1
|
Zhang J, Pan T, Lee J, Goldberg S, King SA, Tang E, Hu Y, Chen L, Hoover A, Zhu L, Eng OS, Dekel B, Huang J, Wu X. Enabling tumor-specific drug delivery by targeting the Warburg effect of cancer. Cell Rep Med 2025; 6:101920. [PMID: 39809265 PMCID: PMC11866520 DOI: 10.1016/j.xcrm.2024.101920] [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: 08/31/2023] [Revised: 02/14/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025]
Abstract
Metabolic reprogramming of tumor cells is an emerging hallmark of cancer. Among all the changes in cancer metabolism, increased glucose uptake and the accumulation of lactate under normoxic conditions (the "Warburg effect") is a common feature of cancer cells. In this study, we develop a lactate-responsive drug delivery platform by targeting the Warburg effect. We design and test a gold/mesoporous silica Janus nanoparticle system as a gated drug carrier, in which the gold particles are functionalized with lactate oxidase and the silica particles are capped with α-cyclodextrin through surface arylboronate modification. In the presence of lactate, the lactate oxidase generates hydrogen peroxide, which induces the self-immolation reaction of arylboronate, leading to uncapping and drug release. Our results demonstrate greatly improved drug delivery specificity and therapeutic efficacy with this platform for the treatment of different cancers. Our findings present an effective approach for drug delivery by metabolic targeting of tumors.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, USA; Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Tony Pan
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Jimmy Lee
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Sanja Goldberg
- Pediatric Stem Cell Research Institute, Safra Children's Hospital, Sheba Medical Center, Tel Aviv, Israel
| | - Sarah Ann King
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Erting Tang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Yifei Hu
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Lifeng Chen
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Alex Hoover
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Linyong Zhu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai 200237, China
| | - Oliver S Eng
- Department of Surgery, University of California, Irvine, Orange, CA 92868, USA
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Safra Children's Hospital, Sheba Medical Center, Tel Aviv, Israel; Division of Pediatric Nephrology and Pediatric Stem Cell Research Institute, Safra Children's Hospital, Sheba Medical Center, Tel Hasomer, Sago Center for Regenerative Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jun Huang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA.
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
2
|
Preethy H A, Rajendran K, Sukumar AJ, Krishnan UM. Emerging paradigms in Alzheimer's therapy. Eur J Pharmacol 2024; 981:176872. [PMID: 39117266 DOI: 10.1016/j.ejphar.2024.176872] [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: 03/08/2024] [Revised: 07/13/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Alzheimer's disease is a neurodegenerative disorder that affects elderly, and its incidence is continuously increasing across the globe. Unfortunately, despite decades of research, a complete cure for Alzheimer's disease continues to elude us. The current medications are mainly symptomatic and slow the disease progression but do not result in reversal of all disease pathologies. The growing body of knowledge on the factors responsible for the onset and progression of the disease has resulted in the identification of new targets that could be targeted for treatment of Alzheimer's disease. This has opened new vistas for treatment of Alzheimer's disease that have moved away from chemotherapeutic agents modulating a single target to biologics and combinations that acted on multiple targets thereby offering better therapeutic outcomes. This review discusses the emerging directions in therapeutic interventions against Alzheimer's disease highlighting their merits that promise to change the treatment paradigm and challenges that limit their clinical translation.
Collapse
Affiliation(s)
- Agnes Preethy H
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Kayalvizhi Rajendran
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Anitha Josephine Sukumar
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India
| | - Uma Maheswari Krishnan
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, India; School of Arts, Sciences, Humanities & Education, SASTRA Deemed University, Thanjavur, India.
| |
Collapse
|
3
|
Song Z, Chen P, Teng L, Wang W, Zhu W. Copper Nanodrugs with Controlled Morphologies through Aqueous Atom Transfer Radical Polymerization. Biomacromolecules 2024; 25:4545-4556. [PMID: 38902858 DOI: 10.1021/acs.biomac.4c00552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Copper (Cu) nanodrugs can be facilely prepared through atom transfer radical polymerization (ATRP) in an aqueous medium. However, it is difficult to control the morphology of Cu nanodrugs and thereby optimize their anticancer activity. In this work, aqueous ATRP was combined with polymerization-induced self-assembly (PISA) to prepare Cu nanodrugs with various morphologies. We mapped the relationship between polymerization condition and product morphology in which each morphology shows a wide preparation window. Decreasing the reaction temperature and feeding more Cu catalysts can improve the mobility of chains, facilitating the morphology evolution from sphere to other high-order morphologies. The resultant Cu nanodrugs with high monomer conversion and high Cu loading efficiency could be easily taken by cancer cells, showing excellent anticancer efficacy in vitro. This work proposed a potential strategy to prepare Cu nanodrugs with a specific morphology in batches, providing the method to optimize the anticancer efficacy through morphology control.
Collapse
Affiliation(s)
- Ziyan Song
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Peng Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Weibin Wang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Weipu Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030000, China
- Key Laboratory of Adsorption and Separation Materials & Technologies of Zhejiang Province, Hangzhou 310058, China
| |
Collapse
|
4
|
Mukherjee D, Raikwar S. Recent Update on Nanocarrier(s) as the Targeted Therapy for Breast Cancer. AAPS PharmSciTech 2024; 25:153. [PMID: 38961013 DOI: 10.1208/s12249-024-02867-x] [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: 11/28/2023] [Accepted: 06/11/2024] [Indexed: 07/05/2024] Open
Abstract
Despite ongoing advances in cancer therapy, the results for the treatment of breast cancer are not satisfactory. The advent of nanotechnology promises to be an essential tool to improve drug delivery effectiveness in cancer therapy. Nanotechnology provides an opportunity to enhance the treatment modality by preventing degradation, improving tumour targeting, and controlling drug release. Recent advances have revealed several strategies to prevent cancer metastasis using nano-drug delivery systems (NDDS). These strategies include the design of appropriate nanocarriers loaded with anti-cancer drugs that target the optimization of physicochemical properties, modulate the tumour microenvironment, and target biomimetic techniques. Nanocarriers have emerged as a preferential approach in the chemotropic treatment for breast cancer due to their pivotal role in safeguarding the therapeutic agents against degradation. They facilitate efficient drug concentration in targeted cells, surmount the resistance of drugs, and possess a small size. Nevertheless, these nanocarrier(s) have some limitations, such as less permeability across the barrier and low bioavailability of loaded drugs. To overcome these challenges, integrating external stimuli has been employed, encompassing infrared light, thermal stimulation, microwaves, and X-rays. Among these stimuli, ultrasound-triggered nanocarriers have gained significant attention due to their cost-effectiveness, non-invasive nature, specificity, ability to penetrate tissues, and capacity to deliver elevated drug concentrations to intended targets. This article comprehensively reviews recent advancements in different nanocarriers for breast cancer chemotherapy. It also delves into the associated hurdles and offers valuable insights into the prospective directions for this innovative field.
Collapse
Affiliation(s)
- Debanjan Mukherjee
- Department of Quality Assurance, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Sarjana Raikwar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
5
|
Quadrado RFN, Silvestri S, de Souza JF, Iglesias BA, Fajardo AR. Advances in porphyrins and chlorins associated with polysaccharides and polysaccharides-based materials for biomedical and pharmaceutical applications. Carbohydr Polym 2024; 334:122017. [PMID: 38553216 DOI: 10.1016/j.carbpol.2024.122017] [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: 01/11/2024] [Revised: 02/26/2024] [Accepted: 03/01/2024] [Indexed: 04/02/2024]
Abstract
Over the last decade, the convergence of advanced materials and innovative applications has fostered notable scientific progress within the biomedical and pharmaceutical fields. Porphyrins and their derivatives, distinguished by an extended conjugated π-electron system, have a relevant role in propelling these advancements, especially in drug delivery systems, photodynamic therapy, wound healing, and (bio)sensing. However, despite their promise, the practical clinical application of these macrocycles is hindered by their inherent challenges of low solubility and instability under physiological conditions. To address this limitation, researchers have exploited the synergistic association of porphyrins and chlorins with polysaccharides by engineering conjugated systems and composite/hybrid materials. This review compiles the principal advances in this growing research field, elucidating fundamental principles and critically examining the applications of such materials within biomedical and pharmaceutical contexts. Additionally, the review addresses the eventual challenges and outlines future perspectives for this poignant research field. It is expected that this review will serve as a comprehensive guide for students and researchers dedicated to exploring state-of-the-art materials for contemporary medicine and pharmaceutical applications.
Collapse
Affiliation(s)
- Rafael F N Quadrado
- Laboratório de Tecnologia e Desenvolvimento de Compósitos e Materiais Poliméricos (LaCoPol), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão s/n, 96010-900 Pelotas, RS, Brazil
| | - Siara Silvestri
- Laboratório de Tecnologia e Desenvolvimento de Compósitos e Materiais Poliméricos (LaCoPol), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão s/n, 96010-900 Pelotas, RS, Brazil; Laboratório de Engenharia de Meio Ambiente (LEMA), Universidade Federal de Santa Maria (UFSM), Campus Camobi, 97105-900 Santa Maria, RS, Brazil
| | - Jaqueline F de Souza
- Laboratório de Bioinorgânica e Materiais Porfirínicos, Universidade Federal de Santa Maria (UFSM), Campus Camobi, 97105-900, Santa Maria, RS, Brazil
| | - Bernardo A Iglesias
- Laboratório de Bioinorgânica e Materiais Porfirínicos, Universidade Federal de Santa Maria (UFSM), Campus Camobi, 97105-900, Santa Maria, RS, Brazil.
| | - André R Fajardo
- Laboratório de Tecnologia e Desenvolvimento de Compósitos e Materiais Poliméricos (LaCoPol), Universidade Federal de Pelotas (UFPel), Campus Capão do Leão s/n, 96010-900 Pelotas, RS, Brazil.
| |
Collapse
|
6
|
Huang S, Xu Z, Zhi W, Li Y, Hu Y, Zhao F, Zhu X, Miao M, Jia Y. pH/GSH dual-responsive nanoparticle for auto-amplified tumor therapy of breast cancer. J Nanobiotechnology 2024; 22:324. [PMID: 38858692 PMCID: PMC11163783 DOI: 10.1186/s12951-024-02588-0] [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: 02/06/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
Breast cancer remains a malignancy that poses a serious threat to human health worldwide. Chemotherapy is one of the most widely effective cancer treatments in clinical practice, but it has some drawbacks such as poor targeting, high toxicity, numerous side effects, and susceptibility to drug resistance. For auto-amplified tumor therapy, a nanoparticle designated GDTF is prepared by wrapping gambogic acid (GA)-loaded dendritic porous silica nanoparticles (DPSNs) with a tannic acid (TA)-Fe(III) coating layer. GDTF possesses the properties of near-infrared (NIR)-enhanced and pH/glutathione (GSH) dual-responsive drug release, photothermal conversion, GSH depletion and hydroxyl radical (·OH) production. When GDTF is exposed to NIR laser irradiation, it can effectively inhibit cell proliferation and tumor growth both in vitro and in vivo with limited toxicity. This may be due to the synergistic effect of enhanced tumor accumulation, and elevated reactive oxygen species (ROS) production, GSH depletion, and TrxR activity reduction. This study highlights the enormous potential of auto-amplified tumor therapy.
