1
|
Shafiei FS, Abroun S. Recent advancements in nanomedicine as a revolutionary approach to treating multiple myeloma. Life Sci 2024; 356:122989. [PMID: 39197575 DOI: 10.1016/j.lfs.2024.122989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024]
Abstract
Multiple myeloma, the second most common hematological malignancy, remains incurable with a 5-year survival rate of approximately 50 % and recurrence rates near 100 %, despite significant attempts to develop effective medicines. Therefore, there is a pressing demand in the medical field for innovative and more efficient treatments for MM. Currently, the standard approach for treating MM involves administering high-dose chemotherapy, which frequently correlates with improved results; however, one major limiting factor is the significant side effects of these medications. Furthermore, the strategies used to deliver medications to tumors limit their efficacy, whether by rapid clearance from circulation or an insufficient concentration in cancer cells. Cancer treatment has shifted from cytotoxic, nonspecific chemotherapy regimens to molecularly targeted, rationally developed drugs with improved efficacy and fewer side effects. Nanomedicines may provide an effective alternative way to avoid these limits by delivering drugs into the complicated bone marrow microenvironment and efficiently reaching myeloma cells. Putting drugs into nanoparticles can make their pharmacokinetic and pharmacodynamic profiles much better. This can increase the drug's effectiveness in tumors, extend its time in circulation in the blood, and lower its off-target toxicity. In this review, we introduce several criteria for the rational design of nanomedicine to achieve the best anti-tumoral therapeutic results. Next, we discuss recent advances in nanomedicine for MM therapy.
Collapse
Affiliation(s)
- Fatemeh Sadat Shafiei
- Department Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Biotechnology, Iranian Research Organization for Science & Technology (IROST), Tehran, Iran
| | - Saeid Abroun
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
2
|
Xu Q, Wang Y, Zheng Y, Zhu Y, Li Z, Liu Y, Ding M. Polymersomes in Drug Delivery─From Experiment to Computational Modeling. Biomacromolecules 2024; 25:2114-2135. [PMID: 38011222 DOI: 10.1021/acs.biomac.3c00903] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Polymersomes, composed of amphiphilic block copolymers, are self-assembled vesicles that have gained attention as potential drug delivery systems due to their good biocompatibility, stability, and versatility. Various experimental techniques have been employed to characterize the self-assembly behaviors and properties of polymersomes. However, they have limitations in revealing molecular details and underlying mechanisms. Computational modeling techniques have emerged as powerful tools to complement experimental studies and enabled researchers to examine drug delivery mechanisms at molecular resolution. This review aims to provide a comprehensive overview of the state of the art in the field of polymersome-based drug delivery systems, with an emphasis on insights gained from both experimental and computational studies. Specifically, we focus on polymersome morphologies, self-assembly kinetics, fusion and fission, behaviors in flow, as well as drug encapsulation and release mechanisms. Furthermore, we also identify existing challenges and limitations in this rapidly evolving field and suggest possible directions for future research.
Collapse
Affiliation(s)
- Qianru Xu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yiwei Wang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yi Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yuling Zhu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Zifen Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yang Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Mingming Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China
| |
Collapse
|
3
|
Zhang Y, Zhong A, Min J, Tu H, Cao Y, Fu J, Li Y, Liu X, Yang Y, Wang J, Liu J, Wu M. Biomimetic Responsive Nanoconverters with Immune Checkpoint Blockade Plus Antiangiogenesis for Advanced Hepatocellular Carcinoma Treatment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:6894-6907. [PMID: 38306190 DOI: 10.1021/acsami.3c18140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2024]
Abstract
The first-line treatment for advanced hepatocellular carcinoma (HCC) combines immune checkpoint inhibitors and antiangiogenesis agents to prolong patient survival. Nonetheless, this approach has several limitations, including stringent inclusion criteria and suboptimal response rates that stem from the severe off-tumor side effects and the unfavorable pharmacodynamics and pharmacokinetics of different drugs delivered systemically. Herein, we propose a single-agent smart nanomedicine-based approach that mimics the therapeutic schedule in a targeted and biocompatible manner to elicit robust antitumor immunity in advanced HCC. Our strategy employed pH-responsive carriers, poly(ethylene glycol)-poly(β-amino esters) amphiphilic block copolymer (PEG-PAEs), for delivering apatinib (an angiogenesis inhibitor), that were surface-coated with plasma membrane derived from engineered cells overexpressing PD-1 proteins (an immune checkpoint inhibitor to block PD-L1). In an advanced HCC mouse model with metastasis, these biomimetic responsive nanoconverters induced significant tumor regression (5/9), liver function recovery, and complete suppression of lung metastasis. Examination of the tumor microenvironment revealed an increased infiltration of immune effector cells (CD8+ and CD4+ T cells) and reduced immunosuppressive cells (myeloid-derived suppressor cells and T regulatory cells) in treated tumors. Importantly, our nanomedicine selectively accumulated in both small and large HCC occupying >50% of the liver volume to exert therapeutic effects with minimal systemic side effects. Overall, these findings highlight the potential of such multifunctional nanoconverters to effectively reshape the tumor microenvironment for advanced HCC treatment.
Collapse
Affiliation(s)
- Yuting Zhang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Aoxue Zhong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Juan Min
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, P. R. China
| | - Haibin Tu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Yanbing Cao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Jinghao Fu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Yonghao Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Yong Yang
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
| | - Jianmin Wang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
| | - Jingfeng Liu
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, P. R. China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| |
Collapse
|
4
|
Bakirdogen G, Selcuk E, Sahkulubey Kahveci EL, Ozbek T, Derman S, Kahveci MU. Fabrication of poly(β-amino ester) and hyaluronic acid based pH responsive nanocomplex as an antibiotic release system. Int J Biol Macromol 2024; 258:129060. [PMID: 38159698 DOI: 10.1016/j.ijbiomac.2023.129060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/18/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
World Health Organization (WHO) warns about antimicrobial resistance (AMR) considered as the most serious threats to global health, food security, and development. There are various efforts for elimination of this serious issue. These efforts include education of individuals, new policies, development of new antimicrobials and new materials for effective delivery. Novel drug delivery systems with ability of local and on-demand delivery are one of the promising approaches for prevention of AMR. In this regard, a pH-responsive antibiotic delivery system based on pH-responsive poly(β-amino ester) (PBAE) and enzyme responsive hyaluronic acid (HA). The polymeric nanocomplexes were obtained via electrostatic complexation of PBAE and HA in the presence of a model antibiotics, colistin and vancomycin. The particle sizes at pH 7.4 were determined in the range of 131-730 nm and 120-400 nm by DLS and STEM, respectively. When pH was switched from 7.4 to 5.5, the hydrodynamic diameter increased 2.5-32 fold. The drug release performances were tested using FITC-labeled antibiotics via fluorescence spectroscopy. The nanocomplexes released the drugs more at pH 5.5 compared to pH 7.4. Antibacterial activity of the system was evaluated on various bacteria. The nanocomplex loaded with the antibiotics exhibited significantly greater efficacy against E. coli and S. aureus.
Collapse
Affiliation(s)
- Gulsah Bakirdogen
- Yildiz Technical University, Davutpasa Campus, Faculty of Chemical and Metallurgical Engineering, Department of Bioengineering, Esenler, 34220, Istanbul, Turkey
| | - Emine Selcuk
- Yildiz Technical University, Davutpasa Campus, Department of Molecular Biology and Genetics, General Biology, Esenler, 34220, Istanbul, Turkey
| | - Elif L Sahkulubey Kahveci
- Yildiz Technical University, Davutpasa Campus, Faculty of Chemical and Metallurgical Engineering, Department of Bioengineering, Esenler, 34220, Istanbul, Turkey
| | - Tulin Ozbek
- Yildiz Technical University, Davutpasa Campus, Department of Molecular Biology and Genetics, General Biology, Esenler, 34220, Istanbul, Turkey
| | - Serap Derman
- Yildiz Technical University, Davutpasa Campus, Faculty of Chemical and Metallurgical Engineering, Department of Bioengineering, Esenler, 34220, Istanbul, Turkey.
| | - Muhammet U Kahveci
- Istanbul Technical University, Faculty of Science and Letters, Department of Chemistry, Maslak, Sariyer, 34467, Istanbul, Turkey.
| |
Collapse
|
5
|
Shymborska Y, Budkowski A, Raczkowska J, Donchak V, Melnyk Y, Vasiichuk V, Stetsyshyn Y. Switching it Up: The Promise of Stimuli-Responsive Polymer Systems in Biomedical Science. CHEM REC 2024; 24:e202300217. [PMID: 37668274 DOI: 10.1002/tcr.202300217] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/20/2023] [Indexed: 09/06/2023]
Abstract
Responsive polymer systems have the ability to change properties or behavior in response to external stimuli. The properties of responsive polymer systems can be fine-tuned by adjusting the stimuli, enabling tailored responses for specific applications. These systems have applications in drug delivery, biosensors, tissue engineering, and more, as their ability to adapt and respond to dynamic environments leads to improved performance. However, challenges such as synthesis complexity, sensitivity limitations, and manufacturing issues need to be addressed for successful implementation. In our review, we provide a comprehensive summary on stimuli-responsive polymer systems, delving into the intricacies of their mechanisms and actions. Future developments should focus on precision medicine, multifunctionality, reversibility, bioinspired designs, and integration with advanced technologies, driving the dynamic growth of sensitive polymer systems in biomedical applications.
