1
|
Ali I, Adil M, Imran M, Qureshi SA, Qureshi S, Hasan N, Ahmad FJ. Nanotechnology in Parkinson's Disease: overcoming drug delivery challenges and enhancing therapeutic outcomes. Drug Deliv Transl Res 2025:10.1007/s13346-025-01799-8. [PMID: 39878857 DOI: 10.1007/s13346-025-01799-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
The global prevalence of Parkinson's Disease (PD) is on the rise, driven by an ageing population and ongoing environmental conditions. To gain a better understanding of PD pathogenesis, it is essential to consider its relationship with the ageing process, as ageing stands out as the most significant risk factor for this neurodegenerative condition. PD risk factors encompass genetic predisposition, exposure to environmental toxins, and lifestyle influences, collectively increasing the chance of PD development. Moreover, early and precise PD diagnosis remains elusive, relying on clinical assessments, neuroimaging techniques, and emerging biomarkers. Conventional management of PD involves dopaminergic medications and surgical interventions, but these treatments often become less effective over time and do not address disease treatment. Challenges persist due to the blood-brain barrier's (BBB) impermeability, hindering drug delivery. Recent advancements in nanotechnology offer promising novel approaches for PD management. Various drug delivery systems (DDS), including nanosized polymers, lipid-based carriers, and nanoparticles (such as metal/metal oxide, protein, and carbonaceous particles), aim to enhance drug and gene delivery. These modifications seek to improve BBB permeability, ultimately benefiting PD patients. This review underscores the critical role of ageing in PD development and explores how age-related neuronal decline contributes to substantia nigra loss and PD manifestation in susceptible individuals. The review also highlights the advancements and ongoing challenges in nanotechnology-based therapies for PD.
Collapse
Affiliation(s)
- Irfan Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammad Adil
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammad Imran
- Faculty of Medicine, Frazer Institute, University of Queensland, Brisbane, 4102, Australia
| | - Saba Asif Qureshi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Saima Qureshi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Nazeer Hasan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Farhan Jalees Ahmad
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
2
|
Thangavelu L, Imran M, Alsharari SH, Abdulaziz AM, Alawlaqi AM, Kamal M, Rekha MM, Kaur M, Soothwal P, Arora I, Kumar MR, Chauhan AS. Exploring hypoxia-induced ncRNAs as biomarkers and therapeutic targets in lung cancer. Pathol Res Pract 2024; 263:155613. [PMID: 39383737 DOI: 10.1016/j.prp.2024.155613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/02/2024] [Accepted: 09/24/2024] [Indexed: 10/11/2024]
Abstract
Lung cancer is a deadly disease, causing nearly 20 % of all cancer deaths globally. A key factor in lung cancer's development and resistance to treatment is hypoxia, a condition where tumor cells experience low oxygen levels. In this low-oxygen environment, special molecules called non-coding RNAs (ncRNAs) become critical players. NcRNAs, including lncRNAs, miRNAs, circRNAs, and siRNAs, control how genes function and how cells behave. Some ncRNAs, like HIF1A-AS2 and HOTAIR, are linked to the aggressive spread of lung cancer, making them potential targets for therapy. Others, like certain miRNAs, show promise as early detection tools due to their influence on tumor blood vessel formation and metabolism. This complex interplay between hypoxia and ncRNAs is crucial for understanding lung cancer. For example, circRNAs can control the activity of miRNAs, impacting how tumors respond to low oxygen. Additionally, siRNAs offer a potential strategy to overcome treatment resistance caused by hypoxia. By studying the intricate relationship between hypoxia and ncRNAs, scientists hope to uncover new biomarkers for lung cancer. This knowledge will pave the way for developing more effective and targeted treatments for this devastating disease.
Collapse
Affiliation(s)
- Lakshmi Thangavelu
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | | | - Akrm M Abdulaziz
- Department of Clinical Pharmacy, King Khalid Hospital, Najran 66262, Saudi Arabia
| | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mandeep Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Pradeep Soothwal
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Isha Arora
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, India.
| |
Collapse
|
3
|
Liu Z, Liu X, Zhang W, Gao R, Wei H, Yu CY. Current advances in modulating tumor hypoxia for enhanced therapeutic efficacy. Acta Biomater 2024; 176:1-27. [PMID: 38232912 DOI: 10.1016/j.actbio.2024.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/08/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Hypoxia is a common feature of most solid tumors, which promotes the proliferation, invasion, metastasis, and therapeutic resistance of tumors. Researchers have been developing advanced strategies and nanoplatforms to modulate tumor hypoxia to enhance therapeutic effects. A timely review of this rapidly developing research topic is therefore highly desirable. For this purpose, this review first introduces the impact of hypoxia on tumor development and therapeutic resistance in detail. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are also systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We provide a detailed discussion of the rationale and research progress of these strategies. Through a review of current trends, it is hoped that this comprehensive overview can provide new prospects for clinical application in tumor treatment. STATEMENT OF SIGNIFICANCE: As a common feature of most solid tumors, hypoxia significantly promotes tumor progression. Advanced nanoplatforms have been developed to modulate tumor hypoxia to enhanced therapeutic effects. In this review, we first introduce the impact of hypoxia on tumor progression. Current developments in the construction of various nanoplatforms to enhance tumor treatment in response to hypoxia are systematically summarized, including hypoxia-overcoming, hypoxia-exploiting, and hypoxia-disregarding strategies. We discuss the rationale and research progress of the above strategies in detail, and finally introduce future challenges for treatment of hypoxic tumors. By reviewing the current trends, this comprehensive overview can provide new prospects for clinical translatable tumor therapy.
Collapse
Affiliation(s)
- Zihan Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xinping Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Wei Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ruijie Gao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
4
|
Yukawa H, Sato K, Baba Y. Theranostics applications of quantum dots in regenerative medicine, cancer medicine, and infectious diseases. Adv Drug Deliv Rev 2023; 200:114863. [PMID: 37156265 DOI: 10.1016/j.addr.2023.114863] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/10/2023]
Abstract
Quantum dots (QDs) have attracted attention for their application and commercialization in all industrial fields, including communications, displays, and solar cells, due to their excellent optical properties based on the quantum size effect. In recent years, the development of QDs that do not contain cadmium which is toxic to cells and living organisms, has progressed, and they have attracted considerable attention in the bio-imaging field for targeting molecules and cells. Furthermore, recently, the need for diagnostics and treatment at the single molecule and single cell level in the medical field has been increasing, and the application of QDs in the medical field is also accelerating. Therefore, this paper outlines the frontiers of diagnostic and therapeutic applications (theranostics) of QDs, especially in advanced medical fields such as regenerative medicine, oncology, and infectious diseases.
Collapse
Affiliation(s)
- Hiroshi Yukawa
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Nagoya University, Tsurumai-cho 65, Showa-ku, Nagoya 466-8550, Japan; Development of Quantum-nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Tsurumai 65, Showa-ku, Nagoya 466-8550, Japan; Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan; Department of Quantum Life Science, Graduate School of Science, Chiba University, Chiba 265-8522, Japan.
| | - Kazuhide Sato
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Nagoya University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 Unit, Nagoya University, Tsurumai-cho 65, Showa-ku, Nagoya 466-8550, Japan; Development of Quantum-nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Tsurumai 65, Showa-ku, Nagoya 466-8550, Japan; Nagoya University Graduate School of Medicine, 65 Tsuruma, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshinobu Baba
- Institute of Nano-Life-Systems, Institutes of Innovation for Future Society, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8603, Japan; Development of Quantum-nano Cancer Photoimmunotherapy for Clinical Application of Refractory Cancer, Nagoya University, Tsurumai 65, Showa-ku, Nagoya 466-8550, Japan; Institute of Quantum Life Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan.
| |
Collapse
|
5
|
Su MC, Nethi SK, Dhanyamraju PK, Prabha S. Nanomedicine Strategies for Targeting Tumor Stroma. Cancers (Basel) 2023; 15:4145. [PMID: 37627173 PMCID: PMC10452920 DOI: 10.3390/cancers15164145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The tumor stroma, or the microenvironment surrounding solid tumors, can significantly impact the effectiveness of cancer therapies. The tumor microenvironment is characterized by high interstitial pressure, a consequence of leaky vasculature, and dense stroma created by excessive deposition of various macromolecules such as collagen, fibronectin, and hyaluronic acid (HA). In addition, non-cancerous cells such as cancer-associated fibroblasts (CAFs) and the extracellular matrix (ECM) itself can promote tumor growth. In recent years, there has been increased interest in combining standard cancer treatments with stromal-targeting strategies or stromal modulators to improve therapeutic outcomes. Furthermore, the use of nanomedicine, which can improve the delivery and retention of drugs in the tumor, has been proposed to target the stroma. This review focuses on how different stromal components contribute to tumor progression and impede chemotherapeutic delivery. Additionally, this review highlights recent advancements in nanomedicine-based stromal modulation and discusses potential future directions for developing more effective stroma-targeted cancer therapies.