Collapse
Affiliation(s)
- Shengnan Huang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province, 450001, P.R. China.
| | - Zhiling Xu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China
| | - Weiwei Zhi
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China
| | - Yijing Li
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China
| | - Yurong Hu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province, 450001, P.R. China
| | - Fengqin Zhao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan Province, 450001, P.R. China
| | - Xiali Zhu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
| | - Yongyan Jia
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, Henan Province, 450046, P. R. China.
| |
Collapse
|
7
|
Naser IH, Zaid M, Ali E, Jabar HI, Mustafa AN, Alubiady MHS, Ramadan MF, Muzammil K, Khalaf RM, Jalal SS, Alawadi AH, Alsalamy A. Unveiling innovative therapeutic strategies and future trajectories on stimuli-responsive drug delivery systems for targeted treatment of breast carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3747-3770. [PMID: 38095649 DOI: 10.1007/s00210-023-02885-9] [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: 09/10/2023] [Accepted: 12/02/2023] [Indexed: 05/23/2024]
Abstract
This comprehensive review delineates the latest advancements in stimuli-responsive drug delivery systems engineered for the targeted treatment of breast carcinoma. The manuscript commences by introducing mammary carcinoma and the current therapeutic methodologies, underscoring the urgency for innovative therapeutic strategies. Subsequently, it elucidates the logic behind the employment of stimuli-responsive drug delivery systems, which promise targeted drug administration and the minimization of adverse reactions. The review proffers an in-depth analysis of diverse types of stimuli-responsive systems, including thermoresponsive, pH-responsive, and enzyme-responsive nanocarriers. The paramount importance of material choice, biocompatibility, and drug loading strategies in the design of these systems is accentuated. The review explores characterization methodologies for stimuli-responsive nanocarriers and probes preclinical evaluations of their efficacy, toxicity, pharmacokinetics, and biodistribution in mammary carcinoma models. Clinical applications of stimuli-responsive systems, ongoing clinical trials, the potential of combination therapies, and the utility of multifunctional nanocarriers for the co-delivery of assorted drugs and therapies are also discussed. The manuscript addresses the persistent challenge of drug resistance in mammary carcinoma and the potential of stimuli-responsive systems in surmounting it. Regulatory and safety considerations, including FDA guidelines and biocompatibility assessments, are outlined. The review concludes by spotlighting future trajectories and emergent technologies in stimuli-responsive drug delivery, focusing on pioneering approaches, advancements in nanotechnology, and personalized medicine considerations. This review aims to serve as a valuable compendium for researchers and clinicians interested in the development of efficacious and safe stimuli-responsive drug delivery systems for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Muhaned Zaid
- Department of Pharmacy, Al-Manara College for Medical Sciences, Maysan, Amarah, Iraq
| | - Eyhab Ali
- Al-Zahraa University for Women, Karbala, Iraq
| | - Hayder Imad Jabar
- Department of Pharmaceutics, College of Pharmacy, University of Al-Ameed, Karbala, Iraq
| | | | | | | | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University, Abha, Saudi Arabia
| | | | - Sarah Salah Jalal
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Ahmed Hussien Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq
- College of Technical Engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq.
| |
Collapse
|
8
|
Chen P, Song Z, Yao X, Wang W, Teng L, Matyjaszewski K, Zhu W. Copper Nanodrugs by Atom Transfer Radical Polymerization. Angew Chem Int Ed Engl 2024; 63:e202402747. [PMID: 38488767 DOI: 10.1002/anie.202402747] [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: 02/07/2024] [Indexed: 04/09/2024]
Abstract
In this study, some copper catalysts used for atom transfer radical polymerization (ATRP) were explored as efficient anti-tumor agents. The aqueous solution of copper-containing nanoparticles with uniform spheric morphology was in situ prepared through a copper-catalyzed activator generated by electron transfer (AGET) ATRP in water. Nanoparticles were then directly injected into tumor-bearing mice for antitumor chemotherapy. The copper nanodrugs had prolonged blood circulation time and enhanced accumulation at tumor sites, thus showing potent antitumor activity. This work provides a novel strategy for precise and large-scale preparation of copper nanodrugs with high antitumor activity.
Collapse
Affiliation(s)
- Peng Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ziyan Song
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xuxia Yao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Weibin Wang
- The First Affiliated Hospital, Department of Surgical Oncology, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lisong Teng
- The First Affiliated Hospital, Department of Surgical Oncology, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Krzysztof Matyjaszewski
- Department of Chemistry, Carnegie Mellon University, Pittsburgh, Pennsylvania, 15213, United States
| | - Weipu Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan, 030000, China
- Key Laboratory of Adsorption and Separation Materials & Technologies of Zhejiang Province, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
9
|
Mim JJ, Hasan M, Chowdhury MS, Ghosh J, Mobarak MH, Khanom F, Hossain N. A comprehensive review on the biomedical frontiers of nanowire applications. Heliyon 2024; 10:e29244. [PMID: 38628721 PMCID: PMC11016983 DOI: 10.1016/j.heliyon.2024.e29244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/19/2024] Open
Abstract
This comprehensive review examines the immense capacity of nanowires, nanostructures characterized by unbounded dimensions, to profoundly transform the field of biomedicine. Nanowires, which are created by combining several materials using techniques such as electrospinning and vapor deposition, possess distinct mechanical, optical, and electrical properties. As a result, they are well-suited for use in nanoscale electronic devices, drug delivery systems, chemical sensors, and other applications. The utilization of techniques such as the vapor-liquid-solid (VLS) approach and template-assisted approaches enables the achievement of precision in synthesis. This precision allows for the customization of characteristics, which in turn enables the capability of intracellular sensing and accurate drug administration. Nanowires exhibit potential in biomedical imaging, neural interfacing, and tissue engineering, despite obstacles related to biocompatibility and scalable manufacturing. They possess multifunctional capabilities that have the potential to greatly influence the intersection of nanotechnology and healthcare. Surmounting present obstacles has the potential to unleash the complete capabilities of nanowires, leading to significant improvements in diagnostics, biosensing, regenerative medicine, and next-generation point-of-care medicines.
Collapse
Affiliation(s)
- Juhi Jannat Mim
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Mehedi Hasan
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Md Shakil Chowdhury
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Jubaraz Ghosh
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Md Hosne Mobarak
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Fahmida Khanom
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| | - Nayem Hossain
- Department of Mechanical Engineering, IUBAT-International University of Business Agriculture and Technology, Bangladesh
| |
Collapse
|
10
|
Qi QR, Tian H, Yue BS, Zhai BT, Zhao F. Research Progress of SN38 Drug Delivery System in Cancer Treatment. Int J Nanomedicine 2024; 19:945-964. [PMID: 38293612 PMCID: PMC10826519 DOI: 10.2147/ijn.s435407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/22/2023] [Indexed: 02/01/2024] Open
Abstract
The active metabolite of irinotecan (CPT-11), 7-ethyl-10-hydroxycamptothecin (SN38), is 100-1000 times more active than CPT-11 and has shown inhibitory effects on a range of cancer cells, including those from the rectal, small cell lung, breast, esophageal, uterine, and ovarian malignancies. Despite SN38's potent anticancer properties, its hydrophobicity and pH instability have caused substantial side effects and anticancer activity loss, which make it difficult to use in clinical settings. To solve the above problems, the construction of SN38-based drug delivery systems is one of the most feasible methods to improve drug solubility, enhance drug stability, increase drug targeting ability, improve drug bioavailability, enhance therapeutic efficacy and reduce adverse drug reactions. Therefore, based on the targeting mechanism of drug delivery systems, this paper reviews SN38 drug delivery systems, including polymeric micelles, liposomal nanoparticles, polymeric nanoparticles, protein nanoparticles, conjugated drug delivery systems targeted by aptamers and ligands, antibody-drug couplings, magnetic targeting, photosensitive targeting, redox-sensitive and multi-stimulus-responsive drug delivery systems, and co-loaded drug delivery systems. The focus of this review is on nanocarrier-based SN38 drug delivery systems. We hope to provide a reference for the clinical translation and application of novel SN38 medications.
Collapse
Affiliation(s)
- Qing-rui Qi
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Huan Tian
- Xi’an Hospital of Traditional Chinese Medicine, Xi’an, 710021, People’s Republic of China
| | - Bao-sen Yue
- Xi’an Hospital of Traditional Chinese Medicine, Xi’an, 710021, People’s Republic of China
| | - Bing-tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Feng Zhao
- Xi’an Hospital of Traditional Chinese Medicine, Xi’an, 710021, People’s Republic of China
| |
Collapse
|
11
|
Liao W, Xiao S, Yang J, Shi X, Zheng Y. Multifunctional nanogel based on carboxymethyl cellulose interfering with cellular redox homeostasis enhances phycocyanobilin photodynamic therapy. Carbohydr Polym 2024; 323:121416. [PMID: 37940295 DOI: 10.1016/j.carbpol.2023.121416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/28/2023] [Accepted: 09/17/2023] [Indexed: 11/10/2023]
Abstract
The redox homeostasis defense mechanism of tumor cells is one of the prime reasons for the unsatisfactory effect of photodynamic therapy (PDT). So far, little attention has been paid to this obstacle. In this work, we reported a synthesizing simple yet versatile nanogel (BCPS), synthesized by cystamine dihydrochloride functionalized sodium carboxymethylcellulose (CMC-SS), bovine serum albumin, and Phycocyanobilin self-assembly. The BCPS reduced the levels of glutathione molecules by reacting with glutathione, thereby interfering with intracellular redox homeostasis and enhancing the sensitivity of tumor cells to PDT. The BCPS was shown to possess excellent serum stability, high blood compatibility, low toxic side effects, and higher reactive oxygen species (ROS) utilization. After irradiation, the BCPS could significantly increase intracellular ROS level by approximately 1.6-fold and decrease the IC50 to HeLa cells by approximately 1.5-fold, compared to the pre-functional drugs BCP. This proposed strategy, based on increasing the utilization rate of ROS in tumor cells is promising for application potentials in tumor therapy.
