1
|
Zhou J, Shen W, Feng W, Zhang X, Wu T, Zhou J, Su Z, Yin T. Temperature Self-Limited Intelligent Thermo-chemotherapeutic Lipid Nanosystem for P-gp Reversal Time Window Matched Pulse Treatment of MDR Tumor. NANO LETTERS 2024; 24:10631-10641. [PMID: 39150779 DOI: 10.1021/acs.nanolett.4c02978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/18/2024]
Abstract
Mild photothermal therapy (PTT) shows the potential for chemosensitization by tumor-localized P-glycoprotein (P-gp) modulation. However, conventional mild PTT struggles with real-time uniform temperature control, obscuring the temperature-performance relationship and resulting in thermal damage. Besides, the time-performance relationship and the underlying mechanism of mild PTT-mediated P-gp reversal remains elusive. Herein, we developed a temperature self-limiting lipid nanosystem (RFE@PD) that integrated a reversible organic heat generator (metal-phenolic complexes) and metal chelator (deferiprone, DFP) encapsulated phase change material. Upon NIR irradiation, RFE@PD released DFP for blocking ligand-metal charge transfer to self-limit temperature below 45 °C, and rapidly reduced P-gp within 3 h via Ubiquitin-proteasome degradation. Consequently, the DOX·HCl-loaded thermo-chemotherapeutic lipid nanosystem (RFE@PD-DOX) led to dramatically improved drug accumulation and 5-fold chemosensitization in MCF-7/ADR tumor models by synchronizing P-gp reversal and drug pulse liberation, achieving a tumor inhibition ratio of 82.42%. This lipid nanosystem integrated with "intrinsic temperature-control" and "temperature-responsive pulse release" casts new light on MDR tumor therapy.
Collapse
Affiliation(s)
- Jiyuan Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Weiyang Shen
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Wenna Feng
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Xin Zhang
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Tongyu Wu
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianping Zhou
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Zhigui Su
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| | - Tingjie Yin
- Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China
| |
Collapse
|
2
|
Dowaidar M. Guidelines for the role of autophagy in drug delivery vectors uptake pathways. Heliyon 2024; 10:e30238. [PMID: 38707383 PMCID: PMC11066435 DOI: 10.1016/j.heliyon.2024.e30238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
The process of autophagy refers to the intracellular absorption of cytoplasm (such as proteins, nucleic acids, tiny molecules, complete organelles, and so on) into the lysosome, followed by the breakdown of that cytoplasm. The majority of cellular proteins are degraded by a process called autophagy, which is both a naturally occurring activity and one that may be induced by cellular stress. Autophagy is a system that can save cells' integrity in stressful situations by restoring metabolic basics and getting rid of subcellular junk. This happens as a component of an endurance response. This mechanism may have an effect on disease, in addition to its contribution to the homeostasis of individual cells and tissues as well as the control of development in higher species. The main aim of this study is to discuss the guidelines for the role of autophagy in drug delivery vector uptake pathways. In this paper, we discuss the meaning and concept of autophagy, the mechanism of autophagy, the role of autophagy in drug delivery vectors, autophagy-modulating drugs, nanostructures for delivery systems of autophagy modulators, etc. Later in this paper, we talk about how to deliver chemotherapeutics, siRNA, and autophagy inducers and inhibitors. We also talk about how hard it is to make a drug delivery system that takes nanocarriers' roles as autophagy modulators into account.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Bioengineering Department, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
- Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
- Biosystems and Machines Research Center, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, 31261, Saudi Arabia
| |
Collapse
|
3
|
Yang C, Ding Y, Mao Z, Wang W. Nanoplatform-Mediated Autophagy Regulation and Combined Anti-Tumor Therapy for Resistant Tumors. Int J Nanomedicine 2024; 19:917-944. [PMID: 38293604 PMCID: PMC10826716 DOI: 10.2147/ijn.s445578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024] Open
Abstract
The overall cancer incidence and death toll have been increasing worldwide. However, the conventional therapies have some obvious limitations, such as non-specific targeting, systemic toxic effects, especially the multidrug resistance (MDR) of tumors, in which, autophagy plays a vital role. Therefore, there is an urgent need for new treatments to reduce adverse reactions, improve the treatment efficacy and expand their therapeutic indications more effectively and accurately. Combination therapy based on autophagy regulators is a very feasible and important method to overcome tumor resistance and sensitize anti-tumor drugs. However, the less improved efficacy, more systemic toxicity and other problems limit its clinical application. Nanotechnology provides a good way to overcome this limitation. Co-delivery of autophagy regulators combined with anti-tumor drugs through nanoplatforms provides a good therapeutic strategy for the treatment of tumors, especially drug-resistant tumors. Notably, the nanomaterials with autophagy regulatory properties have broad therapeutic prospects as carrier platforms, especially in adjuvant therapy. However, further research is still necessary to overcome the difficulties such as the safety, biocompatibility, and side effects of nanomedicine. In addition, clinical research is also indispensable to confirm its application in tumor treatment.
Collapse
Affiliation(s)
- Caixia Yang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
4
|
He K, Chen M, Liu J, Du S, Ren C, Zhang J. Nanomedicine for cancer targeted therapy with autophagy regulation. Front Immunol 2024; 14:1238827. [PMID: 38239356 PMCID: PMC10794438 DOI: 10.3389/fimmu.2023.1238827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 12/13/2023] [Indexed: 01/22/2024] Open
Abstract
Nanoparticles have unique physical and chemical properties and are currently widely used in disease diagnosis, drug delivery, and new drug development in biomedicine. In recent years, the role of nanomedical technology in cancer treatment has become increasingly obvious. Autophagy is a multi-step degradation process in cells and an important pathway for material and energy recovery. It is closely related to the occurrence and development of cancer. Because nanomaterials are highly targeted and biosafe, they can be used as carriers to deliver autophagy regulators; in addition to their favorable physicochemical properties, nanomaterials can be employed to carry autophagy inhibitors, reducing the breakdown of chemotherapy drugs by cancer cells and thereby enhancing the drug's efficacy. Furthermore, certain nanomaterials can induce autophagy, triggering oxidative stress-mediated autophagy enhancement and cell apoptosis, thus constraining the progression of cancer cells.There are various types of nanoparticles, including liposomes, micelles, polymers, metal-based materials, and carbon-based materials. The majority of clinically applicable drugs are liposomes, though other materials are currently undergoing continuous optimization. This review begins with the roles of autophagy in tumor treatment, and then focuses on the application of nanomaterials with autophagy-regulating functions in tumor treatment.
Collapse
Affiliation(s)
- Ketai He
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- West China School of Stomatology, Sichuan University, Sichuan, China
| | - Mingkun Chen
- West China School of Stomatology, Sichuan University, Sichuan, China
| | - Jiao Liu
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Sichuan, China
| | - Shufang Du
- West China School of Stomatology, Sichuan University, Sichuan, China
| | - Changyu Ren
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Sichuan, China
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Li Y, Jia F, Gao Y, Wang X, Cui X, Pan Z, Wang W, Li M, Lu J, Wu Y. Self-assembled nanocomposites of carboxymethyl β-dextran/protamine sulfate for enhanced chemotherapeutic drug sensitivity of triple-negative breast cancer by autophagy inhibition via a ternary collaborative strategy. Int J Biol Macromol 2023; 233:123663. [PMID: 36780963 DOI: 10.1016/j.ijbiomac.2023.123663] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 01/19/2023] [Accepted: 02/09/2023] [Indexed: 02/13/2023]
Abstract
Drug resistance in cancer chemotherapy is a major confounding factor affecting the effectiveness of chemotherapeutic agents, thereby leading to poor clinical outcomes. Most chemotherapeutic drugs can induce protective autophagy and increase the resistance of tumors to chemotherapeutic drugs and reduce effective drug delivery to tumor cells. In this study, a tri-drug nanocomposite (NP) delivery system was devised using carboxymethyl β-dextran (CMD) and protamine sulfate (PS), two natural materials with good bio-compatibility. They were designed to carry the chemotherapeutic drug docetaxel (DTX), the autophagy inhibitor chloroquine (CQ), and Atg5 siRNA to cancer cells. The CQ + DTX + Atg5 siRNA NPs was driven by electrostatic interaction and self-assembly methods. The breast cancer cell line MDA-MB-231 was used for both cell culture and establishing mouse xenograft model. Our findings demonstrated that CQ and Atg5 siRNA encapsulated in NPs could enhance the sensitivity of tumor cells to DTX. The NPs exhibited remarkable considerable therapeutic effects for treating triple-negative breast cancer (TNBC) and good biosafety. Therefore, we established a novel multifunctional nanoplatform based on CMD and PS that enhances chemotherapeutic drug sensitivity through an autophagy inhibition strategy, providing new opportunities to overcome conventional drug resistance and enhance therapeutic efficiency against TNBC.
Collapse
Affiliation(s)
- Yunhao Li
- Department of Medicine, Li Ka Shing Faculty of Medicine, University of HongKong, Hong Kong, China; Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, People's Republic of China
| | - Fan Jia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yujuan Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xuan Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, People's Republic of China.
| | - Xinyue Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, People's Republic of China
| | - Zian Pan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Weifeng Wang
- The First Affiliated Hospital of Jiamusi University, Jiamusi 154003, People's Republic of China
| | - Mingjun Li
- The First Affiliated Hospital of Jiamusi University, Jiamusi 154003, People's Republic of China.
| | - Jianqing Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, People's Republic of China.
| | - Yan Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, No. 11 First North Road, Zhongguancun, Beijing 100190, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China.
| |
Collapse
|
6
|
Le BT, La DD, Nguyen PTH. Ultrasonic-Assisted Fabrication of MIL-100(Fe) Metal-Organic Frameworks as a Carrier for the Controlled Delivery of the Chloroquine Drug. ACS OMEGA 2023; 8:1262-1270. [PMID: 36643433 PMCID: PMC9835187 DOI: 10.1021/acsomega.2c06676] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 12/15/2022] [Indexed: 06/17/2023]
Abstract
Metal-organic framework materials (MOFs) are materials with an ordered crystalline structure and high porosity that have been intensively investigated for many applications, such as gas adsorption, catalysis, sensors, drug delivery, and so on. Among them, the MOF-based drug delivery system has received increasing interest from scientists worldwide. This work presented the preparation of the MIL-100(Fe) metal-organic framework from the organic ligand of trimesic acid and iron ions with ultrasonic assistance. Scanning electron microscopy (SEM), Brunauer-Emmett-Teller surface area (BET), X-ray diffraction (XRD), infrared spectroscopy (FTIR), and Raman spectroscopy were employed to characterize the prepared MIL-100(Fe) material. MIL-100(Fe) materials synthesized by the ultrasonic method have uniform particle morphology ranging from 100 to 300 nm with a surface area of 1033 m2/g. The prepared MIL-100(Fe) was employed as a carrier for delivering chloroquine drug with a maximal loading capacity of 220 mg/g. The MIL-100(Fe)@chloroquine system was also characterized in detail. The delivery system's slow drug release was studied, showing that nearly 80% of chloroquine molecules were released after 7.5 h of immersing time in PBS and simulated gastric solutions and completely detached from the MIL-100(Fe)@chloroquine system only after approximately 80 h. This result shows the ability to control chloroquine drug release of the material, reducing the possibility of drug shock.
