymeric nanocarriers could be further modified as stimuli-responsive systems based on the mechanism of triggered release, i.e., response to a specific stimulus, either endogenous (pH, enzymes, temperature, redox values, hypoxia, glucose levels) or exogenous (light, magnetism, ultrasound, electrical pulses) for the effective biodistribution and controlled release of drugs or genes at specific sites. Various nanoparticles (NPs) have been functionalized and used as templates for imaging systems in the form of metallic NPs, dendrimers, polymeric NPs, quantum dots, and liposomes. The use of polymeric nanocarriers for imaging and to deliver active compounds has attracted considerable interest in various cancer therapy fields. So-called smart nanopolymer systems are built to respond to certain stimuli such as temperature, pH, light intensity and wavelength, and electrical, magnetic and ultrasonic fields. Many imaging techniques have been explored including optical imaging, magnetic resonance imaging (MRI), nuclear imaging, ultrasound, photoacoustic imaging (PAI), single photon emission computed tomography (SPECT), and positron emission tomography (PET). This review reports on the most recent developments in imaging methods by analyzing examples of smart nanopolymers that can be imaged using one or more imaging techniques. Unique features, including nontoxicity, water solubility, biocompatibility, and the presence of multiple functional groups, designate polymeric nanocues as attractive nanomedicine candidates. In this context, we summarize various classes of multifunctional, polymeric, nano-sized formulations such as liposomes, micelles, nanogels, and dendrimers.
Collapse
Affiliation(s)
- Sabya Sachi Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India;
| | - Priyanshu Bharadwaj
- UFR des Sciences de Santé, Université de Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China;
| | - Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman 76175-133, Iran;
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | - Pablo Taboada
- Colloids and Polymers Physics Group, Condensed Matter Physics Area, Particle Physics Department Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain;
- Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania;
| | - George Z. Kyzas
- Department of Chemistry, International Hellenic University, 65404 Kavala, Greece
| |
Collapse
|
7
|
Single enzyme nanoparticle, an effective tool for enzyme replacement therapy. Arch Pharm Res 2020; 43:1-21. [PMID: 31989476 DOI: 10.1007/s12272-020-01216-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/20/2020] [Indexed: 01/10/2023]
Abstract
The term "single enzyme nanoparticle" (SEN) refers to a chemically or biologically engineered single enzyme molecule. SENs are distinguished from conventional protein nanoparticles in that they can maintain their individual structure and enzymatic activity following modification. Furthermore, SENs exhibit enhanced properties as biopharmaceuticals, such as reduced antigenicity, and increased stability and targetability, which are attributed to the introduction of specific moieties, such as poly(ethylene glycol), carbohydrates, and antibodies. Enzyme replacement therapy (ERT) is a crucial therapeutic option for controlling enzyme-deficiency-related disorders. However, the unfavorable properties of enzymes, including immunogenicity, lack of targetability, and instability, can undermine the clinical significance of ERT. As shown in the cases of Adagen®, Revcovi®, Palynziq®, and Strensiq®, SEN can be an effective technology for overcoming these obstacles. Based on these four licensed products, we expect that additional SENs will be introduced for ERT in the near future. In this article, we review the concepts and features of SENs, as well as their preparation methods. Additionally, we summarize different types of enzyme deficiency disorders and the corresponding therapeutic enzymes. Finally, we focus on the current status of SENs in ERT by reviewing FDA-approved products.
Collapse
|
8
|
Faal Maleki M, Jafari A, Mirhadi E, Askarizadeh A, Golichenari B, Hadizadeh F, Jalilzadeh Moghimi SM, Aryan R, Mashreghi M, Jaafari MR. Endogenous stimuli-responsive linkers in nanoliposomal systems for cancer drug targeting. Int J Pharm 2019; 572:118716. [DOI: 10.1016/j.ijpharm.2019.118716] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/11/2022]
|
9
|
Tag and release: strategies for the intracellular cleavage of protein conjugates. Curr Opin Chem Biol 2019; 52:39-46. [DOI: 10.1016/j.cbpa.2019.04.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 01/12/2023]
|
10
|
Dal Corso A, Pignataro L, Belvisi L, Gennari C. Innovative Linker Strategies for Tumor‐Targeted Drug Conjugates. Chemistry 2019; 25:14740-14757. [DOI: 10.1002/chem.201903127] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/15/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Alberto Dal Corso
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| | - Luca Pignataro
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| | - Laura Belvisi
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| | - Cesare Gennari
- Dipartimento di ChimicaUniversità degli Studi di Milano via C. Golgi, 19 20133 Milan Italy
| |
Collapse
|
11
|
Cao Z, Li W, Liu R, Li X, Li H, Liu L, Chen Y, Lv C, Liu Y. pH- and enzyme-triggered drug release as an important process in the design of anti-tumor drug delivery systems. Biomed Pharmacother 2019; 118:109340. [PMID: 31545284 DOI: 10.1016/j.biopha.2019.109340] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/31/2019] [Accepted: 08/05/2019] [Indexed: 12/16/2022] Open
Abstract
It is necessary to design a reasonable drug delivery system(DDS) for targeted release to overcome the potential toxicity and poor selectivity of anti-tumor drug. How a drug is released from a DDS is a critical issue that determines whether the DDS is designed successfully. We all know that the microenvironment of tumors is quite different from normal tissues, such as its acidic environment, different expression levels of some enzymes, etc. These features are widely used in the design of DDSs and play an important role in the drug release process in vivo. Numerous DDSs have been designed and synthesized. This article attention to how drugs are released from DDSs. We summarizes and classify the characteristic enzymes and chemical bonds used in the drug release process by browsing a large number of papers, and describes how they are applied in DDSs with specific examples. By understanding these acid-sensitive chemical bonds and over-expressed enzymes in tumors, different DDSs can be designed for different drug structures to solve specific problems of anti-tumor drugs.
Collapse
Affiliation(s)
- Zhiwen Cao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wen Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Rui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hui Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Linlin Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Youwen Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Cheng Lv
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
12
|
Jafari B, Pourseif MM, Barar J, Rafi MA, Omidi Y. Peptide-mediated drug delivery across the blood-brain barrier for targeting brain tumors. Expert Opin Drug Deliv 2019; 16:583-605. [DOI: 10.1080/17425247.2019.1614911] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Behzad Jafari
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Urmia University of Medical Sciences, Urmia,
Iran
| | - Mohammad M. Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz,
Iran
| | - Mohammad A. Rafi
- Department of Neurology, College of Medicine, Thomas Jefferson University, Philadelphia,
PA, USA
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz,
Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz,
Iran
| |
Collapse
|
13
|
Deirram N, Zhang C, Kermaniyan SS, Johnston APR, Such GK. pH-Responsive Polymer Nanoparticles for Drug Delivery. Macromol Rapid Commun 2019; 40:e1800917. [PMID: 30835923 DOI: 10.1002/marc.201800917] [Citation(s) in RCA: 291] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/31/2019] [Indexed: 01/06/2025]
Abstract
Stimuli-responsive nanoparticles have the potential to improve the delivery of therapeutics to a specific cell or region within the body. There are many stimuli that have shown potential for specific release of cargo, including variation of pH, redox potential, or the presence of enzymes. pH variation has generated significant interest for the synthesis of stimuli-responsive nanoparticles because nanoparticles are internalized into cells via vesicles that are acidified. Additionally, the tumor microenvironment is known to have a lower pH than the surrounding tissue. In this review, different strategies to design pH-responsive nanoparticles are discussed, focusing on the use of charge-shifting polymers, acid labile linkages, and crosslinking.