Collapse
Affiliation(s)
- Yana Shymborska
- Lviv Polytechnic National University, St. George's Square 2, 79013, Lviv, Ukraine
- Jagiellonian University, Faculty of Physics, Astronomy and Applied Computer Science, Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348, Kraków, Poland
- Jagiellonian University, Doctoral School of Exact and Natural Sciences, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Andrzej Budkowski
- Jagiellonian University, Faculty of Physics, Astronomy and Applied Computer Science, Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Joanna Raczkowska
- Jagiellonian University, Faculty of Physics, Astronomy and Applied Computer Science, Smoluchowski Institute of Physics, Łojasiewicza 11, 30-348, Kraków, Poland
| | - Volodymyr Donchak
- Lviv Polytechnic National University, St. George's Square 2, 79013, Lviv, Ukraine
| | - Yuriy Melnyk
- Lviv Polytechnic National University, St. George's Square 2, 79013, Lviv, Ukraine
| | - Viktor Vasiichuk
- Lviv Polytechnic National University, St. George's Square 2, 79013, Lviv, Ukraine
| | - Yurij Stetsyshyn
- Lviv Polytechnic National University, St. George's Square 2, 79013, Lviv, Ukraine
| |
Collapse
|
6
|
Wang Q, Atluri K, Tiwari AK, Babu RJ. Exploring the Application of Micellar Drug Delivery Systems in Cancer Nanomedicine. Pharmaceuticals (Basel) 2023; 16:ph16030433. [PMID: 36986532 PMCID: PMC10052155 DOI: 10.3390/ph16030433] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Various formulations of polymeric micelles, tiny spherical structures made of polymeric materials, are currently being investigated in preclinical and clinical settings for their potential as nanomedicines. They target specific tissues and prolong circulation in the body, making them promising cancer treatment options. This review focuses on the different types of polymeric materials available to synthesize micelles, as well as the different ways that micelles can be tailored to be responsive to different stimuli. The selection of stimuli-sensitive polymers used in micelle preparation is based on the specific conditions found in the tumor microenvironment. Additionally, clinical trends in using micelles to treat cancer are presented, including what happens to micelles after they are administered. Finally, various cancer drug delivery applications involving micelles are discussed along with their regulatory aspects and future outlooks. As part of this discussion, we will examine current research and development in this field. The challenges and barriers they may have to overcome before they can be widely adopted in clinics will also be discussed.
Collapse
Affiliation(s)
- Qi Wang
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Keerthi Atluri
- Product Development Department, Alcami Corporation, Morrisville, NC 27560, USA
| | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH 43614, USA
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Correspondence:
| |
Collapse
|
7
|
Tumor vasculature VS tumor cell targeting: Understanding the latest trends in using functional nanoparticles for cancer treatment. OPENNANO 2023. [DOI: 10.1016/j.onano.2023.100136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
|
8
|
Pradhan R, Dey A, Taliyan R, Puri A, Kharavtekar S, Dubey SK. Recent Advances in Targeted Nanocarriers for the Management of Triple Negative Breast Cancer. Pharmaceutics 2023; 15:pharmaceutics15010246. [PMID: 36678877 PMCID: PMC9866847 DOI: 10.3390/pharmaceutics15010246] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a life-threatening form of breast cancer which has been found to account for 15% of all the subtypes of breast cancer. Currently available treatments are significantly less effective in TNBC management because of several factors such as poor bioavailability, low specificity, multidrug resistance, poor cellular uptake, and unwanted side effects being the major ones. As a rapidly growing field, nano-therapeutics offers promising alternatives for breast cancer treatment. This platform provides a suitable pathway for crossing biological barriers and allowing sustained systemic circulation time and an improved pharmacokinetic profile of the drug. Apart from this, it also provides an optimized target-specific drug delivery system and improves drug accumulation in tumor cells. This review provides insights into the molecular mechanisms associated with the pathogenesis of TNBC, along with summarizing the conventional therapy and recent advances of different nano-carriers for the management of TNBC.
Collapse
Affiliation(s)
- Rajesh Pradhan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
| | - Anuradha Dey
- Medical Research, R&D Healthcare Division, Emami Ltd., Kolkata 700056, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
- Correspondence: (R.T.); (S.K.D.); Tel.: +91-6378-364-745 (R.T.); +91-8239-703-734 (S.K.D.)
| | - Anu Puri
- RNA Structure and Design Section, RNA Biology Laboratory (RBL), Center for Cancer Research, National Cancer Institute—Frederick, Frederick, MD 21702, USA
| | - Sanskruti Kharavtekar
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
- Medical Research, R&D Healthcare Division, Emami Ltd., Kolkata 700056, India
- Correspondence: (R.T.); (S.K.D.); Tel.: +91-6378-364-745 (R.T.); +91-8239-703-734 (S.K.D.)
| |
Collapse
|
9
|
Sadeqi Nezhad M. Poly (beta-amino ester) as an in vivo nanocarrier for therapeutic nucleic acids. Biotechnol Bioeng 2023; 120:95-113. [PMID: 36266918 DOI: 10.1002/bit.28269] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
Abstract
Therapeutic nucleic acids are an emerging class of therapy for treating various diseases through immunomodulation, protein replacement, gene editing, and genetic engineering. However, they need a vector to effectively and safely reach the target cells. Most gene and cell therapies rely on ex vivo gene delivery, which is laborious, time-consuming, and costly; therefore, devising a systematic vector for effective and safe in vivo delivery of therapeutic nucleic acids is required to target the cells of interest in an efficient manner. Synthetic nanoparticle vector poly beta amino ester (PBAE), a class of degradable polymer, is a promising candidate for in vivo gene delivery. PBAE is considered the most potent in vivo vector due to its excellent transfection performance and biodegradability. PBAE nanoparticles showed tunable charge density, diverse structural characteristics, excellent encapsulation capacity, high stability, stimuli-responsive release, site-specific delivery, potent binding to nucleic acids, flexible binding ability to various conjugates, and effective endosomal escape. These unique properties of PBAE are an essential contribution to in vivo gene delivery. The current review discusses each of the components used for PBAE synthesis and the impact of various environmental and physicochemical factors of the body on PBAE nanocarrier.
Collapse
Affiliation(s)
- Muhammad Sadeqi Nezhad
- Clinical and Translational Science Institute, Translational Biomedical Science Department, University of Rochester Medical Center, Rochester, New York, USA.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA.,Department of Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
10
|
Awan UA, Naeem M, Saeed RF, Mumtaz S, Akhtar N. Smart Nanocarrier-Based Cancer Therapeutics. Cancer Treat Res 2023; 185:207-235. [PMID: 37306911 DOI: 10.1007/978-3-031-27156-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Considerable advances in the field of cancer have been made; however, these have not been translated into similar clinical progress which results in the high prevalence and increased cancer-related mortality rate worldwide. Available treatments have several challenges such as off-target side effects, non-specific long-term potential biodisruption, drug resistance, and overall inadequate response rates and high probability of recurrence. The limitations associated with independent cancer diagnosis and therapy can be minimized by an emerging interdisciplinary research field of nanotheranostics which include successful integration of diagnosis and therapy on a single agent using nanoparticles. This may offer a powerful tool in developing innovative strategies to enable "personalized medicine" for diagnosis and treatment of cancer. Nanoparticles have been proven to be powerful imaging tools or potent agents for cancer diagnosis, treatment, and prevention. The nanotheranostic provides minimally invasive in vivo visualization of drug biodistribution and accumulation at the target site with real-time monitoring of therapeutic outcome. This chapter intends to cover several important aspects and the advances in the field of nanoparticles-mediated cancer therapeutics including nanocarrier development, drug/gene delivery, intrinsically active nanoparticles, tumor microenvironment, and nanotoxicity. The chapter represents an overview of challenges associated with cancer treatment, rational for nanotechnology in cancer therapeutics, novel concepts of multifunctional nanomaterials for cancer therapy along with their classification and their clinical prospective in different cancers. A special focus is on the nanotechnology: regulatory perspective for drug development in cancer therapeutics. Obstacles hindering further development of nanomaterials-mediated cancer therapy are also discussed. In general, the objective of this chapter is to improve our perceptive in the design and development of nanotechnology for cancer therapeutics.
Collapse
Affiliation(s)
- Uzma Azeem Awan
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan.
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Muhammad Naeem
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Rida Fatima Saeed
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Sara Mumtaz
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| | - Nosheen Akhtar
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Pakistan
| |
Collapse
|
11
|
Hari SK, Gauba A, Shrivastava N, Tripathi RM, Jain SK, Pandey AK. Polymeric micelles and cancer therapy: an ingenious multimodal tumor-targeted drug delivery system. Drug Deliv Transl Res 2023; 13:135-163. [PMID: 35727533 DOI: 10.1007/s13346-022-01197-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/13/2022]
Abstract
Since the beginning of pharmaceutical research, drug delivery methods have been an integral part of it. Polymeric micelles (PMs) have emerged as multifunctional nanoparticles in the current technological era of nanocarriers, and they have shown promise in a range of scientific fields. They can alter the release profile of integrated pharmacological substances and concentrate them in the target zone due to their improved permeability and retention, making them more suitable for poorly soluble medicines. With their ability to deliver poorly soluble chemotherapeutic drugs, PMs have garnered considerable interest in cancer. As a result of their remarkable biocompatibility, improved permeability, and minimal toxicity to healthy cells, while also their capacity to solubilize a wide range of drugs in their micellar core, PMs are expected to be a successful treatment option for cancer therapy in the future. Their nano-size enables them to accumulate in the tumor microenvironment (TME) via the enhanced permeability and retention (EPR) effect. In this review, our major aim is to focus primarily on the stellar applications of PMs in the field of cancer therapeutics along with its mechanism of action and its latest advancements in drug and gene delivery (DNA/siRNA) for cancer, using various therapeutic strategies such as crossing blood-brain barrier, gene therapy, photothermal therapy (PTT), and immunotherapy. Furthermore, PMs can be employed as "smart drug carriers," allowing them to target specific cancer sites using a variety of stimuli (endogenous and exogenous), which improve the specificity and efficacy of micelle-based targeted drug delivery. All the many types of stimulants, as well as how the complex of PM and various anticancer drugs react to it, and their pharmacodynamics are also reviewed here. In conclusion, commercializing engineered micelle nanoparticles (MNPs) for application in therapy and imaging can be considered as a potential approach to improve the therapeutic index of anticancer drugs. Furthermore, PM has stimulated intense interest in research and clinical practice, and in light of this, we have also highlighted a few PMs that have previously been approved for therapeutic use, while the majority are still being studied in clinical trials for various cancer therapies.
