1
|
Li Y, He J, Liu J, Um W, Ding J. Challenges and opportunities of poly(amino acid) nanomedicines in cancer therapy. Nanomedicine (Lond) 2024; 19:2495-2504. [PMID: 39381990 PMCID: PMC11520535 DOI: 10.1080/17435889.2024.2402677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024] Open
Abstract
Poly(amino acid) nanomedicines hold significant promise for cancer therapy. However, their clinical translation has not matched the extensive efforts of scientists or the burgeoning body of research. The therapeutic outcomes with most nanomedicines often fall short of the promising results observed in animal experiments. This review explores the challenges faced in cancer therapy using poly(amino acid) nanomedicines, particularly addressing the controversies surrounding the enhanced permeability and retention effect and the lack of methods for controlled and reproducible mass production of poly(amino acid) nanomedicines. Furthermore, this review examines the opportunities emerging in this field due to the rapid advancements in artificial intelligence.
Collapse
Affiliation(s)
- Yuce Li
- College of Life Sciences & Health, Wuhan University of Science & Technology, 2 Huangjiahuxi Road, Wuhan, 430065, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Jing He
- College of Life Sciences & Health, Wuhan University of Science & Technology, 2 Huangjiahuxi Road, Wuhan, 430065, P. R. China
| | - Jixiu Liu
- College of Life Sciences & Health, Wuhan University of Science & Technology, 2 Huangjiahuxi Road, Wuhan, 430065, P. R. China
| | - Wooram Um
- Department of Biotechnology, Pukyong National University, 45 Yongso-ro, Nam-Gu, Busan, 48513, Republic of Korea
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| |
Collapse
|
2
|
Nazli A, Irshad Khan MZ, Rácz Á, Béni S. Acid-sensitive prodrugs; a promising approach for site-specific and targeted drug release. Eur J Med Chem 2024; 276:116699. [PMID: 39089000 DOI: 10.1016/j.ejmech.2024.116699] [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: 02/26/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 08/03/2024]
Abstract
Drugs administered through conventional formulations are devoid of targeting and often spread to various undesired sites, leading to sub-lethal concentrations at the site of action and the emergence of undesired effects. Hence, therapeutic agents should be delivered in a controlled manner at target sites. Currently, stimuli-based drug delivery systems have demonstrated a remarkable potential for the site-specific delivery of therapeutic moieties. pH is one of the widely exploited stimuli for drug delivery as several pathogenic conditions such as tumor cells, infectious and inflammatory sites are characterized by a low pH environment. This review article aims to demonstrate various strategies employed in the design of acid-sensitive prodrugs, providing an overview of commercially available acid-sensitive prodrugs. Furthermore, we have compiled the progress made for the development of new acid-sensitive prodrugs currently undergoing clinical trials. These prodrugs include albumin-binding prodrugs (Aldoxorubicin and DK049), polymeric micelle (NC-6300), polymer conjugates (ProLindac™), and an immunoconjugate (IMMU-110). The article encompasses a broad spectrum of studies focused on the development of acid-sensitive prodrugs for anticancer, antibacterial, and anti-inflammatory agents. Finally, the challenges associated with the acid-sensitive prodrug strategy are discussed, along with future directions.
Collapse
Affiliation(s)
- Adila Nazli
- Department of Pharmacognosy, Semmelweis University, 1085, Budapest, Hungary.
| | | | - Ákos Rácz
- Department of Pharmacognosy, Semmelweis University, 1085, Budapest, Hungary.
| | - Szabolcs Béni
- Integrative Health and Environmental Analysis Research Laboratory, Department of Analytical Chemistry, Institute of Chemistry, Eötvös Loránd University, 1117, Budapest, Hungary.
| |
Collapse
|
3
|
Kumar A, Shukla R. Current strategic arsenal and advances in nose to brain nanotheranostics for therapeutic intervention of glioblastoma multiforme. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-35. [PMID: 39250527 DOI: 10.1080/09205063.2024.2396721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/21/2024] [Indexed: 09/11/2024]
Abstract
The fight against Glioblastoma multiforme (GBM) is ongoing and the long-term outlook for GBM remains challenging due to low prognosis but every breakthrough brings us closer to improving patient outcomes. Significant hurdles in GBM are heterogeneity, fortified tumor location, and blood-brain barrier (BBB), hindering adequate drug concentrations within functioning brain regions, thus leading to low survival rates. The nasal passageway has become an appealing location to commence the course of cancer therapy. Utilization of the nose-to-brain (N2B) route for drug delivery takes a sidestep from the BBB to allow therapeutics to directly access the central nervous system (CNS) and enhance drug localization in the vicinity of the tumor. This comprehensive review provides insights into pertinent anatomy and cellular organization of the nasal cavity, present-day diagnostic tools, intracranial invasive therapies, and advancements in intranasal (IN) therapies in GBM models for better clinical outcomes. Also, this review highlights groundbreaking carriers and delivery techniques that could revolutionize GBM management such as biomimetics, image guiding-drug delivery, and photodynamic and photothermal therapies for GBM management.
Collapse
Affiliation(s)
- Ankit Kumar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| |
Collapse
|
4
|
Serras A, Faustino C, Pinheiro L. Functionalized Polymeric Micelles for Targeted Cancer Therapy: Steps from Conceptualization to Clinical Trials. Pharmaceutics 2024; 16:1047. [PMID: 39204392 PMCID: PMC11359152 DOI: 10.3390/pharmaceutics16081047] [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: 07/08/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer is still ranked among the top three causes of death in the 30- to 69-year-old age group in most countries and carries considerable societal and macroeconomic costs that differ depending on the cancer type, geography, and patient gender. Despite advances in several pharmacological approaches, the lack of stability and specificity, dose-related toxicity, and limited bioavailability of chemotherapy (standard therapy) pose major obstacles in cancer treatment, with multidrug resistance being a driving factor in chemotherapy failure. The past three decades have been the stage for intense research activity on the topic of nanomedicine, which has resulted in many nanotherapeutics with reduced toxicity, increased bioavailability, and improved pharmacokinetics and therapeutic efficacy employing smart drug delivery systems (SDDSs). Polymeric micelles (PMs) have become an auspicious DDS for medicinal compounds, being used to encapsulate hydrophobic drugs that also exhibit substantial toxicity. Through preclinical animal testing, PMs improved pharmacokinetic profiles and increased efficacy, resulting in a higher safety profile for therapeutic drugs. This review focuses on PMs that are already in clinical trials, traveling the pathways from preclinical to clinical studies until introduction to the market.
Collapse
Affiliation(s)
| | - Célia Faustino
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa (ULisboa), Avenida Professor Gama PintoGama Pinto, 1649-003 Lisboa, Portugal; (A.S.); (L.P.)
| | | |
Collapse
|
5
|
Chu B, Deng H, Niu T, Qu Y, Qian Z. Stimulus-Responsive Nano-Prodrug Strategies for Cancer Therapy: A Focus on Camptothecin Delivery. SMALL METHODS 2024; 8:e2301271. [PMID: 38085682 DOI: 10.1002/smtd.202301271] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/15/2023] [Indexed: 08/18/2024]
Abstract
Camptothecin (CPT) is a highly cytotoxic molecule with excellent antitumor activity against various cancers. However, its clinical application is severely limited by poor water solubility, easy inactivation, and severe toxicity. Structural modifications and nanoformulations represent two crucial avenues for camptothecin's development. However, the potential for further structural modifications is limited, and camptothecin nanoparticles fabricated via physical loading have the drawbacks of low drug loading and leakage. Prodrug-based CPT nanoformulations have shown unique advantages, including increased drug loading, reduced burst release, improved bioavailability, and minimal toxic side effects. Stimulus-responsive CPT nano-prodrugs that respond to various endogenous or exogenous stimuli by introducing various activatable linkers to achieve spatiotemporally responsive drug release at the tumor site. This review comprehensively summarizes the latest research advances in stimulus-responsive CPT nano-prodrugs, including preparation strategies, responsive release mechanisms, and their applications in cancer therapy. Special focus is placed on the release mechanisms and characteristics of various stimulus-responsive CPT nano-prodrugs and their application in cancer treatment. Furthermore, clinical applications of CPT prodrugs are discussed. Finally, challenges and future research directions for CPT nano-prodrugs are discussed. This review to be valuable to readers engaged in prodrug research is expected.
Collapse
Affiliation(s)
- Bingyang Chu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hanzhi Deng
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Qu
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiyong Qian
- Department of Hematology and Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
6
|
Wu L, Zhao K, Xu L, Cui J, Ruan L, Bei S, Cao J, Qi X, Shen S. Macrophages-mediated delivery of protoporphyrin for sonodynamic therapy of rheumatoid arthritis. ULTRASONICS SONOCHEMISTRY 2024; 107:106928. [PMID: 38820932 PMCID: PMC11179255 DOI: 10.1016/j.ultsonch.2024.106928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/21/2024] [Accepted: 05/25/2024] [Indexed: 06/02/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic inflammatory disease characterized by infiltration of inflammatory cells, hyperplasia of synovium, and destruction of the joint cartilage. Owing to the low drug delivery efficiency and limited immunosuppression effect, complete cure for RA remains a formidable challenge. Here, we show that live macrophages (Mφs) carrying protoporphyrin-loaded Fe3O4 nanoparticles can migrate to the RA tissues and inhibit the inflammation by sonodynamic therapy. The inflammation of RA leads to the release of cytokines, which guides the migration of the Mφs into the RA tissues, realizing precise delivery of therapeutics. The following sonodynamic therapy induced by ultrasound and protoporphyrin destructs the proliferating synovial cells and also infiltrated inflammatory cells, demonstrating significant therapeutic effect for RA. Meanwhile, the cytokines and relapse of RA can be remarkably suppressed because of the efficient damage to the resident inflammatory cells.