Collapse
Affiliation(s)
- Wenqiang Liao
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Siqi Xiao
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China
| | - Yunquan Zheng
- College of Chemistry, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China; International Joint Laboratory of Intelligent Health Care, Fuzhou University, No. 2 Xueyuan Road, Fuzhou 350108, China.
| |
Collapse
|
12
|
Sonam Dongsar T, Tsering Dongsar T, Molugulu N, Annadurai S, Wahab S, Gupta N, Kesharwani P. Targeted therapy of breast tumor by PLGA-based nanostructures: The versatile function in doxorubicin delivery. ENVIRONMENTAL RESEARCH 2023; 233:116455. [PMID: 37356522 DOI: 10.1016/j.envres.2023.116455] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/15/2023] [Accepted: 06/17/2023] [Indexed: 06/27/2023]
Abstract
Breast carcinoma is a molecularly diverse illness, and it is among the most prominent and often reported malignancies in female across the globe. Surgical intervention, chemotherapy, immunotherapy, gene therapy, and endocrine treatment are among the currently viable treatment options for the carcinoma of breast. Chemotherapy is among the most prevalent cancer management strategy. Doxorubicin (DOX) widely employed as a cytostatic medication for the treatment of a variety of malignancies. Despite its widespread acceptance and excellent efficacy against an extensive line up of neoplasia, it has a variety of shortcomings that limit its therapeutic potential in the previously mentioned indications. Employment of nanoparticulate systems has come up as a unique chemo medication delivery strategy and are being considerably explored for the amelioration of breast carcinoma. Polylactic-co-glycolic acid (PLGA)-based nano systems are being utilized in a number of areas within the medical research and medication delivery constitutes one of the primary functions for PLGA given their inherent physiochemical attributes, including their aqueous solubility, biocompatibility, biodegradability, versatility in formulation, and limited toxicity. Herein along with the different application of PLGA-based nano formulations in cancer therapy, the present review intends to describe the various research investigations that have been conducted to enumerate the effectiveness of DOX-encapsulated PLGA nanoparticles (DOX-PLGA NPs) as a feasible treatment option for breast cancer.
Collapse
Affiliation(s)
- Tenzin Sonam Dongsar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Tenzin Tsering Dongsar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nagashekhara Molugulu
- School of Pharmacy, Monash University, Bandar Sunway, Jalan Lagoon Selatan, 47500, Malaysia
| | - Sivakumar Annadurai
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Neelima Gupta
- Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India; Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
13
|
Fan H, Guo Z. Tumor microenvironment-responsive manganese-based nanomaterials for cancer treatment. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
14
|
Ma L, Niu M, Ji Y, Liu L, Gu X, Luo J, Wei G, Yan M. Development of KLA-RGD integrated lipopeptide with the effect of penetrating membrane which target the α vβ 3 receptor and the application of combined antitumor. Colloids Surf B Biointerfaces 2023; 223:113186. [PMID: 36746066 DOI: 10.1016/j.colsurfb.2023.113186] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/23/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023]
Abstract
Herein, an amphiphilic cationic anticancer lipopeptide P17 with α-helical structure was synthesized based on the integration of KLA and RGD peptide which could bind with the receptor of integrin αvβ3. P17 could self assemble into stable spherical aggregates in aqueous solution, and which could encapsulate the anticancer drugs (Such as Dox) to form P17 @ Anticancer drug nanomedicine (P17 @ Dox nanomedicine) which could play the combined therapy of P17 and anticancer drugs (Dox). The encapsulation efficiency of P17 aggregates to Dox was 80.4 ± 3.2 %, and the release behavior of P17 @ Dox nanomedicine in vitro had the characteristics of slow-release and pH responsiveness. The experiments in vitro showed that P17 lipopeptide had low cytotoxicity, high serum stability, low hemolysis and strong penetrating membrane ability. The release of Dox from P17 @ Dox in cells was time-dependment, and the P17 @ Dox nanomedicine had a good anticancer effect. The experiments in vivo showed that P17 and P17 @ Dox nanomedicine both had low hemolysis, and P17 @ Dox nanomedicine could effectively inhibit tumor growth and significantly reduce the toxic and side effects of Dox. Molecular docking experiments showed that P17 could effectively interact with the receptor of integrin αvβ3. In conclusion, P17 lipopeptide could be used as an excellent drug carrier and play the combined anticancer effect of P17 and anticancer drugs.
Collapse
Affiliation(s)
- Linhao Ma
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003,China
| | - Mingcong Niu
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003,China
| | - Yiping Ji
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003,China
| | - Lu Liu
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003,China
| | - XiuLian Gu
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003,China
| | - Junlin Luo
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003,China
| | - Guangcheng Wei
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003,China.
| | - Miaomiao Yan
- Department of Pharmacy Science, Binzhou Medical University, Yantai 264003,China.
| |
Collapse
|
15
|
Karaz S, Senses E. Liposomes Under Shear: Structure, Dynamics, and Drug Delivery Applications. ADVANCED NANOBIOMED RESEARCH 2023. [DOI: 10.1002/anbr.202200101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Selcan Karaz
- Department of Chemical and Biological Engineering Koç University Istanbul 34450 Turkey
| | - Erkan Senses
- Department of Chemical and Biological Engineering Koç University Istanbul 34450 Turkey
| |
Collapse
|
16
|
Li C, Deng Z, Gillies ER. Designing polymers with stimuli-responsive degradation for biomedical applications. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
17
|
Rodrigues Arruda B, Mendes MGA, Freitas PGCD, Reis AVF, Lima T, Crisóstomo LCCF, Nogueira KAB, Pessoa C, Petrilli R, Eloy JO. Nanocarriers for delivery of taxanes: A review on physicochemical and biological aspects. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
18
|
Nanogels: Update on the methods of synthesis and applications for cardiovascular and neurological complications. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
19
|
Zhang C, Kang T, Wang X, Song J, Zhang J, Li G. Stimuli-responsive platinum and ruthenium complexes for lung cancer therapy. Front Pharmacol 2022; 13:1035217. [PMID: 36324675 PMCID: PMC9618881 DOI: 10.3389/fphar.2022.1035217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/04/2022] [Indexed: 11/13/2022] Open
Abstract
Lung cancer is the most common cause of cancer-related deaths worldwide. More efficient treatments are desperately needed. For decades, the success of platinum-based anticancer drugs has promoted the exploration of metal-based agents. Four ruthenium-based complexes have also entered clinical trials as candidates of anticancer metallodrugs. However, systemic toxicity, severe side effects and drug-resistance impeded their applications and efficacy. Stimuli-responsiveness of Pt- and Ru-based complexes provide a great chance to weaken the side effects and strengthen the clinical efficacy in drug design. This review provides an overview on the stimuli-responsive Pt- and Ru-based metallic anticancer drugs for lung cancer. They are categorized as endo-stimuli-responsive, exo-stimuli-responsive, and dual-stimuli-responsive prodrugs based on the nature of stimuli. We describe various representative examples of structure, response mechanism, and potential medical applications in lung cancer. In the end, we discuss the future opportunities and challenges in this field.
Collapse
Affiliation(s)
- Cheng Zhang
- The Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Tong Kang
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xinyi Wang
- The Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jiaqi Song
- Department of Biophysics, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jia Zhang
- The Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Jia Zhang, ; Guanying Li,
| | - Guanying Li
- Department of Biophysics, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- *Correspondence: Jia Zhang, ; Guanying Li,
| |
Collapse
|
20
|
Liu N, Wu L, Zuo W, Lin Q, Liu J, Jin Q, Xiao Z, Chen L, Zhao Y, Zhou J, Zhu X. pH/Thermal-Sensitive Nanoplatform Capable of On-Demand Specific Release to Potentiate Drug Delivery and Combinational Hyperthermia/Chemo/Chemodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:29668-29678. [PMID: 35749592 DOI: 10.1021/acsami.2c09685] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Therapeutic platforms with spatiotemporal control were recently of considerable interest. However, the site-specific regulation of chemotherapeutics release remains an enormous challenge. Herein, a versatile nanoplatform capable of tumor-specific delivery and controlled drug release, coined as PDDFe, was constructed for elevating cancer theranostics. Iron-oxide nanoparticles (IONPs) and doxorubicin (Dox) were encapsulated in pH/thermal-sensitive micelles composed of poly(ethylene)glycol-poly(β-amino esters) and dipalmitoyl phosphatidylcholine to obtain tumor-targeted dual-responsive nanoplatforms. With remarkable magnetic targeting effects, PDDFe specifically accumulated at tumor locations. After internalization by cancer cells, the acidic environment and localized heat generated by hyperthermia therapy would spur PDDFe to become loose and collapse to liberate its payload. In addition to boosting the release, the increased temperature also resulted in direct tumor damage. Meanwhile, the released Dox and IONPs, respectively, stimulated chemotherapy and chemodynamic therapy to jointly destroy cancer, thus leading to a pronounced therapeutic effect. In vivo magnetic resonance/fluorescence/photoacoustic imaging experiments validated that the dual-sensitive nanoplatforms were able to accumulate at the tumor sites. Treatment with PDDFe followed by alternating magnetic field and laser irradiation could prime hyperthermia/chemo/chemodynamic therapy to effectively retard tumor growth. This work presents a nanoplatform with a site-specific controlled release characteristic, showing great promises in potentiating drug delivery and advancing combinational cancer therapy.