Collapse
Affiliation(s)
- Nayeleh Deirram
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Changhe Zhang
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Sarah S Kermaniyan
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Georgina K Such
- School of Chemistry, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
14
|
Mollaev M, Gorokhovets N, Nikolskaya E, Faustova M, Zabolotsky A, Zhunina O, Sokol M, Zamulaeva I, Severin E, Yabbarov N. Type of pH sensitive linker reveals different time-dependent intracellular localization, in vitro and in vivo efficiency in alpha-fetoprotein receptor targeted doxorubicin conjugate. Int J Pharm 2019; 559:138-146. [DOI: 10.1016/j.ijpharm.2018.12.073] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 12/15/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022]
|
15
|
Eom T, Yoo W, Kim S, Khan A. Biologically activatable azobenzene polymers targeted at drug delivery and imaging applications. Biomaterials 2018; 185:333-347. [DOI: 10.1016/j.biomaterials.2018.09.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/30/2022]
|
16
|
Oldham R, Lewis M, Orr D, Liao SK, Ogden J, Hubbard W, Birch R. Individually Specified Drug Immunoconjugates in Cancer Treatment. Int J Biol Markers 2018. [DOI: 10.1177/172460088900400202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Forty-three patients with disseminated refractory malignancies each received an individually-specified combination of either Adriamycin (24 patients) or mitomycin-C (19 patients) conjugated murine monoclonal antibodies. Tumors were typed using a panel of antibodies with both immunohistochemistry and flow cytometry. Cocktails of up to six antibodies were selected based on binding greater than 80% of the malignant cells in the biopsy specimen. These monoclonal antibody cocktails were drug conjugated and administered intravenously. Seventeen out of twenty-four patients had reactions to the administration of Adriamycin immunoconjugates, but these were tolerable in all but two patients. Fever, chills, pruritis and skin rash were by far the most common transitory reactions. All were well controlled with premedication. In several patients it was demonstrated that there was limited antigenic drift among various biopsies within the same patient over time. Up to 1 gram of Adriamycin and up to 5 grams of monoclonal antibody were administered. The limiting factor appeared to be a variable dissociation of active Adriamycin from the antibody which unpredictably caused hemopoietic depression. Similar findings were noted in 19 patients with mitomycin-C conjugates. Thrombocytopenia at a 60mg dose of mitomycin-C in this schedule was dose limiting. Preliminary serological evidence suggests that the development of an IgM antibody which is specific against the mouse monoclonal antibody has the specificity and sensitivity to predict clinical reactions. These antibodies were quantitatively less in mitomycin-C patients. Selected patients were re-treated. One patient with chronic lymphocytic leukemia had re-treatment on three occasions and demonstrated regression of peripheral lymph nodes. Two patients with breast carcinoma had definite improvement in ulcerating skin lesions and two patients with tongue carcinoma had shrinkage of their lesions. No responses were seen with mitomycin-C conjugates but binding was noted to tumors and colon with likely drug induced colitis seen after colon binding. This study demonstrates the feasibility and illustrates technical considerations in preparing drug immunoconjugate cocktails for patients with refractory malignancies. Cocktail formulation and antibody delivery was accomplished. The major technical hurdle appears to be the selection of effective conjugation methods that can be used to optimally bind drugs to monoclonal antibodies for targeted cancer therapy.
Collapse
Affiliation(s)
| | - M. Lewis
- Williamson Medical Center Franklin, TN-USA
| | - D.W. Orr
- Williamson Medical Center Franklin, TN-USA
| | - S-K. Liao
- Williamson Medical Center Franklin, TN-USA
| | - J.R. Ogden
- Williamson Medical Center Franklin, TN-USA
| | | | - R. Birch
- Williamson Medical Center Franklin, TN-USA
| |
Collapse
|
17
|
Zhu J, Wang G, Alves CS, Tomás H, Xiong Z, Shen M, Rodrigues J, Shi X. Multifunctional Dendrimer-Entrapped Gold Nanoparticles Conjugated with Doxorubicin for pH-Responsive Drug Delivery and Targeted Computed Tomography Imaging. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:12428-12435. [PMID: 30251859 DOI: 10.1021/acs.langmuir.8b02901] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Novel theranostic nanocarriers exhibit a desirable potential to treat diseases based on their ability to achieve targeted therapy while allowing for real-time imaging of the disease site. Development of such theranostic platforms is still quite challenging. Herein, we present the construction of multifunctional dendrimer-based theranostic nanosystem to achieve cancer cell chemotherapy and computed tomography (CT) imaging with targeting specificity. Doxorubicin (DOX), a model anticancer drug, was first covalently linked onto the partially acetylated poly(amidoamine) dendrimers of generation 5 (G5) prefunctionalized with folic acid (FA) through acid-sensitive cis-aconityl linkage to form G5·NHAc-FA-DOX conjugates, which were then entrapped with gold (Au) nanoparticles (NPs) to create dendrimer-entrapped Au NPs (Au DENPs). We demonstrate that the prepared DOX-Au DENPs possess an Au core size of 2.8 nm, have 9.0 DOX moieties conjugated onto each dendrimer, and are colloid stable under different conditions. The formed DOX-Au DENPs exhibit a pH-responsive release profile of DOX because of the cis-aconityl linkage, having a faster DOX release rate under a slightly acidic pH condition than under a physiological pH. Importantly, because of the coexistence of targeting ligand FA and Au core NPs as a CT imaging agent, the multifunctional DOX-loaded Au DENPs afford specific chemotherapy and CT imaging of FA receptor-overexpressing cancer cells. The constructed DOX-conjugated Au DENPs hold a promising potential to be utilized for simultaneous chemotherapy and CT imaging of various types of cancer cells.