Collapse
Affiliation(s)
- Lin Wu
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Kai Zhao
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Leyuan Xu
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Junming Cui
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Li Ruan
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Shifang Bei
- Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China.
| | - Jin Cao
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xueyong Qi
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Song Shen
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
7
|
Shang S, Li X, Wang H, Zhou Y, Pang K, Li P, Liu X, Zhang M, Li W, Li Q, Chen X. Targeted therapy of kidney disease with nanoparticle drug delivery materials. Bioact Mater 2024; 37:206-221. [PMID: 38560369 PMCID: PMC10979125 DOI: 10.1016/j.bioactmat.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/09/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
With the development of nanomedicine, nanomaterials have been widely used, offering specific drug delivery to target sites, minimal side effects, and significant therapeutic effects. The kidneys have filtration and reabsorption functions, with various potential target cell types and a complex structural environment, making the strategies for kidney function protection and recovery after injury complex. This also lays the foundation for the application of nanomedicine in kidney diseases. Currently, evidence in preclinical and clinical settings supports the feasibility of targeted therapy for kidney diseases using drug delivery based on nanomaterials. The prerequisite for nanomedicine in treating kidney diseases is the use of carriers with good biocompatibility, including nanoparticles, hydrogels, liposomes, micelles, dendrimer polymers, adenoviruses, lysozymes, and elastin-like polypeptides. These carriers have precise renal uptake, longer half-life, and targeted organ distribution, protecting and improving the efficacy of the drugs they carry. Additionally, attention should also be paid to the toxicity and solubility of the carriers. While the carriers mentioned above have been used in preclinical studies for targeted therapy of kidney diseases both in vivo and in vitro, extensive clinical trials are still needed to ensure the short-term and long-term effects of nano drugs in the human body. This review will discuss the advantages and limitations of nanoscale drug carrier materials in treating kidney diseases, provide a more comprehensive catalog of nanocarrier materials, and offer prospects for their drug-loading efficacy and clinical applications.
Collapse
Affiliation(s)
- Shunlai Shang
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Xiangmeng Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, China
- Peking Union Medical College, Beijing, China
| | - Haoran Wang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Yena Zhou
- School of Medicine, Nankai University, Tianjin, China
| | - Keying Pang
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiaomin Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Min Zhang
- Department of Nephrology, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
| | - Wenge Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Qinggang Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
8
|
Beach M, Nayanathara U, Gao Y, Zhang C, Xiong Y, Wang Y, Such GK. Polymeric Nanoparticles for Drug Delivery. Chem Rev 2024; 124:5505-5616. [PMID: 38626459 PMCID: PMC11086401 DOI: 10.1021/acs.chemrev.3c00705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
9
|
Saddam Hussain M, Khetan R, Clulow AJ, Ganesan R, MacMillan A, Robinson N, Ahmed-Cox A, Krasowska M, Albrecht H, Blencowe A. Teaching an Old Dog New Tricks: A Global Approach to Enhancing the Cytotoxicity of Drug-Loaded, Non-responsive Micelles Using Oligoelectrolytes. ACS APPLIED MATERIALS & INTERFACES 2024; 16:9736-9748. [PMID: 38349780 DOI: 10.1021/acsami.3c16551] [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: 02/15/2024]
Abstract
Polymeric micelles have been extensively studied as vectors for the delivery of hydrophobic drugs for the treatment of cancers and other diseases. Despite intensive research, few formulations provide significant benefits, and even fewer have been clinically approved. While many traditional non-responsive micelles have excellent safety profiles, they lack the ability to respond to the intracellular environment and release their cargo in a spatiotemporally defined manner to effectively deliver large doses of cytotoxic drugs into the cytosol of cells that overwhelm efflux pumps. As a novel and adaptable strategy, we hypothesized that well-established non-responsive polymeric micelles could be augmented with a pH-trigger via the co-encapsulation of cytocompatible oligoelectrolytes, which would allow rapid cargo release in the endosome, leading to increased cytotoxicity. Herein, we demonstrate how this strategy can be applied to render non-responsive micelles pH-responsive, resulting in abrupt cargo release at specific and tunable pH values compatible with endosomal delivery, which significantly increased their cytotoxicity up to 3-fold in an ovarian adenocarcinoma (SKOV-3) cell line compared to non-responsive micelles. In comparison, the oligoelectrolyte-loaded micelles were significantly less toxic to healthy 3T3 fibroblasts, indicating a selective cargo release in cancer cell lines. Oligoelectrolytes can be co-encapsulated in the micelles along with drugs at high encapsulation efficiency percentages, which are both ejected from the micelle core upon oligoelectrolyte ionization. Mechanistically, the increase in cytotoxicity appears to also result from the accelerated endosomal escape of the cargo caused by disruption of the endosomal membrane by the simultaneous release of the oligoelectrolytes from the micelles. Furthermore, we show how this approach is broadly applicable to non-responsive micelles regardless of their composition and various classes of hydrophobic chemotherapeutics. The preliminary studies presented here reveal the versatility and wide scope of oligoelectrolyte-mediated, pH-triggered drug release as a compelling and powerful strategy to enhance the cytotoxicity of non-responsive polymeric micelles.
Collapse
Affiliation(s)
- Md Saddam Hussain
- Applied Chemistry and Translational Biomaterials (ACTB) Group, Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Riya Khetan
- Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Andrew J Clulow
- Australian Synchrotron, Australian Nuclear Science and Technology Organisation (ANSTO), 800 Blackburn Road, Clayton, Victoria 3168, Australia
- Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Raja Ganesan
- Centre for Cancer Biology, UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Alexander MacMillan
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, New South Wales 2033, Australia
| | - Nirmal Robinson
- Centre for Cancer Biology, UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Aria Ahmed-Cox
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, New South Wales 2033, Australia
- Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales 2750, Australia
- Australian Centre for Nanomedicine, Faculty of Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Marta Krasowska
- Surface Interactions and Soft Matter (SISM) Group, Future Industries Institute, UniSA STEM, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Hugo Albrecht
- Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Anton Blencowe
- Applied Chemistry and Translational Biomaterials (ACTB) Group, Centre for Pharmaceutical Innovation (CPI), UniSA CHS, University of South Australia, Adelaide, South Australia 5000, Australia
| |
Collapse
|
10
|
Nag S, Mitra O, Tripathi G, Adur I, Mohanto S, Nama M, Samanta S, Gowda BHJ, Subramaniyan V, Sundararajan V, Kumarasamy V. Nanomaterials-assisted photothermal therapy for breast cancer: State-of-the-art advances and future perspectives. Photodiagnosis Photodyn Ther 2024; 45:103959. [PMID: 38228257 DOI: 10.1016/j.pdpdt.2023.103959] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/16/2023] [Accepted: 12/29/2023] [Indexed: 01/18/2024]
Abstract
Breast cancer (BC) remains an enigmatic fatal modality ubiquitously prevalent in different parts of the world. Contemporary medicines face severe challenges in remediating and healing breast cancer. Due to its spatial specificity and nominal invasive therapeutic regime, photothermal therapy (PTT) has attracted much scientific attention down the lane. PTT utilizes a near-infrared (NIR) light source to irradiate the tumor target intravenously or non-invasively, which is converted into heat energy over an optical fibre. Dynamic progress in nanomaterial synthesis was achieved with specialized visual, physicochemical, biological, and pharmacological features to make up for the inadequacies and expand the horizon of PTT. Numerous nanomaterials have substantial NIR absorption and can function as efficient photothermal transducers. It is achievable to limit the wavelength range of an absorbance peak for specific nanomaterials by manipulating their synthesis, enhancing the precision and quality of PTT. Along the same lines, various nanomaterials are conjugated with a wide range of surface-modifying chemicals, including polymers and antibodies, which may modify the persistence of the nanomaterial and diminish toxicity concerns. In this article, we tend to put forth specific insights and fundamental conceptualizations on pre-existing PTT and its advances upon conjugation with different biocompatible nanomaterials working in synergy to combat breast cancer, encompassing several strategies like immunotherapy, chemotherapy, photodynamic therapy, and radiotherapy coupled with PTT. Additionally, the role or mechanisms of nanoparticles, as well as possible alternatives to PTT, are summarized as a distinctive integral aspect in this article.
Collapse
Affiliation(s)
- Sagnik Nag
- Department of Bio-Sciences, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India; Integrative Multiomics Lab, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India; Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences (JCSMHS), Monash University Malaysia, Bandar Sunway 47500 Selangor Darul Ehsan, Malaysia.
| | - Oishi Mitra
- Department of Bio-Sciences, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India; Integrative Multiomics Lab, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Garima Tripathi
- Department of Bio-Sciences, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Israrahmed Adur
- Department of Bio-Sciences, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Muskan Nama
- Department of Bio-Sciences, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Souvik Samanta
- Department of Bio-Sciences, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - B H Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India; School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences (JCSMHS), Monash University Malaysia, Bandar Sunway 47500 Selangor Darul Ehsan, Malaysia.
| | - Vino Sundararajan
- Integrative Multiomics Lab, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India.
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia.
| |
Collapse
|
11
|
Ioannou P, Baliou S, Samonis G. Nanotechnology in the Diagnosis and Treatment of Antibiotic-Resistant Infections. Antibiotics (Basel) 2024; 13:121. [PMID: 38391507 PMCID: PMC10886108 DOI: 10.3390/antibiotics13020121] [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: 01/04/2024] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
The development of antimicrobial resistance (AMR), along with the relative reduction in the production of new antimicrobials, significantly limits the therapeutic options in infectious diseases. Thus, novel treatments, especially in the current era, where AMR is increasing, are urgently needed. There are several ongoing studies on non-classical therapies for infectious diseases, such as bacteriophages, antimicrobial peptides, and nanotechnology, among others. Nanomaterials involve materials on the nanoscale that could be used in the diagnosis, treatment, and prevention of infectious diseases. This review provides an overview of the applications of nanotechnology in the diagnosis and treatment of infectious diseases from a clinician's perspective, with a focus on pathogens with AMR. Applications of nanomaterials in diagnosis, by taking advantage of their electrochemical, optic, magnetic, and fluorescent properties, are described. Moreover, the potential of metallic or organic nanoparticles (NPs) in the treatment of infections is also addressed. Finally, the potential use of NPs in the development of safe and efficient vaccines is also reviewed. Further studies are needed to prove the safety and efficacy of NPs that would facilitate their approval by regulatory authorities for clinical use.