Collapse
Affiliation(s)
- Mei-Chi Su
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA;
| | - Susheel Kumar Nethi
- Nanovaccine Institute, Department of Chemical & Biological Engineering, Iowa State University, Ames, IA 50011, USA;
| | - Pavan Kumar Dhanyamraju
- Fels Cancer Institute of Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
| | - Swayam Prabha
- Fels Cancer Institute of Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA;
- Department of Cancer and Cellular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Temple University, Philadelphia, PA 19111, USA
| |
Collapse
|
6
|
Zhang J, Tang K, Fang R, Liu J, Liu M, Ma J, Wang H, Ding M, Wang X, Song Y, Yang D. Nanotechnological strategies to increase the oxygen content of the tumor. Front Pharmacol 2023; 14:1140362. [PMID: 36969866 PMCID: PMC10034070 DOI: 10.3389/fphar.2023.1140362] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
Hypoxia is a negative prognostic indicator of solid tumors, which not only changes the survival state of tumors and increases their invasiveness but also remarkably reduces the sensitivity of tumors to treatments such as radiotherapy, chemotherapy and photodynamic therapy. Thus, developing therapeutic strategies to alleviate tumor hypoxia has recently been considered an extremely valuable target in oncology. In this review, nanotechnological strategies to elevate oxygen levels in tumor therapy in recent years are summarized, including (I) improving the hypoxic tumor microenvironment, (II) oxygen delivery to hypoxic tumors, and (III) oxygen generation in hypoxic tumors. Finally, the challenges and prospects of these nanotechnological strategies for alleviating tumor hypoxia are presented.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Kaiyuan Tang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Runqi Fang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Jiaming Liu
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Ming Liu
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Jiayi Ma
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Hui Wang
- School of Fundamental Sciences, Bengbu Medical College, Bengbu, China
| | - Meng Ding
- Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Meng Ding, ; Xiaoxiao Wang, ; Dongliang Yang,
| | - Xiaoxiao Wang
- Biochemical Engineering Research Center, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma’anshan, China
- *Correspondence: Meng Ding, ; Xiaoxiao Wang, ; Dongliang Yang,
| | - Yanni Song
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, China
- *Correspondence: Meng Ding, ; Xiaoxiao Wang, ; Dongliang Yang,
| |
Collapse
|
7
|
Xia Y, Duan S, Han C, Jing C, Xiao Z, Li C. Hypoxia-responsive nanomaterials for tumor imaging and therapy. Front Oncol 2022; 12:1089446. [PMID: 36591450 PMCID: PMC9798000 DOI: 10.3389/fonc.2022.1089446] [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: 11/04/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Hypoxia is an important component of tumor microenvironment and plays a pivotal role in cancer progression. With the distinctive physiochemical properties and biological effects, various nanoparticles targeting hypoxia had raised great interest in cancer imaging, drug delivery, and gene therapy during the last decade. In the current review, we provided a comprehensive view on the latest progress of novel stimuli-responsive nanomaterials targeting hypoxia-tumor microenvironment (TME), and their applications in cancer diagnosis and therapy. Future prospect and challenges of nanomaterials are also discussed.
Collapse
Affiliation(s)
- Yifei Xia
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shao Duan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chaozhe Han
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengwei Jing
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zunyu Xiao
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China,*Correspondence: Chao Li, ; Zunyu Xiao,
| | - Chao Li
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China,*Correspondence: Chao Li, ; Zunyu Xiao,
| |
Collapse
|
8
|
Design of Nanoparticles in Cancer Therapy Based on Tumor Microenvironment Properties. Pharmaceutics 2022; 14:pharmaceutics14122708. [PMID: 36559202 PMCID: PMC9785496 DOI: 10.3390/pharmaceutics14122708] [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: 10/19/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide, and battling cancer has always been a challenging subject in medical sciences. All over the world, scientists from different fields of study try to gain a deeper knowledge about the biology and roots of cancer and, consequently, provide better strategies to fight against it. During the past few decades, nanoparticles (NPs) have attracted much attention for the delivery of therapeutic and diagnostic agents with high efficiency and reduced side effects in cancer treatment. Targeted and stimuli-sensitive nanoparticles have been widely studied for cancer therapy in recent years, and many more studies are ongoing. This review aims to provide a broad view of different nanoparticle systems with characteristics that allow them to target diverse properties of the tumor microenvironment (TME) from nanoparticles that can be activated and release their cargo due to the specific characteristics of the TME (such as low pH, redox, and hypoxia) to nanoparticles that can target different cellular and molecular targets of the present cell and molecules in the TME.
Collapse
|
9
|
Lv SY, He S, Ling XL, Wang YQ, Huang C, Long JR, Wang JQ, Qin Y, Wei H, Yu CY. Review of lipoic acid: From a clinical therapeutic agent to various emerging biomaterials. Int J Pharm 2022; 627:122201. [PMID: 36115465 DOI: 10.1016/j.ijpharm.2022.122201] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/20/2022] [Accepted: 09/09/2022] [Indexed: 01/25/2023]
Abstract
Lipoic acid (LA), an endogenous small molecule in organisms, has been extensively used for the highly efficient clinical treatment of malignant diseases, which include diabetes, Alzheimer's disease, and cancer over the past seven decades. Tremendous progresses have been made on the use of LA in nanomedicine for the development of various biomaterials because of its unique biological properties and highly adaptable structure since the first discovery. However, there are few reviews thus far, to our knowledge, summarizing this hot subject of research of LA and its derived biomaterials. For this purpose, we present herein the first comprehensive summary on the design and development of LA and its derived materials for biomedical applications. This review first discusses the therapeutic use of LA followed by the description of synthesis and preclinical study of LA-derived-small molecules. The applications of various LA and poly (lipoic acid) (PLA)-derived-biomaterials are next summarized in detail with an emphasis on the use of LA for the design of biomaterials and the diverse properties. This review describes the development of LA from a clinical therapeutic agent to a building unit of various biomaterials field, which will promote the further discovery of new therapeutic uses of LA as therapeutic agents and facile development of LA-based derivates with greater performance for biomedical applications.
Collapse
Affiliation(s)
- Shao-Yang Lv
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Suisui He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xiao-Li Ling
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yue-Qin Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Cong Huang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Jin-Rong Long
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Jia-Qi Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yang Qin
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
10
|
Chen Z, Yue Z, Wang R, Yang K, Li S. Nanomaterials: A powerful tool for tumor immunotherapy. Front Immunol 2022; 13:979469. [PMID: 36072591 PMCID: PMC9441741 DOI: 10.3389/fimmu.2022.979469] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Cancer represents the leading global driver of death and is recognized as a critical obstacle to increasing life expectancy. In recent years, with the development of precision medicine, significant progress has been made in cancer treatment. Among them, various therapies developed with the help of the immune system have succeeded in clinical treatment, recognizing and killing cancer cells by stimulating or enhancing the body’s intrinsic immune system. However, low response rates and serious adverse effects, among others, have limited the use of immunotherapy. It also poses problems such as drug resistance and hyper-progression. Fortunately, thanks to the rapid development of nanotechnology, engineered multifunctional nanomaterials and biomaterials have brought breakthroughs in cancer immunotherapy. Unlike conventional cancer immunotherapy, nanomaterials can be rationally designed to trigger specific tumor-killing effects. Simultaneously, improved infiltration of immune cells into metastatic lesions enhances the efficiency of antigen submission and induces a sustained immune reaction. Such a strategy directly reverses the immunological condition of the primary tumor, arrests metastasis and inhibits tumor recurrence through postoperative immunotherapy. This paper discusses several types of nanoscale biomaterials for cancer immunotherapy, and they activate the immune system through material-specific advantages to provide novel therapeutic strategies. In summary, this article will review the latest advances in tumor immunotherapy based on self-assembled, mesoporous, cell membrane modified, metallic, and hydrogel nanomaterials to explore diverse tumor therapies.
Collapse
Affiliation(s)
- Ziyin Chen
- Clinical Medicine, Harbin Medical University, Harbin, China
| | - Ziqi Yue
- Department of Forensic Medicine, Harbin Medical University, Harbin, China
| | - Ronghua Wang
- Department of Outpatient, Dongying People’s Hospital, Dongying, China
| | - Kaiqi Yang
- Clinical Medicine, Harbin Medical University, Harbin, China
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
- *Correspondence: Shenglong Li, ;
| |
Collapse
|
11
|
Fu Z, Wang X, Lu X, Yang Y, Zhao L, Zhou L, Wang K, Fu H. Mannose-decorated ginsenoside Rb1 albumin nanoparticles for targeted anti-inflammatory therapy. Front Bioeng Biotechnol 2022; 10:962380. [PMID: 36046677 PMCID: PMC9420840 DOI: 10.3389/fbioe.2022.962380] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/04/2022] [Indexed: 11/29/2022] Open
Abstract
Ginsenoside Rb1 is a potential anti-inflammatory natural molecule, but its therapeutic efficacy was tremendously hampered by the low solubility and non-targeted delivery. In this study, we innovatively developed a mannose (Man)-modified albumin bovine serum albumin carrier (Man-BSA) to overcome the previously mentioned dilemmas of Rb1. The constructed Man-BSA@Rb1 NPs could improve the solubility and increase the cellular uptake of Rb1, finally leading to the enhanced anti-inflammatory effects. The robust therapeutics of Man-BSA@Rb1 NPs were measured in terms of nitrite, tumor necrosis factor-α (TNF-α), and interleukin-6 (IL-6) levels, which might be achieved by potently inhibiting nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways in lipopolysaccharide (LPS)-induced Raw264.7 cells. Moreover, the therapeutic efficacy of Man-BSA@Rb1 NPs was further confirmed in the d-Gal/LPS-induced liver injury model. The results indicated that Man-BSA may offer a promising system to improve the anti-inflammatory therapy of Rb1.
Collapse
Affiliation(s)
- Zhihui Fu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaohui Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuan Lu
- School of Pharmacy and Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Ying Yang
- School of Pharmacy and Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Lingling Zhao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Zhou
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kaikai Wang
- School of Pharmacy and Affiliated Hospital of Nantong University, Nantong University, Nantong, China
- *Correspondence: Kaikai Wang, ; Hanlin Fu,
| | - Hanlin Fu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Kaikai Wang, ; Hanlin Fu,
| |
Collapse
|
12
|
Yan Y, Li H, Yao H, Cheng X. Nanodelivery Systems Delivering Hypoxia-Inducible Factor-1 Alpha Short Interfering RNA and Antisense Oligonucleotide for Cancer Treatment. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.932976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hypoxia-inducible factor (HIF), which plays a crucial role in oxygen homeostasis, contributes to immunosuppression, tumor angiogenesis, multidrug resistance, photodynamic therapy resistance, and metastasis. HIF as a therapeutic target has attracted scientists’ strong academic research interests. Short interfering RNA (siRNA) and antisense oligonucleotide (ASO) are the more promising and broadly utilized methods for oligonucleotide-based therapy. Their physicochemical characteristics such as hydrophilicity, negative charge, and high molecular weight make them impossible to cross the cell membrane. Moreover, siRNA and ASO are subjected to a rapid deterioration in circulation and cannot translocate into nuclear. Delivery of siRNA and ASO to specific gene targets should be realized without off-target gene silencing and affecting the healthy cells. Nanoparticles as vectors for delivery of siRNA and ASO possess great advantages and flourish in academic research. In this review, we summarized and analyzed regulation mechanisms of HIF under hypoxia, the significant role of HIF in promoting tumor progression, and recent academic research on nanoparticle-based delivery of HIF siRNA and ASO for cancer immunotherapy, antiangiogenesis, reversal of multidrug resistance and radioresistance, potentiating photodynamic therapy, inhibiting tumor metastasis and proliferation, and enhancing apoptosis are reviewed in this thesis. Furthermore, we hope to provide some rewarding suggestions and enlightenments for targeting HIF gene therapy.