Collapse
Affiliation(s)
- Bac Thanh Le
- Institute of Chemistry and
Materials, Nghia Do, Cau Giay, Hanoi 100000, Vietnam
| | - Duong Duc La
- Institute of Chemistry and
Materials, Nghia Do, Cau Giay, Hanoi 100000, Vietnam
| | | |
Collapse
|
7
|
Macrophage-evading and tumor-specific apoptosis inducing nanoparticles for targeted cancer therapy. Acta Pharm Sin B 2023; 13:327-343. [PMID: 36815044 PMCID: PMC9939305 DOI: 10.1016/j.apsb.2022.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/29/2022] [Accepted: 04/08/2022] [Indexed: 12/18/2022] Open
Abstract
Extended circulation of anticancer nanodrugs in blood stream is essential for their clinical applications. However, administered nanoparticles are rapidly sequestered and cleared by cells of the mononuclear phagocyte system (MPS). In this study, we developed a biomimetic nanosystem that is able to efficiently escape MPS and target tumor tissues. The fabricated nanoparticles (TM-CQ/NPs) were coated with fibroblast cell membrane expressing tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL). Coating with this functionalized membrane reduced the endocytosis of nanoparticles by macrophages, but increased the nanoparticle uptake in tumor cells. Importantly, this membrane coating specifically induced tumor cell apoptosis via the interaction of TRAIL and its cognate death receptors. Meanwhile, the encapsulated chloroquine (CQ) further suppressed the uptake of nanoparticles by macrophages, and synergized with TRAIL to induce tumor cell apoptosis. The vigorous antitumor efficacy in two mice tumor models confirmed our nanosystem was an effective approach to address the MPS challenge for cancer therapy. Together, our TM-CQ/NPs nanosystem provides a feasible approach to precisely target tumor tissues and improve anticancer efficacy.
Collapse
|
8
|
Yang X, Wen X, Dai J, Chen Y, Ding W, Wang J, Gu X, Zhang X, Chen J, Sutliff RL, Emory SR, Ruan G. Probing the Intracellular Delivery of Nanoparticles into Hard-to-Transfect Cells. ACS NANO 2022; 16:8751-8765. [PMID: 35579595 DOI: 10.1021/acsnano.1c07648] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Hard-to-transfect cells are cells that are known to present special difficulties in intracellular delivery of exogenous entities. However, the special transport behaviors underlying the special delivery problem in these cells have so far not been examined carefully. Here, we combine single-particle motion analysis, cell biology studies, and mathematical modeling to investigate nanoparticle transport in bone marrow-derived mesenchymal stem cells (BMSCs), a technologically important type of hard-to-transfect cells. Tat peptide-conjugated quantum dots (QDs-Tat) were used as the model nanoparticles. Two different yet complementary single-particle methods, namely, pair-correlation function and single-particle tracking, were conducted on the same cell samples and on the same viewing stage of a confocal microscope. Our results reveal significant differences in each individual step of transport of QDs-Tat in BMSCs vs a commonly used model cell line, HeLa cells. Single-particle motion analysis demonstrates that vesicle escape and cytoplasmic diffusion are dramatically more difficult in BMSCs than in HeLa cells. Cell biology studies show that BMSCs use different biological pathways for the cellular uptake, vesicular transport, and exocytosis of QDs-Tat than HeLa cells. A reaction-diffusion-advection model is employed to mathematically integrate the individual steps of cellular transport and can be used to predict and design nanoparticle delivery in BMSCs. This work provides dissective, quantitative, and mechanistic understandings of nanoparticle transport in BMSCs. The investigative methods described in this work can help to guide the tailored design of nanoparticle-based delivery in specific types and subtypes of hard-to-transfect cells.
Collapse
Affiliation(s)
- Xuan Yang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China 210023
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China 215123
- Nanobiotechnology & Nanomedicine Center, Xi'an Jiaotong-Liverpool University, Suzhou, China 215123
- Institute of Materials Engineering of Nanjing University, Nantong, China 226001
- Shenzhen Research Institute of Nanjing University, Shenzhen, China 518063
- Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, China 210093
| | - Xiaowei Wen
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China 210023
- Institute of Materials Engineering of Nanjing University, Nantong, China 226001
- Shenzhen Research Institute of Nanjing University, Shenzhen, China 518063
- Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, China 210093
| | - Jie Dai
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China 210023
- Institute of Materials Engineering of Nanjing University, Nantong, China 226001
- Shenzhen Research Institute of Nanjing University, Shenzhen, China 518063
- Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, China 210093
| | - Yanming Chen
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China 210023
- Institute of Materials Engineering of Nanjing University, Nantong, China 226001
- Shenzhen Research Institute of Nanjing University, Shenzhen, China 518063
- Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, China 210093
| | - Wanchuan Ding
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China 210023
- Institute of Materials Engineering of Nanjing University, Nantong, China 226001
- Shenzhen Research Institute of Nanjing University, Shenzhen, China 518063
- Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, China 210093
| | - Jun Wang
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China 211166
| | - Xiang Gu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China 210023
- Institute of Materials Engineering of Nanjing University, Nantong, China 226001
- Shenzhen Research Institute of Nanjing University, Shenzhen, China 518063
- Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, China 210093
| | - Xuejin Zhang
- Department of Quantum Electronics and Optical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China 210023
- National Center of Microstructure and Quantum Manipulation, National Lab of Solid State Microstructure, Nanjing University, Nanjing, China 210093
| | - Jin Chen
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China 211166
| | - Roy L Sutliff
- Division of Pulmonary, Allergy, Critical Care, and Sleep, School of Medicine, Emory University, Atlanta, Georgia 30322, United States
| | - Steven R Emory
- Department of Chemistry, Western Washington University, Bellingham, Washington 98225, United States
| | - Gang Ruan
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China 210023
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, China 215123
- Nanobiotechnology & Nanomedicine Center, Xi'an Jiaotong-Liverpool University, Suzhou, China 215123
- Institute of Materials Engineering of Nanjing University, Nantong, China 226001
- Shenzhen Research Institute of Nanjing University, Shenzhen, China 518063
- Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, China 210093
| |
Collapse
|
9
|
Zhang Y, Gao LX, Wang W, Zhang T, Dong FY, Ding WP. M 6 A Demethylase FTO Regulates Cisplatin Resistance of Gastric Cancer by Modulating Autophagy Activation via ULK1. Cancer Sci 2022; 113:3085-3096. [PMID: 35730319 PMCID: PMC9459343 DOI: 10.1111/cas.15469] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 11/30/2022] Open
Abstract
Drug resistance is an important factor for treatment failure of gastric cancer. N6‐methyladenosine (m6A) is the predominant mRNA internal modification in eukaryotes. The roles of m6A modification in drug resistance of gastric cancer remains unclear. In the present study, the m6A methylated RNA level was significantly decreased while the expression of m6A demethylase fat mass and obesity‐associated protein (FTO) was obviously elevated in cisplatin‐resistant (SGC‐7901/DDP) gastric cancer cells. Knockdown of FTO reversed cisplatin resistance of SGC‐7901/DDP cells both in vitro and in vivo, which was attributed to the inhibition of Unc‐51‐like kinase 1 (ULK1)‐mediated autophagy. Mechanistically, ULK1 expression was regulated in an FTO‐m6A‐dependent and YTHDF2‐mediated manner. Collectively, our findings indicate that the FTO/ULK1 axis exerts crucial roles in cisplatin resistance of gastric cancer.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China.,Department of Gastroenterology, The First Aflliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Ling-Xi Gao
- Department of Gastroenterology, The First Aflliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Wen Wang
- Department of Gastroenterology, The First Aflliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Teng Zhang
- Department of Gastroenterology, The First Aflliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| | - Fang-Yi Dong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wen-Ping Ding
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, China.,Department of Radiotherapy, The First Aflliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, China
| |
Collapse
|
10
|
Joint Strategy of PEG-PEI/CDs-E64d Nanoagents for Effective Low-Temperature Photothermal Therapy. J CLUST SCI 2022. [DOI: 10.1007/s10876-022-02262-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
11
|
Zhang X, Wei P, Wang Z, Zhao Y, Xiao W, Bian Y, Liang D, Lin Q, Song W, Jiang W, Wang H. Herceptin-Conjugated DOX-Fe 3O 4/P(NIPAM-AA-MAPEG) Nanogel System for HER2-Targeted Breast Cancer Treatment and Magnetic Resonance Imaging. ACS APPLIED MATERIALS & INTERFACES 2022; 14:15956-15969. [PMID: 35378977 DOI: 10.1021/acsami.1c24770] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
It is essential to synthesize a "diagnosis and therapy" integration nanocarrier for magnetic resonance imaging-guided breast cancer-targeted chemotherapy. Here, we report Fe3O4/P(NIPAM-AA-MAPEG) nanogels (MNLs) based on in situ loading of doxorubicin (DOX) by miniemulsion polymerization. Especially, propyl acrylic acid (AA) moieties were introduced to absorb DOX by electrostatic interactions and conjugated with the antibody herceptin (HER) through the amino-carboxyl coupling reaction. The size and morphology of MNLs could be adjusted by varying the polymerization parameters, such as the monomer feeding ratio, ferrofluid content, and cross-linker content. The MNLs showed superior stability in a physiological environment, but their structures were destroyed in an acidic environment to accelerate DOX release. The dissociation of the HER-DOX-MNLs accelerated the delivery of DOX and enhanced the therapeutic effects. The studies exhibited that the HER-DOX-MNLs could inhibit the tumor growth. In addition, the MNLs with a high magnetic content had the potential advantages in magnetic resonance imaging (MRI) of breast cancer diagnosis. The dual-targeted pH-responsive nanogels were successfully designed as a multifunctional nanocarrier for realizing HER2-positive breast cancer chemotherapy and diagnostics.