Collapse
Affiliation(s)
- Petros Ioannou
- School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Stella Baliou
- School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - George Samonis
- School of Medicine, University of Crete, 71003 Heraklion, Greece
- First Department of Medical Oncology, Metropolitan Hospital of Neon Faliron, 18547 Athens, Greece
| |
Collapse
|
12
|
Chabattula SC, Gupta PK, Govarthanan K, Varadaraj S, Rayala SK, Chakraborty D, Verma RS. Anti-cancer Activity of Biogenic Nat-ZnO Nanoparticles Synthesized Using Nyctanthes arbor-tristis (Nat) Flower Extract. Appl Biochem Biotechnol 2024; 196:382-399. [PMID: 37133677 DOI: 10.1007/s12010-023-04555-1] [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] [Accepted: 04/18/2023] [Indexed: 05/04/2023]
Abstract
Inorganic nanoparticles (NPs) have played an important role as nano-drug delivery systems during cancer therapy in recent years. These NPs can carry cancer therapeutic agents. Due to this, they are considered a promising ancillary to traditional cancer therapies. Among inorganic NPs, Zinc Oxide (ZnO) NPs have been extensively utilized in cellular imaging, gene/drug delivery, anti-microbial, and anti-cancerous applications. In this study, a rapid and cost-effective method was used to synthesize Nat-ZnO NPs using the floral extract of the Nyctanthes arbor-tristis (Nat) plant. Nat-ZnO NPs were physicochemically characterized and tested further on in vitro cancer models. The average hydrodynamic diameter (Zaverage) and the net surface charge of Nat-ZnO NPs were 372.5 ± 70.38 d.nm and -7.03 ± 0.55 mV, respectively. Nat-ZnO NPs exhibited a crystalline nature. HR-TEM analysis showed the triangular shape of NPs. Furthermore, Nat-ZnO NPs were also found to be biocompatible and hemocompatible when tested on mouse fibroblast cells and RBCs. Later, the anti-cancer activity of Nat-ZnO NPs was tested on lung and cervical cancer cells. These NPs displayed potent anti-cancer activity and induced programmed cell death in cancer cells.
Collapse
Affiliation(s)
- Siva Chander Chabattula
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, 600036, India
| | - Piyush Kumar Gupta
- Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248002, India
- Faculty of Health and Life Sciences, INTI International University, 71800, Nilai, Malaysia
| | - Kavitha Govarthanan
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Science and Regenerative Medicine, Bengaluru, Karnataka, 560065, India
| | - Sudha Varadaraj
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, 600036, India
| | - Suresh Kumar Rayala
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, 600036, India.
| | - Debashis Chakraborty
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, Tamilnadu, 600036, India.
| | - Rama Shanker Verma
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamilnadu, 600036, India.
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Teliarganj, Prayagraj, Uttar Pradesh, 211004, India.
| |
Collapse
|
13
|
Wei D, Sun Y, Zhu H, Fu Q. Stimuli-Responsive Polymer-Based Nanosystems for Cancer Theranostics. ACS NANO 2023; 17:23223-23261. [PMID: 38041800 DOI: 10.1021/acsnano.3c06019] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2023]
Abstract
Stimuli-responsive polymers can respond to internal stimuli, such as reactive oxygen species (ROS), glutathione (GSH), and pH, biological stimuli, such as enzymes, and external stimuli, such as lasers and ultrasound, etc., by changing their hydrophobicity/hydrophilicity, degradability, ionizability, etc., and thus have been widely used in biomedical applications. Due to the characteristics of the tumor microenvironment (TME), stimuli-responsive polymers that cater specifically to the TME have been extensively used to prepare smart nanovehicles for the targeted delivery of therapeutic and diagnostic agents to tumor tissues. Compared to conventional drug delivery nanosystems, TME-responsive nanosystems have many advantages, such as high sensitivity, broad applicability among different tumors, functional versatility, and improved biosafety. In recent years, a great deal of research has been devoted to engineering efficient stimuli-responsive polymeric nanosystems, and significant improvement has been made to both cancer diagnosis and therapy. In this review, we summarize some recent research advances involving the use of stimuli-responsive polymer nanocarriers in drug delivery, tumor imaging, therapy, and theranostics. Various chemical stimuli will be described in the context of stimuli-responsive nanosystems. Accordingly, the functional chemical groups responsible for the responsiveness and the strategies to incorporate these groups into the polymer will be discussed in detail. With the research on this topic expending at a fast pace, some innovative concepts, such as sequential and cascade drug release, NIR-II imaging, and multifunctional formulations, have emerged as popular strategies for enhanced performance, which will also be included here with up-to-date illustrations. We hope that this review will offer valuable insights for the selection and optimization of stimuli-responsive polymers to help accelerate their future applications in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Dengshuai Wei
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Hu Zhu
- Maoming People's Hospital, Guangdong 525000, China
| | - Qinrui Fu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
14
|
Shang J, Yang J, Deng Q, Zhou M. Nano-scale drug delivery systems for luteolin: advancements and applications. J Mater Chem B 2023; 11:11198-11216. [PMID: 37986608 DOI: 10.1039/d3tb01753b] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Luteolin (Lu) is a naturally occurring flavonoid compound with a diverse array of pharmacological activities, including anti-tumor, anti-inflammatory, antibacterial, and neuroprotective properties. However, the therapeutic efficacy and clinical application of Lu are significantly hindered by inherent limitations, such as poor water solubility, short half-life, low bioavailability, and potential off-target toxicity. Recent studies have demonstrated that the utilization of nanocarriers presents a promising strategy to enhance the solubility of Lu, prolong its circulation time, and improve its targeting ability. Despite numerous reviews over the past few decades having focused on the source, pharmacological activities, and molecular mechanisms of Lu, there exists a conspicuous gap in the literature regarding a comprehensive review of Lu-loaded nanoformulations and their applications. To address this gap, we present an exhaustive overview of the advancements and applications of nano-scale drug delivery systems specifically designed for Lu. These platforms encompass micelles, nanocarrier-based systems, emulsified drug delivery systems, and vesicular drug delivery systems. We provide detailed insights into the synthetic materials, preparation methods, physicochemical properties, and significant outcomes associated with these nanoformulations. This systematic review will be particularly valuable to researchers seeking novel avenues in the field of nano-delivery strategies and exploring the potential clinical applications of Lu.
Collapse
Affiliation(s)
- Jinlu Shang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Jing Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Qinmin Deng
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
15
|
Guo L, Yang J, Wang H, Yi Y. Multistage Self-Assembled Nanomaterials for Cancer Immunotherapy. Molecules 2023; 28:7750. [PMID: 38067480 PMCID: PMC10707962 DOI: 10.3390/molecules28237750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Advances in nanotechnology have brought innovations to cancer therapy. Nanoparticle-based anticancer drugs have achieved great success from bench to bedside. However, insufficient therapy efficacy due to various physiological barriers in the body remains a key challenge. To overcome these biological barriers and improve the therapeutic efficacy of cancers, multistage self-assembled nanomaterials with advantages of stimuli-responsiveness, programmable delivery, and immune modulations provide great opportunities. In this review, we describe the typical biological barriers for nanomedicines, discuss the recent achievements of multistage self-assembled nanomaterials for stimuli-responsive drug delivery, highlighting the programmable delivery nanomaterials, in situ transformable self-assembled nanomaterials, and immune-reprogramming nanomaterials. Ultimately, we perspective the future opportunities and challenges of multistage self-assembled nanomaterials for cancer immunotherapy.
Collapse
Affiliation(s)
- Lamei Guo
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Jinjun Yang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| |
Collapse
|
16
|
Wang Y, Chen L, Wang Y, Wang X, Qian D, Yan J, Sun Z, Cui P, Yu L, Wu J, He Z. Marine biomaterials in biomedical nano/micro-systems. J Nanobiotechnology 2023; 21:408. [PMID: 37926815 PMCID: PMC10626837 DOI: 10.1186/s12951-023-02112-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/15/2023] [Indexed: 11/07/2023] Open
Abstract
Marine resources in unique marine environments provide abundant, cost-effective natural biomaterials with distinct structures, compositions, and biological activities compared to terrestrial species. These marine-derived raw materials, including polysaccharides, natural protein components, fatty acids, and marine minerals, etc., have shown great potential in preparing, stabilizing, or modifying multifunctional nano-/micro-systems and are widely applied in drug delivery, theragnostic, tissue engineering, etc. This review provides a comprehensive summary of the most current marine biomaterial-based nano-/micro-systems developed over the past three years, primarily focusing on therapeutic delivery studies and highlighting their potential to cure a variety of diseases. Specifically, we first provided a detailed introduction to the physicochemical characteristics and biological activities of natural marine biocomponents in their raw state. Furthermore, the assembly processes, potential functionalities of each building block, and a thorough evaluation of the pharmacokinetics and pharmacodynamics of advanced marine biomaterial-based systems and their effects on molecular pathophysiological processes were fully elucidated. Finally, a list of unresolved issues and pivotal challenges of marine-derived biomaterials applications, such as standardized distinction of raw materials, long-term biosafety in vivo, the feasibility of scale-up, etc., was presented. This review is expected to serve as a roadmap for fundamental research and facilitate the rational design of marine biomaterials for diverse emerging applications.
Collapse
Affiliation(s)
- Yanan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Long Chen
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 55000, Guizhou, China
| | - Yuanzheng Wang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 55000, Guizhou, China.
| | - Xinyuan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Deyao Qian
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Jiahui Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Zeyu Sun
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, 55000, Guizhou, China
| | - Pengfei Cui
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266100, China.
| | - Liangmin Yu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China
| | - Jun Wu
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, 999077, China.
| | - Zhiyu He
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao, 266100, China.
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Sanya, 572024, China.
| |
Collapse
|
17
|
Li G, Song Z, Ru Y, Zhang J, Luo L, Yang W, Wu H, Jin H, Bao X, Wei D, Yan Z, Qu H, Zhu Z, Xue X, Zhou G. Small-molecule nanoprodrug with high drug loading and EGFR, PI3K/AKT dual-inhibiting properties for bladder cancer treatment. EXPLORATION (BEIJING, CHINA) 2023; 3:20220141. [PMID: 37933289 PMCID: PMC10582605 DOI: 10.1002/exp.20220141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/30/2023] [Indexed: 11/08/2023]
Abstract
Bladder cancer (BCa) is one of the most common malignancies worldwide. Although multiple efforts have been made, the 5-year survival rate of patients with BCa remains unchanged in recent years. Overexpression of the epidermal growth factor receptor (EGFR) is found in ≈74% of BCa tissue specimens; however, current EGFR-based targeted therapies show little benefit for BCa patients, as the EGFR downstream pathways appear to be circumvented by other receptor tyrosine kinases (RTKs). In this study, two natural products are identified, namely triptolide (TPL) and hesperidin (HSP), that target and inhibit the EGFR and its downstream PI3K/AKT pathway in BCa. To synergistically combine triptolide and hesperidin, a succinic acid linker was employed to conjugate them and formed an amphiphilic TPL-HSP EGFR-targeting prodrug (THE), which further self-assembled to generate nanoparticles (THE NPs). These NPs allowed the EGFR-targeted delivery of the triptolide and hesperidin, and simultaneous inhibition of the EGFR and PI3K/AKT both in vitro and in vivo. This study provides a promising EGFR-targeted delivery approach with the dual inhibition of the EGFR and PI3K/AKT, while also exhibiting a high drug loading and low toxicity. Our formulation may be a suitable option to deliver natural products for BCa treatment by EGFR-targeted therapy.