Collapse
Affiliation(s)
- Sharath Kumar Hari
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India
| | - Ankita Gauba
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India
| | - Neeraj Shrivastava
- Amity Institute of Microbial Technology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India
| | - Ravi Mani Tripathi
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, 201303, India.
| | - Sudhir Kumar Jain
- School of Studies in Microbiology, Vikram University, Ujjain, Madhya Pradesh, 456010, India
| | - Akhilesh Kumar Pandey
- Department of Biological Sciences, Rani Durgavati University, Jabalpur, M.P, 482001, India.,Vikram University, Ujjain, Madhya Pradesh, 456010, India
| |
Collapse
|
12
|
Hamdy NM, Eskander G, Basalious EB. Insights on the Dynamic Innovative Tumor Targeted-Nanoparticles-Based Drug Delivery Systems Activation Techniques. Int J Nanomedicine 2022; 17:6131-6155. [PMID: 36514378 PMCID: PMC9741821 DOI: 10.2147/ijn.s386037] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Anti-cancer conventional chemotherapeutic drugs novel formula progress, nowadays, uses nano technology for targeted drug delivery, specifically tailored to overcome therapeutic agents' delivery challenges. Polymer drug delivery systems (DDS) play a crucial role in minimizing off-target side effects arising when using standard cytotoxic drugs. Using nano-formula for targeted localized action, permits using larger effective cytotoxic doses on a single special spot, that can seriously cause harm if it was administered systemically. Therefore, various nanoparticles (NPs) specifically have attached groups for targeting capabilities, not seen in bulk materials, which then need activation. In this review, we will present a simple innovative, illustrative, in a cartoon-way, enumeration of NP anti-cancer drug targeting delivery system activation-types. Area(s) covered in this review are the mechanisms of various NP activation techniques.
Collapse
Affiliation(s)
- Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Georgette Eskander
- Faculty of Pharmacy, Ain Shams University, Postgraduate Student, Cairo, Egypt
| | - Emad B Basalious
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
13
|
Sahkulubey Kahveci EL, Kahveci MU, Celebi A, Avsar T, Derman S. Glycopolymer and Poly(β-amino ester)-Based Amphiphilic Block Copolymer as a Drug Carrier. Biomacromolecules 2022; 23:4896-4908. [PMID: 36317475 PMCID: PMC9667500 DOI: 10.1021/acs.biomac.2c01076] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/20/2022] [Indexed: 11/16/2022]
Abstract
Glycopolymers are synthetic macromolecules having pendant sugar moieties and widely utilized to target cancer cells. They are usually considered as a hydrophilic segment of amphiphilic block copolymers to fabricate micelles as drug carriers. A novel amphiphilic block copolymer, namely, poly(2-deoxy-2-methacrylamido-d-glucose-co-2-hydroxyethyl methacrylate)-b-poly(β-amino ester) [P(MAG-co-HEMA)-b-PBAE], with active cancer cell targeting potential and pH responsivity was prepared. Tetrazine end functional P(MAG-co-HEMA) and norbornene end functional PBAE blocks were separately synthesized through reversible addition fragmentation chain transfer polymerization and Michael addition-based poly-condensation, respectively, and followed by end-group transformation. Then, inverse electron demand Diels Alder reaction between the tetrazine and the norbornene groups was performed by simply mixing to obtain the amphiphilic block copolymer. After characterization of the block copolymer in terms of chemical structure, pH responsivity, and drug loading/releasing, pH-responsive micelles were obtained with or without doxorubicin (DOX), a model anticancer drug. The micelles exhibited a sharp protonated/deprotonated transition on tertiary amine groups around pH 6.75 and the pH-specific release of DOX below this value. Eventually, the drug delivery potential was evaluated by cytotoxicity assays on both the noncancerous human umbilical vein endothelial cell (HUVEC) cell line and glioblastoma cell line, U87-MG. While the DOX-loaded polymeric micelles were not toxic in noncancerous HUVEC cells, being toxic only to the cancer cells indicates that it is a potential specific cell targeting strategy in the treatment of cancer.
Collapse
Affiliation(s)
- Elif L. Sahkulubey Kahveci
- Faculty
of Chemical and Metallurgical Engineering, Department of Bioengineering, Yildiz Technical University, Davutpasa Campus, Esenler, 34210Istanbul, Turkey
| | - Muhammet U. Kahveci
- Faculty
of Science and Letters, Department of Chemistry, Istanbul Technical University, Maslak, Sariyer, 34467Istanbul, Turkey
| | - Asuman Celebi
- Department
of Medical Biology, School of Medicine, Bahcesehir University, Goztepe, 34734Istanbul, Turkey
| | - Timucin Avsar
- Department
of Medical Biology, School of Medicine, Bahcesehir University, Goztepe, 34734Istanbul, Turkey
| | - Serap Derman
- Faculty
of Chemical and Metallurgical Engineering, Department of Bioengineering, Yildiz Technical University, Davutpasa Campus, Esenler, 34210Istanbul, Turkey
| |
Collapse
|
14
|
Sim TM. Nanoparticle-assisted targeting of the tumour microenvironment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
15
|
Wang X, Zhang Z, Hadjichristidis N. Poly(amino ester)s as an emerging synthetic biodegradable polymer platform: Recent developments and future trends. Prog Polym Sci 2022. [DOI: 10.1016/j.progpolymsci.2022.101634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
16
|
pH-activated nanoplatform for visualized photodynamic and ferroptosis synergistic therapy of tumors. J Control Release 2022; 350:525-537. [PMID: 36055597 DOI: 10.1016/j.jconrel.2022.08.050] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/10/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022]
Abstract
To overcome drug resistance and improve precision theranostics for hepatocellular carcinoma (HCC), a nanoplatform with an "off/on" function for multimodality imaging (near-infrared-II (NIR-II) fluorescence imaging, magnetic resonance imaging (MRI), and photoacoustic imaging) and synergistic therapy (photodynamic therapy and ferroptosis) activated by an acidic pH in the tumor microenvironment is proposed. Although many photosensitizers with photodynamic effects have been reported, very few of them have outstanding photodynamic effect and high stability with response to endogenous stimuli capable of NIR-II imaging. Herein, a new amphiphilic photosensitizer SR780 derived from croconaine dye, was developed with satisfactory photodynamic effects and pH-responsive NIR-II imaging. Interestingly, it was deactivated by coordination with Fe3+ (SR780@Fe) and activated during their release under mild acidic condition. Ferroptosis can generate hydroxyl free radical and lipid peroxide, which aggravate the oxidative stress of tumor cells and mediate their death while depleting glutathione (GSH) to enhance photodynamic effect. In situ pH-activatable theranostic nanoplatform, SR780@Fe-PAE-GP, was thus developed by loading SR780@Fe with pH-responsive polymers, modified by a glypican-3 (GPC-3) receptor-targeting peptide. The synergistic antitumor effects were confirmed both in vitro and in vivo, and the tumor inhibition rate of the SR780@Fe-PAE-GP + L treatment group reached 98%.
Collapse
|
17
|
He W, Zhang Z, Luo Y, Kwok RTK, Zhao Z, Tang BZ. Recent advances of aggregation-induced emission materials for fluorescence image-guided surgery. Biomaterials 2022; 288:121709. [PMID: 35995625 DOI: 10.1016/j.biomaterials.2022.121709] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 07/23/2022] [Accepted: 07/31/2022] [Indexed: 01/10/2023]
Abstract
Real-time intraoperative guidance is essential during various surgical treatment of many diseases. Aggregation-induced emission (AIE) materials have shown great potential for guiding surgeons during complex interventions, with the merits of deep tissue penetration, high quantum yield, high molar absorptivity, low background, good targeting ability and excellent photostability. Herein, we provided insights to design efficient AIE materials regarding three key parameters, i.e., deep-tissue penetration ability, high brightness of AIE luminogens (AIEgens), and precise tumor/other pathology nidus targeting strategies, for realizing better application of fluorescence image-guided surgery. Representative interdisciplinary achievements were outlined for the demonstration of this emerging field. Challenges and future opportunities of AIE materials were briefly discussed. The aim of this review is to provide a comprehensive view of AIE materials for intraoperative guidance for researchers and surgeons, and to inspire more further correlational studies in the new frontiers of image-guided surgery.