Collapse
Affiliation(s)
- Jingyi Zhu
- Cancer Center , Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai 200072 , People's Republic of China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology , Donghua University , Shanghai 201620 , People's Republic of China
- State Key Laboratory of Material-Oriented Chemical Engineering, School of Pharmaceutical Sciences , Nanjing Tech University , Nanjing 211816 , People's Republic of China
| | - Guoying Wang
- CQM-Centro de Química da Madeira, MMRG , Universidade da Madeira , Campus Universitário da Penteada , 9020-105 Funchal , Portugal
| | - Carla S Alves
- CQM-Centro de Química da Madeira, MMRG , Universidade da Madeira , Campus Universitário da Penteada , 9020-105 Funchal , Portugal
| | - Helena Tomás
- CQM-Centro de Química da Madeira, MMRG , Universidade da Madeira , Campus Universitário da Penteada , 9020-105 Funchal , Portugal
| | - Zhijuan Xiong
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology , Donghua University , Shanghai 201620 , People's Republic of China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology , Donghua University , Shanghai 201620 , People's Republic of China
| | - João Rodrigues
- CQM-Centro de Química da Madeira, MMRG , Universidade da Madeira , Campus Universitário da Penteada , 9020-105 Funchal , Portugal
- School of Materials Science and Engineering/Center for Nano Energy Materials , Northwestern Polytechnical University , Xi'an 710072 , People's Republic of China
| | - Xiangyang Shi
- Cancer Center , Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai 200072 , People's Republic of China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology , Donghua University , Shanghai 201620 , People's Republic of China
- CQM-Centro de Química da Madeira, MMRG , Universidade da Madeira , Campus Universitário da Penteada , 9020-105 Funchal , Portugal
| |
Collapse
|
18
|
Zhu J, Wang G, Alves CS, Tomás H, Xiong Z, Shen M, Rodrigues J, Shi X. Multifunctional Dendrimer-Entrapped Gold Nanoparticles Conjugated with Doxorubicin for pH-Responsive Drug Delivery and Targeted Computed Tomography Imaging. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018. [DOI: https://doi.org/10.1021/acs.langmuir.8b02901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Jingyi Zhu
- Cancer Center, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
- State Key Laboratory of Material-Oriented Chemical Engineering, School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, People’s Republic of China
| | - Guoying Wang
- CQM—Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| | - Carla S. Alves
- CQM—Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| | - Helena Tomás
- CQM—Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| | - Zhijuan Xiong
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
| | - João Rodrigues
- CQM—Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
- School of Materials Science and Engineering/Center for Nano Energy Materials, Northwestern Polytechnical University, Xi’an 710072, People’s Republic of China
| | - Xiangyang Shi
- Cancer Center, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, People’s Republic of China
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, People’s Republic of China
- CQM—Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus Universitário da Penteada, 9020-105 Funchal, Portugal
| |
Collapse
|
19
|
Zhang M, Zhu J, Zheng Y, Guo R, Wang S, Mignani S, Caminade AM, Majoral JP, Shi X. Doxorubicin-Conjugated PAMAM Dendrimers for pH-Responsive Drug Release and Folic Acid-Targeted Cancer Therapy. Pharmaceutics 2018; 10:162. [PMID: 30235881 PMCID: PMC6160908 DOI: 10.3390/pharmaceutics10030162] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 09/11/2018] [Accepted: 09/17/2018] [Indexed: 12/18/2022] Open
Abstract
We present here the development of multifunctional doxorubicin (DOX)-conjugated poly(amidoamine) (PAMAM) dendrimers as a unique platform for pH-responsive drug release and targeted chemotherapy of cancer cells. In this work, we covalently conjugated DOX onto the periphery of partially acetylated and folic acid (FA)-modified generation 5 (G5) PAMAM dendrimers through a pH-sensitive cis-aconityl linkage to form the G5.NHAc-FA-DOX conjugates. The formed dendrimer conjugates were well characterized using different methods. We show that DOX release from the G5.NHAc-FA-DOX conjugates follows an acid-triggered manner with a higher release rate under an acidic pH condition (pH = 5 or 6, close to the acidic pH of tumor microenvironment) than under a physiological pH condition. Both in vitro cytotoxicity evaluation and cell morphological observation demonstrate that the therapeutic activity of dendrimer-DOX conjugates against cancer cells is absolutely related to the DOX drug released. More importantly, the FA conjugation onto the dendrimers allowed a specific targeting to cancer cells overexpressing FA receptors (FAR), and allowed targeted inhibition of cancer cells. The developed G5.NHAc-FA-DOX conjugates may be used as a promising nanodevice for targeted cancer chemotherapy.
Collapse
Affiliation(s)
- Mengen Zhang
- Key Laboratory of Science & Technology of Eco-Textile, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| | - Jingyi Zhu
- College of Materials Science and Engineering, Donghua University, Shanghai 201620, China.
| | - Yun Zheng
- Key Laboratory of Science & Technology of Eco-Textile, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| | - Rui Guo
- Key Laboratory of Science & Technology of Eco-Textile, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| | - Shige Wang
- Key Laboratory of Science & Technology of Eco-Textile, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
| | - Serge Mignani
- Centro de Química da Madeira (CQM), Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal.
| | - Anne-Marie Caminade
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, BP 44099, 31077 Toulouse CEDEX 4, France.
| | - Jean-Pierre Majoral
- Laboratoire de Chimie de Coordination du CNRS, 205 Route de Narbonne, BP 44099, 31077 Toulouse CEDEX 4, France.
| | - Xiangyang Shi
- Key Laboratory of Science & Technology of Eco-Textile, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China.
- Centro de Química da Madeira (CQM), Universidade da Madeira, Campus da Penteada, 9020-105 Funchal, Portugal.
| |
Collapse
|
20
|
A pH-responsive prodrug delivery system self-assembled from acid-labile doxorubicin-conjugated amphiphilic pH-sensitive block copolymers. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 90:27-37. [DOI: 10.1016/j.msec.2018.04.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 01/30/2018] [Accepted: 04/15/2018] [Indexed: 12/20/2022]
|
21
|
Su S, Du FS, Li ZC. Facile Synthesis of a Degradable Poly(ethylene glycol) Platform with Tunable Acid Sensitivity at Physiologically Relevant pH. Macromolecules 2018. [DOI: 10.1021/acs.macromol.8b01053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Shan Su
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Department of Polymer Science & Engineering, College of Chemistry and Molecular Engineering, Center for Soft Matter Science and Engineering, Peking University, Beijing 100871, China
| | - Fu-Sheng Du
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Department of Polymer Science & Engineering, College of Chemistry and Molecular Engineering, Center for Soft Matter Science and Engineering, Peking University, Beijing 100871, China
| | - Zi-Chen Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Department of Polymer Science & Engineering, College of Chemistry and Molecular Engineering, Center for Soft Matter Science and Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
22
|
Gou Y, Zhang Z, Li D, Zhao L, Cai M, Sun Z, Li Y, Zhang Y, Khan H, Sun H, Wang T, Liang H, Yang F. HSA-based multi-target combination therapy: regulating drugs' release from HSA and overcoming single drug resistance in a breast cancer model. Drug Deliv 2018; 25:321-329. [PMID: 29350051 PMCID: PMC6058715 DOI: 10.1080/10717544.2018.1428245] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Multi-drug delivery systems, which may be promising solution to overcome obstacles, have limited the clinical success of multi-drug combination therapies to treat cancer. To this end, we used three different anticancer agents, Cu(BpT)Br, NAMI-A, and doxorubicin (DOX), to build human serum albumin (HSA)-based multi-drug delivery systems in a breast cancer model to investigate the therapeutic efficacy of overcoming single drug (DOX) resistance to cancer cells in vivo, and to regulate the drugs' release from HSA. The HSA complex structure revealed that NAMI-A and Cu(BpT)Br bind to the IB and IIA sub-domain of HSA by N-donor residue replacing a leaving group and coordinating to their metal centers, respectively. The MALDI-TOF mass spectra demonstrated that one DOX molecule is conjugated with lysine of HSA by a pH-sensitive linker. Furthermore, the release behavior of three agents form HSA can be regulated at different pH levels. Importantly, in vivo results revealed that the HSA-NAMI-A-Cu(BpT)Br-DOX complex not only increases the targeting ability compared with a combination of the three agents (the NAMI-A/Cu(BpT)Br/DOX mixture), but it also overcomes DOX resistance to drug-resistant breast cancer cell lines.