Collapse
Affiliation(s)
- Xiaojuan Zhang
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- National Special Superfine Powder Engineering Research Center, Nanjing University of Science and Technology, Nanjing 210094, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Jinling Institute of Technology, Nanjing 211169, China
| | - Pengfei Wei
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
| | - Zhao Wang
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Jinling Institute of Technology, Nanjing 211169, China
| | - Yuan Zhao
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Jinling Institute of Technology, Nanjing 211169, China
| | - Wenke Xiao
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- College of Energy Materials and Chemical Engineering, Hefei University, Hefei 230601, China
| | - Yong Bian
- Laboratory Animal Center, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Dong Liang
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Jinling Institute of Technology, Nanjing 211169, China
| | - Qing Lin
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Jinling Institute of Technology, Nanjing 211169, China
| | - Wenli Song
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Jinling Institute of Technology, Nanjing 211169, China
| | - Wei Jiang
- National Special Superfine Powder Engineering Research Center, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Huan Wang
- School of Material Engineering, Jinling Institute of Technology, Nanjing 211169, China
- Nanjing Key Laboratory of Optometric Materials and Technology, Jinling Institute of Technology, Nanjing 211169, China
| |
Collapse
|
12
|
Paskeh MDA, Entezari M, Clark C, Zabolian A, Ranjbar E, Farahani MV, Saleki H, Sharifzadeh SO, Far FB, Ashrafizadeh M, Samarghandian S, Khan H, Ghavami S, Zarrabi A, Łos MJ. Targeted regulation of autophagy using nanoparticles: New insight into cancer therapy. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166326. [DOI: 10.1016/j.bbadis.2021.166326] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/31/2021] [Accepted: 12/11/2021] [Indexed: 12/12/2022]
|
13
|
Shabani Z, Rahbarghazi R, Karimipour M, Ghadiri T, Salehi R, Sadigh‐Eteghad S, Farhoudi M. Transplantation of bioengineered Reelin-loaded PLGA/PEG micelles can accelerate neural tissue regeneration in photothrombotic stroke model of mouse. Bioeng Transl Med 2021; 7:e10264. [PMID: 35111956 PMCID: PMC8780906 DOI: 10.1002/btm2.10264] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/20/2021] [Accepted: 10/16/2021] [Indexed: 01/09/2023] Open
Abstract
Ischemic stroke is characterized by extensive neuronal loss, glial scar formation, neural tissue degeneration that leading to profound changes in the extracellular matrix, neuronal circuitry, and long-lasting functional disabilities. Although transplanted neural stem cells (NSCs) can recover some of the functional deficit after stroke, retrieval is not complete and repair of lost tissue is negligible. Therefore, the current challenge is to use the combination of NSCs with suitably enriched biomaterials to retain these cells within the infarct cavity and accelerate the formation of a de novo tissue. This study aimed to test the regenerative potential of polylactic-co-glycolic acid-polyethylene glycol (PLGA-PEG) micelle biomaterial enriched with Reelin and embryonic NSCs on photothrombotic stroke model of mice to gain appropriate methods in tissue engineering. For this purpose, two sets of experiments, either in vitro or in vivo models, were performed. In vitro analyses exhibited PLGA-PEG plus Reelin-induced proliferation rate (Ki-67+ NSCs) and neurite outgrowth (axonization and dendritization) compared to PLGA-PEG + NSCs and Reelin + NSCs groups (p < 0.05). Besides, neural differentiation (Map-2+ cells) was high in NSCs cultured in the presence of Reelin-loaded PLGA-PEG micelles (p < 0.05). Double immunofluorescence staining showed that Reelin-loaded PLGA-PEG micelles increased the number of migrating neural progenitor cells (DCX+ cells) and mature neurons (NeuN+ cells) around the lesion site compared to the groups received PLGA-PEG and Reelin alone after 1 month (p < 0.05). Immunohistochemistry results showed that the PLGA/PEG plus Reelin significantly decreased the astrocytic gliosis and increased local angiogenesis (vWF-positive cells) relative to the other groups. These changes led to the reduction of cavity size in the Reelin-loaded PLGA-PEG+NSCs group. Neurobehavioral tests indicated Reelin-loaded PLGA-PEG+NSCs promoted neurological outcome and functional recovery (p < 0.05). These results indicated that Reelin-loaded PLGA-PEG is capable of promoting NSCs dynamic growth, neuronal differentiation, and local angiogenesis following ischemic injury via providing a desirable microenvironment. These features can lead to neural tissue regeneration and functional recovery.
Collapse
Affiliation(s)
- Zahra Shabani
- Neurosciences Research Center (NSRC)Tabriz University of Medical SciencesTabrizIran,Department of Neurosciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Reza Rahbarghazi
- Stem Cell Research CenterTabriz University of Medical SciencesTabrizIran,Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Mohammad Karimipour
- Department of Neurosciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran,Department of Anatomical Sciences, Faculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Tahereh Ghadiri
- Department of Neurosciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Saeed Sadigh‐Eteghad
- Neurosciences Research Center (NSRC)Tabriz University of Medical SciencesTabrizIran
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC)Tabriz University of Medical SciencesTabrizIran
| |
Collapse
|
14
|
Baati T, Njim L, Jaafoura S, Aouane A, Neffati F, Ben Fradj N, Kerkeni A, Hammami M, Hosni K. Assessment of Pharmacokinetics, Toxicity, and Biodistribution of a High Dose of Titanate Nanotubes Following Intravenous Injection in Mice: A Promising Nanosystem of Medical Interest. ACS OMEGA 2021; 6:21872-21883. [PMID: 34497882 PMCID: PMC8412905 DOI: 10.1021/acsomega.1c01733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/02/2021] [Indexed: 05/10/2023]
Abstract
Titanate nanotubes (TiNTs) produced by the static hydrothermal process present a promising nanosystem for nanomedicine. However, the behavior of these nanotubes in vivo is not yet clarified. In this work, for the first time, we investigated the toxicity of these materials, their pharmacokinetic profile, and their biodistribution in mice. A high dose of TiNTs (45 mg/kg) was intravenously injected in mice and monitored from 6 h to 45 days. The histological examination of organs and the analysis of liver and kidney function markers and then the inflammatory response were in agreement with a long-term innocuity of these nanomaterials. The parameters of pharmacokinetics revealed the rapid clarification of TiNTs from the bloodstream after 6 h of the intravenous injection which then mainly accumulated in the liver and spleen, and their degradation and clearance in these tissues were relatively slow (>4 weeks). Interestingly, an important property of these materials is their slow dissolution under the lysosome acid environment, rendering them biodegradable. It is noteworthy that TiNTs were directly eliminated in urine and bile ducts without obvious toxicity in mice. Altogether, all these typical in vivo tests studying the TiNT pharmacokinetics, toxicity, and biodistribution are supporting the use of these biocompatible nanomaterials in the biomedical field, especially as a nanocarrier-based drug delivery system.
Collapse
Affiliation(s)
- Tarek Baati
- Laboratoire
des Substances Naturelles, Institut National
de Recherche et d’Analyse Physico-Chimique, Biotechpole Sidi Thabet 2020, Tunisie
| | - Leila Njim
- Service
d’Anatomie Pathologique, CHU de Monastir, Monastir 5000, Tunisie
| | - Sabra Jaafoura
- Laboratoire
des Substances Naturelles, Institut National
de Recherche et d’Analyse Physico-Chimique, Biotechpole Sidi Thabet 2020, Tunisie
- Laboratoire
ABCDF (LR12ES10), Faculté de Médecine Dentaire, Université de Monastir, Monastir 5000, Tunisie
| | - Aicha Aouane
- Centre
de Microscopie Electronique, Université
Aix-Marseille, IBDML
Campus Luminy, Marseille 3007, France
| | - Fadoua Neffati
- Laboratoire
de Biochimie et de Toxicologie, CHU de Monastir, Monastir 5000, Tunisie
| | - Nadia Ben Fradj
- Laboratoire
de Pharmacologie, Faculté de Médecine
de Monastir, Monastir 5000, Tunisie
| | - Abdelhamid Kerkeni
- Laboratoire
de Biophysique, Faculté de Médecine
de Monastir, Monastir 5000, Tunisie
| | - Mohamed Hammami
- Laboratoire
des Substances Naturelles, Institut National
de Recherche et d’Analyse Physico-Chimique, Biotechpole Sidi Thabet 2020, Tunisie
| | - Karim Hosni
- Laboratoire
des Substances Naturelles, Institut National
de Recherche et d’Analyse Physico-Chimique, Biotechpole Sidi Thabet 2020, Tunisie
| |
Collapse
|
15
|
Xu S, Zhong Y, Nie C, Pan Y, Adeli M, Haag R. Co-Delivery of Doxorubicin and Chloroquine by Polyglycerol Functionalized MoS2 Nanosheets for Efficient Multidrug-Resistant Cancer Therapy. Macromol Biosci 2021; 21:e2100233. [PMID: 34411417 DOI: 10.1002/mabi.202100233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/04/2021] [Indexed: 01/11/2023]
Abstract
2D MoS2 has shown a great potential in biomedical applications, due to its superior loading capacity, photothermal property, and biodegradation. In this work, polyglycerol functionalized MoS2 nanosheets with photothermal and pH dual-stimuli responsive properties are used for the co-delivery of doxorubicin and chloroquine and treatment of multidrug-resistant HeLa (HeLa-R) cells. The polyglycerol functionalized MoS2 nanosheets with 80 nm average size show a high biocompatibility and loading efficiency (≈90%) for both drugs. The release of drugs from the nanosheets at pH 5.5 is significantly promoted by laser irradiation leading to efficient destruction of incubated HeLa-R cells. In vitro evaluation shows that the designed nanoplatform has a high ability to kill HeLa-R cells. Confocal experiments demonstrate that the synthesized drug delivery system enhances the cellular uptake of DOX via folic acid targeting ligand. Taking advantage of the combined properties including biocompatibility and targeting ability as well as high loading capacity and photothermal release, this multifunctional nanosystem is a promising candidate for anticancer therapy.
Collapse
Affiliation(s)
- Shaohui Xu
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Yinan Zhong
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Chuanxiong Nie
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Yuanwei Pan
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Mohsen Adeli
- Faculty of Science, Department of Chemistry, Lorestan University, Khorramabad, Iran
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| |
Collapse
|
16
|
Sun J, Tian Q, Liu M, Su Y, Liu X, Deng Y, Song Y. Evaluation of the Antitumor Effect and Immune Response of Micelles Modified with a Polysialic Acid-D-α-Tocopheryl Polyethylene Glycol 1000 Succinate Conjugate. AAPS PharmSciTech 2021; 22:223. [PMID: 34409520 DOI: 10.1208/s12249-021-02047-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/11/2021] [Indexed: 11/30/2022] Open
Abstract
D-α-Tocopheryl polyethylene glycol 1000 succinate (TPGS) has shown potential applications in cancer therapy owing to its attractive properties, including reversal of multi-drug resistance and synergistic effects with antitumor drugs. However, its associated shortcomings cannot be underestimated, including activation of the body's immune response and acceleration of blood clearance of polyethylene glycolylated preparations. Polysialic acid (PSA) is a polysaccharide homopolymer, with the dual function of immune camouflage and tumor targeting. PSA and TPGS conjugates (PSA-TPGS) were synthesized to weaken the immune risks of TPGS. We developed PSA-TPGS and TPGS self-assembled mixed micelles and encapsulated the classical antineoplastic, docetaxel. The particle size of docetaxel-loaded mixed micelles was 16.3 ± 2.0 nm, with entrapment efficiency of 99.0 ± 0.9% and drug-loading efficiency of 3.20 ± 0.03%. Antitumor activity studies revealed that the mixed micelles showed better tumor inhibition than Tween 80 and TPGS micelles. Detection of the accelerated blood clearance (ABC) phenomenon demonstrated that insertion of PSA-TPGS into the micelles weakened the ABC phenomenon induced by TPGS. In summary, PSA-TPGS could be a potential nanocarrier to improve antitumor activity and weaken immune responses.