Collapse
Affiliation(s)
- Wei He
- School of Science and Engineering, Clinical Translational Research Center of Aggregation-Induced Emission, School of Medicine, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China; Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; HKUST Shenzhen Research Institute, No. 9 Yuexing 1st RD, South Area Hi-tech Park, Nanshan, Shenzhen, 518057, China; Center for Aggregation-Induced Emission and State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, 510640, China.
| | - Zicong Zhang
- School of Science and Engineering, Clinical Translational Research Center of Aggregation-Induced Emission, School of Medicine, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Yumei Luo
- School of Science and Engineering, Clinical Translational Research Center of Aggregation-Induced Emission, School of Medicine, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Ryan Tsz Kin Kwok
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; HKUST Shenzhen Research Institute, No. 9 Yuexing 1st RD, South Area Hi-tech Park, Nanshan, Shenzhen, 518057, China.
| | - Zheng Zhao
- School of Science and Engineering, Clinical Translational Research Center of Aggregation-Induced Emission, School of Medicine, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China; HKUST Shenzhen Research Institute, No. 9 Yuexing 1st RD, South Area Hi-tech Park, Nanshan, Shenzhen, 518057, China.
| | - Ben Zhong Tang
- School of Science and Engineering, Clinical Translational Research Center of Aggregation-Induced Emission, School of Medicine, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China; Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; HKUST Shenzhen Research Institute, No. 9 Yuexing 1st RD, South Area Hi-tech Park, Nanshan, Shenzhen, 518057, China; Center for Aggregation-Induced Emission and State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou, 510640, China.
| |
Collapse
|
18
|
Nanomedicine and regenerative medicine approaches in osteoarthritis therapy. Aging Clin Exp Res 2022; 34:2305-2315. [PMID: 35867240 DOI: 10.1007/s40520-022-02199-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 07/06/2022] [Indexed: 11/01/2022]
Abstract
Osteoarthritis (OA), the most common chronic joint disease, is a degenerative disease that affects 7% of the worldwide population, more than 500 million people all over the world. OA is the main factor of disability in elderly people which decreases the quality of life of patients. It is characterized by joint pain, low bone density, and deterioration of the joint structure. Despite ongoing novel advances in drug discovery and drug delivery, OA therapy is still a big challenge since there is no available effective treatment and the existing therapies mainly focus on pain and symptomatic management rather than improving and/or suppressing its progression. This review aims to summarize the currently available and novel emerging therapies for OA including regenerative medicine and nanotechnology-based materials and formulations at the clinical and experimental levels. Applications of regenerative medicine and novel technologies such as nanotechnology in OA treatments have opened a new window to support OA patients by offering treatments that could halt or delay OA progression satisfactorily or provide an effective cure in near future. Nanomedicine and regenerative medicine suggest novel alternatives in the regeneration of cartilage, repair of bone damage, and control of chronic pain in OA therapy.
Collapse
|
19
|
Gupta M, Sharma V, Sharma K, Kumar A, Sharma A, Kazmi I, Al-Abbasi FA, Alzarea SI, Afzal O, Altamimi ASA, Singh SK, Gupta G, Paudel KR, Hansbro PM, Dua K. A kNGR Peptide-Tethered Lipid-Polymer Hybrid Nanocarrier-Based Synergistic Approach for Effective Tumor Therapy: Development, Characterization, Ex-Vivo, and In-Vivo Assessment. Pharmaceutics 2022; 14:1401. [PMID: 35890297 PMCID: PMC9320317 DOI: 10.3390/pharmaceutics14071401] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
The present study aims to design, develop and characterize kNGR (Asn-Gly-Arg) peptide-conjugated lipid-polymer-based nanoparticles for the target-specific delivery of anticancer bioactive(s), i.e., Paclitaxel (PTX). The kNGR-PEG-DSPE conjugate was synthesized and characterized by using spectral analysis. The dual-targeted PLGA-lecithin-PEG core-shell nanoparticles (PLNs-kNGR-NPs) were synthesized using a modified nanoprecipitation process, and their physiological properties were determined. The results support that, compared to other NPs, PLNs-kNGR-NPs are highly cytotoxic, owing to higher apoptosis and intracellular uptake. The significance of rational nanoparticle design for synergistic treatment is shown by the higher tumor volume inhibition percentage rate (59.7%), compared to other designed formulations in Balb/c mice in the HT-1080 tumor-induced model. The overall results indicate that the PLNs-kNGR-NPs-based hybrid lipid-polymer nanoparticles present the highest therapeutic efficacy against solid tumor overexpressing the CD13 receptors.
Collapse
Affiliation(s)
- Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, Pushp Vihar Sector-3, MB Road, New Delhi 110017, India; (M.G.); (K.S.); (A.K.)
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. H.S. Gour University (A Central University), Sagar 470003, India
| | - Vikas Sharma
- Divine International Group of Institutions-Pharmacy, Gwalior 474001, India;
| | - Kalicharan Sharma
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, Pushp Vihar Sector-3, MB Road, New Delhi 110017, India; (M.G.); (K.S.); (A.K.)
| | - Anoop Kumar
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, Pushp Vihar Sector-3, MB Road, New Delhi 110017, India; (M.G.); (K.S.); (A.K.)
| | - Ajay Sharma
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, Pushp Vihar Sector-3, MB Road, New Delhi 110017, India; (M.G.); (K.S.); (A.K.)
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (I.K.); (F.A.A.-A.)
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (I.K.); (F.A.A.-A.)
| | - Sami I. Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.)
| | - Abdulmalik Saleh Alfawaz Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.)
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India;
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo 2007, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jaipur 302017, India;
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun 248007, India
| | - Keshav Raj Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney 2007, Australia;
| | - Philip M. Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Faculty of Science, School of Life Sciences, Sydney 2007, Australia;
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney 2007, Australia
| |
Collapse
|
20
|
Dutta G, Manickam S, Sugumaran A. Stimuli-Responsive Hybrid Metal Nanocomposite - A Promising Technology for Effective Anticancer Therapy. Int J Pharm 2022; 624:121966. [PMID: 35764265 DOI: 10.1016/j.ijpharm.2022.121966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/19/2022]
Abstract
Cancer is one of the most challenging, life-threatening illnesses to cure, with over 10 million new cases diagnosed each year globally. Improved diagnostic cum treatment with common side-effects are warranting for successful therapy. Nanomaterials are recognized to improve early diagnosis, imaging, and treatment. Recently, multifunctional nanocomposites attracted considerable interest due to their low-cost production, and ideal thermal and chemical stability, and will be beneficial in future diagnostics and customized treatment capacity. Stimuli-Responsive Hybrid Metal Nanocomposites (SRHMNs) based nanocomposite materials pose the on/off delivery of bioactive compounds such as medications, genes, RNA, and DNA to specific tissue or organs and reduce toxicity. They simultaneously serve as sophisticated imaging and diagnostic tools when certain stimuli (e.g., temperature, pH, redox, ultrasound, or enzymes) activate the nanocomposite, resulting in the imaging-guided transport of the payload at defined sites. This review in detail addresses the recent advancements in the design and mechanism of internal breakdown processes of the functional moiety from stimuli-responsive systems in response to a range of stimuli coupled with metal nanoparticles. Also, it provides a thorough understanding of SRHMNs, enabling non-invasive interventional therapy by resolving several difficulties in cancer theranostics.
Collapse
Affiliation(s)
- Gouranga Dutta
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Sivakumar Manickam
- Petroleum and Chemical Engineering, Faculty of Engineering, Universiti Teknologi Brunei, Jalan Tungku Link Gadong, BE1410, Brunei Darussalam
| | - Abimanyu Sugumaran
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, India.
| |
Collapse
|
21
|
Akib AA, Shakil R, Rumon MMH, Roy CK, Chowdhury EH, Chowdhury AN. Natural and Synthetic Micelles for Delivery of Small Molecule Drugs, Imaging Agents and Nucleic Acids. Curr Pharm Des 2022; 28:1389-1405. [PMID: 35524674 DOI: 10.2174/1381612828666220506135301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/02/2022] [Indexed: 11/22/2022]
Abstract
The poor solubility, lack of targetability, quick renal clearance, and degradability of many therapeutic and imaging agents strongly limit their applications inside the human body. Amphiphilic copolymers having self-assembling properties can form core-shell structures called micelles, a promising nanocarrier for hydrophobic drugs, plasmid DNA, oligonucleotides, small interfering RNAs (siRNAs) and imaging agents. Fabrication of micelles loaded with different pharmaceutical agents provides numerous advantages including therapeutic efficacy, diagnostic sensitivity, and controlled release to the desired tissues. Moreover, due to their smaller particle size (10-100 nm) and modified surfaces with different functional groups (such as ligands) help them to accumulate easily in the target location, enhancing cellular uptake and reducing unwanted side effects. Furthermore, the release of the encapsulated agents may also be triggered from stimuli-sensitive micelles at different physiological conditions or by an external stimulus. In this review article, we discuss the recent advancement in formulating and targeting different natural and synthetic micelles including block copolymer micelles, cationic micelles, and dendrimers-, polysaccharide- and protein-based micelles for the delivery of different therapeutic and diagnostic agents. Finally, their applications, outcomes, and future perspectives have been summarized.
Collapse
Affiliation(s)
- Anwarul Azim Akib
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Ragib Shakil
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Md Mahamudul Hasan Rumon
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Chanchal Kumar Roy
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| | - Ezharul Hoque Chowdhury
- Jeffrey Cheah School of Medicine and Health Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Malaysia
| | - Al-Nakib Chowdhury
- Department of Chemistry, Bangladesh University of Engineering and Technology, Dhaka-1000, Bangladesh
| |
Collapse
|
22
|
Bai M, Yang M, Gong J, Xu H, Wei Z. Progress and Principle of Drug Nanocrystals for Tumor Targeted Delivery. AAPS PharmSciTech 2021; 23:41. [PMID: 34964079 DOI: 10.1208/s12249-021-02200-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/09/2021] [Indexed: 12/26/2022] Open
Abstract
Drugs are referred to as drug nanocrystals when they exist as nanoscale crystal structures. This kind of nanocarrier has been widely utilized to increase the solubility and absorption for poorly aqueous soluble drugs after oral administration, or prolong the drug circulation when intravenous administration. The systemic cytotoxicity caused by antitumor drugs usually come from the nonspecific drug distribution. To solve the disadvantage of poor targetability, drug nanocrystals for tumor targeted delivery have been developed in recent years. In this review, the targeting mechanisms of various surface modified drug nanocrystals are introduced with the focus on passive targeting, active targeting and stimuli-responsive targeting in details. Function and application of common surface modified materials are also discussed.