Collapse
|
13
|
Huang H, Shao L, Chen Y, Han W, Zhou Y, Liu T, Gu J, Zhu H. Sequential Dual Delivery System Based on siCOX-2-Loaded Gold Nanostar and Thermal-Sensitive Liposomes Overcome Hypoxia-Mediated Multidrug Resistance in Tumors. Mol Pharm 2022; 19:2390-2405. [PMID: 35639669 DOI: 10.1021/acs.molpharmaceut.2c00164] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Reversing hypoxia-mediated multidrug resistance (MDR) presents a unique challenge in clinical chemotherapy. Here, a sequential dual delivery system composited with Cyclooxygenase-2 siRNA (siCOX-2) in poly-d-arginine (9R)/2-deoxyglucose (DG)-loaded gold nanostar (GNS) (siCOX-2@RDG) and paclitaxel (PTX)-loaded thermosensitive liposome (PTSL) was proposed to conquer the hypoxia-mediated MDR in tumors. As a result, the prepared siCOX-2@RDG exhibited a starlike morphology with a uniform particle size of 194.36 ± 1.44 nm and a ζ-potential of -11.83 ± 2.01 mV. In vitro, PTSL displayed expected thermal-responsive release properties. As expected, siCOX-2@RDG displayed exceptional DG-mediated hypoxia-targeting capability both in vitro and in vivo and downregulated the expression of COX-2 successfully. Meanwhile, GNS-triggered hyperthermia elevated the cellular uptake of PTSL in PTX-resistant HepG2(HepG2/PTX) cells in vitro and enhanced the permeability of tumor tissues, thus elevating the valid retention of PTX into solid tumors. Finally, we demonstrated that the sequential dual systems composed of siCOX-2@RDG and PTSL could reverse hypoxia-mediated MDR and exhibit excellent synergistic antitumor effects both in vitro and in vivo, prolonging the survival of tumor-bearing mice. The devised sequential dual systems, composed of two independent nanosystems, have a promising potential to overcome hypoxia-mediated MDR in clinical practice.
Collapse
Affiliation(s)
- Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Lanlan Shao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yan Chen
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Weili Han
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yao Zhou
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
| | - Jinhua Gu
- Department of Clinical Pharmacy, The Affiliated Maternal and Child Health Hospital of Nantong University/Nantong Children's Hospital, Nantong 226001, China
| | - Hongyan Zhu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
14
|
Emerging Nanotherapeutic Approaches to Overcome Drug Resistance in Cancers with Update on Clinical Trials. Pharmaceutics 2022; 14:pharmaceutics14040866. [PMID: 35456698 PMCID: PMC9028322 DOI: 10.3390/pharmaceutics14040866] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
A key issue with modern cancer treatments is the emergence of resistance to conventional chemotherapy and molecularly targeted medicines. Cancer nanotherapeutics were created in order to overcome the inherent limitations of traditional chemotherapeutics. Over the last few decades, cancer nanotherapeutics provided unparalleled opportunities to understand and overcome drug resistance through clinical assessment of rationally designed nanoparticulate delivery systems. In this context, various design strategies such as passive targeting, active targeting, nano-drug, and multimodal nano-drug combination therapy provided effective cancer treatment. Even though cancer nanotherapy has made great technological progress, tumor biology complexity and heterogeneity and a lack of comprehensive knowledge of nano-bio interactions remain important roadblocks to future clinical translation and commercialization. The current developments and advancements in cancer nanotherapeutics employing a wide variety of nanomaterial-based platforms to overcome cancer treatment resistance are discussed in this article. There is also a review of various nanotherapeutics-based approaches to cancer therapy, including targeting strategies for the tumor microenvironment and its components, advanced delivery systems for specific targeting of cancer stem cells (CSC), as well as exosomes for delivery strategies, and an update on clinical trials. Finally, challenges and the future perspective of the cancer nanotherapeutics to reverse cancer drug resistance are discussed.
Collapse
|
15
|
Zhang X, He C, Xiang G. Engineering nanomedicines to inhibit hypoxia-inducible Factor-1 for cancer therapy. Cancer Lett 2022; 530:110-127. [PMID: 35041892 DOI: 10.1016/j.canlet.2022.01.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/18/2021] [Accepted: 01/10/2022] [Indexed: 11/02/2022]
Abstract
Hypoxia-inducible factor-1 (HIF-1), an essential promoter of tumor progression, has attracted increasing attention as a therapeutic target. In addition to hypoxic cellular conditions, HIF-1 activation can be triggered by cancer treatment, which causes drug tolerance and therapeutic failure. To date, a series of effective strategies have been explored to suppress HIF-1 function, including silencing the HIF-1α gene, inhibiting HIF-1α protein translation, degrading HIF-1α protein, and inhibiting HIF-1 transcription. Furthermore, nanoparticle-based drug delivery systems have been widely developed to improve the stability and pharmacokinetics of HIF-1 inhibitors or achieve HIF-1-targeted combination therapies as a nanoplatform. In this review, we summarize the current literature on nanomedicines targeting HIF-1 to combat cancer and discuss their potential for future development.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chuanchuan He
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
16
|
Zhou M, Yuan M, Zhang M, Lei C, Aras O, Zhang X, An F. Combining histone deacetylase inhibitors (HDACis) with other therapies for cancer therapy. Eur J Med Chem 2021; 226:113825. [PMID: 34562854 DOI: 10.1016/j.ejmech.2021.113825] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/30/2022]
Abstract
Histone deacetylases (HDACs) play an important role in regulating the expression of genes involved in tumorigenesis and tumor maintenance, and hence they have been considered as key targets in cancer therapy. As a novel category of antitumor agents, histone deacetylase inhibitors (HDACis) can induce cell cycle arrest, apoptosis, and differentiation in cancer cells, ultimately combating cancer. Although in the United States, the use of HDACis for the treatment of certain cancers has been approved, the therapeutic efficacy of HDACis as a single therapeutic agent in solid tumorshas been unsatisfactory and drug resistance may yet occur. To enhance therapeutic efficacy and limit drug resistance, numerous combination therapies involving HDACis in synergy with other antitumor therapies have been studied. In this review, we describe the classification of HDACs. Moreover, we summarize the antitumor mechanism of the HDACis for targeting key cellular processes of cancers (cell cycle, apoptosis, angiogenesis, DNA repair, and immune response). In addition, we outline the major developments of other antitumor therapies in combination with HDACis, including chemotherapy, radiotherapy, phototherapy, targeted therapy, and immunotherapy. Finally, we discuss the current state and challenges of HDACis-drugs combinations in future clinical studies, with the aim of optimizing the antitumor effect of such combinations.
Collapse
Affiliation(s)
- Mengjiao Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Minjian Yuan
- Department of Pharmacology, School of Pharmacy, Nantong University, 226000, Nantong, Jiangsu, PR China
| | - Meng Zhang
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Chenyi Lei
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, United States
| | - Xiaohong Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, PR China.
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, No.76 Yanta West Road, Xi'an, 710061, Shaanxi, People's Republic of China; Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, 199 Ren'ai Road, Suzhou, 215123, Jiangsu, PR China.
| |
Collapse
|
17
|
Sun Y, Zhou Z, Yang S, Yang H. Modulating hypoxia inducible factor-1 by nanomaterials for effective cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1766. [PMID: 34713633 DOI: 10.1002/wnan.1766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/18/2022]
Abstract
Hypoxia, which is induced by abnormal tumor growth when it outstrips its oxygen supply, is a major character of cancer. The reaction of cells against hypoxia is mainly concentrated on the hypoxia-induced transcription factors (HIFs), especially HIF-1, which remain stabilized during hypoxia. Additionally, the oxygen-independent mechanism of regulating HIF-1 acts a vital part in different stages of tumor progression as well as chemo-/radio-/PDT resistance, resulting in poor curative effects and prognosis. In this review, we will outline the up-to-date information about how HIF-1 interferes with tumor metastasis and therapy resistance, followed by a detailed introduction of motivating techniques based on various nanomaterials to interfere with HIF signaling for effective cancer therapy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yun Sun
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| | - Zhiguo Zhou
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| | - Shiping Yang
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| | - Hong Yang
- The Education Ministry Key Lab of Resource Chemistry, Joint International Research Laboratory of Resource Chemistry, Ministry of Education, and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai, China
| |
Collapse
|
18
|
Recent advances in active targeting of nanomaterials for anticancer drug delivery. Adv Colloid Interface Sci 2021; 296:102509. [PMID: 34455211 DOI: 10.1016/j.cis.2021.102509] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/24/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022]
Abstract
One of the challenges in cancer chemotherapy is the low target to non-target ratio of therapeutic agents which incur severe adverse effect on the healthy tissues. In this regard, nanomaterials have tremendous potential for impacting cancer therapy by altering the toxicity profile of the drug. Some of the striking advantages provided by the nanocarriers mediated targeted drug delivery are relatively high build-up of drug concentration at the tumor site, improved drug content in the formulation and enhanced colloidal stability. Further, nanocarriers with tumor-specific moieties can be targeted to the cancer cell through cell surface receptors, tumor antigens and tumor vasculatures with high affinity and accuracy. Moreover, it overcomes the bottleneck of aimless drug biodistribution, undesired toxicity and heavy dosage of administration. This review discusses the recent developments in active targeting of nanomaterials for anticancer drug delivery through cancer cell surface targeting, organelle specific targeting and tumor microenvironment targeting strategies. Special emphasis has been given towards cancer cell surface and organelle specific targeting as delivery of anticancer drugs through these routes have made paradigm change in cancer management. Further, the current challenges and future prospects of nanocarriers mediated active drug targeting are also demonstrated.