Collapse
Affiliation(s)
- Yi Gou
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China.,b School of Pharmacy , Nantong University , Nantong , Jiangsu , China
| | - Zhenlei Zhang
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China
| | - Dongyang Li
- c Department of Biology , Southern University of Science and Technology , Shenzhen , Guangdong , China
| | - Lei Zhao
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China
| | - Meiling Cai
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China
| | - Zhewen Sun
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China
| | - Yongping Li
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China
| | - Yao Zhang
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China
| | - Hamid Khan
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China
| | - Hongbing Sun
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China.,d Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease , China Pharmaceutical University , Nanjing , Jiangsu , China
| | - Tao Wang
- c Department of Biology , Southern University of Science and Technology , Shenzhen , Guangdong , China
| | - Hong Liang
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China
| | - Feng Yang
- a State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China , Guangxi Normal University , Guilin , Guangxi , China
| |
Collapse
|
23
|
Dhavale RP, Waifalkar PP, Sharma A, Dhavale RP, Sahoo SC, Kollu P, Chougale AD, Zahn DRT, Salvan G, Patil PS, Patil PB. Monolayer grafting of aminosilane on magnetic nanoparticles: An efficient approach for targeted drug delivery system. J Colloid Interface Sci 2018; 529:415-425. [PMID: 29940324 DOI: 10.1016/j.jcis.2018.06.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/03/2018] [Accepted: 06/04/2018] [Indexed: 12/12/2022]
Abstract
Magnetic nanoparticles (MNPs) with higher magnetization are highly desirable for targeted drug delivery (TDD) systems, as it helps accumulation of drug at the target site. However, functionalization of MNPs for drug binding reduces the magnetization which affects the efficacy of TDD. Herein we report direct functionalization of MNPs with (3-Aminopropyl)triethoxysilane (APTES) which preserves the magnetization. Grafting density estimated by TGA and BET analysis showed monolayer grafting of APTES on MNP surface. MNPs were comprehensively characterized by XRD, HR-TEM, SQUID-VSM and FTIR. Anti-cancerous drug telmisartan (TEL) was loaded on monolayer APTES grafted MNPs. In-vitro controlled drug release and cytotoxicity study on PC-3 human prostate cancer cell line of TEL conjugated MNPs are also discussed. This functionalization strategy can be extended to other biomedical applications where higher magnetization is desired.
Collapse
Affiliation(s)
- R P Dhavale
- School of Nanoscience and Technology, Shivaji University, Kolhapur, Maharashtra 416004, India
| | - P P Waifalkar
- Department of Physics, Shivaji University, Kolhapur, Maharashtra 416004, India
| | - Apoorva Sharma
- Institute of Physics, Chemnitz University of Technology, 09107 Chemnitz, Germany
| | - R P Dhavale
- Department of Pharmaceutics, Bharati Vidyapeeth College of Pharmacy, Kolhapur, Maharashtra 416013, India
| | - Subasa C Sahoo
- Department of Physics, Central University of Kerala, Kasaragod, Kerala 671314, India
| | - P Kollu
- CASEST, School of Physics, University of Hyderabad, Gachibowli, Hyderabad, Telangana 500046, India; Thin Film Magnetism Group, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - A D Chougale
- Department of Chemistry, The New College, Shivaji University, Kolhapur, Maharashtra 416012, India
| | - D R T Zahn
- Institute of Physics, Chemnitz University of Technology, 09107 Chemnitz, Germany
| | - G Salvan
- Institute of Physics, Chemnitz University of Technology, 09107 Chemnitz, Germany
| | - P S Patil
- School of Nanoscience and Technology, Shivaji University, Kolhapur, Maharashtra 416004, India; Department of Physics, Shivaji University, Kolhapur, Maharashtra 416004, India
| | - P B Patil
- Department of Physics, The New College, Shivaji University, Kolhapur, Maharashtra 416012, India.
| |
Collapse
|
24
|
Hassanzadeh P, Atyabi F, Dinarvand R. Linkers: The key elements for the creation of efficient nanotherapeutics. J Control Release 2018; 270:260-267. [DOI: 10.1016/j.jconrel.2017.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/09/2017] [Accepted: 12/11/2017] [Indexed: 01/16/2023]
|
25
|
Towards antibody-drug conjugates and prodrug strategies with extracellular stimuli-responsive drug delivery in the tumor microenvironment for cancer therapy. Eur J Med Chem 2017; 142:393-415. [DOI: 10.1016/j.ejmech.2017.08.049] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/21/2017] [Accepted: 08/22/2017] [Indexed: 11/20/2022]
|
26
|
Bazban-Shotorbani S, Hasani-Sadrabadi MM, Karkhaneh A, Serpooshan V, Jacob KI, Moshaverinia A, Mahmoudi M. Revisiting structure-property relationship of pH-responsive polymers for drug delivery applications. J Control Release 2017; 253:46-63. [DOI: 10.1016/j.jconrel.2017.02.021] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 01/23/2017] [Accepted: 02/19/2017] [Indexed: 12/17/2022]
|
27
|
Yabbarov NG, Nikolskaya ED, Zhunina OA, Kondrasheva IG, Zamulaeva IA, Severin ES. Polyamidoamine dendrimers with different surface charge as carriers in anticancer drug delivery. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2017. [DOI: 10.1134/s1068162017020182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Su S, Du FS, Li ZC. Synthesis and pH-dependent hydrolysis profiles of mono- and dialkyl substituted maleamic acids. Org Biomol Chem 2017; 15:8384-8392. [DOI: 10.1039/c7ob02188g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Controlled synthesis and in-depth study on pH-dependent hydrolysis profiles of substituted maleamic acid derivatives.
Collapse
Affiliation(s)
- Shan Su
- Beijing National Laboratory for Molecular Sciences (BNLMS)
- Key Laboratory of Polymer Chemistry & Physics of Ministry of Education
- Department of Polymer Science & Engineering
- College of Chemistry and Molecular Engineering
- Peking University
| | - Fu-Sheng Du
- Beijing National Laboratory for Molecular Sciences (BNLMS)
- Key Laboratory of Polymer Chemistry & Physics of Ministry of Education
- Department of Polymer Science & Engineering
- College of Chemistry and Molecular Engineering
- Peking University
| | - Zi-Chen Li
- Beijing National Laboratory for Molecular Sciences (BNLMS)
- Key Laboratory of Polymer Chemistry & Physics of Ministry of Education
- Department of Polymer Science & Engineering
- College of Chemistry and Molecular Engineering
- Peking University
| |
Collapse
|
29
|
Eom T, Yoo W, Lee YD, Park JH, Choe Y, Bang J, Kim S, Khan A. An activatable anticancer polymer–drug conjugate based on the self-immolative azobenzene motif. J Mater Chem B 2017; 5:4574-4578. [DOI: 10.1039/c7tb01250k] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Triggered cellular uptake of a synthetic graft copolymer carrying an anticancer drug is achieved through self-immolation of the side-chain azobenzene groups.