Collapse
|
23
|
Handa M, Singh A, Flora SJS, Shukla R. Stimuli-responsive Polymeric nanosystems for therapeutic applications. Curr Pharm Des 2021; 28:910-921. [PMID: 34879797 DOI: 10.2174/1381612827666211208150210] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Recent past decades have reported emerging of polymeric nanoparticles as a promising technique for controlled and targeted drug delivery. As nanocarriers, they have high drug loading and delivery to the specific site or targeted cells with an advantage of no drug leakage within en route and unloading of a drug in a sustained fashion at the site. These stimuli-responsive systems are functionalized in dendrimers, metallic nanoparticles, polymeric nanoparticles, liposomal nanoparticles, quantum dots. PURPOSE OF REVIEW The authors reviewed the potential of smart stimuli-responsive carriers for therapeutic application and their behavior in external or internal stimuli like pH, temperature, redox, light, and magnet. These stimuli-responsive drug delivery systems behave differently in In vitro and In vivo drug release patterns. Stimuli-responsive nanosystems include both hydrophilic and hydrophobic systems. This review highlights the recent development of the physical properties and their application in specific drug delivery. CONCLUSION The stimuli (smart, intelligent, programmed) drug delivery systems provide site-specific drug delivery with potential therapy for cancer, neurodegenerative, lifestyle disorders. As development and innovation, the stimuli-responsive based nanocarriers are moving at a fast pace and huge demand for biocompatible and biodegradable responsive polymers for effective and safe delivery.
Collapse
Affiliation(s)
- Mayank Handa
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh 226002. India
| | - Ajit Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh 226002. India
| | - S J S Flora
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh 226002. India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, Uttar Pradesh 226002. India
| |
Collapse
|
24
|
Li J, Wang J, Zhang J, Han T, Hu X, Lee MMS, Wang D, Tang BZ. A Facile Strategy of Boosting Photothermal Conversion Efficiency through State Transformation for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2105999. [PMID: 34651361 DOI: 10.1002/adma.202105999] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/11/2021] [Indexed: 06/13/2023]
Abstract
Improving photothermal conversion efficiency (PCE) is critical to facilitate therapeutic performance during photothermal therapy (PTT). However, current strategies of prompting PCE always involve complex synthesis or modification of photothermal agents, thereby significantly inhibiting the practical applications and fundamental understanding of photothermal conversion. A facile strategy is herein present for boosting PCE by transforming photothermal agents from aggregated state to dispersed state. Compared to aggregated state, the developed photothermal agents with semiconducting nature can rotate freely in dispersed state, which allows for an efficient nonradiative dissipation through twisted intramolecular charge transfer (TICT) effect, consequentially offering excellent photothermal performance. Noteworthy, the state transformation can be achieved by virtue of releasing photothermal molecules from nanoparticles on the basis of a pH-responsive polymer nanocarrier, and the PCE is elevated from 43% to 60% upon changing the pH values from 7.4 to 5.0. Moreover, the nanoparticle disassembly and state transformation behaviors can also smoothly proceed in lysosome of cancer cells, demonstrating a distinct photothermal therapeutic performance for cancer ablation. It is hoped that this strategy of transforming state to boost PCE would be a new platform for practical applications of PTT technique.
Collapse
Affiliation(s)
- Jie Li
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jianxing Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jianyu Zhang
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Ting Han
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Xiyao Hu
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Michelle M S Lee
- Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, 999077, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Ben Zhong Tang
- Shenzhen Institute of Aggregate Science and Technology, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| |
Collapse
|
25
|
Gas-filled protein nanostructures as cavitation nuclei for molecule-specific sonodynamic therapy. Acta Biomater 2021; 136:533-545. [PMID: 34530143 DOI: 10.1016/j.actbio.2021.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022]
Abstract
Sonodynamic therapy (SDT) is a promising alternative for cancer therapy, understood to exert cytotoxicity through cavitation and subsequent production of large amounts of reactive oxygen species (ROS). Gas-filled protein nanostructures (gas vesicles or GVs) produced by cyanobacteria have a hollow structure similar to microbubbles and have demonstrated comparable enhancement of ultrasound imaging contrast. We thus hypothesized that GVs may act as stable nuclei for inertial cavitation to enhance SDT with improved enhanced permeability and retention (EPR) effects due to their nanometer scale. The function of GVs to mediate cavitation, ROS production, and cell-targeted toxicity under SDT was determined. In solution, we found that GVs successfully increased cavitation and enhanced ROS production in a dose- and time-dependent manner. Then, GV surfaces were modified (FGVs) to specifically target CD44+ cells and accumulate preferentially at the tumor site. In vitro sonodynamic therapy (SDT) showed ROS production and tumor cell toxicity substantially elevated in the presence of FGVs, and the addition of FGVs was found to enhance cavitation and subsequently inhibit tumor growth and exert greater damage to tumors under SDT in vivo. Our results thus demonstrate that FGVs can function as stable, nanosized, nuclei for spatially accurate and cell-targeted SDT. STATEMENT OF SIGNIFICANCE: The initiation of inertial cavitation is critical for ROS generation and subsequent cellular toxicity in SDT. Thus, precise control of the occurrence of cavitation is a key factor in increasing SDT's therapeutic efficacy. We explored nanometer-sized gas vesicles (GVs) as a new class of cavitation nuclei for molecule-specific sonodynamic therapy. Our results showed that GV-mediated SDT treatment enabled targeted disruption of specific cells expressing a known surface marker within the area of insonation, providing a spatially specific and targeted SDT treatment.
Collapse
|
26
|
Delivery of extracellular matrix-enriched stem cells encapsulated with enzyme-free pH-sensitive polymer for enhancing therapeutic angiogenesis. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2021.08.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
27
|
Grace VMB, Wilson DD, Guruvayoorappan C, Danisha JP, Bonati L. Liposome nano-formulation with cationic polar lipid DOTAP and cholesterol as a suitable pH-responsive carrier for molecular therapeutic drug (all-trans retinoic acid) delivery to lung cancer cells. IET Nanobiotechnol 2021; 15:380-390. [PMID: 34694713 PMCID: PMC8675848 DOI: 10.1049/nbt2.12028] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/27/2020] [Accepted: 01/17/2021] [Indexed: 12/13/2022] Open
Abstract
The molecular targeted drug ATRA demands a suitable carrier that delivers to the cancer site due to its poor bioavailability and drug resistance. ATRA, being a lipid with carboxylic acid, has been nano‐formulated as a cationic lipo‐ATRA with DOTAP:cholesterol:ATRA (5:4:1) and its pH‐responsive release, intracellular drug accumulation, and anticancer effect on human lung cancer (A549) cell line analysed. The analysis of the physicochemical characteristics of the developed lipo‐ATRA (0.8 µmol) revealed that the size of 231 ± 2.35 d.nm had a zeta potential of 6.4 ± 1.19 and an encapsulation efficiency of 93.7 ± 3.6%. The ATRA release from lipo‐ATRA in vitro was significantly (p ≤ 0.05) higher at acidic pH 6 compared to pH 7.5. The intracellular uptake of ATRA into lipo‐ATRA‐treated A549 cells was seven‐fold higher (0.007 ± 0.001 mg/ml) while only three‐fold uptake was observed in free ATRA treatment (0.003 ± 0.002 mg/ml). The lipo‐ATRA treatment caused a highly significant (p ≤ 0.001) decrease in percent cell viability at 48 h when compared with the free ATRA treatment. Overall, the results proved that the developed lipo‐ATRA has suitable physicochemical properties with enhanced ATRA release at acidic pH, while maintaining stability at physiologic pH and temperature. This resulted in an increased ATRA uptake by lung cancer cells with enhanced treatment efficiency. Hence, it is concluded that DOTAP lipo‐ATRA is a suitable carrier for ATRA delivery to solid cancer cells.
Collapse
Affiliation(s)
| | - Devarajan David Wilson
- School of Science, Arts, Media and Management, Karunya Institute of Technology and Sciences, Coimbatore, Tamil Nadu, India
| | - Chandrasekharan Guruvayoorappan
- Laboratory of Immunopharmacology and Experimental Therapeutics, Division of Cancer Research, Regional Cancer Centre, Medical College Campus, Thiruvananthapuram, Kerala, India
| | - Jesubatham Perinba Danisha
- Department of Biotechnology, Karunya Institute of Technology and Sciences, Coimbatore, Tamil Nadu, India
| | - Lucia Bonati
- IAESTE Intern at Department of Biotechnology, Karunya Institute of Technology and Sciences, Coimbatore, Tamil Nadu, India
| |
Collapse
|
28
|
Rahmati MA, Rashidzadeh H, Hosseini MJ, Sadighian S, Kermanian M. Self-assembled magnetic polymeric micelles for delivery of quercetin: Toxicity evaluation on isolated rat liver mitochondria. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 33:279-298. [PMID: 34547988 DOI: 10.1080/09205063.2021.1982644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Multifunctional nanocarriers as a promising platform could provide numerous opportunities in the field of drug delivery. Drug carriers loaded with both magnetic nanoparticles (MNPs) and therapeutic agents would allow the combination of chemotherapy with the possibility of monitoring or controlling the distribution of the nano vehicles in the body which may improve the effectiveness of the therapy. Furthermore, by applying these strategies, triggering drug release and/or synergistic hyperthermia treatment are also reachable. This study aimed to explore the potential of the quercetin (QUR) loaded magnetic nano-micelles for improving drug bioavailability while reducing the drug adverse effects. The bio-safety of developed QUR loaded magnetic nano-micelles (QMNMs) were conducted via mitochondrial toxicity using isolated rat liver mitochondria including glutathione (GSH), malondialdehyde (MDA), and the ferric reducing ability of plasma (FRAP). QMNMs with a mean particle size of 85 nm (PDI value of 0.269) and great physical stability were produced. Also, TEM images indicated that the prepared QMNMs were semi-spherical in shape. These findings also showed that the constructed QMNMs, as a pH-sensitive drug delivery system, exhibited a stable and high rate of QUR release under mildly acidic conditions pH (5.3) compared to neutral pH (7.4). The most striking result to emerge from the data is that an investigation of various mitochondrial functional parameters revealed that both QMNMs and QUR have no specific mitochondrial toxicity. Altogether, these results offer overwhelming evidence for the bio-safety of QMNMs and might be used as an effective drug delivery system for targeting and stimuli-responsive QUR delivery.