Collapse
Affiliation(s)
- Guoyin Li
- College of Life Science and AgronomyZhoukou Normal UniversityZhoukouHenanChina
- Department of Biochemistry and Molecular BiologyState Key Laboratory of Cancer BiologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Zewen Song
- Department of OncologyCentral South University Third Xiangya HospitalChangshaHunanChina
| | - Yi Ru
- Department of Biochemistry and Molecular BiologyState Key Laboratory of Cancer BiologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Jing Zhang
- Department of PathologyXijing HospitalState Key Laboratory of Cancer BiologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Lianxiang Luo
- The Marine Biomedical Research InstituteGuangdong Medical UniversityZhanjiangGuangdongChina
| | - Wei Yang
- Warshel Institute for Computational BiologySchool of Science and EngineeringThe Chinese University of Hong KongShenzhenChina
| | - Hao Wu
- School of Basic Medical SciencesXi'an Key Laboratory of Immune Related DiseasesXi'an Jiaotong UniversityXi'anShaanxiChina
| | - Haibao Jin
- Shanghai Key Laboratory of Advanced Polymeric MaterialsSchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghaiChina
| | - Xuanwen Bao
- Department of Medical OncologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Di Wei
- Graduate SchoolDepartment of Biochemistry and Molecular BiologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Zhao Yan
- Graduate SchoolDepartment of Biochemistry and Molecular BiologyThe Fourth Military Medical UniversityXi'anShaanxiChina
| | - Haijing Qu
- School of PharmacyShanghai Jiao Tong UniversityXi'anShanghaiChina
| | - Zheng Zhu
- Department of MedicineHarvard Medical SchoolBostonMassachusettsUSA
| | - Xiangdong Xue
- School of PharmacyShanghai Jiao Tong UniversityXi'anShanghaiChina
| | - Gang Zhou
- National Translational Science Center for Molecular MedicineDepartment of Cell BiologyState Key Laboratory of Cancer BiologyFourth Military Medical UniversityXi'anShaanxiChina
| |
Collapse
|
18
|
Peng X, Li X, Xie B, Lai Y, Sosnik A, Boucetta H, Chen Z, He W. Gout therapeutics and drug delivery. J Control Release 2023; 362:728-754. [PMID: 37690697 DOI: 10.1016/j.jconrel.2023.09.011] [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: 05/28/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Gout is a common inflammatory arthritis caused by persistently elevated uric acid levels. With the improvement of people's living standards, the consumption of processed food and the widespread use of drugs that induce elevated uric acid, gout rates are increasing, seriously affecting the human quality of life, and becoming a burden to health systems worldwide. Since the pathological mechanism of gout has been elucidated, there are relatively effective drug treatments in clinical practice. However, due to (bio)pharmaceutical shortcomings of these drugs, such as poor chemical stability and limited ability to target the pathophysiological pathways, traditional drug treatment strategies show low efficacy and safety. In this scenario, drug delivery systems (DDS) design that overcome these drawbacks is urgently called for. In this review, we initially describe the pathological features, the therapeutic targets, and the drugs currently in clinical use and under investigation to treat gout. We also comprehensively summarize recent research efforts utilizing lipid, polymeric and inorganic carriers to develop advanced DDS for improved gout management and therapy.
Collapse
Affiliation(s)
- Xiuju Peng
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Xiaotong Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Bing Xie
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Yaoyao Lai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Alejandro Sosnik
- Department of Materials Science and Engineering, Technion - Israel Institute of Technology, Technion City, Haifa 3200003, Israel
| | - Hamza Boucetta
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, PR China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China.
| |
Collapse
|
19
|
Ma M, Zeng H, Yang P, Xu J, Zhang X, He W. Drug Delivery and Therapy Strategies for Osteoporosis Intervention. Molecules 2023; 28:6652. [PMID: 37764428 PMCID: PMC10534890 DOI: 10.3390/molecules28186652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
With the advent of the aging society, osteoporosis (OP) risk increases yearly. Currently, the clinical usage of anti-OP drugs is challenged by recurrent side effects and poor patient compliance, regardless of oral, intravenous, or subcutaneous administration. Properly using a drug delivery system or formulation strategy can achieve targeted drug delivery to the bone, diminish side effects, improve bioavailability, and prolong the in vivo residence time, thus effectively curing osteoporosis. This review expounds on the pathogenesis of OP and the clinical medicaments used for OP intervention, proposes the design approach for anti-OP drug delivery, emphatically discusses emerging novel anti-OP drug delivery systems, and enumerates anti-OP preparations under clinical investigation. Our findings may contribute to engineering anti-OP drug delivery and OP-targeting therapy.
Collapse
Affiliation(s)
- Mingyang Ma
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (M.M.); (H.Z.)
| | - Huiling Zeng
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; (M.M.); (H.Z.)
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 211198, China;
| | - Jiabing Xu
- Taizhou Institute for Drug Control, Taizhou 225316, China;
| | - Xingwang Zhang
- Department of Pharmaceutics, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
20
|
Cai R, Zhang L, Chi H. Recent development of polymer nanomicelles in the treatment of eye diseases. Front Bioeng Biotechnol 2023; 11:1246974. [PMID: 37600322 PMCID: PMC10436511 DOI: 10.3389/fbioe.2023.1246974] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023] Open
Abstract
The eye, being one of the most intricate organs in the human body, hosts numerous anatomical barriers and clearance mechanisms. This highlights the importance of devising a secure and efficacious ocular medication delivery system. Over the past several decades, advancements have been made in the development of a nano-delivery platform based on polymeric micelles. These advancements encompass diverse innovations such as poloxamer, chitosan, hydrogel-encapsulated micelles, and contact lenses embedded with micelles. Such technological evolutions allow for sustained medication retention and facilitate enhanced permeation within the eye, thereby standing as the avant-garde in ocular medication technology. This review provides a comprehensive consolidation of ocular medications predicated on polymer nanomicelles from 2014 to 2023. Additionally, it explores the challenges they pose in clinical applications, a discussion intended to aid the design of future clinical research concerning ocular medication delivery formulations.
Collapse
Affiliation(s)
- Ruijun Cai
- Department of Pharmacy, The People’s Hospital of Jiuquan, Jiuquan, Gansu, China
| | - Ling Zhang
- Department of Pharmacy, The People’s Hospital of Jiuquan, Jiuquan, Gansu, China
| | - Hao Chi
- Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
21
|
Tiwari H, Rai N, Singh S, Gupta P, Verma A, Singh AK, Kajal, Salvi P, Singh SK, Gautam V. Recent Advances in Nanomaterials-Based Targeted Drug Delivery for Preclinical Cancer Diagnosis and Therapeutics. Bioengineering (Basel) 2023; 10:760. [PMID: 37508788 PMCID: PMC10376516 DOI: 10.3390/bioengineering10070760] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Nano-oncology is a branch of biomedical research and engineering that focuses on using nanotechnology in cancer diagnosis and treatment. Nanomaterials are extensively employed in the field of oncology because of their minute size and ultra-specificity. A wide range of nanocarriers, such as dendrimers, micelles, PEGylated liposomes, and polymeric nanoparticles are used to facilitate the efficient transport of anti-cancer drugs at the target tumor site. Real-time labeling and monitoring of cancer cells using quantum dots is essential for determining the level of therapy needed for treatment. The drug is targeted to the tumor site either by passive or active means. Passive targeting makes use of the tumor microenvironment and enhanced permeability and retention effect, while active targeting involves the use of ligand-coated nanoparticles. Nanotechnology is being used to diagnose the early stage of cancer by detecting cancer-specific biomarkers using tumor imaging. The implication of nanotechnology in cancer therapy employs photoinduced nanosensitizers, reverse multidrug resistance, and enabling efficient delivery of CRISPR/Cas9 and RNA molecules for therapeutic applications. However, despite recent advancements in nano-oncology, there is a need to delve deeper into the domain of designing and applying nanoparticles for improved cancer diagnostics.
Collapse
Affiliation(s)
- Harshita Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- Department of Botany, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Nilesh Rai
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Swati Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Priyamvada Gupta
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Ashish Verma
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Akhilesh Kumar Singh
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Sciences, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Kajal
- Department of Agriculture Biotechnology, National Agri-Food Biotechnology Institute, Sahibzada Ajit Singh Nagar 140306, India
| | - Prafull Salvi
- Department of Agriculture Biotechnology, National Agri-Food Biotechnology Institute, Sahibzada Ajit Singh Nagar 140306, India
| | - Santosh Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vibhav Gautam
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
22
|
Guo M, Peng T, Wu C, Pan X, Huang Z. Engineering Ferroptosis Inhibitors as Inhalable Nanomedicines for the Highly Efficient Treatment of Idiopathic Pulmonary Fibrosis. Bioengineering (Basel) 2023; 10:727. [PMID: 37370658 DOI: 10.3390/bioengineering10060727] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/10/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) refers to chronic progressive fibrotic interstitial pneumonia. It is called a "tumor-like disease" and cannot be cured using existing clinical drugs. Therefore, new treatment options are urgently needed. Studies have proven that ferroptosis is closely related to the development of IPF, and ferroptosis inhibitors can slow down the occurrence of IPF by chelating iron or reducing lipid peroxidation. For example, the ferroptosis inhibitor deferoxamine (DFO) was used to treat a mouse model of pulmonary fibrosis, and DFO successfully reversed the IPF phenotype and increased the survival rate of mice from 50% to 90%. Given this, we perceive that the treatment of IPF by delivering ferroptosis inhibitors is a promising option. However, the delivery of ferroptosis inhibitors faces two bottlenecks: low solubility and targeting. For one thing, we consider preparing ferroptosis inhibitors into nanomedicines to improve solubility. For another thing, we propose to deliver nanomedicines through pulmonary drug-delivery system (PDDS) to improve targeting. Compared with oral or injection administration, PDDS can achieve better delivery and accumulation in the lung, while reducing the systemic exposure of the drug, and is an efficient and safe drug-delivery method. In this paper, three possible nanomedicines for PDDS and the preparation methods thereof are proposed to deliver ferroptosis inhibitors for the treatment of IPF. Proper administration devices and challenges in future applications are also discussed. In general, this perspective proposes a promising strategy for the treatment of IPF based on inhalable nanomedicines carrying ferroptosis inhibitors, which can inspire new ideas in the field of drug development and therapy of IPF.