Collapse
Affiliation(s)
- Taejun Eom
- Department of Chemical and Biological Engineering
- Korea University
- Seoul
- Korea
| | - Wonjae Yoo
- Center for Theragnosis
- Korea Institute of Science and Technology
- Seoul
- Korea
- School of Chemical Engineering
| | - Yong-Deok Lee
- Department of Chemical and Biological Engineering
- Korea University
- Seoul
- Korea
- Center for Theragnosis
| | - Jae Hyung Park
- School of Chemical Engineering
- Sungkyunkwan University
- Suwon
- Korea
| | - Youngson Choe
- Department of Chemical Engineering
- Pusan National University
- Pusan
- Korea
| | - Joona Bang
- Department of Chemical and Biological Engineering
- Korea University
- Seoul
- Korea
| | - Sehoon Kim
- Center for Theragnosis
- Korea Institute of Science and Technology
- Seoul
- Korea
| | - Anzar Khan
- Department of Chemical and Biological Engineering
- Korea University
- Seoul
- Korea
| |
Collapse
|
30
|
Zagorodko O, Arroyo-Crespo JJ, Nebot VJ, Vicent MJ. Polypeptide-Based Conjugates as Therapeutics: Opportunities and Challenges. Macromol Biosci 2016; 17. [DOI: 10.1002/mabi.201600316] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/02/2016] [Indexed: 12/16/2022]
Affiliation(s)
- Oleksandr Zagorodko
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
| | - Juan José Arroyo-Crespo
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
| | - Vicent J. Nebot
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
- Polypeptide Therapeutic Solutions SL; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
| | - María J. Vicent
- Polymer Therapeutics Laboratory; Centro de Investigación Príncipe Felipe; Valencia 46012 Spain
| |
Collapse
|
31
|
Yang J, Kopeček J. Design of smart HPMA copolymer-based nanomedicines. J Control Release 2016; 240:9-23. [DOI: 10.1016/j.jconrel.2015.10.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/29/2015] [Accepted: 10/01/2015] [Indexed: 01/13/2023]
|
32
|
Choy CJ, Ley CR, Davis AL, Backer BS, Geruntho JJ, Clowers BH, Berkman CE. Second-Generation Tunable pH-Sensitive Phosphoramidate-Based Linkers for Controlled Release. Bioconjug Chem 2016; 27:2206-13. [DOI: 10.1021/acs.bioconjchem.6b00422] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Cindy J. Choy
- Washington State University, Department
of Chemistry, P.O. Box 644630, Pullman, Washington 99164-4630, United States
| | - Corinne R. Ley
- Washington State University, Department
of Chemistry, P.O. Box 644630, Pullman, Washington 99164-4630, United States
| | - Austen L. Davis
- Washington State University, Department
of Chemistry, P.O. Box 644630, Pullman, Washington 99164-4630, United States
| | - Brian S. Backer
- Washington State University, Department
of Chemistry, P.O. Box 644630, Pullman, Washington 99164-4630, United States
| | - Jonathan J. Geruntho
- Washington State University, Department
of Chemistry, P.O. Box 644630, Pullman, Washington 99164-4630, United States
| | - Brian H. Clowers
- Washington State University, Department
of Chemistry, P.O. Box 644630, Pullman, Washington 99164-4630, United States
| | - Clifford E. Berkman
- Washington State University, Department
of Chemistry, P.O. Box 644630, Pullman, Washington 99164-4630, United States
| |
Collapse
|
33
|
Guo X, Wang L, Wei X, Zhou S. Polymer-based drug delivery systems for cancer treatment. ACTA ACUST UNITED AC 2016. [DOI: 10.1002/pola.28252] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Xing Guo
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education; School of Materials Science and Engineering, Southwest Jiaotong University; Chengdu 610031 China
| | - Lin Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education; School of Materials Science and Engineering, Southwest Jiaotong University; Chengdu 610031 China
| | - Xiao Wei
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education; School of Materials Science and Engineering, Southwest Jiaotong University; Chengdu 610031 China
| | - Shaobing Zhou
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education; School of Materials Science and Engineering, Southwest Jiaotong University; Chengdu 610031 China
| |
Collapse
|
34
|
Pierre TS, Chiellini E. Review : Biodegradability of Synthetic Polymers for Medical and Pharmaceutical Applications: Part 3—Pendent Group Hydrolysis and General Conclusions. J BIOACT COMPAT POL 2016. [DOI: 10.1177/088391158700200305] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Thomas St. Pierre
- Department of Chemistry and Industrial Chemistry Pisa University Pisa, Italy
| | - Emo Chiellini
- Department of Chemistry and Industrial Chemistry Pisa University Pisa, Italy
| |
Collapse
|
35
|
Choi WM, Kopečková P, Minko T, Kopeček J. Synthesis of HPMA Copolymer Containing Adriamycin Bound via an Acid-Labile Spacer and its Activity toward Human Ovarian Carcinoma Cells. J BIOACT COMPAT POL 2016. [DOI: 10.1177/088391159901400601] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
N-(2-Hydroxypropyl)methacrylamide (HPMA) copolymer-adriamycin (ADR) conjugate (P-aconityl-ADR) was synthesized by the attachment of cis-aconityl-ADR to an HPMA copolymer precursor using 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC) as the condensing agent. The ADR release from the HPMA copolymer conjugate was pH sensitive. After 48 h incubation at pH 5, 6, and 7, the amount of ADR released was 63.4, 9.2, and 2.8% respectively. The in vitro cytotoxicity of the conjugate was evaluated toward A2780 sensitive and A2780/AD resistant human ovarian carcinoma cells. An HPMA copolymer, containing ADR bound via a tetrapeptide (GFLG) sequence susceptible to cleavage catalyzed by lysosomal enzymes (P-GFLG-ADR), was used as control. The IC50 doses seemed to indicate that the total hydrolysis of P-aconityl-ADR in prelysosomal and lysosomal compartments proceeded faster than the release of ADR from P-GFLG-ADR catalyzed by lysomal cysteine proteinases. Both HPMA copolymer-ADR conjugates appeared to overcome the ATP-driven P-glycoprotein efflux pump expressed in A2780/AD cells.
Collapse
Affiliation(s)
- Won-Moon Choi
- Departments of Pharmaceutics and Pharmaceutical Chemistry/CCCD and of Bioengineering, University of Utah, Salt Lake City, UT 84112
| | - Pavla Kopečková
- Departments of Pharmaceutics and Pharmaceutical Chemistry/CCCD and of Bioengineering, University of Utah, Salt Lake City, UT 84112
| | - Tamara Minko
- Departments of Pharmaceutics and Pharmaceutical Chemistry/CCCD and of Bioengineering, University of Utah, Salt Lake City, UT 84112
| | - Jindřich Kopeček
- Departments of Pharmaceutics and Pharmaceutical Chemistry/CCCD and of Bioengineering, University of Utah, Salt Lake City, UT 84112
| |
Collapse
|
36
|
Yin J, Chen Y, Zhang ZH, Han X. Stimuli-Responsive Block Copolymer-Based Assemblies for Cargo Delivery and Theranostic Applications. Polymers (Basel) 2016; 8:E268. [PMID: 30974545 PMCID: PMC6432437 DOI: 10.3390/polym8070268] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 02/03/2023] Open
Abstract
Although a number of tactics towards the fabrication and biomedical exploration of stimuli-responsive polymeric assemblies being responsive and adaptive to various factors have appeared, the controlled preparation of assemblies with well-defined physicochemical properties and tailor-made functions are still challenges. These responsive polymeric assemblies, which are triggered by stimuli, always exhibited reversible or irreversible changes in chemical structures and physical properties. However, simple drug/polymer nanocomplexes cannot deliver or release drugs into the diseased sites and cells on-demand due to the inevitable biological barriers. Hence, utilizing therapeutic or imaging agents-loaded stimuli-responsive block copolymer assemblies that are responsive to tumor internal microenvironments (pH, redox, enzyme, and temperature, etc.) or external stimuli (light and electromagnetic field, etc.) have emerged to be an important solution to improve therapeutic efficacy and imaging sensitivity through rationally designing as well as self-assembling approaches. In this review, we summarize a portion of recent progress in tumor and intracellular microenvironment responsive block copolymer assemblies and their applications in anticancer drug delivery and triggered release and enhanced imaging sensitivity. The outlook on future developments is also discussed. We hope that this review can stimulate more revolutionary ideas and novel concepts and meet the significant interest to diverse readers.