Collapse
Affiliation(s)
- Mohammad-Amin Rahmati
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamid Rashidzadeh
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mir-Jamal Hosseini
- Zanjan applied pharmacology research center, Zanjan university of medical sciences, Zanjan, Iran
| | - Somayeh Sadighian
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehraneh Kermanian
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
29
|
Yang Z, Zhao H, Wang D, Yin L, Cai K, Lin Z, Chen T, Yang C. DPD simulations on mixed polymeric DOX-loaded micelles assembled from PCL-SS-PPEGMA/PDEA-PPEGMA and their dual pH/reduction-responsive release. Phys Chem Chem Phys 2021; 23:19011-19021. [PMID: 34612439 DOI: 10.1039/d1cp02750f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The design of mixed polymeric micelles by a combination of two or more dissimilar polymers is a potential strategy to achieve multiple stimuli-response for anti-cancer drug delivery. However, their drug loading co-micellization behavior and multiple stimuli-responsive drug release mechanism have been poorly understood at the mesoscopic level, especially in the system that involves reduction-response due to the difficulty of simulation on the cleavage of chemical bonds. In this work, the co-micellization behavior, drug distribution regularities and dual pH/reduction-responsive drug release process of mixed micelles formed by disulfide-linked polycaprolactone-b-polyethylene glycol methyl ether methacrylate (PCL-SS-PPEGMA) and poly(ethylene glycol) methyl ether-b-poly(N,N-diethylamino ethyl methacrylate) (PDEA-PPEGMA) were studied by dissipative particle dynamics (DPD) mesoscopic simulations. A dedicated bond-breaking script was employed to accomplish the disulfide bond-breaking simulations. The results showed that PCL55-SS-PPEGMA10 and PDEA34-PPEGMA11 could be well mixed to form superior DOX-loaded micelles with good drug-loading capacity and drug-controlled release performance. To prepare the DOX-loaded micelles with optimized properties, the simulation results suggested the feed ratio of DOX:PCL55-SS-PPEGMA10:PDEA34-PPEGMA11 set to 3:4:4. Compared with the two single stimuli-response, the dual pH/reduction-response process perfectly combined both pH-response and reduction-response together, providing a higher release rate of DOX. Therefore, this study provides theoretical guidance aimed at the property optimization and micellar structure design of the dual pH/reduction-responsive mixed micelles.
Collapse
Affiliation(s)
- Zexiong Yang
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Lee J, Kim B, Park B, Won Y, Kim SY, Lee S. Real-time cancer diagnosis of breast cancer using fluorescence lifetime endoscopy based on the pH. Sci Rep 2021; 11:16864. [PMID: 34413447 PMCID: PMC8376886 DOI: 10.1038/s41598-021-96531-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 08/04/2021] [Indexed: 12/11/2022] Open
Abstract
A biopsy is often performed for the diagnosis of cancer during a surgical operation. In addition, pathological biopsy is required to discriminate the margin between cancer tissues and normal tissues in surgical specimens. In this study, we presented a novel method for discriminating between tumor and normal tissues using fluorescence lifetime endoscopy (FLE). We demonstrated the relationship between the fluorescence lifetime and pH in fluorescein using the proposed fluorescence lifetime measurement system. We also showed that cancer could be diagnosed based on this relationship by assessing differences in pH based fluorescence lifetime between cancer and normal tissues using two different types of tumor such as breast tumors (MDA-MB-361) and skin tumors (A375), where cancer tissues have ranged in pH from 4.5 to 7.0 and normal tissues have ranged in pH from 7.0 to 7.4. To support this approach, we performed hematoxylin and eosin (H&E) staining test of normal and cancer tissues within a certain area. From these results, we showed the ability to diagnose a cancer using FLE technique, which were consistent with the diagnosis of a cancer with H&E staining test. In summary, the proposed pH-based FLE technique could provide a real time, in vivo, and in-situ clinical diagnostic method for the cancer surgical and could be presented as an alternative to biopsy procedures.
Collapse
Affiliation(s)
- Jooran Lee
- Medical Device Development Center, Osong Medical Innovation Foundation, Cheongju, Chungbuk, 28160, South Korea
| | - Byungyeon Kim
- Medical Device Development Center, Osong Medical Innovation Foundation, Cheongju, Chungbuk, 28160, South Korea
| | - Byungjun Park
- Medical Device Development Center, Osong Medical Innovation Foundation, Cheongju, Chungbuk, 28160, South Korea
| | - Youngjae Won
- Medical Device Development Center, Osong Medical Innovation Foundation, Cheongju, Chungbuk, 28160, South Korea
- Intek-Medi, 123, Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, South Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro, 43-gil, Songpa-gu, Seoul, 138-736, South Korea
| | - Seungrag Lee
- Medical Device Development Center, Osong Medical Innovation Foundation, Cheongju, Chungbuk, 28160, South Korea.
| |
Collapse
|
31
|
Pérez-Herrero E, Fernández-Medarde A. The reversed intra- and extracellular pH in tumors as a unified strategy to chemotherapeutic delivery using targeted nanocarriers. Acta Pharm Sin B 2021; 11:2243-2264. [PMID: 34522586 PMCID: PMC8424227 DOI: 10.1016/j.apsb.2021.01.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/11/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023] Open
Abstract
Solid tumors are complex entities, comprising a wide variety of malignancies with very different molecular alterations. Despite this, they share a set of characteristics known as "hallmarks of cancer" that can be used as common therapeutic targets. Thus, every tumor needs to change its metabolism in order to obtain the energy levels required for its high proliferative rates, and these adaptations lead to alterations in extra- and intracellular pH. These changes in pH are common to all solid tumors, and can be used either as therapeutic targets, blocking the cell proton transporters and reversing the pH changes, or as means to specifically deliver anticancer drugs. In this review we will describe how proton transport inhibitors in association with nanocarriers have been designed to block the pH changes that are needed for cancer cells to survive after their metabolic adaptations. We will also describe studies aiming to decrease intracellular pH in cancer using nanoparticles as molecular cages for protons which will be released upon UV or IR light exposure. Finally, we will comment on several studies that have used the extracellular pH in cancer for an enhanced cell internalization and tumor penetration of nanocarriers and a controlled drug delivery, describing how nanocarriers are being used to increase drug stability and specificity.
Collapse
Affiliation(s)
- Edgar Pérez-Herrero
- Departamento de Ingeniería Química y Tecnología Farmacéutica, Universidad de La Laguna, La Laguna 38206, Tenerife, Spain
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna 38206, Tenerife, Spain
- Instituto Universitario de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna 38200, Tenerife, Spain
| | - Alberto Fernández-Medarde
- Instituto de Biología Molecular y Celular Del Cáncer, Centro de Investigación Del Cáncer (USAL-CSIC), Salamanca 37007, Spain
| |
Collapse
|
32
|
Singh R, Bhateria R. Core-shell nanostructures: a simplest two-component system with enhanced properties and multiple applications. ENVIRONMENTAL GEOCHEMISTRY AND HEALTH 2021; 43:2459-2482. [PMID: 33161517 DOI: 10.1007/s10653-020-00766-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 10/30/2020] [Indexed: 06/11/2023]
Abstract
With the pace of time, synthesis of nanomaterials has paved paths to blend two or more materials having different properties into hybrid nanoparticles. Therefore, it has become possible to combine two different functionalities in a single nanoparticle and their properties can be enhanced or modified by coupling of two different components. Core-shell technology has now represented a new trend in analytical sciences. Core-shell nanostructures are in demand due to their specific design and geometry. They have internal core of one component (metal or biomolecules) surrounded by a shell of another component. Core-shell nanoparticles have great importance due to their high thermal stability, high solubility and lower toxicity. In this review, recent progress in development of new and sophisticated core-shell nanostructures has been explored. The first section covers introduction throwing light on basics of core-shell nanoparticles. Following section classifies core-shell nanostructures into single core/shell, multicore/single shell, single core/multishell and multicore/multishell nanostructures. Next main section gives a brief description on types of core-shell nanomaterials followed by processes for the synthesis of core-shell nanostructures. Ultimately, the final section focuses on the application areas such as drug delivery, bioimaging, solar cell applications etc.
Collapse
Affiliation(s)
- Rimmy Singh
- Department of Environmental Sciences, MDU, Rohtak, India
| | | |
Collapse
|
33
|
Hyun J, Eom J, Song J, Seo I, Um SH, Park KM, Bhang SH. Poly(amino ester)-Based Polymers for Gene and Drug Delivery Systems and Further Application toward Cell Culture System. Macromol Biosci 2021; 21:e2100106. [PMID: 34117832 DOI: 10.1002/mabi.202100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/20/2021] [Indexed: 11/10/2022]
Abstract
Various synthetic polymers based on poly(amino ester) (PAE) are suggested as candidates for gene and drug delivery owing to their pH-responsiveness, which contributes to efficient delivery performance. PAE-based pH-responsive polymers are more biodegradable and hydrophilic than other types of pH-responsive polymers. The functionality of PAE-based polymers can be reinforced by using different chemical modifications to improve the efficiency of gene and drug delivery. Additionally, PAE-based polymers are used in many ways in the biomedical field, such as in transdermal delivery and stem cell culture systems. Here, the recent novel PAE-based polymers designed for gene and drug delivery systems along with their further applications toward adult stem cell culture systems are reviewed. The synthetic tactics are contemplated and pros and cons of each type of polymer are analyzed, and detailed examples of the different types are analyzed.