Collapse
|
17
|
Cai H, Wang R, Guo X, Song M, Yan F, Ji B, Liu Y. Combining Gemcitabine-Loaded Macrophage-like Nanoparticles and Erlotinib for Pancreatic Cancer Therapy. Mol Pharm 2021; 18:2495-2506. [PMID: 34078087 DOI: 10.1021/acs.molpharmaceut.0c01225] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pancreatic cancer is a lethal malignancy with a dismal prognosis. Gemcitabine is currently used to treat pancreatic cancer, but it is limited by significant toxicity. Clinical trials on the combination of gemcitabine and erlotinib reported unsatisfactory outcomes along with concerns of toxicity. The encapsulation of chemotherapy drugs in polylactic-co-glycolic acid (PLGA) nanoparticles (NPs) can alleviate toxicity through targeted delivery and sustained release. In addition, camouflaging the NPs with a macrophage membrane can evade the immune system and further improve tumor homing. We designed gemcitabine-loaded PLGA NPs with a macrophage membrane coating (MPGNPs) to reduce drug toxicity and increase the accumulation in the tumor. The combination of MPGNPs and erlotinib synergistically inhibited pancreatic cancer cell proliferation in vitro and in vivo by targeting the PI3K/AKT/mTOR and Ras/Raf/MEK/ERK signaling pathways. The MPGNPs were also able to evade phagocytosis and achieve passive targeting to the pancreatic tumors. The combination of MPGNPs and erlotinib showed synergistic anti-tumor efficacy in vitro and in vivo. This study provides a proof-of-concept for treating pancreatic cancer with a combination of MPGNPs and erlotinib.
Collapse
Affiliation(s)
- Hongqiao Cai
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Ruobing Wang
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Xingren Guo
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Meiyu Song
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, International Joint Research Laboratory of Nano-Micro Architecture Chemistry (NMAC), International Research Center for Chemistry-Medicine Joint Innovation, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, International Joint Research Laboratory of Nano-Micro Architecture Chemistry (NMAC), International Research Center for Chemistry-Medicine Joint Innovation, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, China
| | - Bai Ji
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China
| |
Collapse
|
18
|
Cai Q, Jiang J, Zhang H, Ge P, Yang L, Zhu W. Reduction-Responsive Anticancer Nanodrug Using a Full Poly(ethylene glycol) Carrier. ACS APPLIED MATERIALS & INTERFACES 2021; 13:19387-19397. [PMID: 33876927 DOI: 10.1021/acsami.1c04648] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Poly(ethylene glycol) (PEG) is applied extensively in biomedical fields because of its nontoxic, nonimmunogenic, and protein resistance properties. However, the strong hydrophilicity of PEG prevents it from self-assembling into an amphiphilic micelle in water, making it a challenge to fabricate a full-PEG carrier to deliver hydrophobic anticancer drugs. Herein, a paclitaxel (PTX)-loaded nanodrug was readily prepared through self-assembly of PTX and an amphiphilic PEG derivative, which was synthesized via melt polycondensation of two PEG diols (i.e., PEG200 and PEG10k) and mercaptosuccinic acid. The full PEG component endows the nanocarrier with good biocompatibility. Furthermore, because of the core cross-linked structure via the oxidation of mercapto groups, the nanodrug can be selectively disassociated under an intratumor reductive microenvironment through the reduction of disulfide bonds to release the loaded PTX and kill the cancer cells while maintaining high stability under the extratumor physiological condition. Additionally, it was confirmed that the nanodrug not only prolongs the biocirculation time of PTX but also possesses excellent in vivo antitumor efficacy while avoiding side effects of free PTX, for example, liver damage, which is promising for delivering clinical hydrophobic drugs to treat a variety of malignant tumors.
Collapse
Affiliation(s)
- Qiuquan Cai
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jiahong Jiang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Hongjie Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Pengfei Ge
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Liu Yang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Weipu Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
- Key Laboratory of Adsorption and Separation Materials & Technologies of Zhejiang Province, Hangzhou 310027, China
| |
Collapse
|
19
|
Alven S, Aderibigbe BA. The Therapeutic Efficacy of Dendrimer and Micelle Formulations for Breast Cancer Treatment. Pharmaceutics 2020; 12:E1212. [PMID: 33333778 PMCID: PMC7765183 DOI: 10.3390/pharmaceutics12121212] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is among the most common types of cancer in women and it is the cause of a high rate of mortality globally. The use of anticancer drugs is the standard treatment approach used for this type of cancer. However, most of these drugs are limited by multi-drug resistance, drug toxicity, poor drug bioavailability, low water solubility, poor pharmacokinetics, etc. To overcome multi-drug resistance, combinations of two or more anticancer drugs are used. However, the combination of two or more anticancer drugs produce toxic side effects. Micelles and dendrimers are promising drug delivery systems that can overcome the limitations associated with the currently used anticancer drugs. They have the capability to overcome drug resistance, reduce drug toxicity, improve the drug solubility and bioavailability. Different classes of anticancer drugs have been loaded into micelles and dendrimers, resulting in targeted drug delivery, sustained drug release mechanism, increased cellular uptake, reduced toxic side effects of the loaded drugs with enhanced anticancer activity in vitro and in vivo. This review article reports the biological outcomes of dendrimers and micelles loaded with different known anticancer agents on breast cancer in vitro and in vivo.
Collapse
Affiliation(s)
| | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa;
| |
Collapse
|
20
|
Liu P, Liu X, Cheng Y, Zhong S, Shi X, Wang S, Liu M, Ding J, Zhou W. Core-Shell Nanosystems for Self-Activated Drug-Gene Combinations against Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2020; 12:53654-53664. [PMID: 33205940 DOI: 10.1021/acsami.0c15089] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The combination of gene therapy with chemotherapeutics provides an efficacious strategy for enhanced tumor therapy. RNA-cleaving DNAzyme has been recognized as a promising gene-silencing tool, while its combination with chemotherapeutic drugs has been limited by the lack of an effective codelivery system to allow sufficient intracellular DNAzyme activation, which requires specific metal ions as a cofactor. Here, a self-activatable DNAzyme/drug core-shell codelivery system is fabricated to combat triple-negative breast cancer (TNBC). The hydrophobic chemotherapeutic, rapamycin (RAP), is self-assembled into the pure drug nanocore, and the metal-organic framework (MOF) shell based on coordination between Mn2+ and tannic acid (TA) is coated on the surface to coload an autophagy-inhibiting DNAzyme. The nanosystem efficiently delivers the payloads into tumor cells, and upon endocytosis, the MOF shell is disintegrated to release the therapeutics in response to an acidic endo/lysosome environment and intracellular glutathione (GSH). Notably, the coreleased Mn2+ serves as the cofactor of DNAzyme for effective self-activation, which suppresses the expression of Beclin 1 protein, the key initiator of autophagy, resulting in a significantly strengthened antitumor effect of RAP. Using tumor-bearing mouse models, the nanosystem could passively accumulate into the tumor tissue, impose potent gene-silencing efficacy, and thus sensitize chemotherapy to inhibit tumor growth upon intravenous administration, providing opportunities for combined gene-drug TNBC therapy.
Collapse
Affiliation(s)
- Peng Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Xuanjun Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Yan Cheng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Shenghui Zhong
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
- School of Medicine, Yichun University, Yichun, Jiangxi 336000, China
| | - Xinyi Shi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Shengfeng Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
- Department of Pharmacy, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Miao Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
- Department of Pharmacy, Hunan Cancer Hospital, Changsha, Hunan 410011, China
| | - Jinsong Ding
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Wenhu Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| |
Collapse
|
21
|
Yang B, Shi J. Developing New Cancer Nanomedicines by Repurposing Old Drugs. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202004317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Bowen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
| |
Collapse
|
22
|
Yang B, Shi J. Developing New Cancer Nanomedicines by Repurposing Old Drugs. Angew Chem Int Ed Engl 2020; 59:21829-21838. [DOI: 10.1002/anie.202004317] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Bowen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
| |
Collapse
|
23
|
Raychaudhuri R, Pandey A, Hegde A, Abdul Fayaz SM, Chellappan DK, Dua K, Mutalik S. Factors affecting the morphology of some organic and inorganic nanostructures for drug delivery: characterization, modifications, and toxicological perspectives. Expert Opin Drug Deliv 2020; 17:1737-1765. [PMID: 32878492 DOI: 10.1080/17425247.2020.1819237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Introduction: In this review, we aim to highlight the impact of various processes and formulation variables influencing the characteristics of certain surfactant-based nanoconstructs for drug delivery. Areas covered: The review includes the discussion on processing parameters for the preparation of nanoconstructs, especially those made up of surfactants. Articles published in last 15 years (437) were reviewed, 381 articles were selected for data review and most appropriate articles (215) were included in article. Effect of variables such as surfactant concentration and type, membrane additives, temperature, and pH-dependent transitions on morphology has been highlighted along with effect of shape on nanoparticle uptake by cells. Various characterization techniques explored for these nanostructures with respect to size, morphology, lamellarity, distribution, etc., and a separate section on polymeric vesicles and the influence of block copolymers, type of block copolymer, control of block length, interaction of multiple block copolymers on the structure of polymersomes and chimeric nanostructures have been discussed. Finally, applications, modification, degradation, and toxicological aspects of these drug delivery systems have been highlighted. Expert opinion: Parameters influencing the morphology of micelles and vesicles can directly or indirectly affect the efficacy of small molecule cellular internalization as well as uptake in the case of biologicals.[Figure: see text].