Collapse
Affiliation(s)
- Nian Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| | - Liang Wu
- School of Medicine, Xiamen University, Xiamen 361102, P. R. China
| | - Wenbao Zuo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| | - Qian Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| | - Jinxue Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| | - Quanyi Jin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| | - Zhimei Xiao
- School of Medicine, Xiamen University, Xiamen 361102, P. R. China
| | - Luping Chen
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518116, P. R. China
| | - Yilin Zhao
- School of Medicine, Xiamen University, Xiamen 361102, P. R. China
- Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma (Xiamen University Affiliated ZhongShan Hospital), Xiamen 361004, P. R. China
| | - Jun Zhou
- Department of Pharmacy, South China Hospital of Shenzhen University, Shenzhen, Guangdong 518116, P. R. China
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, P. R. China
| |
Collapse
|
21
|
Rezaei A, Rafieian F, Akbari-Alavijeh S, Kharazmi MS, Jafari SM. Release of bioactive compounds from delivery systems by stimuli-responsive approaches; triggering factors, mechanisms, and applications. Adv Colloid Interface Sci 2022; 307:102728. [PMID: 35843031 DOI: 10.1016/j.cis.2022.102728] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 11/01/2022]
Abstract
Recent advances in emerging nanocarriers and stimuli-responsive (SR) delivery systems have brought about a revolution in the food and pharmaceutical industries. SR carriers are able to release the encapsulated bioactive compounds (bioactives) upon an external trigger. The potential of releasing the loaded bioactives in site-specific is of great importance for the pharmaceutical industry and medicine that can deliver the cargo in an appropriate condition. For the food industry, release of encapsulated bioactives is considerably important in processing or storage of food products and can be used in their formulation or packaging. There are various stimuli to control the favorite release of bioactives. In this review, we will shed light on the effect of different stimuli such as temperature, humidity, pH, light, enzymatic hydrolysis, redox, and also multiple stimuli on the release of encapsulated cargo and their potential applications in the food and pharmaceutical industries. An overview of cargo release mechanisms is also discussed. Furthermore, various alternatives to manipulate the controlled release of bioactives from carriers and the perspective of more progress in these SR carriers are highlighted.
Collapse
Affiliation(s)
- Atefe Rezaei
- Food Security Research Center, Department of Food Science and Technology, School of Nutrition and Food Science, Isfahan University of Medical Sciences, P.O. Box: 81746-73461, Isfahan, Iran.
| | - Fatemeh Rafieian
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Safoura Akbari-Alavijeh
- Department of Food Science and Technology, Faculty of Agriculture and Natural Resources, University of Mohaghegh Ardabili, P.O. Box 56199-11367, Ardabil, Iran
| | | | - Seid Mahdi Jafari
- Department of Food Materials and Process Design Engineering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran; Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, E-32004 Ourense, Spain.
| |
Collapse
|
22
|
Nanomaterials as Ultrasound Theragnostic Tools for Heart Disease Treatment/Diagnosis. Int J Mol Sci 2022; 23:ijms23031683. [PMID: 35163604 PMCID: PMC8835969 DOI: 10.3390/ijms23031683] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 01/27/2023] Open
Abstract
A variety of different nanomaterials (NMs) such as microbubbles (MBs), nanobubbles (NBs), nanodroplets (NDs), and silica hollow meso-structures have been tested as ultrasound contrast agents for the detection of heart diseases. The inner part of these NMs is made gaseous to yield an ultrasound contrast, which arises from the difference in acoustic impedance between the interior and exterior of such a structure. Furthermore, to specifically achieve a contrast in the diseased heart region (DHR), NMs can be designed to target this region in essentially three different ways (i.e., passively when NMs are small enough to diffuse through the holes of the vessels supplying the DHR, actively by being associated with a ligand that recognizes a receptor of the DHR, or magnetically by applying a magnetic field orientated in the direction of the DHR on a NM responding to such stimulus). The localization and resolution of ultrasound imaging can be further improved by applying ultrasounds in the DHR, by increasing the ultrasound frequency, or by using harmonic, sub-harmonic, or super-resolution imaging. Local imaging can be achieved with other non-gaseous NMs of metallic composition (i.e., essentially made of Au) by using photoacoustic imaging, thus widening the range of NMs usable for cardiac applications. These contrast agents may also have a therapeutic efficacy by carrying/activating/releasing a heart disease drug, by triggering ultrasound targeted microbubble destruction or enhanced cavitation in the DHR, for example, resulting in thrombolysis or helping to prevent heart transplant rejection.
Collapse
|
23
|
Cheng Z, Huang Y, Shao P, Wang L, Zhu S, Yu J, Lu W. Hypoxia-Activated Albumin-Binding Exatecan Prodrug for Cancer Therapy. ACS OMEGA 2022; 7:1082-1089. [PMID: 35036771 PMCID: PMC8757358 DOI: 10.1021/acsomega.1c05671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
As an effective drug delivery strategy for traditional antitumor drugs, the stimulus-responsive albumin-based prodrugs are getting more and more attention. These prodrugs only release drugs in specific tumor microenvironments, which can prevent premature release of the drug in the circulation. Tumor hypoxia is a fundamental feature of the solid tumor microenvironment. As a hypoxia-activated linker, the 5-position branched linker of 1-methyl-2-nitro-5-hydroxymethylimidazole can be a trigger for albumin-based prodrugs. In this study, we report the synthesis and biological evaluation of the hypoxia-activated albumin-binding prodrug Mal-azo-Exatecan. After intravenous administration, the maleimide on the side chain can rapidly bind to endogenous albumin, enabling the prodrugs to accumulate in tumors, where tumor-associated hypoxia microenvironments trigger the selective release of Exatecan. The 5-position branched linker of 1-methyl-2-nitro-5-hydroxymethylimidazole as a cleavable linker has high plasma stability and does not cause Exatecan release from HSA-azo-Exatecan during circulation in vivo, avoiding systemic side effects caused by Exatecan.
Collapse
|
24
|
Berillo D, Zharkinbekov Z, Kim Y, Raziyeva K, Temirkhanova K, Saparov A. Stimuli-Responsive Polymers for Transdermal, Transmucosal and Ocular Drug Delivery. Pharmaceutics 2021; 13:2050. [PMID: 34959332 PMCID: PMC8708789 DOI: 10.3390/pharmaceutics13122050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022] Open
Abstract
Despite their conventional and widespread use, oral and intravenous routes of drug administration face several limitations. In particular, orally administered drugs undergo enzymatic degradation in the gastrointestinal tract and first-pass metabolism in the liver, which tend to decrease their bioavailability. Intravenous infusions of medications are invasive, painful and stressful for patients and carry the risk of infections, tissue damage and other adverse reactions. In order to account for these disadvantages, alternative routes of drug delivery, such as transdermal, nasal, oromucosal, ocular and others, have been considered. Moreover, drug formulations have been modified in order to improve their storage stability, solubility, absorption and safety. Recently, stimuli-responsive polymers have been shown to achieve controlled release and enhance the bioavailability of multiple drugs. In this review, we discuss the most up-to-date use of stimuli-responsive materials in order to optimize the delivery of medications that are unstable to pH or undergo primary metabolism via transdermal, nasal, oromucosal and ocular routes. Release kinetics, diffusion parameters and permeation rate of the drug via the mucosa or skin are discussed as well.
Collapse
Affiliation(s)
- Dmitriy Berillo
- Department of Pharmaceutical and Toxicological Chemistry, Pharmacognosy and Botany School of Pharmacy, Asfendiyarov Kazakh National Medical University, Almaty 050000, Kazakhstan
| | - Zharylkasyn Zharkinbekov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Z.Z.); (Y.K.); (K.R.); (K.T.)
| | - Yevgeniy Kim
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Z.Z.); (Y.K.); (K.R.); (K.T.)
| | - Kamila Raziyeva
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Z.Z.); (Y.K.); (K.R.); (K.T.)
| | - Kamila Temirkhanova
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Z.Z.); (Y.K.); (K.R.); (K.T.)
| | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (Z.Z.); (Y.K.); (K.R.); (K.T.)
| |
Collapse
|
25
|
Mena-Giraldo P, Orozco J. Polymeric Micro/Nanocarriers and Motors for Cargo Transport and Phototriggered Delivery. Polymers (Basel) 2021; 13:3920. [PMID: 34833219 PMCID: PMC8621231 DOI: 10.3390/polym13223920] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 02/07/2023] Open
Abstract
Smart polymer-based micro/nanoassemblies have emerged as a promising alternative for transporting and delivering a myriad of cargo. Cargo encapsulation into (or linked to) polymeric micro/nanocarrier (PC) strategies may help to conserve cargo activity and functionality when interacting with its surroundings in its journey to the target. PCs for cargo phototriggering allow for excellent spatiotemporal control via irradiation as an external stimulus, thus regulating the delivery kinetics of cargo and potentially increasing its therapeutic effect. Micromotors based on PCs offer an accelerated cargo-medium interaction for biomedical, environmental, and many other applications. This review collects the recent achievements in PC development based on nanomicelles, nanospheres, and nanopolymersomes, among others, with enhanced properties to increase cargo protection and cargo release efficiency triggered by ultraviolet (UV) and near-infrared (NIR) irradiation, including light-stimulated polymeric micromotors for propulsion, cargo transport, biosensing, and photo-thermal therapy. We emphasize the challenges of positioning PCs as drug delivery systems, as well as the outstanding opportunities of light-stimulated polymeric micromotors for practical applications.