Collapse
Affiliation(s)
- Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jiin Eom
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jihun Song
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Inwoo Seo
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyung Min Park
- Division of Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
34
|
ATP/Hyals dually responsive core-shell hyaluronan/chitosan-based drug nanocarrier for potential application in breast cancer therapy. Int J Biol Macromol 2021; 183:839-851. [PMID: 33965490 DOI: 10.1016/j.ijbiomac.2021.05.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/25/2021] [Accepted: 05/02/2021] [Indexed: 12/18/2022]
Abstract
The stability of self-assembled drug nanocarriers during blood circulation and the controlled intracellular drug delivery are two challenges in cancer therapy. In this paper, we constructed an adenosine triphosphate (ATP)/hyaluronidase(Hyals) dually responsive core-shell hyaluronan/chitosan-based drug nanocarrier for breast cancer therapy, using SNX-loaded 3-fluoro-4-carboxyphenylboronic acid-conjugated quaternary ammonium chitosan nanoparticles (SNX@HTCC-FPBA NPs) as the core and crosslinked polyethylene glycol-/methacrylate-modified hyaluronic acid (mHA-PEG) as the shell. The formed SNX@HTCC-FPBA/mHA-PEG NPs were stable against salt ion strength, pH values and human plasma mimicking the bloodstream, but ATP/Hyals dually sensitive with a drug delivery of 85% within 48 h in the mimicking intracellular environment of breast cancer cells. These nanoparticles showed a low hemolysis of less than 3%, a high resistance to bovine serum albumin adsorption of 0.06 mg/mg, and an efficient internalization by two breast cancer cell lines (MCF-7 and MDA-MB-453). The cell culture indicated that they were friendly to human skin fibroblasts, but presented a close IC50 value to SNX for MCF-7 (0.14 μg mL-1) and MDA-MB-453 (0.05 μg mL-1) at 48 h, respectively. Thus, SNX@HTCC-FPBA/mHA-PEG NPs were potential drug nanocarriers for breast tumor therapy.
Collapse
|
35
|
Guan Y, Fan P, Dong W, Shang D. A covalent triazine-based framework containing hydrogen-bonding for highly drug loading and pH-responsive release. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2021. [DOI: 10.1080/10601325.2021.1922085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Yue Guan
- School of Life Science, Liaoning Normal University, Dalian, China
| | - Peng Fan
- School of Life Science, Liaoning Normal University, Dalian, China
| | - Weibing Dong
- School of Life Science, Liaoning Normal University, Dalian, China
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| | - Dejing Shang
- School of Life Science, Liaoning Normal University, Dalian, China
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| |
Collapse
|
36
|
Ghanbari-Movahed M, Kaceli T, Mondal A, Farzaei MH, Bishayee A. Recent Advances in Improved Anticancer Efficacies of Camptothecin Nano-Formulations: A Systematic Review. Biomedicines 2021; 9:480. [PMID: 33925750 PMCID: PMC8146681 DOI: 10.3390/biomedicines9050480] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 04/21/2021] [Accepted: 04/24/2021] [Indexed: 12/12/2022] Open
Abstract
Camptothecin (CPT), a natural plant alkaloid, has indicated potent antitumor activities via targeting intracellular topoisomerase I. The promise that CPT holds in therapies is restricted through factors that include lactone ring instability and water insolubility, which limits the drug oral solubility and bioavailability in blood plasma. Novel strategies involving CPT pharmacological and low doses combined with nanoparticles have indicated potent anticancer activity in vitro and in vivo. This systematic review aims to provide a comprehensive and critical evaluation of the anticancer ability of nano-CPT in various cancers as a novel and more efficient natural compound for drug development. Studies were identified through systematic searches of PubMed, Scopus, and ScienceDirect. Eligibility checks were performed based on predefined selection criteria. Eighty-two papers were included in this systematic review. There was strong evidence for the association between antitumor activity and CPT treatment. Furthermore, studies indicated that CPT nano-formulations have higher antitumor activity in comparison to free CPT, which results in enhanced efficacy for cancer treatment. The results of our study indicate that CPT nano-formulations are a potent candidate for cancer treatment and may provide further support for the clinical application of natural antitumor agents with passive targeting of tumors in the future.
Collapse
Affiliation(s)
- Maryam Ghanbari-Movahed
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
- Department of Biology, Faculty of Science, University of Guilan, Rasht 4193833697, Iran
| | - Tea Kaceli
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| | - Arijit Mondal
- Department of Pharmaceutical Chemistry, Bengal College of Pharmaceutical Technology, Dubrajpur 731123, India;
| | - Mohammad Hosein Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA;
| |
Collapse
|
37
|
Sun Y, Davis E. Nanoplatforms for Targeted Stimuli-Responsive Drug Delivery: A Review of Platform Materials and Stimuli-Responsive Release and Targeting Mechanisms. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:746. [PMID: 33809633 PMCID: PMC8000772 DOI: 10.3390/nano11030746] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
To achieve the promise of stimuli-responsive drug delivery systems for the treatment of cancer, they should (1) avoid premature clearance; (2) accumulate in tumors and undergo endocytosis by cancer cells; and (3) exhibit appropriate stimuli-responsive release of the payload. It is challenging to address all of these requirements simultaneously. However, the numerous proof-of-concept studies addressing one or more of these requirements reported every year have dramatically expanded the toolbox available for the design of drug delivery systems. This review highlights recent advances in the targeting and stimuli-responsiveness of drug delivery systems. It begins with a discussion of nanocarrier types and an overview of the factors influencing nanocarrier biodistribution. On-demand release strategies and their application to each type of nanocarrier are reviewed, including both endogenous and exogenous stimuli. Recent developments in stimuli-responsive targeting strategies are also discussed. The remaining challenges and prospective solutions in the field are discussed throughout the review, which is intended to assist researchers in overcoming interdisciplinary knowledge barriers and increase the speed of development. This review presents a nanocarrier-based drug delivery systems toolbox that enables the application of techniques across platforms and inspires researchers with interdisciplinary information to boost the development of multifunctional therapeutic nanoplatforms for cancer therapy.
Collapse
Affiliation(s)
| | - Edward Davis
- Materials Engineering Program, Mechanical Engineering Department, Auburn University, 101 Wilmore Drive, Auburn, AL 36830, USA;
| |
Collapse
|
38
|
Dharmayanti C, Gillam TA, Klingler-Hoffmann M, Albrecht H, Blencowe A. Strategies for the Development of pH-Responsive Synthetic Polypeptides and Polymer-Peptide Hybrids: Recent Advancements. Polymers (Basel) 2021; 13:624. [PMID: 33669548 PMCID: PMC7921987 DOI: 10.3390/polym13040624] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/11/2021] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Synthetic polypeptides and polymer-peptide hybrid materials have been successfully implemented in an array of biomedical applications owing to their biocompatibility, biodegradability and ability to mimic natural proteins. In addition, these materials have the capacity to form complex supramolecular structures, facilitate specific biological interactions, and incorporate a diverse selection of functional groups that can be used as the basis for further synthetic modification. Like conventional synthetic polymers, polypeptide-based materials can be designed to respond to external stimuli (e.g., light and temperature) or changes in the environmental conditions (e.g., redox reactions and pH). In particular, pH-responsive polypeptide-based systems represent an interesting avenue for the preparation of novel drug delivery systems that can exploit physiological or pathological pH variations within the body, such as those that arise in the extracellular tumour microenvironment, intracellularly within endosomes/lysosomes, or during tissue inflammation. Here, we review the significant progress made in advancing pH-responsive polypeptides and polymer-peptide hybrid materials during the last five years, with a particular emphasis on the manipulation of ionisable functional groups, pH-labile linkages, pH-sensitive changes to secondary structure, and supramolecular interactions.
Collapse
Affiliation(s)
- Cintya Dharmayanti
- Applied Chemistry and Translational Biomaterials Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (C.D.); (T.A.G.)
| | - Todd A. Gillam
- Applied Chemistry and Translational Biomaterials Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (C.D.); (T.A.G.)
- Surface Interactions and Soft Matter Group, Future Industries Institute, University of South Australia, Mawson Lakes, SA 5095, Australia
| | | | - Hugo Albrecht
- Drug Discovery and Development Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Anton Blencowe
- Applied Chemistry and Translational Biomaterials Group, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia; (C.D.); (T.A.G.)
| |
Collapse
|
39
|
Ofridam F, Tarhini M, Lebaz N, Gagnière É, Mangin D, Elaissari A. pH
‐sensitive polymers: Classification and some fine potential applications. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5230] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Fabrice Ofridam
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Mohamad Tarhini
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, ISA UMR 5280 Villeurbanne France
| | - Noureddine Lebaz
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Émilie Gagnière
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Denis Mangin
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, LAGEPP UMR 5007 Villeurbanne France
| | - Abdelhamid Elaissari
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, ISA UMR 5280 Villeurbanne France
| |
Collapse
|
40
|
Alven S, Aderibigbe BA. The Therapeutic Efficacy of Dendrimer and Micelle Formulations for Breast Cancer Treatment. Pharmaceutics 2020; 12:E1212. [PMID: 33333778 PMCID: PMC7765183 DOI: 10.3390/pharmaceutics12121212] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is among the most common types of cancer in women and it is the cause of a high rate of mortality globally. The use of anticancer drugs is the standard treatment approach used for this type of cancer. However, most of these drugs are limited by multi-drug resistance, drug toxicity, poor drug bioavailability, low water solubility, poor pharmacokinetics, etc. To overcome multi-drug resistance, combinations of two or more anticancer drugs are used. However, the combination of two or more anticancer drugs produce toxic side effects. Micelles and dendrimers are promising drug delivery systems that can overcome the limitations associated with the currently used anticancer drugs. They have the capability to overcome drug resistance, reduce drug toxicity, improve the drug solubility and bioavailability. Different classes of anticancer drugs have been loaded into micelles and dendrimers, resulting in targeted drug delivery, sustained drug release mechanism, increased cellular uptake, reduced toxic side effects of the loaded drugs with enhanced anticancer activity in vitro and in vivo. This review article reports the biological outcomes of dendrimers and micelles loaded with different known anticancer agents on breast cancer in vitro and in vivo.