Collapse
Affiliation(s)
- Jun Yin
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Hefei University of Technology and Anhui Key Laboratory of Advanced Functional Materials and Devices, Hefei 230009, China.
| | - Yu Chen
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Hefei University of Technology and Anhui Key Laboratory of Advanced Functional Materials and Devices, Hefei 230009, China.
| | - Zhi-Huang Zhang
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Hefei University of Technology and Anhui Key Laboratory of Advanced Functional Materials and Devices, Hefei 230009, China.
| | - Xin Han
- Department of Polymer Science and Engineering, School of Chemistry and Chemical Engineering, Hefei University of Technology and Anhui Key Laboratory of Advanced Functional Materials and Devices, Hefei 230009, China.
| |
Collapse
|
37
|
Zhong Q, da Rocha SRP. Poly(amidoamine) Dendrimer-Doxorubicin Conjugates: In Vitro Characteristics and Pseudosolution Formulation in Pressurized Metered-Dose Inhalers. Mol Pharm 2016; 13:1058-72. [PMID: 26832992 DOI: 10.1021/acs.molpharmaceut.5b00876] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Lung cancers are the leading cause of cancer death for both men and women. A series of PEGylated poly(amidoamine) dendrimer-based doxorubicin (DOX) nanocarriers (G3NH2-mPEG-nDOX) were synthesized and their chemistry tailored for the development of novel pseudosolution formulations in propellant-based metered-dose inhalers (pMDIs) with enhanced aerosol characteristics. A pH-labile bond was used to conjugate DOX to dendrimer for controlled intracellular release. We employed a two-step PEGylation strategy to cover a range of DOX loading and PEGylation density. We investigated the impact of pH, PEGylation density, and DOX payload on the release of DOX from the conjugate. We also determined the cellular internalization of the conjugate, the intracellular release kinetics of DOX from the conjugate, and their ability to kill human alveolar carcinoma cells (A549). The acid-labile conjugates sustained the release of DOX in acidic medium, and also intracellularly, as determined by nuclear colocalization studies with confocal microscopy. Meanwhile, DOX was retained in the conjugate at extracellular physiological conditions, indicating their potential to achieve spatial and temporal controlled release profiles. We also observed that the kinetics of cellular entry of the conjugates with DOX increased significantly compared to free DOX. Due to controlled release, the G3NH2-mPEG-nDOX conjugates showed time-dependent cell kill, but their cell kill ability was comparable to free DOX, which suggests their potential in vivo as compared to free DOX. The conjugates were formulated in pMDIs as pseudosolution formulations, with the help of a minimum amount of cosolvent (ethanol; <0.4%; v/v). The physical stability and aerosol characteristics of the conjugates were controlled by the PEGylation density of the carriers: the higher the PEG density, the better the dispersibility and the better the deep lung deposition of the conjugates (fine particle fraction up to ca. 80%).
Collapse
Affiliation(s)
- Qian Zhong
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University , 5050 Anthony Wayne Drive, Detroit, Michigan 48202, United States
| | - Sandro R P da Rocha
- Department of Chemical Engineering and Materials Science, College of Engineering, Wayne State University , 5050 Anthony Wayne Drive, Detroit, Michigan 48202, United States.,Pharmaceutics and Chemical and Life Science Engineering, Virginia Commonwealth University , 410 N 12th Street, Richmond, Virginia 23298-0533, United States
| |
Collapse
|
38
|
Mechanisms and biomaterials in pH-responsive tumour targeted drug delivery: A review. Biomaterials 2016; 85:152-67. [PMID: 26871891 DOI: 10.1016/j.biomaterials.2016.01.061] [Citation(s) in RCA: 637] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 01/25/2016] [Accepted: 01/27/2016] [Indexed: 12/12/2022]
Abstract
As the mainstay in the treatment of various cancers, chemotherapy plays a vital role, but still faces many challenges, such as poor tumour selectivity and multidrug resistance (MDR). Targeted drug delivery using nanotechnology has provided a new strategy for addressing the limitations of the conventional chemotherapy. In the last decade, the volume of research published in this area has increased tremendously, especially with functional nano drug delivery systems (nanocarriers). Coupling a specific stimuli-triggered drug release mechanism with these delivery systems is one of the most prevalent approaches for improving therapeutic outcomes. Among the various stimuli, pH triggered delivery is regarded as the most general strategy, targeting the acidic extracellular microenvironment and intracellular organelles of solid tumours. In this review, we discuss recent advances in the development of pH-sensitive nanocarriers for tumour-targeted drug delivery. The review focuses on the chemical design of pH-sensitive biomaterials, which are used to fabricate nanocarriers for extracellular and/or intracellular tumour site-specific drug release. The pH-responsive biomaterials bring forth conformational changes in these nanocarriers through various mechanisms such as protonation, charge reversal or cleavage of a chemical bond, facilitating tumour specific cell uptake or drug release. A greater understanding of these mechanisms will help to design more efficient drug delivery systems to address the challenges encountered in conventional chemotherapy.
Collapse
|
39
|
pH-responsive polymer–drug conjugates: Design and progress. J Control Release 2016; 222:116-29. [DOI: 10.1016/j.jconrel.2015.12.024] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 01/31/2023]
|
40
|
Jacques SA, Leriche G, Mosser M, Nothisen M, Muller CD, Remy JS, Wagner A. From solution to in-cell study of the chemical reactivity of acid sensitive functional groups: a rational approach towards improved cleavable linkers for biospecific endosomal release. Org Biomol Chem 2016; 14:4794-803. [DOI: 10.1039/c6ob00846a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
pH-Sensitive linkers designed to undergo selective hydrolysis at acidic pH compared to physiological pH can be used for the selective release of therapeutics at their site of action.