Collapse
Affiliation(s)
| | - Jahir Orozco
- Max Planck Tandem Group in Nanobioengineering, Institute of Chemistry, Faculty of Natural and Exact Sciences, University of Antioquia, Complejo Ruta N, Calle 67 # 52-20, Medellin 050010, Colombia;
| |
Collapse
|
26
|
Shishir MRI, Gowd V, Suo H, Wang M, Wang Q, Chen F, Cheng KW. Advances in smart delivery of food bioactive compounds using stimuli-responsive carriers: Responsive mechanism, contemporary challenges, and prospects. Compr Rev Food Sci Food Saf 2021; 20:5449-5488. [PMID: 34668321 DOI: 10.1111/1541-4337.12851] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/12/2021] [Accepted: 09/03/2021] [Indexed: 12/19/2022]
Abstract
Many important food bioactive compounds are plant secondary metabolites that have traditional applications for health promotion and disease prevention. However, the chemical instability and poor bioavailability of these compounds represent major challenges to researchers. In the last decade, therefore, major impetus has been given for the research and development of advanced carrier systems for the delivery of natural bioactive molecules. Among them, stimuli-responsive carriers hold great promise for simultaneously improving stability, bioavailability, and more importantly delivery and on-demand release of intact bioactive phytochemicals to target sites in response to certain stimuli or combination of them (e.g., pH, temperature, oxidant, enzyme, and irradiation) that would eventually enhance therapeutic outcomes and reduce side effects. Hybrid formulations (e.g., inorganic-organic complexes) and multi-stimuli-responsive formulations have demonstrated great potential for future studies. Therefore, this review systematically compiles and assesses the recent advances on the smart delivery of food bioactive compounds, particularly quercetin, curcumin, and resveratrol through stimuli-responsive carriers, and critically reviews their functionality, underlying triggered-release mechanism, and therapeutic potential. Finally, major limitations, contemporary challenges, and possible solutions/future research directions are highlighted. Much more research is needed to optimize the processing parameters of existing formulations and to develop novel ones for lead food bioactive compounds to facilitate their food and nutraceutical applications.
Collapse
Affiliation(s)
- Mohammad Rezaul Islam Shishir
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen, China.,Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China.,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Vemana Gowd
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China.,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Hao Suo
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China.,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Mingfu Wang
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China.,School of Biological Sciences, The University of Hong Kong, Hong Kong, P. R. China
| | - Qiang Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture, Beijing, China
| | - Feng Chen
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China.,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| | - Ka-Wing Cheng
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen, China.,Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen, China
| |
Collapse
|
27
|
Guan L, Chen J, Tian Z, Zhu M, Bian Y, Zhu Y. Mesoporous organosilica nanoparticles: Degradation strategies and application in tumor therapy. VIEW 2021. [DOI: 10.1002/viw.20200117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Lei Guan
- School of Materials Science and Engineering University of Shanghai for Science and Technology Shanghai China
| | - Jiajie Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai China
| | - Zhengfang Tian
- Hubei Key Laboratory of Processing and Application of Catalytic Materials, College of Chemical Engineering Huanggang Normal University Huanggang Hubei Province China
| | - Min Zhu
- School of Materials Science and Engineering University of Shanghai for Science and Technology Shanghai China
| | - Yuhai Bian
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Yufang Zhu
- Hubei Key Laboratory of Processing and Application of Catalytic Materials, College of Chemical Engineering Huanggang Normal University Huanggang Hubei Province China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai China
| |
Collapse
|
28
|
Uebel F, Thérien-Aubin H, Landfester K. Tailoring the mechanoresponsive release from silica nanocapsules. NANOSCALE 2021; 13:15415-15421. [PMID: 34499058 DOI: 10.1039/d1nr04697g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Triggering the release of encapsulated cargos using mechanical stress acting on a nanocarrier is a strategy with potential applications from drug delivery to self-healing coatings. The mechanically triggered release of encapsulated molecules can be controlled by tuning the mechanical properties of the nanocapsules, which are strongly linked to the nanocapsule architecture. Here, silica nanocarriers were designed to tune precisely the release initiated by mechanical stress. We synthesized silica nanocapsules (SiNCs) with a finely tunable diameter and shell thickness and performed AFM nanoindentation experiments to determine the breaking force of single SiNCs. We demonstrated that it is possible to trigger the release of encapsulated payload by the application of an external mechanical force on the SiNCs. Furthermore, we successfully controlled the breaking force and the amount of released payload by tailoring the architecture of the nanocarriers, illustrating how such mechanoresponsive SiNCs could be used as responsive nanocarriers for the delivery of molecular cargos.
Collapse
Affiliation(s)
- Fabian Uebel
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Héloïse Thérien-Aubin
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
- Department of Chemistry, Memorial University of Newfoundland, 283 Prince Philip Dr, St. John's, NL, A1B 3X7, Canada.
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| |
Collapse
|
29
|
Ultrasound-Responsive Smart Drug Delivery System of Lipid Coated Mesoporous Silica Nanoparticles. Pharmaceutics 2021; 13:pharmaceutics13091396. [PMID: 34575472 PMCID: PMC8468042 DOI: 10.3390/pharmaceutics13091396] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/23/2022] Open
Abstract
The immediate release of chemotherapeutics at the target site, along with no premature release in circulation is always challenging. The purpose of this study was to develop a stimuli responsive drug delivery system, composed of lipid supported mesoporous silica nanoparticles (MSNPs) for triggered drug release at the target site and simultaneously avoiding the premature release. MSNPs with a higher drug loading capacity and very slow release were designed so as to enhance release by FDA approved US-irradiation. Doxorubicin, as a model drug, and perfluoropentane (PFP) as a US responsive material, were entrapped in the porous structure of MSNPs. Lipid coating enhanced the cellular uptake and in addition provided a gatekeeping effect at the pore opening to reduce premature release. The mechanical and thermal effects of US induced the conversion of liquid PFP to a gaseous form that was able to rupture the lipid layer, resulting in triggered drug release. The prolonged stability profile and non-toxic behavior made them suitable candidate for the delivery of anticancer drugs. This smart system, with the abilities of better cellular uptake and higher cytotoxic effects on US-irradiation, would be a good addition to the applied side of chemotherapeutic advanced drug delivery systems.
Collapse
|
30
|
Kapalatiya H, Madav Y, Tambe VS, Wairkar S. Enzyme-responsive smart nanocarriers for targeted chemotherapy: an overview. Drug Deliv Transl Res 2021; 12:1293-1305. [PMID: 34251612 DOI: 10.1007/s13346-021-01020-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2021] [Indexed: 02/02/2023]
Abstract
Nanocarriers play pivotal roles in the field of biomedical applications, particularly in anticancer therapy. One of the prominent strategies for the transport of anticancer drugs with site-specific release and improved therapeutic efficacy is the use of an enzyme-responsive drug delivery system. There is an emerging class of cancer therapeutics engineered to control the release of a drug via enzymatic degradation. Enzymes, being an essential component of bio-nanotechnology toolbox, hold exceptional biorecognition abilities as well as outstanding catalytic properties. Often, abnormal enzyme expression observed in cancer offers many opportunities in designing nanocarriers modified with enzyme-labile linkage. Through altered physical or chemical characteristics of these nanocarriers or cleavage of the drug in response to the bio-action of enzyme, an on-demand drug release can be obtained. In this review, several classes of enzymes performing critical roles in cancer such as hydrolases, lipases, and oxidoreductases are summarized. Insights on various approaches that interfere with the mechanism of these enzymes have also been included. Finally, various smart nanocarriers such as mesoporous silica nanoparticles, gold nanoparticles, carbon-nanotubes, micelles, liposomes, and dendrimers serving as excellent platforms for enzyme-responsive formulations have been discussed.
Collapse
Affiliation(s)
- Hiral Kapalatiya
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Yamini Madav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Varunesh Sanjay Tambe
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
31
|
Herdiana Y, Wathoni N, Shamsuddin S, Joni IM, Muchtaridi M. Chitosan-Based Nanoparticles of Targeted Drug Delivery System in Breast Cancer Treatment. Polymers (Basel) 2021; 13:1717. [PMID: 34074020 PMCID: PMC8197416 DOI: 10.3390/polym13111717] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/18/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Breast cancer remains one of the world's most dangerous diseases because of the difficulty of finding cost-effective and specific targets for effective and efficient treatment methods. The biodegradability and biocompatibility properties of chitosan-based nanoparticles (ChNPs) have good prospects for targeted drug delivery systems. ChNPs can transfer various antitumor drugs to targeted sites via passive and active targeting pathways. The modification of ChNPs has attracted the researcher to the loading of drugs to targeted cancer cells. The objective of our review was to summarize and discuss the modification in ChNPs in delivering anticancer drugs against breast cancer cells from published papers recorded in Scopus, PubMed, and Google Scholar. In order to improve cellular uptake, drug accumulation, cytotoxicity, and selectivity, we examined different kinds of modification of ChNPs. Notably, these forms of ChNPs use the characteristics of the enhanced permeability and retention (EPR) effect as a proper parameter and different biological ligands, such as proteins, peptides, monoclonal antibodies, and small particles. In addition, as a targeted delivery system, ChNPs provided and significantly improved the delivery of drugs into specific breast cancer cells (MDA-MB-231, 4T1 cells, SK-BR-3, MCF-7, T47D). In conclusion, a promising technique is presented for increasing the efficacy, selectivity, and effectiveness of candidate drug carriers in the treatment of breast cancer.
Collapse
Affiliation(s)
- Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (Y.H.); (N.W.)
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (Y.H.); (N.W.)
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia;
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), USM, Penang 11800, Malaysia
- USM-RIKEN Interdisciplinary Collaboration on Advanced Sciences (URICAS), USM, Penang 11800, Malaysia
| | - I Made Joni
- Departement of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jl. Raya Bandung Sumedang KM.21 Jatinangor, Sumedang 45363, Indonesia;
- Functional Nano Powder University Center of Excellence, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| |
Collapse
|
32
|
Wang Q, Bao Y. Nanodelivery of natural isothiocyanates as a cancer therapeutic. Free Radic Biol Med 2021; 167:125-140. [PMID: 33711418 DOI: 10.1016/j.freeradbiomed.2021.02.044] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/31/2021] [Accepted: 02/26/2021] [Indexed: 12/18/2022]
Abstract
Natural isothiocyanates (ITCs) are phytochemicals abundant in cruciferous vegetables with the general structure, R-NCS. They are bioactive organosulfur compounds derived from the hydrolysis of glucosinolates by myrosinase. A significant number of isothiocyanates have been isolated from different plant sources that include broccoli, Brussels sprouts, cabbage, cauliflower, kale, mustard, wasabi, and watercress. Several ITCs have been demonstrated to possess significant pharmacological properties including: antioxidant, anti-inflammatory, anti-cancer and antimicrobial activities. Due to their chemopreventive effects on many types of cancer, ITCs have been regarded as a promising anti-cancer therapeutic agent without major toxicity concerns. However, their clinical application has been hindered by several factors including their low aqueous solubility, low bioavailability, instability as well as their hormetic effect. Moreover, the typical dietary uptake of ITCs consumed for promotion of good health may be far from their bioactive (or cytotoxic) dose necessary for cancer prevention and/or treatment. Nanotechnology is one of best options to attain enhanced efficacy and minimize hormetic effect for ITCs. Nanoformulation of ITCs leads to enhance stability of ITCs in plasma and emphasize on their chemopreventive effects. This review provides a summary of the potential bioactivities of ITCs, their mechanisms of action for the prevention and treatment of cancer, as well as the recent research progress in their nanodelivery strategies to enhance solubility, bioavailability, and anti-cancer efficacy.