Collapse
|
19
|
Zhang S, Gan Y, Shao L, Liu T, Wei D, Yu Y, Guo H, Zhu H. Virus Mimetic Shell-Sheddable Chitosan Micelles for siVEGF Delivery and FRET-Traceable Acid-Triggered Release. ACS APPLIED MATERIALS & INTERFACES 2020; 12:53598-53614. [PMID: 33201664 DOI: 10.1021/acsami.0c13023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Targeting vascular endothelial growth factor (VEGF) using small interfering RNA (siVEGF) has shown great potential in inhibiting the growth, proliferation, and migration of tumors by reducing the proliferation of blood vessels. On the basis of bionic principles, a novel pH-responsive and virus mimetic shell-sheddable chitosan (CS) micelles (CMs) as siRNA delivery system was introduced in this study. The cyclo(Arg-Gly-Asp-d-Phe-Lys) (cRGD) modified poly(enthylene glycol) (PEG) was conjugated to the HA2 modified chitosan via a hydrazone linkage (cRGD-PEG-Hz-CS-HA2). The cRGD-PEG-Hz-CS-HA2 conjugate could form micelles by interacting with the complex of octanal, Boc-l-lysine, and 9-d-arginine (9R) (octyl-Lys-9R) as a hydrophodic core forming agent, termed as cRGD-PEG-Hz-CS-HA2/octyl-Lys-9R (abbreviated as cRGD/HA2/Hz-CMs).The CMs modified with cRGD can accurately target glioma cells (U87MG cells) with high expression of αvβ3. The payloads of siVEGF were packed into the core of cRGD/HA2/Hz-CMs via electrostatic interaction and hydrophobic interaction. The intracellular cargo release was achieved by the pH-responsive lysis of the hydrazone bond in acidic environment of endosome. Moreover, the exposed HA2, as a pH-sensitive membrane-disruptive peptide, assists the escape of the carriers from endosome into cytosol. In addition, cRGD/HA2/Hz-CMs can effectively deliver siVEGF and silence VEGF gene expression in U87MG cells, leading to the significant tumor growth inhibition. This study demonstrates that cRGD/HA2/Hz-CMs can deliver and release siVEGF in a controlled manner, which was traced by the fluorescence resonance energy transfer (FRET) system in order to achieve RNAi-based anti-angiogenic treatment of cancer in vivo.
Collapse
Affiliation(s)
- Shengyu Zhang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
- Department of Pharmacy, Haimen People's Hospital, Nantong 226100, China
| | - Ye Gan
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Lanlan Shao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Tianqing Liu
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 1006, Australia
| | - Danyi Wei
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yanyan Yu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Hongwei Guo
- College of Pharmacy, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education &Center for Translational Medicine, Guangxi Medical University, Nanning 530021, China
| | - Hongyan Zhu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
20
|
Zhu G, Wang K, Qin H, Zhao X, Chen W, Xu L, Cao W, Guo H. Internal cross-linked polymeric nanoparticles with dual sensitivity for combination therapy of muscle-invasive bladder cancer. J Nanobiotechnology 2020; 18:124. [PMID: 32887622 PMCID: PMC7472706 DOI: 10.1186/s12951-020-00686-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/28/2020] [Indexed: 01/03/2023] Open
Abstract
Background Chemotherapy is a standard cancer treatment which uses anti-cancer drugs to destroy or slow the growth of cancer cells. However, chemotherapy has limited therapeutic effects in bladder cancer. One of the reasons of this resistance to chemotherapy is that higher levels of glutathione in invasive bladder cancer cells. We have fabricated nanoparticles that respond to high concentrations of glutathione and near-infrared laser irradiation in order to increase the drug accumulation at the tumor sites and combine chemotherapy with photothermal therapy to overcome the challenges of bladder cancer treatment. Methods The DOX&IR780@PEG-PCL-SS NPs were prepared by co-precipitation method. We investigated the tumor targeting capability of NPs in vitro and in vivo. The orthotopic bladder cancer model in C57BL/6 mice was established for in vivo study and the photothermal effects and therapeutic efficacy of NPs were evaluated. Results The DOX&IR780@PEG-PCL-SS NPs were synthesized using internal cross-linking strategy to increase the stability of nanoparticles. Nanoparticles can be ingested by tumor cells in a short time. The DOX&IR780@PEG-PCL-SS NPs have dual sensitivity to high levels of glutathione in bladder cancer cells and near-infrared laser irradiation. Glutathione triggers chemical structural changes of nanoparticles and preliminarily releases drugs, Near-infrared laser irradiation can promote the complete release of the drugs from the nanoparticles and induce a photothermal effect, leading to destroying the tumor cells. Given the excellent tumor-targeting ability and negligible toxicity to normal tissue, DOX&IR780@PEG-PCL-SS NPs can greatly increase the concentration of the anti-cancer drugs in tumor cells. The mice treated with DOX&IR780@PEG-PCL-SS NPs have a significant reduction in tumor volume. The DOX&IR780@PEG-PCL-SS NPs can be tracked by in vivo imaging system and have good tumor targeting ability, to facilitate our assessment during the experiment. Conclusion A nanoparticle delivery system with dual sensitivity to glutathione and near-infrared laser irradiation was developed for delivering IR780 and DOX. Chemo-photothermal synergistic therapy of both primary bladder cancer and their metastases was achieved using this advanced delivery system.![]()
Collapse
Affiliation(s)
- Guanchen Zhu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210009, China
| | - Kaikai Wang
- School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Haixiang Qin
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210009, China
| | - Xiaozhi Zhao
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210009, China
| | - Wei Chen
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210009, China
| | - Linfeng Xu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210009, China
| | - Wenmin Cao
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210009, China
| | - Hongqian Guo
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, 210009, China.
| |
Collapse
|
21
|
Phung CD, Tran TH, Pham LM, Nguyen HT, Jeong JH, Yong CS, Kim JO. Current developments in nanotechnology for improved cancer treatment, focusing on tumor hypoxia. J Control Release 2020; 324:413-429. [PMID: 32461115 DOI: 10.1016/j.jconrel.2020.05.029] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/12/2022]
Abstract
Hypoxia is a common feature of the tumor microenvironment, which is characterized by tissue oxygen deficiency due to an aggressive proliferation of cancer cells. Hypoxia activates hypoxia-inducible factor-dependent signaling, which in turn regulates metabolic reprogramming, immune suppression, resistance to apoptosis, angiogenesis, metastasis, and invasion to secondary sites. In this review, we provide an overview of the use of nanotechnology to harmonize intra-tumoral oxygen or suppress hypoxia-related signaling for an improved efficacy of cancer treatment. The biological background was followed by conducting a literature review on the (1) nanoparticles responsible for enhancing oxygen levels within the tumor, (2) nanoparticles sensitizing hypoxia, (3) nanoparticles suppressing hypoxia-inducing factor, (4) nanoparticles that relieve tumor hypoxia for enhancement of chemotherapy, photodynamic therapy, and immunotherapy, either individually or in combination. Lastly, the heterogeneity of cancer and limitations of nanotechnology are discussed to facilitate translational therapeutic treatment.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Yen Nghia, Ha Dong, Hanoi 12116, Viet Nam; PHENIKAA Research and Technology Institute (PRATI), A&A Green Phoenix Group JSC, No.167 Hoang Ngan, Trung Hoa, Cau Giay, Hanoi 11313, Viet Nam
| | - Le Minh Pham
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Hanh Thuy Nguyen
- Department of Industrial & Physical Pharmacy, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, United States
| | - Jee-Heon Jeong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
22
|
Zhao Y, Chen G, Meng Z, Gong G, Zhao W, Wang K, Liu T. A novel nanoparticle drug delivery system based on PEGylated hemoglobin for cancer therapy. Drug Deliv 2020; 26:717-723. [PMID: 31293178 PMCID: PMC6691922 DOI: 10.1080/10717544.2019.1639846] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Proteins such as albumin, gelatin, casein, transferrin, and collagen are widely used as drug delivery systems. However, only albumin-based paclitaxel (PTX) formulation Abraxane® (PTX-albumin NPs prepared by nab-technology) has been successfully developed for treating metastatic breast cancer clinically due to abundant materials, simple industrial scale-up process, and well tumor-targeting ability. Hemoglobin (Hb) is another protein used for drug delivery with similar advantages. In this study, we successfully synthesized PEG-Hb nanoparticles loading with PTX based on previously well-established acid-denatured method. PEG-Hb-PTX NPs showed enhanced cellular uptake and great cellular inhibition ability in vitro. Moreover, our animal study showed that PEGylated NPs greatly accumulated in tumor tissues and exhibited excellent anticancer activity in vivo. We found that PEG-Hb-PTX NPs possess a better in vivo antitumor effect than the commercially available Taxol® formulation. We believe that PEG-Hb has great potential as an efficient drug delivery system for further clinic study.