Collapse
Affiliation(s)
- Sylvain A. Jacques
- LFCS Laboratory
- CAMB UMR 7199 CNRS University of Strasbourg
- LabEx Medalis
- icFRC
- Faculty of Pharmacy
| | - Geoffray Leriche
- LFCS Laboratory
- CAMB UMR 7199 CNRS University of Strasbourg
- LabEx Medalis
- icFRC
- Faculty of Pharmacy
| | - Michel Mosser
- LFCS Laboratory
- CAMB UMR 7199 CNRS University of Strasbourg
- LabEx Medalis
- icFRC
- Faculty of Pharmacy
| | - Marc Nothisen
- V-SAT Laboratory
- CAMB UMR 7199 CNRS University of Strasbourg
- LabEx Medalis
- icFRC
- Faculty of Pharmacy
| | - Christian D. Muller
- Laboraroire d'Innovation Thérapeutique
- UMR 7200
- CNRS University of Strasbourg
- Faculty of Pharmacy
- 67400 Illkirch
| | - Jean-Serge Remy
- V-SAT Laboratory
- CAMB UMR 7199 CNRS University of Strasbourg
- LabEx Medalis
- icFRC
- Faculty of Pharmacy
| | - Alain Wagner
- LFCS Laboratory
- CAMB UMR 7199 CNRS University of Strasbourg
- LabEx Medalis
- icFRC
- Faculty of Pharmacy
| |
Collapse
|
41
|
Zaro JL, Shen WC. Cationic and amphipathic cell-penetrating peptides (CPPs): Their structures and in vivo studies in drug delivery. Front Chem Sci Eng 2015. [DOI: 10.1007/s11705-015-1538-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
42
|
Laga R, Janoušková O, Ulbrich K, Pola R, Blažková J, Filippov SK, Etrych T, Pechar M. Thermoresponsive Polymer Micelles as Potential Nanosized Cancerostatics. Biomacromolecules 2015; 16:2493-505. [PMID: 26153904 DOI: 10.1021/acs.biomac.5b00764] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An effective chemotherapy for neoplastic diseases requires the use of drugs that can reach the site of action at a therapeutically efficacious concentration and maintain it at a constant level over a sufficient period of time with minimal side effects. Currently, conjugates of high-molecular-weight hydrophilic polymers or biocompatible nanoparticles with stimuli-releasable anticancer drugs are considered to be some of the most promising systems capable of fulfilling these criteria. In this work, conjugates of thermoresponsive diblock copolymers with the covalently bound cancerostatic drug pirarubicin (PIR) were synthesized as a reversible micelle-forming drug delivery system combining the benefits of the above-mentioned carriers. The diblock copolymer carriers were composed of hydrophilic poly[N-(2-hydroxypropyl)methacrylamide]-based block containing a small amount (∼ 5 mol %) of comonomer units with reactive hydrazide groups and a thermoresponsive poly[2-(2-methoxyethoxy)ethyl methacrylate] block. PIR was attached to the hydrophilic block of the copolymer through the pH-sensitive hydrazone bond designed to be stable in the bloodstream at pH 7.4 but to be degraded in an intratumoral/intracellular environment at pH 5-6. The temperature-induced conformation change of the thermoresponsive block (coil-globule transition), followed by self-assembly of the copolymer into a micellar structure, was controlled by the thermoresponsive block length and PIR content. The cytotoxicity and intracellular transport of the conjugates as well as the release of PIR from the conjugates inside the cells, followed by its accumulation in the cell nuclei, were evaluated in vitro using human colon adenocarcinoma (DLD-1) cell lines. It was demonstrated that the studied conjugates have a great potential to become efficacious in vivo pharmaceuticals.
Collapse
Affiliation(s)
- Richard Laga
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Department of Biomedical Polymers, Heyrovský square 2, 162 06 Prague, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Department of Biomedical Polymers, Heyrovský square 2, 162 06 Prague, Czech Republic
| | - Karel Ulbrich
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Department of Biomedical Polymers, Heyrovský square 2, 162 06 Prague, Czech Republic
| | - Robert Pola
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Department of Biomedical Polymers, Heyrovský square 2, 162 06 Prague, Czech Republic
| | - Jana Blažková
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Department of Biomedical Polymers, Heyrovský square 2, 162 06 Prague, Czech Republic
| | - Sergey K Filippov
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Department of Biomedical Polymers, Heyrovský square 2, 162 06 Prague, Czech Republic
| | - Tomáš Etrych
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Department of Biomedical Polymers, Heyrovský square 2, 162 06 Prague, Czech Republic
| | - Michal Pechar
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic, v.v.i., Department of Biomedical Polymers, Heyrovský square 2, 162 06 Prague, Czech Republic
| |
Collapse
|
43
|
Leriche G, Nothisen M, Baumlin N, Muller CD, Bagnard D, Remy JS, Jacques SA, Wagner A. Spiro Diorthoester (SpiDo), a Human Plasma Stable Acid-Sensitive Cleavable Linker for Lysosomal Release. Bioconjug Chem 2015; 26:1461-5. [DOI: 10.1021/acs.bioconjchem.5b00280] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | - Nadège Baumlin
- INSERM U1109 − MN3T Lab, University of Strasbourg, LabEx Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Christian D. Muller
- Laboratoire d’Innovation
Thérapeutique,
UMR 7200, CNRS University of Strasbourg, Faculty of Pharmacy, 74 route du Rhin, 67400 Illkirch, France
| | - Dominique Bagnard
- INSERM U1109 − MN3T Lab, University of Strasbourg, LabEx Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | | | | | | |
Collapse
|
44
|
Wong PT, Choi SK. Mechanisms of Drug Release in Nanotherapeutic Delivery Systems. Chem Rev 2015; 115:3388-432. [DOI: 10.1021/cr5004634] [Citation(s) in RCA: 349] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Pamela T. Wong
- Michigan
Nanotechnology Institute
for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Seok Ki Choi
- Michigan
Nanotechnology Institute
for Medicine and Biological Sciences, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
45
|
Coyne CP, Jones T, Bear R. Simultaneous Dual Selective Targeted Delivery of Two Covalent Gemcitabine Immunochemotherapeutics and Complementary Anti-Neoplastic Potency of [Se]-Methylselenocysteine. JOURNAL OF CANCER THERAPY 2015; 6:62-89. [PMID: 25821636 PMCID: PMC4376018 DOI: 10.4236/jct.2015.61009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The anti-metabolite chemotherapeutic, gemcitabine is relatively effective for a spectrum of neoplastic conditions that include various forms of leukemia and adenocarcinoma/carcinoma. Rapid systemic deamination of gemcitabine accounts for a brief plasma half-life but its sustained administration is often curtailed by sequelae and chemotherapeutic-resistance. A molecular strategy that diminishes these limitations is the molecular design and synthetic production of covalent gemcitabine immunochemotherapeutics that possess properties of selective "targeted" delivery. The simultaneous dual selective "targeted" delivery of gemcitabine at two separate sites on the external surface membrane of a single cancer cell types represents a therapeutic approach that can increase cytosol chemotherapeutic deposition; prolong chemotherapeutic plasma half-life (reduces administration frequency); minimize innocent exposure of normal tissues and healthy organ systems; and ultimately enhance more rapid and thorough resolution of neoplastic cell populations. MATERIALS AND METHODS A light-reactive gemcitabine intermediate synthesized utilizing succinimidyl 4,4-azipentanoate was covalently bound to anti-EGFR or anti-HER2/neu IgG by exposure to UV light (354-nm) resulting in the synthesis of covalent immunochemotherapeutics, gemcitabine-(C4-amide)-[anti-EGFR] and gemcitabine-(C4-amide)-[anti-HER2/neu]. Cytotoxic anti-neoplastic potency of gemcitabine-(C4-amide)-[anti-EGFR] and gemcitabine-(C4-amide)-[anti-HER2/neu] between gemcitabine-equivalent concentrations of 10-12 M and 10-6 M was determined utilizing chemotherapeutic-resistant mammary adenocarcinoma (SKRr-3). The organoselenium compound, [Se]-methylselenocysteine was evaluated to determine if it complemented the anti-neoplastic potency of the covalent gemcitabine immunochemotherapeutics. RESULTS Gemcitabine-(C4-amide)-[anti-EGFR], gemcitabine-(C4-amide)-[anti-HER2/neu] and the dual simultaneous combination of gemcitabine-(C4-amide)-[anti-EGFR] with gemcitabine-(C4-amide)-[anti-HER2/neu] all had anti-neoplastic cytotoxic potency against mammary adenocarcinoma. Gemcitabine-(C4-amide)-[anti-EGFR] and gemcitabine-(C4-amide)-[anti-HER2/neu] produced progressive increases in anti-neoplastic cytotoxicity that were greatest between gemcitabine-equivalent concentrations of 10-9 M and 10-6 M. Dual simultaneous combinations of gemcitabine-(C4-amide)-[anti-EGFR] with gemcitabine-(C4-amide)-[anti-HER2/neu] produced levels of anti-neoplastic cytotoxicity intermediate between each of the individual covalent gemcitabine immunochemotherapeutics. Total anti-neoplastic cytotoxicity of the dual simultaneous combination of gemcitabine-(C4-amide)-[anti-EGFR] and gemcitabine-(C4-amide)-[anti-HER2/neu] against chemotherapeutic-resistant mammary adenocarcinoma (SKBr-3) was substantially higher when formulated with [Se]-methylsele-nocysteine.