Collapse
Affiliation(s)
- Qi Wang
- Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK.
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich NR4 7UQ, UK.
| |
Collapse
|
33
|
Thermosensitive Drug Delivery System SBA-15-PEI for Controlled Release of Nonsteroidal Anti-Inflammatory Drug Diclofenac Sodium Salt: A Comparative Study. MATERIALS 2021; 14:ma14081880. [PMID: 33918907 PMCID: PMC8068836 DOI: 10.3390/ma14081880] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/19/2022]
Abstract
Mesoporous SBA-15 silica material was prepared by the sol–gel method and functionalized with thermosensitive polyethylenimine polymers with different molecular weight (g·mol−1): 800 (SBA-15(C)-800), 1300 (SBA-15(C)-1300) and 2000 (SBA-15(C)-2000). The nonsteroidal anti-inflammatory drug (NSAID) diclofenac sodium was selected as a model drug and encapsulated into the pores of prepared supports. Materials were characterized by the combination of infrared spectroscopy (IR), atomic force microscopy (AFM), transmission electron microscopy (TEM), photon cross-correlation spectroscopy (PCCS), nitrogen adsorption/desorption analysis, thermogravimetry (TG), differential scanning calorimetry (DSC) and small-angle X-ray diffraction (SA-XRD) experiments. The drug release from prepared matrixes was realized in two model media differing in pH, namely small intestine environment/simulated body fluid (pH = 7.4) and simulated gastric fluid (pH = 2), and at different temperatures, namely normal body temperature (T = 37 °C) and inflammatory temperature (T = 42 °C). The process of drug loading into the pores of prepared materials from the diclofenac sodium salt solutions with different concentrations and subsequent quantitative determination of released drugs was analyzed by UV-VIS spectroscopy. Analysis of prepared SBA-15 materials modified with polyethylenimines in solution showed a high ability to store large amounts of the drug, up to 230 wt.%. Experimental results showed their high drug release into the solution at pH = 7.4 for both temperatures, which is related to the high solubility of diclofenac sodium in a slightly alkaline environment. At pH = 2, a difference in drug release rate was observed between both temperatures. Indeed, at a higher temperature, the release rates and the amount of released drug were 2–3 times higher than those observed at a lower temperature. Different kinetic models were used to fit the obtained drug release data to determine the drug release rate and its release mechanism. Moreover, the drug release properties of prepared compounds were compared to a commercially available medicament under the same experimental conditions.
Collapse
|
34
|
Kim T, Nam K, Kim YM, Yang K, Roh YH. DNA-Assisted Smart Nanocarriers: Progress, Challenges, and Opportunities. ACS NANO 2021; 15:1942-1951. [PMID: 33492127 DOI: 10.1021/acsnano.0c08905] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Due to powerful breakthroughs in nanotechnology, smart delivery mechanisms have rapidly emerged for use in diverse applications across biomedical research and therapeutic development. Recent efforts toward understanding stimuli-responsive strategies have led to substantial improvements in their conceptual application and in vitro efficiency. Because disease targets for therapy are often localized in specific cells, organs, or tissues, an enhanced permeability and retention (EPR)-based strategy remains inadequate for accurate drug delivery and release to target regions, resulting in an insufficient drug concentration reaching the target region and undesired side effects. To address these issues, more precise and remote-controlled stimuli-responsive systems, which recognize and react to changes in the pathophysiological microenvironment, were recently elucidated as feasible on-demand drug-delivery systems. In this Perspective, we focus on progress toward stimuli-responsive drug-delivery systems that utilize dynamic DNA molecules by exploiting DNA nanotechnology. DNA structures can be precisely reconfigured by external and internal stimuli to drive the release of a loaded drug in a target region with appropriate microenvironments. We describe the chemical, physical, and biological engineering principles and strategies for constructing DNA-assisted nanocarriers. We also provide a summary of smart nanocarrier systems, organized with respect to the structural changes in the DNA strand in the microenvironment, resulting from changes in pH and temperature and the presence of intracellular oligonucleotides. To do so, we highlight recent advances in related biomedical research and applications as well as discuss major challenges and opportunities for DNA-assisted nanocarriers to guide the development of future in vivo therapies and clinical translation strategies.
Collapse
Affiliation(s)
- Taehyung Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Keonwook Nam
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Young Min Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Kyungjik Yang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Young Hoon Roh
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
35
|
Li L, Fu J, Wang X, Chen Q, Zhang W, Cao Y, Ran H. Biomimetic "Nanoplatelets" as a Targeted Drug Delivery Platform for Breast Cancer Theranostics. ACS APPLIED MATERIALS & INTERFACES 2021; 13:3605-3621. [PMID: 33449625 DOI: 10.1021/acsami.0c19259] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Breast cancer is a major threat to health and lives of females. Biomimetic nanotechnology brought brighter hope for early diagnosis and treatment of breast cancer. Here, we proposed a platelet (PLT) membrane-derived strategy for enhanced photoacoustic (PA)/ultrasonic (US)/fluorescence (FL) multimodal imaging and augmented synergistic photothermal/chemotherapeutic efficacy in tumor cells. A PA imaging contrast and photothermal agent, nanocarbons (CNs), a chemotherapeutic and FL material, doxorubicin (DOX), and perfluoropentane (PFP) were coencapsulated into the poly(lactic-co-glycolic) acid (PLGA) skeletons. Then, the PLT membranes were coated onto the PLGA NPs, which were named as "nanoplatelets" (DOX-PFP-CNs@PLGA/PM NPs). The "nanoplatelets", which conserved the structural advantages and inherent properties of PLTs, could not only escape from phagocytosis of macrophages but also actively targeted tumor cells by the way of antigen-antibody interactions between P-selectin on the PM and CD44 receptors of the tumor cells. With CNs and DOX loaded in, these "nanoplatelets" could serve as an excellent contrast agent for PA/FL imaging. Under laser irradiation, the "nanoplatelets" could turn light energy into heat energy. The laser-triggered photothermal effect, on the one hand, could ablate the tumor cells immediately, and on the other hand, could initiate the optical droplet vaporization of PFP, which subsequently enhanced US imaging and promoted the discharge of encapsulated DOX from the "nanoplatelets" for remarkably strengthening photothermal therapeutic power in turn. In this work, as compared with the bare drug-loaded nanoparticles, the "nanoplatelets" exhibited much more accumulation in the tumor cells, demonstrating superior multimodal imaging capability and preferable synergistic therapeutic performance. In conclusion, the "nanoplatelets" could serve as contrast agents for US imaging and PA imaging to guide the therapy. What is more, the bioinspired PLT-derived, targeted, and nontoxic "nanoplatelets", which were exploited for multimodal PA/US/FL imaging-guided synergistic photothermal/chemo therapy, will be of great value to breast cancer theranostics in the days to come.
Collapse
Affiliation(s)
- Lin Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Jian Fu
- Vascular Surgery Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Xingyue Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Qiaoqi Chen
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Wei Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P. R. China
| |
Collapse
|
36
|
Huda S, Alam MA, Sharma PK. Smart nanocarriers-based drug delivery for cancer therapy: An innovative and developing strategy. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.102018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Li Y, Mei T, Han S, Han T, Sun Y, Zhang H, An F. Cathepsin B-responsive nanodrug delivery systems for precise diagnosis and targeted therapy of malignant tumors. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2020.05.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
38
|
Miao H, Chen X, Luan Y. Small Molecular Gemcitabine Prodrugs for Cancer Therapy. Curr Med Chem 2020; 27:5562-5582. [PMID: 31419928 DOI: 10.2174/0929867326666190816230650] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/22/2019] [Accepted: 07/09/2019] [Indexed: 02/04/2023]
Abstract
Gemcitabine as a pyrimidine nucleoside analog anticancer drug has high efficacy for a broad spectrum of solid tumors. Gemcitabine is activated within tumor cells by sequential phosphorylation carried out by deoxycytidine kinase to mono-, di-, and triphosphate nucleotides with the last one as the active form. But the instability, drug resistance and toxicity severely limited its utilization in clinics. In the field of medicinal chemistry, prodrugs have proven to be a very effective means for elevating drug stability and decrease undesirable side effects including the nucleoside anticancer drug such as gemcitabine. Many works have been accomplished in design and synthesis of gemcitabine prodrugs, majority of which were summarized in this review.
Collapse
Affiliation(s)
- He Miao
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Shandong Province, Qingdao, China
| | - Xuehong Chen
- Department of Pharmacology, College of Basic Medicine, Qingdao University, Shandong Province, Qingdao, China
| | - Yepeng Luan
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Shandong Province, Qingdao, China
| |
Collapse
|
39
|
Wang W, Liu X, Zheng X, Jin HJ, Li X. Biomineralization: An Opportunity and Challenge of Nanoparticle Drug Delivery Systems for Cancer Therapy. Adv Healthc Mater 2020; 9:e2001117. [PMID: 33043640 DOI: 10.1002/adhm.202001117] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/29/2020] [Indexed: 12/12/2022]
Abstract
Biomineralization is a common process in organisms to produce hard biomaterials by combining inorganic ions with biomacromolecules. Multifunctional nanoplatforms are developed based on the mechanism of biomineralization in many biomedical applications. In the past few years, biomineralization-based nanoparticle drug delivery systems for the cancer treatment have gained a lot of research attention due to the advantages including simple preparation, good biocompatibility, degradability, easy modification, versatility, and targeting. In this review, the research trends of biomineralization-based nanoparticle drug delivery systems and their applications in cancer therapy are summarized. This work aims to promote future researches on cancer therapy based on biomineralization. Rational design of nanoparticle drug delivery systems can overcome the bottleneck in the clinical transformation of nanomaterials. At the same time, biomineralization has also provided new research ideas for cancer treatment, i.e., targeted therapy, which has significantly better performance.