Collapse
Affiliation(s)
- Ruchira Raychaudhuri
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal, Karnataka State, India
| | - Abhjieet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal, Karnataka State, India
| | - Aswathi Hegde
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal, Karnataka State, India
| | - Shaik Mohammad Abdul Fayaz
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education , Manipal, Karnataka State, India
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University , Bukit Jalil, Kuala Lumpur, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney , Broadway, NSW, Australia
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education , Manipal, Karnataka State, India
| |
Collapse
|
24
|
Peyvand P, Vaezi Z, Sedghi M, Dalir N, Ma’mani L, Naderi-Manesh H. Imidazolium-based ionic liquid functionalized mesoporous silica nanoparticles as a promising nano-carrier: response surface strategy to investigate and optimize loading and release process for Lapatinib delivery. Pharm Dev Technol 2020; 25:1150-1161. [DOI: 10.1080/10837450.2020.1803909] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Parvaneh Peyvand
- Department of Biophysics/Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Zahra Vaezi
- Department of Biophysics/Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Mosslim Sedghi
- Department of Biophysics/Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| | - Nima Dalir
- Department of Physical Chemistry, Faculty of Basic Science, Tarbiat Modares University, Tehran, Iran
| | - Leila Ma’mani
- Department of Nanotechnology, Agricultural Biotechnology Research Institute of Iran (ABRII), Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Hossein Naderi-Manesh
- Department of Biophysics/Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
25
|
Sepand MR, Ranjbar S, Kempson IM, Akbariani M, Muganda WCA, Müller M, Ghahremani MH, Raoufi M. Targeting non-apoptotic cell death in cancer treatment by nanomaterials: Recent advances and future outlook. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102243. [PMID: 32623018 DOI: 10.1016/j.nano.2020.102243] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/29/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022]
Abstract
Many tumors develop resistance to most of the apoptosis-based cancer therapies. In this sense targeting non-apoptotic forms of cell death including necroptosis, autophagy and ferroptosis may have therapeutic benefits in apoptosis-defective cancer cells. Nanomaterials have shown great advantages in cancer treatment owing to their unique characteristics. Besides, the capability of nanomaterials to induce different forms of cell death has gained widespread attention in cancer treatment. Reports in this field reflect the therapeutic potential of necroptotic cell death induced by nanomaterials in cancer. Also, autophagic cell death induced by nanomaterials alone and as a part of chemo-, radio- and photothermal therapy holds great promise as anticancer therapeutic option. Besides, ferroptosis induction by iron-based nanomaterials in drug delivery, immunotherapy, hyperthermia and imaging systems shows promising results in malignancies. Hence, this review is devoted to the latest efforts and the challenges in this field of research and its clinical merits.
Collapse
Affiliation(s)
- Mohammad Reza Sepand
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sheyda Ranjbar
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ivan M Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia; School of Pharmacy and Medical Sciences, University of South Australia, SA, Australia
| | - Mostafa Akbariani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mareike Müller
- Physical Chemistry I and Research Center of Micro and Nanochemistry (Cμ), University of Siegen, Siegen, Germany
| | - Mohammad Hossein Ghahremani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Raoufi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Physical Chemistry I and Research Center of Micro and Nanochemistry (Cμ), University of Siegen, Siegen, Germany.
| |
Collapse
|
26
|
Raj EN, Lin Y, Chen C, Liu K, Chao J. Selective Autophagy Pathway of Nanoparticles and Nanodrugs: Drug Delivery and Pathophysiological Effects. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Emmanuel Naveen Raj
- Institute of Molecular Medicine and Bioengineering National Chiao Tung University Hsinchu 30068 Taiwan
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Yu‐Wei Lin
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Chien‐Hung Chen
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Kuang‐Kai Liu
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
| | - Jui‐I Chao
- Institute of Molecular Medicine and Bioengineering National Chiao Tung University Hsinchu 30068 Taiwan
- Department of Biological Science and Technology National Chiao Tung University Hsinchu 30068 Taiwan
- Center For Intelligent Drug Systems and Smart Bio‐devices National Chiao Tung University Hsinchu 30068 Taiwan
| |
Collapse
|
27
|
Xu F, Ye ML, Zhang YP, Li WJ, Li MT, Wang HZ, Qiu X, Xu Y, Yin JW, Hu Q, Wei WH, Chang Y, Liu L, Zhao Q. MicroRNA-375-3p enhances chemosensitivity to 5-fluorouracil by targeting thymidylate synthase in colorectal cancer. Cancer Sci 2020; 111:1528-1541. [PMID: 32073706 PMCID: PMC7226198 DOI: 10.1111/cas.14356] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/19/2022] Open
Abstract
Resistance to chemotherapy is a major challenge for the treatment of patients with colorectal cancer (CRC). Previous studies have found that microRNAs (miRNAs) play key roles in drug resistance; however, the role of miRNA‐373‐3p (miR‐375‐3p) in CRC remains unclear. The current study aimed to explore the potential function of miR‐375‐3p in 5‐fluorouracil (5‐FU) resistance. MicroRNA‐375‐3p was found to be widely downregulated in human CRC cell lines and tissues and to promote the sensitivity of CRC cells to 5‐FU by inducing colon cancer cell apoptosis and cycle arrest and by inhibiting cell growth, migration, and invasion in vitro. Thymidylate synthase (TYMS) was found to be a direct target of miR‐375‐3p, and TYMS knockdown exerted similar effects as miR‐375‐3p overexpression on the CRC cellular response to 5‐FU. Lipid‐coated calcium carbonate nanoparticles (NPs) were designed to cotransport 5‐FU and miR‐375‐3p into cells efficiently and rapidly and to release the drugs in a weakly acidic tumor microenvironment. The therapeutic effect of combined miR‐375 + 5‐FU/NPs was significantly higher than that of the individual treatments in mouse s.c. xenografts derived from HCT116 cells. Our results suggest that restoring miR‐375‐3p levels could be a future novel therapeutic strategy to enhance chemosensitivity to 5‐FU.
Collapse
Affiliation(s)
- Fei Xu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Ming-Liang Ye
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Yu-Peng Zhang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Wen-Jie Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Meng-Ting Li
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Hai-Zhou Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Xiao Qiu
- Department of Hematology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Yan Xu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Jin-Wen Yin
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Qian Hu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Wan-Hui Wei
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Ying Chang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Lan Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, China
| |
Collapse
|
28
|
Kommalapati VK, Kumar D, Tangutur AD. Inhibition of JNJ-26481585-mediated autophagy induces apoptosis via ROS activation and mitochondrial membrane potential disruption in neuroblastoma cells. Mol Cell Biochem 2020; 468:21-34. [PMID: 32146648 DOI: 10.1007/s11010-020-03708-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/21/2020] [Indexed: 01/14/2023]
Abstract
Neuroblastoma (NB) is the common pediatric tumor of the sympathetic nervous system characterized by poor prognosis. Owing to the challenges such as high tumor heterogeneity, multidrug resistance, minimal residual disease, etc., there is an immediate need for exploring new therapeutic strategies and effective treatments for NB. Herein, in the current study, we explored the unexplored response of NB cells to the second-generation histone deacetylase inhibitor (HDACi) JNJ-26481585(JNJ) and the lysosomotropic agent, Chloroquine (CQ) alone and upon JNJ/CQ treatment as a plausible therapeutic. We identify that while JNJ alone induced autophagy in NB cells, JNJ/CQ treatment decreased the viability and proliferation of NB cells in vitro by switching from autophagy to apoptosis. Further we found that autophagy inhibition by CQ pre-treatment led to the generation of ROS and a decrease in the mitochondrial membrane potential (MMP) that subsequently caused caspase-3-mediated apoptotic cell death in NB cells. Corroborating the above observations, we found that the ROS scavenger N-acetylcysteine (NAC) countered caspase-3 activity and the cells were rescued from apoptosis. Finally, these observations establish that JNJ/CQ treatment resulted in cell death in NB cells by triggering the formation of ROS and disruption of MMP, suggesting that modulation of JNJ-induced autophagy by CQ represents a promising new therapeutic approach in NB.
Collapse
Affiliation(s)
- Vamsi Krishna Kommalapati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500007, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana, India
| | - Dinesh Kumar
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500007, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana, India
| | - Anjana Devi Tangutur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500007, India. .,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, Telangana, India.
| |
Collapse
|
29
|
Sun K, Yu W, Ji B, Chen C, Yang H, Du Y, Song M, Cai H, Yan F, Su R. Saikosaponin D loaded macrophage membrane-biomimetic nanoparticles target angiogenic signaling for breast cancer therapy. APPLIED MATERIALS TODAY 2020; 18:100505. [DOI: 10.1016/j.apmt.2019.100505] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
|
30
|
Wu Y, Si Y, Xiang Y, Zhou T, Liu X, Wu M, Li W, Zhang T, Xiang K, Zhang L, Zhao H, Liu Y. Polyphyllin I activates AMPK to suppress the growth of non-small-cell lung cancer via induction of autophagy. Arch Biochem Biophys 2020; 687:108285. [PMID: 32074500 DOI: 10.1016/j.abb.2020.108285] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/07/2020] [Accepted: 01/29/2020] [Indexed: 12/14/2022]
Abstract
Polyphyllin I (PPI), a bioactive constituent extracted from the rhizomes of Paris polyphylla, is cytotoxic to several cancer types. This study was designed to explore whether PPI prevents non-small-cell lung cancer (NSCLC) growth and to investigate the molecular mechanism. AMP-activated protein kinase (AMPK) has been implicated in the activation of autophagy in distinct tissues. In cultured human NSCLC cell lines, PPI induces autophagy by activating AMPK and then inhibiting mTOR signaling in a concentration-dependent manner. Furthermore, the activation of autophagy induced by PPI was reversed by the AMPK inhibitor compound C. Computational docking showed that PPI directly interacted with the allosteric drug and metabolite site of AMPK to stabilize its activation. Microscale thermophoresis and Drug Affinity Responsive Targeting Stability (DARTS) assay further confirmed the high affinity between PPI and AMPK. In vivo studies indicated that PPI suppressed the growth of NSCLC and increased the levels of LC3-II and phosphorylated AMPK in tumors isolated from a xenograft model of NSCLC in mice. Moreover, PPI exhibited favorable pharmacokinetics in rats. In summary, PPI conclusively acts as a direct AMPK activator to induce cell autophagy which inhibits the growth of NSCLC cells. In the future, PPI therapy should be applied to treat patients with NSCLC.