Collapse
Affiliation(s)
- Yongmei Zhao
- a School of Pharmacy, Nantong University , Nantong , China
| | - Gang Chen
- b Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University , Yangzhou , China
| | - Zhengjie Meng
- c College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University , Nanjing , China
| | - Guangming Gong
- d Department of Pharmaceutics , Jinling Hospital, Nanjing University School of Medicine , Nanjing , China
| | - Wei Zhao
- e School of Polymer Science and Engineering, Qingdao University of Science and Technology , Qingdao , China
| | - Kaikai Wang
- a School of Pharmacy, Nantong University , Nantong , China
| | - Tianqing Liu
- f QIMR Berghofer Medical Research Institute , Brisbane , Australia
| |
Collapse
|
23
|
Self-assembled PEI nanomicelles with a fluorinated core for improved siRNA delivery. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101403] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
24
|
Tumor microenvironment targeted nanotherapeutics for cancer therapy and diagnosis: A review. Acta Biomater 2020; 101:43-68. [PMID: 31518706 DOI: 10.1016/j.actbio.2019.09.009] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 08/09/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Recent findings suggest that the cellular and extracellular materials surrounding the cancerous cells from an atypical tumor microenvironment (TM) play a pivotal role in the process of tumor initiation and progression. TM comprises an intricate system involving diverse cell types including endothelial cells, pericytes, smooth muscle cells, fibroblasts, various inflammatory cells, dendritic cells, and cancer stem cells (CSCs). The TM-forming cells dynamically interact with the cancerous cells through various signaling mechanisms and pathways. The existence of this dynamic cellular communication is responsible for creating an environment suitable for sustaining a reasonably high cellular proliferation. Presently, researchers are showing interest to use these TM conditions to mediate effective targeting measures for cancer therapy. The use of nanotherapeutics-based combination therapy; stimuli-responsive nanotherapeutics targeting acidic pH, hypoxic environment; and nanoparticle-induced hyperthermia are some of the approaches that are under intense investigation for cancer therapy. This review discusses TM and its role in cancer progression and crosstalk understanding, opportunities, and epigenetic modifications involved therein to materialize the capability of nanotherapeutics to target cancer by availing TM. STATEMENT OF SIGNIFICANCE: This article presents various recent reports, proof-of-concept studies, patents, and clinical trials on the concept of tumor microenvironment for mediating the cancer-specific delivery of nanotechnology-based systems bearing anticancer drug and diagnostics. We highlight the potential of tumor microenvironment; its role in disease progression, opportunities, challenges, and allied treatment strategies for effective cancer therapy by conceptual understanding of tumor microenvironment and epigenetic modifications involved. Specifically, nanoparticle-based approaches to target various processes related to tumor microenvironment (pH responsive, hypoxic environment responsive, targeting of specific cells involved in tumor microenvironment, etc.) are dealt in detail.
Collapse
|
25
|
Combined Modality Therapy Based on Hybrid Gold Nanostars Coated with Temperature Sensitive Liposomes to Overcome Paclitaxel-Resistance in Hepatic Carcinoma. Pharmaceutics 2019; 11:pharmaceutics11120683. [PMID: 31847496 PMCID: PMC6969923 DOI: 10.3390/pharmaceutics11120683] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022] Open
Abstract
In this study, we prepared gold nanostar (GNS) composite nanoparticles containing siRNA of cyclooxygenase-2(siCOX-2) that were modified by tumor targeting ligand 2-deoxyglucose (DG) and transmembrane peptide 9-poly-D-arginine (9R) to form siCOX-2(9R/DG-GNS). Paclitaxel loaded temperature sensitive liposomes (PTX-TSL) were surface-modified to produce PTX-TSL-siCOX-2(9R/DG-GNS) displaying homogeneous star-shaped structures of suitable size (293.93 nm ± 3.21) and zeta potentials (2.47 mV ± 0.22). PTX-TSL-siCOX-2(9R/DG-GNS) had a high thermal conversion efficiency under 808 nm laser radiation and a superior transfection efficiency, which may be related to the targeting effects of DG and increased heat induced membrane permeability. COX-2 expression in HepG2/PTX cells was significantly suppressed by PTX-TSL-siCOX-2(9R/DG-GNS) in high temperatures. The co-delivery system inhibited drug-resistant cell growth rates by ≥77% and increased the cell apoptosis rate about 47% at elevated temperatures. PTX-TSL and siCOX-2 loaded gold nanostar particles, therefore, show promise for overcoming tumor resistance.
Collapse
|
26
|
Zhao L, Gu C, Gan Y, Shao L, Chen H, Zhu H. Exosome-mediated siRNA delivery to suppress postoperative breast cancer metastasis. J Control Release 2019; 318:1-15. [PMID: 31830541 DOI: 10.1016/j.jconrel.2019.12.005] [Citation(s) in RCA: 275] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 11/29/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
High recurrence and metastasis of triple-negative breast cancer (TNBC) after operation is a leading cause of breast cancer related death. The pre-metastatic niche (PMN) is an environment in a secondary organ conducive to the metastasis of a primary tumor. Herein, we identify exosomes from autologous breast cancer cells that show effective lung targeting ability. Based on this, we developed the biomimetic nanoparticles (cationic bovine serum albumin (CBSA) conjugated siS100A4 and exosome membrane coated nanoparticles, CBSA/siS100A4@Exosome) to improve drug delivery to the lung PMN. CBSA/siS100A4@Exosome self-assembled nanoparticles formed homogeneous sizes of ~200 nm, protected siRNA from degradation, and showed excellent biocompatibility. Further in vivo studies showed that CBSA/siS100A4@Exosome had a higher affinity toward lung in comparison to the CBSA/siS100A4@Liposome, and exhibited outstanding gene-silencing effects that significantly inhibited the growth of malignant breast cancer cells. Taken together, these results indicate that CBSA/siS100A4@Exosome self-assembled nanoparticles are a promising strategy to suppress postoperative breast cancer metastasis.
Collapse
Affiliation(s)
- Liuwan Zhao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Chunyan Gu
- Department of Pathology, Affiliated Nantong Third Hospital of Nantong University, Nantong 226006, China
| | - Ye Gan
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Lanlan Shao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Hongwei Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48108, USA.
| | - Hongyan Zhu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China.
| |
Collapse
|
27
|
Hypoxia-tropic nanozymes as oxygen generators for tumor-favoring theranostics. Biomaterials 2019; 230:119635. [PMID: 31767443 DOI: 10.1016/j.biomaterials.2019.119635] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/22/2019] [Accepted: 11/16/2019] [Indexed: 12/16/2022]
Abstract
Oxygen deficiency is the main obstacle of hypoxia-related theranostics, thus this is a considerable amount of research focusing on the development of methods to supply oxygen by taking advantage of hypoxia-responsive properties of nanoparticles. However, strategies to properly penetrate hypoxic regions by the nanoparticles remains an unmet challenge. In this work, a biomimetic nanozyme capable of possessing catalase-like activity and the efficient direct penetration of hypoxic areas in tumor tissues was developed to supply oxygen based on catalytic tumor microenvironment-responsive reaction, providing substantial tumor hypoxia relief with nearly 3-fold reduction compared to untreated tumor tissues. To demonstrate the advantages of the nanozymes in overcoming hypoxia, a theranostic nanosystem model composed of the core/shell nanozymes and aggregation-induced emission (AIE) molecules was designed. The nanosystem was able to present multi-modal imaging of tumors and modulated the tumor microenvironment for improved photodynamic therapy (PDT) by cascade reactions of therapeutic effector molecules, thereby providing significantly enhanced therapeutic benefits in inhibiting tumor growth and lung metastasis of orthotopic breast cancer. This conceptual study showed the multifaceted features of biomimetic nanozymes as tumor therapeutics and demonstrated the encouraging potential for modulating hypoxia as an application for tumor theranostics.
Collapse
|
28
|
Versatile electrostatically assembled polymeric siRNA nanovectors: Can they overcome the limits of siRNA tumor delivery? Int J Pharm 2019; 567:118432. [DOI: 10.1016/j.ijpharm.2019.06.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 11/20/2022]
|
29
|
Yu Y, Zhou Y, Zhu M, Liu M, Zhu H, Chen Y, Su G, Chen W, Peng H. Programming a split G-quadruplex in a DNA nanocage and its microRNA imaging in live cells. Chem Commun (Camb) 2019; 55:5131-5134. [PMID: 30973555 DOI: 10.1039/c9cc02096a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A novel approach to program target-responsive devices by incorporating the split G4 motifs in a DNA nanocage has been developed. The rigid prism outcompetes the flexible one in reaction kinetics and signal/background ratios, which can be easily internalized by cells and successfully applied in microRNA imaging in live cells.
Collapse
Affiliation(s)
- Yanyan Yu
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hua M, Yang S, Ma J, He W, Kuang L, Hua D. Highly selective and sensitive determination of uranyl ion by the probe of CdTe quantum dot with a specific size. Talanta 2018; 190:278-283. [DOI: 10.1016/j.talanta.2018.08.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/03/2018] [Accepted: 08/03/2018] [Indexed: 01/03/2023]
|
31
|
Theerasilp M, Chalermpanapun P, Sunintaboon P, Sungkarat W, Nasongkla N. Glucose-installed biodegradable polymeric micelles for cancer-targeted drug delivery system: synthesis, characterization and in vitro evaluation. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2018; 29:177. [PMID: 30506149 DOI: 10.1007/s10856-018-6177-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Glucose metabolism of cancer can be used as a strategy to target cancer cells which exhibit altered glycolytic rate. The facilitated glucose transporter (Glut) plays an important role in enhancement glycolytic rate resulting in increased glucose uptake into cancer cells. 18FGD-PET image is an example for using Glut as a targeting to diagnose the high glycolytic rate of tumor. Thus, Glut may be adapted to target cancer cells for drug delivery system. Herein, biodegradation polymeric micelles target cancer cells by Glut was fabricated. The amphiphilic block copolymer of poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-b-PCL) was synthesized where terminal group of the PEG chain was installed with glucose molecules. The 1H-NMR confirmed the existence of glucose moiety from two distinct peaks (5.2 and 4.7 ppm) of protons at anomeric carbon of glucose. Glucose-PEG-b-PCL spontaneously forms micelles in an aqueous solution. The size and zeta potential were 22 nm and -7 mv, respectively. Glucose-micelles have high stability, and no evidence of cytotoxicity was found after incubation for 7 days. Doxorubicin, used as a fluorescent probe, was loaded into glucose-micelles. The enhanced amount of doxorubicin as a result of glucose-micelles in PC-3, MCF-7 and HepG2 was evaluated by fluorescence microscopy and flow cytometer. Glucose molecules on the surface of micelles increased internalization and enhanced uptake of micelles via bypassing endocytosis pathway. These results show the use of glucose as a targeting ligand on the micelle surface to target cancer cells via Glut.
Collapse
Affiliation(s)
- Man Theerasilp
- Department of Biomedical Engineering, Mahidol University, Puttamonthon, Nakorn Pathom, 73170, Thailand
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Mahidol University, Bangkok, 10400, Thailand
| | - Punlop Chalermpanapun
- Department of Biomedical Engineering, Mahidol University, Puttamonthon, Nakorn Pathom, 73170, Thailand
| | - Panya Sunintaboon
- Department of Chemistry, Mahidol University, Nakorn patom, 73170, Thailand
| | - Witaya Sungkarat
- Department of Radiology, Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Norased Nasongkla
- Department of Biomedical Engineering, Mahidol University, Puttamonthon, Nakorn Pathom, 73170, Thailand.