Collapse
Affiliation(s)
- Weicai Wang
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Xiaofan Liu
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Xiangjiang Zheng
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| | - Hyung Jong Jin
- Department of Bioscience and Biotechnology The University of Suwon Hwaseong Gyeonggi‐Do 18323 Republic of Korea
| | - Xuemei Li
- Collaborative Innovation Center of Tumor Marker Detection Technology Equipment and Diagnosis‐Therapy Integration in Universities of Shandong Shandong Province Key Laboratory of Detection Technology for Tumor Makers School of Chemistry and Chemical Engineering Linyi University Linyi Shandong 276005 China
| |
Collapse
|
40
|
Azo-inserted responsive hybrid liposomes for hypoxia-specific drug delivery. Acta Biomater 2020; 115:343-357. [PMID: 32771598 DOI: 10.1016/j.actbio.2020.07.061] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 11/21/2022]
Abstract
Stimuli-responsive drug delivery systems using endogenous stimuli from tumor microenvironments such as acidic pH, over-expressed enzyme, and high redox potential as triggers have shown tremendous promise in cancer therapy. However, their clinical application is severely limited because of tumor heterogeneity. Hypoxia, a physiological feature observed in almost all solid tumors and even in nodules with very small size, has currently emerged as a more general but efficient stimulus to trigger release. Herein, we developed hypoxia-responsive hybrid liposomes (HR-HLPs), composed of azo-inserted organokoxysilane-based lipid analogue as a responsive component and commercial phospholipid for reducing the rigidity of liposomal membrane caused by azo, for drug delivery targeting tumor hypoxia. HR-HLPs had the advantages of high structural stability to avoid premature drug leakage when circulating in the blood and high sensitivity in responding to hypoxia once reaching tumor sites. HR-HLPs exhibit deep tumor penetration capability, enabling effective delivery to hypoxic regions distant from tumor vessels. Moreover, HR-HLPs could selectively release their payload, co-localizing with over-expressed hypoxia inducible factor 1α (HIF-1α) in vitro and in vivo. As a result, HR-HLPs showed improved therapeutic outcome accompanied by reduced adverse effects. The results highlighted the potential application of azo-inserted responsive hybrid liposomes for hypoxia-targeted drug delivery. STATEMENT OF SIGNIFICANCE.
Collapse
|
41
|
Li D, Zhang R, Liu G, Kang Y, Wu J. Redox-Responsive Self-Assembled Nanoparticles for Cancer Therapy. Adv Healthc Mater 2020; 9:e2000605. [PMID: 32893506 DOI: 10.1002/adhm.202000605] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/16/2020] [Indexed: 12/21/2022]
Abstract
Chemotherapy, combined with other treatments, is widely applied in the clinical treatment of cancer. However, deficiencies inherited from the traditional route of administration limit its successful application. With the development of nanotechnology, a series of smart nanodelivery systems have been developed to utilize the unique tumor environment (pH changes, different enzymes, and redox potential gradients) and exogenous stimuli (thermal changes, magnetic fields, and light) to improve the curative effect of anticancer drugs. In this review, endogenous and exogenous stimuli are briefly introduced. Among these stimuli, various redox-sensitive linkages are primarily described in detail, and their application with self-assembled nanoparticles is recounted. Finally, the application of redox-responsive self-assembled nanoparticles in cancer therapy is summarized.
Collapse
Affiliation(s)
- Dandan Li
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province School of Biomedical Engineering Sun Yat‐sen University Guangzhou 510006 P. R. China
- The Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen 518107 P. R. China
| | - Ruhe Zhang
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province School of Biomedical Engineering Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Guiting Liu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province School of Biomedical Engineering Sun Yat‐sen University Guangzhou 510006 P. R. China
| | - Yang Kang
- The Seventh Affiliated Hospital Sun Yat‐sen University Shenzhen 518107 P. R. China
| | - Jun Wu
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province School of Biomedical Engineering Sun Yat‐sen University Guangzhou 510006 P. R. China
| |
Collapse
|
42
|
Zohreh N, Rastegaran Z, Hosseini SH, Akhlaghi M, Istrate C, Busuioc C. pH-triggered intracellular release of doxorubicin by a poly(glycidyl methacrylate)-based double-shell magnetic nanocarrier. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111498. [PMID: 33255062 DOI: 10.1016/j.msec.2020.111498] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/25/2020] [Accepted: 09/04/2020] [Indexed: 12/26/2022]
Abstract
Two core-double-shell pH-sensitive nanocarriers were fabricated using Fe3O4 as magnetic core, poly(glycidyl methacrylate-PEG) and salep dialdehyde as the first and the second shell, and doxorubicin as the hydrophobic anticancer drug. Two nanocarriers were different in the drug loading steps. The interaction between the first and the second shell assumed to be pH-sensitive via acetal cross linkages. The structure of nanocarriers, organic shell loading, magnetic responsibility, morphology, size, dispersibility, and drug loading content were investigated by IR, NMR, TG, VSM, XRD, DLS, HRTEM and UV-Vis analyses. The long-term drug release profiles of both nanocarriers showed that the drug loading before cross-linking between the first and second shell led to a more pH-sensitive nanocarrier exhibiting higher control on DOX release. Cellular toxicity assay (MTT) showed that DOX-free nanocarrier is biocompatible having cell viability greater than 80% for HEK-293 and MCF-7 cell lines. Besides, high cytotoxic effect observed for drug-loaded nanocarrier on MCF-7 cancer cells. Cellular uptake analysis showed that the nanocarrier is able to transport DOX into the cytoplasm and perinuclear regions of MCF-7 cells. In vitro hemolysis and coagulation assays demonstrated high blood compatibility of nanocarrier. The results also suggested that low concentration of nanocarrier have a great potential as a contrast agent in magnetic resonance imaging (MRI).
Collapse
Affiliation(s)
- Nasrin Zohreh
- Department of Chemistry, Faculty of Science, University of Qom, P. O. Box: 37185-359, Qom, Iran.
| | - Zahra Rastegaran
- Department of Chemistry, Faculty of Science, University of Qom, P. O. Box: 37185-359, Qom, Iran
| | - Seyed Hassan Hosseini
- Department of Chemical Engineering, University of Science and Technology of Mazandaran, Behshahr, Iran.
| | - Mehdi Akhlaghi
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran 1414713135, Iran
| | - Cosmin Istrate
- Laboratory of Atomic Structures and Defects in Advanced Materials, National Institute of Materials Physics, Magurele, Romania
| | - Cristina Busuioc
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, Bucharest, Romania
| |
Collapse
|
43
|
Tian B, Liu Y, Liu J. Smart stimuli-responsive drug delivery systems based on cyclodextrin: A review. Carbohydr Polym 2020; 251:116871. [PMID: 33142550 DOI: 10.1016/j.carbpol.2020.116871] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/29/2022]
Abstract
Stimulated by researches in materials chemistry and medicine fields, drug delivery has entered a new stage of development. Drug delivery systems have been extensively studied according to the differences in the drug therapeutic environment such as pH, light, temperature, magnet, redox, enzymes, etc. Cyclodextrin is a smart tool that has been proven to be used in the preparation of drug delivery, and has become a new area of concern in recent years. In this review, we discuss recent research advances in smart stimuli-responsive cyclodextrin-based drug delivery. First, different stimuli-responsive drug delivery systems based on cyclodextrin are introduced and classified. Then, the characteristics of different types of stimuli-responsive drug delivery systems are described, and their applications are emphasized. Finally, current challenges and future development opportunities of smart stimuli-responsive drug delivery systems based on cyclodextrin are discussed.
Collapse
Affiliation(s)
- Bingren Tian
- College of Chemistry and Chemical Engineering, Xinjiang University, Urumchi, 830001, China.
| | - Yumei Liu
- College of Chemistry and Chemical Engineering, Xinjiang University, Urumchi, 830001, China.
| | - Jiayue Liu
- School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
44
|
Wang D, Wang S, Xia Y, Liu S, Jia R, Xu G, Zhan J, Lu Y. Preparation of ROS-responsive core crosslinked polycarbonate micelles with thioketal linkage. Colloids Surf B Biointerfaces 2020; 195:111276. [PMID: 32763765 DOI: 10.1016/j.colsurfb.2020.111276] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 11/17/2022]
Abstract
Herein, we prepared novel reactive oxygen species (ROS) responsive core crosslinked (CCL/TK) polycarbonate micelles conveniently by click reaction between amphiphilic diblock copolymer poly(ethylene glycol)-poly(5-methyl-5-propargylxycar-bonyl-1,3-dioxane-2-one) (PEG-PMPC) with pendant alkynyl group and thioketal containing azide derivative bis (2-azidoethyl) 3, 3'- (propane-2, 2-diylbis (sulfanediyl)) dipropanoate (TK-N3). The CCL/TK micelles were obtained with small size of 146.4 nm, showing excellent stability against dilution and high doxorubicin (DOX) loading. In vitro toxicity tests demonstrated that the obtained CCL/TK micelles have good biocompatibility and low toxicity with cell viability above 95 %. Furthermore, DOX-loaded CCL/TK micelles showed significantly superior toxicity with IC50 values for HeLa and MCF-7 cells about 3.74 μg/mL and 3.91 μg/mL, respectively. Confocal laser scanning microscope (CLSM) and flow cytometry showed excellent internalization efficiency and intracellular drug release of DOX-loaded CCL/TK micelles. The obtained ROS-responsive CCL/TK micelles showed great potential for anticancer drug delivery.