Collapse
Affiliation(s)
| | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa;
| |
Collapse
|
41
|
Iqbal S, Qu Y, Dong Z, Zhao J, Rauf Khan A, Rehman S, Zhao Z. Poly (β‐amino esters) based potential drug delivery and targeting polymer; an overview and perspectives (review). Eur Polym J 2020. [DOI: 10.1016/j.eurpolymj.2020.110097] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
42
|
Recent advancement and development of chitin and chitosan-based nanocomposite for drug delivery: Critical approach to clinical research. ARAB J CHEM 2020. [DOI: 10.1016/j.arabjc.2020.10.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
43
|
Luo K, Yin S, Zhang R, Yu H, Wang G, Li J. Multifunctional composite nanoparticles based on hyaluronic acid-paclitaxel conjugates for enhanced cancer therapy. Int J Pharm 2020; 589:119870. [DOI: 10.1016/j.ijpharm.2020.119870] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/05/2020] [Accepted: 09/06/2020] [Indexed: 12/22/2022]
|
44
|
Binary blended co-delivery nanoparticles with the characteristics of precise pH-responsive acting on tumor microenvironment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 117:111370. [PMID: 32919698 DOI: 10.1016/j.msec.2020.111370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 07/19/2020] [Accepted: 07/30/2020] [Indexed: 01/12/2023]
Abstract
Although combined chemotherapy had achieved the ideal efficacy in clinical anti-cancer therapeutic, the issues that need to be addressed are non-targeting and toxic-side effects of small molecule chemical drug (SMCD). In this study, we designed and prepared a novel binary blended co-delivered nanoparticles (BBCD NPs) with pH-responsive feature on tumor microenvironment. The BBCD NPs consists of two kind of drug-loaded NPs, in one of which carboxymethyl chitosan (CMC) and Poly (lactic-co-glycolic acid) (PLGA) were chosen as delivery carrier to load anti-cancer drug vincristine (VCR), named CMC-PLGA-VCR NPs (or CPNPVCR); and in the other of which methoxy poly(ethylene glycol)-poly(β-amino ester) (mPEG-PAE) were chosen as delivery carrier to load anti-fibrotic drug pirfenidone (PFD), named mPEG-PAE-PFD NPs (or PPNPPFD). Then, the two types of NPs (CPNPVCR and PPNPPFD) were physically mixed in mass ratios to form BBCD NPs, which was named CPNPVCR&PPNPPFD. CPNPVCR&PPNPPFD had good encapsulation efficiency and loading capacity, and the particle size distribution was uniform. In cytotoxicity experiments and non-contact co-culture studies in vitro, the model drugs loaded in CPNPVCR&PPNPPFD could respectively target cancer cell and cancer associated fibroblast (CAF) owing to the precise pH-sensitive drug release in the pharmacological targets and show stronger synergism than that of the combined treatment of two free drugs. As a modularity and assemble ability feature in design, BBCD NPs would have the advantages on the terms of concise on preparation process, controllable on quality standard, stable in natural environment storage. The research results can provide scientific evidence for the further development of a novel drug co-delivery system with multi-type cell targets.
Collapse
|
45
|
Li M, Zhao G, Su WK, Shuai Q. Enzyme-Responsive Nanoparticles for Anti-tumor Drug Delivery. Front Chem 2020; 8:647. [PMID: 32850662 PMCID: PMC7406800 DOI: 10.3389/fchem.2020.00647] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/22/2020] [Indexed: 01/10/2023] Open
Abstract
The past few decades have seen great progress in the exploration of nanoparticles (NPs) as novel tools for cancer treatments and diagnosis. Practical and reliable application of nanoparticle-based technology in clinical transformation remains nevertheless an ongoing challenge. The design, preparation, and evaluation of various smart NPs with specific physicochemical responses in tumor-related physiological conditions have been of great interests in both academic and clinical research. Of particular, smart enzyme-responsive nanoparticles can predictively and selectively react with specific enzymes expressed in tumor tissues, leading to targeted delivery of anti-tumor drugs, reduced systemic toxicity, and improved therapeutic effect. In addition, NPs interact with internal enzymes usually under mild conditions (low temperature, aqueous media, neutral or close to neutral pH) with high efficiency. In this review, recent advances in the past 5 years in enzyme-responsive nanoparticles for anti-tumor drug delivery are summarized and discussed. The following contents are divided based on the different action sites of enzymes toward NPs, notably hydrophobic core, cleavable/uncleavable linker, hydrophilic crown, and targeting ligand. Enzyme-engaged destruction of any component of these delicate nanoparticle structures could result in either targeting drug delivery or controlled drug release.
Collapse
Affiliation(s)
- Mengqian Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Guangkuo Zhao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Wei-Ke Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Qi Shuai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
46
|
Emerging era of “somes”: polymersomes as versatile drug delivery carrier for cancer diagnostics and therapy. Drug Deliv Transl Res 2020; 10:1171-1190. [PMID: 32504410 DOI: 10.1007/s13346-020-00789-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Over the past two decades, polymersomes have been widely investigated for the delivery of diagnostic and therapeutic agents in cancer therapy. Polymersomes are stable polymeric vesicles, which are prepared using amphiphilic block polymers of different molecular weights. The use of high molecular weight amphiphilic copolymers allows for possible manipulation of membrane characteristics, which in turn enhances the efficiency of drug delivery. Polymersomes are more stable in comparison with liposomes and show less toxicity in vivo. Furthermore, their ability to encapsulate both hydrophilic and hydrophobic drugs, significant biocompatibility, robustness, high colloidal stability, and simple methods for ligands conjugation make polymersomes a promising candidate for therapeutic drug delivery in cancer therapy. This review is focused on current development in the application of polymersomes for cancer therapy and diagnosis. Graphical abstract.
Collapse
|
47
|
Wang MZ, Niu J, Ma HJ, Dad HA, Shao HT, Yuan TJ, Peng LH. Transdermal siRNA delivery by pH-switchable micelles with targeting effect suppress skin melanoma progression. J Control Release 2020; 322:95-107. [DOI: 10.1016/j.jconrel.2020.03.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/29/2020] [Accepted: 03/16/2020] [Indexed: 01/29/2023]
|
48
|
Guan Y, Wang LY, Wang B, Ding MH, Bao YL, Tan SW. Recent Advances of D-α-tocopherol Polyethylene Glycol 1000 Succinate Based Stimuli-responsive Nanomedicine for Cancer Treatment. Curr Med Sci 2020; 40:218-231. [PMID: 32337683 DOI: 10.1007/s11596-020-2185-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/07/2020] [Indexed: 01/13/2023]
Abstract
D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) is a pharmaceutical excipient approved by Chinese NMPA and FDA of USA. It's widely applied as a multifunctional drug carrier for nanomedicine. The advantages of TPGS include P-glycoprotein (P-gp) inhibition, penetration promotion, apoptosis induction via mitochondrial-associated apoptotic pathways, multidrug resistant (MDR) reversion, metastasis inhibition and so on. TPGS-based drug delivery systems which are responding to external stimulus can combine the inhibitory functions of TPGS towards P-gp with the environmentally responsive controlled release property and thus exerts a synergistic anti-cancer effect, through increased intracellular drug concentration in tumors cells and well-controlled drug release behavior. In this review, TPGS-based nano-sized delivery systems responsive to different stimuli were summarized and discussed, including pH-responsive, redoxresponsive and multi-responsive systems in various formulations. The achievements, mechanisms and different characteristics of TPGS-based stimuli-responsive drug-delivery systems in tumor therapy were also outlined.
Collapse
Affiliation(s)
- Yang Guan
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lin-Yan Wang
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bo Wang
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Mei-Hong Ding
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yu-Ling Bao
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Song-Wei Tan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
49
|
Yu C, Wang L, Xu Z, Teng W, Wu Z, Xiong D. Smart micelles self-assembled from four-arm star polymers as potential drug carriers for pH-triggered DOX release. JOURNAL OF POLYMER RESEARCH 2020. [DOI: 10.1007/s10965-020-02108-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
50
|
Wang G, Song L, Hou X, Kala S, Wong KF, Tang L, Dai Y, Sun L. Surface-modified GVs as nanosized contrast agents for molecular ultrasound imaging of tumor. Biomaterials 2020; 236:119803. [PMID: 32028170 DOI: 10.1016/j.biomaterials.2020.119803] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/11/2020] [Accepted: 01/19/2020] [Indexed: 12/13/2022]
Abstract
Nanobubbles, as a kind of new ultrasound contrast agent (UCAs), have shown promise to penetrate tumor vasculature to allow for targeted imaging. However, their inherent physical instability is an ongoing concern that could weaken their imaging ability with ultrasound. Gas vesicles (GVs), which are genetically encoded, naturally stable nanostructures, have been developed as the first ultrasonic biomolecular reporters which showed strong contrast enhancement. However, further development of tumor imaging with GVs is limited by the quick clearance of GVs by the reticuloendothelial system (RES). Here, we developed PEGylated HA-GVs (PH-GVs) for in-tumor molecular ultrasound imaging by integrating polyethylene glycol (PEG) and hyaluronic acid (HA) in GV shells. PH-GVs were observed to accumulate around CD44-positive cells (SCC7) but not be internalized by macrophage cell line RAW 264.7. Green fluorescence from PH-GVs was found around cell nuclei in the tumor site after 6 h and the signal was sustained over 48 h following tail injection, demonstrating PH-GVs' ability to escape the clearance from the RES and to penetrate tumor vasculature through enhanced permeability and retention (EPR) effects. Further, PH-GVs produced strong ultrasound contrast in the tumor site in vivo, with no obvious side-effects detected following intravenous injection. Thus, we demonstrate the potential of PH-GVs as novel, nanosized and targeted UCAs for efficient and specific molecular tumor imaging, paving the way for the application of GVs in precise and personalized medicine.
Collapse
Affiliation(s)
- Guohao Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Lin Song
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Xuandi Hou
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Shashwati Kala
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Kin Fung Wong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Liya Tang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region
| | - Yunlu Dai
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau
| | - Lei Sun
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong Special Administrative Region.
| |
Collapse
|