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Mahidol University, Bangkok, 10400, Thailand.
| |
Collapse
|
32
|
Tang B, Qian Y, Gou Y, Cheng G, Fang G. VE-Albumin Core-Shell Nanoparticles for Paclitaxel Delivery to Treat MDR Breast Cancer. Molecules 2018; 23:E2760. [PMID: 30366367 PMCID: PMC6278303 DOI: 10.3390/molecules23112760] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/18/2018] [Accepted: 10/20/2018] [Indexed: 11/17/2022] Open
Abstract
Multi-drug resistance (MDR) presents a serious problem in cancer chemotherapy. In this study, Vitamin E (VE)-Albumin core-shell nanoparticles were developed for paclitaxel (PTX) delivery to improve the chemotherapy efficacy in an MDR breast cancer model. The PTX-loaded VE-Albumin core-shell nanoparticles (PTX-VE NPs) had small particle sizes (about 100 nm), high drug entrapment efficiency (95.7%) and loading capacity (12.5%), and showed sustained release profiles, in vitro. Docking studies indicated that the hydrophobic interaction and hydrogen bonds play a significant role in the formation of the PTX-VE NPs. The results of confocal laser scanning microscopy analysis demonstrated that the cell uptake of PTX was significantly increased by the PTX-VE NPs, compared with the NPs without VE (PTX NPs). The PTX-VE NPs also exhibited stronger cytotoxicity, compared with PTX NPs with an increased accumulation of PTX in the MCF-7/ADR cells. Importantly, the PTX-VE NPs showed a higher anti-cancer efficacy in MCF-7/ADR tumor xenograft model than the PTX NPs and the PTX solutions. Overall, the VE-Albumin core-shell nanoparticles could be a promising nanocarrier for PTX delivery to improve the chemotherapeutic efficacy of MDR cancer.
Collapse
Affiliation(s)
- Bo Tang
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Yu Qian
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Yi Gou
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, China.
| | - Gang Cheng
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, Liaoning, China.
| | - Guihua Fang
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
33
|
Su G, Jiang H, Xu B, Yu Y, Chen X. Effects of Protein Corona on Active and Passive Targeting of Cyclic RGD Peptide-Functionalized PEGylation Nanoparticles. Mol Pharm 2018; 15:5019-5030. [PMID: 30222356 DOI: 10.1021/acs.molpharmaceut.8b00612] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein corona can alter the physiochemical properties of targeting nanoparticles (NPs), as well as their physiological responses and targeting functionality. Herein, we synthesized 20 types of NPs with diverse surface chemistry in order to study the impacts of protein corona on targeting functionality of NPs functionalized with cyclic RGD peptides and their relationships to the polyethylene glycol (PEG) length and grafting density of targeting ligands. After protein adsorption, cyclic RGD on the surface of NP was still able to bind its receptors with increased targeted cellular uptake, even at a relatively low density. However, the cellular uptake was reduced from 26 to 76% when compared with protein nonbound NPs, which was caused by the shielding effect of the outer layer adsorbed proteins. NPs functionalized with short PEG molecules and moderate cyclic RGD density performed a better targeting efficiency. Due to PEG conjugation, the protein corona was demonstrated to be beneficial for passive targeting by decreasing macrophage cellular uptake. These relationships between surface chemistry and targeting functionality will provide guidelines for the design of targeting nanoformulations in nanomedicine.
Collapse
Affiliation(s)
- Gaoxing Su
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210093 , China.,School of Pharmacy , Nantong University , Nantong 226001 , China
| | - Huaqiao Jiang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210093 , China.,School of Pharmacy , Nantong University , Nantong 226001 , China
| | - Bohui Xu
- School of Pharmacy , Nantong University , Nantong 226001 , China
| | - Yanyan Yu
- School of Pharmacy , Nantong University , Nantong 226001 , China
| | - Xueqin Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210093 , China
| |
Collapse
|
34
|
Sahu A, Choi WI, Tae G. Recent Progress in the Design of Hypoxia-Specific Nano Drug Delivery Systems for Cancer Therapy. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800026] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Abhishek Sahu
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| | - Won Il Choi
- Center for Convergence Bioceramic Materials; Convergence R&D Division; Korea Institute of Ceramic Engineering and Technology; 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu Cheongju Chungbuk 28160 Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering; Gwangju Institute of Science and Technology; 123 Cheomdan-gwagiro, Buk-gu Gwangju 61005 Republic of Korea
| |
Collapse
|
35
|
|
36
|
Gou Y, Miao D, Zhou M, Wang L, Zhou H, Su G. Bio-Inspired Protein-Based Nanoformulations for Cancer Theranostics. Front Pharmacol 2018; 9:421. [PMID: 29755355 PMCID: PMC5934525 DOI: 10.3389/fphar.2018.00421] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 04/11/2018] [Indexed: 01/09/2023] Open
Abstract
Over the past decade, more interests have been aroused in engineering protein-based nanoformulations for cancer treatment. This excitement originates from the success of FDA approved Abraxane (Albumin-based paclitaxel nanoparticles) in 2005. The new generation of biocompatible endogenous protein-based nanoformulations is currently constructed through delivering cancer therapeutic and diagnostic agents simultaneously, as named potential theranostics. Protein nanoformulations are commonly incorporated with dyes, contrast agents, drug payloads or inorganic nanoclusters, serving as imaging-guided combinatorial cancer therapeutics. Employing the nature identity of proteins, the theranostics, escape the clearance by reticuloendothelial cells and have a long blood circulation time. The nanoscale sizet allows them to be penetrated deeply into tumor tissues. In addition, stimuli release and targeted molecules are incorporated to improve the delivery efficiency. The ongoing advancement of protein-based nanoformulations for cancer theranostics in recent 5 years is reviewed in this paper. Fine-designed nanoformulations based on albumin, ferritin, gelatin, and transferrin are highlighted from the literature. Finally, the current challenges are identified in translating protein-based nanoformulations from laboratory to clinical trials.
Collapse
Affiliation(s)
- Yi Gou
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Targets, School of Pharmacy, Nantong University, Nantong, China
| | - Dandan Miao
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Targets, School of Pharmacy, Nantong University, Nantong, China
| | - Min Zhou
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Targets, School of Pharmacy, Nantong University, Nantong, China
| | - Lijuan Wang
- Guangzhou Key Laboratory of Environmental Exposure and Health and Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, China
| | - Hongyu Zhou
- Guangzhou Key Laboratory of Environmental Exposure and Health and Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, China
| | - Gaoxing Su
- Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Targets, School of Pharmacy, Nantong University, Nantong, China
| |
Collapse
|
37
|
Xu B, Xu Y, Su G, Zhu H, Zong L. A multifunctional nanoparticle constructed with a detachable albumin outer shell and a redox-sensitive inner core for efficient siRNA delivery to hepatocellular carcinoma cells. J Drug Target 2018; 26:941-954. [PMID: 29564911 DOI: 10.1080/1061186x.2018.1455840] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Successful delivery of small interfering RNA (siRNA) into the cytoplasm of target cells relies on biocompatible and efficient vectors. In this study, a novel multifunctional core/shell nanoparticle [CS-SS-9R/BSA-c(RGDyK)] was developed to effectively deliver siVEGF to hepatocellular carcinoma cells (Bel-7402 cells). To improve the gene payload and transfection efficiency, a positively charged inner core (CS-SS-9R) was constructed by grafting nona-arginine (9R) onto chitosan (CS) using disulphide bonds. The negatively charged outer shell [BSA-c(RGDyK)] assembled on the surface of the inner core by electrostatic forces that shielded high cationic charges and provided improved targeting. The protein outer shell gradually detached from the inner core in the acidic lysosomal environment, leaving the cationic inner core exposed in order to escape from lysosomes. The nanoparticles were capable of delivering siVEGF into Bel-7402 cells via integrin receptor-mediated endocytosis. Successful lysosomal escape of the inner core and the rapid release of siVEGF into the cytoplasm resulted in a 78.9% decrease in VEGF expression and 81.2% inhibition of tumour cell proliferation. In conclusion, this nanoparticle is responsive to the intracellular environment and accurately delivered siRNA into the cytoplasm, providing a safe and highly efficient gene delivery strategy for cancer therapy.
Collapse
Affiliation(s)
- Bohui Xu
- a School of Pharmacy , China Pharmaceutical University , Nanjing , China.,b School of Pharmacy , Nantong University , Nantong , China
| | - Yan Xu
- b School of Pharmacy , Nantong University , Nantong , China
| | - Gaoxing Su
- b School of Pharmacy , Nantong University , Nantong , China
| | - Hongyan Zhu
- b School of Pharmacy , Nantong University , Nantong , China
| | - Li Zong
- a School of Pharmacy , China Pharmaceutical University , Nanjing , China
| |
Collapse
|
38
|
Shen J, Zhang W, Qi R, Mao ZW, Shen H. Engineering functional inorganic-organic hybrid systems: advances in siRNA therapeutics. Chem Soc Rev 2018; 47:1969-1995. [PMID: 29417968 PMCID: PMC5861001 DOI: 10.1039/c7cs00479f] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer treatment still faces a lot of obstacles such as tumor heterogeneity, drug resistance and systemic toxicities. Beyond the traditional treatment modalities, exploitation of RNA interference (RNAi) as an emerging approach has immense potential for the treatment of various gene-caused diseases including cancer. The last decade has witnessed enormous research and achievements focused on RNAi biotechnology. However, delivery of small interference RNA (siRNA) remains a key challenge in the development of clinical RNAi therapeutics. Indeed, functional nanomaterials play an important role in siRNA delivery, which could overcome a wide range of sequential physiological and biological obstacles. Nanomaterial-formulated siRNA systems have potential applications in protection of siRNA from degradation, improving the accumulation in the target tissues, enhancing the siRNA therapy and reducing the side effects. In this review, we explore and summarize the role of functional inorganic-organic hybrid systems involved in the siRNA therapeutic advancements. Additionally, we gather the surface engineering strategies of hybrid systems to optimize for siRNA delivery. Major progress in the field of inorganic-organic hybrid platforms including metallic/non-metallic cores modified with organic shells or further fabrication as the vectors for siRNA delivery is discussed to give credit to the interdisciplinary cooperation between chemistry, pharmacy, biology and medicine.