Collapse
Affiliation(s)
- C P Coyne
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, USA
| | - Toni Jones
- Department of Basic Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, USA
| | - Ryan Bear
- Wise Center, Mississippi State University, Mississippi State, USA
| |
Collapse
|
46
|
|
47
|
Nakayama M, Akimoto J, Okano T. Polymeric micelles with stimuli-triggering systems for advanced cancer drug targeting. J Drug Target 2015; 22:584-99. [PMID: 25012066 DOI: 10.3109/1061186x.2014.936872] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Since the 1990s, nanoscale drug carriers have played a pivotal role in cancer chemotherapy, acting through passive drug delivery mechanisms and subsequent pharmaceutical action at tumor tissues with reduction of adverse effects. Polymeric micelles, as supramolecular assemblies of amphiphilic polymers, have been considerably developed as promising drug carrier candidates, and a number of clinical studies of anticancer drug-loaded polymeric micelle carriers for cancer chemotherapy applications are now in progress. However, these systems still face several issues; at present, the simultaneous control of target-selective delivery and release of incorporated drugs remains difficult. To resolve these points, the introduction of stimuli-responsive mechanisms to drug carrier systems is believed to be a promising approach to provide better solutions for future tumor drug targeting strategies. As possible trigger signals, biological acidic pH, light, heating/cooling and ultrasound actively play significant roles in signal-triggering drug release and carrier interaction with target cells. This review article summarizes several molecular designs for stimuli-responsive polymeric micelles in response to variation of pH, light and temperature and discusses their potentials as next-generation tumor drug targeting systems.
Collapse
Affiliation(s)
- Masamichi Nakayama
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University (TWIns) , Kawada-Cho, Shinjuku-ku, Tokyo , Japan
| | | | | |
Collapse
|
48
|
Cheng T, Ma R, Zhang Y, Ding Y, Liu J, Ou H, An Y, Liu J, Shi L. A surface-adaptive nanocarrier to prolong circulation time and enhance cellular uptake. Chem Commun (Camb) 2015; 51:14985-8. [DOI: 10.1039/c5cc05854f] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mixed-shell micelles (MSMs) with adaptive surfaces could rapidly and reversibly change surface properties to prolong circulation time and enhance cellular uptake.
Collapse
Affiliation(s)
- Tangjian Cheng
- State Key Laboratory of Medicinal Chemical Biology
- Key Laboratory of Functional Polymer Materials
- Ministry of Education
- Institute of Polymer Chemistry
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
| | - Rujiang Ma
- State Key Laboratory of Medicinal Chemical Biology
- Key Laboratory of Functional Polymer Materials
- Ministry of Education
- Institute of Polymer Chemistry
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
| | - Yumin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine
- Institute of Radiation Medicine
- Chinese Academy of Medical Science & Peking Union Medical College
- Tianjin
- P. R. China
| | - Yuxun Ding
- State Key Laboratory of Medicinal Chemical Biology
- Key Laboratory of Functional Polymer Materials
- Ministry of Education
- Institute of Polymer Chemistry
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
| | - Jinjian Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine
- Institute of Radiation Medicine
- Chinese Academy of Medical Science & Peking Union Medical College
- Tianjin
- P. R. China
| | - Hanlin Ou
- State Key Laboratory of Medicinal Chemical Biology
- Key Laboratory of Functional Polymer Materials
- Ministry of Education
- Institute of Polymer Chemistry
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
| | - Yingli An
- State Key Laboratory of Medicinal Chemical Biology
- Key Laboratory of Functional Polymer Materials
- Ministry of Education
- Institute of Polymer Chemistry
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine
- Institute of Radiation Medicine
- Chinese Academy of Medical Science & Peking Union Medical College
- Tianjin
- P. R. China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology
- Key Laboratory of Functional Polymer Materials
- Ministry of Education
- Institute of Polymer Chemistry
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin)
| |
Collapse
|
49
|
Aina V, Malavasi G, Magistris C, Cerrato G, Martra G, Viscardi G, Menabue L, Lusvardi G. Conjugation of amino-bioactive glasses with 5-aminofluorescein as probe molecule for the development of pH sensitive stimuli-responsive biomaterials. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2014; 25:2243-2253. [PMID: 24722810 DOI: 10.1007/s10856-014-5206-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/27/2014] [Indexed: 06/03/2023]
Abstract
Bioceramics, such as silica-based glasses, are widely used in bone and teeth restoration. Nowadays, the association between nanotechnology and pharmacology is one of the most promising research fields in cancer therapy. The advanced processing methods and new chemical strategies allow the incorporation of drugs within them or on their functionalized surfaces. Bioceramics can act as local drug delivery systems to treat bone and teeth diseases. The present paper reports data related to the development of a pH-stimuli responsive bioactive glass. The glass conjugation with 5-aminofluorescein (5-AF), through a pH-sensitive organic spacer, allows to produce a pH-responsive bioactive biomaterial: when it is exposed to specific pH changes, it can favour the release of 5-AF directly at the target site. 5-AF has been chosen as a simple, low cost, non toxic model to simulate doxorubicin, an anticancer drug. As doxorubicin, 5-AF contains an amino group in its structure in order to form an amide bond with the carboxylic functionalities of the glass. Raman spectroscopy and thermal analysis confirm the glass conjugation of 5-AF by means of an amide bond; the amount of 5-AF loaded was very high (≈ 65 and 44 wt%). The release tests at two different pH (4.2 and 7.4) show that the amount of released 5-AF is higher at acid pH with respect to physiological one. This preliminary datum evidenced that a pH-sensitive drug delivery system has been developed. The low amount of 5-AF released (<1 wt% of the total 5-AF) is due to the very low solubility of 5-AF in aqueous medium. This disadvantage, may be overcome in a dynamic environment (physiological conditions), where it is possible to obtain a drug release system ensuring an effective therapeutic dose for long times and, at the same time, avoiding the drug toxicity.
Collapse
Affiliation(s)
- Valentina Aina
- Department of Chemistry, Interdepartmental NIS (Nanostructured Interfaces and Surfaces); INSTM (Italian National Consortium for Materials Science and Technology), University of Torino, Via P. Giuria 7, 10125, Turin, Italy,
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Yang J, Kopeček J. Macromolecular therapeutics. J Control Release 2014; 190:288-303. [PMID: 24747162 PMCID: PMC4142088 DOI: 10.1016/j.jconrel.2014.04.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 12/13/2022]
Abstract
This review covers water-soluble polymer-drug conjugates and macromolecules that possess biological activity without attached low molecular weight drugs. The main design principles of traditional and backbone degradable polymer-drug conjugates as well as the development of a new paradigm in nanomedicines - (low molecular weight) drug-free macromolecular therapeutics are discussed. To address the biological features of cancer, macromolecular therapeutics directed to stem/progenitor cells and the tumor microenvironment are deliberated. Finally, the future perspectives of the field are briefly debated.
Collapse
Affiliation(s)
- Jiyuan Yang
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City 84112, USA
| | - Jindřich Kopeček
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City 84112, USA; Department of Bioengineering, University of Utah, Salt Lake City 84112, USA.
| |
Collapse
|