Collapse
Affiliation(s)
- Mengqin Guo
- College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Tingting Peng
- College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511436, China
| |
Collapse
|
23
|
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: 18] [Impact Index Per Article: 18.0] [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
|
24
|
Li Y, Li X, Yi J, Cao Y, Qin Z, Zhong Z, Yang W. Nanoparticle-Mediated STING Activation for Cancer Immunotherapy. Adv Healthc Mater 2023:e2300260. [PMID: 36905358 DOI: 10.1002/adhm.202300260] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Indexed: 03/12/2023]
Abstract
As the first line of host defense against pathogenic infections, innate immunity plays a key role in antitumor immunotherapy. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) (cGAS-STING) pathway has attracted much attention because of the secretion of various proinflammatory cytokines and chemokines. Many STING agonists have been identified and applied into preclinical or clinical trials for cancer immunotherapy. However, the fast excretion, low bioavailability, nonspecificity, and adverse effects of the small molecule STING agonists limit their therapeutic efficacy and in vivo application. Nanodelivery systems with appropriate size, charge, and surface modification are capable of addressing these dilemmas. In this review, the mechanism of the cGAS-STING pathway is discussed and the STING agonists, focusing on nanoparticle-mediated STING therapy and combined therapy for cancers, are summarized. Finally, the future direction and challenges of nano-STING therapy are expounded, emphasizing the pivotal scientific problems and technical bottlenecks and hoping to provide general guidance for its clinical application.
Collapse
Affiliation(s)
- Yongjuan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xinyan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jinmeng Yi
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yongjian Cao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Weijing Yang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan, 450001, China
| |
Collapse
|
25
|
Nanoplatform-based cellular reactive oxygen species regulation for enhanced oncotherapy and tumor resistance alleviation. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
26
|
Figueiredo P, González RD, Carvalho ATP. Insights into the Degradation of Polymer-Drug Conjugates by an Overexpressed Enzyme in Cancer Cells. J Med Chem 2023; 66:2761-2772. [PMID: 36787193 PMCID: PMC9969400 DOI: 10.1021/acs.jmedchem.2c01781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Intensive efforts have been made to provide better treatments to cancer patients. Currently, nanoparticle-based drug delivery systems have gained propulsion, as they can overcome the drawbacks of free drugs. However, drug stability inside the nanocapsule must be ensured to prevent burst release. To overcome this, drugs conjugated to amphiphilic copolymers, assembled into nanoparticles, can provide a sustained release if endogenously degraded. Thus, we have designed and assessed the drug release viability of polymer-drug conjugates by the human Carboxylesterase 2, for a targeted drug activation. We performed molecular dynamics simulations applying a quantum mechanics/molecular mechanics potential to study the degradation profiles of 30 designed conjugates, where six were predicted to be hydrolyzed by this enzyme. We further analyzed the enzyme-substrate environment to delve into what structural features may lead to successful hydrolysis. These findings contribute to the development of new medicines ensuring effective cancer treatments with fewer side effects.
Collapse
Affiliation(s)
- Pedro
R. Figueiredo
- CNC
− Center for Neuroscience and Cell Biology, Institute for Interdisciplinary
Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal,PhD
Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary
Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Ricardo D. González
- CNC
− Center for Neuroscience and Cell Biology, Institute for Interdisciplinary
Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal,PhD
Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary
Research (IIIUC), University of Coimbra, Casa Costa Alemão, 3030-789 Coimbra, Portugal
| | - Alexandra T. P. Carvalho
- CNC
− Center for Neuroscience and Cell Biology, Institute for Interdisciplinary
Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal,Department
of Biocatalysis and Isotope Chemistry, Almac
Sciences, Almac House, 20 Seagoe Industrial Estate, Craigavon BT63 5QD, Northern Ireland, United Kingdom,
| |
Collapse
|
27
|
Brito ME, Mikhtaniuk SE, Neelov IM, Borisov OV, Holm C. Implicit-Solvent Coarse-Grained Simulations of Linear-Dendritic Block Copolymer Micelles. Int J Mol Sci 2023; 24:2763. [PMID: 36769091 PMCID: PMC9917066 DOI: 10.3390/ijms24032763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/20/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The design of nanoassemblies can be conveniently achieved by tuning the strength of the hydrophobic interactions of block copolymers in selective solvents. These block copolymer micelles form supramolecular aggregates, which have attracted great attention in the area of drug delivery and imaging in biomedicine due to their easy-to-tune properties and straightforward large-scale production. In the present work, we have investigated the micellization process of linear-dendritic block copolymers in order to elucidate the effect of branching on the micellar properties. We focus on block copolymers formed by linear hydrophobic blocks attached to either dendritic neutral or charged hydrophilic blocks. We have implemented a simple protocol for determining the equilibrium micellar size, which permits the study of linear-dendritic block copolymers in a wide range of block morphologies in an efficient and parallelizable manner. We have explored the impact of different topological and charge properties of the hydrophilic blocks on the equilibrium micellar properties and compared them to predictions from self-consistent field theory and scaling theory. We have found that, at higher degrees of branching in the corona and for short polymer chains, excluded volume interactions strongly influence the micellar aggregation as well as their effective charge.
Collapse
Affiliation(s)
- Mariano E. Brito
- Institute for Computational Physics, University of Stuttgart, D-70569 Stuttgart, Germany
| | - Sofia E. Mikhtaniuk
- School of Computer Technologies and Control, St. Petersburg National Research University of Information Technologies, Mechanics and Optics, 197101 St. Petersburg, Russia
| | - Igor M. Neelov
- School of Computer Technologies and Control, St. Petersburg National Research University of Information Technologies, Mechanics and Optics, 197101 St. Petersburg, Russia
| | - Oleg V. Borisov
- School of Computer Technologies and Control, St. Petersburg National Research University of Information Technologies, Mechanics and Optics, 197101 St. Petersburg, Russia
- Institut des Sciences Analytiques et de Physico-Chimie pour l’Environnement et les Matériaux, UMR 5254 CNRS UPPA, 64053 Pau, France
| | - Christian Holm
- Institute for Computational Physics, University of Stuttgart, D-70569 Stuttgart, Germany
| |
Collapse
|
28
|
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: 45] [Impact Index Per Article: 45.0] [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
|
29
|
Ye J, Li L, Yin J, Wang H, Li R, Yang Y, Guan Y, Xia X, Liu Y. Tumor-targeting intravenous lipid emulsion of paclitaxel: Characteristics, stability, toxicity, and toxicokinetics. J Pharm Anal 2022; 12:901-912. [PMID: 36605580 PMCID: PMC9805944 DOI: 10.1016/j.jpha.2022.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 11/07/2022] Open
Abstract
Lipid nanoemulsions are promising nanodrug delivery carriers that can improve the efficacy and safety of paclitaxel (PTX). However, no intravenous lipid emulsion of PTX has been approved for clinical treatment, and systemic safety profiles have not yet been reported. Here we outline the development of a PTX-loaded tumor-targeting intravenous lipid emulsion (PTX Emul) and describe its characteristics, colloidal stability, and systemic safety profiles in terms of acute toxicity, long-term toxicity, and toxicokinetics. We also compare PTX Emul with conventional PTX injection. Results showed that PTX Emul exhibited an ideal average particle size (approximately 160 nm) with narrow size distribution and robust colloidal stability under different conditions. Hypersensitivity reaction and hemolysis tests revealed that PTX Emul did not induce hypersensitivity reactions and had no hemolytic potential. In addition, where the alleviated systemic toxicity of PTX Emul may be attributed to the altered toxicokinetic characteristics in beagle dogs, including the decreased AUC and increased plasma clearance and volume of distribution, PTX Emul alleviated acute and long-term toxicity as evidenced by the enhanced the median lethal dose and approximate lethal dose, moderate body weight change, decreased bone marrow suppression and organ toxicity compared with those under PTX injection at the same dose. A fundamental understanding of the systemic safety profiles, high tumor-targeting efficiency, and superior antitumor activity in vivo of PTX Emul can provide powerful evidence of its therapeutic potential as a future treatment for breast cancer.
Collapse
Affiliation(s)
- Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lin Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jiye Yin
- National Beijing Center for Drug Safety Evaluation and Research, Beijing Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Renjie Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yongbiao Guan
- National Beijing Center for Drug Safety Evaluation and Research, Beijing Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China,Corresponding author.
| | - Xuejun Xia
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Corresponding author.
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Corresponding author.
| |
Collapse
|
30
|
Li B, Tan T, Chu W, Zhang Y, Ye Y, Wang S, Qin Y, Tang J, Cao X. Co-delivery of paclitaxel (PTX) and docosahexaenoic acid (DHA) by targeting lipid nanoemulsions for cancer therapy. Drug Deliv 2022; 29:75-88. [PMID: 34964421 PMCID: PMC8735879 DOI: 10.1080/10717544.2021.2018523] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/06/2021] [Accepted: 12/06/2021] [Indexed: 10/26/2022] Open
Abstract
Breast cancer is one of the most common types of cancer in female patients with high morbidity and mortality. Multi-drug chemotherapy has significant advantages in the treatment of malignant tumors, especially in reducing drug toxicity, increasing drug sensitivity and reducing drug resistance. The objective of this research is to fabricate lipid nanoemulsions (LNs) for the co-delivery of PTX and docosahexaenoic acid (DHA) with folic acid (FA) decorating (PTX/DHA-FA-LNs), and investigate the anti-tumor activity of the PTX/DHA-FA-LNs against breast cancer both in vitro and in vivo. PTX/DHA-FA-LNs showed a steady release of PTX and DHA from the drug delivery system (DDS) without any burst effect. Furthermore, the PTX/DHA-FA-LNs exhibited a dose-dependent cytotoxicity and a higher rate of apoptosis as compared with the other groups in MCF-7 cells. The cellular uptake study revealed that this LNs were more readily uptaken by MCF-7 cells and M2 macrophages in vitro. Additionally, the targeted effect of PTX/DHA-FA-LNs was aided by FA receptor-mediated endocytosis, and its cytotoxicity was proportional to the cellular uptake efficiency. The anti-tumor efficiency results showed that PTX/DHA-FA-LNs significant inhibited tumor volume growth, prolonged survival time, and reduced toxicity when compared with the other groups. These results indicated that DHA increases the sensitivity of tumor cells and tumor-associated macrophages (ATM2) to PTX, and synergistic effects of folate modification in breast cancer treatment, thus PTX/DHA-FA-LNs may be a promising nanocarrier for breast cancer treatment.