Collapse
Affiliation(s)
- Jianliang Shen
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China. and School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325035, China and Wenzhou Institute of Biomaterials and Engineering, Chinese Academy of Science, Wenzhou, 325001, China and Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA.
| | - Wei Zhang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Ruogu Qi
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA.
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, China. and Department of Applied Chemistry, South China Agricultural University, Guangzhou 510642, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, USA. and Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY10065, USA
| |
Collapse
|
39
|
Tofani LB, Depieri LV, Campos PM, Riul TB, Antonietto KS, de Abreu Fantini MC, Bentley MVLB. In Vitro TyRP-1 Knockdown Based on siRNA Carried by Liquid Crystalline Nanodispersions: an Alternative Approach for Topical Treatment of Vitiligo. Pharm Res 2018; 35:104. [PMID: 29560584 DOI: 10.1007/s11095-017-2330-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 12/12/2017] [Indexed: 01/27/2023]
Abstract
PURPOSE Vitiligo is a skin disease characterized by depigmentation and the presence of white patches that are associated with the loss of melanocytes. The most common explanation for the cause of this condition is that it is an autoimmune condition. TyRP-1 is involved in melanin pigment synthesis but can also function as a melanocyte differentiation antigen. This protein plays a role in the autoimmune destruction of melanocytes, which results in the depigmentation, characteristic of this disease. In this study, we evaluated liquid crystalline nanodispersions as non-viral vectors to deliver siRNA-TyRP-1 as an alternative for topical treatment of vitiligo. METHODS Liquid crystalline nanodispersions were obtained and characterized with respect to their physical-chemical parameters including size, PdI and zeta potential, as well as Small Angle X-ray Scattering and complexing to siRNA. The effects of the liquid crystalline nanodispersions on the cellular viability, cell uptake and levels of the knockdown target TyRP-1 were evaluated in melan-A cells after 24 h of treatment. RESULTS The liquid crystalline nanodispersions demonstrated adequate physical-chemical parameters including nanometer size and a PdI below 0.38. These systems promoted a high rate of cell uptake and an impressive TyRP-1 target knockdown (> 80%) associated with suitable loading of TyRp-1 siRNA. CONCLUSIONS We demonstrated that the liquid crystalline nanodispersions showed promising alternative for the topical treatment of vitiligo due to their physical parameters and ability in knockdown the target protein involved with autoimmune destruction of melanocytes.
Collapse
Affiliation(s)
- Larissa Bueno Tofani
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Lívia Vieira Depieri
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Patrícia Mazureki Campos
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Thalita Bachelli Riul
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | - Kamilla Swiech Antonietto
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, s/n, Ribeirão Preto, SP, 14040-903, Brazil
| | | | - Maria Vitória Lopes Badra Bentley
- School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, s/n, Ribeirão Preto, SP, 14040-903, Brazil.
| |
Collapse
|
40
|
Xia Y, Wang C, Xu T, Li Y, Guo M, Lin Z, Zhao M, Zhu B. Targeted delivery of HES5-siRNA with novel polypeptide-modified nanoparticles for hepatocellular carcinoma therapy. RSC Adv 2018; 8:1917-1926. [PMID: 35542585 PMCID: PMC9077277 DOI: 10.1039/c7ra12461a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/15/2017] [Indexed: 12/18/2022] Open
Abstract
For actively targeted delivery of small interfering RNA (siRNA) to solid tumors, we fabricated functionalized selenium nanoparticles (SeNPs) decorated with the polypeptide RGDfC. Herein, RGDfC was used as tumor-targeted moiety and installed onto the surface of SeNPs to enhance the cellular uptake. RGDfC-SeNPs@siRNA were internalized into the HepG2 cell mainly through clathrin-mediated endocytosis. The active efficacy of the RGDfC-SeNPs@siRNA was confirmed via gene silencing assay, MTT assay and flow cytometry analysis. Owing to the tumor-targeting effect of RGDfC, RGDfC-SeNPs@siRNA achieved an obvious improvement in gene silencing ability, which led to significant growth inhibition of HepG2 cells. Furthermore, treatment with RGDfC-SeNPs@siRNA resulted in greater antitumor efficacy than lipofectamine 2000@siRNA in vitro and in vivo. In addition, the RGDfC-SeNPs@siRNA was almost non-toxic to the key organs of mice. In sum, these findings provide an alternative therapeutic route for targeted cancer treatments. A novel polypeptide RGDfC-modified selenium nanoparticle was fabricated to selectively deliver HES5-siRNA to tumors for hepatocellular carcinoma therapy.![]()
Collapse
Affiliation(s)
- Yu Xia
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Changbing Wang
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Tiantian Xu
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Yinghua Li
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Min Guo
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Zhengfang Lin
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Mingqi Zhao
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Bing Zhu
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| |
Collapse
|
41
|
Shi NQ, Li Y, Zhang Y, Shen N, Qi L, Wang SR, Qi XR. Intelligent "Peptide-Gathering Mechanical Arm" Tames Wild "Trojan-Horse" Peptides for the Controlled Delivery of Cancer Nanotherapeutics. ACS APPLIED MATERIALS & INTERFACES 2017; 9:41767-41781. [PMID: 29161013 DOI: 10.1021/acsami.7b15523] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Cell-penetrating peptides (CPPs), also called "Trojan-Horse" peptides, have been used for facilitating intracellular delivery of numerous diverse cargoes and even nanocarriers. However, the lack of targeting specificity ("wildness" or nonselectivity) of CPP-nanocarriers remains an intractable challenge for many in vivo applications. In this work, we used an intelligent "peptide-gathering mechanical arm" (Int PMA) to curb CPPs' wildness and enhance the selectivity of R9-liposome-based cargo delivery for tumor targeting. The peptide NGR, serving as a cell-targeting peptide for anchoring, and peptide PLGLAG, serving as a substrate peptide for deanchoring, were embedded in the Int PMA motif. The Int PMA construct was designed to be sensitive to tumor microenvironmental stimuli, including aminopeptidase N (CD13) and matrix metalloproteinases (MMP-2/9). Moreover, Int PMA could be specifically recognized by tumor tissues via CD13-mediated anchoring and released for cell entry by MMP-2/9-mediated deanchoring. To test the Int PMA design, a series of experiments were conducted in vitro and in vivo. Functional conjugates Int PMA-R9-poly(ethylene glycol) (PEG)2000-distearoylphosphatidyl-ethanolamine (DSPE) and R9-PEG2000-DSPE were synthesized by Michael addition reaction and were characterized by thin-layer chromatography and matrix-assisted laser desorption ionization-time-of-flight mass spectrometry. The Int PMA-R9-modified doxorubicin-loaded liposomes (Int PMA-R9-Lip-DOX) exhibited a proper particle diameter (approximately 155 nm) with in vitro sustained release characteristics. Cleavage assay showed that Int PMA-R9 peptide molecules could be cleaved by MMP-2/9 for completion of deanchoring. Flow cytometry and confocal microscopy studies indicated that Int PMA-R9-Lip-DOX can respond to both endogenous and exogenous stimuli in the presence/absence of excess MMP-2/9 and MMP-2/9 inhibitor (GM6001) and effectively function under competitive receptor-binding conditions. Moreover, Int PMA-R9-Lip-DOX generated more significant subcellular dispersions that were especially evident within endoplasmic reticulum (ER) and Golgi apparatus. Notably, Int PMA-R9-Lip-DOX could induce enhanced apoptosis, during which caspase 3/7 might be activated. In addition, Int PMA-R9-Lip-DOX displayed enhanced in vitro and in vivo antitumor efficacy versus "wild" R9-Lip-DOX. On the basis of investigations at the molecular level, cellular level, and animals' level, the control of Int PMA was effective and promoted selective delivery of R9-liposome cargo to the target site and reduced nonspecific uptake. This Int PMA-controlled strategy based on aminopeptidase-guided anchoring and protease-triggered deanchoring effectively curbed the wildness of CPPs and bolstered their effectiveness for in vivo delivery of nanotherapeutics. The specific nanocarrier delivery system used here could be adapted using a variety of intelligent designs based on combinations of multifunctional peptides that would specifically and preferentially bind to tumors versus nontumor tissues for tumor-localized accumulation in vivo. Thus, CPPs have a strong advantage for the development of intelligent nanomedicines for targeted tumor therapy.
Collapse
Affiliation(s)
- Nian-Qiu Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University , Tianjin 300071, China
| | | | - Yong Zhang
- College of Life Science, Jilin University , 2699 Qianjin Street, Changchun 130012, Jilin Province, China
| | | | | | | | - Xian-Rong Qi
- Department of Pharmaceutics, School of Pharmaceutical Science, Peking University , Beijing 100191, China
| |
Collapse
|
42
|
Targeted Delivery of siRNA with pH-Responsive Hybrid Gold Nanostars for Cancer Treatment. Int J Mol Sci 2017; 18:ijms18102029. [PMID: 28937584 PMCID: PMC5666711 DOI: 10.3390/ijms18102029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/12/2022] Open
Abstract
In this work, we report the engineering of gold nanostars (GNS) to deliver small interfering RNA (siRNA) into HepG2 cells. The ligand DG-PEG-Lipoic acid (LA)-Lys-9R (hydrazone) was designed to functionalize GNS, and create the nanoparticles named as 9R/DG-GNS (hydrazone). In the ligand, 2-deoxyglucose (DG) is the targeting molecule, polyethylene glycol (PEG) helps to improve the dispersity and biocompatibility, 9-poly-d-arginine (9R) is employed to provide a positive surface charge and adsorb negative siRNA, and hydrazone bonds are pH-responsive and can avoid receptor-mediated endosomal recycling. Compared to GNS alone, 9R/DG-GNS (hydrazone) showed superior transfection efficiency. The expressions of cyclooxygenase-2 (COX-2) in HepG2 and SGC7901 cells were significantly suppressed by siRNA/9R/DG-GNS (hydrazone) complex. Notably, 9R/DG-GNS (hydrazone) possessed low cytotoxicity even at high concentrations in both normal cells and tumor cells. The combination treatment of siRNA/9R/DG-GNS (hydrazone) complex inhibited the cell growth rate by more than 75%. These results verified that the pH-responsive GNS complex is a promising siRNA delivery system for cancer therapy, and it is anticipated that near-infrared absorbing GNS with good photothermal conversion efficiency can be potentially used for photothermal therapy of tumors.