Collapse
Affiliation(s)
- Yezi Wu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yuchen Xiang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Tong Zhou
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuewen Liu
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Mingwei Wu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Wenjuan Li
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research and Institute of Medicinal Chemistry, Hubei University of Medicine, Shiyan, Hubei, China
| | - Te Zhang
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research and Institute of Medicinal Chemistry, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ke Xiang
- Department of Science and Education, Gucheng People's Hospital, Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Liang Zhang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Huzi Zhao
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei, China; Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, Hubei, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research and Institute of Medicinal Chemistry, Hubei University of Medicine, Shiyan, Hubei, China.
| |
Collapse
|
31
|
Zhu YX, Jia HR, Gao G, Pan GY, Jiang YW, Li P, Zhou N, Li C, She C, Ulrich NW, Chen Z, Wu FG. Mitochondria-acting nanomicelles for destruction of cancer cells via excessive mitophagy/autophagy-driven lethal energy depletion and phototherapy. Biomaterials 2020; 232:119668. [DOI: 10.1016/j.biomaterials.2019.119668] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/01/2019] [Accepted: 12/04/2019] [Indexed: 12/18/2022]
|
32
|
Li X, Lu C, Xia W, Quan G, Huang Y, Bai X, Yu F, Xu Q, Qin W, Liu D, Pan X. Poly(L-Glutamic Acid)-Based Brush Copolymers: Fabrication, Self-assembly, and Evaluation as Efficient Nanocarriers for Cationic Protein Drug Delivery. AAPS PharmSciTech 2020; 21:78. [PMID: 31970547 DOI: 10.1208/s12249-020-1624-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/08/2020] [Indexed: 02/05/2023] Open
Abstract
Protein drugs were considered to be the first choice to treat many human diseases, but their clinical application was usually limited by their short half-life and lack of validated targeted therapy. Here, a series of folate-functionalized poly(ethylene glycol)-b-(poly(2-aminoethyl-L-glutamate)-g-poly(L-glutamic acid))s (FA-PEG-b-(PELG-g-PLGA)s) were designed as tumor-targeted carriers for cationic protein delivery. Compared with traditional copolymers consisting of PEG and linear charged hydrophilic blocks, FA-PEG-b-(PELG-g-PLGA) with brush-like polyelectrolyte segments were beneficial to improving their electrostatic interactions with loading protein molecules, thus increasing drug-loading stability and protecting encapsulated proteins from degradation. The designed polymer brushes could efficiently encapsulate cytochrome C (CytC), a cationic model protein, to form polyion complex (PIC) micelles with an average particle size of approximately 200 nm. An in vitro drug release study showed that the drug-loading stability of the formed PIC micelles was largely improved. The functionalization of the block copolymer carriers with a targeting folate group enhanced the tumor cell growth inhibition and total apoptotic rates induced by CytC. Our results shed light on the unique advantages of brush-like polymer carriers in delivering cationic proteins, and the poly(L-glutamic acid)-based linear-brush diblock copolymers could be applied as a versatile delivery platform for molecular targeting in cancer therapy.
Collapse
Affiliation(s)
- Xin Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China
| | - Chao Lu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Wenquan Xia
- Shantou University Medical College, Shantou, 515041, China
| | - Guilan Quan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ying Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xuequn Bai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Feiyuan Yu
- Shantou University Medical College, Shantou, 515041, China
| | - Qian Xu
- Shantou University Medical College, Shantou, 515041, China
| | - Wanbing Qin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Daojun Liu
- Shantou University Medical College, Shantou, 515041, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| |
Collapse
|
33
|
Nelemans LC, Gurevich L. Drug Delivery with Polymeric Nanocarriers-Cellular Uptake Mechanisms. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E366. [PMID: 31941006 PMCID: PMC7013754 DOI: 10.3390/ma13020366] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/17/2022]
Abstract
Nanocarrier-based systems hold a promise to become "Dr. Ehrlich's Magic Bullet" capable of delivering drugs, proteins and genetic materials intact to a specific location in an organism down to subcellular level. The key question, however, how a nanocarrier is internalized by cells and how its intracellular trafficking and the fate in the cell can be controlled remains yet to be answered. In this review we survey drug delivery systems based on various polymeric nanocarriers, their uptake mechanisms, as well as the experimental techniques and common pathway inhibitors applied for internalization studies. While energy-dependent endocytosis is observed as the main uptake pathway, the integrity of a drug-loaded nanocarrier upon its internalization appears to be a seldomly addressed problem that can drastically affect the uptake kinetics and toxicity of the system in vitro and in vivo.
Collapse
Affiliation(s)
| | - Leonid Gurevich
- Department of Materials and Production, Aalborg University, 9220 Aalborg, Denmark;
| |
Collapse
|
34
|
Chen X, Tong R, Liu B, Liu H, Feng X, Ding S, Lei Q, Tang G, Wu J, Fang W. Duo of (-)-epigallocatechin-3-gallate and doxorubicin loaded by polydopamine coating ZIF-8 in the regulation of autophagy for chemo-photothermal synergistic therapy. Biomater Sci 2020; 8:1380-1393. [PMID: 31916560 DOI: 10.1039/c9bm01614g] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To achieve highly systemic therapeutic efficacy, chemotherapy is combined with photothermal therapy for chemo-photothermal synergistic therapy; however, this strategy suffers from high toxicity and unsatisfactory sensitivity for cancer cells. Herein, we developed a pH- and photothermal-responsive zeolitic imidazolate framework (ZIF-8) compound for loading a dual-drug in the tumor site and improving their curative effects. Since autophagy always accompanies tumor progression and metastasis, there is an unmet need for an anticancer treatment related to the regulation of autophagy. Green tea polyphenols, namely, (-)-epigallocatechin-3-gallate (EGCG) and doxorubicin (DOX), both of which exhibit anticancer activity, were dual-loaded via polydopamine (PDA) coating ZIF-8 (EGCG@ZIF-PDA-PEG-DOX, EZPPD for short) through hierarchical self-assembly. PDA could transfer photothermal energy to increase the temperature under near-infrared (NIR) laser irradiation. Due to its pH-response, EZPPD released EGCG and DOX in the tumor microenvironment, wherein the temperature increased with the help of PDA and NIR laser irradiation. The duo of DOX and EGCG induced autophagic flux and accelerated the formation of autophagosomes. In a mouse HeLa tumor model, photothermal-chemotherapy could ablate the tumor with a significant synergistic effect and potentiate the anticancer efficacy. Thus, the results indicate that EZPPD renders the key traits of a clinically promising candidate to address the challenges associated with synergistic chemotherapy and photothermal utilization in antitumor therapy.
Collapse
Affiliation(s)
- Xuerui Chen
- Department of Chemistry, Zhejiang University, Hangzhou 310027, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Si Y, Wang J, Liu X, Zhou T, Xiang Y, Zhang T, Wang X, Feng T, Xu L, Yu Q, Zhao H, Liu Y. Ethoxysanguinarine, a Novel Direct Activator of AMP-Activated Protein Kinase, Induces Autophagy and Exhibits Therapeutic Potential in Breast Cancer Cells. Front Pharmacol 2020; 10:1503. [PMID: 31969821 PMCID: PMC6960228 DOI: 10.3389/fphar.2019.01503] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/20/2019] [Indexed: 12/24/2022] Open
Abstract
Ethoxysanguinarine (Eth) is a benzophenanthridine alkaloid extracted from Macleaya cordata (Willd) R. Br. It possesses antibacterial and antiviral activities and offers therapeutic benefits for the treatment of respiratory syndrome virus-induced cytopathic effects. However, the effect of Eth on human tumors and its pharmacological effects remain to be elucidated, together with its cellular target. Here, we examined the effects of Eth on breast cancer (BC) cells. We found that at low doses, Eth strongly inhibited the viability of BC cell lines and induced autophagy. Mechanistic studies showed that Eth induced autophagy by upregulating the activity of the AMP-activated protein kinase (AMPK). The AMPK inhibitor compound C significantly attenuated Eth-induced autophagy and inhibited proliferation. Meanwhile, the AMPK activator metformin significantly enhanced Eth-induced autophagy and inhibited proliferation. Computational docking and affinity assays showed that Eth directly interacted with the allosteric drug and metabolite site of AMPK to stabilize its activation. AMPK was less activated in tumor samples compared to normal breast tissues and was inversely associated with the prognosis of the patients. Moreover, Eth exhibited potent anti-BC activity in nude mice and favorable pharmacokinetics in rats. These characteristics render Eth as a promising candidate drug for further development and for designing new effective AMPK activators.
Collapse
Affiliation(s)
- Yuan Si
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Jiu Wang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research and Institute of Medicinal Chemistry, Hubei University of Medicine, Shiyan, China
| | - Xuewen Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Tong Zhou
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Yuchen Xiang
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Te Zhang
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Xianhui Wang
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Tingting Feng
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Li Xu
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Qingqing Yu
- Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Huzi Zhao
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China
| | - Ying Liu
- Laboratory of Molecular Target Therapy of Cancer, Institute of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Laboratory of Molecular Target Therapy of Cancer, Biomedical Research Institute, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research and Institute of Medicinal Chemistry, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
36
|
Chang H, Zou Z, Wang Q, Li J, Jin H, Yin Q, Xing D. Targeting and Specific Activation of Antigen-Presenting Cells by Endogenous Antigen-Loaded Nanoparticles Elicits Tumor-Specific Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1900069. [PMID: 31921548 PMCID: PMC6947714 DOI: 10.1002/advs.201900069] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 08/03/2019] [Indexed: 05/19/2023]
Abstract
Immunotherapy has shown tremendous promise for improving cancer treatment. Unfortunately, antigen-presenting cells (APCs) in cancer patients cannot effectively recognize and process tumor antigens to activate host immune responses. In this study, an approach is developed to improve cancer immunotherapy that utilizes endogenous antigen-carrying nanoparticles (EAC-NPs), which encompasses a set of antigens isolated from solid tumors and adjuvants. The EAC-NPs specifically target APCs and subsequently result in enhanced T cell responses and improved antitumor efficacy. Mechanistic studies reveal that the EAC-NPs enhance and prolong the presence of immune compounds in APCs, which ensure persistent antigen loading and stimulation, induce a rapid proliferation of CD4+ and CD8+ T cells, and significantly increase the ratios of intratumoral CD4+ T/Treg and CD8+ T/Treg. The work using nanotechnology provides a promising strategy in improving antitumor immunity by enhancing the immunogenicity and presentation of tumor self-antigens for cancer immunotherapy.
Collapse
Affiliation(s)
- Hao‐Cai Chang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Zheng‐Zhi Zou
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Qiu‐Hong Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Jie Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Huan Jin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Qian‐Xia Yin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| |
Collapse
|
37
|
Xin X, Du X, Xiao Q, Azevedo HS, He W, Yin L. Drug Nanorod-Mediated Intracellular Delivery of microRNA-101 for Self-sensitization via Autophagy Inhibition. NANO-MICRO LETTERS 2019; 11:82. [PMID: 34138035 PMCID: PMC7770860 DOI: 10.1007/s40820-019-0310-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 08/30/2019] [Indexed: 05/20/2023]
Abstract
Autophagy is closely related to the drug resistance and metastasis in cancer therapy. Nanoparticle-mediated co-delivery of combinatorial therapy with small-molecular drugs and nucleic acids is promising to address drug resistance. Here, a drug-delivering-drug (DDD) platform consisting of anti-tumor-drug nanorods as a vehicle for cytosol delivery of nucleic acid (miR-101) with potent autophagic-inhibition activity is reported for combinatorial therapy. The developed 180-nm nanoplatform, with total drug loading of up to 66%, delivers miR-101 to cancer cells, with threefold increase in intracellular level compared to conventional gene carriers and inhibits the autophagy significantly, along with above twofold reduction in LC3II mRNA and approximately fivefold increase in p62 mRNA over the control demonstrated in the results in vivo. And in turn, the delivery of miR-101 potentiates the drug's ability to kill cancer cells, with a threefold increase in apoptosis over that of chemotherapy alone. The anti-tumor study in vivo indicates the combined therapy that enables a reduction of 80% in tumor volume and > twofold increase in apoptosis than of the single-drug strategy. In summary, via the carrier-free strategy of DDD, this work provides a delivery platform that can be easily customized to overcome drug resistance and facilitates the delivery of combined therapy of small-molecular drugs and nucleic acids.