Collapse
Affiliation(s)
- Deqi Wang
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Song Wang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, PR China
| | - Yingchun Xia
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Simeng Liu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Ruixin Jia
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Gege Xu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Junjie Zhan
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Yanbing Lu
- College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China; State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha, 410082, PR China.
| |
Collapse
|
45
|
Caramazza L, Nardoni M, De Angelis A, Paolicelli P, Liberti M, Apollonio F, Petralito S. Proof-of-Concept of Electrical Activation of Liposome Nanocarriers: From Dry to Wet Experiments. Front Bioeng Biotechnol 2020; 8:819. [PMID: 32793572 PMCID: PMC7390969 DOI: 10.3389/fbioe.2020.00819] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/26/2020] [Indexed: 12/19/2022] Open
Abstract
The increasing interest toward biocompatible nanotechnologies in medicine, combined with electric fields stimulation, is leading to the development of electro-sensitive smart systems for drug delivery applications. To this regard, recently the use of pulsed electric fields to trigger release across phospholipid membranes of liposomes has been numerically studied, for a deeper understanding of the phenomena at the molecular scale. Aim of this work is to give an experimental validation of the feasibility to control the release from liposome vesicles, using nanosecond pulsed electric fields characterized by a 10 ns duration and intensity in the order of MV/m. The results are supported by multiphysics simulations which consider the coupling of three physics (electromagnetics, thermal and pore kinetics) in order to explain the occurring physical interactions at the microscopic level and provide useful information on the characteristics of the train of pulses needed to obtain quantitative results in terms of liposome electropermeabilization. Finally, a complete characterization of the exposure system is also provided to support the reliability and validity of the study.
Collapse
Affiliation(s)
- Laura Caramazza
- ICEmB at DIET, Sapienza University of Rome, Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Martina Nardoni
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Annalisa De Angelis
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Patrizia Paolicelli
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Micaela Liberti
- ICEmB at DIET, Sapienza University of Rome, Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Francesca Apollonio
- ICEmB at DIET, Sapienza University of Rome, Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Stefania Petralito
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
46
|
Pham SH, Choi Y, Choi J. Stimuli-Responsive Nanomaterials for Application in Antitumor Therapy and Drug Delivery. Pharmaceutics 2020; 12:E630. [PMID: 32635539 PMCID: PMC7408499 DOI: 10.3390/pharmaceutics12070630] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 01/14/2023] Open
Abstract
The new era of nanotechnology has produced advanced nanomaterials applicable to various fields of medicine, including diagnostic bio-imaging, chemotherapy, targeted drug delivery, and biosensors. Various materials are formed into nanoparticles, such as gold nanomaterials, carbon quantum dots, and liposomes. The nanomaterials have been functionalized and widely used because they are biocompatible and easy to design and prepare. This review mainly focuses on nanomaterials responsive to the external stimuli used in drug-delivery systems. To overcome the drawbacks of conventional therapeutics to a tumor, the dual- and multi-responsive behaviors of nanoparticles have been harnessed to improve efficiency from a drug delivery point of view. Issues and future research related to these nanomaterial-based stimuli sensitivities and the scope of stimuli-responsive systems for nanomedicine applications are discussed.
Collapse
Affiliation(s)
| | | | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea; (S.H.P.); (Y.C.)
| |
Collapse
|
47
|
Meng X, Xu Y, Lu Q, Sun L, An X, Zhang J, Chen J, Gao Y, Zhang Y, Ning X. Ultrasound-responsive alkaline nanorobots for the treatment of lactic acidosis-mediated doxorubicin resistance. NANOSCALE 2020; 12:13801-13810. [PMID: 32573588 DOI: 10.1039/d0nr03726e] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Lactic acidosis is one of the key characteristics of the tumor microenvironment (TME), and plays a critical role in therapy resistance, making it an attractive target for enhancing anticancer treatment. However, no effective systems exhibit the ability to selectively neutralize tumor lactic acidosis in a controlled manner. Here, we have developed novel ultrasound-responsive alkaline nanorobots (AN-DSP), composed of PLGA nanoparticles containing doxorubicin (DOX), sodium carbonate (Na2CO3) and perfluorocarbon (PFC), for recovering from lactic acidosis-mediated drug resistance. AN-DSP exhibit sensitive response to ultrasound stimulation, and rapidly release Na2CO3 to neutralize lactic acidosis, consequently enhancing DOX susceptibility in vitro and in vivo. Particularly, our nanorobots autonomously accumulate in tumors by an enhanced permeability and retention effect, and can specifically disrupt the tumor acidic microenvironment in response to external ultrasonic powering, resulting in the inhibition of tumor growth with minimal adverse effects. Therefore, AN-DSP represent a promising approach for selectively overcoming tumor lactic acidosis induced therapeutic resistance.
Collapse
Affiliation(s)
- Xia Meng
- National Laboratory of Solid State Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, 210093, Nanjing, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Vaiserman A, Koliada A, Lushchak O. Neuroinflammation in pathogenesis of Alzheimer's disease: Phytochemicals as potential therapeutics. Mech Ageing Dev 2020; 189:111259. [PMID: 32450086 DOI: 10.1016/j.mad.2020.111259] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/02/2020] [Accepted: 05/02/2020] [Indexed: 02/07/2023]
Abstract
Accumulation of neurotoxic forms of amyloid-β proteins in senile plaques and hyperphosphorylated tau proteins in neurofibrillary tangles is a well-known pathophysiological hallmark of Alzheimer's disease (AD). However, clinical trials with drugs targeting amyloid-β and tau have failed to demonstrate efficacy in treating AD. All currently FDA-approved anti-AD drugs have symptomatic effects only and are not able to cure this disease. This makes necessary to search for alternative therapeutic targets. Accumulating evidence suggests that systemic inflammation and related vascular dysfunction play important etiological roles in AD and precede its clinical manifestation. Therefore, novel therapeutic modalities targeted at these pathophysiological components of AD are intensively developed now. Phytochemicals such as resveratrol, curcumin, quercetin, genistein and catechins are promising anti-AD therapeutics due to their ability to affect major pathogenetic mechanisms of AD, including oxidative stress, neuroinflammation and mitochondrial dysfunction. The implementation of innovative approaches for phytochemical delivery, including the nanotechnology-based ones which enable to significantly enhance their oral bioavailability, would likely provide an opportunity to address many challenges of conventional anti-AD therapies. In this review, roles of inflammation and vascular dysregulation in AD are described and phytobioactive compound-based treatment strategies for AD are discussed.
Collapse
Affiliation(s)
- Alexander Vaiserman
- Laboratory of Epigenetics, D.F. Chebotarev Institute of Gerontology, NAMS, 67 Vyshgorodska str., Kyiv, 04114, Ukraine.
| | - Alexander Koliada
- Laboratory of Epigenetics, D.F. Chebotarev Institute of Gerontology, NAMS, 67 Vyshgorodska str., Kyiv, 04114, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str., Ivano-Frankivsk, 76018, Ukraine
| |
Collapse
|
49
|
Kumar K, Kumar Shyamlal BR, Verma R, Kondaiah P, Chaudhary S. Reduction-Triggered Doxorubicin Delivery by Self-Assembled Nanospheres of Lipoylated Caffeine. ChemMedChem 2020; 15:733-737. [PMID: 32162419 DOI: 10.1002/cmdc.202000070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/06/2020] [Indexed: 01/09/2023]
Abstract
This study reports a new amphiphilic bioconjugate (CAFF-LA) derived from the lipoylation of a hydroxyethyl derivative of caffeine. In water, CAFF-LA self-assembles into nanospheres with an average size of 155 nm, as evidenced from dynamic light scattering and electron microscopy studies. The nanospheres are stable in serum and could be disintegrated upon exposure to the reducing environment of dithiothreitol (DTT; 10 mM) and glutathione (GSH; 10 mM). These nanospheres easily encapsulate the chemotherapy medication, doxorubicin (DOX), and demonstrate an efficacious transport into doxorubicin-resistant cervical cancer (HeLa) cells, wherein a marked induction in apoptosis and significantly lower IC50 have been observed when compared to that of free drug. The in vitro assessment of cell viability and hemocompatibility present these nanospheres as potentially safe and efficient intracellular reduction stimulus-responsive drug-delivery vehicles.
Collapse
Affiliation(s)
- Krishan Kumar
- Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur, 302017, India
| | - Bharti Rajesh Kumar Shyamlal
- Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur, 302017, India
| | - Rajbala Verma
- Department of Zoology, University of Rajasthan, Jaipur, 302004, India
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, 560012, India
| | - Sandeep Chaudhary
- Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur, 302017, India
| |
Collapse
|
50
|
Huang Y, Zhang W, Xu Y, Zhu S, Wu Y, Chen T, Xiao Y, Lu W, Zhang X, Yu J. Dynamic core crosslinked camptothecin prodrug micelles with reduction sensitivity and boronic acid-mediated enhanced endocytosis: An intelligent tumor-targeted delivery nanoplatform. Int J Pharm 2020; 580:119250. [PMID: 32209369 DOI: 10.1016/j.ijpharm.2020.119250] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/14/2020] [Accepted: 03/20/2020] [Indexed: 12/31/2022]
Abstract
The physicochemical properties of camptothecin (CPT) limit its clinical application. To maximize drug efficacy, a novel intelligent prodrug delivery nanoplatform with a tumor microenvironment-cleavable core crosslinking strategy was proposed based on a phenylboronic acid (PBA) modified polyethylene glycol (PEG)-polyglutamic acid (PGlu) polymer with disulfide-bonded CPT, called PBA-PEG-P(Glu-co-GlussCPT). The fabricated nanoplatform was a spherical micelle that could withstand dilution and carry a large number of therapeutic molecules to the tumor tissues, thereby minimizing premature drug release. Moreover, the nanoplatform release 6.2 ± 0.62, 12.4 ± 1.8, 46.7 ± 0.33, and 79.2 ± 1.58% of CPT after incubation in 0.02, 1, 5, and 10 mM dithiothreitol for 24 h, respectively, exhibiting good reduction-sensitivity. Moreover, the nanoplatform exhibited significant antiproliferative activity against tumor cells. In addition, with PBA modification, the nanoplatform demonstrated enhanced endocytosis efficiency. This prodrug nanoplatform also exhibited significant in vivo antitumor efficacy on both murine and human hepatoma xenograft models, without showing significant systemic toxicity but demonstrating good biocompatibility. In other words, this novel intelligent prodrug delivery nanoplatform with tumor microenvironment-cleavable core crosslinking strategy and active targeting strategy based on prodrug polymer PBA-PEG-P(Glu-co-GlussCPT) demonstrated multiple functions and significant potential for antitumor drug delivery.
Collapse
Affiliation(s)
- Yushu Huang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Wanli Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Yanyun Xu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Shulei Zhu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Yanqian Wu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Tiandong Chen
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Yi Xiao
- Department of Radiology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, PR China
| | - Wei Lu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Xiongwen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China
| | - Jiahui Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, PR China.
| |
Collapse
|