Collapse
Affiliation(s)
- Bo Li
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Tingfei Tan
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Weiwei Chu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Ying Zhang
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuanzi Ye
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shanshan Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| | - Yan Qin
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Jihui Tang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Xi Cao
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, People’s Republic of China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, People’s Republic of China
| |
Collapse
|
31
|
Yang Z, Du Y, Lei L, Xia X, Wang X, Tong F, Li Y, Gao H. Co-delivery of ibrutinib and hydroxychloroquine by albumin nanoparticles for enhanced chemotherapy of glioma. Int J Pharm 2022; 630:122436. [PMID: 36436742 DOI: 10.1016/j.ijpharm.2022.122436] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 10/18/2022] [Accepted: 11/20/2022] [Indexed: 11/25/2022]
Abstract
Ibrutinib (IBR) is an oral covalent inhibitor of Bruton's tyrosine kinase (BTK) that has been approved for the treatment of hematological malignancies. It was reported that IBR exhibited great therapeutic potential for glioma. However, the poor water solubility and high hepatic first-pass effect restrict its anti-glioma application. Meanwhile, IBR induces cytoprotective autophagy through Akt/mTOR signaling pathway, thus leading to a compromised antitumor effect. Herein, we aimed to develop a human serum albumin (HSA) based co-delivery system (IBR&HCQ HSA NPs) encapsulating IBR and hydroxychloroquine (HCQ). The bioavailability of IBR was largely improved, and enhanced sensitivity of glioma to IBR was achieved due to inhibition effect of HCQ on IBR-induced pro-survival autophagy. The physicochemical properties of IBR&HCQ HSA NPs were characterized to optimize the formulation. Biodistribution investigation revealed that HSA NPs (20 mg/kg, i.v.) dramatically increased the accumulation of IBR in glioma, which was 5.59 times higher than that of free IBR (100 mg/kg, i.g.). CCK-8 and apoptosis assays demonstrated that IBR&HCQ HSA NPs showed maximal cytotoxicity to C6 cells. In vivo studies indicated that the survival time was significantly prolonged in IBR&HCQ HSA NPs treated mice compared to those treated with IBR HSA NPs. Taken together, the HSA-based drug delivery system of IBR and HCQ opens a new avenue for efficient treatment of glioma.
Collapse
Affiliation(s)
- Zhihang Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Yufan Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Lei Lei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Xue Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Xiaorong Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Fan Tong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China
| | - Yuan Li
- Gynecology and Obstetrics Department, Peking University Third Hospital, Beijing 100191, PR China.
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, PR China.
| |
Collapse
|
32
|
Guo Z, Sui J, Li Y, Wei Q, Wei C, Xiu L, Zhu R, Sun Y, Hu J, Li JL. GE11 peptide-decorated acidity-responsive micelles for improved drug delivery and enhanced combination therapy of metastatic breast cancer. J Mater Chem B 2022; 10:9266-9279. [PMID: 36342458 DOI: 10.1039/d2tb01816k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nanotechnology-mediated drug delivery systems suffer from insufficient retention in tumor tissues and unreliable drug release at specific target sites. Herein, we developed an epidermal growth factor receptor-targeted multifunctional micellar nanoplatform (GE11-DOX+CEL-M) by encapsulating celecoxib into polymeric micelles based on the conjugate of GE11-poly(ethylene glycol)-b-poly(trimethylene carbonate) with doxorubicin to suppress tumor growth and metastasis. The polymeric micelles maintained stable nanostructures under physiological conditions but quickly disintegrated in a weakly acidic environment, which is conducive to controlled drug release. Importantly, GE11-DOX+CEL-M micelles effectively delivered the drug combination to tumor sites and enhanced tumor cell uptake through GE11-mediated active tumor targeting. Subsequently, GE11-DOX+CEL-M micelles dissociated in response to intracellular slightly acidic microenvironmental stimuli, resulting in rapid release of celecoxib and doxorubicin to synergistically inhibit the proliferation and migration of tumor cells. Systemic administration of GE11-DOX+CEL-M micelles into mice bearing subcutaneous 4T1 tumor models resulted in higher tumor growth suppression and decreased lung metastasis of tumor cells compared with micelles without GE11 decoration or delivering only doxorubicin. Furthermore, the micelles effectively reduced the systemic toxicity of the chemotherapy drugs. This nanotherapeutic system provides a promising strategy for safe and effective cancer therapy.
Collapse
Affiliation(s)
- Zhihao Guo
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China. .,National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, China. .,Center for Molecular Science and Engineering, College of Science, Northeastern University, 3-11 Wenhua Road, Shenyang, 110819, China
| | - Junhui Sui
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, China. .,College of Life Science and Technology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang, 453003, China
| | - Yumei Li
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China.
| | - Qinchuan Wei
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China.
| | - Cailing Wei
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China.
| | - Linyun Xiu
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China.
| | - Ruohua Zhu
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China.
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, China.
| | - Jianshe Hu
- Center for Molecular Science and Engineering, College of Science, Northeastern University, 3-11 Wenhua Road, Shenyang, 110819, China
| | - Ji-Liang Li
- School of Biomedical Engineering, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, 325027, China. .,Wenzhou Institute, University of Chinese Academy of Sciences, 1 Jinlian Road, Wenzhou, 325000, China.
| |
Collapse
|
33
|
Liu W, Cheng M, Yuan F, He J, Feng Y, Jin Y, Feng J, Yang S, Tu L. Enhancing oral bioavailability of andrographolide via sodium dodecyl sulfate and D-α-Tocopherol polyethylene glycol 1000 succinate copolymer modified nanocrystals. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
34
|
Wang S, Song Y, Ma J, Chen X, Guan Y, Peng H, Yan G, Tang R. Dynamic crosslinked polymeric nano-prodrugs for highly selective synergistic chemotherapy. Asian J Pharm Sci 2022; 17:880-891. [PMID: 36600901 PMCID: PMC9800956 DOI: 10.1016/j.ajps.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 09/12/2022] [Accepted: 09/27/2022] [Indexed: 01/07/2023] Open
Abstract
To achieve highly selective synergistic chemotherapy attractive for clinical translation, the precise polymeric nano-prodrugs (PPD-NPs) were successfully constructed via the facile crosslinking reaction between pH-sensitive poly(ortho ester)s and reduction-sensitive small molecule synergistic prodrug (Pt(IV)-1). PPD-NPs endowed the defined structure and high drug loading of cisplatin and demethylcantharidin (DMC). Moreover, PPD-NPs exhibited steady long-term storage and circulation via the crosslinked structure, suitable negative potentials and low critical micelle concentration (CMC), improved selective tumour accumulation and cellular internalization via dynamic size transition and surficial amino protonation at tumoural extracellular pH, promoted efficient disintegration and drug release at tumoural intracellular pH/glutathione, and enhanced cytotoxicity via the synergistic effect between cisplatin and DMC with the feed ratio of 1:2, achieving significant tumour suppression while decreasing the side effects. Thus, the dynamic crosslinked polymeric nano-prodrugs exhibit tremendous potential for clinically targeted synergistic cancer therapy.
Collapse
Affiliation(s)
- Shi Wang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei 230601, China
| | - Yining Song
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu Medical College, Bengbu 233030, China
| | - Jingge Ma
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei 230601, China
| | - Xinyang Chen
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei 230601, China
| | - Yuanhui Guan
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei 230601, China
| | - Hui Peng
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei 230601, China
| | - Guoqing Yan
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei 230601, China,Corresponding authors.
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei 230601, China,Corresponding authors.
| |
Collapse
|
35
|
Alhaj-Suliman SO, Wafa EI, Salem AK. Engineering nanosystems to overcome barriers to cancer diagnosis and treatment. Adv Drug Deliv Rev 2022; 189:114482. [PMID: 35944587 DOI: 10.1016/j.addr.2022.114482] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/30/2022] [Accepted: 08/03/2022] [Indexed: 01/24/2023]
Abstract
Over the past two decades, multidisciplinary investigations into the development of nanoparticles for medical applications have continually increased. However, nanoparticles are still subject to biological barriers and biodistribution challenges, which limit their overall clinical potential. This has motivated the implementation of innovational modifications to a range of nanoparticle formulations designed for cancer imaging and/or cancer treatment to overcome specific barriers and shift the accumulation of payloads toward the diseased tissues. In recent years, novel technological and chemical approaches have been employed to modify or functionalize the surface of nanoparticles or manipulate the characteristics of nanoparticles. Combining these approaches with the identification of critical biomarkers provides new strategies for enhancing nanoparticle specificity for both cancer diagnostic and therapeutic applications. This review discusses the most recent advances in the design and engineering of nanoparticles as well as future directions for developing the next generation of nanomedicines.
Collapse
Affiliation(s)
- Suhaila O Alhaj-Suliman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Emad I Wafa
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States
| | - Aliasger K Salem
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, United States; Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA 52242, United States.
| |
Collapse
|
36
|
Zhang R, You X, Luo M, Zhang X, Fang Y, Huang H, Kang Y, Wu J. Poly(β-cyclodextrin)/platinum prodrug supramolecular nano system for enhanced cancer therapy: Synthesis and in vivo study. Carbohydr Polym 2022; 292:119695. [PMID: 35725183 DOI: 10.1016/j.carbpol.2022.119695] [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: 02/14/2022] [Revised: 05/30/2022] [Accepted: 06/01/2022] [Indexed: 11/02/2022]
Abstract
The use of cisplatin is restricted by systemic toxicity and drug resistance. Supramolecular nano-drug delivery systems involving drugs as building blocks circumvent these limitations promisingly. Herein, we describe a novel supramolecular system [Pt(IV)-SSNPs] based on poly(β-cyclodextrin), which was synthesized for efficient loading of adamantly-functionalized platinum(IV) prodrug [Pt(IV)-ADA2] via the host-guest interaction between β-cyclodextrin and adamantyl. Pt(IV)-ADA2 can be converted to active cisplatin in reducing environment in cancer cells, which further reduces systemic toxicity. The introduction of the adamantane group-tethered mPEG2k endowed the Pt(IV)-SSNPs with a longer blood circulation time. In vitro assays exhibited that the Pt(IV)-SSNPs could be uptaken by CT26 cells, resulting in cell cycle arrest in the G2/M and S phases, together with apoptosis. Furthermore, the Pt(IV)-SSNPs showed effective tumor accumulation, better antitumor effect, and negligible cytotoxicity to major organs. These results indicate that supramolecular nanoparticles are a promising platform for efficient cisplatin delivery and cancer treatment.