Collapse
Affiliation(s)
- Xiaofei Xin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Xiaoqing Du
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Qingqing Xiao
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Helena S Azevedo
- School of Engineering and Materials Science, Institute of Bioengineering, Queen Mary, University of London, London, E1 4NS, UK
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| | - Lifang Yin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
38
|
Tavakol S, Ashrafizadeh M, Deng S, Azarian M, Abdoli A, Motavaf M, Poormoghadam D, Khanbabaei H, Afshar EG, Mandegary A, Pardakhty A, Yap CT, Mohammadinejad R, Kumar AP. Autophagy Modulators: Mechanistic Aspects and Drug Delivery Systems. Biomolecules 2019; 9:E530. [PMID: 31557936 PMCID: PMC6843293 DOI: 10.3390/biom9100530] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 09/14/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy modulation is considered to be a promising programmed cell death mechanism to prevent and cure a great number of disorders and diseases. The crucial step in designing an effective therapeutic approach is to understand the correct and accurate causes of diseases and to understand whether autophagy plays a cytoprotective or cytotoxic/cytostatic role in the progression and prevention of disease. This knowledge will help scientists find approaches to manipulate tumor and pathologic cells in order to enhance cellular sensitivity to therapeutics and treat them. Although some conventional therapeutics suffer from poor solubility, bioavailability and controlled release mechanisms, it appears that novel nanoplatforms overcome these obstacles and have led to the design of a theranostic-controlled drug release system with high solubility and active targeting and stimuli-responsive potentials. In this review, we discuss autophagy modulators-related signaling pathways and some of the drug delivery strategies that have been applied to the field of therapeutic application of autophagy modulators. Moreover, we describe how therapeutics will target various steps of the autophagic machinery. Furthermore, nano drug delivery platforms for autophagy targeting and co-delivery of autophagy modulators with chemotherapeutics/siRNA, are also discussed.
Collapse
Affiliation(s)
- Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Milad Ashrafizadeh
- Department of basic science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran.
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Maryam Azarian
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
- Departament de Bioquímica i Biologia Molecular, Institut de Biotecnologia i Biomedicina (IBB), Universitat Autónoma de Barcelona, Barcelona, Spain.
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | - Mahsa Motavaf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Delaram Poormoghadam
- Department of Medical Nanotechnology, Faculty of Advanced Sciences & Technology, Pharmaceutical Sciences Branch, Islamic Azad University, (IAUPS), Tehran, Iran.
| | - Hashem Khanbabaei
- Medical Physics Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Elham Ghasemipour Afshar
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Ali Mandegary
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Abbas Pardakhty
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Celestial T Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.
| |
Collapse
|
39
|
Birhan YS, Hailemeskel BZ, Mekonnen TW, Hanurry EY, Darge HF, Andrgie AT, Chou HY, Lai JY, Hsiue GH, Tsai HC. Fabrication of redox-responsive Bi(mPEG-PLGA)-Se 2 micelles for doxorubicin delivery. Int J Pharm 2019; 567:118486. [PMID: 31260783 DOI: 10.1016/j.ijpharm.2019.118486] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/15/2019] [Accepted: 06/28/2019] [Indexed: 12/12/2022]
Abstract
Stimuli-responsive polymeric nanostructures have emerged as potential drug carriers for cancer therapy. Herein, we synthesized redox-responsive diselenide bond containing amphiphilic polymer, Bi(mPEG-PLGA)-Se2 from mPEG-PLGA and 3,3'-diselanediyldipropanoic acid (DSeDPA) using DCC/DMAP as coupling agents. Due to its amphiphilic nature, Bi(mPEG-PLGA)-Se2 self-assembled in to stable micelles in aqueous solution with a hydrodynamic size of 123.9 ± 0.85 nm. The Bi(mPEG-PLGA)-Se2 micelles exhibited DOX-loading content (DLC) of 6.61 wt% and encapsulation efficiency (EE) of 54.9%. The DOX-loaded Bi(mPEG-PLGA)-Se2 micelles released 73.94% and 69.54% of their cargo within 72 h upon treatment with 6 mM GSH and 0.1% H2O2, respectively, at pH 7.4 and 37 °C. The MTT assay results demonstrated that Bi(mPEG-PLGA)-Se2 was devoid of any inherent toxicity and the DOX-loaded micelles showed pronounced antitumor activities against HeLa cells, 44.46% of cells were viable at maximum dose of 7.5 µg/mL. The cellular uptake experiment further confirmed the internalization of DOX-loaded Bi(mPEG-PLGA)-Se2 micelles and endowed redox stimuli triggered drug release in cytosol and nuclei of cancer cells. Overall, the results suggested that the smart, biocompatible Bi(mPEG-PLGA)-Se2 copolymer could serve as potential drug delivery biomaterial for the controlled release of hydrophobic drugs in cancer cells.
Collapse
Affiliation(s)
- Yihenew Simegniew Birhan
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Balkew Zewge Hailemeskel
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Tefera Worku Mekonnen
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Endiries Yibru Hanurry
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Haile Fentahun Darge
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Abegaz Tizazu Andrgie
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Hsiao-Ying Chou
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC
| | - Juin-Yih Lai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC; Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC; R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan, ROC
| | - Ging-Ho Hsiue
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan, ROC.
| | - Hsieh-Chih Tsai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC; Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei 106, Taiwan, ROC.
| |
Collapse
|
40
|
Rao J, Mei L, Liu J, Tang X, Yin S, Xia C, Wei J, Wan D, Wang X, Wang Y, Li M, Zhang Z, He Q. Size-adjustable micelles co-loaded with a chemotherapeutic agent and an autophagy inhibitor for enhancing cancer treatment via increased tumor retention. Acta Biomater 2019; 89:300-312. [PMID: 30878446 DOI: 10.1016/j.actbio.2019.03.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/01/2019] [Accepted: 03/12/2019] [Indexed: 12/16/2022]
Abstract
Autophagy plays a key role in the stress response of tumor cells, which contributes to cancer cell survival and resistance to chemotherapy by degrading cytoplasmic proteins to provide energy and clear the hazardous substances. Therefore, combined treatment of chemotherapeutics and autophagy inhibitors is thought to obtain a desirable antitumor effect. Nanoparticles (NPs) show potential in tumor-targeting drug delivery because of the enhanced permeability and retention (EPR) effect. However, NPs with fixed particle size cannot achieve optimal delivery effect. Herein, a strategy based on Cu (I)-catalyzed click chemistry-triggered aggregation of azide/alkyne-modified micelles was developed for the co-delivery of the chemotherapeutic drug doxorubicin (Dox) and the autophagy inhibitor wortmannin (Wtmn). In vitro experiments showed that the size of micelles increased in a time-dependent manner, which enhanced micelle accumulation in both B16F10 and 4 T1 cells. The fluorescence resonance energy transfer (FRET) experiment and biodistribution study further demonstrated that the aggregation of micelles through click cycloaddition significantly improved the accumulation of drug-loading micelles at the tumor region. Furthermore, the decreased amount of autophagosomes observed by transmission electron microscopy (TEM), the declined expression of LC3-II, and the increased level of p62 by western blotting and immunohistochemistry (IHC) confirmed the obvious inhibition of autophagy induced by Dox/Wtmn co-loaded size-adjustable micelles, which had a synergistic effect in cancer suppression. In addition, the co-loaded size-adjustable micelles showed outstanding cytotoxicity and antitumor effect. Therefore, this strategy effectively suppressed melanoma and breast cancer in mice. STATEMENT OF SIGNIFICANCE: The therapeutic effects of chemotherapy can be limited by autophagy; hence, combined use of autophagy inhibitors with chemotherapeutics achieves desirable anticancer efficacy. In the present study, we designed size-adjustable micelles by modifying the click reaction substrate azide group and the alkyne group on the surface of micelles, and subsequently, the autophagy inhibitor wortmannin and the chemotherapeutic drug doxorubicin were co-loaded. The micelles could aggregate by click reaction at the tumor site when the catalysts were intratumorally injected. The results showed that the size-adjustable micelles achieved efficient drug delivery, penetration, and retention in tumors; through the combined effect of wortmannin-mediated autophagy inhibition and doxorubicin-mediated cytotoxicity, this strategy exerted significant anticancer effect in melanoma and breast cancer treatment.
Collapse
|
41
|
Sun X, Jiang L, Wang C, Sun S, Mei L, Huang L. Systematic investigation of intracellular trafficking behavior of one-dimensional alumina nanotubes. J Mater Chem B 2019; 7:2043-2053. [PMID: 32254808 DOI: 10.1039/c8tb03349h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanotube materials exhibit high drug loading capacity and controlled drug release properties, providing new opportunities for drug delivery. However, the intracellular trafficking paths of 1-dimensional (1D) nanostructured materials are poorly understood compared to their spherical counterparts, impeding the broad application of 1D materials as drug carriers. Here, we report the intracellular trafficking mechanism of nontoxic and biocompatible nanomaterials called anodic alumina nanotubes (AANTs), a model for 1D materials with a geometry that can be precisely engineered. The results indicated that AANTs enter the cells mainly by caveolin endocytosis and micropinocytosis and that cells use a novel macropinocytosis-late endosomes (LEs)-lysosomes route to transport AANTs. Moreover, liposomes (marked by DsRed-Rab18) are fully involved in the classical pathway of early endosomes (EEs)/LEs developing into lysosomes. The AANTs were delivered to the cells via two pathways: slow endocytosis recycling and GLUT4 exocytosis vesicles. The AANTs also induced intracellular autophagy and then degraded via the endolysosomal pathway. Blocking endolysosomal pathways using autophagy inhibitors prevented the degradation of AANTs through lysosomes. Our results add new insights into the pathways and mechanisms of intracellular trafficking of AANTs, and suggest that intracellular trafficking and lysosomal degradation are highly interdependent and important for efficient drug delivery, and should be evaluated together for drug carrier development.