Collapse
|
43
|
|
44
|
Zhang S, Zhao L, Qiu N, Liu Y, Xu B, Zhu H. On the hypoxic tumor targeting ability of two chitosan micelles loaded with oil-soluble CdSe quantum dots. Pharm Dev Technol 2017; 23:87-95. [PMID: 28782455 DOI: 10.1080/10837450.2017.1365082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hypoxia, an outstanding characteristic of various solid tumors, has been considered a critical factor of aggressive tumor phenotypes, poor clinical prognosis, and increased expression of the multidrug-resistant gene. Therefore, it is critical to develop a drug delivery system to enhance the delivery effect of the antitumor drug in the hypoxic tumor. We constructed two types of tumor targeting micelles based on chitosan and evaluated their properties in targeting hypoxic tumors. Chitosan-based micelles consisted of a hydrophobic group octyl group, a hydrophilic polyethylene glycol, tumor targeting ligands glucosamine or folic acid, and a transmembrane peptide 9-d-arginine. The molecular structure, morphology, size distribution, zeta potential, and biosafety of two micelles were characterized. Oil-soluble CdSe quantum dots were used as a fluorescent probe to evaluate the hypoxic tumor cell targeting properties of the micelles. Moreover, HepG2 human hepatocellular carcinoma cells and HeLa human cervical carcinoma cells were used as in vitro models. We demonstrated that, under hypoxic conditions, two chitosan micelles showed better targeting ability to HepG2 and HeLa cells, which enhanced the effect of antitumor drugs by specifically targeting transport in hypoxic tumors. Therefore, chitosan micelles may be a potential drug delivery system that can be used to deliver antitumor drugs to hypoxic tumors.
Collapse
Affiliation(s)
- Shengyu Zhang
- a School of Pharmacy , Nantong University , Nantong , PR China
| | - Liuwan Zhao
- a School of Pharmacy , Nantong University , Nantong , PR China
| | - Nanqin Qiu
- a School of Pharmacy , Nantong University , Nantong , PR China
| | - Yanjun Liu
- a School of Pharmacy , Nantong University , Nantong , PR China
| | - Bohui Xu
- a School of Pharmacy , Nantong University , Nantong , PR China
| | - Hongyan Zhu
- a School of Pharmacy , Nantong University , Nantong , PR China
| |
Collapse
|
45
|
Jahangirian H, Lemraski EG, Webster TJ, Rafiee-Moghaddam R, Abdollahi Y. A review of drug delivery systems based on nanotechnology and green chemistry: green nanomedicine. Int J Nanomedicine 2017; 12:2957-2978. [PMID: 28442906 PMCID: PMC5396976 DOI: 10.2147/ijn.s127683] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This review discusses the impact of green and environmentally safe chemistry on the field of nanotechnology-driven drug delivery in a new field termed "green nanomedicine". Studies have shown that among many examples of green nanotechnology-driven drug delivery systems, those receiving the greatest amount of attention include nanometal particles, polymers, and biological materials. Furthermore, green nanodrug delivery systems based on environmentally safe chemical reactions or using natural biomaterials (such as plant extracts and microorganisms) are now producing innovative materials revolutionizing the field. In this review, the use of green chemistry design, synthesis, and application principles and eco-friendly synthesis techniques with low side effects are discussed. The review ends with a description of key future efforts that must ensue for this field to continue to grow.
Collapse
Affiliation(s)
- Hossein Jahangirian
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | | | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Roshanak Rafiee-Moghaddam
- School of Chemical Sciences and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Selangor
| | - Yadollah Abdollahi
- Department of Electrical Engineering, Faculty of Engineering, University of Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
46
|
Tang Y, Zeng Z, He X, Wang T, Ning X, Feng X. SiRNA Crosslinked Nanoparticles for the Treatment of Inflammation-induced Liver Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2017; 4:1600228. [PMID: 28251047 PMCID: PMC5323819 DOI: 10.1002/advs.201600228] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 07/29/2016] [Indexed: 05/20/2023]
Abstract
RNA interference mediated by small interfering RNA (siRNA) provides a powerful tool for gene regulation, and has a broad potential as a promising therapeutic strategy. However, therapeutics based on siRNA have had limited clinical success due to their undesirable pharmacokinetic properties. This study presents pH-sensitive nanoparticles-based siRNA delivery systems (PNSDS), which are positive-charge-free nanocarriers, composed of siRNA chemically crosslinked with multi-armed poly(ethylene glycol) carriers via acid-labile acetal linkers. The unique siRNA crosslinked structure of PNSDS allows it to have minimal cytotoxicity, high siRNA loading efficiency, and a stimulus-responsive property that enables the selective intracellular release of siRNA in response to pH conditions. This study demonstrates that PNSDS can deliver tumor necrosis factor alpha (TNF-α) siRNA into macrophages and induce the efficient down regulation of the targeted gene in complete cell culture media. Moreover, PNSDS with mannose targeting moieties can selectively accumulate in mice liver, induce specific inhibition of macrophage TNF-α expression in vivo, and consequently protect mice from inflammation-induced liver damages. Therefore, this novel siRNA delivering platform would greatly improve the therapeutic potential of RNAi based therapies.
Collapse
Affiliation(s)
- Yaqin Tang
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| | - Ziying Zeng
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| | - Xiao He
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| | - Tingting Wang
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| | - Xinghai Ning
- Department of Biomedical EngineeringNanjing UniversityNanjing210093P. R. China
| | - Xuli Feng
- Innovative Drug Research CenterChongqing UniversityChongqing401331P. R. China
| |
Collapse
|
47
|
Zang X, Ding H, Zhao X, Li X, Du Z, Hu H, Qiao M, Chen D, Deng Y, Zhao X. Anti-EphA10 antibody-conjugated pH-sensitive liposomes for specific intracellular delivery of siRNA. Int J Nanomedicine 2016; 11:3951-67. [PMID: 27574425 PMCID: PMC4993279 DOI: 10.2147/ijn.s107952] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Therapeutic delivery of small interfering RNA (siRNA) is a major challenge that limits its potential clinical application. Here, a pH-sensitive cholesterol–Schiff base–polyethylene glycol (Chol–SIB–PEG)-modified cationic liposome–siRNA complex, conjugated with the recombinant humanized anti-EphA10 antibody (Eph), was developed as an efficient nonviral siRNA delivery system. Chol–SIB–PEG was successfully synthesized and confirmed with FTIR and 1H-NMR. An Eph–PEG–SIB–Chol-modified liposome–siRNA complex (EPSLR) was prepared and characterized by size, zeta potential, gel retardation, and encapsulation efficiency. Electrophoresis results showed that EPSLR was resistant to heparin replacement and protected siRNA from fetal bovine serum digestion. EPSLR exhibited only minor cytotoxicity in MCF-7/ADR cells. The results of flow cytometry and confocal laser scanning microscopy suggested that EPSLR enhanced siRNA transfection in MCF-7/ADR cells. Intracellular distribution experiment revealed that EPSLR could escape from the endo-lysosomal organelle and release siRNA into cytoplasm at 4 hours posttransfection. Western blot experiment demonstrated that EPSLR was able to significantly reduce the levels of MDR1 protein in MCF-7/ADR cells. The in vivo study of DIR-labeled complexes in mice bearing MCF-7/ADR tumor indicated that EPSLR could reach the tumor site rather than other organs more effectively. All these results demonstrate that EPSLR has much potential for effective siRNA delivery and may facilitate its therapeutic application.
Collapse
Affiliation(s)
| | - Huaiwei Ding
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, People's Republic of China
| | - Xiufeng Zhao
- Hongqi Hospital affiliated to Mudanjiang Medical University, Mudanjiang, People's Republic of China
| | - Xiaowei Li
- Department of Pharmaceutics, School of Pharmacy
| | - Zhouqi Du
- Department of Pharmaceutics, School of Pharmacy
| | - Haiyang Hu
- Department of Pharmaceutics, School of Pharmacy
| | - Mingxi Qiao
- Department of Pharmaceutics, School of Pharmacy
| | - Dawei Chen
- Department of Pharmaceutics, School of Pharmacy
| | - Yuihui Deng
- Department of Pharmaceutics, School of Pharmacy
| | - Xiuli Zhao
- Department of Pharmaceutics, School of Pharmacy
| |
Collapse
|
48
|
Development of Drugs and Technology for Radiation Theragnosis. NUCLEAR ENGINEERING AND TECHNOLOGY 2016. [DOI: 10.1016/j.net.2016.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
49
|
Li Y, Yang HY, Lee DS. Polymer-Based and pH-Sensitive Nanobiosensors for Imaging and Therapy of Acidic Pathological Areas. Pharm Res 2016; 33:2358-72. [DOI: 10.1007/s11095-016-1944-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/11/2016] [Indexed: 12/27/2022]
|
50
|
Kim S, Na HK, Won C, Min DH. In-depth study on the gene silencing capability of silica nanoparticles with different pore sizes: degree and duration of RNA interference. RSC Adv 2016. [DOI: 10.1039/c5ra27777a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The mesoporous silica nanoparticles (MSN) having different pore sizes were synthesized and utilized for siRNA delivery system capable of controlling kinetics of RNA interference.
Collapse
Affiliation(s)
- Seongchan Kim
- Center for RNA Research
- Institute for Basic Science (IBS)
- Department of Chemistry
- Seoul National University
- Seoul 151-747
| | - Hee-Kyung Na
- Center for RNA Research
- Institute for Basic Science (IBS)
- Department of Chemistry
- Seoul National University
- Seoul 151-747
| | - Cheolhee Won
- Institute of Nanobio Convergence Technology
- Lemonex Inc
- Seoul 151-742
- Republic of Korea
| | - Dal-Hee Min
- Center for RNA Research
- Institute for Basic Science (IBS)
- Department of Chemistry
- Seoul National University
- Seoul 151-747
| |
Collapse
|