Collapse
Affiliation(s)
- Ruhe Zhang
- School of Biomedical Engineering; State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, China
| | - Xinru You
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Moucheng Luo
- School of Biomedical Engineering; State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, China
| | - Xinyu Zhang
- School of Biomedical Engineering; State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, China
| | - Yifen Fang
- Department of Cardiology, The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Yang Kang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| | - Jun Wu
- School of Biomedical Engineering; State Key Laboratory of Oncology in South China, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
37
|
Junnuthula V, Kolimi P, Nyavanandi D, Sampathi S, Vora LK, Dyawanapelly S. Polymeric Micelles for Breast Cancer Therapy: Recent Updates, Clinical Translation and Regulatory Considerations. Pharmaceutics 2022; 14:1860. [PMID: 36145608 PMCID: PMC9501124 DOI: 10.3390/pharmaceutics14091860] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/15/2022] [Accepted: 09/01/2022] [Indexed: 12/13/2022] Open
Abstract
With the growing burden of cancer, parallel advancements in anticancer nanotechnological solutions have been witnessed. Among the different types of cancers, breast cancer accounts for approximately 25% and leads to 15% of deaths. Nanomedicine and its allied fields of material science have revolutionized the science of medicine in the 21st century. Novel treatments have paved the way for improved drug delivery systems that have better efficacy and reduced adverse effects. A variety of nanoformulations using lipids, polymers, inorganic, and peptide-based nanomedicines with various functionalities are being synthesized. Thus, elaborate knowledge of these intelligent nanomedicines for highly promising drug delivery systems is of prime importance. Polymeric micelles (PMs) are generally easy to prepare with good solubilization properties; hence, they appear to be an attractive alternative over the other nanosystems. Although an overall perspective of PM systems has been presented in recent reviews, a brief discussion has been provided on PMs for breast cancer. This review provides a discussion of the state-of-the-art PMs together with the most recent advances in this field. Furthermore, special emphasis is placed on regulatory guidelines, clinical translation potential, and future aspects of the use of PMs in breast cancer treatment. The recent developments in micelle formulations look promising, with regulatory guidelines that are now more clearly defined; hence, we anticipate early clinical translation in the near future.
Collapse
Affiliation(s)
| | - Praveen Kolimi
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA
| | - Dinesh Nyavanandi
- Pharmaceutical Development Services, Thermo Fisher Scientific, Cincinnati, OH 45237, USA
| | - Sunitha Sampathi
- GITAM School of Pharmacy, GITAM Deemed to be University, Hyderabad 502329, India
| | | | - Sathish Dyawanapelly
- Department of Pharmaceutical Science and Technology, Institute of Chemical Technology, Mumbai 400019, India
| |
Collapse
|
38
|
Yang YN, Cheng JJ, He J, Lu WG. Novel Docetaxel-Loaded Micelles Based on all-trans-Retinoic Acid: Preparation and Pharmacokinetic Study in Rats. PHARMACEUTICAL FRONTS 2022. [DOI: 10.1055/s-0042-1757511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
AbstractDocetaxel (DTX) is a poorly soluble drug. The purpose of this study was to explore a DTX-loaded micelle delivery system using N-(all-trans-retinoyl)-L-cysteic acid methyl ester sodium salt (XMeNa) as the carrier materials. In this study, amphiphilic surfactant XMeNa was synthesized. Then, the blood biocompatibility and the value of critical micelle concentration (CMC) were assessed by a hemolysis test and pyrene-based fluorescent probe techniques, respectively. The XM-DTX micelles were prepared using the method of thin-film hydration, and characterized by dynamic light scattering and transmission electron microscopy (TEM). The entrapment efficiency (EE) and drug loading efficiency (DLE) were assessed by the ultrafiltration method. In vitro release and pharmacokinetic behaviors of XM-DTX micelles were performed in rats using Taxotere (a commercialized DTX injection) as a control. Our data confirmed the excellent blood biocompatibility of XMeNa as a carrier. XMeNa can self-assemble into micelles in aqueous media with a very low CMC (6.2 μg/mL). The average size and zeta potential of the XM-DTX micelles were 17.3 ± 0.2 nm, and −41.6 ± 0.3 mV, respectively. EE and DLE reached up to 95.3 ± 0.7% and 22.4 ± 0.2%, respectively, which may account for the high solubility of DTX in normal saline. The micelles were spherical in TEM with good dispersion and no aggregation and adhesion, and exhibited good stability after reconstitution over 8 hours. Results from in vitro release assay suggested a much slower release behavior of XM-DTX micelles in comparison to Taxotere. Additionally, XM-DTX micelles prolonged DTX retention in blood circulation, increased the area under the curve by 2.4-fold, and significantly decreased the clearance of the drug. Given above, the XM-DTX micelles could improve the solubility and the release of DTX. The amphiphilic surfactant XMeNa also exhibited great potential as a vehicle for exploring delivery of poorly water soluble drugs in the future.
Collapse
Affiliation(s)
- Ya-Ni Yang
- National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Jia-Jia Cheng
- National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Jun He
- National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| | - Wei-Gen Lu
- National Pharmaceutical Engineering Research Center, China State Institute of Pharmaceutical Industry, Shanghai, People's Republic of China
| |
Collapse
|
39
|
Shi H, Zhu Y, Xing C, Li S, Bao Z, Lei L, Lin D, Wang Y, Chen H, Xu X. An injectable thermosensitive hydrogel for dual delivery of diclofenac and Avastin® to effectively suppress inflammatory corneal neovascularization. Int J Pharm 2022; 625:122081. [PMID: 35934166 DOI: 10.1016/j.ijpharm.2022.122081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 10/16/2022]
Abstract
Corneal neovascularization (CNV) is a sequela of anterior segment inflammation, which could lead to vision impairment and even blindness. In the present study, the dual delivery of anti-inflammatory agent (i.e., diclofenac; DIC) and anti-VEGF antibody (i.e., Avastin®; Ava) by the thermosensitive hydrogel (Poly(dl-lactide)-poly(ethylene glycol)-poly(dl-lactide); PDLLA-PEG-PDLLA) is expected to effectively inhibit CNV via their synergistic effects. The optimal DIC micelles were formulated and then mixed with Ava and PDLLA-PEG-PDLLA aqueous solution to generate various DIC@Ava-loaded hydrogels. The co-encapsulation of DIC micelles and Ava did not influence the gelling behavior of the system, and the resulting DIC@Ava-loaded hydrogel provided sustained drug release of both DIC and Ava without compromising their pharmacological activity over 19 days. As indicated by in vitro cytotoxicity and in vivo ocular biocompatibility test, the proposed PDLLA-PEG-PDLLA hydrogel caused minimal cytotoxicity against all tested cell lines at a polymeric concentration ranging from 0.05 mg/mL to 0.8 mg/mL and demonstrated good ocular biocompatibility after a single subconjunctival injection. Using the rabbit CNV model, we documented the superior anti-angiogenic effects of the DIC@Ava-loaded hydrogel over Ava alone medication (treatment with Ava solution and Ava-loaded hydrogel) due to synergistic effects of anti-VEGF and anti-inflammatory action. Overall, the proposed DIC@Ava-loaded hydrogel might be a powerful strategy to reduce CNV.
Collapse
|
40
|
Effects of Polymer Molecular Weight on In Vitro and In Vivo Performance of Nanoparticle Drug Carriers for Lymphoma Therapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.07.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
41
|
The in vivo fate of polymeric micelles. Adv Drug Deliv Rev 2022; 188:114463. [PMID: 35905947 DOI: 10.1016/j.addr.2022.114463] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 06/10/2022] [Accepted: 07/15/2022] [Indexed: 12/12/2022]
Abstract
This review aims to provide a systemic analysis of the in vivo, as well as subcellular, fate of polymeric micelles (PMs), starting from the entry of PMs into the body. Few PMs are able to cross the biological barriers intact and reach the circulation. In the blood, PMs demonstrate fairly good stability mainly owing to formation of protein corona despite controversial results reported by different groups. Although the exterior hydrophilic shells render PMs "long-circulating", the biodistribution of PMs into the mononuclear phagocyte systems (MPS) is dominant as compared with non-MPS organs and tissues. Evidence emerges to support that the copolymer poly(ethylene glycol)-poly(lactic acid) (PEG-PLA) is first broken down into pieces of PEG and PLA and then remnants to be eliminated from the body finally. At the cellular level, PMs tend to be internalized via endocytosis due to their particulate nature and disassembled and degraded within the cell. Recent findings on the effect of particle size, surface characteristics and shape are also reviewed. It is envisaged that unraveling the in vivo and subcellular fate sheds light on the performing mechanisms and gears up the clinical translation of PMs.
Collapse
|
42
|
Single subcutaneous injection of the minocycline nanocomposite-loaded thermosensitive hydrogel for the effective attenuation of experimental autoimmune uveitis. Int J Pharm 2022; 622:121836. [PMID: 35597394 DOI: 10.1016/j.ijpharm.2022.121836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/30/2022] [Accepted: 05/14/2022] [Indexed: 11/24/2022]
Abstract
Autoimmune uveitis induces a serious pathological and inflammatory response in the retina/choroid and results in vision impairment and blindness. Here, we report a minocycline (Mino) nanocomposite-loaded hydrogel offering a high drug payload and sustained drug release for the effective control of ocular inflammation via a single subcutaneous injection. In the presence of divalent cations (i.e., Ca2+), Mino was found to co-assemble with a phosphorylated peptide (i.e., NapGFFpY) via electrostatic interaction and consequently generating Mino nanocomposite. The drug entrapment efficiency (EE) of the Mino nanocomposite varied from 29.93±0.76% to 67.90±6.57%, depending on different component concentrations. After incorporation into 30 wt% poly (D,L-lactide)-b-poly (ethylene glycol)-b-poly (D,L-lactide) (PDLLA-PEG-PDLLA) thermosensitive hydrogel, the resulting Mino nanocomposite-loaded hydrogel provided a sustained drug release over 21 days. In the experimental autoimmune uveitis (EAU) rat model, a single subcutaneous injection of the Mino nanocomposite-loaded hydrogel effectively alleviated ocular inflammation in a dose-dependent manner. As indicated by optical coherence tomography (OCT) and electroretinogram (ERG) measurements, the Mino nanocomposite-loaded hydrogel treatment not only remarkably reduced destruction of the retina by EAU, but also greatly rescued retinal functions. Moreover, the proposed Mino nanocomposite-loaded hydrogel exerted its therapeutic effect on EAU primarily through a significant reduction of the influx of leukocytes and Th17 cells as well as suppression of microglia activation and apoptosis in the retina. Overall, the proposed Mino nanocomposite-loaded hydrogel might be a promising strategy for the clinical management of EAU.