Collapse
Affiliation(s)
- Xiangyu Sun
- Department of Physics, Tsinghua University, Beijing 10008, China.
| | | | | | | | | | | |
Collapse
|
42
|
Sun JH, Ye C, Bai EH, Zhang LL, Huo SJ, Yu HH, Xiang SY, Yu SQ. Co-delivery nanoparticles of doxorubicin and chloroquine for improving the anti-cancer effect in vitro. NANOTECHNOLOGY 2019; 30:085101. [PMID: 30523865 DOI: 10.1088/1361-6528/aaf51b] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
To increase the efficacy of small molecule chemotherapeutic drug (SMCD) and reduce its toxic and side effects, we selected two model drugs doxorubicin (DOX) and chloroquine (CQ). DOX is a SMCD and CQis a chemosensitizer with autophagy inhibition. Poly(lactic-co-glycolic acid) (PLGA) and alpha-tocopherol polyethylene glycol 1000 succinate were chosen as delivery carriers to design and prepare a novel type of drug co-delivery single-nanoparticles by emulsification-solvent volatilisation, named NPDOX+CQ. The physicochemical properties of NPDOX+CQ were characterised. Then A549 cells and A549/Taxol cells were used for the in vitro anti-cancer effect study. At the same time, cellular uptake, intracellular migration and anti-cancer mechanism of nanoparticles were studied. The NPs showed a uniform spherical shape with good dispersibility, and both drugs had good encapsulation efficiency and loading capacity. In all formulations, NPDOX+CQ showed the highest in vitro cytotoxicity. The results showed that NPs could protect drugs from being recognised and excluded by P-glycoprotein (P-gp). Moreover, the results of the mechanistic study demonstrated that NPs were transported by autophagy process after being taken up by the cells. Therefore, during the migration of NPDOX+CQ, CQ could exert its efficacy and block autophagy so that DOX would not be hit by autophagy. Western Blot results showed that NPDOX+CQ had the best inhibition effect of autophagy. It can be concluded that the system can prevent the drug from being recognised and excluded by P-gp, and CQ blocks the process of autophagy so that the DOX is protected and more distributed to the nucleus of multidrug resistance (MDR) cell. The NPDOX+CQ constructed in this study provides a feasible strategy for reversing MDR in tumour cells.
Collapse
Affiliation(s)
- Jia-Hui Sun
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Zhang M, Zhang W, Tang G, Wang H, Wu M, Yu W, Zhou Z, Mou Y, Liu X. Targeted Codelivery of Docetaxel and Atg7 siRNA for Autophagy Inhibition and Pancreatic Cancer Treatment. ACS APPLIED BIO MATERIALS 2019; 2:1168-1176. [PMID: 35021365 DOI: 10.1021/acsabm.8b00764] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Miaozun Zhang
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo 315041, China
| | - Wei Zhang
- Department of Gastroenterology, Ningbo No.2 Hospital, Ningbo 315010, China
| | - Guping Tang
- Department of Chemistry, Zhejiang University, Hangzhou 310028, China
| | - Hebin Wang
- College of Life Sciences, Tarim University, Alar 843300, China
| | - Min Wu
- Department of Chemistry, Zhejiang University, Hangzhou 310028, China
| | - Weiming Yu
- Department of General Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo 315041, China
| | - Zhenfeng Zhou
- Department of Anesthesiology, Zhejiang Provincial People’s Hospital, Hangzhou 310014, China
| | - Yiping Mou
- Department of General Surgery, Zhejiang Provincial People’s Hospital, Hangzhou 310014, China
| | - Xingang Liu
- Department of Chemistry, Zhejiang University, Hangzhou 310028, China
| |
Collapse
|
44
|
Miller MA, Mikula H, Luthria G, Li R, Kronister S, Prytyskach M, Kohler RH, Mitchison T, Weissleder R. Modular Nanoparticulate Prodrug Design Enables Efficient Treatment of Solid Tumors Using Bioorthogonal Activation. ACS NANO 2018; 12:12814-12826. [PMID: 30550257 PMCID: PMC6307086 DOI: 10.1021/acsnano.8b07954] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/04/2018] [Indexed: 05/18/2023]
Abstract
Prodrug strategies that facilitate localized and controlled activity of small-molecule therapeutics can reduce systemic exposure and improve pharmacokinetics, yet limitations in activation chemistry have made it difficult to assign tunable multifunctionality to prodrugs. Here, we present the design and application of a modular small-molecule caging strategy that couples bioorthogonal cleavage with a self-immolative linker and an aliphatic anchor. This strategy leverages recently discovered in vivo catalysis by a nanoencapsulated palladium compound (Pd-NP), which mediates alloxylcarbamate cleavage and triggers release of the activated drug. The aliphatic anchor enables >90% nanoencapsulation efficiency of the prodrug, while also allowing >104-fold increased cytotoxicity upon prodrug activation. We apply the strategy to a prodrug formulation of monomethyl auristatin E (MMAE), demonstrating its ability to target microtubules and kill cancer cells only after selective activation by Pd-NP. Computational pharmacokinetic modeling provides a mechanistic basis for the observation that the nanotherapeutic prodrug strategy can lead to more selective activation in the tumor, yet in a manner that is more sensitive to variable enhanced permeability and retention (EPR) effects. Combination treatment with the nanoencapsulated MMAE prodrug and Pd-NP safely blocks tumor growth, especially when combined with a local radiation therapy regimen that is known to improve EPR effects, and represents a conceptual step forward in prodrug design.
Collapse
Affiliation(s)
- Miles A. Miller
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
- E-mail:
| | - Hannes Mikula
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Institute
of Applied Synthetic Chemistry, Vienna University
of Technology (TU Wien), Vienna 1060, Austria
| | - Gaurav Luthria
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Biomedical Informatics, Harvard Medical
School, Boston, Massachusetts 02115, United States
| | - Ran Li
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Stefan Kronister
- Institute
of Applied Synthetic Chemistry, Vienna University
of Technology (TU Wien), Vienna 1060, Austria
| | - Mark Prytyskach
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Rainer H. Kohler
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| | - Timothy Mitchison
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ralph Weissleder
- Center
for Systems Biology, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
- Department
of Radiology, Massachusetts General Hospital
and Harvard Medical School, Boston, Massachusetts 02114, United States
- Department
of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
45
|
Improving Encapsulation of Hydrophilic Chloroquine Diphosphate into Biodegradable Nanoparticles: A Promising Approach against Herpes Virus Simplex-1 Infection. Pharmaceutics 2018; 10:pharmaceutics10040255. [PMID: 30513856 PMCID: PMC6320969 DOI: 10.3390/pharmaceutics10040255] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023] Open
Abstract
Chloroquine diphosphate (CQ) is a hydrophilic drug with low entrapment efficiency in hydrophobic nanoparticles (NP). Herpes simplex virus type 1 (HSV-1) is an enveloped double-stranded DNA virus worldwide known as a common human pathogen. This study aims to develop chloroquine-loaded poly(lactic acid) (PLA) nanoparticles (CQ-NP) to improve the chloroquine anti- HSV-1 efficacy. CQ-NP were successfully prepared using a modified emulsification-solvent evaporation method. Physicochemical properties of the NP were monitored using dynamic light scattering, atomic force microscopy, drug loading efficiency, and drug release studies. Spherical nanoparticles were produced with modal diameter of <300 nm, zeta potential of −20 mv and encapsulation efficiency of 64.1%. In vitro assays of CQ-NP performed in Vero E6 cells, using the MTT-assay, revealed different cytotoxicity levels. Blank nanoparticles (B-NP) were biocompatible. Finally, the antiviral activity tested by the plaque reduction assay revealed greater efficacy for CQ-NP compared to CQ at concentrations equal to or lower than 20 µg mL−1 (p < 0.001). On the other hand, the B-NP had no antiviral activity. The CQ-NP has shown feasible properties and great potential to improve the antiviral activity of drugs.
Collapse
|
46
|
Wei F, Duan Y. Crosstalk between Autophagy and Nanomaterials: Internalization, Activation, Termination. ACTA ACUST UNITED AC 2018; 3:e1800259. [PMID: 32627344 DOI: 10.1002/adbi.201800259] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Nanomaterials (NMs) are comprehensively applied in biomedicine due to their unique physical and chemical properties. Autophagy, as an evolutionarily conserved cellular quality control process, is closely associated with the effect of NMs on cells. In this review, the recent advances in NM-induced/inhibited autophagy (NM-phagy) are summarized, with an aim to present a comprehensive description of the mechanisms of NM-phagy from the perspective of internalization, activation, and termination, thereby bridging autophagy and nanomaterials. Several possible mechanisms are extensively reviewed including the endocytosis pathway of NMs and the related cross components (clathrin and adaptor protein 2 (AP-2), adenosine diphosphate (ADP)-ribosylation factor 6 (Arf6), Rab, UV radiation resistance associated gene (UVRAG)), three main stress mechanisms (oxidative stress, damaged organelles stress, and toxicity stress), and several signal pathway-related molecules. The mechanistic insight is beneficial to understand the autophagic response to NMs or NMs' regulation of autophagy. The challenges currently encountered and research trend in the field of NM-phagy are also highlighted. It is hoped that the NM-phagy discussion in this review with the focus on the mechanistic aspects may serve as a guideline for future research in this field.
Collapse
Affiliation(s)
- Fujing Wei
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-enviroment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P. R. China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-enviroment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P. R. China
| |
Collapse
|
47
|
Karami Ghaleseiedi Z, Dadkhah Tehrani A, Parsamanesh M. Starch-based dual amphiphilic graft copolymer as a new pH-sensitive maltidrug co-delivery system. Int J Biol Macromol 2018; 118:913-920. [DOI: 10.1016/j.ijbiomac.2018.06.156] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/02/2018] [Accepted: 06/27/2018] [Indexed: 01/07/2023]
|
48
|
|
49
|
Autophagy differentially regulates TNF receptor Fn14 by distinct mammalian Atg8 proteins. Nat Commun 2018; 9:3744. [PMID: 30218067 PMCID: PMC6138730 DOI: 10.1038/s41467-018-06275-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 07/26/2018] [Indexed: 02/07/2023] Open
Abstract
Autophagy, a conserved membrane trafficking process, sequesters cytoplasmic components into autophagosomes and targets them for lysosomal degradation. The TNF receptor Fn14 participates in multiple intracellular signaling pathways and is strongly induced upon tissue injury and solid tumorigenesis. While Fn14 is a short-lived protein, the regulation of its levels is largely obscure. Here we uncover a role for autophagy in Fn14 turnover, wherein specific core autophagy Atg8 proteins play distinct roles: Fn14 accumulates in the ERGIC in absence of GABARAP but within endosomes in the vicinity of autophagic membranes in absence of GATE-16. Moreover, GABARAP regulates overall cellular levels of Fn14, whereas GATE-16 regulates TWEAK signaling by Fn14 and thereby NF-κB activity. These findings not only implicate different Atg8 proteins in distinct roles within the mechanism of selective autophagic regulation of Fn14, but may also provide a more general view of their role in mediating autophagosome biogenesis from different membrane sources.
Collapse
|
50
|
Wang Y, Ren J, Liu Y, Liu R, Wang L, Yuan Q, He J, Nie Y, Xu J, Yu J. Preparation and evaluation of folic acid modified succinylated gelatin micelles for targeted delivery of doxorubicin. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|