Collapse
|
43
|
Lu M, Huang X, Cai X, Sun J, Liu X, Weng L, Zhu L, Luo Q, Chen Z. Hypoxia-Responsive Stereocomplex Polymeric Micelles with Improved Drug Loading Inhibit Breast Cancer Metastasis in an Orthotopic Murine Model. ACS APPLIED MATERIALS & INTERFACES 2022; 14:20551-20565. [PMID: 35476401 DOI: 10.1021/acsami.1c23737] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor metastasis is a leading cause of breast cancer-related death. Taxane-loaded polymeric formulations, such as Genexol PM and Nanoxel M using poly(ethylene glycol)-poly(d,l-lactide) (PEG-PLA) micelles as drug carriers, have been approved for the treatment of metastatic breast cancer. Unfortunately, the physical instability of PEG-PLA micelles, leading to poor drug loading, premature drug leakage, and consequently limited drug delivery to tumors, largely hinders their therapeutic outcome. Inspired by the enantiomeric nature of PLA, this work developed stereocomplex PEG-PLA micelles through stereoselective interactions of enantiomeric PLA, which are further incorporated with a hypoxia-responsive moiety used as a hypoxia-cleavable linker of PEG and PLA, to maximize therapeutic outcomes. The results showed that the obtained micelles had high structural stability, showing improved drug loading for effective drug delivery to tumors as well as other tissues. Especially, they were capable of sensitively responding to the hypoxic tumor environment for drug release, reversing hypoxia-induced drug resistance and hypoxia-promoted cell migration for enhanced bioavailability under hypoxia. In vivo results further showed that the micelles, especially at a high dose, inhibited the growth of the primary tumor and improved tumor pathological conditions, consequently remarkably inhibiting its metastasis to the lungs and liver, while not causing any systemic toxicity. Hypoxia-responsive stereocomplex micelles thus emerge as a reliable drug delivery system to treat breast cancer metastasis.
Collapse
Affiliation(s)
- Min Lu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Xu Huang
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Xiaohui Cai
- Department of Hematology, Nanjing Medical University, Affiliated Changzhou No. 2 People's Hospital, Changzhou 213000, People's Republic of China
| | - Jiajia Sun
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Xuemeng Liu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Lingyan Weng
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Li Zhu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Qianqian Luo
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Zhongping Chen
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| |
Collapse
|
44
|
Liu R, Luo C, Pang Z, Zhang J, Ruan S, Wu M, Wang L, Sun T, Li N, Han L, Shi J, Huang Y, Guo W, Peng S, Zhou W, Gao H. Advances of nanoparticles as drug delivery systems for disease diagnosis and treatment. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.05.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Huang Y, Zou L, Wang J, Jin Q, Ji J. Stimuli-responsive nanoplatforms for antibacterial applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1775. [PMID: 35142071 DOI: 10.1002/wnan.1775] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022]
Abstract
The continuously increasing bacterial resistance has become a big threat to public health worldwide, which makes it urgent to develop innovative antibacterial strategies. Nanotechnology-based drug delivery systems are considered as promising strategies in combating bacterial infections which are expected to improve the therapeutic efficacy and minimize the side effects. Unfortunately, the conventional nanodrug delivery systems always suffer from practical dilemmas, including incomplete and slow drug release, insufficient accumulation in infected sites, and weak biofilm penetration ability. Stimuli-responsive nanoplatforms are hence developed to overcome the disadvantages of conventional nanoparticles. In this review, we provide an extensive review of the recent progress of endogenous and exogenous stimuli-responsive nanoplatforms in the antibacterial area, including planktonic bacteria, intracellular bacteria, and bacterial biofilms. Taking advantage of the specific infected microenvironment (pH, enzyme, redox, and toxin), the mechanisms and strategies of the design of endogenous stimuli-responsive nanoplatforms are discussed, with an emphasis on how to improve the therapeutic efficacy and minimize side effects. How to realize controlled drug delivery using exogenous stimuli-responsive nanoplatforms especially light-responsive nanoparticles for improved antibacterial effects is another topic of this review. We especially highlight photothermal-triggered drug delivery systems by the combination of photothermal agents and thermo-responsive materials. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Lingyun Zou
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Jing Wang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
46
|
Huang Y, Xu Z, Wei Y, Han S, Cai X, Chen D. Albumin-Embellished Arsenic Trioxide-Loaded Polymeric Nanoparticles Enhance Tumor Accumulation and Anticancer Efficacy via Transcytosis for Hepatocellular Carcinoma Therapy. AAPS PharmSciTech 2022; 23:111. [PMID: 35411416 DOI: 10.1208/s12249-022-02254-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022] Open
Abstract
Arsenic trioxide (ATO) has efficient anticancer effect on hepatocellular carcinoma (HCC) in clinical trials, but its off-target distribution and side effects have limited its use. Here, we demonstrate an albumin-embellished ATO-loaded polyethylene glycol-polycaprolactone-polyethyleneimine (PEG-PCL-PEI) nanoparticle (AATONP) to enhance the tumor distribution and intratumor drug release of ATO for HCC therapy. AATONP is prepared by surface embellishment with albumin on the cationic ATO-loaded PEG-PCL-PEI nanoparticles (CATONP). Albumin embellishment can reduce the cationic material's hemolytic toxicity in blood cells while maintaining the rapid internalization and lysosome escape abilities of the positively charged CATONP. AATONP provides sustained and low pH-responsive drug release, facilitating the targeted drug release in the intratumor acidic microenvironment. Moreover, AATONP can significantly improve the circulation time and tumor distribution of ATO via albumin-mediated transcytosis in HCC tumor-bearing mice. Compared with free ATO and the clinically used nanomedicine Genexol/PM, AATONP shows potent antitumor activity against a human HCC xenograft mouse model, leading to a higher tumor inhibition rate of 89.4% in HCC therapy. In conclusion, this work presents an efficient strategy to achieve tumor accumulation and the intratumor drug release of ATO for HCC therapy. An albumin-embellished arsenic trioxide (ATO)-loaded polyethylene glycol-polycaprolactone-polyethyleneimine nanoparticle (AATONP) is designed to enhance tumor distribution and intratumor drug release of ATO for hepatocellular carcinoma therapy. AATONP can achieve enhanced tumor distribution via albumin-mediated transcytosis and exhibit intratumor drug release of ATO via tumor acidic microenvironment-response, leading to potent antitumor activity.
Collapse
|
47
|
Suzuki M, Takebe G, Takagi T, Tsukada H. Characterization of Novel Paclitaxel Nanoparticles Prepared by Laser Irradiation. Chem Pharm Bull (Tokyo) 2022; 70:269-276. [DOI: 10.1248/cpb.c21-00994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Masumi Suzuki
- Central Research Laboratory, Hamamatsu Photonics K.K
| | - Gen Takebe
- Central Research Laboratory, Hamamatsu Photonics K.K
| | - Tokio Takagi
- Central Research Laboratory, Hamamatsu Photonics K.K
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics K.K
| |
Collapse
|
48
|
Coassembling functionalized glycopolypeptides to prepare pH-responsive self-indicating nanocomplexes to manipulate self-assembly/drug delivery and visualize intracellular drug release. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2022; 134:112711. [DOI: 10.1016/j.msec.2022.112711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/24/2022] [Accepted: 02/08/2022] [Indexed: 11/21/2022]
|
49
|
Song Y, Huang Y, Zhou F, Ding J, Zhou W. Macrophage-targeted nanomedicine for chronic diseases immunotherapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2021.08.090] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
50
|
Guo C, Zhang Y, Yuan H, Zhang Y, Yin T, He H, Gou J, Tang X. Improved Core Viscosity Achieved by PDLLA 10kCo-Incorporation Promoted Drug Loading and Stability of mPEG 2k-b-PDLLA 2.4k Micelles. Pharm Res 2022; 39:369-379. [PMID: 35118566 DOI: 10.1007/s11095-022-03174-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/20/2022] [Indexed: 10/19/2022]
Abstract
PURPOSE This study aims to investigate the effect of poly(D, L-lactic acid)10K (PDLLA10K) incorporation on the drug loading and stability of poly(ethylene glycol)2K-block-poly(D, L-lactide)2.4K (mPEG2k-b-PDLLA2.4k) micelles. In addition, a suitable lyophilization protector was screened for this micelle to obtain favorable lyophilized products. METHODS The incorporation ratios of PDLLA10k were screened based on the particle size and drug loading. The dynamic stability, core viscosity, drug release, stability in albumin, and in vivo pharmacokinetic characteristics of PDLLA10k incorporated micelles were compared with the original micelles. In addition, the particle size variation was used as an indicator to screen the most suitable lyophilization protectant for the micelles. DSC, FTIR, XRD were used to illustrate the mechanism of the lyophilized protectants. RESULTS After the incorporation of 5 wt% PDLLA10K, the maximum loading of mPEG2k-b-PDLLA2.4k micelles for TM-2 was increased from 26 wt% to 32 wt%, and the in vivo half-life was increased by 2.25-fold. Various stability of micelles was improved. Also, the micelles with hydroxypropyl-β-cyclodextrin (HP-β-CD) as lyophilization protectants had minimal variation in particle size. CONCLUSIONS PDLLA10k incorporation can be employed as a strategy to increase the stability of mPEG2k-b-PDLLA2.4k micelles, which can be attributed to the viscosity building effect. HP-β-CD can be used as an effective lyophilization protectant since mPEG and HP-β-CD form the pseudopolyrotaxanesque inclusion complexes.
Collapse
Affiliation(s)
- Chen Guo
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Ying Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Haoyang Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China.
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, 110016, Liaoning, People's Republic of China.
| |
Collapse
|